Engineered Biomaterials Ebook
Engineered Biomaterials Ebook
Rishabha Malviya
Sonali Sundram Editors
Engineered
Biomaterials
Synthesis and Applications
Engineering Materials
This series provides topical information on innovative, structural and functional
materials and composites with applications in optical, electrical, mechanical, civil,
aeronautical, medical, bio- and nano-engineering. The individual volumes are
complete, comprehensive monographs covering the structure, properties, manufac-
turing process and applications of these materials. This multidisciplinary series is
devoted to professionals, students and all those interested in the latest developments
in the Materials Science field, that look for a carefully selected collection of high
quality review articles on their respective field of expertise.
Indexed at Compendex (2021) and Scopus (2022)
Rishabha Malviya · Sonali Sundram
Editors
Engineered Biomaterials
Synthesis and Applications
Editors
Rishabha Malviya Sonali Sundram
Department of Pharmacy Department of Pharmacy
Galgotias University Galgotias University
Greater Noida, Uttar Pradesh, India Greater Noida, Uttar Pradesh, India
© The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature
Singapore Pte Ltd. 2023
This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher, whether
the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse
of illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way, and
transmission or information storage and retrieval, electronic adaptation, computer software, or by similar
or dissimilar methodology now known or hereafter developed.
The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication
does not imply, even in the absence of a specific statement, that such names are exempt from the relevant
protective laws and regulations and therefore free for general use.
The publisher, the authors, and the editors are safe to assume that the advice and information in this book
are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or
the editors give a warranty, expressed or implied, with respect to the material contained herein or for any
errors or omissions that may have been made. The publisher remains neutral with regard to jurisdictional
claims in published maps and institutional affiliations.
This Springer imprint is published by the registered company Springer Nature Singapore Pte Ltd.
The registered company address is: 152 Beach Road, #21-01/04 Gateway East, Singapore 189721,
Singapore
Biomaterials are materials with novel properties that can enable them to come in
contact with the living tissue. They can be synthesised by a variety of processes and
engineered according to a specific use and application in order to mimic biological
phenomena or else be employed for some other process based on its use. There are
immense benefits of Biomaterials particularly in improving the quality of life bene-
fits apart from others. The progress in this field is dependent on interdisciplinary
research with scientists from diverse backgrounds like chemists, physicists, biolo-
gists, pharmaceutical scientists, medical doctors, engineers, material producers and
manufacturers etc. For medicinal purposes it can either be a diagnostic purpose or
therapeutic purpose. The main rationale behind writing this book is to cover all the
diverse aspects and the scientific knowledge associated with engineering of bioma-
terials as well as its utilisation in the healthcare field. This book attempts to explain
in detail the principles and foremost methods for the synthesis of biomaterials and
its application interconnected with prevention, mitigation, diagnosis and treatment
purposes. The readers of this book will get an idea about how biomaterials are
growing wider in terms of application as well as the increasing future opportunities
associated with them. This book can also serve as a useful source of information
for studying engineering biomaterials. Various methods for the synthesis and devel-
opment of biomaterials are provided in the contributed chapters. These methods
include those related to genetic engineering for the development of novel bioma-
terials, environmentally friendly approaches for the development of biomaterials
based on bone and tissue engineering, gold nanoparticles derived from microorgan-
isms as a synthetic method for developing biomaterials, and biomimetic approaches
for producing biomaterials.
Each chapter of this book highlights methods of synthesizing biomaterials and
how to apply them in healthcare. Not only the opportunities and advantages have
been covered but the possible challenges have been addressed so that the readers and
researchers get an idea about the obstacles and their means to overcome them.
It is my pleasure to present this forward for this book entitled Engineered Bioma-
terials: Synthetic Approach and Applications Edited by Dr. Rishabha Malviya.
vii
viii Foreword
Different chapters in this book have been contributed by the experts in their respective
fields.
I highly acknowledge all the authors as well as the editors for their endeavour and
efforts to compile this book and wish them all the very best.
Biomaterial can be defined as a material which is engineered for interacting with the
biological system for the healthcare purpose in diagnosis, mitigation and treatment
motives. Engineering biomaterials means the study of biomaterials. Biomaterials
have a property of biocompatibility which makes them suitable as well as safe for
use. Methods for the synthesis of biomaterials and all the aspects associated with it
are covered in this book. In the initial chapter information is given about the biomate-
rials which are derived naturally, its advances as well as opportunities. Then various
techniques are given for synthesis and development of biomaterials which includes
techniques associated with genetic engineering for the development of novel bioma-
terials, green methods for the development of bone and tissue engineering based
biomaterials, gold nanoparticles from a microorganism—a synthetic approach for
development of biomaterials and the biomimetic approaches for manufacturing of
biomaterials. A detail description about genetically induced biomaterial advances in
medical science is given in one of the chapters. Nowadays biomaterials are applied
widely in healthcare applications which are growing wider along with time. Along
with the synthetic approaches, the applications of biomaterials are also covered in
this book. The usage of nano-sized biomaterials in drug delivery systems, recent
trends associated with it as well as the future opportunities are covered here. Usage
of biomaterials in implant devices, stimuli responsive materials in controlled release
of drug, advanced tissue engineering with novel engineered biomaterials, collagen
based nanomaterials for delivering drug, photo responsive material for 4D printing
in tissue engineering, surface modified biomaterials in developing medical devices,
nano-porous metal foams as versatile nanoplatforms for drug delivery applications
and its properties, recent progress as well as challenges related to it, development
of cardiovascular biomaterials from collagenous tissues, stimuli responsive material
in controlled release of drug, gold nanoparticles along with their clinical applica-
tions and morphological study of silver nanoparticles supported on ti1-xcexo2, their
synthesis and antimicrobial applications are covered in the chapters of this book. The
students as well as researchers reading this book will get a detailed idea about what
are biomaterials, how they are synthesized and how they can be applied in health-
care for medical purposes. The readers will find the information useful and precise.
ix
x Preface
Careful attention has been given by the authors to avoid making the information
complex so that readers do not get confused while reading the book. Authors have
tried to cover recent points linked to biomaterials so that latest information can be
conveyed through this book. We thank the authors for putting their efforts in writing
this book.
Applications of Biomaterials
Nanostructured Biomaterials in Drug Delivery . . . . . . . . . . . . . . . . . . . . . . . 233
İbrahim Mizan Kahyaoğlu, Erdi Can Aytar, Alper Durmaz,
and Selcan Karakuş
xi
xii Contents
xv
xvi About the Editors
Ainil Hawa Jasni, Azlin Suhaida Azmi, Noor Illi Mohamad Puad,
Fathilah Ali, and Yusilawati Ahmad Nor
Abstract Biomaterials are materials that have been formed from or created by
biological organisms such as plants, animals, bacteria, fungus, and other forms of life
are referred to as biologically derived materials. Biomaterials are normally designed
to interface with biological systems, for the treatment, augmentation, or replace-
ment of biological functions. Across billions of years, life has been composed of and
existed within these biomaterial molecules, monomers, and polymers. For instance,
biomaterials of polysaccharides are sugars or starch polymers. Cellulose is the most
ubiquitous and abundant polysaccharide. Polysaccharides are found in the tissues
of both trees and humans. Meanwhile, natural biomaterials are substances that are
derived from natural sources such as plants, animals, or minerals, and are used
in medical and healthcare applications. Examples of natural biomaterials include
collagen, chitosan, silk, cellulose, hyaluronic acid, and bone minerals such as hydrox-
yapatite. These materials are attractive in the field of regenerative medicine and tissue
engineering due to their biocompatibility and biodegradability. Additionally, some
natural biomaterials can mimic the physical and chemical properties of the body’s
natural tissues, making them ideal for use in implants and scaffolds. Recent advances
in the production of natural biomaterials include the development of more efficient
and scalable manufacturing processes, which has made them more widely available
and accessible for use in medical applications. In addition, advances in the under-
standing of the biological interactions between these materials and the body have
allowed for the development of new and improved medical devices and therapies.
The use of natural biomaterials also provides unique opportunities for customization
and personalization in medical treatment. For example, natural biomaterials such
as collagen and hyaluronic acid can be engineered to meet specific patient needs,
such as tissue repair and regeneration, wound healing, and drug delivery. Overall,
natural biomaterials have shown great promise in many fields. This chapter’s goal
is to give readers a quick introduction to naturally derived biomaterials and their
advances and opportunities. For example, recent developments in the production of
natural biomaterials have made them more widely available and accessible for use
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 3
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_1
4 A. H. Jasni et al.
1 Introduction
Naturally derived biomaterials also offer promising solutions for sustainable mate-
rials. Many conventional materials used in industry rely on non-renewable resources
and often result in negative environmental impacts. In contrast, biomaterials can be
produced sustainably and may even be biodegradable. By leveraging these materials,
we can reduce our impact on the environment while still producing high-quality prod-
ucts. Advances in naturally derived biomaterials represent an incredible opportunity
for improving human livelihoods while also protecting our environment. As research
continues to explore the full range of applications for these materials, we can look
forward to even more exciting discoveries and innovations in the years to come. It’s
clear that the possibilities are truly remarkable, and we should all be excited about
what the future holds for this cutting-edge field.
Naturally derived biomaterials are materials that are sourced from biological sources
such as plants, animals, and microorganisms [1]. These materials are usually biocom-
patible and biodegradable, meaning they can be safely used in living organisms
without causing harm and can eventually be broken down by biological processes.
Biomaterials are materials that interact with biological systems for various purposes,
for instance the delivery of drugs, medical devices, and tissue engineering [2]. Natu-
rally derived biomaterials, obtained from natural sources such as animals, plants,
and microorganisms [3] have undergone substantial research and are utilised in
biomedical applications for their biocompatibility, biodegradability, and bioactivity.
Here are some of the reasons why naturally derived biomaterials are important in
various applications (Table 1).
This chapter covers a variety of closely connected topics to the development, prop-
erties, and applications of biomaterials that are derived from natural sources. The
topics which are covered in this chapter include:
6 A. H. Jasni et al.
6 Polysaccharides
6.1 Starch
Starch is a polysaccharide that acts as the main component for storing energy in
plants. Amylose and amylopectin, two different forms of glucose polymers, make up
the substance [15]. Amylopectin is a branching chain of glucose molecules, whereas
amylose is a linear chain [16]. Starch is a naturally occurring polymer composed
of glucose units that can be found in a variety of plant-based products, including
corn, wheat, rice, potatoes, and cassava [17]. It is frequently utilised as a thickening,
stabilizer, and gelling agent in the food sector because of its unique chemical and
physical properties [18].
Starch is insoluble in cold water but can be partially solubilized by heat or acid
hydrolysis, breaking down the starch into smaller glucose units. Starch has a high
molecular weight, which makes it viscous and difficult to dissolve [19] especially
in the electrospinning and membrane casting applications. Starch can be effectively
dissolved using ionic liquids, 4-methylmorpholine 4-oxide (NMMO), and dimethyl
sulfoxide (DMSO), with DMSO being the most cost-effective and soluble [20].
Raw starch must be chemically changed to make it more hydrophobic, stronger
Naturally Derived Biomaterials: Advances and Opportunities 9
Starch has a white, odourless, and tasteless appearance [21]. Starch has a large
capacity to store water and can hold up to 20 times its weight in liquid [22]. Starch
can form a gel when heated in water, allowing it to thicken food products and increase
their viscosity [23]. Starch is a renewable and biodegradable resource, making it
an attractive material for use in various applications beyond food. Natural derived
biomaterials derived from starch can include:
1. Bioplastics: As an alternative, starch-based bioplastics can be used to replace
traditional petroleum-based plastics and can be used in packaging materials [24],
disposable utensils, and other single-use products
2. Adhesives: Starch-based adhesives are used in paper and cardboard production
and can replace synthetic adhesives [25].
3. Textiles: Starch-based materials can be used in textiles as a sizing agent, which
helps prevent shrinkage during washing and ironing [26].
4. Medicine: Starch can be used as a binder in tablet formulations and as a
disintegrant to help tablets dissolve in the body [27].
5. Agriculture: Materials made of starch can be utilised as a biodegradable substitute
to synthetic mulch films used in agriculture [28].
6.2 Glycogen
6.3 Peptidoglycan
6.4 Cellulose
Insects, crustaceans, and other arthropods’ exoskeletons are made of the carbohy-
drate chitin It is also present in some fungi cell walls [41]. Chitin provides strength
and protection to these organisms. Chitin can be processed into nanofibers, nanopar-
ticles, and other nanostructures, which have special characteristics including a large
surface area, mechanical strength, and biocompatibility [42]. These nanomaterials
have potential applications in wound healing, tissue engineering, and drug delivery.
Another recent development in the use of chitin is when it comes to food packing
[43]. In the not-too-distant future, it has been reviewed by authors in [44] that a
significant amount of chitin and chitosan can be derived from mushrooms [44]. There
are bright prospects for the industrial use of fungus-sourced chitosan, especially
within the food and pharmaceutical industries, as a result of the comparisons made
between the physicochemical, functional, and biological traits of that substance and
that of marine chitin and chitosan [44].
Chitin has also been explored for its potential use in agriculture as a biopesticide.
Chitosan can act as a natural insecticide, disrupting the growth and development of
pests while being safe for humans and environment [45]. Additionally, chitin can be
used as a soil conditioner, enhancing soil fertility and plant growth [46]. It was found
that the exuviae of a particular insect species (mainly chitin) had enhanced bacteria
that grew after the topsoil underwent treatment with 10 g/kg in the rhizosphere of B.
oleracea. However, at a ratio of 1 g/kg, there were no appreciable changes in bacterial
abundance. The results are consistent with earlier research on chitin alterations, which
has shown to increase bacterial population abundance [47].
12 A. H. Jasni et al.
6.6 Alginate
Brown seaweed is the source of the natural polymer known as alginate, such as
kelp [52]. This environmentally friendly and renewable biomaterial exhibits several
intriguing qualities, including good mild gelation when adding divalent cations (such
as Ca2+ ), biocompatibility, low toxicity, and cost effectiveness. High-molecular-
weight alginate’s poor solubility and high viscosity, the aqueous solution’s poly-
electrolyte nature, the lack of suitable organic solvents, the substantial amount of
intra- and intermolecular hydrogen bonds, continue to be problems in alginate alone
applications [53]. Thus, it must be coupled with other polymers to enhance its
values [54] that had developed natural polymer high-strength hydrogels using gelatin
and hydrazide alginate (HAlg), which were then crosslinked to mimic the struc-
ture of collagen and glycosaminoglycans (GAGs) within the extracellular matrix,
respectively. The Gelatin-HAlg-DN hydrogels exhibit excellent biodegradability and
swelling consistency in physiological conditions, as well as the capacity to facilitate
cell binding and proliferation. In a rat model with critical size bone defects, psoralen-
loaded gelatin-HAlg-DN hydrogels effectively induced bone regeneration offering
intriguing potential as tissue engineering scaffolds [54].
A polymer present in the connective tissues called hyaluronic acid (HA) aids in
maintaining hydration and lubrication. It is also used in some cosmetic products
for its moisturizing properties [55]. Bioink additions include HA and multi-walled
carbon nanotubes (CNTs) in 3D bioprinting for fabrication of cartilage had been
reported [56]. HA acts as a molecule that is both structural and signalling. It possesses
extraordinary water-retention abilities that create a gel-like environment within the
tissue and give the entire structure flexibility for the structure [57].
Naturally Derived Biomaterials: Advances and Opportunities 13
6.8 Others
6.8.1 Inulin
6.8.2 Xylan
6.8.3 Proteins
Proteins are substantial, intricate molecules consisting of long chains of amino acids
[62]. They are found in every cell of every living organism and are involved in a variety
of functions, such as providing structural support, catalysing chemical reactions,
and transporting molecules within cells and throughout the body. A protein’s linear
sequence of amino acids makes up its primary structural component. These consistent
folding structures, alpha helices and beta sheets, make up a protein’s secondary
structure. The collection of curves and folds in a single linear chain of amino acids,
sometimes referred to as a polypeptide, is the tertiary structure of a protein. Last but
not least, macromolecules having multiple polypeptide chain structures or subunits
are known as proteins [63]. Figure 1 is the illustration each type of protein structure.
14 A. H. Jasni et al.
Fig. 1 Illustration of type of protein structure (Taken from open source, https://doi.org/10.1016/j.
bea.2021.100021)
Here are some examples of protein-based naturally derived biomaterials and their
functions:
• Structural proteins: Structural proteins, such as collagen, keratin and elastin,
provide support and shape to tissues and organs in the body [64].
• Transport proteins: Transport proteins, such as haemoglobin and albumin, bind
to and transport molecules, such as oxygen and nutrients, throughout the body.
For example, one of the most prevalent proteins in blood plasma, serum albumin
is crucial to all biological activities and has been used in several biomedical
procedures. Human albumin, bovine albumin, and ovalbumin, which are used to
manufacture biomaterials, are appropriate for use in bone regeneration because
they have the right microstructure, hydrophilicity, and biocompatibility [65].
• Contractile proteins: Actin and myosin are examples of contractile proteins that
cause muscles to contract and move.
In summary, proteins are necessary molecules that have several functions in living
things. Their unique properties, such as their ability to provide structural support,
and transport molecules, make them important in many material science applications,
such as food, medicine, and biotechnology.
Naturally Derived Biomaterials: Advances and Opportunities 15
Fig. 2 Collagen fibre structure, from fibrils and molecules to chains, together with amino acid
sequence of Hydroxyproline, Proline, and Glycine. (Created with biorender)
6.8.4 Collagen
The protein collagen, which is the most prevalent in the human body, gives numerous
tissues their structural support [66]. It has been frequently employed beyond the field
of tissue engineering owing to its remarkable biocompatibility, low immunogenicity,
and ability to support cell adhesion and proliferation. Collagen is made up of three
chains. The chains are linked to form a triple helix. Because glycine is the smallest
amino acid, it permits the chain to form a tight shape and endure stress [67]. Figure 2
is the structure of the collagen fibre.
Silk is a naturally occurring protein fibre produced by silkworms. It has been used in
tissue engineering, implant coating and others due to its biocompatibility, biodegrad-
ability, and ability to form scaffolds with tuneable mechanical properties [68]. A
recent study reported the possibility of encouraging the development of bone-like
tissue from human dental pulp stromal cells (hDPSCs) both in in vitro as well as
in vivo within porous Bombyx Mori silk structures by employing the specific HDAC2
and 3 inhibitor MI192. Fabricated and evaluated were scaffolds made of 2 and 5
wt.% silk. In contrast to the 2 wt% silk scaffolds, the scaffolds with a weight of
5% exhibit larger internal lamellae, slower rates of expansion and deterioration, and
higher torsional moduli. The alkaline phosphatase activity of hDPSCs on a 5 wt%
16 A. H. Jasni et al.
Fig. 3 The technique used for fabrication silk scaffolds for biomedical applications (adapted from
open source, https://doi.org/10.3390/polym14153110)
silk scaffold was dramatically increased by pre-treatment with MI192 (ALP). Histo-
logical examination proved that, in contrast to the control group, the use of MI192-
pretreated hDPSCs-silk scaffolds improved bone extracellular matrix (ALP, Col1a,
and OCN) deposition and mineralization. After six weeks of subcutaneous implanta-
tion in mice, the MI192-pretreated hDPSCs-silk scaffold constructs increased vascu-
larization and extracellular matrix mineralization compared to the untreated control.
After six weeks of subcutaneous implantation in nude mice, the MI192-pretreated
hDPSCs-silk scaffold constructs increased vascularization and extracellular matrix
mineralization compared to the untreated control [69]. Figure 3 is the workflow of
silk fabrication and application.
6.8.6 Gelatin
6.8.7 Elastin
6.9 Others
6.9.1 Fibrin
Fibrin is a naturally occurring protein that aids in the formation of clots and wound
healing. It is utilized in tissue engineering to form hydrogels that can support cell
growth and migration [71].
6.9.2 Lipids
Lipids are a wide class of naturally occurring macromolecules present in plants and
animals [72]. They are distinguished by their hydrophobic characteristics and are
used in a number of biomaterials research applications. One major use of lipids
is in the creation of liposomes. Liposomes are spherical vesicles consisting of a
bilayer of phospholipids that can be used to encapsulate drugs and other therapeutic
molecules for targeted drug delivery [73]. The inside of the liposome is formed by
the hydrophilic portions of the phospholipids, which contain the drug, while the
hydrophilic head groups form the outer shell, allowing the liposome to interact with
biological systems [74].
Lipids are also used in the creation of biodegradable polymers. Lipid-based poly-
mers can be designed to degrade in vivo, making them suitable for use in the fields of
tissue engineering and drug delivery [75]. They are additionally employed to produce
micro- and nanoparticle for targeted medication delivery [76]. Another use of lipids is
the formation of lipid membranes. Lipid membranes are used in biomimetic systems
to mimic the properties of natural cell membranes [77]. They can be used for a
variety of purposes, including water treatment, biological sensors, and drug detection
[78]. Lipids are additionally useful to create hydrogels. Hydrogels comprise three-
dimensional, water-swollen polymer networks that can be useful for drug delivery,
wound care, and tissue engineering. Lipids can be incorporated into hydrogels to
provide mechanical support and improve cell adhesion and proliferation [79].
Overall, lipids are a versatile biomaterial with many potential applications in
biomaterial science. Their hydrophobic properties and ability to self-assemble make
them appealing candidates for drug delivery, tissue engineering, and biomimetic
system.
Fatty acids are naturally occurring biomolecules that are commonly found in plant
and animal tissues. They are composed of long hydrocarbon chains with a carboxyl
group at one end, making them amphipathic like phospholipids [72]. Fatty acids have
many uses in biomaterial science due to their biocompatibility, biodegradability, and
versatility.
18 A. H. Jasni et al.
One major use of fatty acids is in the creation of bio-based polymers. Fatty acids
can be used as monomers to create polymers such as polyesters, polyamides, and
polyurethanes. These polymers are used for drug delivery, tissue engineering, and
exterior coatings thar are only a few of the potential applications [80]. Hydrogels are
also manufactured using fatty acids. Hydrogels are cross-linked networks of liquid-
swollen polymers which are able to be utilized for the administration of drugs, wound
healing, and other applications. tissue engineering. Fatty acids can be incorporated
into hydrogels to provide mechanical support and improve cell adhesion and prolif-
eration. A recent study showed that hydrogenated phosphatidylcholine and oleic acid
were used to create a highly hydrated hydrogel (95% water) for potential biomedical
uses [81].
Another application of fatty acids is in the production of surfactants. Surfactants
are molecules that can reduce the surface tension of liquids and increase their ability to
mix with other substances. Fatty acids can be used to create natural surfactants that are
biodegradable and non-toxic, making them suitable for use in personal care products,
detergents, and other industrial applications. It has been known that oleochemical-
based surfactants outperform petroleum-based ones in terms of biocompatibility
and biodegradability [82]. Sorbitan esters are commercially known as “Span” in
the contemporary industrial sector, specifically Span 80, a biodegradable surfactant
made from oleic acid and sorbitol-based sugar alcohol. These esters are commonly
used in the development and formulation of water-in-oil emulsions due to their rela-
tive hydrophobicity. Sorbitan esters are widely derivatized by combining with ethy-
lene oxide to form sorbitan ester ethoxylates, which enhances their hydrophilicity.
Polyethoxylated sorbitan monoesters, popularly known as “Tween” in the industry,
are surfactants that are ideal for creating oil-in-water emulsions [83, 84].
Overall, fatty acids are a versatile biomaterial with many potential applications
in biomaterials science. Their biocompatibility, biodegradability, and versatility
make them an attractive option for the development of sustainable and biologically
compatible materials.
6.9.4 Phospholipids
Phospholipids are naturally occurring biomolecules that are found in cell membranes
and other biological structures. They are amphipathic, meaning that they have both
hydrophilic (water-loving) and hydrophobic (water-repelling) regions [85]. This
unique property makes phospholipids useful in a variety of applications in biomaterial
science.
One major use of phospholipids is in drug delivery systems. Phospholipid-based
liposomes can be used to encapsulate drugs and other therapeutic molecules, allowing
for targeted delivery to specific tissues or cells. The liposome’s outer shell is formed
by the hydrophilic head regions of the phospholipids, while the hydrophobic tails
form the interior, where the drug is contained. The liposome can then be designed to be
released by the reaction to certain stimuli, such as changes in pH or temperature, and
the medication is released. The optimum phospholipid for managing the release of
Naturally Derived Biomaterials: Advances and Opportunities 19
the anaesthetic bupivacaine (BUP) medication from liposomal depots and controlling
the regulated aggregation of the liposomes is phosphatidylglycerol (PG) [86].
Phospholipids are also used in the creation of biomimetic membranes, which are
synthetic structures designed to mimic the properties of natural cell membranes.
These membranes have a wide range of uses, including water filtration, biosensors,
and drug screening. The amphipathic nature of phospholipids allows them to form
bilayers, which can be used as the foundation for these biomimetic membranes. The
best exemplar of this is a study done by [87] where the cell membrane phospho-
lipids are functions as dentin’s mineralization sites and potential raw materials for
bottom-up strategies aiming for quicker and more intricate production of dentin-like
structures.
Phospholipids are also used in tissue engineering applications. They can be added
to scaffolds to offer structural support while also promoting cell adhesion and prolif-
eration. Because of their improved blood compatibility and cytocompatibility, elec-
trospun poly (lactic-co-glycolic acid) (PLGA) membranes were a suitable case in
point [88]. Overall, phospholipids’ distinct features make them flexible biomaterials
with numerous uses in pharmaceutical delivery, tissue engineering, and biomimetic
membrane design.
6.9.5 Waxes
Wax is a simple lipid that is generated by esterifying a long-chain alcohol with a fatty
acid. Alcohol can have 12–32 carbon atoms. Waxes are found in nature as coatings
on plants and stems. The wax protects the plant from excessive water loss [89]. In
practise, cellulose combined with a trace amount of plant-derived wax (nonacosane-
10-ol and nonacosane-5, 10-diol) has a higher mechanical strength and modulus of
elasticity [90] than synthetic plastic. The diffusion coefficients of oxygen, nitrogen,
and water molecules through this material were found to be at least 50% lower than
those found in polyethylene. It is an environmentally friendly, long-lasting packaging
material with outstanding mechanical, thermal, and barrier properties [90].
For example, collagen can be extracted from animal tissues using acid or enzy-
matic digestion [92], while cellulose can be extracted from plants using chemical
or mechanical methods [93].
2. Purification: After extraction, the material is usually purified to remove impurities
and obtain a pure form of the material. Purification methods include filtration,
centrifugation, and chromatography [94].
3. Modification: Modification of naturally derived biomaterials is frequently
required to modify their qualities and adapt them to certain uses. Modification
can be achieved through chemical or physical methods. For example, cross-
linking of collagen can improve its mechanical strength and stability, while
surface modification of cellulose can improve its biocompatibility [95].
4. Characterization: Characterization of naturally derived biomaterials is essen-
tial to determine their properties and suitability for the intended application.
Characterization methods include spectroscopy, microscopy, and mechanical
testing.
5. Quantification: Quantification is the act of assigning a numerical value to a
measurement of anything, or counting the quanta of whatever is being measured.
Quantification creates a defined method of measurement that permits statistical
operations and mathematical computations to be performed [96].
6. Pilot production: A pilot study, pilot project, pilot test, pilot production or pilot
experiment is a small-scale preliminary study undertaken prior to the performance
of a full-scale research project to evaluate feasibility, duration, cost, adverse
occurrences, and to enhance the study design [97]. Figure 4 is the summary of
biomaterials synthesis workflow.
8 Chemical Modification
9 Physical Modification
10 Enzymatic Modification
2. Degradation: This involves the use of enzymes to selectively degrade the bioma-
terials to control the rate of release of bioactive molecules, drugs, or cells from
the biomaterial. For example, enzymes like metalloproteinase [111] collagenase,
hyaluronidase, and chondroitinase can be used to selectively degrade collagen,
hyaluronic acid, and chondroitin sulphate, respectively [112].
3. Cross-linking: This involves the use of enzymes to cross-link biomaterials to
enhance their mechanical and structural properties. For example, enzymes like
transglutaminase can be used to cross-link proteins, such as gelatin and collagen,
to form stable hydrogels [113].
4. Surface cleaning: This involves the use of enzymes to remove contaminants and
impurities from the surface of the biomaterial to improve its biocompatibility.
For example, enzymes like lipase and protease can be used to remove lipids and
proteins from the surface of the biomaterial [114].
Enzymatic modification of biomaterials is an intriguing approach for improving
biocompatibility and performance for a variety of purposes, namely drug delivery,
tissue engineering, and medical devices.
12 Industrial Applications
Table 2 (continued)
No. Type of biomaterials Findings Study
2 Collagen • Collagen nanostructures using the [119]
scales of fish were produced for
the very first time using
desolvation techniques
• According to the histological and
macroscopical study, the synthetic
fish scale’s collagen nanomaterials
aided in the healing process with
little toxicity compared to the
saline group
3 Alginate-chitosan+HA • Alginate-chitosan microbeads and [120]
thermo-responsive hyaluronic
acid-poly(N-isopropylacrylamide)
(HA-pNIPAM) hydrogels have
been created as bioresorbable local
bacteriophage (Staphylococcus
aureus phage ISP and
Pseudomonas aeruginosa phage
LUZ19) delivery systems to
permit both delayed and quick
phage release
• Both phages were gradually
released hydrogels HA-pNIPAM
during the course of 21 days. The
short-tailed LUZ19 was
continuously released from the
microbeads for 21 days, whereas
the long-tailed ISP released in
bursts at first and then at a
declining rate over time
4 Fibrin • Research on the drug release and [121]
antibacterial properties of
platelet-rich fibrin (PRF), a natural
vehicle for the administration of
antibiotics
• Following oral surgery, using
antibiotic-loaded PRF may reduce
the risk of post-operative infection,
replace or enhance systemic
antibiotic therapy, and maintain
the healing properties of PRF
(continued)
26 A. H. Jasni et al.
Table 2 (continued)
No. Type of biomaterials Findings Study
5 Cellulose • Using an in-situ co-precipitation [122]
approach, Magnetic iron oxide
(MIO)-incorporated waste tissue
sheets (WTP) and sugarcane
bagasse (SCB) were incorporated
to make WTP/MIO and SCB/MIO
nanocomposite particles (NCPs)
• By including MIO-NPs into the
cellulose matrix, the swelling
capacity, drug loading capacity,
and drug release time were all
improved
Implant coatings
1 Laminin • A poly (D, L-lactide) [123]
(PDLLA)-Laminin 332 (LN332)
hybrid film that displayed timed
release of LN332for up to 28 days
was successfully created using a
layer-by-layer manufacturing
process
• It accelerated gingival
mesenchymal stem cells’ epithelial
growth and promoted their
adhesion, dissemination, and
proliferation
2 Hydroxyapatite • 10 implants featuring 10 implants [124]
having a dual acid-etching (DAA)
surface and a nanostructured
hydroxyapatite coating (HAnano).
was given to ten lambs (aged 2–4)
• Low-density bone in sheep after
28 days with HAnano enhances
bone development as compared to
DAA surface
3 HA • Tannic acid (TA) and hyaluronic [125]
acid (HYA) use is necessary to
lower the rate of corrosion of
implants made of magnesium
alloys
• The magnesium alloy’s corrosion
rate was lowered by the TA-HYA
coating from 7.379 mm/year to
0.204 mm/year in in vitro
corrosion testing employing Tafel
polarisation
(continued)
Naturally Derived Biomaterials: Advances and Opportunities 27
Table 2 (continued)
No. Type of biomaterials Findings Study
4 Silk • FibroFix Cartilage P™ [126]
(FibroFix™)—commercial brand
name
• Substantial bone repair and strong
implant tissue integration, which,
in our opinion, will speed up the
healing process (promote tissue
regeneration)
5 Collagen • An innovative injectable medicinal [127]
product on primary cultures of
human gingival fibroblasts (hGF),
dental SKIN BioRegulation (Guna
S.p.A., Milan, Italy) was put to the
test
• It is made of type I collagen of
porcine origin
• Significant improvement in 48-h
viability of hGF-grown cells (p =
0.05) and 24-h wound healing (p
= 0.001)
Table 3 (continued)
No. Sample type Findings References
5. Cellulose • Bio sequins fabricated [132]
from bioplastic derived
from tree woods namely
‘Bio Iridescent’
• Traditional sequins are
produced of polyvinyl
chloride (PVC), a
polyester film, often
known as Mylar, which
poses serious
environmental and health
dangers by creating
hazardous, bioaccumulate
compounds, including
hormone disruptors and
carcinogens like
phthalates
Packaging industry
6. Glycol • monoethylene glycols [133]
(meg) and monopropylene
glycols (mpg) from woods
• a genuine greener option
for making recyclable pet,
polyesters, and industrial
liquids
Cosmetic industry
7. Glycol • A fully environmentally [133]
friendly option for the
manufacture of carrier
liquids. the secret to
providing a generation of
resins, detergents,
de-icing agents, and
cosmetics that is
sustainable and renewable
8. PLA+Polyhydroxyalkanoates (PHA) • The cosmetic case is made [134]
of polylactic acid (PLA)
and amorphous PHA
technology for a private
brand, ‘Wakemake’
• An alternative substitute
of acrylonitrile butadiene
styrene (ABS) casing
(continued)
30 A. H. Jasni et al.
Table 3 (continued)
No. Sample type Findings References
Medical industry
9. HA • HA-coated gold [135]
nanoparticles (AuNP-HA)
synthesised in one pot.
Usage as a drug
(Sulfasalazine) (SSZ),
delivery vehicle
• AuNP-HA exhibits a
promising material as a
modern medicine delivery
method with
characteristics of
controlled release
10. Collagen+Glycogen+Hydroxyapatite • Composite hydrogels [31]
functions as bone repair
scaffold
• When cultivated on the
composite hydrogels,
bone mesenchymal stem
cells displayed favourable
cell adhesion, vitality, and
proliferation
Construction industry
11. Cellulose • Food leftovers are [136]
combined with water and
seasoning, then heated up
and pressed into a mould.
It is called ‘Fabula’
• Has a bending strength
four times more than that
of concrete and is
primarily made of
vacuum-dried, ground-up
fruit peels and coffee
grounds
12. Alginate+Oyster shells • ‘Ostra’-A biomaterial [136]
resembling ceramic and
cement that is created
from used oyster shells
and seaweed extract
• Sourced from local
seafood restaurants in Italy
Naturally Derived Biomaterials: Advances and Opportunities 31
Naturally derived biomaterials hold great promise for a variety of commercial and
biological uses. However, there are also several concerns that must be completely
addressed to realize their potential.
Some of the challenges and future opportunities associated with naturally derived
biomaterials are:
• Standardization and quality control: Naturally derived biomaterials can have
batch-to-batch variability, which can affect their properties and performance.
Standardization and quality control measures need to be developed to ensure
consistent quality of naturally derived biomaterials [138].
• Cost-effectiveness: Naturally derived biomaterials can be expensive to produce
and process, which can limit their use in industrial applications [139]. Improve-
ments in production efficiency and processing methods can help to reduce costs
and increase the scalability of naturally derived biomaterials.
• Mechanical and chemical properties: The mechanical and chemical properties
of naturally derived biomaterials can vary widely, which can make it difficult to
control their properties for specific applications. Advances in processing methods
and functionalization techniques can help to overcome these limitations [140].
32 A. H. Jasni et al.
Advances in biotechnology and material science are likely to drive new opportu-
nities for the development of naturally derived biomaterials. For example, genetic
engineering and synthetic biology approaches can be used to tailor the natural bioma-
terials’ characteristics for particular uses, while utilising novel processing techniques
that can help to overcome challenges related to scalability and processing efficiency.
Genetic engineering allows for the modification of an organism’s DNA to produce
specific traits or characteristics that can be beneficial for certain applications [142].
For example, scientists can engineer bacteria to produce large quantities of proteins
or other biomolecules that can be used in medical or industrial settings. Similarly,
genetic engineering can be used to modify the properties of natural biomaterials such
as silk or collagen, allowing them to be tailored for specific applications. Synthetic
biology, on the other hand, involves creating new biological systems and designing
existing ones or organisms with desired properties or functions [143]. This approach
can be used to create entirely new biomaterials that do not exist in nature, such as
biofuels or biodegradable plastics.
Both genetic engineering and synthetic biology can be used to improve the prop-
erties of natural biomaterials for specific applications. For example, scientists can
use genetic engineering to modify the properties of silk to make it more elastic or
stronger. Synthetic biology can also be used to create new biomaterials with unique
properties, such as self-healing materials or materials that respond to changes in their
environment.
In addition to genetic engineering and synthetic biology, novel processing tech-
niques can also be used to enhance the functionality and performance of natural
biomaterials. For example, scientists can use electrospinning or 3D printing to
construct scaffolds for delivery of drugs or tissue engineering. These methods enable
fine-grained control over the composition and characteristics of the biomaterials,
enabling them to be tailored for specific applications.
The capacity of natural biomaterials to combine with nanotechnology is perhaps
the most fascinating part of them. By combining the unique properties of natural mate-
rials with the precision of nanotechnology, researchers can create smart biomaterials
Naturally Derived Biomaterials: Advances and Opportunities 33
that having the capacity to revolutionize a vast area of industries. The possibilities
are truly endless.
14 Conclusion
In summary, while there are challenges associated with naturally derived bioma-
terials, ongoing research and development efforts hold significant promise for the
future of these materials in an extensive array of manufacturing and biomedical
industries. Overall, this chapter delivers a summary of the synthesis of natural bioma-
terials and their possible applications, emphasizing the need for continued research
in this field. Regardless of the approach used, sustainability is always an important
consideration when synthesizing biomaterials. By utilizing renewable resources and
minimizing environmental impact, researchers can create more sustainable bioma-
terials that benefit both the users and the planet. This is particularly significant given
the growing concern about environment variation besides the requirement for more
eco-friendly solutions in all areas of life. As more research is conducted, we will
undoubtedly discover even more applications for these incredible materials, making
a positive impact on human health and our planet’s future.
Naturally Derived Biomaterials: Advances and Opportunities 35
References
1. Coppola, G., Gaudio, M.T., Lopresto, C.G. et al.: Earth Syst. Environ. 5, 231–251 (2021)
2. Sciences, Penn State College of Agricultural. What is biomaterial? Department of Agricul-
tural Economics, Sociology and Education (2023). https://aese.psu.edu/teachag/curriculum/
modules/biomaterials/what-is-a-biomaterial#:~:text=A%20material%20derived%20from%
2C%20or,injury%20or%20growing%20biological%20cells
3. Leong, K., Quek, C.H., Basu, B., Chan, B., Goodman, S.B., Le Visage, C., Liang, X.-J.:
Science Direct. Biomaterials (2023). https://www.sciencedirect.com/journal/biomaterials
4. Birajdar, M.S., Joo, H., Koh, W.G. et al.: Biomater. Res. 8, 25 (2021)
5. Fowler, S.: Introduction to the Chemistry of Life. Charles Molnar and Jane Gair. Concepts of
Biology (2021)
6. Cole, L., Kramer, P.R.: Macronutrients. Human Physiology, Biochemistry and Basic
Medicine, pp. 157–164. Academic Press (2016)
7. Biology. Lipid. Biology Online (2022). https://www.biologyonline.com/dictionary/lipid
8. Ngwuluka, N.C.: Responsive polysaccharides and polysaccharides-based nanoparticles for
drug delivery. [book auth.] Nedal Y. Abu-Thabit Abdel Salam Hamdy Makhlouf. Stimuli
Responsive Polymeric Nanocarriers for Drug Delivery Applications, pp. 531–554. Woodhead
Publishing (2018)
9. Wang, F., Li, P., Chu, H.C., Lo, P.K.: Biosensors (Basel) 12(2), 93 (2022)
10. Muncie, J.M., Weaver, V.M.: Curr. Top. Dev. Biol. 130, 1–37 (2018)
11. Badylak, L.T., SaldinMadeline, C., CramerSachin, S., VelankarLisa, J., WhiteStephen, F.:
Elsevier BV, pp. 1–46 (2017)
12. Chempages. Biomolecules: Carbohydrates. Polysaccharides (2023). https://www2.chem.
wisc.edu/deptfiles/genchem/netorial/modules/biomolecules/modules/carbs/carb6.htm
13. Mohammed, A.S.A., Naveed, M., Jost, N.: J. Polym. Environ. 29(8), 2359–2371 (2021)
14. Helmenstine, A.M.: Polysaccharide Definition and Functions. Thought Co. (2020). https://
www.thoughtco.com/polysaccharide-definition-and-functions-4780155
15. Zhang, Y.H.P.: 9—Starch: A high-density chemical hydrogen storage compound for PEM
fuel cells. [book auth.] Antonio M. Chaparro Paloma Ferreira-Aparicio. Portable Hydrogen
Energy Systems, pp. 161–173. Academic Press (2018)
16. Kadokawa, J.: Enzymatic synthesis of functional amylosic materials and amylose analog
polysaccharides. [book auth.] Katja Loos Nico Bruns. Methods in Enzymology, pp. 189–213.
Academic Press (2019)
17. Falua, K.J., Pokharel, A., Babaei-Ghazvini, A., Ai, Y., Acharya, B.: Polymers 14(11), 2215
(2022)
18. Omoregie Egharevba, H.: IntechOpen (2020)
19. Tian, J., Deng, H., Huang, M., Liu, R., Yi, Y., Dong, X.: Electrospun Nanofibers for Food
and Food Packaging Technology. [book auth.] Xianfeng Wang, Jianyong Yu Bin Ding. Micro
and Nano Technologies: Electrospinning: Nanofabrication and Applications, pp. 455–516.
William Andrew Publishing (2019)
20. Ren, F., Wang, J., Yu, J., Zhong, C., Xie, F., Wang, S.: ACS Omega 6(41) (2021)
21. Cengage. Encyclopedia.com. Organic Chemistry: Starch (2018). https://www.encyclopedia.
com/science-and-technology/chemistry/organic-chemistry/starch
22. Annie, M.: Moisture and Mosses. Mountain Moss (2023). https://www.mountainmoss.com/
pages/moisture-and-mosses
23. Bakerpedia. What is Starch? Starch (2023). https://bakerpedia.com/ingredients/starch/
24. do Val Siqueira, L., La Fuente Arias, C.I., Maniglia, B.C., Tadini, C.C.: Current Opinion in
Food Science 122–130 (2021)
25. Gadhave, R., Gadhave, C.: Open J. Polym. Chem. 12, 55–79 (2022)
26. Leverette, M.M.: How to Use Laundry Starch and Sizing? The Spruce, New York (2021)
27. Apeji, Y.E., Kaigama, R.T., Ibrahim, S.H., Anyebe, S.N., Abdussalam, A.O., Oyi, A.R.: Turk.
J. Pharm. Sci. 19(5), 513–520 (2022)
36 A. H. Jasni et al.
28. Gamage, A., Liyanapathiranage, A., Manamperi, A., Gunathilake, C., Mani, S., Merah, O.,
Madhujith, T.: Sustainability 14(10), 6085 (2022)
29. FlexBooks. Polysaccharides. Chemistry For High School (2023). https://flexbooks.ck12.org/
cbook/ck-12-chemistry-flexbook-2.0/section/26.3/primary/lesson/polysaccharides-chem/
30. Gagliardi, A., Giuliano, E., Venkateswararao, E., Fresta, M., Bulotta, S., Awasthi, V., Cosco,
D.: Front. Pharmacol. 12, 601626 (2021)
31. Zhang, X., Zhou, J., Ying, H., Zhou, Y., Lai, J., Chen, J.: ACS Sustain. Chem. Eng. 8(4),
2106–2114 (2020)
32. Salton, M.R.J., Kim, K.S.: Structure. In: Baron, S. (Ed.) Medical Microbiology, 4th edn.
Galveston (TX): [book auth.] Baron S. Medical Microbiology 4th Edition. University of
Texas Medical Branch at Galveston (1996)
33. Kim, S.J., Chang, J., Singh, M.: Biochim. Biophys. Acta 1848(1 Pt B), 350–62 (2015)
34. Garde, S., Chodisetti, P.K., Reddy, M.: EcoSal Plus 9(2) (2021)
35. Davis, K.M., Weiser, J.N.: Infect. Immun. 79(2), 562–70 (2011)
36. Godoy, M.G., Amorim, G.M., Barreto, M.S., Freire, D.M.G.: Agricultural Residues as
Animal Feed: Protein Enrichment and Detoxification Using Solid-State Fermentation. [book
auth.] Christian Larroche, Carlos Ricardo Soccol Ashok Pandey. Current Developments in
Biotechnology and Bioengineering, pp. 235–256. Elsevier (2018)
37. Klemm, D., Heublein, B., Fink, H.-P., Bohn, A.: Angewandte Chemie (International ed. in
English) 44, 3358–93 (2005)
38. Norizan, M.N., Shazleen, S.S., Alias, A.H., Sabaruddin, F.A., Asyraf, M.R.M., Zainudin,
E.S., Abdullah, N., Samsudin, M.S., Kamarudin, S.H., Norrrahim, M.N.F.: Nanomaterials
(Basel) 12(19), 3483 (2022)
39. Deng, L., Huang, Y., Chen, S., Han, Z., Han, Z., Jin, M., Qu, X., Wang, B., Wang, H., Gu, S.:
Carbohydr. Polym. 308, 120647 (2023)
40. Bierach, C., Coelho, A.A., Turrin, M. et al.: Archit. Struct. Constr. (2023)
41. Elieh-Ali-Komi, D., Hamblin, M.R.: Int. J. Adv. Res. (Indore) 411–427 (2016)
42. Arockianathan, M.: 4 - Chitin-based nanomaterials. [book auth.] Ashok Kumar, Tuan Anh
Nguyen, Swati Sharma, Yassine Slimani Shamsher Kanwar. In: Micro and Nano Technologies:
Biopolymeric Nanomaterials, pp. 61–99. Elsevier (2021)
43. Stefanowska, K., Woźniak, M., Dobrucka, R., Ratajczak, I.: Materials, vol. 16, p. 1579 (2023)
44. Alimi, B.A., Pathania, S., Wilson, J., Duffy, B., Frias, J.M.C.: Int. J. Biol. Macromol. 237,
124195 (2023)
45. Saberi Riseh, R., Hassanisaadi, M., Vatankhah, M., Babaki, S., Ait Barka, E.: Int. J. Biol.
Macromol. 220 (2022)
46. Application of Chitosan in Agriculture. Linkedin. (2022). https://www.linkedin.com/pulse/
application-chitosan-agriculture-cathy-liu
47. Wantulla, M., Joop J.A. van Loon, Dicke, M.: Appl. Soil Ecol. 188 (2023). ISSN 0929-1393
48. Iber, B., Kasan, N., Torsabo, D., Omuwa, J.: J. Renew. Mater. 10, 42–49 (2021)
49. Parvin, N. et al.: 9, 64 (2023)
50. López-Valverde, N., Aragoneses, J., López-Valverde, A., Rodríguez, C., Macedo de Sousa,
B., Aragoneses, J.M.: Front. Bioeng. Biotechnol. 10, 907589 (907589)
51. Teixeira-Santos, R., Lima, M., Gomes, L.C., Mergulhão, F.J.: iScience 24(12), 103480 (2022)
52. Pereira, L., Cotas, J.: Introductory Chapter: Alginates—Alginates—Recent Uses of This
Natural Polymer. [A General Overview [Internet].] IntechOpen (2020). https://doi.org/10.
5772/intechopen.88381
53. Farshidfar, N., Iravani, S., Varma, R.S.: Alginate-Based Biomaterials in Tissue Engineering
and Regenerative Medicine 21(3), 189 (2023). Marine Drugs. https://doi.org/10.3390/md2
1030189
54. Wu, T., Liu, L., Gao, Z., Cui, C., Fan, C., Liu, Y., Mingyuan Di, A., Yang, Q., Xu, Z., Liua,
W.: Biomaterials Science (2023)
55. Best Summer Skincare Products of 2023. Outlook India (2023). https://www.outlookindia.
com/business-spotlight/best-summer-skincare-products-of-2023-news-271961
56. Szymański, T., Semba, J.A., Mieloch, A.A. et al.: Sci. Rep. 13, 646 (2023)
Naturally Derived Biomaterials: Advances and Opportunities 37
57. Kwon, H., Brown, W.E., Lee, C.A. et al.: Nat. Rev. Rheumatol. 15, 550–570 (2019)
58. Hussain, M.A., Haseeb, M.T., Muhammad, G., Tahir, M.N.: Inulin Type Fructan: A Versatile
Functional Material for Food and Healthcare. [book auth.] M., Sheardown, H., Al-Ahmed,
A. Jafar Mazumder. Functional Biopolymers. Polymers and Polymeric Composite (2019)
59. Parın, F.N. et al.: Polymers 15, 1002 (2023)
60. Paz-Gómez, G., del Caño-Ochoa, J.C., Rodríguez-Alabanda, O., Romero, P.E., Cabrerizo-
Vílchez, M., Guerrero-Vaca, G., Rodríguez-Valverde, M.A.: Coatings 9, 5–29 (2019)
61. Martin Gericke, L.H.S., Heinze, T.: Carbohydrate Polymers 300, 120251 (2023). ISSN 0144-
8617
62. What are proteins and what do they do? Medline Plus (2023). https://medlineplus.gov/gen
etics/understanding/howgeneswork/protein/#:~:text=Proteins%20are%20made%20up%20o
f,combined%20to%20make%20a%20protein
63. Education, Nature. Protein Structure. Scitable (2023). https://www.nature.com/scitable/
topicpage/protein-structure-14122136/#:~:text=The%20overall%20structure%20of%20t
he,and%20beta%20sheets%20(red).&text=The%20primary%20structure%20of%20a,protei
n’s%20unique%20three%2Ddimensional%20shape
64. Elastin. Cleveland Clinic (2023). https://my.clevelandclinic.org/health/body/22482-elastin
65. Mengmeng Jin, I., Zhu, S., Hou, Y.: ACS Biomaterials Science and Engineering Article ASAP
(2023)
66. What is collagen, and why do people use it? Medical News Today. [Online] Healthline Media
(2022). https://www.medicalnewstoday.com/articles/262881
67. Wu, M., Cronin, K., Crane, J.S.: Biochemistry, Collagen Synthesis. Treasure Island (FL).
StatPearls Publishing (2023)
68. Manoukian, O.S., Sardashti, N., Stedman, T., Gailiunas, K., Ojha, A., Penalosa, A., Mancuso,
C., Hobert, M., Kumbar, S.G.: Biomaterials for Tissue Engineering and Regenerative
Medicine. [book auth.] Roger Narayan. Encyclopedia of Biomedical Engineering. Elsevier
(2019)
69. Man, K., Joukhdar, H., Manz, X.D. et al.: Cell Tissue Res. 388, 565–581 (2022)
70. Loureiro dos Santos, L.A.: Natural Polymeric Biomaterials: Processing and Properties.
Reference Module in Materials Science and Materials Engineering. Elsevier (2017)
71. Murphy, K.C., Whitehead, J., Zhou, D., Ho, S.S., Leach, J.K.: Acta Biomater. 64, 176–186
(2017)
72. Ahmed, S., Shah, P., Ahmed, O.: Biochemistry, Lipids. StatPearls Publishing, Treasure Island
(FL) (2023)
73. Nsairat, H., Khater, D., Sayed, U., Odeh, F., Al Bawab, A., Alshaer, W.: 8(5), e09394 (2022).
Heliyon
74. Nakhaei, P., Margiana, R., Bokov, D.O., Abdelbasset, W.K., Jadidi Kouhbanani, M.A., Varma,
R.S., Marofi, F., Jarahian, M., Beheshtkhoo, N.: Front. Bioeng. Biotechnol. (2021)
75. Farasati Far, B., Naimi-Jamal, M.R., Sedaghat, M., Hoseini, A., Mohammadi, N., Bodaghi,
M.: J. Funct. Biomater. 14(2), 115 (2023)
76. Musielak, E., Feliczak-Guzik, A., Nowak, I.: Materials (Basel) 15(2), 682 (2022)
77. Luchini, A., Vitiello, G.: Biomimetics (Basel) 6(1), 3 (2021)
78. Nikoleli, G.P., Nikolelis, D.P., Siontorou, C.G., Nikolelis, M.T., Karapetis, S.: Membranes
(Basel) 8(4), 108 (2018)
79. Syed Azhar, S.N.A., Ashari, S.E., Zainuddin, N., Hassan, M.: UPM, Serdang, Selangor.
Molecules 27(1), 289 (2022)
80. Maisonneuve, L., Lebarbé, T., Cramail, E.G.H.: Structure–properties relationship of fatty
acid-based thermoplastics as synthetic polymer mimics (2013)
81. Talló, K., Bosch, M., Pons, R., Cocera, M., López, O.: J. Mater. Chem. B 8(1), 61–167 (2020)
82. Nagtode, V.S., Cardoza, C., Yasin, H.K.A., Mali, S.N., Tambe, S.M., Roy, P., Singh, K., Goel,
A., Amin, P.D., Thorat, B.R., Cruz, J.N., Pratap, A.P.: ACS Omega 8(13), 11674–99 (2023)
83. Hill, K., Rhode, O.: Lipid/Fett 101(1), 25–33 (1999)
84. Allen, D.K., Tao, B.Y.: Carbohydrate-alkyl ester derivatives as biosurfactants. J. Surfact.
Deterg 2(3), 383–390 (1999)
38 A. H. Jasni et al.
85. Alberts, B., Johnson, A., Lewis, J., et al.: Molecular Biology of the Cell, 4th edn. Garland
Science, New York (2002)
86. Aleandri, S., Rahnfeld, L., Chatzikleanthous, D., Bergadano, A., Bühr, C., Detotto, C.,
Fuochi, S., Weber-Wilk, K., Schürch, S., van Hoogevest, P., Luciani, P.: Eur. J. Pharmaceut.
Biopharmaceut. 181, 300–309 (2022). ISSN 0939-6411
87. Anada, R., Hara, E.S., Nagaoka, N., Okada, M., Kamioka, H., Matsumoto, T.: J. Mater. Chem.
B (2022)
88. Mao, Y., Guidoin, R., li, Y., Brochu, G., Zhang, Z., Wang, L.: Mater. Des. 205, 109737 (2021)
89. Wax. Chemistry. [Online] LibreText (2022). https://chem.libretexts.org/Bookshelves/Biolog
ical_Chemistry/Supplemental_Modules_(Biological_Chemistry)/Lipids/Non-glyceride_Lip
ids/Wax#:~:text=A%20wax%20is%20a%20simple,coatings%20on%20leaves%20and%20s
tems
90. Madhuranthakam, C.M.R., Fernandes, S.Q., Piozzi, A., Francolini, I.: Int. J. Mol. Sci. 23(16),
9501 (2022)
91. Zhang, Q.W., Lin, L.G., Ye, W.C.: Chin. Med. 13, 20 (2018)
92. Matinong, A.M.E., Chisti, Y., Pickering, K.L., Haverkamp, R.G.: Biology (Basel) 11(6), 905
(2022)
93. Ranganagowda, R.P.G., Kamath, S.S., Bennehalli, B.: Mat. Sci. Res. India 16(1) (2019)
94. Sepmag. Purification Techniques. Purification Techniques. [Online] (2023). https://www.sep
mag.eu/blog/purification-techniques
95. Jiang, Y.H., Lou, Y.Y., Li, T.H., Liu, B.Z., Chen, K., Zhang, D., Li, T.: Am. J. Transl. Res.
14(2), 1146–1159 (2022)
96. Quantifiers and Quantification. Stanford Encyclopedia of Philosophy. [Online] Stanford
Education (2022). https://plato.stanford.edu/entries/quantification/#:~:text=Quantifier%20e
xpressions%20are%20marks%20of,most%20common%20examples%20of%20quantifica
tion
97. Lewis, M., Bromley, K., Sutton, C.J., McCray, G., Myers, H.L., Lancaster, G.A.: Pilot Feasibil.
Stud. 7, 1 (2021)
98. Troy, E., Tilbury, M.A., Power, A.M., Wall, J.G.: Polymers (Basel) 13(19), 3321 (2021)
99. Carmona, P., et al.: Gels 7, 186 (2021)
100. Taaca, K.L.M., Prieto, E.I., Vasquez, M.R.: Polymers 14 13, 2560 (2022)
101. Aranaz, I., Alcántara, A.R., Civera, M.C., Arias, C., Elorza, B., Heras Caballero, A., Acosta,
N.: Polymers (Basel) 13(19), 3256 (2021)
102. Purohit, P., Bhatt, A., Mittal, R.K., Abdellattif, M.H., Farghaly, T.A.: Front. Bioeng.
Biotechnol. 10, 1044927 (2023)
103. Sahin Kehribar, E., Isilak, M.E., Bozkurt, E.U., Adamcik, J., Mezzenga, R., Seker, U.O.S.:
Biomater. Sci. 9(10), 3650–3661 (2021)
104. Bose, S., Robertson, S.F., Bandyopadhyay, A.: Acta Biomater. 66, 6–22 (2018)
105. Jasni, A.H.: Fabrication of nanostructures by physical techniques. [book auth.] Preetha Balakr-
ishnan Sabu Thomas. In: Micro and Nano Technologies Nanoscale Processing. Elsevier
(2021)
106. Gopi, S., Balakrishnan, P.: Liposomal nanostructures: Properties and applications. [book auth.]
Preetha Balakrishnan Sabu Thomas. In: Micro and Nano Technologies Nanoscale Processing.
Elsevier (2021)
107. Ilangovan, R., Subha, V., Earnest Ravindran, R.S., Kirubanandan, S., Renganathan, S.: Nano-
materials: Synthesis, physicochemical characterization, and biopharmaceutical applications.
[book auth.] Preetha Balakrishnan Sabu Thomas. In: Micro and Nano Technologies. Elsevier
(2021)
108. Gerwig, G.J., Poele, E.M., Dijkhuizen, L., Kamerling, J.P.: Stevia Glycosides: Chem-
ical and Enzymatic Modifications of Their Carbohydrate Moieties to Improve the Sweet-
Tasting Quality,. [book auth.] David C. Baker. Advances in Carbohydrate Chemistry and
Biochemistry. Academic Press (2016)
109. Giri, B. : Simultaneous Determination of Protein and Glucose in Urine Sample Using a
Paper-Based Bioanalytical Device. Laboratory Methods in Microfluidics. Elsevier (2017)
Naturally Derived Biomaterials: Advances and Opportunities 39
110. Perluigi, M., Marco, F., Foppoli, C., Coccia, R., Blarzino, C., Marcante, M., Cini, C.:
Biochemical and biophysical research communications 3 (2003)
111. Brouns, J.E.P., Dankers, P.Y.W.: Biomacromolecules 22(1), 4–23 (2021)
112. Cell Dissociation Methods for Disaggregation of Tissue: Mechanical vs Enzymatic vs
Chemical. Akadeum Life Science. [Online] Mteric Marketing, March 2021. https://www.
akadeum.com/blog/cell-dissociation/#:~:text=Enzymatic%20dissociation%20uses%20spec
ific%20proteins,combination%20leads%20to%20optimal%20results
113. Duarte, L., Matte, C.R., Bizarro, C.V. et al.: World J. Microbiol. Biotechnol. 36, 11 (2020)
114. Chandra, P., Enespa, S.R., Arora, P.K.: Microb. Cell Fact. 19(1), 169 (2020)
115. Bacakova, M., Pajorova, J., Sopuch, T., Bacakova, L.: Materials (Basel) 11(11), 2314 (2018)
116. Widiyanti, P., Priskawati, Y.C.A.: Int. J. Biomater. (2023)
117. Qi, P., Ning, Z., Zhang, X.: IET Nanobiotechnol. 1–8 (2022)
118. Kang, W., Shi, Y., Yang, Z., Yin, X., Zhao, Y., Weng, L., Teng, Z.: RSC Adv. 13, 5609–5618
(2023)
119. Shalaby, M., Ghareeb, A.Z., Khedr, S.M., Mostafa, H.M., Saeed, H., Hamouda, D.
120. Rotman, S.G., Post, V., Foster, A.L., Lavigne, R., Wagemans, J., Trampuz, A., Gonzalez
Moreno, M., Metsemakers, W.-J., Grijpma, D.W., Richards, R.G., Eglin, D., Moriarty, T.F.:
J. Drug Deliv. Sci. Technol. 79, 103991 (2023). ISSN 1773-2247
121. Bennardo, F., Gallelli, L., Palleria, C., Colosimo, M., Fortunato, L., De Sarro, G., Giudice,
A.: BMC Oral Health 23(1), 134 (2023)
122. Naznin, A., Dhar, P.K., Dutta, S.K., Chakrabarty, S., Karmakar, U.K., Kundu, P., Hossain,
M.S., Barai, H.R., Haque, M.R.: Pharmaceutics 15(3), 732 (2023)
123. Liu, L., Wang, J., Li, Y., Liu, B., Zhang, W., An, W., Wang, Q., Xu, B., Zhao, L., Ma, C.:
Regenerat. Biomater. 9, rbac054 (2022)
124. Almeida, D., Sartoretto, S.C., Calasans-Maia, J.D.A., Ghiraldini, B., Bezerra, F.J.B.,
Granjeiro, J.M., et al.: PLoS ONE 18(2), e0282067 (2023)
125. Salsabila, A., et al.: Metals 13, 494 (2023)
126. Centre, Bristol Biomedical Research. First in human study to assess knee carti-
lage repair implant launches at Southmead Hospital. NIHR. [Online] July 13,
2022. https://www.bristolbrc.nihr.ac.uk/news/first-in-human-study-to-assess-knee-cartilage-
repair-implant-launches-at-southmead-hospital/.
127. Romasco, T., et al.: Biomedicines 11, 786 (2023)
128. Biswas, A., et al.: Polymers 15, 1425 (2023)
129. Zhatkanbayev, Y., Zhatkanbayeva, Z., Iskakova, Z., Kolpek, A., Serikov, A., Moldagulova,
N., Danlybayeva, G., Sarsenova, A.: Int. J. Biomater. Hindawi (2023)
130. Castim, D.: Vegan Biomaterials Could Replace Chemicals In Crop Management. Vegan
Biomaterials Could Replace Chemicals in Crop Management (2022)
131. Carlson, C.: Modern Synthesis uses bacteria to create biomaterial fabric. Dezeen. [Online]
(2023). https://www.dezeen.com/2023/04/03/modern-synthesis-bacteria-biomaterial-fabric/
132. Finney, A.: Stella McCartney releases jumpsuit made with iridescent BioSequins. Dezeen.
[Online] (2023). https://www.dezeen.com/2019/08/02/bio-iridescent-sequin-elissa-brunato-
sustainable-fashion/
133. Future proof your product and elevate your brand - with woodbased renewable Glycols. UPM
Biochemicals. [Online] (2023). https://www.upmbiochemicals.com/glycols/
134. Mueller, J.: CJ Biomaterials Develops Cosmetic Case Featuring Bio-sourced Materials for CJ
Olive Young. Global Cosmetic Industry (2022). https://www.gcimagazine.com/packaging/
containers/news/22618509/cj-bio-cj-biomaterials-inc-develops-cosmetic-case-featuring-bio
sourced-materials
135. Kim, H.-M., Park, J.H., Choi, Y.J., Ohb, J.-M., Park, J.: RSC Adv. 8 (2023)
136. Bagshaw, E.: Top 23 biomaterial designers to watch in 2023. Mater. Source (2022)
137. Markos: Oyster shell composite. Material exploration for moulding. Instagram, Italy (2020)
138. Troy, E., et al.: Polymers 13 (2021)
139. O’Brien, F.J.: Mater. Today 14(3), 88–95I (2011). SSN 1369-7021
40 A. H. Jasni et al.
140. Yusoff, N.H.M., Chong, C.H., Wan, Y.K., Cheah, K.H., Wong, V.-L.: J. Water Process Eng.
51, 103410 (2023). ISSN 2214-7144
141. Mariani, E., Lisignoli, G., Borzì, R.M., Pulsatelli, L.: Int. J. Mol. Sci. 20(3), 636 (2019)
142. Genetic Engineering. National Human Genome Research Institute. [Online] USA (2023).
https://www.genome.gov/genetics-glossary/Genetic-Engineering
143. Joyce, S., Mazza, A.-M., Kendall, S.: Rapporteurs. Synthetic Biology: Science and Tech-
nology for the New Millennium. Positioning Synthetic Biology to Meet the Challenges of the
21st Century: Summary Report of a Six Academies Symposium Series. National Academic
Press (2013)
144. Majumder, K.: Biomaterials. Times of India (2021)
145. National Academies of Sciences, Engineering, and Medicine. Evaluation of the Army
Research Laboratory: Interim Report. The National Academies Press, Washington (2014)
146. Pereira, C.S., Thompson, J.A., Xavier, K.B.: FEMS Microbiol. Rev. 37(2), 156–81 (2013)
147. What Does The Future Have In Store For Photonics?. Stensborg. [Online] Stensborg (2022).
https://www.stensborg.com/post/what-does-the-future-have-in-store-for-photonics#:~:text=
The%20future%20of%20photonics&text=Right%20now%2C%20research%20is%20ongo
ing,optical%20internet%20to%20quantum%20communications
148. Choi, S.: Batteries 9(2), 119 (2023)
149. Ng, S., Kurisawa, M.: Acta Biomaterialia 108–129 (2021). ISSN 1742-7061
Abstract Biomaterials are garnering huge success in the modern era of unforeseen
disease conditions. They find their utility in specifically interacting with biolog-
ical systems for therapeutic or diagnostic purposes. They help patients recuperate
from disease or injury by restoring tissue function thereby, sometimes also leading
to regeneration. Some of the biomaterials are living (microorganisms based) and
can be prepared through microencapsulation, 3D printing, coating, and spinning by
which they can be inculcated into matrices. On the other hand, microbes can even
produce their own matrix which can be genetically engineered to have control over
the responses of the body. Genetic engineering and biotechnological approaches are
an ideal amalgamation of systems for preparing biomaterials that handle accuracy
and complexity quite conveniently. The biomaterials generated by the use of geneti-
cally engineered techniques have proven to be a boon for the regenerative medicines
market, gene delivery systems, tissue regeneration platforms, and controlled drug
delivery systems. In the current scenario, researchers have focused their attention
on molecular evolution for the preparation of advanced designs of genetically engi-
neered biomaterials and the development of inert biomaterials that can be properly
integrated into devices. The advancements and research carried out in genetic engi-
neering continue to assist researchers to gain insights into a specific change in the
structure of employed molecules for the preparation of biomaterials and the way it
will affect the development of biomaterials and their function in the longer run. The
manufacturing sector of biomaterials is a domain that has significantly evolved and
is still being researched.
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 43
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_2
44 A. Chhina et al.
Abbreviations
1 Introduction
The main goal of medical research aims to convert multifaceted engineering science
along with the biological science into procedures for the substitution, rejuvenate, cell
repair, tissue repair, or organs in order to restore compromised function. Biomaterials
are any substances, whether organic or inorganic, living or dead, typically composed
of several parts that engage with the body system. In medical uses, such intelligent
materials are frequently used to supplement or take the place of a natural function.
To state it in a more simplified manner, a biomaterial is a substance that has been
developed to take on a shape that, either on its own or as part of a complex system,
is utilized to regulate interactions with living systems and has the potential aid in the
growth of any curative or clinical method. Smart biomaterials are used in medicinal
uses to brace, improve, and restore a biological functionality or tissue that’s damaged.
They can be natural or synthetic. Contrary to what the term “biomaterial” might
imply, a biomaterial need not be biological or composed of materials linked to life.
The actual substance may be made of metal, plastic, or different types of composites,
but it may also be bioinspired and derived from natural sources. They are intended
to interact with the body or live in a biological environment. Biomaterial sciences
deal with investigations regarding biomaterials. They have expanded steadily and
significantly during the course of their existence as a result of various enterprises
making considerable financial expenditures in the development of new stuff. Aspects
of tissue engineering, biology, chemistry, and materials science are all included in
the discipline of biomaterials science.
Due to the incorporation of genetic engineering, cell-based biomaterials have
attracted attention from all over the globe for their potential use in a variety of
medical disciplines. These engineered biomaterials can change shape or mechanical
characteristics, release bioactive ions, and actively discharge chemicals as a result of
physiological triggers. All of these material-induced activities may result in native
cells and influence the immune system These designed biomaterials can be utilized
in the deposition of extracellular matrix (ECM) to aid in the repair of tissues, and
enhance cell adhesion and proliferation by incorporating biophysical and biochem-
ical signals [1]. Evaluation of the short- and long-lasting cellular and when creating
the next generation of bio-responsive materials, understanding how materials interact
at the molecular level is essential as material capabilities increase [2].
Medications, genetic materials, and proteins are all delivered therapeutically
through the use of injectable biomaterials. They provide the opportunity to address
a range of conditions by delivering medications precisely and preventing immune
system uptake. Injectable biomaterials with both synthetic and organically derived
components are being investigated for use in managing heart attacks, cancer, and
bone defects. Widespread chronic illnesses like type 1 diabetes, an autoimmune
disease in which the body’s defenses attack pancreatic cells that produce insulin,
may be combated with the aid of immunomodulating biomaterials. An injectable
synthetic biomaterial recently created by researchers reversed type 1 diabetes in
non-obese diabetic mice. This is a significant move toward creating a biodegradable
46 A. Chhina et al.
platform to help manage the disease’s effects. Applications for genetically engi-
neered materials include the delivery of drug, nanoparticle coatings, carriers that are
macromolecular, and hydrogels. Engineering of the tissue has a significant influence
on novel cutting-edge approaches to gene engineering. Studies supported various
approaches to obtaining, creating, and using these materials while highlighting the
evolved functions and properties.
Biomaterials are more frequently integrated into devices than they are used as
simple materials in medical uses. Supramolecular biomaterials, which are collec-
tions of molecules that go beyond the capabilities of individual molecules, have the
capacity to detect and react, making them perfect materials for treating disease or
injury. Supramolecular biomaterials that can respond to physiological signals by
activating or deactivating or that mimic biological signaling are being investigated
by researchers.
The method of creating biomaterials has changed over time. Many of the bioma-
terials that are used in medicine today were not intended to be used in medicine;
instead, they were discovered by clinicians to help treat patients. Thus, cellulose
acetate, a common plastic, was used to create the first dialysis conduit. Initial
vascular grafts employed polymers like Dacron, which were taken from fabrics.
Initially, polyurethanes of industrial quality were used as the basis for the mate-
rials for artificial hearts. These resources made it possible to handle critical medical
issues. However, they also brought about challenges. Platelets and the complement
system may be activated by dialysis tubing; vascular grafts made of dacron that are at
least 6 mm thick can be used; otherwise, biological processes at the blood-material
and tissue-material interfaces could result in occlusion; additionally, blood-material
interactions could cause clot development in an artificial heart, which would raise
the risk of stroke and other complications [3]. Recent studies are based on drug and
cell transporters made of a variety of biomaterials. In tissue engineering, it may be
necessary to combine medications, cells, and an appropriate carrier with a tailored
degradation profile, specialized macroscopic features, and specific biological cues to
promote healthy tissue growth [4]. Biological activity can be added to biomaterials
by incorporating oligopeptides that promote binding.
Traditional biomaterials like polytetrafluoroethylene, silicone rubber, or polyethy-
lene are implicitly recognized by cells in vivo. Non-specific protein adsorption
from bodily secretions occurs on the surface of surfaces, and adsorbed homolo-
gous adhesive proteins encourage adherence of the cell by binding to cell surface
receptors. The explicit power of adhesion of cells on intelligent materials is possible
by preventing nonspecific protein adhesion on the material surface and utilizing
adhesion-promoting peptides that are unique to particular cell types [5]. Short primary
segments of these peptides are derived from the domains that are receptor bound with
the adhesion proteins. The tri-peptide sequence RGD is the adhesion peptide that is
Different Techniques of Genetic Engineering Used for the Development … 47
Genetically engineered biomaterials have been a topic of research ever since recom-
binant DNA technology has come into play. Such biomaterials can be of animal or
plant origin with properties of strength, firmness, and ability to adapt to the environ-
ment. Where genetically engineered techniques aid in developing new, relevant, and
functional biomaterials, there they also help in developing a better version of already
existing biomaterials along with enhancing chemical and physical qualities leading
to an increase in the applicability of biomaterials which have a significant influ-
ence on biomedical applications such vaccine production, engineering of tissue and
regeneration, drug administration, and zoological interpretations, etc. In the creation
of hydrogels, development of films, formulation of matrices, development of phar-
maceuticals, as well as several physiological and anatomical implants, the use of
Different Techniques of Genetic Engineering Used for the Development … 49
genetically altered proteins can provide good quality materials with excellent multi-
functional properties, broadening the range of applications and need for developing
better genetically engineered biomaterials [13]. Moreover, electrical functions can
also be inculcated into the biomaterials through the use of technology which helps
in excellent antibacterial activities which could be in vitro or in vivo.
Multi-stage moderation of smart biomaterials, which includes gene alteration
(e.g., modifying gene sequence, molecular weight, and chain size), chemical conju-
gation (e.g., adding light-sensitive molecules, oil-loving and/or water-loving side
chains), and mesoscopic blending (e.g., doping with metallic nanoparticles, GR, and
CNT), allows for the integration of biochemical and electrical capabilities into the
material system based on biopolymers [14]. To circumvent the limitations of viral
vectors for NK cell engineering, attempts have been made to substitute nanoparticles
for viral vectors as they did not show any efficacy due to a transduction efficiency of
less than 20%. Due to the distinct physicochemical features of genetic modification,
nanoparticles can add imaging modality to the vector while avoiding many of the
possible safety problems associated with viral vectors [15].
Living-engineered biomaterials that have microorganisms involved have the
capacity to respond in complicated ways to environmental cues, and they can be
genetically modified to enable user control of behavior and the incorporation of
a variety of inputs. The altered microbes can either produce their matrix, as in
biofilms, or they can be integrated into matrices utilizing numerous innovations,
including coating, 3D printing, spinning, and microencapsulation [16]. In addition,
the use of biomaterials in clinical environments may be hampered by a few drawbacks
that have been observed. For instance, a significant hurdle impeding effective tissue
repair and regeneration is biomaterial-mediated inflammation; as a result, biocompat-
ibility is being continuously researched and enhanced which can be fulfilled through
the process of genetic engineering [17]. Various sorts of immunomodulatory genes
were introduced for alteration to stop biomaterials from being immunologically
rejected. Transmembrane proteins, cytokines, and genes genes-producing all have
immunomodulatory effects and other proteins have been proven in several investiga-
tions and these genes acted in a variety of ways to exert influence that is suppressive
or agitative on the immune system elements [18].
Live cells can now be incorporated into structures and allowed to flourish owing to
developments in 3D bioprinting. Also, chimeric biomaterials can be 3D printed more
accurately and intricately. The conventional biomaterials used as non-conventional
biomaterials can be produced with more precision and finer qualities and hence
can also help in saving cost [19]. Consequently, there are many uses for intelligent
biomaterials, ranging from health (such as engineering of the tissues, and delivery
of the drugs, and biosensors) to more newly researched environmental uses like
ecosystem restoration (e.g., coral reefs and environmental remediation) [20]. The
development of physicochemical features of protein-based drug delivery devices
that are accurately calibrated is made possible by genetic engineering approaches,
offering a higher degree of customization than is possible with synthetic polymers.
Elastin-like proteins (ELP), silk-like proteins (SLP), and silk-elastin-like proteins
(SELP) due to their inbuilt physical and chemical attributes and the simplicity of
50 A. Chhina et al.
4.1 Scaffolding
Both organic and synthetic nanoengineered biomaterials that are polymeric are
employed in biomedical utilization like targeted and smart drug delivery with
prolonged and controlled release. Natural polymeric nanocomposites are frequently
employed in medicine because they are biodegradable, nearly biocompatible, and not
poisonous. Applications for synthetic biopolymer nanocomposites are numerous. By
combining with biomolecules, they can be altered to become more biocompatible
and harmless to the body [24].
Sequencing in the genetic engineering of biomaterials is one of the significant tech-
niques as the properties of biomaterials depend on the sequences that are employed
in the biomaterials which is an emerging topic for research recently. Examples
of synthetic methods for sequence, control include iterative methods where each
monomer unit is added sequentially, regulated polymerization techniques using both
chain and step-growth mechanisms, and bioinspired and supported methodologies
like molecular imprinting and templated syntheses. In the case of genetic engi-
neering of biomaterials. Biomaterials made of particular peptidomimetics and statis-
tical copolymers have both been made using the comparatively simple process of
sequence selection known as “bioabstraction,” or replicating minimum functional
DNA or protein sequences. Synthetic analogs have been produced using this tech-
nique which are biologically important compounds, including host defense peptides
(HDPs), cell-penetrating peptides (CPPs), and antimicrobial peptides (AMPs) [25].
layout, and length of the protein biomaterial’s chain. This quickly developing tech-
nology has given rise to effective tools for creating unique smart biomaterials with
preset 3D structures [26]. However, several issues need to be resolved to synthesize
protein chains using genetic engineering techniques: Repetitive DNA sequences may
be altered and deleted in various ways, sequences of messenger RNA (mRNA) neces-
sary for a certain codon for a particular amino acid residue. The host cell may break
down the target protein before sufficient levels are synthesized, reducing the yield.
Biomaterials that have been genetically modified have many benefits. By carefully
regulating the length, stereochemistry, and 3D structure of the polymer chain, it is
possible to create materials with specific, distinctive characteristics.
4.4 Decellularization
Recently, novel methods based on genetic changes have been developed, including
decellularized biomaterials. Decellularization is the elimination of cells and
constituents involving the nuclear ones, of the tissue, using a variety of chemi-
cals while conserving the ultrastructure of the collagen, elastin, microfibrils, proteo-
glycans, glycosaminoglycans (GAGs), and other growth factor components found
in the extracellular matrix [27]. Decellularization seeks to reduce the immuno-
genicity, calcification-inducing potential, and cytotoxicity of native extracellular
matrix (ECM) by removing the majority of cells from it with the aid of physical,
chemical, and biological methods [28]. Extracellular matrix (ECM)-based bioma-
terials now have new prospects in the transfusion of organs and tissue regrowth in
preclinical and clinical settings because of advances in decellularization techniques.
The resultant dECM scaffold should ideally maintain its functional content and three-
dimensional (3D) structure for tissue restoration purposes. Before dECM scaffold
transplantation, it was important to recellularize the dECM with a particular cell
type to increase cell survival and reduce immunological rejection [29]. Steps in the
decellularization are given in Fig. 3.
Decullarization occurs either through physical methods such as freeze-thaw,
hydrostatic pressure, ultrahydrostatic pressure, and oscillation, chemical methods
like hypertonic/hypotonic solutions, detergents namely sodium dodecyl sulfate
(SDS), alcohols or triton X-100 and enzymatic methods such as RNases, DNases,
and trypsin which aids in separating a tissue’s extracellular matrix (ECM) from the
cells that reside there, leaving behind an ECM scaffold that can be employed in arti-
ficial organ and tissue regeneration [29]. Crosslinking after decellularization occurs
through a number of crosslinking agents which can be physical, chemical, or natural.
Thermal dehydrogenation and photo-oxidation crosslinking are the two basic types of
physical crosslinking. Thermal dehydrogenation was frequently utilized in the begin-
ning and now, this approach is frequently employed as a backup strategy because
of the difficult crosslinking conditions. The crosslinking conditions are difficult to
manage using the photo-oxidation approach. This makes it more frequently employed
in the decellularization of tumor tissue, which is typically done for disease modeling
Different Techniques of Genetic Engineering Used for the Development … 53
purposes. The dECM of the tumor was crosslinked by Lü et al. using photo-oxidation
mediated by methylene blue [14]. This was the first study to show whether photo-
oxidative crosslinking affected the tumor dECM scaffold’s structural, physical, and
biological characteristics. Epoxy compounds, carbodiimide (CDI), glutaraldehyde,
and hexamethylene diamine carbamate (HMDC) are examples of crosslinkers which
are employed in chemical crosslinking but have huge side effects due to which better
crosslinking agents need to be used [29].
Sterilization being the third step is the most crucial step when decellularization is
involved as the applicability of different biomaterials is put off due to limited steril-
ization methods for a period that is more than expected. The structure of the ECM
will inevitably change as a result of current sterilization techniques, such as exposing
the material to radiations such as ethylene oxide, gamma, or electron beam, and this
could negatively impact the biomaterial’s mechanical properties [29]. With respect
to the list provided above, a more suitable option is provided by the development
of supercritical carbon dioxide. [30]. The impact of three widely used sterilizing
techniques, including ethylene oxide (EtO), peracetic acid (PAA), and gamma irra-
diation (GI), on material qualities, was examined by Matuska et al. In this procedure,
they studied the early cellular contacts and discovered that, unlike GI and EtO, PAA
showed advantages in cell adhesion but did not significantly alter the structure of the
cell. According to this, the choice of sterilizing technique is important for modifying
the properties of biomaterials in a way that promotes cellular adherence and is highly
relevant to the use of biomaterials in vivo [31]. There is currently no sterilization
method that can completely stop the incidence of undesirable consequences [32].
54 A. Chhina et al.
Consequently, selecting the best sterilizing process is a crucial step for using decel-
lularized scaffolds in the future for therapeutic applications, and more research is
still required.
Future uses of the decellularized matrix-based materials will depend on the final
step of preservation in the process of decellularization as well. The manufactured
decellularized scaffolds were now typically stored in PBS alongside antibiotics and
antimycotics at a temperature of 4°C for a brief length of time, on standby, or at
20 °C for long-term preservation [29, 33]. The degradation of active substances and
structural damage, however, seems to be unavoidable at the moment and require
urgently more work. The ideal characteristics of a tissue for repair in wound repair
include strong bioactivity, adequate mechanical characteristics, excellent degrada-
tion, and outstanding biocompatibility are all desirable characteristics in a scaffold
for tissue repair. Decellularized ECM (dECM) is a three-dimensional natural scaf-
fold produced from autologous, allogeneic, and xenogenic tissues that retains its
intrinsic tissue structure despite having had the cellular components removed. The
dECM is promising in translational medicine [34] because of exceptional physio-
logical activity, high biocompatibility, lack of immunogenicity, and use as a diverse
source of basic materials [35].
New advancements in the biomaterial’s domain in the realms of regenerative
medicine and engineering of tissue, dECM-based smart biomaterials have had great
success. Also, scientists were inspired to create a novel standard for the application
of dECM biomaterials, such as constructing a safe and biocompatible method for
delivering therapeutic agents like drugs and cells. For instance, dECM can be ground
into nano- or microparticle size and used as a therapeutic delivery equipment to repair
anastomoses [29]. Clinically, both Axogen’s Avance nerve transplant and AlloDerm,
an acellular skin matrix from Biohorizon is a regenerative tissue matrix intended to
hasten skin regeneration. are used to treat wounded nerves and are two examples of
emerging clinical applications of dECM. DECM can also be utilized to make bioink-
related hydrogels. Of the inks now on the market for 3D bioprinting, bioinks based on
dECM exhibit the highest level of biomimetic capability. The building block for 3D
printing a functioning human organ is made of dECM bioinks in conjunction with the
recent development of bioprinters. It is fascinating that multiple studies have been
published employing ECM hydrogels synthesized from almost every organ where
dECM bioinks are employed for bioprinting. ECM hydrogels could be employed in
minimally invasive deliveries as an easily administered pre-gel viscous solution into
a patient utilizing a catheter or syringe [36].
antimicrobials and growth factors are two examples of biologically active chemi-
cals that can improve cellular and tissue reactions [27]. When introducing bioactive
compounds, such as growth hormones or extracellular matrix proteins, through dip
coating, to the surface of scaffolds physical adsorption, a straightforward immobi-
lization technique, is commonly used. Topography of surface, functional groups,
pH, wettability, and electrical charge of the substance are just a few examples of the
physical and chemical characteristics that affect adsorption efficiency. Since many
nanomaterials are hydrophobic, techniques are required to increase their wettability
and turn them into hydrophilic materials. Chemical interactions between carbon and
non-carbon atoms are broken down using physical techniques like ion bombard-
ment, UV light, and plasma modification. As a result, oxygen reacts with unsatu-
rated bonds and radicals to promote hydrophilicity and reactivity toward substances
that are biological. Since they contain a large number of reactive chemical groups
(hydroxyl, carboxyl, and amide), natural polymers have the benefit of being less
hydrophobic and able to interact with physiological molecules. Materials composed
of proteins, such as collagen and silk, have been customized by the adsorption of
biologically active compounds, such as vascular endothelial growth factor (VEGF)
[37], basic fibroblast growth factor (bFGF) [38], and bone morphogenetic proteins
(BMPs) [39], and pharmaceuticals like antibiotics, and heparin [40].
Van der Waals, hydrogen, and hydrophobic interactions serve a purpose in adsorp-
tion while they are electrostatic in nature, external factors can affect how stable the
adsorbed molecules’ bond is relatively weak or moderate. In this manner, uncon-
trolled release of the immobilized species can be brought on by changes in the pH,
strength of ions, and the number of adsorbed species in the surrounding media. For
instance, large doses of bone morphogenetic proteins (BMPs) in order to trigger the
correct osteogenic response, substances that diffuse away from the fracture location
are required. The BMP-2 release profile from collagen sponges exhibits an early
spike within the first ten minutes, during which 30% of the BMP-2 is lost by the
messenger carrier, and is thereafter sluggish over the course of the next 3–5 days.
Clinical conditions like soft tissue hematomas, ectopic bone growth, and receding
bones may result from this original burst release [41].
between the mechanical and electrical states linearly while responding to mechan-
ical stress and are made of crystalline materials lacking inversion symmetry [43].
Pyroelectric nanogenerators (PyENGs), which transform variations in heat energy
into electrical energy, come in third [44]. Recently, it was shown that TENGs and
PyENGs can work together by harnessing the human body’s mechanical and thermal
energy to generate electrical signals [45]. Nonetheless, triboelectric nanogenerators
are frequently chosen in comparison to their counterparts piezoelectric nanogenera-
tors or pyroelectric nanogenerators due to their conditions of operation for regions of
low frequency/amplitude, ecosystem quality, more extensive resources, and material
selection [46].
Polymers, such as polyvinylidene fluoride, polytetrafluoroethylene, poly-
dimethylsiloxane, and others, make up the majority of triboelectric nanogenerators
because they all include the element fluorine, which has a great affinity for elec-
trons. Although these polymers can produce significant triboelectricity, they can be
challenging to handle and decompose, and their accumulation and disposal could be
hazardous to both human health and the environment. The ecologically friendly and
biodegradable TENGs (bio-TENGs) will create smart portable tools to address these
issues, setting a new standard for the establishment of a smart and green society.
TENGs in which the triboelectric layer components are partially or entirely made of
natural substances or their constituents are classified as bio-TENGs. Firstly, bioma-
terials having high biocompatibility and biodegradability are obtained from natural
species and they can be decomposed by microorganisms after being disposed of
in nature. Additionally, because TENGs have an innate ability to generate green
energy from their environment and biologic processes, these intelligent bio-TENGs
can be used to power autonomous driving sensors or smart electronics. Finally, there
are numerous sources of biological materials, ranging from marine products like
shellfish and seaweed to agricultural by-products like straw, rice husks, bark, and
bagasse [47]. The majority of biodegradable polymers have low toxicity and strong
biocompatibility, which makes BD-TENGs effective in biomedical applications. The
implantations for medical devices should be removed promptly when their purpose
has been served in the human body. Biodegradable devices are typically employed
to treat disorders of the heart and there is no requirement of additional surgery,
in contrast to non-degradable devices that can only be removed through procedures,
which causes subsequent injury to the patients [48]. They are also employed in tissue
regeneration.
For the current triboelectric series to be expanded, new triboelectric materials
must be developed. The design of positive and negative materials for placement
in the triboelectric series is under the limelight. The synthesis of novel materials
containing significantly more accurate halogenic elements is the main focus of tech-
niques for creating novel tribo-negative materials. As an illustration, Lee et al. stated
that fluorinated polymeric sulfur outperformed commercial PTFE, which is ranked
the worst in the series of triboelectric materials. A disadvantage associated with tribo-
negative materials is their insulation. The creation of materials that are tribo-positive
is inspired by the emergence of the highest positive charges in polymers comprising
nitrogen and oxygen with pyridine amide, amine, or hydroxyl groups. Based on this
Different Techniques of Genetic Engineering Used for the Development … 57
hypothesis, Zhao et al. prepared an aniline formaldehyde resin (AFR) with sufficient
mechanical strength and controlled microstructures for TENG device applications
[43].
By capturing high entropy energy, TENGs offer a fresh approach to the problem
of supply of energy. Nevertheless, flexible, humidity-resistant, and affordable TENG
must meet strict standards for wearable electronic devices. Here, a straightforward
and environmentally benign process was used to create the multipurpose wheat starch
TENG (S-TENG). The S-TENG opens the door for the mass production of multifunc-
tional biomaterials-based TENG as well as the real-world use of wearable electronics
and self-powered sensors [49].
There is a reported way for easily improving the quality of a chitosan-diatom-
based biomaterial TENG (CD-TENG) by controlling the weight ratio of the diatom
frustule, which can increase the density of charge in the chitosan-diatom composite
film [50]. This is useful to apply in wearable devices that might be easily attached to
human skin and will not be associated with any adverse implications. Nanocarriers
are stimuli-responsive biopolymers that are commonly used in invasive and trans-
dermal drug delivery (TDD) systems that are stimulated and regulated by TENGs
power supply. Other common drug-delivery agents include nanoparticles, micronee-
dles, and liposomes. TENG is therefore utilized to deliver drugs with organelles and
biomaterials for offering a target-oriented controlled drug release [51]. Paper-based
cellulose TENGs, nano cellulose TENGs, and micro/nano composite TENGs are
other advancements in biomaterials based on the triboelectric nanogenerator prin-
ciple. The advantages of cellulose as a naturally occurring polymer are cost effective-
ness, processability, good mechanical flexibility, biocompatibility, and biodegrad-
ability. Additionally, it may display a special combination of chemical, structural,
dielectric, and optical characteristics. These benefits may transform cellulose-based
functional materials into appealing substrates or TENG elements [52].
The creation of a durable, stable encapsulating covering is necessary for the
commercialization of biomaterial TENG devices as clinical implants since it will
shield the device from the hostile in vivo environment. This is because overcoming
the immune system’s response to a foreign body is challenging, and fibroblast growth
may eventually cause the implanted TENGs to respond to motion stimuli less effec-
tively [53]. To construct well-designed TENG sensors, researchers will eventually
need to use fabric, shape-memory polymers made of latex or rubber, hydrogel, and
other cutting-edge functional materials. In addition, significant progress in the area
of flexible electrodes is required to increase the wearability of TENGs both inside
and outside the living systems. Hard and soft materials, including carbonaceous
nanoparticles, polymeric substances, 2D materials, inorganic and ceramic materials,
and polymers, are used to create today’s naturally flexible electrodes [54]. The TENG-
based healthcare system benefits greatly from TENG-based biomaterials, which have
a bright future in terms of consistency, sustainability, comfort, and adaptability.
58 A. Chhina et al.
Organic matrix macromolecules are essential for improving the mechanical char-
acteristics of biomineralized compounds like bone and teeth in nature. Because
there is still a lot to learn about how natural matrix components work, creating
artificial matrix counterparts is both exciting and difficult. Biomimetic matrices can
be created utilizing genetically produced elastin-like polypeptides (ELPs) instead
of natural components for the design of mechanically robust ELP-hydroxyapatite
(HAP) composites. ELPs with clearly defined backbone charge distributions can
be made by intermittently including negative, positive, or neutral side chains or
HAP-binding octa glutamic acid patterns at one or both protein termini. ELPs have
sequence-specific properties that affect how they interact with ions, bind HAP, and
spread HAP nanoparticles. With ELPs that bind with HAP, calcium phosphate cement
can be further improved to provide materials with enhanced mechanical strength,
injectability, and anti-washout properties [55].
Elastin-like polypeptides possess some adaptable properties which enable them
to form different nanostructures, including nanoparticles, nanofibers, and nanocom-
posites [56, 57]. When utilized in drug delivery, these nanotechnologies increase
the effectiveness of the therapy because the nanostructure can target the molecule,
increasing its stability and increasing contact surface. This could boost these struc-
tures’ activity, which is essential for maintaining their thermodynamic properties
[58, 59].
4.8 CRISPR-Cas 9
Table 2 Biomaterial vectors along with their Cas9 cargoes: Applications and Advantages [63]
Cas9 Cargo Delivery System Application Advantages
DNA Palmstearin derived Analyzing The PS-Lips cationic lipid
fatty acyl chains nanoparticle molecular nanocarrier system’s
architecture effect on liposomes successfully
Cas9 plasmid delivery carried genome-editing
and characterization of tools containing CRISPR/
delivery to HEK-293 Cas9 encoded pDNA
cells
DNA Turbofect Identification of Nf-1 Effective pain
pain-related management
therapeutic targets
mRNA Extracellular vesicles Targeting in MOLM13 In bothhuman cells and
derived RBCs cells xenograft animal models,
RNA medication delivery
with RBCEVs exhibits
highly strong microRNA
inhibition and
CRISPR-Cas9 genome
editing with no
detectableharm
Protein Nanoclews involving Disruption of EGFP in Enhanced delivery of
yarn-like DNA U2OS-EGFP cells CRISPR-Cas9
in vitro and in vivo
DNA Liposome-Exosome Lipofectamine 2000 Exosome-liposome hybrid
hybrids and nanoparticles are
HEK293FT-derived promising for in vivo gene
exosome performing editing since they can
targeting in transport the
mesenchymal stem CRISPR-Cas9 system to
cells MSCs
mRNA Zwitter-ionic lipids Lung cancer cell Improvement in safety
amino delivery and in vivo and effective usage of
targeting gene editing
Protein-tagged Arginine coated Works by targeting The method assures the
NLS and glutamic cationic gold Hela cells and development of
acid nanoparticles disrupting signals for “weaponized”
(ArgNPs) cancer survival macrophages for cancer
immunotherapy due to the
enhanced attack and
destruction of cancer cells
mRNAwith Cholesterol,DSPC In TTR amydoilosis Helps in reducing
modified sgRNA lipid, PEG-2000 for gene targeting Transthyretin (TTR)
DMG and LP01 lipid serum protein
concurrently with in vivo
genome-editing levels that
are clinically significant
(continued)
60 A. Chhina et al.
Table 2 (continued)
Cas9 Cargo Delivery System Application Advantages
Highly negative Lipofectamine 2000, Delivery which is When compared to DNA
GFP fused protein RNAiMAX in vivo to mice cochlea transfection, delivery of
unaltered complexes
resulted in genomic
modifications of up to
80%
DNA Cationic polypeptide Disrupting GFP and The system provides a
made with surface targeting in HeLa flexible gene-editing
PEGylation tumor-bearing mice platform for biological
research and therapeutic
applications, enabling
multiplex gene knock-out,
gene knock-in, and gene
activation in vitro and in
vivo
associated RNA sequence, is the system that is used for the majority of CRISPR appli-
cations in bacteria. This method causes an incision in the designated area, allowing for
DNA editing. The DNA that recognizes the protospacer is bound by the Cas9-sgRNA
complex, which leads to a slow unraveling of the DNA. The CRISPR-Cas9 tech-
nology has the advantage of enabling researchers to more precisely control how the
CRISPR/Cas systems are activated by targeting transcriptional activators to specific
genomic regions. The latest advances in the interaction of Porphyromonas gingivalis
(P. gingivalis), Enterococcus faecalis (E. faecalis), and Streptococcus mutans (S.
mutans) with bacterial functions and CRISPR/Cas systems are relevant to dentistry
[61].
Delivering the CRISPR-Cas complex to particular cells or tissues is challenging
and laborious because its components must transcend tissue and cell membrane
barriers to enter the nucleus, where they can affect the nuclear genome. The two
broadly used physical techniques for introducing the CRISPR-Cas complex into
cells are electroporation and single-cell microinjection. These techniques are crucial
for editing genes in developing embryos and creating transgenic animals [62].
Due to their adaptability, biocompatibility, and rising transfection effectiveness,
biomaterials are increasingly being used as non-viral vectors. Capacity restrictions
are not an issue, and nanoparticles can be further tailored to enhance nuclear transport
and tissue selectivity. The capacity of biomaterials to be tuned offers considerable
potential for enhancing CRISPR-Cas9 delivery for in vivo gene editing. Since there
are practically no restrictions on how biomaterials can be customized, this gives phys-
ical and viral delivery a particular advantage. This is due to the fact that several organ
systems along with their types of cells will eventually need gene editing methods
that are specially tailored to their microenvironments [63]. By incorporating various
biomaterials into them, Cas9 can be supplied in the form of DNA, plasmids, mRNA,
and proteins.
Different Techniques of Genetic Engineering Used for the Development … 61
4.9 3D Printing
The methods of genetic engineering are utilized to extract and mix particular DNA
from diverse materials to create chimera DNA. When the chimeric DNA is sent back
with the help of plasmids to bacterial cells, it can then be duplicated. The survival of
cells carrying chimeric DNA can be backed by bioprinting techniques like inkjet-,
extrusion-, and laser-based printing. They can design chimeric biomaterials for 3D
printing in a controlled and cost-effective way. Recombinant DNA technology allows
engineers to employ chimeric biomaterials in a number of biological research and
development applications by taking their desirable traits from nonhuman origin. The
creation of fake CAR T cells for use in adoptive cell therapy for the treatment of
cancer is the most well-known application of chimeric biomaterials. Particularly,
Fc-chimeric proteins may be utilized for the identification of desired stem cells
and as test compounds in advanced 3D printers. Chimeric organoids that mimic the
tumor microenvironment can be designed by bioprinting cancer cells and healthy
mammary epithelial cells. Furthermore, chimeric biomaterials can be included in
drug delivery systems to improve the therapeutic potency of previously accessible
proteins and drugs. Recently, attempts have been made to combine chimera biomate-
rials with biopolymers [19]. Despite being incredibly effective in CAR T-cell therapy
as a customized approach to cancer treatment, chimeric biomaterials can potentially
have unfavorable side effects and flaws in patients. Additionally, because of its high
cost and drawn-out approach, CAR T-cell treatment is not commonly available.
The ethical ramifications of research using human organoids and/or the introduction
and integration of chimera material into a host mammal are also complex. Sound
ethical principles must therefore be upheld as chimeric biomaterials are increasingly
used in modern science. It is necessary to harvest DNA from numerous genomes in
order to create chimeric biomaterials, which calls for the employment of additional
3D printing settings that allow high cell survival and density. Across all printing
techniques, there is a definite trade-off between cell survival, resolution, and price.
Chimeric biomaterials are designed using recombinant DNA technology, which
reconstructs genes from many species into desired effects by programming them
at specified gene sequences. Chimeric polymers have a much greater degree of
control over the amino acid sequences than their chemically synthesized equiva-
lents, which is much greater than what is possible with solution or solid-phase peptide
synthesis. Incorporating stimuli-responsive motifs into a chimeric polymer’s back-
bone allows for accurate phase changes in response to changing temperature, pH,
and ionic strength. Additionally, this addition might enhance interactions between
an implant and already-existing cells. Chimeric polymer-based biodegradable struc-
tures can also maintain their temporal and spatial precision so that breakdown only
happens in the presence of specific enzymes. This can be achieved through the addi-
tion of amino acids which are prone to breakdown [64]. Engineers could advance
the creation of treatments that address a variety of unresolved medical issues that
have plagued society since the dawn of humankind by leveraging desirable properties
from other sources.
62 A. Chhina et al.
gene expression of cells that come into contact with the material. Substances in tissue
engineering that may affect epigenetic pathways are titanium, silica, PLGA, bioglass,
and ceramics [69]. Another organic material that has undergone epigenetic function-
alization is silk. James et al. examined an anti-sense-miRNA-214 silk device utilizing
surface coating, which led to a continuous release of miRNA inhibitors up to 7 days
in vitro and increased the expression of osteogenic genes in the human mesenchymal
stem cells implanted on these devices [70]. These findings would suggest that this
unique technique could be helpful for localized bone tissue creation and for promoting
osteogenesis at the implant surface. Collagen sponges and macroporous biphasic
calcium phosphate scaffolds combined with HDAC inhibitors (HDACi) caused the
creation of woven bone and newly produced bone at the scaffold’s interface, which is
another epigenetic mechanism for collagen functionalization that has been explored
[71]. Although Cre recombinases are widely utilized, optogenetics has been found to
be beneficial for a variety of phylogenetic histone deacetylases, methyltransferases,
acetyltransferase inhibitors, and proteins that recruit these enzymes. Included in this
group of proteins are KYP, TgSET8, NUE, PHF19, Sin3a, Sirt3, NcoR, HDAC8,
RPD3, and Sir2a [72].
Genetic code expansion (GCE), a related approach, is comparable to it. Next-
generation protein engineering, in particular GCE, enables heterologous expression
of proteins with NCAA/UNAA inclusion in proteins in both prokaryotic and eukary-
otic organisms [73]. GCE promotes biomedical and tissue engineering research by
developing innovative protein-based biopolymers with broad structural and func-
tional flexibility. Silk, elastin, collagen, and fibrin are used to create a variety of
biomaterials that are created using the GCE process. The ability to enhance the
production of biomaterial proteins by GCE is expected to usher in a new era of tissue
engineering and biotechnology. According to recent advancements in tissue engi-
neering, customized tissue constructs with anisotropic architecture, heterogeneous
cells, and composite nano/biomaterials are being created for regenerating functional
tissue and organotypic tissue models [74]. Because they enable precise control over
the loading of bioactive moieties, microstructure, and handling, biomaterials with
spatiotemporal variations are essential for 3D manufacturing. GCE improves the
possibility of producing biomaterials made of growth factors and congener proteins
that act as brokers for interactions between cells and the matrix. GCE presents a
unique opportunity for the creation of bioinspired proteins and customized biomate-
rials that show potential for use in precision and personalized medicine. GCE creates
protein biomaterials with all-inclusive qualities to ensure enhanced interactions
between the components of the tissue engineering trio [74].
64 A. Chhina et al.
antigens. They can therefore make excellent use of the immune system’s numerous
internal regulating mechanisms. One way to promote tolerance is through inducing
energy or deletion of self-reactive lymphocytes. Additionally, targeting helper T cell
activation toward the regulatory pathway can help prevent the activation of other T
cells’ undesired effector functions.
Biomaterials allow for engineered solutions to complex biological issues, as was
the case with vaccine development, and they hold hope for the future of our ability to
effectively treat autoimmune diseases and aberrant immune activity [81]. Macroscale
scaffolds based on biomaterials can be utilized to disseminate immunomodula-
tory elements like proteins (like cytokines and antibodies), oligonucleotides (such
silencing RNA and plasmid DNA), and scavengers that eliminate or reroute harmful
substances (like ROS) locally. Future biomaterials-based cell carriers and niches
may be created to monitor certain immune responses for diagnostic, investigation
and observation, and clinical study purposes together with their use for therapeutic
goals. For instance, the delivery of a pancreatic-islet lysate to gather, separate, and
analyze the repertoire of T cells that are diabetogenic has been investigated using
a PLGA-based microporous scaffold. Similar to this, a locally applied adjuvant-
and antigen-loaded alginate microneedle patches can attract T cells indicating active
systemic responses. [82].
By combining a suitable biomaterial (which is an essential part of the extracel-
lular matrix involving the airway system) using airway organoids harboring disease-
relevant cell types, it is possible to construct a structure like the tissues, with favor-
able functional and morphological attributes. Lung-on-a-chip systems also need to be
enhanced and optimized in order to replicate and imitate the critical pulmonary tissue
microenvironment conditions. This could aid in the discovery of innovative medica-
tions and therapeutics for challenging lung diseases like the newer SARS-CoV-2. A
promising method for determining potential side effects of recently created SARS-
CoV-2 medications and vaccinations is to combine lung-on-a-chip technology with
heart, liver, and kidney organs-on-a-chip systems [83]. An acellular dermal matrix
created by the usage of skin from the human deceased when mixed fat grafts in skin
graft and breast reconstruction surgeries, to enhance the localization and survival of
transplanted fat cells procedures to improve the localization and survival of trans-
planted fat cells. In the commercially available product Epigraft, collagen has been
used to enable the organization of composite cell types within tissue grafts, such
as skin grafts. Lately, cardiomyocytes derived from stem cells and collagen sheet-
laden epicardium cells have been used in combination for myocardial tissue healing
following infarction [84].
Hence it is believed that biomaterials will eventually be used for more sophisti-
cated purposes, such as signal transducers that trigger cellular roles. Early viability
studies in animals suggest that remote control of cell activity is possible using external
actuators including ultrasonic, magnetic fields, and electronic inputs. These external
actuators generate forces that the body can withstand [1]. These technologies are
in their nascent stages, so advancements in the production of external signals will
be required for propelling their development, and therefore, genetically modified
biomaterials will reach heights in the near future.
66 A. Chhina et al.
Over the past fifteen years, there has been a lot of research into genetically modified
biomaterials, and CRISPR has already begun to play a significant part in the advance-
ment of this field. With the advancement of manufacturing techniques that produce
genetically engineered biomaterials, new ways have been paved for harnessing this
potential of the manufacturing sector and formulating genetically engineered bioma-
terials from it. Scalable production has provided hope for the synthesis of genetically
engineered nanofibrous materials. With features like biocompatibility, biodegrad-
ability, prospective energy collection, thermal, optical, and carcinogenic detection
potentialities, among others, these methodologies may arise in the not too-distant
future. Intelligent and genetically generated biomaterials and medicine delivery
systems have created tremendous strides in the last few years. It is being tested
to create new materials with unique properties. Researchers presently concentrate
mostly on designs found in nature, such as spider silk motifs, in order to develop
materials with outstanding mechanical properties. The design of novel materials, not
present in nature, that are based on an understanding of the relationship between
the structure of protein-based materials and biorecognition, self-assembly, and char-
acteristics has the most potential, though [1]. Bio adaptability would be the best
factor for selecting the optimal biomaterials that prove to be useful in certain tissue
repair. Biomaterials with exceptional bio adaptability must possess some essential
qualities, such as adaptable components that mimic cell response and tissue recon-
struction, matched mechanical properties with natural tissues, suitable surfaces with
advantageous tissue reaction, and biomimetic multi-level structures.
Tissue engineering and regenerative medicine, two techniques that are now in
development, will someday transform medical implants. Tissue engineering will
make it possible to create functional, living alternatives for many tissues and organs.
Tissue engineering will benefit from developments in biodegradable polymers, fast
prototyping, drug administration, cell culture, stem cell procedures, angiogenesis,
and biomimetic approaches for building extracellular, matrix-like biomimetics. The
potential applications of clever systems and micro- and nanofabrication are almost
endless. Future in vitro systems will be built on a sophisticated knowledge of how
biological systems engage with synthetic surfaces. These novel technologies will
also be carefully and morally addressed because they have the potential to alter
expectations for humans. Another area of study that will improve the functionality
of implants is lowering the risk of infection. This can be assisted by antibacterial
surface coatings, non-fouling surfaces, and antibiotic-controlled release mechanisms.
The development of devices that function as both a device and a drug will also be
made possible by advancements in controlled drug release in general.
Different Techniques of Genetic Engineering Used for the Development … 67
7 Conclusion
References
1. Bashor, C.J., Hilton, I.B., Bandukwala, H., Smith, D.M., Veiseh, O.: Engineering the next
generation of cell-based therapeutics. Nat. Rev. Drug Discov. 21(9), 655–675 (2022)
2. Kersey, A.L., Nguyen, T.U., Nayak, B., Singh, I., Gaharwar, A.K.: Omics-based approaches
to guide the design of biomaterials. Mater. Today. 2023.
68 A. Chhina et al.
3. Peppas, N.A., Langer, R.: New challenges in biomaterials. Science 263(5154), 1715–1720
(1994)
4. Ratner, B.D., Bryant, S.J.: Biomaterials: where we have been and where we are going. Annu.
Rev. Biomed. Eng. 6(1), 41–75 (2004)
5. Purbrick, M.D.: Book review: Surface modification of polymeric biomaterials. Edited by B. D.
Ratner and D. G. Castner. Plenum Publishing Corporation, New York, 1997. Polym Int. 46(1):
78–78 (1998)
6. Pierschbacher, M.D., Ruoslahti, E.: Cell attachment activity of fibronectin can be duplicated
by small synthetic fragments of the molecule. Nat. 309(5963), 30–33 (1984)
7. Sikavitsas, V.I., Temenoff, J.S., Mikos, A.G.: Biomaterials and bone mechanotransduction.
Biomaterials 22(19), 2581–2593 (2001)
8. Lloyd, A.W., Faragher, R.G., Denyer, S.P.: Ocular biomaterials and implants. Biomaterials
22(8), 769–785 (2001)
9. Ziats, N.P., Miller, K.M., Anderson, J.M.: In vitro and in vivo interactions of cells with
biomaterials. Biomaterials 9(1), 5–13 (1988)
10. Hench, L.L.: Biomaterials: a forecast for the future. Biomaterials 19(16), 1419–1423 (1998)
11. Patel, N.R., Gohil, P.P.: A review on biomaterials: scope, applications & human anatomy
significance. Int. J. Emerg. Technol. Adv. Eng. 2(4):91–101 (2012)
12. Biswal, T., BadJena, S.K., Pradhan, D.: Sustainable biomaterials and their applications: a short
review. Mater. Today: Proc. 1(30), 274–282 (2020)
13. Chutia, S.J., Yashwanth, B.S., Kumar, M., Nath, R.J., Bora, G., Sarma, D.K.: Adv. Genet. Eng.
(2020)
14. Zhang, Y., Zhou, Z., Sun, L., Liu, Z., Xia, X., Tao, T.H.: “Genetically engineered” biofunctional
triboelectric nanogenerators using recombinant spider silk. Adv. Mater. 30(50), 1805722 (2018)
15. Kim, K.S., Han, J.H., Park, J.H., Kim, H.K., Choi, S.H., Kim, G.R., Song, H., An, H.J.,
Han, D.K., Park, W., Park, K.S.: Multifunctional nanoparticles for genetic engineering and
bioimaging of natural killer (NK) cell therapeutics. Biomaterials 1(221), 119418 (2019)
16. Aleixandre, R.N., Shrikrishnan, S., Dalby, M.J., Salmeron-Sanchez, M.: Engineered living
biomaterials. Nat. Rev. Mater. 6(12), 1175–1190 (2021)
17. Zhu, D., Jiang, Z., Li, N., Wang, X., Ren, L., Ye, Y., Pan, Y., Yang, G.: Insights into the
use of genetically modified decellularized biomaterials for tissue engineering and regenerative
medicine. Adv. Drug Delivery Rev. 28, 114413 (2022)
18. Jiang, Z., Fu, M., Zhu, D., Wang, X., Li, N., Ren, L., He, J., Yang, G.: Genetically modified
immunomodulatory cell-based biomaterials in tissue regeneration and engineering. Cytokine
Growth Factor Rev. 2022
19. Dobres, S., Mula, G., Sauer, J., Zhu, D.: Applications of 3D printed chimeric DNA biomaterials.
ER 3(1), 13–23 (2022)
20. de León, E.H., Valle-Pérez, A.U., Khan, Z.N., Hauser, C.A.: Intelligent and smart biomaterials
for sustainable 3D printing applications. Curr. Opin. Biomed. Eng. 4, 100450 (2023)
21. Chambre, L., Martín-Moldes, Z., Parker, R.N., Kaplan, D.L.: Bioengineered elastin-and silk-
biomaterials for drug and gene delivery. Adv. Drug Delivery Rev. 1(160), 186–198 (2020)
22. Sengupta, D., Heilshorn, S.C.: Protein-engineered biomaterials: highly tunable tissue engi-
neering scaffolds. Tissue Eng. B Rev. 16(3), 285–293 (2010)
23. Bzymek, M., Lovett, S.T.: Instability of repetitive DNA sequences: the role of replication in
multiple mechanisms. Proc. Natl. Acad. Sci. U.S.A. 98(15), 8319–8325 (2001)
24. Maurya, A.K., Mishra, A., Mishra, N.: Nanoengineered polymeric biomaterials for drug
delivery system. In: Nanoengineered Biomaterials for Advanced Drug Delivery. Elsevier;
pp. 109–143 (2020)
25. Austin, M.J., Rosales, A.M.: Tunable biomaterials from synthetic, sequence-controlled
polymers. Biomater. Sci. 7(2), 490–505 (2019)
26. McGrath, K.P., Fournier, M.J., Mason, T.L., Tirrell, D.A.: Genetically directed syntheses of new
polymeric materials. Expression of artificial genes encoding proteins with repeating-(AlaGly)
3ProGluGly-elements. J. Am. Chem. Soc. 114(2), 727–33 (1992)
Different Techniques of Genetic Engineering Used for the Development … 69
27. Choudhury, D., Yee, M., Sheng, Z.L., Amirul, A., Naing, M.W.: Decellularization systems and
devices: State-of-the-art. Acta Biomater. 1(115), 51–59 (2020)
28. Kawecki, M., Łabuś, W., Klama-Baryla, A., Kitala, D., Kraut, M., Glik, J., Misiuga, M., Nowak,
M., Bielecki, T., Kasperczyk, A.: A review of decellurization methods caused by an urgent
need for quality control of cell-free extracellular matrix’scaffolds and their role in regenerative
medicine. J. Biomed. Mater. Res. Part B Appl. Biomater. 106(2), 909–23 (2018)
29. Yao, Q., Zheng, Y.W., Lan, Q.H., Kou, L., Xu, H.L., Zhao, Y.Z.: Recent development and
biomedical applications of decellularized extracellular matrix biomaterials. Mater. Sci. Eng. C
1(104), 109942 (2019)
30. Lü, W.D., Sun, R.F., Hu, Y.R., Lu, J.R., Gu, L., Liu, Z.G., Lei, G.Y., Qiang, Z., Cai, L.: Photoox-
idatively crosslinked acellular tumor extracellular matrices as potential tumor engineering
scaffolds. Acta Biomater. 15(71), 460–473 (2018)
31. Matuska, A.M., McFetridge, P.S.: The effect of terminal sterilization on structural and biophys-
ical properties of a decellularized collagen-based scaffold; implications for stem cell adhesion.
J. Biomed. Mater. Res. Part B Appl. Biomater. 103(2):397–406 (2015)
32. Keane, T.J., Swinehart, I.T., Badylak, S.F.: Methods of tissue decellularization used for
preparation of biologic scaffolds and in vivo relevance. Methods 1(84), 25–34 (2015)
33. Zou, J.L., Liu, S., Sun, J.H., Yang, W.H., Xu, Y.W., Rao, Z.L., Jiang, B., Zhu, Q.T., Liu, X.L.,
Wu, J.L., Chang, C.: Peripheral nerve-derived matrix hydrogel promotes remyelination and
inhibits synapse formation. Adv. Funct. Mater. 28(13), 1705739 (2018)
34. Yu, R., Zhang, H., Guo, B.: Conductive biomaterials as bioactive wound dressing for wound
healing and skin tissue engineering. Nano-Micro Lett. 14, 1–46 (2022)
35. Zhang, X., Chen, X., Hong, H., Hu, R., Liu, J., Liu, C.: Decellularized extracellular matrix
scaffolds: recent trends and emerging strategies in tissue engineering. Bioact. Mater. 1(10),
15–31 (2022)
36. Gomes, S., Leonor, I.B., Mano, J.F., Reis, R.L., Kaplan, D.L.: Natural and genetically
engineered proteins for tissue engineering. Prog. Polym. Sci. 37(1), 1–17 (2012)
37. Kleinheinz, J., Jung, S., Wermker, K., Fischer, C., Joos, U.: Release kinetics of VEGF165 from
a collagen matrix and structural matrix changes in a circulation model. Head Face Med. 6(1),
1–7 (2010)
38. Wongpanit, P., Ueda, H., Tabata, Y., Rujiravanit, R.: In vitro and in vivo release of basic
fibroblast growth factor using a silk fibroin scaffold as delivery carrier. J. Biomater. Sci. Polym.
Ed. 21(11), 1403–1419 (2010)
39. Friess, W., Uludag, H., Foskett, S., Biron, R., Sargeant, C.: Characterization of absorbable
collagen sponges as recombinant human bone morphogenetic protein-2 carriers. Int. J. Pharm.
185(1), 51–60 (1999)
40. Wang, X., Zhang, X., Castellot, J., Herman, I., Iafrati, M., Kaplan, D.L.: Controlled release
from multilayer silk biomaterial coatings to modulate vascular cell responses. Biomaterials
29(7), 894–903 (2008)
41. Shields, L.B., Raque, G.H., Glassman, S.D., Campbell, M., Vitaz, T., Harpring, J., Shields, C.B.:
Adverse effects associated with high-dose recombinant human bone morphogenetic protein-2
use in anterior cervical spine fusion. Spine J. 31(5), 542–547 (2006)
42. Lyu, Y., Wang, Y.: Output optimization of biodegradable triboelectric nanogenerators. Nano
Energy 16, 107811 (2022)
43. Briscoe, J., Dunn, S.: Piezoelectric nanogenerators–a review of nanostructured piezoelectric
energy harvesters. Nano Energy 1(14), 15–29 (2015)
44. Korkmaz, S., Kariper, İA.: Pyroelectric nanogenerators (PyNGs) in converting thermal energy
into electrical energy: Fundamentals and current status. Nano Energy 1(84), 105888 (2021)
45. Jiang, D., Su, Y., Wang, K., Wang, Y., Xu, M., Dong, M., Chen, G.: A triboelectric and pyroelec-
tric hybrid energy harvester for recovering energy from low-grade waste fluids. Nano Energy
1(70), 104459 (2020)
46. Ahmed, A., Hassan, I., Helal, A.S., Sencadas, V., Radhi, A., Jeong, C.K., El-Kady, M.F.: Tribo-
electric nanogenerator versus piezoelectric generator at low frequency (< 4 Hz): a quantitative
comparison. Iscience. 23(7), 101286 (2020)
70 A. Chhina et al.
47. Li, X., Jiang, C., Ying, Y., Ping, J.: Biotriboelectric nanogenerators: materials, structures, and
applications. Adv. Energy Mater. 10(44), 2002001 (2020)
48. Zhou, Y., Deng, W., Xu, J., Chen, J.: Engineering materials at the nanoscale for triboelectric
nanogenerators. Cell Rep. Phys. Sci. 1(8) (2020)
49. Zheng, N., Xue, J., Jie, Y., Cao, X., Wang, Z.L.: Wearable and humidity-resistant biomaterials-
based triboelectric nanogenerator for high entropy energy harvesting and self-powered sensing.
Nano Res. 15(7), 6213–6219 (2022)
50. Kim, J.N., Lee, J., Go, T.W., Rajabi-Abhari, A., Mahato, M., Park, J.Y., Lee, H., Oh, I.K.: Skin-
attachable and biofriendly chitosan-diatom triboelectric nanogenerator. Nano Energy 1(75),
104904 (2020)
51. Adhikary, P., Mahmud, M.P., Solaiman, T., Wang, Z.L.: Recent advances on biomechanical
motion- driven triboelectric nanogenerators for drug delivery. Nano Today 1(45), 101513 (2022)
52. Niu, Z., Cheng, W., Cao, M., Wang, D., Wang, Q., Han, J., Long, Y., Han, G.: Recent advances
in cellulose-based flexible triboelectric nanogenerators. Nano Energy 1(87), 106175 (2021)
53. Correa-Gallegos, D., Jiang, D., Rinkevich, Y.: Fibroblasts as confederates of the immune
system. Immunol. Rev. 302(1), 147–162 (2021)
54. Nidhi, S., Joseph, N., Vighnesh, N.P., Sabarinath, P.J., John, J., John, H., Padmanabhan, N.T.:
Recent updates on triboelectric nanogenerator based advanced biomedical technologies: A
short review. RINENG 100782 (2022)
55. Wang, E., Lee, S.H., Lee, S.W.: Elastin-like polypeptide based hydroxyapatite bionanocom-
posites. Biomacromol 12(3), 672–680 (2011)
56. Peddi, S., Roberts, S.K., MacKay, J.A.: Nanotoxicology of an elastin-like polypeptide
rapamycin formulation for breast cancer. Biomacromol 21(3), 1091–1102 (2020)
57. Lin, Y., Jin, W., Qiu, Y., Zhang, G.: Programmable stimuli-responsive polypeptides for
biomimetic synthesis of silica nanocomposites and enzyme self-immobilization. Int. J. Biol.
Macromol. 1(134), 1156–1169 (2019)
58. Butcher, N.J., Mortimer, G.M., Minchin, R.F.: Unravelling the stealth effect. Nat. Nanotechnol.
11(4), 310–311 (2016)
59. Schöttler, S., Becker, G., Winzen, S., Steinbach, T., Mohr, K., Landfester, K., Mailänder, V.,
Wurm, F.R.: Protein adsorption is required for stealth effect of poly (ethylene glycol)-and poly
(phosphoester)-coated nanocarriers. Nat. Nanotechnol. 11(4), 372–377 (2016)
60. Nidhi, S., Anand, U., Oleksak, P., Tripathi, P., Lal, J.A., Thomas, G., Kuca, K., Tripathi, V.:
Novel CRISPR–Cas systems: an updated review of the current achievements, applications, and
future research perspectives. Int. J. Mol. Sci. 22(7), 3327 (2021)
61. Chavez-Granados, P.A., Manisekaran, R., Acosta-Torres, L.S., Garcia-Contreras, R.: CRISPR/
Cas gene-editing technology and its advances in dentistry. Biochim. 1(194), 96–107 (2022)
62. Bhattacharjee, R., Jana, A., Nandi, A., Sinha, A., Bhattacharjee, A., Mitra, S., Kar, S., Dey,
A., Singh, S.K., Varma, S.R., Panda, P.K.: Synergy of Nanocarriers with CRISPR-Cas9 in an
Emerging Technology Platform for Biomedical Appliances: Current Insights and Perspectives.
Mater. Des. 24, 111415 (2022)
63. Eoh, J., Gu, L.: Biomaterials as vectors for the delivery of CRISPR–Cas9. Biomater. Sci. 7(4),
1240–1261 (2019)
64. Ghandehari, H.: Recombinant biomaterials for pharmaceutical and biomedical applications.
Pharm. Res. 25, 672–673 (2008)
65. Shadish, J.A., DeForest, C.A.: Site-selective protein modification: from functionalized proteins
to functional biomaterials. Matter. 2(1), 50–77 (2020)
66. Shim, G., Kim, D., Park, G.T., Jin, H., Suh, S.K., Oh, Y.K.: Therapeutic gene editing: delivery
and regulatory perspectives. Acta Pharmacol. Sin. 38(6), 738–753 (2017)
67. Gaharwar, A.K., Singh, I., Khademhosseini, A.: Engineered biomaterials for in situ tissue
regeneration. Nat. Rev. Mater. 5(9), 686–705 (2020)
68. Dhaliwal, A., Pelka, S., Gray, D.S., Moghe, P.V.: Engineering lineage potency and plasticity
of stem cells using epigenetic molecules. Sci. Rep. 8(1), 16289 (2018)
69. Larsson, L., Pilipchuk, S.P., Giannobile, W.V., Castilho, R.M.: When epigenetics meets bioengi-
neering—A material characteristics and surface topography perspective. J. Biomed. Mater. Res.
Part B Appl. Biomater. 106(5), 2065–71 (2018)
Different Techniques of Genetic Engineering Used for the Development … 71
70. James, E.N., Van Doren, E., Li, C., Kaplan, D.L.: Silk biomaterials-mediated miRNA
functionalized orthopedic devices. Tissue Eng. Part A 25(1–2), 12–23 (2019)
71. Asa’ad, F., Pelanyte, G., Philip, J., Dahlin, C., Larsson, L.: The role of epigenetic function-
alization of implants and biomaterials in osseointegration and bone regeneration—A review.
Molecules 25(24), 5879 (2020)
72. Hu, W., Li, Q., Li, B., Ma, K., Zhang, C., Fu, X.: Optogenetics sheds new light on tissue
engineering and regenerative medicine. Biomaterials 1(227), 119546 (2020)
73. Williams, T.L., Iskandar, D.J., Nödling, A.R., Tan, Y., Luk, L.Y., Tsai, Y.H.: Transferability of
N-terminal mutations of pyrrolysyl-tRNA synthetase in one species to that in another species
on unnatural amino acid incorporation efficiency. Amino Acids 53, 89–96 (2021)
74. Sisila, V., Indhu, M., Radhakrishnan, J., Ayyadurai, N.: Building biomaterials through genetic
code expansion. Trends Biotechnol. (2022)
75. Haq-Siddiqi, N.A., Britton, D., Montclare, J.K.: Protein-engineered biomaterials for cartilage
therapeutics and repair. Adv. Drug Deliv. Rev. 9, 114647 (2022)
76. Kowalski, P.S., Bhattacharya, C., Afewerki, S., Langer, R.: Smart biomaterials: recent advances
and future directions. ACS Biomater. Sci. Eng. 4(11), 3809–3817 (2018)
77. Lyu, S., Fang, J., Duan, T., Fu, L., Liu, J., Li, H.: Optically controlled reversible protein
hydrogels based on photoswitchable fluorescent protein Dronpa. ChemComm. 53(100), 13375–
13378 (2017)
78. Finch, L., Harris, S., Solomou, G., Sen, J., Tzerakis, N., Emes, R.D., Lane, C.S., Hart, S.R.,
Adams, C.F., Chari, D.M.: Safe nanoengineering and incorporation of transplant populations
in a neurosurgical grade biomaterial, DuraGen PlusTM , for protected cell therapy applications.
JCR 10(321), 553–563 (2020)
79. Diomede, F., D’Aurora, M., Gugliandolo, A., Merciaro, I., Orsini, T., Gatta, V., Piattelli, A.,
Trubiani, O., Mazzon, E.: Biofunctionalized scaffold in bone tissue repair. Int. J. Mol. Sci.
19(4), 1022 (2018)
80. Ribeiro-Filho, L.A., Sievert, K.D.: Acellular matrix in urethral reconstruction. Adv. Drug Deliv.
Rev. 1(82), 38–46 (2015)
81. Thomas, S.N., Archer, P.A., Manspeaker, M.P.: Biomaterials for Immunoengineering. In:
Biomaterials Science. Academic Press, pp. 1199–1215 (2020)
82. Dellacherie, M.O., Seo, B.R., Mooney, D.J.: Macroscale biomaterials strategies for local
immunomodulation. Nat. Rev. Mater. 4(6), 379–397 (2019)
83. Seyfoori, A., Amereh, M., Dabiri, S.M., Askari, E., Walsh, T., Akbari, M.: The role of biomate-
rials and three dimensional (3D) in vitro tissue models in fighting against COVID-19. Biomater.
Sci. 9(4), 1217–1226 (2021)
84. Kopeček, J.: Smart and genetically engineered biomaterials and drug delivery systems. Eur. J.
Pharm. Sci. 20(1), 1–6 (2003)
Abbreviations
TE Tissue Engineering
BE Bone Engineering
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 73
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_3
74 A. Hazra et al.
1 Introduction
With an increase in the rates of diseases affecting various organs of the human body,
and the rate expected to double by the end of 2030, the need for the ability to replace
any such organ has become imperative [1]. It has led to the expansion of the field of
medicine known as regenerative medicine, which involves tissue engineering (TE),
regeneration of bones, and self-healing through activation of the host’s immune
system [2]. It is a multidisciplinary discipline that integrates physics concepts with
principles derived from life science to provide clinical treatment strategies. The
concept of regenerative medicine emerged in the early 1990s, and since then, efforts
have been made to strengthen research in the field.
Although bone bioengineering is a type of tissue engineering, it is considered
distinct from tissue engineering of other organs in today’s context, with bone engi-
neering being one of the most developed arenas. The creation of bio-active artificial
cells and tissues for the substitution and regeneration of faulty tissues or organs is the
focus of tissue engineering [3]. This includes isolation of suitable cells, development
of compatible scaffolds, and implantation of the molecules into the tissues to alter
functions or initiate repair of damaged organs and tissues [2]. Bone engineering (BE)
involves the synergistic effect of engineered biomaterials with the host’s recovery
factors to regenerate bone tissues. It appears to have no side effects so far and allows
for healing of bone fractures and other flaws using the host’s cells [4]. However, with
Green Methods for the Development of Bone and Tissue … 75
the progress and improvement in the field, there has been a distinct rate of pollution
because of it and has led to the environmental depletion.
The raw materials being used, the chemicals being depleted, and the destruction of
faulty end products have led to soil erosion as well as major water pollution. It has also
been known to affect the health of individuals being involved in the process which
has now led to the thought of developing a process that is responsible toward human
health and environment- green practice. Green practices involve the implementation
of resource- and environmentally conscious methods throughout the entire lifecycle,
from design to construction, operation, maintenance, renovation, and destruction [5].
The traditional considerations of economy, usefulness, durability, and comfort are
expanded and are now being complemented with green practices.
The primary concept of green practices was presented by Anastas and Warner who
put forward the foundation based on 12 principles, which revolves around reducing
waste, hazardous products and derivatives, balancing atom economy and energy
efficiency, developing designs that make use of catalysis and based on the usage of
benign compounds and renewable sources, and always having a detailed plan about
degradation of the product, pollution control, and accident prevention. Thus, in the
light of 2023, today scientists from all fields are working on improving the production
of biomaterials such that it keeps the environment stress free [6].
not every substance can be modified in them, reducing the range of sources for
producing scaffolds in tissue engineering [18]. Among each of the other tissue engi-
neering processes, bone regeneration is the most critical one as it is founded on
numerous principles of molecular and cellular biology, biological development, and
structural biology, all of which are monitored by biomechanics and bioengineering.
This is because bone tissues are responsible for dynamics of the body and must be
adjusted according to its different locations. The most difficult aspect of bone tissue
development is retaining vascularization of blood vessels by making the scaffolding
porous and integrating it with the host system. With current synthetic mechanisms,
only the periphery of a scaffolding is suitable to be used as an implantation thus
limiting its use. Owing to these reasons, the synthetic mechanisms for production of
bone tissues are not widely in use [19].
Usually, the current strategies are based on either the cellular or source material
mechanism, thus for the usage to be widespread all the factors must be collaborated.
Bone engineering is also an expensive process due to it being patient-dependent
and poses health issues due to erroneous cell isolations, seeding, culturing, and
implantation [20]. Methods need to be established such that all the factors can be
culminated, and they impose no adverse effects on human health [4]. Moreover,
the growth in the field of tissue regeneration has been quite stagnant over the past
two decades. The clinical translations of the basic research for bone, cartilage, and
ligament regeneration have met a roadblock ever since the involvement of stem
cells had been implemented. Though the synthetic polymers at one time seemed
like the most promising solution to tissue engineering, in the current scenario there
seems to be an unresolved failure at the translation stage for application of the
methods [4, 21]. Also, for the current approaches to be effective a proper cell for the
source must be determined for the model to be designed. However, the separation of
human cells and culturing it in vitro such that its sensitivity is maintained, and its
differentiation capacity is not reduced is a tedious task. Also, for some cells to work
they need to be converted into their embryonic forms. Furthermore, the different
methods are influenced by the tissue origin, the patient’s age, and the individual’s
health condition. Also, the environment around the cell must be replicated exactly
to establish the correct behavior of the polymer of the scaffold. Thus is the need of
designing materials from natural sources which can be used as scaffolds by exploiting
their chemical, physical, and biological properties [22–29]. These natural fibers can
also be combined with synthetic materials depending on the need of the tissue at the
site [30].
Another issue that crops up while developing regenerated tissues is the innervation
of those tissues. For the structures to attain complete functionality after implantation,
the structures need to get consistent with other growing tissues in the region [31].
These requirements of innervations have been seen to be very critical for regions like
bladders and small intestines [32]. This association not only develops an interlink
with the nervous system but also rejuvenates the tissue regenerated [33, 34]. For
the bioengineering of peripheral nerves, an important criterion is the inculcation of
neurotrophic factors which would ultimately lead to synapse formation, even muscle
Green Methods for the Development of Bone and Tissue … 77
tissues require electric impulse for stimulation. These are critical steps and are diffi-
cult to implement as well [35, 36]. Some grafted organs require neovascularization
to occur, such that the branches developed after morphogenesis can help in accep-
tance of the implantation, but it is more than often seen to be not there with synthetic
polymers. However, in other cases the synthetic materials sometimes exceed the
functionality rate of native tissues, leading to some complications [37, 38]. Overall
many trials with synthetic fibers have been postponed owing to the financial factors
as some of the synthetic fibers which are best suited for human use have a high cost
[39].
The idea of developing environmentally friendly techniques for bone and tissue
engineering arose because of the dangers that synthetic techniques posed to the
human body and the environment [40]. This resulted in the creation of engineered
biomaterials and scaffolds composed of natural polymers that offer little to no risk to
the environment or human health. Green approaches involve using healthy, non-toxic
materials and procedures to lessen the hazardous and damaging effects of scientific
study. The reduction of pollution’s harmful impacts on the environment and the
human body is a key objective of green technologies and practices [41]. The goal
of bone tissue engineering (BTE) is to create new, functional bone tissues using
knowledge of the structure of bones, bone mechanics, and tissue building. To put it
another way, if you want to efficiently regenerate as well as repair bone, you must
first understand the biology of bone and how it develops. The ideas for biologically
active chemicals, scaffolds, engineering, and material production are also combined
to build, repair, or treat injured tissues or organs [42–44].
Because of their inherent bioactivity, including cell adhesion and proliferation,
as well as the fact that they are harmless to native, healthy tissue, natural biomate-
rials are critical for tissue engineering. Biomaterials made from natural substances
include those made from glycosaminoglycans, cellulose, chitin, collagen, chitosan,
elastin, alginate, fibrinogen, dextran, gelatin, laminin, and silk. Among these mate-
rials, collagen, fibrinogen, glycosaminoglycans, elastin, and laminin are mimics of
original extracellular matrix (ECM) components. Extracellular matrix is crucial for
tissue regeneration because it supports cells physically and controls cellular processes
including growth, proliferation, differentiation, and homeostasis, enabling the body
to repair tiny flaws in cells, tissues, and organs [45, 46].
The human body is unable to regenerate deficiencies because of a shortage of
extracellular matrix (ECM) to make up large defects. Natural biomaterials were
created in this manner to be employed in applications involving tissue engineering.
These natural biomaterials are employed in hydrogels, that additionally serve as an
excellent tool for tissue engineering. Hydrogels are capable of being made from
both synthetic and naturally occurring biomaterials. Artificial polymers include poly
78 A. Hazra et al.
HEMA, PEO, and PVA. Naturally occurring biomaterials used in hydrogel prepa-
ration include collagen, chitosan, gelatin, fibrin, alginate, agarose, and many more
[47].
3.1 Collagen
3.2 Chitosan
material for bone scaffolds because it promotes osteoblast cell adhesion and prolif-
eration as well as the development of mineralized bone matrix. Because chitosan
is a polymer composed of amino acids and polysaccharides, it contains breakable
glycosidic bonds. In vivochitosan degradation is typically carried out by the enzyme
lysozyme [60].
Chitosan has been combined with a variety of delivery components, including algi-
nate, hydroxyapatite, hyaluronic acid, calcium phosphate, PMMA, poly-l-lactic acid
(PLLA), and growth factors, for potential use in orthopedics. Chitosan, in general,
offers a wide range of options for cell-based tissue creation. Chitosan can be made
into 3D highly interconnected, porous structures with a high degree of porosity using
a variety of technologies, allowing cells to proliferate and move nutrients. Porous
scaffolds, for example, can be created by lyophilizing a frozen chitosan solution in
acetic acid [61]. Chitosan and acetic acid may phase split and form ice crystals when
frozen. The scaffold develops holes as a result of the sublimation of acetic acid ice
crystals during the freeze-drying process. Another method is to use porogens (salts,
sugar) in conjunction with chitosan in a melt-based technique [62].
Rapid prototyping technology can also be used to create chitosan scaffolds. This
is a computer-aided method [63]. The use of chitosan-based scaffolds that mimic
some bone characteristics can improve osteoblast adhesion and proliferation. For
example, chitosan scaffolds combined with bioactive ceramics can impart osteo-
conductive and even osteo-inductive properties to the final structure, directing bone
development engineering [64]. Chitosan, in combination with hydroxyapatite and
tricalcium phosphates, has shown promising results in mesenchymal stem cell (MSC)
adhesion, proliferation, and differentiation, as well as intriguing results in bone tissue
engineering [65]. Chitosan has been used to improve the biological and mechanical
properties of marine-derived polymers in order to provide an alternative to collagen
derived from animal sources. When combined with polyesters, chitosan increased cell
viability and adhesion [62]. Development of scaffolds from chitosan is in flowchart
shown in Fig. 2.
3.3 Gelatin
Gelatin is a naturally occurring biopolymer created when animal collagen from skin,
bones, and tendons is partially hydrolyzed in an acid or alkaline solution, and the
structure is very similar to collagen. It is comprised of various proteins and amino
acids. Regardless of how collagen is hydrolyzed to create gelatin, both polymers
include up to 20 distinct amino acids in varying amounts in their basic structure.
This basic structure provides a three-amino-acid detection sequence for integrin-
mediated adhesion of cells. The properties of gelatin are determined by the source
of collagen, the type of collagen, and the method of collagen conversion to gelatin.
Various methods are implemented for the creation of gelatin scaffolds. One such
process includes electrospinning. The production of polyelectrolyte colloidal sol
makes it very challenging to electro-spun gelatin using water as the solvent.
Green Methods for the Development of Bone and Tissue … 81
Fig. 2 Development of
Scaffolds from Chitosan
The silky gelatin nanofibers were created using a variety of polar organic solvents,
including trifluoroethanol and hexafluoro isopropanol [66]. Biomaterials made of
gelatin are widely used in tissue engineering. Gelatin scaffolds demonstrated high
DNA, GAG, and collagen contents and enabled the development of chondrogenic
cells. Gelatin used with chitosan was used in skin tissue engineering [67]. Much
interest has been shown in electro-spun nanofibrous scaffolds as prospective plat-
forms for bone repair and regeneration. Degradable polymers, whether they are
synthetic or natural, were used as a starting point for research into the advanta-
geous properties of nanofibrous matrices. Many synthetic polymers’ hydrophobic
properties restrict cell affinities and early cell adhesion. Another method to increase
hydrophilicity and, consequently, cell compatibility in polymers is to combine
synthetic and natural polymers. This is done using a composite of polycaprolactone-
gelatin scaffolds. Gelatin-apatite composite was also used since it mimicked the
properties of bone ECM [68].
Silk fibroin has been extensively researched for tissue engineering applications due
to its superior biological and mechanical strength, as well as biocompatibility and
biodegradability.
Silk is made up of fibrous protein (SF) and sericin. Silk is produced by a variety
of arthropods, including spiders and silkworms with the most used silk produced
by the silkworm Bombyx mori. SF has been shown to be very biocompatible. After
observing its attachment to fibroblast cells and growth on SF matrices, its role as a
biomaterial was evaluated. Silk fibroin can be easily separated from sericin by using
a steaming alkaline or surfacting solution (degumming). The resulting fibers have a
82 A. Hazra et al.
diameter of 10–25 mm and are composed of a hydrophobic heavy chain (350 kDa)
and a hydrophilic light chain (25 kDa).
Depending on the type of scaffold required, silk fibroin can be created using
a variety of techniques. One such technique is salt leaching using NaCl particles
for scaffolds with spherical pores and for scaffolds with lamellar pores a process
involving freezing, and lyophilizing was used which resulted in the formation of beta
sheets. Another technique included electrospinning which produced fibers with low
tensile strength [69]. Silk fibroin is a very useful biomaterial in tissue engineering.
By making the fibers stiffer, it was seen to increase osteogenic induction. Silk used
along with calcium phosphate in engineering of bone showed increased stability, and
bioactivity with no toxicity. An osteo-inductive composite was created by combining
silk and HAp, the important mineral seen in bone, to replicate the composition of
bone. HAp improved the scaffolds’ compressive strength as well as the ability of
cells to proliferate and produce bone. Other biofunctional substances for restoration
or therapy of tissues of bone are titanium dioxide (TiO2 ) nanoparticle and natural
silk fibroin-based nanocomposite scaffolds [70, 71]. The development of scaffolds
from silk fibroin in flowchart is shown in Fig. 3.
3.5 Starch
starch, which contains 15–30% amylose, and higher content of amylose, which
contains more than 50% amylose. Crystallinity can range from 15% in high-amylose
starch to 50% in waxy starch [72]. Starch’s brittleness prevents it from being used in
most situations. As a result, starch must be modified or combined with other polymers
to improve its properties.
Starch scaffolds can be produced by traditional casting in solvent procedures. In
this procedure, the starch is dissolved in chloroform and cast into a mold. Cellu-
lose and starch were combined by leaching with salt and freeze-drying methods
in a different approach. Wet spinning method is also used to create scaffolds of
desired pore size which include mixing of starch with other polymers. Chitosan-starch
microparticles can be obtained from water in oil emulsification techniques. Starch
can also be used along with HAp and chitosan which is processed by co-precipitation
[73]. Several studies on bone tissue engineering have used starch-based scaffolds.
When inserted into a critical-sized defect, scaffolds made of polycaprolactone, starch,
and stem cells boosted the repair of bone. In comparison to scaffolds loaded with
no differentiated cells, scaffolds loaded with osteoblastic cells produced more new
bone. Modified starch scaffolds and polycaprolactone with silanol group showed
improved osteogenic differentiation and bone healing. 3D scaffolds promoted adhe-
sion and recruitment of cells and promoted tissue regeneration [74]. The development
of scaffolds from starch in Flowchart is shown in Fig. 4.
The goal of numerous studies is to create techniques that are cost-effective, low-
energy, or sustainable to produce various tissues while decreasing the consumption
of harmful ingredients. One of the safest ways to create tissues and organs is to apply
natural substances taken from plants or animal bones [75]. Synthetic materials exhibit
physicochemical properties that may be regulated during the production process.
They are also robust, affordable, dependable, frequently easily electro spinnable,
and strong. Their lack of cell-recognition sites, however, results in a low affinity for
cell attachment.
Thus, natural polymers are favored because they resemble the macromolecular
components of the human body [76]. The main benefits of bio-based and biodegrad-
able polymers include reducing GHG emissions, utilizing local resources, reusing
by products, and preserving fossil fuels. Without any prior processing, biopolymers
print beautifully and have good scent and fat barriers. Polymers made of starch offer
excellent surface finishing and antistatic characteristics. Naturally occurring biopoly-
mers (collagen, chitosan, starch, silk) have a better role in cell adhesion, proliferation,
and less cytotoxicity, hence are better materials for scaffold preparation for tissue
engineering. Polymers made from natural materials outperform semi-synthetic or
synthetic polymers in replicating the ECM and interacting with tissues, owing to
their high resemblance to the tissue environment [77].
Despite having the above said advantages, natural polymers do have some disad-
vantages, such as often high production costs (collagen, hyaluronic acid). Its complex
structural and chemical makeup, as well as macromolecular complexity, architec-
ture and shape, unpredictable hydration rate, resource limitations, and potential for
microbial deterioration are further disadvantages that may limit their use in tissue
engineering. Furthermore, because of their low stability, the rate of disintegration
and catabolization is high of some spontaneously created scaffolds is higher than the
pace of regeneration of the host tissue. Certain natural polymers, like cellulose and
chitosan, have limited mechanical characteristics and some have poor processability
e.g., polypeptides. Synthetic biopolymers have a few advantages over natural poly-
mers, such as a tunable and engineerable hydrophilic/hydrophobic ratio, a faster rate
of breakdown, and better mechanical properties. Another disadvantage of natural
biopolymers is degradation, so it can’t be used for a long period of time [78].
Though there has been overwhelming progress in the field of green bioengineering,
some constraints have always been present. The acquisition of the natural sources and
its behavior has always been a point of criticality. Current focus is being shifted to
use perennial plants as source material so that the restriction due to seasonal changes
can be avoided [79]. This would be helpful as the production would not be limited
Green Methods for the Development of Bone and Tissue … 85
by time frames. Also, plant sources are being tried to be extracted from plants which
have broader availability, that it is found in multiple regions to make the production
uniform [80, 81]. Also, to make the environment a safer place and reduce economical
stress, waste utilization has cropped up as a concept over the years, where agricultural
wastes are being worked upon to lead to the conformation of different scaffolding
frameworks [82].
Also, currently most production processes require high energy, which differs
depending on the source material that would be extracted. One of the current aims
is to develop methodologies or extract forms that can be molded at less than 100 °C
[83, 84]. Despite a lot of challenges in the field, the major challenges that need to be
combated include, developing the natural polymer in a way that it imitates all struc-
tural, functional, and morphological properties of the said tissue, and can also show
protein adsorption as well as inter- and intra-cellular interactions. Also, they must be
suitable for drug delivery as well as having a window for chemical and biomechanical
modifications [85, 86]. Thus, though the research in the field has made remarkable
discoveries with respect to the use of natural polymers, they are now trying to make
it more feasible and extraction and development to be easier as well as affordable
[87].
6 Conclusion
Current research in this field has dealt with the extraction, development, and modifica-
tion of natural polymers to produce particulars with concerns to bone and engineering
of tissue. The use of polysaccharides and hydrogels has given a new dimension to
discovery in this field and has contributed toward reducing environmental damage
and quite a bit of the economic load. These new compositions have been seen to have
better congregation with human tissues and thus can work toward the replacement of
faulty ones. The progression of them into organ transplantation remains to be envis-
aged into. The multiple methodologies that have been developed for regeneration are
still left to be optimally standardized and its reach to be increased. The novel strate-
gies and newly constructed biomaterials have played a pivotal role in turning engi-
neered templates for tissue replacement, into fully functional independent systems.
They possess all characteristics including biocompatibility, conductivity of impulses,
cellular interactions, and integration with host’s native cells. Further research aims
at investing the natural polymers into other complicated surgeries and formulations
of degradable, injectable, and mechanically stronger components for restoration of
functionality of various organs.
86 A. Hazra et al.
References
1. Baroli, B.: From natural bone grafts to tissue engineering therapeutics: brainstorming on
pharmaceutical formulative requirements and challenges. J. Pharm. Sci. 98(4), 1317–1375
(2009)
2. Carleton, P.F., Schachter, S., Parrish, J.A., Collins, J.M., Crocker, J.B., Dixon, R.F., Edgman-
Levitan, S., Lewandrowski, K.B., Stahl, J.E., Klapperich, C., Cabodi, M.: National institute of
biomedical imaging and bioengineering point-of-care technology research network: advancing
precision medicine. IEEE J. Trans. Eng. Health Med. 16(4), 1–4 (2016)
3. Panebianco, C.J., Dutta, P., Frost, J.R., Huang, A., Kim, O.S., Iatridis, J.C., Vernengo, A.J.,
x7uWeiser, J.R.: Teaching tissue repair through an inquiry-based learning bioadhesives module.
Biomed. Eng. Educ. 4, 1–4 (2022)
4. Amini, A.R., Laurencin, C.T., Nukavarapu, S.P.: Bone tissue engineering: recent advances and
challenges. Crit. Rev. Biomed. Eng. 40(5) (2012)
5. Anastas, P.T., Warner, J.C.: Principles of green chemistry. Green Chem. Theory Pract. 29 (1998)
6. DeVierno, K.A., House-Knight, T., Whitford, J., Ponnusamy, E., Miller, P., Jesse, N., Roden-
born, R., Sayag, S., Gebel, M., Aped, I., Sharfstein, I.: A method for assessing greener alterna-
tives between chemical products following the 12 principles of green chemistry. ACS Sustain.
Chem. Eng. 5(4), 2927–2935 (2017)
7. De Angelis, G., Medeghini, L., Conte, A.M., Mignardi, S.: Recycling of eggshell waste into
low-cost adsorbent for Ni removal from wastewater. J. Clean. Prod. 15(164), 1497–1506 (2017)
8. Oberbek, P., Bolek, T., Chlanda, A., Hirano, S., Kusnieruk, S., Rogowska-Tylman, J., Nechy-
porenko, G., Zinchenko, V., Swieszkowski, W., Puzyn, T.: Characterization and influence of
hydroxyapatite nanopowders on living cells. Beilstein J. Nanotechnol. 9(1), 3079–3094 (2018)
9. Dulińska-Molak, I., Mao, H., Kawazoe, N., Chen, G.: Effect of single-wall carbon nanotubes
on mechanical property of chondrocytes. J. Nanosci. Nanotechnol. 14(3), 2459–2465 (2014)
10. Hembrick-Holloman, V., Samuel, T., Mohammed, Z., Jeelani, S., Rangari, V.K.: Ecofriendly
production of bioactive tissue engineering scaffolds derived from egg-and seashells. J. Market.
Res. 9(6), 13729–13739 (2020)
11. Huang, R., Zhu, X., Tu, H., Wan, A.: The crystallization behavior of porous poly (lactic acid)
prepared by modified solvent casting/particulate leaching technique for potential use of tissue
engineering scaffold. Mater. Lett. 1(136), 126–129 (2014)
12. Patricio, T., Domingos, M., Gloria, A., Bártolo, P.: Characterisation of PCL and PCL/PLA
scaffolds for tissue engineering. Proc. Cirp. 1(5), 110–114 (2013)
13. Johari, N., Fathi, M.H., Golozar, M.A.: Fabrication, characterization and evaluation of the
mechanical properties of poly (E-caprolactone)/nano-fluoridated hydroxyapatite scaffold for
bone tissue engineering. Compos. B Eng. 43(3), 1671–1675 (2012)
14. Zhu, X., Zhong, T., Huang, R., Wan, A.: Preparation of hydrophilic poly (lactic acid) tissue
engineering scaffold via (PLA)-(PLA-b-PEG)-(PEG) solution casting and thermal-induced
surface structural transformation. J. Biomater. Sci. Polym. Ed. 26(17), 1286–1296 (2015)
15. Loh, Q.L., Choong, C.: Three-dimensional scaffolds for tissue engineering applications: role
of porosity and pore size. Tissue Eng. Part B Rev. 19(6), 485–502 (2013). https://doi.org/10.
1089/ten.TEB.2012.0437
16. Shuai, C., Liu, G., Yang, Y., Qi, F., Peng, S., Yang, W., He, C., Wang, G., Qian, G.: A strawberry-
like Ag-decorated barium titanate enhances piezoelectric and antibacterial activities of polymer
scaffold. Nano Energy 1(74), 104825 (2020)
17. Yang, Y., Lu, C., Peng, S., Shen, L., Wang, D., Qi, F., Shuai, C.: Laser additive manufacturing
of Mg-based composite with improved degradation behaviour. Virtual Phys. Prototyp. 15(3),
278–293 (2020)
18. Yang, S., Leong, K.F., Du, Z., Chua, C.K.: The design of scaffolds for use in tissue engineering.
Part II. Rapid prototyping techniques. Tissue Eng. 8(1), 1–1 (2002)
19. Hollister, S.J., Murphy, W.L.: Scaffold translation: barriers between concept and clinic. Tissue
Eng. Part B Rev. 17(6), 459–474 (2011)
Green Methods for the Development of Bone and Tissue … 87
20. Alman, B.A., Kelley, S.P., Nam, D.: Heal thyself: using endogenous regeneration to repair
bone. Tissue Eng. Part B Rev. 17(6), 431–436 (2011)
21. Quarto, R., Giannoni, P.: Bone tissue engineering: past–present–future. Mesenchymal Stem
Cells Meth. Protoc. 21–33 (2016)
22. Baino, F., Vitale-Brovarone, C.: Three-dimensional glass-derived scaffolds for bone tissue
engineering: current trends and forecasts for the future. J. Biomed. Mater. Res. A 97(4), 514–535
(2011). https://doi.org/10.1002/jbm.a.33072
23. Baino, F., Novajra, G., Vitale-Brovarone, C.: Bioceramics and scaffolds: a winning combination
for tissue engineering. Front. Bioeng. Biotechnol. 17(3), 202 (2015)
24. Boffito, M., Sartori, S., Ciardelli, G.: Polymeric scaffolds for cardiac tissue engineering:
requirements and fabrication technologies. Polym. Int. 63(1), 2–11 (2014)
25. Boffito, M., Bernardi, E., Sartori, S., Ciardelli, G., Sassi, M.P.: A mechanical characterization
of polymer scaffolds and films at the macroscale and nanoscale. J. Biomed. Mater. Res., Part
A 103(1), 162–169 (2015)
26. Chiono, V., Sartori, S., Calzone, S., Boffito, M., Tonda-Turo, C., Mattu, C., Gentile, P.,
Ciardelli, G.: Synthetic biodegradable medical polyurethanes. In: Science and Principles of
Biodegradable and Bioresorbable Medical Polymers, pp. 189–216. Woodhead Publishing
(2017)
27. Sartori, S., Boffito, M., Serafini, P., Caporale, A., Silvestri, A., Bernardi, E., Sassi, M.P.,
Boccafoschi, F., Ciardelli, G.: Synthesis and structure–property relationship of polyester-
urethanes and their evaluation for the regeneration of contractile tissues. React. Funct. Polym.
73(10), 1366–1376 (2013)
28. Silvestri, A., Sartori, S., Boffito, M., Mattu, C., Di Rienzo, A.M., Boccafoschi, F., Ciardelli,
G.: Biomimetic myocardial patches fabricated with poly (E-caprolactone) and polyethylene
glycol-based polyurethanes. J. Biomed. Mater. Res. B Appl. Biomater. 102(5), 1002–1013
(2014)
29. Sionkowska, A.: Current research on the blends of natural and synthetic polymers as new
biomaterials. Prog. Polym. Sci. 36(9), 1254–1276 (2011)
30. Caddeo, S., Boffito, M., Sartori, S.: Tissue engineering approaches in the design of healthy and
pathological in vitro tissue models. Front. Bioeng. Biotechnol. 26(5), 40 (2017)
31. Oberpenning, F., Meng, J., Yoo, J.J., Atala, A.: De novo reconstitution of a functional
mammalian urinary bladder by tissue engineering. Nat. Biotechnol. 17(2), 149–155 (1999)
32. Nakase, Y., Nakamura, T., Kin, S., Nakashima, S., Yoshikawa, T., Kuriu, Y., Miyagawa, K.,
Sakakura, C., Otsuji, E., Ikada, Y., Yamagishi, H.: Endocrine cell and nerve regeneration in
autologous in situ tissue-engineered small intestine. J. Surg. Res. 137(1), 61–68 (2007)
33. Auger, F.A., Berthod, F., Moulin, V., Pouliot, R., Germain, L.: Tissue-engineered skin substi-
tutes: from in vitro constructs to in vivo applications. Biotechnol. Appl. Biochem. 39(3),
263–275 (2004)
34. Suuronen, E.J., McLaughlin, C.R., Stys, P.K., Nakamura, M., Munger, R., Griffith, M.: Func-
tional innervation in tissue engineered models for in vitro study and testing purposes. Toxicol.
Sci. 82(2), 525–533 (2004)
35. Burdick, J.A., Ward, M., Liang, E., Young, M.J., Langer, R.: Stimulation of neurite outgrowth
by neurotrophins delivered from degradable hydrogels. Biomaterials 27(3), 452–459 (2006)
36. Dennis, R.G., Dow, D.E.: Excitability of skeletal muscle during development, denervation, and
tissue culture. Tissue Eng. 13(10), 2395–2404 (2007)
37. Risau, W.: Mechanisms of angiogenesis. Nature 386(6626), 671–674 (1997)
38. Lutolf, M.P., Gilbert, P.M., Blau, H.M.: Designing materials to direct stem-cell fate. Nature
462(7272), 433–441 (2009)
39. Olson, J.K., Boldyrev, A.I.: Ab initio characterization of the flexural B3H8-anion found in the
reversible dehydrogenation. Comput. Theor. Chem. 967(1), 1–4 (2011)
40. Chahal, S., Kumar, A., Hussian, F.S.: Development of biomimetic electrospun polymeric
biomaterials for bone tissue engineering. a review. J. Biomater. Sci. Polym. Ed. 30(14),
1308–1355 (2019)
88 A. Hazra et al.
41. Yu, X., Tang, X., Gohil, S.V., Laurencin, C.T.: Biomaterials for bone regenerative engineering.
Adv. Healthcare Mater. 4(9), 1268–1285 (2015)
42. Li, J.J., Kaplan, D.L., Zreiqat, H.: Scaffold-based regeneration of skeletal tissues to meet clinical
challenges. J Mater Chem B. 2, 7272–7306 (2014)
43. Dimitriou, R., Jones, E., McGonagle, D., Giannoudis, P.V.: Bone regeneration: current concepts
and future directions. BMC Med. 9(1), 1–0 (2011)
44. Iravani, A., Akbari, M.H., Zohoori, M.: Advantages and disadvantages of green technology;
goals, challenges and strengths. Int. J. Sci. Eng. Appl. 6(9), 272–284 (2017)
45. Mukherjee, S., Reddy Venugopal, J., Ravichandran, R., Ramakrishna, S., Raghunath, M.: Eval-
uation of the biocompatibility of PLACL/collagen nanostructured matrices with cardiomy-
ocytes as a model for the regeneration of infarcted myocardium. Adv. Func. Mater. 21(12),
2291–2300 (2011)
46. Theocharis, A.D., Skandalis, S.S., Gialeli, C., Karamanos, N.K.: Extracellular matrix structure.
Adv. Drug Deliv. Rev. 1(97), 4–27 (2016)
47. Ullah, S., Chen, X.: Fabrication, applications and challenges of natural biomaterials in tissue
engineering. Appl. Mater. Today 1(20), 100656 (2020)
48. Brodsky, B., Werkmeister, J.A., Ramshaw, J.A.: Collagens and gelatins. Biopolymers online.
Wiley-VCH Verlag GmbH & Co. KgaA, Germany (2005)
49. Matthews, J.A., Wnek, G.E., Simpson, D.G., et al.: Electrospinning of collagen nanofibers.
Biomacromol 3, 232–238 (2002)
50. Parry, D.A., Craig, A.S.: Collagen fibrils during development and maturation and their contri-
bution to the mechanical attributes of connective tissue. In: Collagen: Volume II Biochemistry
and Biomechanics, pp. 1–23. CRC Press (2018)
51. Xie, J., Lou, X., Wang, X., Yang, L., Zhang, Y.: Electrospun nanofibers of hydroxyapatite/
collagen/chitosan promote osteogenic differentiation of the induced pluripotent stem cell-
derived mesenchymal stem cells. J. Control. Release Off. J. Control. Release Soc. 213, e53
(2015)
52. Maia, F.R., Correlo, V.M., Oliveira, J.M., Reis, R.L.: Natural origin materials for bone tissue
engineering: properties, processing, and performance. In: Principles of Regenerative Medicine,
pp. 535–558. Academic Press (2019)
53. Liu, D., Nikoo, M., Boran, G., Zhou, P., Regenstein, J.M.: Collagen and gelatin. Ann. Rev.
Food Sci. Technol. 6, 527e57 (2015)
54. Croisier, F., Jérôme, C.: Chitosan-based biomaterials for tissue engineering. Eur. Polymer J.
49(4), 780–792 (2013)
55. Aravamudhan, A., Ramos, D.M., Nip, J., Harmon, M.D., James, R., Deng, M., et al.: Cellulose
and collagen derived micro-nano structured scaffolds for bone tissue engineering. J. Biomed.
Nanotechnol. 9(4), 719e31 (2013)
56. Miguel, F.B., de Almeida Barbosa Ju´nior, A., de Paula, F.L., Barreto, I.C., Goissis, G., Rosa,
F.P.: Regeneration of critical bone defects with anionic collagen matrix as scaffolds. J. Mater.
Sci. Mater. Med. 24(11), 2567e75 (2013)
57. Venkatesan, J., Vinodhini, P.A., Sudha, P.N., Kim, S.K.: Chitin and chitosan composites for
bone tissue regeneration. Adv. Food Nutr. Res. 73, 59e81 (2014)
58. Rodríguez-Vázquez, M., Vega-Ruiz, B., Ramos-Zúñiga, R., Saldaña-Koppel, D.A., Quiñones-
Olvera, L.F.: Chitosan and its potential use as a scaffold for tissue engineering in regenerative
medicine. Biomed. Res. Int. 4, 2015 (2015)
59. Aranaz, I., Gutiérrez, M.C., Ferrer, M.L., Del Monte, F.: Preparation of chitosan nanocom-
posites with a macroporous structure by unidirectional freezing and subsequent freeze-drying.
Mar. Drugs 12(11), 5619–5642 (2014)
60. Aranaz, I., Mengíbar, M., Harris, R., Paños, I., Miralles, B., Acosta, N., Galed, G., Heras, Á.:
Functional characterization of chitin and chitosan. Curr. Chem. Biol. 3(2), 203–230 (2009)
61. Jana, S., Florczyk, S.J., Leung, M., Zhang, M.: High-strength pristine porous chitosan scaffolds
for tissue engineering. J. Mater. Chem. 22(13), 6291e9 (2012)
62. Costa-Pinto, A.R., Vargel, I., Tuzlakoglu, K., Correlo, V.M., Sol, P.C., Faria, S., et al.: Influ-
ence of scaffold composition over in vitro osteogenic differentiation of hBMSCs and in vivo
inflammatory response. J. Biomater. Appl. 28(9), 1430e42 (2014)
Green Methods for the Development of Bone and Tissue … 89
63. Li, L., Li, B., Zhao, M., Ding, S., Zhou, C.: Single-step mineralization of woodpile chitosan
scaffolds with improved cell compatibility. J. Biomed. Mater. Res. B Appl. Biomater. 98B(2),
230e7 (2011)
64. Mota, J., Yu, N., Caridade, S.G., Luz, G.M., Gomes, M.E., Reis, R.L., et al.: Chitosan/bioactive
glass nanoparticle composite membranes for periodontal regeneration. Acta Biomater. 8(11),
4173e80 (2012)
65. Shavandi, A., Bekhit Ael, D., Ali, M.A., Sun, Z., Gould, M.: Development and characterization
of hydroxyapatite/beta-TCP/chitosan composites for tissue engineering applications. Mater.
Sci. Eng. C Mater. Biol. Appl. 56, 481e93 (2015)
66. Hoque, M.E., Nuge, T., Yeow, T.K., et al.: Gelatin based scaffolds for tissue engineering—a
review. Polym. Res. J. 9, 15 (2015)
67. Awad, H.A., Wickham, M.Q., Leddy, H.A., Gimble, J.M., Guilak, F.: Chondrogenic differentia-
tion of adipose-derived adult stem cells in agarose, alginate, and gelatin scaffolds. Biomaterials
25(16), 3211–3222 (2004)
68. Aldana, A.A., Abraham, G.A.: Current advances in electrospun gelatin-based scaffolds for
tissue engineering applications. Int. J. Pharm. 523(2), 441–453 (2017)
69. Correia, C., Bhumiratana, S., Yan, L.-P., Oliveira, A.L., Gimble, J.M., Rockwood, D., et al.:
Development of silk-based scaffolds for tissue engineering of bone from human adipose-derived
stem cells. Acta Biomater. 8(7), 2483e92 (2012)
70. Huang, X., Bai, S., Lu, Q., Liu, X., Liu, S., Zhu, H.: Osteoinductive-nanoscaled silk/HA
composite scaffolds for bone tissue engineering application. J. Biomed. Mater. Res. Part B
Appl. Biomater. 103(7), 1402e14 (2015)
71. Yang, J., Yang, X., Wang, L., Zhang, W., Yu, W., Wang, N., Peng, B., Zheng, W., Yang, G.,
Jiang, X.: Biomimetic nanofibers can construct effective tissue-engineered intervertebral discs
for therapeutic implantation. Nanoscale 9(35), 13095–13103 (2017)
72. Alca´zar-Alay, S.C., Meireles, M.A.A.: Physicochemical properties, modifications and appli-
cations of starches from different botanical sources. Food Sci. Technol. (Campinas) 35, 215e36
(2015)
73. Balmayor, E.R., Baran, T.E., Unger, M., Marques, A.P., Azevedo, H.S., Reis, R.L.: Pres-
ence of starch enhances in vitro biodegradation and biocompatibility of a gentamicin delivery
formulation. J. Biomed. Mater. Res. B Appl. Biomater. 103(8), 1610e20 (2015)
74. Requicha, J.F., Moura, T., Leonor, I.B., Martins, T., Munoz, F., Reis, R.L., et al.: Evaluation
of a starch-based double layer scaffold for bone regeneration in a rat model. J. Orthop. Res.
32(7), 904e9 (2014)
75. Jahangirian, H., Lemraski, E.G., Rafiee-Moghaddam, R., Webster, T.J.: A review of using green
chemistry methods for biomaterials in tissue engineering. Int. J. Nanomed. 13, 5953 (2018)
76. De Vrieze, S., Van Camp, T., Nelvig, A., Hagström, B., Westbroek, P., De Clerck, K.: The
effect of temperature and humidity on electrospinning. J. Mater. Sci. 44, 1357–1362 (2009)
77. Malafaya, P.B., Silva, G.A., Reis, R.L.: Natural–origin polymers as carriers and scaffolds
for biomolecules and cell delivery in tissue engineering applications. Adv. Drug Deliv. Rev.
59(4–5), 207–233 (2007)
78. Abbasian, M., Massoumi, B., Mohammad-Rezaei, R., Samadian, H., Jaymand, M.: Scaffolding
polymeric biomaterials: Are naturally occurring biological macromolecules more appropriate
for tissue engineering? Int. J. Biol. Macromol. 1(134), 673–694 (2019)
79. Sharmila, G., Fathima, M.F., Haries, S., Geetha, S., Kumar, N.M., Muthukumaran, C.: Green
synthesis, characterization and antibacterial efficacy of palladium nanoparticles synthesized
using Filicium decipiens leaf extract. J. Mol. Struct. 15(1138), 35–40 (2017)
80. Edison, T.N., Lee, Y.R., Sethuraman, M.G.: Green synthesis of silver nanoparticles using
Terminalia cuneata and its catalytic action in reduction of direct yellow-12 dye. Spectrochim.
Acta Part A Mol. Biomol. Spectrosc. 5(161), 122–129 (2016)
81. Rostami-Vartooni, A., Nasrollahzadeh, M., Alizadeh, M.: Green synthesis of seashell supported
silver nanoparticles using Bunium persicum seeds extract: application of the particles for
catalytic reduction of organic dyes. J. Colloid Interface Sci. 15(470), 268–275 (2016)
90 A. Hazra et al.
82. Wei, Y., Fang, Z., Zheng, L., Tsang, E.P.: Biosynthesized iron nanoparticles in aqueous extracts
of Eichhornia crassipes and its mechanism in the hexavalent chromium removal. Appl. Surf.
Sci. 31(399), 322–329 (2017)
83. Naseem, T., Farrukh, M.A.: Antibacterial activity of green synthesis of iron nanoparticles using
Lawsonia inermis and Gardenia jasminoides leaves extract. J. Chem. 1, 2015 (2015)
84. Nasrollahzadeh, M., Sajadi, S.M., Maham, M.: Green synthesis of palladium nanoparticles
using Hippophae rhamnoides Linn leaf extract and their catalytic activity for the Suzuki-
Miyaura coupling in water. J. Mol. Catal. A: Chem. 1(396), 297–303 (2015)
85. Kargozar, S., Ramakrishna, S., Mozafari, M.: Chemistry of biomaterials: prospects. Curr.
Opinion Biomed. Eng. 1(10), 181–190 (2019)
86. Mohammadinejad, R., Maleki, H., Larraneta, E., Fajardo, A.R., Nik, A.B., Shavandi, A.,
Sheikhi, A., Ghorbanpour, M., Farokhi, M., Govindh, P., Cabane, E.: Status and future scope
of plant-based green hydrogels in biomedical engineering. Appl. Mater. Today 1(16), 213–246
(2019)
87. Ebrahiminezhad, A., Zare-Hoseinabadi, A., Berenjian, A., Ghasemi, Y.: Green synthesis and
characterization of zero-valent iron nanoparticles using stinging nettle (Urtica dioica) leaf
extract. Green Process. Synth. 6(5), 469–475 (2017)
Abstract Protein-based monomers are among the most alluring options for
achieving improved results with cutting-edge biomaterials since recent advances
in bioartificial and microbiology technologies enable the very complicated, accurate
design, and fabrication of protein-based biomaterials. These sequences are easily
enhanced with bioactive motifs that improve their functionalities, material-host inter-
actions, and basic biological requirements since their sequences are built from archi-
tectural protein-based modules. These polypeptides have gained increasing interest
for usage in biomedical activities such as cell cultures, bioengineering, separation,
and purification design is vital, as well as controlled drug administration because of
their versatility, self-assembly behavior, cued, and biocompatibility. The biopolymers
discussed in this piece are biomimetic materials and genetic algorithms created from
elastin-derived proteins. These genetically programmed polymers’ design, manufac-
turing, and characterization as well as their potential uses in regenerative medicine,
bioinformatics, targeted medication delivery, and stem cell therapy will also be
covered.
1 Introduction
Medical science, biology, chemistry, materials science, and engineering are all inter-
related fields in the study of biomaterials. It has been said that “biopolymer is utilized
to create systems that substitute an element or a function that the body performs
in a secure, dependable, cost-effective, and physiologically reasonable way.” An
E. Kaushik (B)
Information Technology and Engineering Department, Dr. Akhilesh Das Gupta Institute of
Technology & Management, New Delhi, India
e-mail: Kaushikeva0026@gmail.com
R. Kaushik
CS and Mathematics Department, Data Analytics Program, University of Illinois, Illinois, USA
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 95
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_4
96 E. Kaushik and R. Kaushik
of the mechanisms dictating the body’s systems and advances in genomic science,
allowing the use of refined methods that take inspiration from the natural world to
mimic the mechanical and biological attributes of the tumor site while ensuring secu-
rity. Depending on the purpose, biomedical technologies for muscle restoration and
medication development could be produced or natural, degradable, or not. Protein-
based galactose, which was inspired by naturally occurring proteins discovered in the
ECM components, stands out as a leading candidate for use as essential components
of cutting-edge medical technology and improved replacements in tissue regenera-
tion because their characteristics are determined by the physicochemical character-
istics and loop of constituent monomers. Genetic manipulation still offers significant
control over the component organic chemicals used to make delivery, transportation,
and skin substitutes, despite the significant advances made in the field of polyelec-
trolyte materials in terms of advancements in the effectiveness of polymeric materials
research methods and lower polydispersity’s [5]. According to sequence-structure
interactions, the length and concentration of genetically created drug carriers affect
a variety of macro, micro, and nanoscale characteristics. Protein polymers made
of elastin-like polypeptides are used as well-developed genetic engineering drug
carriers as well as personalized supports for tissue restoration. Polymers known as
elastin-like peptides (ELPs) have brief pentapeptide repetitions that mimic those in
real elastin. They are influenced biologically. The most frequent recurrence of any of
98 E. Kaushik and R. Kaushik
these amino acid residues is VPGXG, where X can be any other important ingredient
than L-proline. The term “elastin-like recombinases” (ELRs), a recent invention, is
already in use to describe genetically modified ELPs. With absolute control over
the sequence of amino acids and the possibility to include a range of functions and
bioactive sequences, ELRs can be made utilizing DNA technology. Because of their
biological and thermal properties, such as biomaterials, degradability, and behavior
that is responsive to temperatures and environment, ELRs, which are microbiologi-
cally biomaterials, present an appealing alternative for the development of innovative
biomedical devices [6, 7]. ELRs also can self-assemble sterically over a temperature
slowly referred to as the low ambient temperatures (Tt), which causes a conforma-
tional reorganization at the cellular scale and the reversible development of steric
effects. The inverse thermal transformation (ITT) of ELRs is the name given to
this characteristic. Numerous elements, such as variations in temperature, pH, light,
ionic imbalances, etc., can worsen the polymer backbone. Furthermore, because it
is controlled by the molecular size and make-up of the recombinant, the activity
that responds to stimuli can be altered. They are ideal materials for the majority of
materials technology and bioscience applications, particularly for controlled tissue
engineering and drug delivery because of their metabolic behavior, flexible mechan-
ical characteristics, heat sensitivity, and capacity for self-assembly. In this study,
we explore the application of ELRs as cutting-edge drug delivery systems to safely
deliver therapeutic drugs to targeted areas. ELRs’ diverse topologies, sequencing,
and assembly enable the creation of specialized pharmacological devices such as
glucose molecules, nanoparticles, and three-dimensional assemblages. It will also
be discussed how monomeric ELRs can be used as aqueous delivery methods for
certain peptides that act as knh or cell-penetrating sections. Such systems are suit-
able for systemic distribution since they are designed to self-assemble in response to
environmental cues. They are very beneficial in tumor tissue therapy because of their
thermoresponsive activity. ELRs can self-assemble similarly to how they can produce
nanoparticles (NPs). Thus, the usage of bases and hydrogels as a 3D framework for
drug-delivery devices as well as the applications of NPs as drug nanomaterials will
be discussed [8]. Utilization of NPs as drug solid lipid nanoparticles when deliv-
ering therapeutic approaches, proteins, or signaling molecules to the skin’s surface,
or those arrangements that can internalize the medication, bioactive constituents,
or even DNA to maximize absorption and persistence effects. Problems with inad-
equate drug absorption loading can be solved by building macroscopic scaffolds
using chemical and physical ELR chain bridging, which also enables controllable
localization of the reactive drug release region and rate.
Tissue engineered and tissue regeneration are very beneficial methods for the preser-
vation, repair, or enhancement of functional body materials or complete systems
after they have exhausted their viability. The development of support systems that
Genetically Induced Biomaterial Advances in Medical Sciences 99
are “allowed to exist” when applied directly to injured tissues but are unable to
recreate on their own has progressed in recent years in the field of nanomaterials
for tissue regeneration. Systems that can interact with the organism and reproduce
biological environments are being created using contemporary technology. To build
functional tissues, these diverse fields of nanomaterials research are progressively
combining the use of cutting-edge scaffolds, the right cells, and bioactive chemi-
cals [9, 10]. For the construction of a functional scaffold, a few components have
been identified as being crucial, including the following: Factors include the needed
three-dimensional graphene, its porosity, its unique material performance, its water
sorption and permeable, epithelial connection via chemical signals, its hydrophilicity
and permeation, and its length of residence before host tissue remodeling.
Elastin-like scaffolds have been created with the goal to mimic some of the char-
acteristics of the ECM, including the architectural surface features, appropriate firm-
ness and porosity, improvement of ligand binding and viability, sensitivity to cell
signaling, processability, and cued to variations in their environment, among others.
The skeletal system, the vascular system, as well as the rejuvenation of soft tissues
are among the areas of the body where elastin has been designed to obtain specialized
support. These methods are covered in this study. The following discussion examines
a few recent cases.
a vital part of the coagulation system required for platelet adherence to collagen,
was detected using immunohistochemistry testing. Additionally, the immunological
response brought on by post-ICH activated microglia was dramatically diminished
by the RGD-containing ELR therapy, as well as vWF overexpression.
These substances constitute a significant portion of the bone and ligaments that
make up the bones and joints, of the body’s skeleton. Upon injury or in response
to ongoing modifying stimuli, the mature bone does have the innate capacity to
repair. Despite this ability to regenerate, some conditions or diseases that affect
how efficiently the body can heal itself, such as inflammation brought on by the
severity of the injury or oncogenic or pathologic bone problems, may impede the
process. Tissue-engineered supports are necessary for these circumstances because
of their osteogenic, osteoinduction, and osteogenic components, which provide struc-
tural functioning and promote bone healing. Due to their amphiphilic characteristics,
which enable them to attach to thermo-responsive hydrogels in physiological circum-
stances, two distinct ELRs have been produced. The domains from self-aware fibrin
typically contain the Arg-Gly-Asp (RGD) themes in addition to the osteoblastic and
osteogenic bone technologist and technology protein-2 (BMP-2) motif. Elastase-
sensitive action scenes are included in the segments of these ELRs to investigate the
rate of degradation of the ELR molecules. While the RGD sequencing helped host
cells adhere to the substrate and increase cell survival, the BMP-2 component encour-
aged osteoblast development. The deterioration of the artificial scaffold’s organic
composition occurred concurrently with bone regeneration, which was used in this
work to show how host bone remodeling affects how rapidly the scaffold dissolves.
In vivo experiments on female Zealand white bunnies revealed hematologic marrow
and fibroblast layer infiltration and multiplication, suggesting a large potential for
bone graft recovery and outstanding biocompatibility. The V40C2 ELR block, a
pentapeptide made of valine and disulfide, was previously combined with human
recombinant BMP-2 to increase the osteoblastic and osteoinductive capabilities of
the ELR scaffold. At 37 °C, this ELR block is identified [15, 16]. The main goal
was to maintain the bioactivity of BMP-2 in the fusion protein. It was demonstrated
that despite the protein’s diminished biocompatibility, however, neither ELR gene
product nor the commercially available pure BMP-2 lost any of their cytocompat-
ibility. BMP-V40C2 showed stronger calcium deposition than the control during a
21-day treatment regimen, as well as increased mRNA concentrations of the tran-
scription factors Osterix (OSX), which is only expressed in osteoblasts. The produc-
tion of artificial bone tissue utilizing hydrogel hydroxyapatite (Si-HA) scaffolding
and ELRs is an additional intriguing method. Due to its osteoblasts, osteogenic, and
accessibility, which encourage bone development and osseointegration, HA—a vital
element of vertebrate bone and teeth—is widely utilized in orthopedic, dental, and
cranial operations. The 3D degradable scaffolds employed in this investigation were
102 E. Kaushik and R. Kaushik
coated with two bioactive ELRs that were produced and analyzed. It has been demon-
strated that these ELRs mediate biochemical mechanisms at the interface of minerals
and have a strong affinity for calcium phosphorus, much like HA. The production
of the osteoblast markers OC and ALP, however, showed that the Si-HA-SNA15-
containing scaffold was particularly effective at directing the BMSCs toward the
osteoblastic lineage when osteoblast differentiation was assessed [17]. All studied
scaffolds allowed cells to colonize and proliferate, but the functionalized ELR gener-
ated a biochemical pathway that markedly enhanced the scaffolds’ ability to stimulate
osteoconduction. Unlike bone, some tissues, like enamel, cannot regenerate. An ELR,
a tiny protein that stimulates enamel remineralization, contains statherin. In contrast
to the untreated substrates, the resultant STNA15-ELR formed mineral platelets after
8 days of growth in a mineralization solution at 37 °C. ELRs may therefore be crucial
for preserving the mineral’s stability. On day 8, SEM pictures verified the presence
of a sodium fluoride and fluorapatite combination [18]. Fluorapatite was also visible
in TEM images. This exploratory study might lead to the creation of a useful enamel
biomineralization substance. One of the most challenging academic problems has
long been developing the finest biomechanically feasible skeletal scaffolding for
tissue engineering. To produce a hydrogel with sufficient wear resistance over time,
cysteines and the domain peptide were included in this case. After 21 days in culture,
the human mesenchymal stem cells (hMSCs) on the biomaterials appeared to still
be alive, proving that contraction rather than erosion was the cause of the loss of
volume.
These hMSCs also had higher levels of osteogenesis markers such as osteopontin
and osteocalcin than hMSCs that were placed on glass slides. Chondrocytes were
put to the gel after 28 days of growth and displayed 86% more vitality, respectable
architecture, and elevated activity of alkaline phosphatase than the control [19, 20].
As we mentioned at the beginning, one of the most intriguing applications of elastin-
like particles is the creation of scaffolding that mimics biological structures.
The final design of a 3D spheroid using an ELR chemically coupled to
polyethyleneimine (PEI) was compared with that of a 2D monolayer culture, even
though 2D cultures cannot replicate the physiological 3D environment. To create
3D microspheres as a 3D model of culture, these scientists plated hybrid fat-derived
stem cells (hASCs) on normal styrene coated with the ions with a positive charge
ELR-PEI. Viability tests were performed after both had been grown in differentiation
media, and they revealed the same number of cells as for the 2D layer. Scaffolds or
scaffolding for glycosidic bonds are also produced as composites to regulate bone
regeneration because of the advantages mentioned above. The mechanical proper-
ties of the scaffolds can be improved by the stiffness and flexibility of ELRs [21].
Four distinct composites with various ELR/collagen ratios were examined in this
experiment. The composite with the highest ELR/collagen content (18 mg/mL) had
the highest levels of ALP activity, OCN transcription, and stiffer matrix even though
all these composites contained hASCs that were proliferating and differentiating.
Composites constructed entirely of chondro had substantially less matrix and took
Genetically Induced Biomaterial Advances in Medical Sciences 103
longer to mature than those with lower ELR/collagen ratios. Currently, one of the
most prevalent causes of discomfort and incapacity in the elderly is the degeneration
of joint cartilage.
Due to collagen’s perivascular nature, it is challenging to identify articular struc-
tures that are harmed because of trauma or aging joints because collagen has a limited
capacity to rebuild [22, 23].
Like this, hydrogels are intriguing scaffolds because of their mechanical prop-
erties and ease of manipulation, especially those that can self-gelate under physi-
ological conditions and may precisely integrate a device in situ. Available internal
and external ELR-HA hydrogel foundations have been created to compare scaffolds
with varying component ratios. The stiffness of the supports remained consistent for
all the ratios that were taken into consideration. When HA concentration in chon-
drocytes increased, there was a dose-dependent rise in the transcription of cartilage
marker genes like aggrecan, SOX9, and type 2 collagen. The importance of these
chondrogenic indicators must be stressed because chondrocytes are the only cells
present in the knees that have the charge of controlling the extracellular matrix. The
hydrogel’s chondrocytes also showed increasing levels of type 1 collagen, a marker
of an undesirable fibrous connective tissue, and metallopeptidase-13, a marker of
matrix disintegration and remodeling, when the concentration of HA was reduced
[24]. Moreover, chondrogenic multiplication was boosted in biomaterials with lower
HA concentrations, whereas sGAG (sulfated glycosaminoglycan) deposition was
promoted in hydrogels with higher HA contents. This system (5% HAhydrogel) was
shown to be the best scaffold since it had the highest peak of cartilage indicators,
the largest sGAG deposit, the least amount of cellular proliferation, and the fewest
of type 1 collagen and fibrous tissue phenotypic development.
The various ELR architectures are examined in this chapter as the building blocks
for numerous drug delivery systems. In these subchapters, devices spanning between
monomeric ELRs to amphoteric ELRs that can create nanoparticles have been
discussed. The chapter is concluded with macroscale ELRs for the delivery of drugs
in the forms of depots or hydrogels.
with a pH-responsive component that can also be stabilized with Zn 2 + ions [31,
32]. When compared to ELRBCs made of [VG7A8]-64/[V]-120, a non-pH-sensitive
ELR block copolymer, in vivo penetration, and distribution in tumors. The solid
tumor tissues usually have extracellular pH ranges of 6.2–6.9 as opposed to the
normal tissue’s circulatory pH range of 7.2–7.4, therefore the ionic intensity ELR-
based polyamide microspheres break down at this lower pH. The 60 pentapeptide
repeats in the diblock, which include the hydroxyl group as a guest residue, form a
hydrophobic block, whereas the 60 core protein residues, which contain glutamine
and glycine in a 1:1 ratio, form a hydrophilic block. Then, Silaffin R5 and the
hydrophilic region of the ELR were genetically connected. When phosphorus ions
are present in phosphate-buffered saline, the ELR-R5 is crosslinked, resulting in the
formation of silica nanoparticles. Afterward, silicic acid is added to the solution of
the made-up templates to make the silica polycondensation. Many researchers have
used the strain-promoted azide-alkyne cycloaddition (SPAAC) method to enhance
the formation procedure by connecting the chain at the particle’s outer shell. To
ensure peripheral crosslinking, a clearly defined ELR was created by joining the more
hydrophobic block to a lysine-rich area. The arginine amine groups were converted
into azide units using the diazo-transfer method, and the NPs were subsequently stabi-
lized through exergonic crosslinking using the chemical bis-cyclooctyne. Additional
approaches involve employing membrane transporters made of polymer nanoparti-
cles and genetically altered drug receptors after the prescription has been coupled to
the receptor [33]. The basis of this innovative encapsulation method, developed by
MacKay and colleagues, is the high selectivity between a given molecular medication
and its protein sequence targets attached to the corona of protein polymer nanopar-
ticles. The drug is administered gradually once it has been securely attached to the
carrier. Rapamycin (Rapa), an immunosuppressive macrolide antibiotic, is applied
over stents to prevent organ transplant rejection. Rapa’s anti-proliferative qualities,
constrained dispersion, low oral absorption, and quick systemic clearance made it
the ideal candidate for these studies. 23 days after the initial dose, the rodents lost
over 15% of their entire body mass, demonstrating the high toxicity of free Rapa.
The drug for the prostate xenograft mouse model showed superior antitumor action,
showing notably lower cytotoxicity in contrast to free Rapa, without exhibiting any
symptoms of behavioral issues or body weight loss. Eventually, diverse proteins and
small molecules will be encapsulated, targeted, and released using this technique.
More and more sophisticated devices have been developed that use ELR-based tech-
nologies to recognize complex NPs that can transport a variety of small molecules
to improve regulated medication delivery systems. One of the best ways to incor-
porate bits into macro material is by self-assembly, which can transform nanoscale
Genetically Induced Biomaterial Advances in Medical Sciences 107
into devices with unique capabilities. Coworkers created the H6-ELR-CP triblock
polymer in this situation, which consists of a polypeptide block with thermal respon-
siveness, a hex histidine block with the potential to bind ionic species, and the capsid
peptides of the cowpea erythema mottle virus, for whom the self-assembly may
be pH-dependent [34, 35]. New nanocarriers built on recombinamers that mimic
elastin require inventive ways. Active targeting abilities can also be added thanks
to the customization of these ELRs with fibroblast ligands. To facilitate active cell-
targeting, ELR with a polymeric acid tail (ELR-D) and then added epidermal growth
factor EGF (ELR-D-E). The resultant nanoparticles have been shown to have a
specific biological interaction, and in vitro results have been encouraging using A549
cells, a normal lung cancer epithelium line that pushes the limits of the EGF recep-
tors. It looked at how to distribute suicide genes using a plasmid that contains the
Rna for the toxin PAP-S.
With excellent selectivity for MCF-7 cancer cell lines as opposed to a MUC-1-
negative tumor line, this technique effectively transported the plasmid into tumor
cells while protecting normal human cells. This aptamer-ELR vector’s ability to
transfect cells, as demonstrated by micropinocytosis uptake, opens the door for the
use of ELRs in suicidal genome therapy to cure cancer and further opens the door
for their use in the biomedical sector.
One form of treatment where a prolonged local treatment of the tumor is required
for success is anticancer therapy. Therapeutic drugs must navigate several biological
barriers on their way to the tumor to be successful, and systemic medicines usually
have high rates of toxicity as a side effect. Thank goodness, localized anticancer
therapy exposure and reduced drug storage inside the tumor can be achieved through
intertumoral injection, minimizing negative side effects. An enticing alternative to
local delivery is the creation of a biocompatible substance that may be injected as a
medication delivery deposit at room temperature. The thermosensitive ELR sequence
in this instance carried a biopolymer into a tumor when soluble, and because of air
temp coacervation, produced a depot in situ [36]. Intertumoral radiation therapy has
a very good response rate in solid tumors, especially aggressive prostate cancers
(brachytherapy). By introducing radioactive “seeds” into or close to the tumor, this
technique gives the tumor a significant dosage of radiation while limiting unneeded
exposure to the neighboring healthy tissues. Despite the method’s overall benefits,
existing seed implantations are permanent and can only be used in the treatment of
low- or intermediate-grade malignancies. They are ineffective for treating high-grade
tumors. The development of polymeric devices that function in conjunction with
irradiation has undergone several attempts. It is preferable to avoid the problems with
conventional brachytherapy. Avoiding the issues with traditional brachytherapy is
preferred-based systems that have recently been taken into consideration for topically
applied delivery, particularly in the context of creating new capabilities considering
108 E. Kaushik and R. Kaushik
2.6 Hydrogels
[1] Frappier et al. [40]; Atomically small material alterations enable a variety of
nanoelectronics devices, such as resonant tunneling diodes (RTDs), quantum well-
integrating photodetectors (QWIPs), subatomic good lasers, and heterojunction junc-
tion effect transistors (HFETs). For the development and creation of such devices,
a fundamental knowledge of electron propagation in such wavelengths is required.
Based on a basic nonequilibrium electron transport theory, NEMO is a flexible tool
for designing and studying quantum devices. EMO was linked with the parallel
processing evolutionary algorithm application PGAPACK to modify the properties
of the structural materials to match a certain set of experimental data. A numer-
ical experiment is done to generate structural parameters like layer thicknesses and
doping levels to evaluate an experimental current–voltage characteristic. We find
that the basic and doping characteristics of the heNEMO model parameters closely
reflect the genetic algorithm. Synthesis is achievable with such precise agreement
between theory and technique are known.
[2] Schloss et al. [41]; In order to create new kinds of allergy vaccinations, DNA
Fragment was used in allergens research. Details on the genetic sequencing and
makeup were made available. One technique that is widely employed in the creation
of antimicrobial peptides that meet certain T- or B-cell epitopes. A new technique
for developing hypersensitive vaccinations that can provide a better and much less
replication of the epitopes is genetic engineering allergen synthesis. To show how
well these hypo-allergens reduce allergenicity, numerous stimulating skin and nasal
testing have been employed. The vaccines’ capacity to generate anti-allergen IgG
antibodies and sustain T fibroblast activity have both frequently been shown, despite
the diminished immunoglobulin E (IgE)-binding reactivity. The main hypoallergenic
have been polypeptide segments and tetrameric birch allergen structures.
[3] Olorunniji et al. [42]; Atomic size differences in the materials enable devices like
resonance tunneling diodes (RTDs), quantum well-infrared photodetectors (QWIPs),
classical good lasers, and heterojunction field effect transistors (HFETs). Such
heterostructure devices call for an in-depth knowledge of electron transport in
110 E. Kaushik and R. Kaushik
quantum states for their design and optimization. This problem is addressed by
the overall design and analysis tool for nanodevices known as the Nanoelectronics
Modeling Tool (NEMO). NEMO was used with PGAPACK, a library of parallel
processing neural network methods, to improve structural and material properties.
The quasi-band effects in the longitudinal and transverse dimensions as well as the
Wkb charge personality are included in the electron transport calculations displayed
here.
[4] Tang [43]; A popular biocompatible substance for stem-cell cartilage healing
is elastin-like polypeptides (ELPs), a class of artificial polypeptides with special
characteristics. The pentapeptide Val-Pro-Gly-Xaa-Gly makes up ELPs, and Xaa
can be any other amino acid besides Pro that passes through an inverted temperature
phase change. They can dissolve in water when it is below their transition temperature
(T t). Yet, when the temperature of water surpasses their T t, they congeal. This study
evaluates the rheological characteristics of an uncross-linked ELP below and above
its T t as well as the effectiveness of ELP in triggering chondrogenesis in vitro.
Recombinant DNA techniques were used to create an ELP with a T t of 35 °C.
4 Application
A natural polymeric protein called silk sericin is derived from the Bombyx mori silk-
worm. Sericin can be retrieved and used in other ways during the many processes used
to make raw silk and textiles. Additionally, sericin recovery lessens the impact of silk
production on the environment. Sericin protein has advantages due to its characteris-
tics. The protein is UV-resistant, antimicrobial, and resistant to oxidation [43]. It also
readily collects and releases moisture. To create products with better qualities, sericin
proteins can be polymerized, cross-linked, and combined with other biomolecules,
notably synthetic polymers. Additionally, the protein can be used to enhance or
coat both natural and synthetic fibers, fabrics, and products. Bioplastics, biomedical
112 E. Kaushik and R. Kaushik
Investigated were the effects of o2 radio wave glow discharge (rfGD) on the interfaces
of various starch-based biomaterials (SBB) as well as the effects of protein adsorp-
tion on controlling bone-cell activity. Both simple and complicated protein systems
used fibronectin, vitronectin, and bovine serum albumin. Surfaces coated with RfGD
demonstrated greater hydrophilicity and interface energy as compared to untreated
SBB. In cornstarch-based biodegradable polymeric blends, polycaprolactone (SPCL;
30/70 weight percent), poly (ethylene alcohol (SEVA-C; 50/50 weight percent),
and methylcellulose (SCA; 50/50 weight percent) were investigated. The maximum
degree of change was seen in SCA and SCA reinforced with 10% fluorapatite (HA)
after rfGD treatment [44]. On SCA, it was discovered that cell adhesion and multipli-
cation were increased compared to untreated surfaces, and the plasma modification
had no effect on SCA + 10%HA. BSA, FN, and VN single solutions enhanced cell
adherence to SCA surfaces, and a similar impact was observed for ternary systems.
Moreover, when compared to the untreated surfaces, SEVA-C treated with blood
shows better adhesion and proliferation. MG63 cell multiplication was clearly aided
Genetically Induced Biomaterial Advances in Medical Sciences 113
5 Case Study
One of the important features that distinguish the genetic algorithm from other algo-
rithms like evolutionary algorithms is that its main search operators are crossover and
mutation. The two main genetic operators utilized in genetic algorithms are crossover
and mutation. The objective of the current study was to investigate the impacts of
genetic algorithm operators such as mutation and crossover (P c & P), population
size (n), and the number of iterations (I) used to forecast the biological material
extruder’s lowest hardness (N). The second-order polynomial regression equation
developed for the extrudate characteristic hardness in terms of independent factors
such as temperature, screw speed, the fish proportion of the feed, and feed moisture
content was employed as the objective function in the GA analysis [47]. A simple
genetic algorithm (SGA) based on crossover and mutation operators was used in the
current study. A rank-based fitness selection algorithm was built into a C language
program for an SGA. The maximum number of iterations and inhabitants was set to
100. It was discovered that a medium community of 50, iterations of the algorithm,
and the lowest hardness values were feasible. According to the Pareto charts, P c
Genetically Induced Biomaterial Advances in Medical Sciences 115
was found to have a greater impact when the population was below 50, while P m
was crucial when the population was below ten. The threshold values for a crossover
chance of less than 50% and a mutation probability of less than 5%. Physical state
of biomaterial for myocardium tissue engineering is shown in Table 3.
One of the biggest causes of death in the globe is cardiovascular disease. After a
myocardial infarction, cardiac tissue containing deceased or damaged cardiac cells
downstream of the occluded channel does not recover because adult cardiac cells
have a restricted capacity for proliferating. The heart becomes feeble when non-
functional fibrotic scar tissue takes the place of the original cardiac tissue [48].
Researchers are looking into the possibility of stem cells to regenerate damaged
cardiac tissue because host cardiac cells have a restricted capacity for proliferation.
This project has made tremendous progress. Currently, there is no agreement on the
ideal stem cell type, matrix material, or microenvironmental stimuli for cells. This
article provides a summary of the various biofunctional compounds and bioactive
matrices that, when combined with stem cells, have demonstrated promise for the
renewal and reinforcement of cardiac tissue. Engineered biomaterials are also used
in cardiac tissue engineering, which uses stem cells and biomaterial scaffolds to
generate tissues in vitro that can be used for drug testing or eventual implantation. This
study highlights the advantages of repairing injured myocardium with biomaterials
and stem cells and gives a quick overview of the characteristics of these polymers
that make them such useful instruments in the field.
116 E. Kaushik and R. Kaushik
6 Challenges
is one cell type for every 100,000 cells), and diminished developmental capacity with
aging for some stem cell types. The control of stem cell development is a potential
use of mechanistically customized bioactive materials that may also serve as the
rationale for stem cell-based therapy.
• Bio Ceramics: Control of Infection
Due to bacterial adherence to biomaterials, which causes biomaterial-centered infec-
tion and insufficient tissue integration, several medical devices have a short lifespan.
Globally, the problem of the spread of bacteria resistant to common antibiotics is
getting worse. The increased incidence of chronic foot and limb lesions, which
frequently require amputation, is a substantial barrier. The bacteria and inflammation
that lead to cellular dedifferentiation must be controlled locally. AgBG concentra-
tions of 0.05−0.20 mg per microliter of culture medium suppress the development
of these bacteria. AgBG has a quick antibacterial effect in addition to being bacterio-
static. At AgBG concentrations of 10 mg ml1, a full bactericidal action was evoked
within the first hours of incubation. 45S5 Both Bioglass and BG had no impact on
the viability or proliferation of bacteria. Only the leaching of Ag+ ions from the glass
matrix is responsible for AgBG’s antibacterial activity [52]. Analytical measurements
exclude any involvement of pH changes, ionic strength changes, or the dissolution
of other ionic species from the biomaterials in AgBG-mediated bacterial death. The
patterns of Ag+ dissolution from AgBG in the addition and exclusion of bacteria
lend credence to the notion that the microbes are storing silver. According to XRD
patterns, FTIR spectra, and ICP data, the Ag-doped gel glass exhibits the same bioac-
tive behavior as bioactive gel glasses that have Approval from the FDA for use in
bone repair.
• Predicted in vitro testing of the biomaterials’ and nanoparticles’ toxicity and
biocompatibility
For both ethical and budgetary reasons, society is worried about the existing reliance
on in vivo testing on animals to assess the safety of novel biomaterials, TE structures,
and nanoparticles. The development of reliable and cost-effective predictive in vitro
experiments based on human cells is a major challenge for the twenty-first century.
There are a variety of issues with in vitro research that need to be resolved to ensure
relevance to prospective in vivo applications [53]. A mature cell phenotypic in culture
must, first and foremost, be representative of the same form of human cells in vivo.
Cell culture research currently frequently uses eternal cell lines, which are unable
to express the intricate protein arrays necessary for mature phenotypes. Third, while
testing, the mature cell phenotype in the cultured cells must be preserved. The drug
being tested might not be toxic enough to solely kill the cells; instead, it could cause
de-differentiation and impair the tissues’ ability to repair. These demand monitoring
of cell morphologies, preferably in situ. Fourth, the in vitro testing should contain
details regarding the molecular biological alterations that the chemical causes in
the cells. Fifth, the in vitro tests ought to be able to do statistical analysis to discern
between minute alterations in the cell population. Sixth, it’s important to take pricing
and usability into account.
118 E. Kaushik and R. Kaushik
7 Future Scope
8 Conclusion
It has also been studied how to distribute therapeutic drugs safely and effectively
to certain places, as well as a variety of ELR-based architectures and their prospec-
tive application as superior drug delivery systems. Whether utilized with transgenic
biopharma in a liquid delivery mechanism or as a pharmacokinetics enhancer, either
via chemical linkage or by creating fusion proteins of the drug, monomeric elastin-
like proteins that are implicated have been explored. These polypeptides can increase
the half-life of medications when utilized as therapeutic systems. Genetic engineering
also makes it possible for better cellular reception of nanostructures with therapeutic
functions in damaged tissues by inserting targeting peptides like knh or fibroblast
domains. Additionally, the right amino acid selection enables the existence of reactive
groups that are beneficial for the covalent connections formed during the conjugation
of molecules with proven therapeutic efficacy, with drugs and peptides being among
the most popular choices. ELRs can be designed to identify in response to external
stimuli, making them suited for systemic and local delivery of therapeutic drugs to
the injured tissue. ELRs that are available at body temperature after thermorespon-
sive transition have been demonstrated to accumulate in cancerous tissue because of
local overheating. In fact, by utilizing the porosity and perfusion of the vasculature,
therapeutic hyperthermia has improved the local distribution of ELR prodrugs into
tumor tissues. According to the diverse device structures, the biological applications
of NPs—in particular, ELR-based nanoparticles having signaling pathways that can
interact with organisms on their surfaces—have been studied. When used to heal
brain injury, and persistent skin damage, or to promote bone regeneration, several
of these have shown positive results. Lacritin nanomaterials are just one type of
NP that have been employed as therapeutic agents and are coupled to proteins that
really are visible on their surface. They support the integrity of the epithelium and
Genetically Induced Biomaterial Advances in Medical Sciences 119
corneal tissue healing. Last but just not least, studies on 3D ELR have been conducted.
These include depots, hydrogels, and macromolecular carriers. Because they provide
highly localized treatment, these are particularly promising for anticancer therapy. To
create injectable depots for venous distribution, ELRs can collaborate. This enhances
therapy options, localized anticancer medicine exposure, and drug stores in the
tumor. As macromolecular carriers, fusion-based ELR depots can carry curcumin
or glucagon-like peptide-1, which represents a substantial development in the treat-
ment of t2dm and pro therapy, etc. [55, 56]. The benefit of utilizing biomaterials that
have been chemically and structurally copolymerized is their ability to be positioned
close to the area of interest and disperse embedded drugs over a long period of time
locally, minimizing any negative effects. Because they enable the regulated release
of several bioactive compounds like hormones, antibodies, signaling pathways, and
other therapeutic biomolecules with possible biological uses, hydrogels containing
a domain sensitive to proteolytic disintegration have drawn particular interest. In
addition, the use of ELR-based 3D, notably hydrogels, has been investigated for
tissue-engineered with an emphasis on skeletal regeneration. Numerous strategies
have been examined, including the utilization of physiological conditions that repli-
cate the affected body part, such as the musculoskeletal or circulatory systems, or
the use of special scaffolds created from amphiphilic ELRs to restore soft tissues.
Because they can introduce various bioactive patterns into the specifically provided
sequencing or because they possess the appropriate cells for the regeneration of func-
tional tissues, these creative and biomimetic scaffolds are very significant for tissue
engineering. Studies have demonstrated that ELRs are excellent candidates for tissue
engineering because of the synergy that occurs when they are combined with bicom-
ponent scaffolds like ELR-collagen, ELR-silicon replacement perovskite, or ELR-
hyaluronic acid hydrogel scaffolds. For bone reconstructive surgery, new methods
based on precisely defined combinations of osteogenic materials, osteoinductive
chemicals, and/or osteoblastic cells are highly desired. The objective is to effectively
reproduce the original tissue without going through the bone harvesting process [57,
58]. To incorporate all the aforementioned qualities, a carefully controlled hybrid
material must yet be developed. Further developments require cooperation between
experts from a variety of disciplines, including stem cell biology genetics, diagnostic
devices, material sciences, medicine release, synthetic biology, and surgery.
9 Discussion
Materials that have potential for use in biomedical and therapeutic applications are
referred to as biomaterials [59]. They could be any or all the following, depending
on the application:
• Bioactive: They actively interact with bodily systems. This is mostly considered
in applications for tissue repair.
120 E. Kaushik and R. Kaushik
• Bioinert: This phrase implies that the substance in question does not affect or
upset the body system.
• Bioresolvable/Biodegradable: To prevent environmental pollution, the material
used for clinical or medicinal usage must be easily decomposed after use.
• Biocompatible: The material must be appropriate for its use, which is the most
crucial requirement of all [60, 60]. The material that is best for a certain application
will have mechanical characteristics that are like those of the real body component
that is being used or replaced.
• Mechanical Properties: The mechanical qualities of a biomaterial reveal the
molecule’s biocompatibility with the body system by describing the material’s
toughness, strength, and ductility.
References
1. Taberna, T.B., Zandstra, P.W.: Hematopoietic stem/progenitor cells and engineering: Producing
T cell competent blood progenitor cells from pluripotent stem cells in scalable dynamic
suspension culture. Cytotherapy, 25(6). https://doi.org/10.1016/s1465-3249(23)00385-7
2. Acknowledgment to the reviewers of Applied Microbiology in 2022.: Appl. Microbiol., 3(1),
76–78 (2023). https://doi.org/10.3390/applmicrobiol3010006
3. Ahearne, M.: Mechanical testing of Hydrogels. Mech. Hydrogels, 73–90 (2022). https://doi.
org/10.1016/b978-0-08-102862-9.00003-8
4. Zhang, Q.: Multi-scale instrumented indentation of Hydrogels. Mech. Hydrogels, 91–118
(2022). https://doi.org/10.1016/b978-0-08-102862-9.00001-4
5. Gao, M., Xie, X., Huang, T., Zhang, N., Wang, Y.: Glutaraldehyde-assisted crosslinking in
regenerated cellulose films toward high dielectric and mechanical properties. Cellulose 29(15),
8177–8194 (2022). https://doi.org/10.1007/s10570-022-04785-2
6. Singh, R., Malhotra, A., Bansal, R.: Synthetic cytotoxic drugs as cancer chemotherapeutic
agents. Med. Chem. Chemother. Agents, 499–537 (2023). https://doi.org/10.1016/b978-0-323-
90575-6.00010-7
7. Medicinal Chemistry of Chemotherapeutic Agents. (2023). https://doi.org/10.1016/c2020-0-
03892-2
8. Importance of human anatomy and physiology in biomaterial science.: Biomater. Sci., 1–44
(2022). https://doi.org/10.1515/9783110655377-001
9. Peutzfeldt, A., Dahl, J.: Editorial announcement. Biomater. Investig. Dent., 9(1), 110–110
(2022). https://doi.org/10.1080/26415275.2022.2152939
10. Paulussen, F.M., Grossmann, T.N.: Peptide-based covalent inhibitors of protein–protein
interactions. J. Pept. Sci., 29(1), (2022). https://doi.org/10.1002/psc.3457
11. Numata, K.: Biopolymer material and composite. Biopolym. Sci. Proteins Pept., 205–246
(2021). https://doi.org/10.1016/b978-0-12-820555-6.00009-4
12. Processing sericin.: Sustainable Use of Byproducts from Silk Processing, pp. 39–99. https://
doi.org/10.1002/9783527828760.ch2
13. Peutzfeldt, A., Dahl, J.E.: Editorial. Biomater. Investig. Dent., 8(1), 180–180 (2021). https://
doi.org/10.1080/26415275.2021.2010366
14. Nanoparticle-functionalized poly-methyl methacrylate bone cement for sustained chemother-
apeutic drug delivery.: OrthoMedia. (2021). https://doi.org/10.1302/3114-210284
15. González de Torre, I., González-Pérez, M., Alonso, M., Rodríguez-Cabello, J.C.: Elastin-like
recombinamers (elrs) for biomedical applications. Soft Matter Biomed. Appl., 205–235 (2021).
https://doi.org/10.1039/9781839161124-00205
Genetically Induced Biomaterial Advances in Medical Sciences 121
16. Miguel, G.A., Álvarez-López, C.: Extraction and antioxidant activity of sericin, a protein from
silk. Braz. J. Food Technol., 23. https://doi.org/10.1590/1981-6723.05819
17. Pharmacodynamics.: Definitions. (2020c). https://doi.org/10.32388/yfuyn4
18. Wei, G.: Characterization techniques of protein and peptide nanofibers: Self-assembly kinetics.
Artif. Protein Pept. Nanofibers, 99–118 (2020). https://doi.org/10.1016/b978-0-08-102850-6.
00005-x
19. Krishnan, U.M.: Protein and peptide nanostructures for drug and Gene Delivery. Artif. Protein
Pept. Nanofibers, 279–325 (2020). https://doi.org/10.1016/b978-0-08-102850-6.00013-9
20. Brodin, P.: Cell microbiology interview. Cell. Microbiol., 23(3), (2020). https://doi.org/10.
1111/cmi.13288
21. Chemotherapeutic perfusion.: Definitions. (2020). https://doi.org/10.32388/x77s0i
22. Chemotherapeutic topical agent.: Definitions. (2020b). https://doi.org/10.32388/hz6rdt
23. Peutzfeldt, A., Dahl, J.E.: Editorial. Biomater. Investig. Dent., 7(1), 158–158 (2020). https://
doi.org/10.1080/26415275.2020.1831296
24. Petersen, H.S.: Non-specific protein adsorption on polyetheretherketones -challenges for appli-
cation as biomaterial. Mod. Approaches Mater. Sci., 1(4). https://doi.org/10.32474/mams.2019.
01.000119
25. Lary, J.M.: Hyperthermia and Teratogenicity. Hyperth. Cancer Treat., 107–126 (2019). https://
doi.org/10.1201/9780429266539-6
26. Horbett, T.A.: Protein adsorption to Hydrogels. Hydrogels Med. Pharm., 127–171 (2019).
https://doi.org/10.1201/9780429285097-6
27. Levanič, J., Gericke, M., Heinze, T., Poljanšek, I., Oven, P.: Stable nanocellulose gels prepared
by crosslinking of surface charged cellulose nanofibrils with di- and triiodoalkanes. Cellulose,
27(4), 2053–2068 (2019). https://doi.org/10.1007/s10570-019-02947-3
28. Qiang, Y., Patel, A., Manas-Zloczower, I.: Enhancing microfibrillated cellulose reinforcing
efficiency in epoxy composites by graphene oxide crosslinking. Cellulose 27(4), 2211–2224
(2019). https://doi.org/10.1007/s10570-019-02916-w
29. Diehl, B.G., Watts, H.D., Kubicki, J.D., Regner, M.R., Ralph, J., Brown, N.R.: Correction to:
Towards lignin-protein crosslinking: Amino acid adducts of a lignin model Quinone Methide.
Cellulose 26(11), 7025–7025 (2019). https://doi.org/10.1007/s10570-019-02493-y
30. Biophysical investigations of molecular mechanisms of chemotherapeutic agents action. 1.
chemotherapeutic and antiviral agents (review).: Biophys. Bull., (42), (2019). https://doi.org/
10.26565/2075-3810-2019-42-02
31. Nutt, D.J., Nestor, L.J.: Pharmacodynamics of addictive substances. Oxf. Med. Online (2018).
https://doi.org/10.1093/med/9780198797746.003.0006
32. Tarr, G.E.: Manual methods for protein/peptide sequence analysis. Protein/Pept. Seq. Anal.:
Curr. Methodol., 35–48. https://doi.org/10.1201/9781351076081-2
33. Krutzsch, H.C.: Protein/peptide sequence analysis by mass spectrometry. Protein/Pept. Seq.
Anal.: Curr. Methodol., 161–180 (2018). https://doi.org/10.1201/9781351076081-9
34. Hayashi, R.: Enzymatic methods of protein/peptide sequencing from Carboxyterminal End.
Protein/Pept. Seq. Anal.: Curr. Methodol., 145–160 (2018). https://doi.org/10.1201/978135107
6081-8
35. Mazin, A.V., Mazina, O.M.: RAD51 and dmc1 recombinases. Mol. Life Sci., 1009–1016
(2018). https://doi.org/10.1007/978-1-4614-1531-2-67
36. Manian, A.P., Aldred, A.K., Lenninger, M., Bechtold, T.: Alkali pretreatments and crosslinking
of Lyocell Fabrics. Cellulose, 24(9), 3991–4002 (2017). https://doi.org/10.1007/s10570-017-
1384-9
37. Aranda-Espinoza, H., Adlerz, K.: The cell as an inspiration in biomaterial design. Biomater.
Mech., 103–116 (2017). https://doi.org/10.1201/9781315152585-5
38. K, S.:. Tamoxifen: Pharmacokinetics and pharmacodynamics. Open Access J. Pharm. Res.,
1(8), (2017a). https://doi.org/10.23880/oajpr-16000143
39. Shahbaz, K.: Tamoxifen: Pharmacokinetics and pharmacodynamics. Open Access J. Pharm.
Res., 1(8), (2017). https://doi.org/10.23880/oajpr-16000143
122 E. Kaushik and R. Kaushik
40. Frappier, V., Duran, M., Keating, A.E.: PixelDB: Protein-peptide complexes annotated with
structural conservation of the peptide binding mode. Protein Sci. 27(1), 276–285 (2017). https://
doi.org/10.1002/pro.3320
41. Schloss, P.D.: Preprinting Microbiology. (2017). https://doi.org/10.1101/110858
42. Olorunniji, F.J., Rosser, S.J., Marshall Stark, W.: Purification and in vitro characterization of
zinc finger recombinases. Methods Mol. Biol., 229–245 (2017). https://doi.org/10.1007/978-
1-4939-7169-5-15
43. Tang, J.C., Rudolph, S., Cepko, C.L.: Viral delivery of GFP-dependent recombinases to the
Mouse Brain. Methods Mol. Biol., 109–126 (2017). https://doi.org/10.1007/978-1-4939-716
9-5-8
44. Voziyanova, E., Anderson, R.P., Voziyanov, Y.: (2017). Dual recombinase-mediated cassette
exchange by tyrosine site-specific recombinases. Methods Mol. Biol., 53–67 (2017). https://
doi.org/10.1007/978-1-4939-7169-5-4
45. Liu, W., Tuck, L.R., Wright, J.M., Cai, Y.: Using purified tyrosine site-specific recombinases
in vitro to rapidly construct and diversify metabolic pathways. Methods Mol. Biol., 285–302
(2017). https://doi.org/10.1007/978-1-4939-7169-5_18
46. Biopolymers, 106(1), (2016). https://doi.org/10.1002/bip.v106.1
47. Vaclaw, M.C.: Linking protein-catechol-chitin interactions to the physical properties of beetle
elytral cuticle, a multicomponent biomaterial. In: 2016 International Congress of Entomology.
(2016). https://doi.org/10.1603/ice.2016.95574
48. Abdalla, M.: Important factors influencing protein crystallization. Glob. J. Biotechnol.
Biomater. Sci., 025–028 (2016). https://doi.org/10.17352/gjbbs.000008
49. Li, W., Su, X., Zhong, Q., Liu, Z., Cai, Y., Yao, J.: Influence of reaction conditions on the
self-assembly of the natural silk sericin protein. Microsc. Res. Tech., 80 (3), 298–304 (2016).
https://doi.org/10.1002/jemt.22666
50. David, K.: Advances and challenges with fibrous protein biomaterial designs. Front. Bioeng.
Biotechnol., 4. https://doi.org/10.3389/conf.fbioe.2016.01.02613
51. Chemotherapeutic.: Encycl. Nanotechnol., 524–524 (2016). https://doi.org/10.1007/978-94-
017-9780-1_100176
52. Market scenario of biomaterial-based devices.: Smart Biomater. Devices, 207–217 (2016).
https://doi.org/10.1201/9781315371559-10
53. Badylak, S.F.: In Host response to biomaterials: The impact of host response on biomaterial
selection. Elsevier/AP, Amsterdam (2015)
54. Xiang, Z., Anthony, R., Lan, W., Runge, T.: Glutaraldehyde crosslinking of Arabinoxylan
produced from corn ethanol residuals. Cellulose 23(1), 307–321 (2015). https://doi.org/10.
1007/s10570-015-0828-3
55. Donev, R.: Advances in protein chemistry and Structural Biology. Academic Press is an imprint
of Elsevier, Waltham, MA (2015)
56. Fusion protein (fused protein, chimeric protein, fused peptide, fusion peptide, hybrid protein).:
Dict. Genomics, Transcr. Proteomics, 1–1 (2015). https://doi.org/10.1002/9783527678679.dg0
4683
57. Makvilay, S., Sanongraj, W., Khamwichit, W.: Turbidity removl using silk sericin and silk
sericin powder as coagulant aid. Advanced Materials Research 931–932, 276–280 (2014).
https://doi.org/10.4028/www.scientific.net/amr.931-932.276
58. Dunn, B.: Preface. Protein Pept. Lett. 20(1), 1–1 (2013). https://doi.org/10.2174/092986613
804096883
59. Tatemastu, K.: Utilization of transgenic silkworms for recombinant protein production. J.
Biotechnol. & Biomater., s9(01), (2012). https://doi.org/10.4172/2155-952x.s9-004
60. Protein adsorption at the biomaterial–tissue interface.: An Introd. Biomater., 139–156 (2011).
https://doi.org/10.1201/b11561-11
61. Pereira, P.: Peptide and protein application in tissue repair and regeneration. Pept. Protein
Deliv., 291–311 (2011). https://doi.org/10.1016/b978-0-12-384935-9.10012-4
Genetically Induced Biomaterial Advances in Medical Sciences 123
Abstract In recent times, the concept of biomimetics has gained widespread accep-
tance across different industries. This interdisciplinary field combines principles
from biology, engineering, and chemistry to develop materials, devices, or artifi-
cial systems that imitate biological processes. In the medical field, Biomaterials
play an important part in the recovery process by restoring function and speeding
up healing. These components, which might be natural or synthetic, are utilized to
preserve, enhance, or replace damaged tissues or biological processes. Additionally,
biomimetic technology has highlighted the importance of organelle attachment and
detachment in an organism’s ability to adapt to its environment. Tissue engineering’s
major goal is to make successful tissue grafts that can replace or repair damaged or
deteriorated tissues and organs. Currently, there are ongoing pre-clinical studies and
clinical applications of engineered tissues such as bones, cartilage, skin, skeletal
muscles, blood vessels, and bladder. In this chapter, the interconnection between
regenerative biology and engineering is examined, with emphasis on the utilization
of biomimetics in tissue engineering and the creation of functional tissue transplants
for regenerative medicine.
Abbreviation
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 125
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_5
126 S. Choudhury et al.
T tendon
L ligament
TEN tissue-engineered nerve
CRISPR clustered regularly interspaced short palindromic repeats
Cas9 clustered regularly interspaced short palindromic repeats-associated
9
ZIF-8 zeolitic imidazolate framework
ELPs Elastin-like polypeptides
PGA poly-glycolic acid
PLA poly-lactic acid
OATS osteochondral autograft transfer system
NYSERDA New York State Energy Research and Development Authority
BIONIS Biomimetics Network for Industrial Sustainability
PLMC Poly D, L-lactide-co-trimethylene carbonate
1 Introduction
Human design has been “inspired by nature” for over three thousand years [1]. The
phrase “learning from nature” can be referred to in a variety of ways, and as a
systematic methodology, it is still in its infancy as a subject of study, particularly in
engineering design. Biomimetics covers a wide range of study areas, has an impact
on a number of application areas, and is thought to significantly improve the quality
of life on the scientific, social, and economic levels [2]. The majority of important
findings, however, have remained in their respective fields because of the large and
dispersed nature of research topics. According to studies, individual parties rather
than institutions have tended to practice biomimetics [3].
Nature has long served as an inspiration for the creation of useful materials and
systems [4]. Biomimetics involves drawing inspiration, adapting, or deriving design
ideas from nature. It is a process of developing innovative technologies by studying
biological systems. Through biomimetics, concepts from the field of biology are
applied to engineering, enabling the development of technologies that benefit from
millions of years of development evolution in natural biological systems as a result of
natural selection [2]. It denotes the investigation and imitation of natural processes,
systems, and approaches [5].
Biomimetics is a relatively recent topic of study that entails applying biological
scientific concepts and functions to engineering, design, chemistry, electronics, and
other disciplines [1]. Biologists, physicists, chemists, material scientists, engineers,
and other professionals combine to create biomimetics. Exploring the functions,
structures, and principles of several natural things, as well as engineering and engi-
neering materials and techniques with commercial uses, are all part of this multidis-
ciplinary discipline [6]. In 1974, the term “biomimetics” first appeared in print in
Webster’s dictionary [7].
Biomimetic Approaches for Biomaterials Development 127
2 Terminology
2.1 Bioinspiration
2.2 Biomimicry
materials [11]. The field of engineered biomimicry is rapidly evolving and involves
various disciplines, including biology, materials science, and manufacturing, making
it a challenging area of study [12].
2.3 Biomimetics
The collaboration among diverse fields such as biology, technology, and other innova-
tive areas has the goal of addressing practical issues by analyzing biological systems,
creating models of them, and transferring and implementing these models to discover
solutions [3]. “Biomimetics” refers to the imitation of life or nature. It comes from
the Greek word bio mimesis [6].
2.4 Bionics
The term “bionics” was coined by Jack Steele of the US Air Force Medical Division in
1960 by combining the words “biology” and “technics”. Bionics refers to a technical
domain that aims to replicate, enhance, or replace biological functions with elec-
tronic and/or mechanical equivalents [13]. In addition, there is a growing interest
in researching the improved regenerative abilities of bionic tissue-engineered
nerve (TEN) grafts, with a specific focus on the bionics of their structure and
components [14].
2.5 Biomedicine
The application of engineering principles to the medical field is the focus of biomed-
ical engineering, which is an engineering discipline that has gained increasing accep-
tance within the field [16]. Another fast-developing field of biomedical engineering
is tissue engineering [17].
Biomimetic Approaches for Biomaterials Development 129
2.8 Biomechatronics
Fig. 1 Interlink between biology and biomimetic approaches (Made via powerpoint)
130 S. Choudhury et al.
3 Background to Biomimetics
4 Biological Materials
The body is a chemical laboratory that converts natural chemicals into energy,
construction materials, trash, and a variety of multifunctional structures [26]. Humans
have long recognized natural materials as sources of food, clothing, comfort, and
other essentials [27]. Despite their tiny size, animals and insects may create huge
quantities of natural products such as fur, leather, honey, wax, milk, and silk to suit
human requirements [5]. Due to their closely regulated structure and better-oriented
connections, many biological materials are less dense than synthetic materials with
equivalent qualities [7].
For centuries, natural materials have been utilized due to their appealing char-
acteristics like strength, durability, and beauty. In order to replicate these benefits,
attempts have been made to produce artificial imitations of these materials in any
quantity required. Natural materials possess unique features such as self-replication,
self-healing, reconfigurability, multifunctionality, and chemical equilibrium. Unlike
man-made materials that are typically processed through heating and pressuriza-
tion, natural materials are created under ambient conditions. Materials derived from
biological sources result in minimal waste and pollution, and the final product is
usually biodegradable and recyclable by nature. Mastering the handling of these
materials will expand our range of material options and enable us to produce more
environmentally friendly and recyclable materials [5].
Biomaterials play a crucial role in wound healing by acting as carriers for protein
and gene delivery, as well as providing scaffolds for cell attachment, growth, and
differentiation. They have been utilized to transport growth factors, small organic
compounds, and genes for the treatment of various diseases and wound healing [28].
Spider with their incredible webs is shown in Fig. 2.
Fig. 2 Spiders are impressive “manufacturing engineers” of the natural world, able to create mate-
rials with remarkable efficiency, as evidenced by their incredible webs [5] The silk material used
in the spider’s web is five times stronger than steel, when compared by weight. (Original picture
captured by author)
Fig. 3 The biologically inspired design employs two analogical processes: a An analogy based on
a solution
Fig. 4 The biologically inspired design employs two analogical processes: b analogy based on an
issue
potential design solutions. The principles learned through fundamental research can
then be applied to technology. The transfer of scientific knowledge to industry is
closely tied to this approach [3] (Fig. 3).
Biomimetic Approaches for Biomaterials Development 133
The subsequent steps in the design process that is driven by solutions and inspired
by biology [30]:
Step 1: Recognition of Biological Solutions
Designers start with a certain biological solution in mind [30]
Step 2: Definition of Biological Solution
Step 3: Extraction of the Principle
Step 4: Reframing the Problem
Creators are compelled to reinterpret their work in order to assess how consumers
will appreciate the biological purpose attained [30].
Step 5: Look for a Problem
In contrast to search in the biological world, which entails searching through
some finite space of biological answers, it may encompass inventing whole new
challenges [30].
Step 6: Difficulties Identification
Step 7: Implementation of the Principle
Tissue engineering enables the creation of biological substitutes for diseased and
damaged tissues using a number of techniques [31]. To attain the desired outcome,
numerous approaches are being investigated in musculoskeletal tissue engineering.
Among these, implanting 3D synthetic polymeric scaffolds at the site of tissue
damage is a widely used method. These scaffolds provide a framework that facili-
tates cell attachment, proliferation, and extracellular matrix (ECM) synthesis [32],
to accelerate tissue regeneration [33]. Many researchers have created scaffolds using
standard approaches such as phase separation salt leaching [34, 35].
The discovery of biomimetic scaffolds that mimic the architecture of the extra-
cellular matrix (ECM) has recently emerged as a promising strategy for tissue regen-
eration. To fabricate these 3D biomimetic scaffolds, advanced techniques such as
electrospinning and rapid prototyping have been employed. These scaffolds possess
Biomimetic Approaches for Biomaterials Development 135
problem. This technique involves using cell membranes obtained from lymphocytes,
macrophages, thrombocytes, and other sources to overcome the challenges related
to protein adsorption [8]. The biological composition of the cell membrane surface
is crucial in controlling the adsorption of biological components on particles, which
affects the circulation time, immune-mediated degradation, and biocompatibility of
the particles [40].
Stretchable and flexible pressure sensors rely on biomimetic materials with micro-
and nano-hierarchical structures to work. These sensors have high sensitivity,
stability, and a wide pressure range [8]. Stimuli-responsive actuators, which can
transform diverse forms of energy such as thermal, optical, and electrical energy into
mechanical energy, are another application of biomimetic materials. These actu-
ators are highly versatile and can perform intricate mechanical movements in a
precise sequence, making them useful for bionic devices, artificial muscles, and intel-
ligent robots [41]. In recent times, there has been a growing focus on multi-stimulus
actuators instead of single-stimulus actuators [41]. Bilayer actuators are an attrac-
tive candidate for biomimetic applications because of their outstanding mechanical
characteristics and significant photo-induced stress [8].
7 Tissue Engineering
One of the most successful approaches for replacing or repairing damaged skin
and other biological components is tissue engineering [42]. In the artificial tissue
regeneration process, both in vivo and in vitro procedures are utilized [43]. It offers a
significant treatment alternative by combining the patient’s own cells with a scaffold
for bone healing [42]. The purpose of biomimetic tissue engineering approaches is
to use biomaterials to actively drive tissue repair and regeneration [44]. To guarantee
effective regeneration of the target tissue, the biomaterials used in tissue-engineered
constructs (TECs) and drug delivery must fulfill particular biological and mechanical
parameters [45].
Biomimetic materials that mimic the characteristics of the natural extracellular
matrix (ECM) have been created with the help of recent advancements in biomaterials
and tissue engineering [46].
Scaffolding plays a crucial role in facilitating cell adhesion, growth, and prolifer-
ation during both in vivo and in vitro artificial tissue regeneration procedures [42].
The intricate nature of tissue engineering interfaces involves a multi-scale organi-
zation of various tissue types, which necessitates collaboration among professionals
from diverse fields such as biomedical engineering, materials science, and orthopedic
surgery [47].
Biomimetic Approaches for Biomaterials Development 137
Fig. 5 Biomaterials, both natural and synthetic, for tissue engineering (Made via PowerPoint)
employed in bone tissue engineering, there is a growing need for modern bioma-
terials with improved mechanical and functional properties to overcome emerging
challenges. Aliphatic polycarbonates are one such promising biomaterial, possessing
desirable characteristics like biodegradability and biocompatibility, which make
them increasingly valuable in medical applications [8].
The mechanical qualities of the musculoskeletal system’s meniscus (M), tendon (T),
and ligament (L) tissues are elastic and flexible (Fig. 6). Due to their relatively weaker
circulatory system, these tissues require a lower amount of oxygen and nutrients for
their regeneration and repair, in contrast to other tissues [47]. Due to the poor ability
for spontaneous healing in these tissues, surgical treatments such as autografts and
allografts are frequently required in situations of damage [51]. Traditional treatments
have drawbacks, such as graft rejection and poor health consequences have prompted
the investigation of biomaterial engineering for M/T/L tissues as a viable alternative.
Biomimetic Approaches for Biomaterials Development 139
Fig. 6 Meniscus (M), tendon (T), and ligament (L) are all elastic tissues in the musculoskeletal
system (L). (Images from Google used under Creative Commons Licenses)
The stabilization of joints and the restriction of bone movement are critical functions
of ligaments, while tendons transmit forces generated by contracting muscles to
bones. These tissue types’ interfaces or junctions, which anchor soft tissue to bone,
are critical components of this process [51].
Biomimetic biomaterials such as collagen and elastin are frequently used in
the engineering of elastic tissues [49]. Elastin is a highly desirable biomaterial
because of its exceptional elasticity, durability, and responsiveness to stimuli such as
LCST. Elastin-like polypeptides (ELPs) are extensively generated through chemical
synthesis and recombinant DNA expression, and serve as valuable model systems for
research and biomaterials in numerous biomedical domains, including protein purifi-
cation, drug delivery, and tissue engineering. By combining or crosslinking these
polymers, biomimetic materials with varying levels of crosslinking or combination
can be produced [25].
Fig. 7 Teeth, bone, and seashells are examples of hard tissues. (Pictures taken from Google under
Creative Commons Licenses)
In addition to strength, toughness is a vital attribute that influences the tensile prop-
erties of hard materials. There have been about 60 biogenic minerals found, which
are minerals that originate from biological processes. The most common biogenic
minerals include hydroxyapatite (found in bones and teeth), calcium carbonate
(found in seashells), and silica (found in sub-millimeter marine organisms such as
radiolarians and diatoms). These minerals come in different shapes, amounts, and
sizes [52].
Hard materials are essential in the design or restoration of various components
of the musculoskeletal system, such as bone tissue. Orthopedic treatments, which
are becoming increasingly prevalent, employ a variety of materials, each with its
own set of benefits and drawbacks. Ceramics and bioglass were the initial hard
biomaterials used in hard tissues [54, 55]. Alternatives include biomaterials made
of calcium sulfate and calcium phosphate, which are biocompatible and absorbable.
Several calcium and phosphate combinations have been investigated for orthopedic
purposes, including bone cement. As these materials decompose, they produce safe
ions such as calcium, phosphate, and sulfate that are already present in the body.
Among the different forms of calcium phosphate, hydroxyapatite has gained more
attention. Consequently, researchers have developed composite scaffolds by blending
different forms of hydroxyapatite with organic or inorganic biodegradable polymers
for use in hard tissues such as bone and osteochondral [49] (Fig. 7).
Several issues, such as tissue resorption and implant rupture or contracture, limit
the use of autologous tissue transplants and synthetic implants in current soft tissue
reconstruction techniques [56]. In order to construct soft tissue structures like muscle
and cartilage in the musculoskeletal system, a combination of natural and synthetic
biomaterials is utilized (Fig. 8). Collagen, gelatin, hyaluronic acid, chitosan, and
acellular matrix are some of the most often employed natural compounds for this
Biomimetic Approaches for Biomaterials Development 141
Fig. 8 Collagen is a natural substance found in soft tissues such as muscle and cartilage. (Picture
taken from Google under creative commons licenses)
purpose [56]. Hydrogel structures and sponges made from a diverse range of mate-
rials, including alginate, agarose, collagen, hyaluronan, fibrin gels, poly (glycolic
acid) (PGA), and poly (lactic acid) (PLA), are commonly utilized in cartilage tissue
engineering [57].
The self-healing capacity of adult articular cartilage is limited, and even minor
injuries or damage can lead to the degeneration of joints and result in severe pain
and functional limitations. Despite advances in cartilage tissue engineering during
the previous two decades, properly treating articular cartilage defects remains a
substantial therapeutic problem. Despite considerable efforts to develop innovative
biological solutions, there are currently limited therapeutic options available [57]
(Fig. 8).
9 Tissue Grafting
Replacement of sick or damaged tissue removed from one portion of the body with
healthy skin, bone, or other tissue.
There are three types of tissue grafts: autograft, allograft, and xenograft. Autograft
involves transferring tissue from one area of the body to another, like using the
semitendinosus tendon from the same patient to fix the anterior cruciate tendon. In
contrast, allograft involves utilizing tissue from a donor to reconstruct and repair
tissue in another person. Depending on the intended use, the donated tissue can be
fresh, processed, or frozen. Strict screening, processing, and testing are conducted
to ensure safety and minimize the risk of disease transmission, which may involve
washing and irradiation [58].
142 S. Choudhury et al.
Stem cell therapy is concerned with the restoration of normal function by replacing
or regenerating human cells, tissues, or organs. With the aid of pioneering methods,
this field holds promise in effectively curing or treating ailments that were previously
challenging to manage with conventional medications and procedures. As a result,
regenerative medicine has the potential to transform human medicine [60].
Biomimetic Approaches for Biomaterials Development 143
Approximately six decades ago, Dr. Joseph Murray, John Merrill, and J. Hartwell
Harrison spearheaded a team that executed the first triumphant organ transplant in
Boston, effectively inaugurating this field. This momentous feat heralded a new age
in organ transplantation and presented a remedy for patients with end-stage organ
disease, an unprecedented achievement in human history [59].
In the late 1950s and 1960s, the first successful cell therapies, including bone
marrow transplantation, were discovered [61].
Regenerative medicine encompasses several biomedical methodologies directed
at mending injured organs or tissues, evoking both positive expectations and ethical
controversies. The ethical predicaments surrounding this field have resulted in a
schism between traditional Christian societies and post-Christian secular societies. In
addressing bioethical disagreements in regenerative medicine, it is essential to recog-
nize the distinct moral and metaphysical perspectives that underpin these debates.
Each side presents individual interpretations of human nature. While mortality
persists as an inevitability, regenerative medicine holds promise in prolonging human
lifespans [9].
Research in regenerative medicine includes the study of self-healing, where the
body employs its own mechanisms, possibly with the help of foreign biological
material, to restore tissues and organs. As the focus of the field moves toward devel-
oping cures for complex and chronic illnesses, The phrases “tissue engineering” and
“regenerative medicine” have grown more interchangeable [62].
While the regenerative medicine industry is developing, it has been noted that
there are not only therapeutic applications but also non-therapeutic ones, including
the use of tissues as biosensors to detect biological or chemical hazards, as well as
tissue chips that can be used to assess the toxicity of new drugs [1].
Cells are the fundamental unit of the human body, and they work together to form
tissues. These tissues contain cells that produce an extracellular matrix or scaffold,
which offers support to the cells and serves as a platform for signaling molecules
to communicate with the cells. These signals trigger cellular responses that regulate
their functions. By studying how cells interact with their environment, organize
themselves into tissues, and respond to signals, scientists have been able to develop
techniques to repair damaged tissues and generate new ones [63].
The first step in tissue engineering often involves the creation of a scaffold
using a variety of materials, including proteins or polymers. Following the scaf-
fold’s construction, cells can be introduced either with or without growth factors,
depending on the desired outcome. If the environmental conditions are favorable, the
cells can self-organize into tissue. Alternatively, all three components—cells, scaf-
fold, and growth factors—can be mixed simultaneously, resulting in self-assembly
of the tissue [64].
144 S. Choudhury et al.
Although tissue engineering has shown promise in generating implants for certain
body parts such as bladders, tiny arteries, skin grafts, and even trachea, it is still not
a widely used treatment in patient care due to its experimental nature and high costs.
Although laboratory-engineered tissues such as heart, lung, and liver tissues have
been developed, they are still far from being entirely reproducible for implantation
into humans. However, these tissues can be beneficial for drug development research,
allowing for the use of active human tissue to test drug candidates, saving money,
reducing the usage of research animals, and potentially speeding up the development
of personalized therapy [2].
York State Energy Research and Development Authority (NYSERDA) has made
biomimicry a requirement to tackle several energy-related challenges [39].
Biomimetics has received significant attention in Germany, where 28 research
institutions collaborate in BIOKON and the Federal Ministry of Education and
Research oversees 35 initiatives. In the United Kingdom, BIONIS serves as
a connector between academic institutions and businesses, while in Japan, the
Century Center of Excellence, under the supervision of the Ministry of Education,
Culture, Sports, Science, and Technology, focuses on biomimetic manufacturing and
innovative use of biological resources in agriculture [33].
Biomimetics, which takes inspiration from living organisms, has led to the develop-
ment of many useful products that have enhanced human life. The combination of
biomedical engineering, medicine, science, and biomimetics is anticipated to have a
significant impact on managing diseases, disabilities, and injuries in the future, partic-
ularly in fields such as tissue engineering and regenerative medicine. Biomimetic
principles and functions, like the adhesive and regenerative properties of spider webs,
leukocyte migration and adhesion during inflammation, and the regenerative capa-
bilities of lizards and buckhorns, can be employed in biomedical engineering [59].
The emerging field of next-generation biomimetics is using a combination of
biology and technology to tackle various issues. Nanotechnology is becoming
increasingly important in understanding the structures of materials and accelerating
the formation of secondary structures of proteins. To promote bone regeneration,
researchers have designed multifunctional fibrous scaffolds that mimic the archi-
tecture of native tissue. One recent study examined the potential of using poly D,
L-lactide-co-trimethylene carbonate (PLMC) nanofibers as scaffold materials for
tissue repair and regeneration, and found that their biomimetic properties improved
efficiency. By incorporating biomimetics into biomedical engineering, technology is
being advanced in numerous ways [4].
13 Conclusion
parts of our life. While some of the ideas may appear to be science fiction given
our current technology constraints, these notions may get closer to reality than we
realize as we develop a better knowledge of nature and enhance our skills.
References
1. Vincent, J.F., Bogatyreva, O.A., Bogatyrev, N.R., Bowyer, A., Pahl, A.K.: Biomimetics: its
practice and theory. J. R. Soc. Interface. 3(9), 471–482 (2006)
2. Lepora, N.F., Verschure, P., Prescott, T.J.: The state of the art in biomimetics. Bioinspir. Biomim.
8(1), 013001 (2013)
3. Fayemi, P.E., Wanieck, K., Zollfrank, C., Maranzana, N., Aoussat, A.: Biomimetics: Process,
tools and practice. Bioinspir. Biomim. 12(1), 011002 (2017)
4. Sarikaya, M., Tamerler, C., Jen, A.K., Schulten, K., Baneyx, F.: Molecular biomimetics:
nanotechnology through biology. Nat. Mater. 2(9), 577–585 (2003)
5. Bar-Cohen Y.: Biomimetics: biologically inspired technologies. CRC press (2005)
6. Bhushan, B.: Biomimetics: lessons from nature–an overview. Philos. Trans. R. Soc. A: Math.,
Phys. Eng. Sci. 367(1893), 1445–1486 (2009)
7. Vincent, J.F.: Biomimetics—a review. Proc. Inst. Mech. Eng. [H] 223(8), 919–939 (2009)
8. Suresh Kumar, N., Padma Suvarna, R., Chandra Babu Naidu, K., Banerjee, P., Ratnamala, A.,
Manjunatha, H.: A review on biological and biomimetic materials and their applications. Appl.
Phys. A. 126(6), 445 (2020)
9. Ferrari, M., Martin, D.K.: Nanobiotechnology of biomimetic membranes. Springer Science
(2007)
10. Naik, R.R., Singamaneni, S.: Introduction: bioinspired and biomimetic materials. Chem. Rev.
117(20), 12581–12583 (2017)
11. Cohen, Y.H., Reich, Y.: Biomimetic design method for innovation and sustainability. Springer,
Berlin, Germany (2016)
12. Martín-Palma, R.J., Lakhtakia, A.: Progress on bioinspired, biomimetic, and bioreplication
routes to harvest solar energy. Appl. Phys. Rev. 4(2), 021103 (2017)
13. Iouguina, A.: Biologically informed disciplines: a comparative analysis of terminology within
the fields of bionics, biomimetics, and biomimicry (Doctoral dissertation, Carleton University)
14. Qi, T., Zhang, X., Gu, X., Cui, S.: Experimental study on repairing peripheral nerve defects
with novel bionic tissue engineering. Adv. Healthcare Mater. 5, 2203199 (2023)
15. Gaines, A.D., Davis-Floyd, R.: Biomedicine. Encycl. Med. Anthropol. 1, 95–109 (2004)
16. Cha, C., Shin, S.R., Annabi, N., Dokmeci, M.R., Khademhosseini, A.: Carbon-based nano-
materials: multifunctional materials for biomedical engineering. ACS Nano 7(4), 2891–2897
(2013)
17. Melchels, F.P., Feijen, J., Grijpma, D.W.: A review on stereolithography and its applications
in biomedical engineering. Biomaterials 31(24), 6121–6130 (2010)
18. Vattam, S.S., Helms, M.E., Goel, A.K.: Compound analogical design: interaction between
problem decomposition and analogical transfer in biologically inspired design. Des. Comput.
Cogn. 23(8), 377–396 (2008)
19. Witte, H., Lutherdt, S., Schilling, C.: Biomechatronics: how much biology does the engineer
need? In: Proceedings of the 2004 IEEE International conference on control applications, vol.
2, pp. 944–948. IEEE (2004)
20. Witte, H., Fremerey, M., Weyrich, S., Mämpel, J., Fischheiter, L., Voges, D., Zimmermann,
K., Schilling, C.: Biomechatronics is not just biomimetics. In: 9th International workshop on
robot motion and control, pp. 74–79. IEEE (2013)
21. Cassie, A.B., Baxter, S.: Wettability of porous surfaces. Trans. Faraday Soc. 40, 546–551 (1944)
22. Zhang, J., Sheng, X., Jiang, L.: The dewetting properties of lotus leaves. Langmuir 25(3),
1371–1376 (2009)
148 S. Choudhury et al.
23. Bixler, G.D., Bhushan, B.: Bioinspired rice leaf and butterfly wing surface structures combining
shark skin and lotus effects. Soft Matter 8(44), 11271–11284 (2012)
24. Panchanathan, D., Rajappan, A., Varanasi, K.K., McKinley, G.H.: Plastron regeneration on
submerged superhydrophobic surfaces using in situ gas generation by chemical reaction. ACS
Appl. Mater. Interfaces 10(39), 33684–33692 (2018)
25. Kushner, A.M., Guan, Z.: Modular design in natural and biomimetic soft materials. Angew.
Chem. Int. Ed. 50(39), 9026–9057 (2011)
26. Mann, S., (ed.).: Biomimetic materials chemistry. John Wiley & Sons, (1996)
27. Carlson, J., Ghaey, S., Moran, S., Tran, C.A., Kaplan, D.L.: Biological materials in engineering
mechanisms. In: Biomimetics, pp. 383–398, CRC Press, (2005)
28. Andreadis, S.T., Geer, D.J.: Skin structure and physiology. Trends Biotechnol. 7(24), 331–337
(2006)
29. Zhang, G.: Biomimicry in biomedical research
30. Helms, M., Vattam, S., Goel, A.K., Yen, J., Weissburg, M.: Problem-driven and solution-based
design: twin processes of biologically inspired design. In: ACADIA08 Conference, (2008)
31. Salgado, A.J., Coutinho, O.P., Reis, R.L.: Bone tissue engineering: state of the art and future
trends. Macromol. Biosci. 4(8), 743–765 (2004)
32. Agrawal, C.M., Ray, R.B.: Biodegradable polymeric scaffolds for musculoskeletal tissue engi-
neering. J. Biomed. Mater. Res.: Off. J. Soc. Biomater., Jpn. Soc. Biomater., Aust. Soc.
Biomater. Korean Soc. Biomater. 55(2), 141–150 (2001)
33. Loh, Q.L., Choong, C.: Three-dimensional scaffolds for tissue engineering applications: role
of porosity and pore size
34. Diba, M., Kharaziha, M., Fathi, M.H., Gholipourmalekabadi, M., Samadikuchaksaraei,
A.J.: Preparation and characterization of polycaprolactone/forsterite nanocomposite porous
scaffolds designed for bone tissue regeneration. Compos. Sci. Technol. 72(6), 716–723 (2012)
35. Golafshan, N., Gharibi, H., Kharaziha, M., Fathi, M.: A facile one-step strategy for development
of a double network fibrous scaffold for nerve tissue engineering. Biofabrication 9(2), 025008
(2017)
36. Hardy, J.G., Ghezzi, C.E., Saballos, R.J., Kaplan, D.L., Schmidt, C.E.: Supracolloidal assem-
blies as sacrificial templates for porous silk-based biomaterials. Int. J. Mol. Sci. 16(9),
20511–20522 (2015)
37. Rouet, R., Thuma, B.A., Roy, M.D., Lintner, N.G., Rubitski, D.M., Finley, J.E., Wisniewska,
H.M., Mendonsa, R., Hirsh, A., de Onate, L., Compte, B.J.: Receptor-mediated delivery of
CRISPR-Cas9 endonuclease for cell-type-specific gene editing. J. Am. Chem. Soc. 140(21),
6596–6603 (2018)
38. Wells, D.J.: Gene therapy progress and prospects: electroporation and other physical methods.
Gene Ther. 11(18), 1363–1369 (2004)
39. Stride, E., Saffari, N.: Microbubble ultrasound contrast agents: a review. Proc. Inst. Mech. Eng.
[H] 217(6), 429–447 (2003)
40. Su, G., Zhou, X., Zhou, H., Li, Y., Zhang, X., Liu, Y., Cao, D., Yan, B.: Size-dependent
facilitation of cancer cell targeting by proteins adsorbed on nanoparticles. ACS Appl. Mater.
Interfaces 8(44), 30037–30047 (2016)
41. Hou, Y., Chang, H., Song, K., Lu, C., Zhang, P., Wang, Y., Wang, Q., Rao, W., Liu, J.: Coloration
of liquid-metal soft robots: From silver-white to iridescent. ACS Appl. Mater. Interfaces 10(48),
41627–41636 (2018)
42. Ng, J., Spiller, K., Bernhard, J., Vunjak-Novakovic, G.: Biomimetic approaches for bone tissue
engineering. Tissue Eng. Part B Rev. 23(5), 480–493 (2017)
43. Reddy, R., Reddy, N.: Biomimetic approaches for tissue engineering. J. Biomater. Sci. Polym.
Ed. 29(14), 1667–1685 (2018)
44. Thibeault, S.L., Klemuk, S.A., Smith, M.E., Leugers, C., Prestwich, G.: In vivo comparison
of biomimetic approaches for tissue regeneration of the scarred vocal fold. Tissue Eng. Part A
15(7), 1481–1487 (2009)
45. Bas, O., Catelas, I., De-Juan-Pardo, E.M., Hutmacher, D.W.: The quest for mechanically and
biologically functional soft biomaterials via soft network composites. Adv. Drug Deliv. Rev.
1(132), 214–234 (2018)
Biomimetic Approaches for Biomaterials Development 149
46. Rahmany, M.B., Van Dyke, M.: Biomimetic approaches to modulate cellular adhesion in
biomaterials: A review. Acta Biomater. 9(3), 5431–5437 (2013)
47. Boys, A.J., McCorry, M.C., Rodeo, S., Bonassar, L.J., Estroff, L.A.: Next generation tissue
engineering of orthopedic soft tissue-to-bone interfaces. MRS Communs. 7(3), 289–308 (2017)
48. Gingery, A., Killian, M.L.: * Special focus issue on strategic directions in musculoskeletal
tissue engineering. Tissue Eng. Part A 23(17–18), 873 (2017)
49. Del Bakhshayesh, A.R., Asadi, N., Alihemmati, A., Tayefi Nasrabadi, H., Montaseri, A.,
Davaran, S., Saghati, S., Akbarzadeh, A., Abedelahi, A.: An overview of advanced biocom-
patible and biomimetic materials for creation of replacement structures in the musculoskeletal
systems: focusing on cartilage tissue engineering. J. Biol. Eng. 13, 1–21 (2019)
50. Camargo, P.H., Satyanarayana, K.G., Wypych, F.: Nanocomposites: synthesis, structure,
properties and new application opportunities. Mater. Res. 12, 1–39 (2009)
51. Patel, S., Caldwell, J.M., Doty, S.B., Levine, W.N., Rodeo, S., Soslowsky, L.J., Thomopoulos,
S., Lu, H.H.: Integrating soft and hard tissues via interface tissue engineering. J. Orthop.
Research® . 36(4), 1069–1077 (2018)
52. Barthelat, F.: Biomimetics for next generation materials. Philos. Trans. R. Soc. A: Math., Phys.
Eng. Sci., 365(1861), 2907–2919 (2007)
53. Zhang, K., Wang, S., Zhou, C., Cheng, L., Gao, X., Xie, X., Sun, J., Wang, H., Weir, M.D.,
Reynolds, M.A., Zhang, N.: Advanced smart biomaterials and constructs for hard tissue
engineering and regeneration. Bone research. 6(1), 31 (2018)
54. Hench, L.L.: Bioceramics: from concept to clinic. J. Am. Ceram. Soc. 72, 93–98 (1993)
55. Hench, L.L.: Opening paper 2015-some comments on bioglass: four eras of discovery and
development. Biomed. Glass. 1(1), (2015)
56. Bressan, E., Favero, V., Gardin, C., Ferroni, L., Iacobellis, L., Favero, L., Vindigni, V., Berengo,
M., Sivolella, S., Zavan, B.: Biopolymers for hard and soft engineered tissues: application in
odontoiatric and plastic surgery field. Polymers 3(1), 509–526 (2011)
57. Johnstone, B., Alini, M., Cucchiarini, M., Dodge, G.R., Eglin, D., Guilak, F., Madry, H.,
Mata, A., Mauck, R.L., Semino, C.E., Stoddart, M.J.: Tissue engineering for articular cartilage
repair—the state of the art. Eur. Cell. Mater. 25(248), e67 (2013)
58. Jones, L.C., Topoleski, L.T., Tsao, A.K.: Biomaterials in orthopaedic implants. In: Mechanical
testing of orthopaedic implants, pp. 17–32. Woodhead Publishing, (2017)
59. Parker, A.R., Townley, H.E.: Biomimetics of photonic nanostructures. Nat. Nanotechnol. 2(6),
347–353 (2007)
60. Hua, Q., Sun, J., Liu, H., Bao, R., Yu, R., Zhai, J., Pan, C., Wang, Z.L.: Skin-inspired highly
stretchable and conformable matrix networks for multifunctional sensing. Nat. Commun. 9(1),
244 (2018)
61. Mao, A.S., Mooney, D.J.: Regenerative medicine: Current therapies and future directions. Proc.
Natl. Acad. Sci. 112(47), 14452–14459 (2015)
62. Pangarkar, S., Pham, Q.G., Eapen, B.C. (eds.): Pain care essentials and innovations E-Book.
Elsevier Health Sciences, (2020)
63. Stearns-Reider, K.M., Hicks, M.R., Hammond, K.G., Reynolds, J.C., Maity, A., Kurman-
galiyev, Y.Z., Chin, J., Stieg, A.Z., Geisse, N.A., Hohlbauch, S., Kaemmer, S.: Myoscaf-
folds reveal laminin scarring is detrimental for stem cell function while sarcospan induces
compensatory fibrosis. NPJ Regenerative Medicine. 8(1), 16 (2023)
64. Zydney, J.M.: Scaffolding. Encycl. Sci. Learn., 2913–2916 (2012)
65. Hwang, J., Jeong, Y., Park, J.M., Lee, K.H., Hong, J.W., Choi, J.: Biomimetics: forecasting the
future of science, engineering, and medicine. Int. J. Nanomed., 5701–5713 (2015)
66. Wu, M., Liu, X., Bai, H., Lai, L., Chen, Q., Huang, G., Liu, B., Tang, G.: Surface-layer
protein-enhanced immunotherapy based on cell membrane-coated nanoparticles for the effec-
tive inhibition of tumor growth and metastasis. ACS Appl. Mater. Interfaces 11(10), 9850–9859
(2019)
67. Sridhar, K., Bouhallab, S., Croguennec, T., Renard, D., Lechevalier, V.: Application of high-
pressure and ultrasound technologies for legume proteins as wall material in microencapsula-
tion: new insights and advances. Trends Food Sci. & Technol., (2022)
150 S. Choudhury et al.
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 153
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_6
154 A. Izadyari Aghmiuni et al.
1 Introduction
In the biomedical field, natural biomaterials based on their origin include the
following 5 categories (Fig. 1).
• Polysaccharides: chitin, chitosan, cellulose, agarose, etc.
• Proteins: silk (fibroin and sericin), collagen, etc.
• Glycosaminoglycans
• Cell/organ-derived matrices such as decellularized skin, liver, heart valves, etc.
• Herbal active ingredients
Plant-Based Biomaterials in Tissue Engineering and Drug Delivery … 155
All these groups play an impact on the decrease of the use of xenografts, allografts,
and autografts, and can overcome the limitations such as the reduction of donor
sites, and the creation of immune reactions. In this regard, decellularization is one of
the methods that can be effectively used as homografts and xenografts [15]. In this
method, the tissues or the organs lose all their cell components and leave intact ECMs
[12]. In these conditions, the ECM can maintain its role in mechanical supports,
signaling, and improvement of cell adherence [16]. Such matrices can be applied
in regenerating tissues. Moreover, the design of natural polymer-based substrates,
substitutes, or scaffolds can be also useful in achieving such a matrix that can imitate
the function and structure of native ECMS [8].
These biopolymers also can act as carriers of drugs, genes, growth factors,
macromolecules, etc. for biomedical applications [14, 17]. Notably, medicinal herbs
themselves can be used as therapeutic agents or factors for improving cellular
behavior.
However, despite the nature of these materials, natural biomaterials possess unique
properties which led to an increase in their use of biomedical, tissue engineering
technologies, and drug delivery systems. The characterizations and classification of
these biomaterials have been listed in Table 1.
thermomechanical properties
Agarose This biomaterial is a main constituent of Red algae and seaweed [37–41]
agar that possesses beneficial features
similar to alginate and can act as a
differentiation medium for stem cells, as
well as an excellent candidate to apply
in neural and cardiac tissue engineering
(continued)
157
Table 1 (continued)
158
Chondroitin sulfate This biomaterial is comprised of Extracted and purified from [53–55]
repeated di-saccharide units, as well as bovine, shark, porcine, chicken,
sulfated at positions 4 or 6. This and skate cartilage
biopolymer can play a key role in
treating cartilage
(continued)
159
Table 1 (continued)
160
[119]. Pharmaceutical systems are considered the main approaches in this field that
can be systemically and orally administered to deliver hydrophobic and hydrophilic
therapeutic agents. These systems as effective therapeutic actions play a key role in
controlled drug release, especially for low concentrations of drugs. A drug carrier
in these systems provides a rate of controlled release and complete rehabilitation of
the drug in more time [120–122]. These carriers can be injectable and/or implanted
in the damaged or diseased tissue/cell to improve delivery (Fig. 2) [123].
One of the methods for achieving this aim is the surface modification of carriers or
the design of hybrid/composite carriers to prevent the rapid destruction/degradation
of the carrier, encapsulate the drug for a lengthened release, and sustain drug stability
[124, 125]. In this regard, biomaterials and their products can be suitable options due
to their biocompatible properties and nontoxic, lower processing costs, etc.
Many studies in this field indicate the interest of researchers in using these attrac-
tive materials. The study of Kilicarslan et al. is one of these cases that is about
complex films prepared with alginate-chitosan to treat periodontal diseases [126].
Accordingly, it is known that the increase in alginate concentration and chitosan
molecular weight can increase the adhesiveness of film content. Their reports indi-
cated that a 3:1 concentration of alginate: chitosan along with the lower molecular
weight of chitosan can provide more controlled release.
The release of ciprofloxacin antibiotic from alginate–gelatin microspheres is
another study performed by Islan et al. [127]. They stated that the encapsulation of this
antibiotic can decrease gastrointestinal irritations related to its oral administration.
Based on their results, these microspheres indicated ~80% encapsulation efficiency
Plant-Based Biomaterials in Tissue Engineering and Drug Delivery … 167
and release of less than 10% in 15 min (under simulated gastric conditions). More-
over, the results of this study indicated that Ca2+ /glutaraldehyde-crosslinked alginate-
gelatin microspheres can provide more suitable surfaces for decorating bioactive
molecules.
Chen et al. also illustrated that chitosan/alginate-coated complexes can provide
the controlled release of insulin and overcome challenges of insulin delivery [128].
Based on their results, the self-assembly of chitosan and alginate-coated nanoparti-
cles through the double emulsion method provides nanoparticles in the 200–300 nm
range and prolonged insulin release.
There are many pieces of research on the matter of targeted drug delivery for
diabetes treatment, and the development of metformin nano-emulsions is one of
these studies. Accordingly, Kumar et al. stated that alginate nanocapsules-loaded
metformin with an encapsulation efficiency of 78% can improve gastrointestinal
absorption and intestinal permeability and promote the efficacy of the drugs (3 times
higher than that of pure metformin) [129].
Varadharaj et al. demonstrated that starch-based nanoparticles possess anti-
diabetic and antioxidant properties. Such that 100 μg/mL concentrations indicated
the highest antioxidant properties (~75%) [130]. They reported that synthesized
nanoparticles possess potential properties against diabetes due to their high activity
in alpha-amylase inhibition (58.56%).
Starch-modified nanoemulsions loaded with borage seed oil are another study for
targeted delivery systems that can be applied in food industries and the production
of dietary supplements [131]. Accordingly, Rehman et al. illustrated that such nano-
emulsions along with peppermint oil can be used as a natural antioxidant (maximum
antioxidant capacity: 99.42 μg Trolox/mL).
The development of polymer micelles loaded with curcumin is another study
on treating diabetic ulcers [132]. In this study, Usman Akbar et al. showed
that these polymer micelles as therapeutic carriers can overcome limitations in
the use of curcumin like poor solubility and low bio-availability. Moreover, the
curcumin-loaded micelles can accelerate the wound healing process compared to pure
curcumin. Yang et al. have also reported that insulin-loaded polysaccharide hydrogels
can be effective in treating type 2 diabetes mellitus [133]. Accordingly, the authors
stated that oral administration of encapsulated insulin not only can significantly
reduce the disease symptoms in diabetic mice but also improve insulin sensitivity
and regulate lipid metabolism.
Some food gums such as Salcan can be also suitable candied for oral drug delivery.
The study of Qi et al. is one sample in this field to deliver insulin orally [134]. Based
on their reports, the acidic pHs led to an inhibition of insulin release, while neutral
conditions significantly increased drug release. Furthermore, oral administration of
these hydrogels illustrated a sustained reduction in the level of fasting blood glucose
in diabetic rats (after 6 h).
Studies have demonstrated that herbal polysaccharides-based nano-hydrogels can
also play an impact on dermal regeneration. In this regard, bio-composite nano-
hydrogels based on bamboo prepared by Padwad et al. were known as a suitable
therapeutic factor for healing wounds in streptococci that can regulate the expression
168 A. Izadyari Aghmiuni et al.
The reports have indicated that natural polymers and bioactive materials can be
promising strategies in biomedical applications due to their role in the improve-
ment of cell–cell biological responses and the imitation of functional and structural
properties of the target tissues [163]. Indeed, recent efforts in the fabrication of
naturally derived biomaterials-based substitutes [164–169] have provided significant
opportunities to generate novel tissue substitutes [15, 170].
Medicinal herbs are known as one of these biomaterials which gaining signifi-
cant attention among researchers due to their potential for increasing cell prolifera-
tion and controlled differentiation [171, 172]. The biopolymers similar to signaling
biomolecules of tissue extracellular matrix (ECM) (i.e. glycoproteins, proteoglycans,
and glycosaminoglycans) are other samples from these materials. In this field silk
fibers [173] and hyaluronic acid [174–176] can imitate the behavior of glycosamino-
glycans and proteoglycans in the tissue ECMs and be led to an increase in cell-to-cell
or cell-to-scaffold signaling [175, 177, 178].
According to studies, naturally derived biomaterials can be effective in cellular
functions like signaling, growth, differentiation, etc. Generally, the tremendous
differentiation of stem cells acts as a double-edged sword, hence therapeutic strate-
gies that led to the controlled differentiation of cells play a crucial role in this field.
One of these strategies includes the use of biomaterials to design tissue substitutes [2,
11]. Such an approach provides suitable physicochemical and mechanical properties
for cell differentiation and results in a sustained delivery and local of differentiated
cells to the targeted tissues.
In this regard, the studies of Izadyari Aghmiuni et al. indicate that herbs-derived
biomaterials possess high therapeutic potential [171]. Although, herbal ingredients-
based different formulas such as gels, creams, ointments, etc., cannot be directly
used in damaged tissues like bone, cartilage, nerve, cornea, tendon, etc. However,
this research team showed that some mucilage such as quince seed possesses good
restorative potential when used in the structure of tissue substitutes. Our results illus-
trated that quince seed mucilage-based hybrid scaffolds not only can provide a better
porous network than chitosan-based scaffolds but also support dermal fibroblasts
and improve the absorption capacity of water or wound secretions on the scaffold.
They also found that the combination of quince seed mucilage and PEG can increase
the transduction of mechanical force-induced signals and promotes the biological
signals to induce stem cell differentiation into targeted cells such as dermal cells.
170 A. Izadyari Aghmiuni et al.
Table 2 (continued)
Industry Carrier/entrapped material Function Refs.
Eudragit RL-100/ Increase of controlled drug release and [149]
gelatin-based nanoparticles/ improvement of ketorolac
Ketorolac tromethamine tromethamine bioavailability for ocular
applications
Gelatin/Silver nanoparticles The increase in stability and [150]
antibacterial activities against
Staphylococcus aureus; inhibition of
angiogenesis disturbance and corneal
neovascularization; a dual functional
nanotherapeutic (i.e. antimicrobial and
antiangiogenic) to treat eye-related
microbial infections
Egg-phosphatidyl A long-term sustained release of the [151]
choline-derived Liposomes/ drug via subconjunctival injection;
Latanoprost lowers intraocular pressure for up to 90
days
Carbopol 934 gel-based The clear, stable, and transparent [152]
self-nano emulsifying/ nanoemulsion with spreadability of
6-Gingerol ~234 g cm/sec, an increase of
permeation of skin and solubility;
better topical application; management
of wound healing due to the better
anti-inflammatory properties
Food Nanoemulsions/Essential oils The increase in stability of essential [153]
oils; supports effective delivery
systems
Oil-in-water nanoemulsions/ Physically stable up to more than 1 [154]
Eugenol oil month as an antifungal agent against
Fusarium oxysporum f. sp. vasinfectum
Nanoemulsion/Oregano oil The stability of the oil; inactivating and [155]
controlling the growth of foodborne
bacteria sustainably
Soybean-based Thyme oil was stable for 60 days (21 [156]
polysaccharide emulsions/ °C); thyme oil emulsion possessed a
Thyme oil better antimicrobial property compared
to pure oil of this herb; such emulsions
can act as antimicrobial delivery
systems for increasing the
microbiological safety of products in
the food industry
(continued)
172 A. Izadyari Aghmiuni et al.
Table 2 (continued)
Industry Carrier/entrapped material Function Refs.
Cosmetic Nanostructured lipid carriers Liquid lipids such as fatty acid [157]
based on Glyceryl triglyceride provide a higher solubility
palmito-stearate and fatty acid for idebenone compared to solid lipids;
triglycerides/Idebenone this process can help the stability of
drugs and be led to an increase in their
applications in the cosmetic industry as
the materials of antioxidant, anti-aging,
anti-acne, etc.
Nanostructured lipid carriers Improvement of the water solubility [158]
based on the liquid oil/ and photo-stability of Coenzyme Q10
Coenzyme Q10 in aqueous solution (>80%) than
Coenzyme Q10 itself (<33.5%), so that
photo-stability of this antioxidant was
~65% after 5 months; hence the
nanostructured system can
considerably increase the stability of
materials for various applications
Nanoparticles and The size of nanoparticles and [159]
nanoemulsion based on oleic nano-emulsion were 141–295 nm with
acid/Lornoxicam physically stable for 6 months (various
temperatures) with a release
mechanism of Fickian drug diffusion
along with a high rate of drug
penetration. Such systems can be
effective in the transdermal delivery of
drugs to treat inflammatory and painful
diseases of the skin
Nanostructured lipid carriers These nanostructured lipids possessed [160]
based on high photo-stability (93.9%) for 30
glycerin monostearate, days that free alpha-lipoic acid
glyceryl triacetate/ (42.5%); such systems can act as
Alpha-lipoic acid carriers for loading unstable lipophilic
active ingredients such as antioxidant
materials and increase their
effectiveness in cosmetic productions
Lipid nanocarriers based on Such carriers can be led to a high [161]
stearic acid, Virgin coconut photo-stability (~88%) in 90 days and
oil/Phenylethyl Resorcinol an increase in the retention amount of
Phenylethyl Resorcinol in the skin
Nanostructured lipid carriers/ Such lipid nanocarriers can provide [162]
Podophyllotoxin high dermal targeting efficiency and
safety, as well as improve the
penetration of hydrophobic drugs for
topical application
Plant-Based Biomaterials in Tissue Engineering and Drug Delivery … 173
Another study in this field is aloe vera-based substrates [179, 180]; the reports
of Kallyanashis Paul et al. indicate that aloe vera-based hydrogels can alleviate
maternal birth injuries [181]. Accordingly, aloe vera-alginate hydrogels can act as
an immediate treatment by delivering stem cells and regenerating damaged tissue.
Indeed, the authors stated local injection of such hydrogels (with or without stem
cells) can be significantly effective in repairing birth injuries and act as a therapeutic
strategy for preventing pelvic organ prolapse or healing birth injuries.
Suganya et al. demonstrated that fibers of poly(caprolactone) along with aloe vera
powder (10 wt%) have better hydrophilic properties, higher tensile strength (6.28
MPa), and elastic modulus similar to dermal tissue. Our reports indicated that the
electrospun meshes of aloe vera can increase cell proliferation, secretion of collagen,
and expression of F-actin compared with polycaprolactone-collagen fibers [182].
Based on the studies of Tahmasebi et al., nano-fibrous polymer substrates blended
with aloe vera gel can be also used in hard tissue engineering [183]. The results
illustrated the better biocompatibility of the scaffold when blended with aloe vera.
Moreover, they stated that aloe vera gel possesses osteoinductive potential and can
be used for acceleration of bone regeneration as a bio-implant.
Based on the studies, the use of aloe vera along with other herbal compounds
can accelerate the wound healing process. Hybrid nano-fibrous scaffolds based on
Aaloe vera and curcumin are the sample in this field that was designed by Ezhilarasu
et al. These scaffolds can provide a synergistic effect on the fibroblast behavior and
improve the antimicrobial properties of the scaffold [184].
According to the studies, herbal active ingredients can play an impact in improving
the biological characteristics of substrates, when these bioactive ingredients were
used along with other naturally derived biomaterials. Such that, Zadegan et al. illus-
trated that silk fibroin-base nano-fibers containing Urtica dioica L. (nettle) possess
better water uptake and cellular attachment, as well as higher cellular proliferation
than that silk fibroin nano-fiber [185]. Based on the reports of Ghiyasi et al., scaf-
folds containing Nettle/ZnO-nanoparticle can also provide the synergy effects for
the increase of antibacterial activities. They indicated that the addition of nettle and
ZnO-nanoparticles to poly(caprolactone)-based substrates can increase the tensile
strength up to 2.54 MPa, improve water uptake ability, and promote fibroblast L929
cell proliferation [186].
Hajiali et al. also indicated that sodium alginate/lavender essential oil-based dress-
ings led to a high efficacy for the treatment of UVB-induced dermal burn [187]. Such
dressings are also able to decrease and control the inflammatory responses, induced
in the skin fibroblasts due to UVB exposure.
Some reports illustrate that animal fats can also improve the differentiation and
proliferation of cells; so that, they can be a suitable candidate to fabricate wound
dressings and/or bio-engineered substitutes containing stem cells for skin tissue repair
[188]. There are many studies on this field, some examples have been listed in Table 3.
174 A. Izadyari Aghmiuni et al.
Table 3 (continued)
Herb Scaffolds, substrates, or gels Function Refs.
Xylan Polyvinyl alcohol-based nano-fibers The improvement of [196]
containing xylan fibroblast proliferation and
adhesion, mechanical
properties, and the
biodegradable rate of the
scaffold; the suitable for
skin tissue regeneration
Cissus Polycaprolactone scaffold incorporated The increase of osteogenic [197]
quadrangularis with Hydroxyapatite and Cissus activities and hard tissue
quadrangularis regeneration, improvement
of cell growth, and
mineralization; the
suitable for bone tissue
regeneration
Spinacia Alginate-carboxymethyl cellulose The improvement of [198]
oleracea scaffold containing Spinacia oleracea mechanical stability and
extract biocompatibility of
scaffold, the control of
degradation rate; the
increase in the
proliferation of human
osteosarcoma cells,
suitable for bone tissue
regeneration
Gum tragacanth Electrospun nano-fibers containing Gum The increase of the [199]
and curcumin tragacanth-curcumin antibacterial properties, a
decrease of the epithelial
gap; acceleration in wound
closure, as well as, the
formation of granulation
tissue, hair follicles, and
sweat glands
Arnebia Polycaprolactone-chitosan–polyethylene High potential in burn [200]
euchroma oxide scaffolds containing arnebia wound healing, controlled
euchroma degradation rate;
improvement of swelling
and the mechanical
property; the enhancement
of antibacterial activity,
suitable for applications of
dermal tissue engineering
176 A. Izadyari Aghmiuni et al.
Tissue engineering is a potential strategy to address the demand for tissue substitutes
resulting from trauma and diseases. However, the use of therapeutic agents in combi-
nation with this strategy plays a key role in the creation of new generations of engi-
neered substrates that can regenerate the speed and quality of the formation of new
tissues (Fig. 3). Some of the recent advances resulting from the combination of drug
delivery systems and tissue engineering strategy have been described in this section.
The studies of Karimi et al. are a sample in this field. They reported that hydrogel
dressings containing silk fibroin/polydopamine nanoparticles as a combined method
(drug delivery and tissue engineering) can play an impact on delivering antibiotics
such as Ciprofloxacin [201]. The results of their study indicated such dressings can
preserve the bioactivity of the released antibiotics (>80%) and be led to inhibition of
the bacteria growth. These hydrogel dressings can also support fibroblast cells due
to the existence of various nanoparticles.
Moreover, the electrospun scaffolds have recently received attention in topical
drug delivery. Such an approach not only can significantly decrease the systemic
absorption of the pharmaceutical agents but provides localized therapeutic effects at
lower concentrations, as well.
Core–shell electrospun scaffolds are another sample that developed due to their
therapeutic application. Indeed, according to this strategy, one component of the fiber
or some biologically active molecules can be encapsulated inside another component
[202] and the shell not only controls the release kinetics of the therapeutic agent but
protects the core biomaterial as well [203, 204].
Zhang et al. assessed the protein encapsulation into PCL nanofibers by the elec-
trospinning technique [205]. They reported that the core–shell nanofiber suppresses
the burst release of the drug and be led to sustained release. Notably, the release rate
was highly dependent on the degradation rate of the outer fiber, the shell.
Emulsion electrospinning also is known as a new technique that can create core–
shell fibers via stable polymeric emulsion. Yang et al. indicated that the use of the
emulsion electrospinning method plays a crucial role in using core–shell-based ultra-
fine fibers as the carrier for therapeutic agents, such as proteins [206]. This technique
leads to the creation of porous fibrous mats with bead-free, integrally core–shell
structures.
The core–shell electrospun fibrous also can provide a composite carrier by forming
a polymeric core and an inorganic shell. In this matter, Cui et al. demonstrated that
the incubation of PDLLA electrospun fibers containing calcium nitrate in phosphate
solution form calcium phosphate with a good dispersion on the fibers [207]. They
stated that this core–shell nano-composite fibrous as a scaffold or filler possesses
potential applications in tissue engineering and biomedical.
Grafting the amino groups on the mentioned fibers is another sample in this matter,
performed by Cui et al. via an optimized aminolysis process [208]. The authors found
that the incubation of PDLLA fibers and gelatin containing hydroxyapatite into the
simulated body fluid can provide the core–shell composites with hydroxyapatite
nucleation and growth. They also indicated that the core–shell composite of calcium
phosphate is created through layer-by-layer self-assembly of polymer fibers and
gelatin. It seems that such composites can be effective in bone regeneration and
mineralization [209, 210].
Nowadays, there are many polymers (degradable and non-degradable) in this field
that can act as carriers for local or systemic delivery of drugs by electrospinning
process. Indeed, the advantage of this technique is in maintaining the bioactivities
and molecular structures of the incorporated drugs or biomolecules that can be due
to process conditions.
Hence, it seems that a combination of both tissue engineering and drug delivery
techniques can be a promising approach to treat diseases or damaged tissues through
local, systemic, or implant treatments.
strength is paramount [211] because one of the problems of naturally derived bioma-
terials is rapid biodegradation rate and low stability in an aqueous and physiological
environment.
In this field crosslinking method of the materials as the most common approach
plays a key role in achieving the modified polymeric matrices/structures (Fig. 4).
[212]. Glutaraldehyde is one of these crosslinkers that although led to cytotoxicity
or undesirable changes in some of the biomaterial functions, however, can create
permanent crosslinks between proteins, decrease the antigenicity activity of ECM
biomaterials, and result in the non-biodegradable matrix [211, 213–216]. Hence,
before seeding the cell, synthesized matrices with this crosslinker should be washed.
Notably, such matrices are usually used for permanent implants in the body.
In this regard, the use of other chemical crosslinkers like sodium metaphosphates,
epichlorohydrins, carbodiimides, polyepoxy, and carboxylic acids such as citric acid
can provide suitable mechanical strength and stiffness along with the decrease of
cytotoxic effect, as well as improvement of cell attachment and stability of matrix
[211, 214, 217]. Although, in-vivo responses to such chemically altered matrice are
much different compared to native ECM.
Physical crosslinking is another method for modifying matrice properties and
comprises several steps of heating or compression. Although the advantage of this
method is the absence of chemical residue in the matrix structure, however, physical
crosslinks are not as stable as ones made with other methods [211].
Enzymatic crosslinking also is a natural method to create crosslinks between
polymers. In this matter, transglutaminase (TGase) is known as one of the crosslinkers
used by many organisms [211].
Table 4 The design of new biomaterials by crosslinking
Crosslinking Description/Reason for using the Most widely used Effects Refs.
biomaterials to form crosslinking technique biopolymers and related
various structures crosslinkers
Films and membranes The easiest biomaterial structures for Biomaterials: collagen The increase in tensile strength [218, 220, 221]
application of tissue engineering and Crosslinkers: EDC/NHS, (57%) and modulus (~17-fold),
controlled release/The improvement of acid chlorides as well as a decrease in swelling
mechanical property of substrate and of the structures via controlling
decrease of degradation rate crosslinking conditions
Porous and These structures are known as composite/ Biomaterials: collagen, The interconnected 3D [222–225]
sponge-liked structure hybrid substrates and are used in the hyaluronic acid structures, a decrease in
applications of hard tissue engineering, as Crosslinkers: swelling and enzymatic
well as the design of wound dressings and glutaraldehyde, EDC/NHS degradation without affecting
periodontal matrices/To improve thermal the cell viability
stability and increase the rate of
biodegradation
Biopolymeric The structures with a high capacity to retain Biomaterials: gelatin, The decrease of enzymatic [214, 226–229]
hydrogels water and apply in biomedicine/To avoid collagen, chitosan degradation, an increase of the
the dissolution of hydrogels Crosslinkers: viscoelastic properties and the
glutaraldehyde, carboxylic breaking modulus of the gel,
acid, dextran aldehyde EDC/ and promotion of adhesion and
NHS growth of cell
Plant-Based Biomaterials in Tissue Engineering and Drug Delivery …
Regular fibers via The fibers with different diameters for Biomaterials: collagen, The improvement of [230–232]
wet/melt spinning biomedical applications and tissue herbal proteins like mechanical properties and cell
engineering and soy proteins, gelatin, adhesion
silk fibroin
Crosslinkers: EDC/NHS,
carboxylic acids
(continued)
179
Table 4 (continued)
180
Crosslinking Description/Reason for using the Most widely used Effects Refs.
biomaterials to form crosslinking technique biopolymers and related
various structures crosslinkers
Ultrafine fibers These fibers possess unique properties for Biomaterials: proteins and Promotion of cellular [216, 218, 233–237]
biomedical applications and are very polysaccharides (like starch, attachment and proliferation, a
similar to ECM networks/To mimic the pullan, dextran) decrease of degradation rate
structure and fabrications of ECMs, as well Crosslinkers: EDC/NHS,
as increase tensile strength glutaraldehyde, citric acid,
glycerol, glycerol
phosphate, tripolyphosphate,
tannic acid, acetic acid,
trisodium metaphosphite
Micro/nanoparticles These particles derived from biomaterials Biomaterials: chitosan, The increase in stability, a [238–242]
are used for in-vivo pharmaceutical delivery carboxymethyl pullulan, decrease of particle size and
and other therapeutic agents/The hyaluronic acid, starch, degradation rate, modification
improvement of the performance of alginate, collagen, gelatin of physicochemical properties
nanoparticles Crosslinkers: of particles like low viscosity,
glutaraldehyde, sulfate swelling, controlled release of
anions, sodium drugs
metaphosphate,
tripolyphosphate,
epichlorohydrin,
carbodiimide
A. Izadyari Aghmiuni et al.
Plant-Based Biomaterials in Tissue Engineering and Drug Delivery … 181
Generally, crosslinking methods are used for loading drugs, nutraceuticals, genes,
growth factors, etc., as well as designing electrospun structures, fibers, films, hydro-
gels, 2D/3D scaffolds or substrates, and micro/nano-particles from biomaterials for
in-vitro/in-vivo applications [214, 218, 219]. Table 4 indicates the chemicals and
techniques used for crosslinking biomaterials and medical applications.
Generally, the design of ideal matrices (scaffolds, substrates, substitutes, carriers,
etc.) for applications of tissue engineering or drug delivery systems can play a key
role in the regeneration of damaged tissues and the treatment of diseases. These
matrices should be able to facilitate cell attachment, induce cell proliferation and
differentiation, as well as form extracellular matrix (ECM) deposition, as well as
lead to a controlled release of therapeutic agents in drug delivery systems [243].
In this matter, mechanical stability, modification of function and structure of nano/
micro-carriers, as well as delivering and localizing the biomolecules (cells, growth
factors, and bio-actives into damaged tissue) are the main properties of the designed
matrices.
Some of the design criteria of such matrices for biomedical applications involve
the following:
4.1 Biocompatibility.
4.2 Ability to adhere, differentiate, proliferate, and deliver cells.
4.3 Biodegradability.
4.4 Controlled release.
4.5 Mechanical properties depended on the biomaterial properties.
4.6 Porosity of matrix [the creation of porous structures (>90% along with a
pore size of 300–500 μm) to penetrate and grow cells, and deliver nutrients
for applications of tissue engineering, and nano/micro-size for drug delivery
systems.
4.7 Fabrication capability (easy formation of biomaterials).
Table 5 indicates some of these parameters that can be effective in the design of
matrices.
Table 5 (continued)
Bio-matrices Pore size, porosity Mechanical Morphology Refs.
properties
Gelatin-PCL along with The average fiber The increase in Fibrous scaffolds [250]
nanohydroxyapatite diameter of ~615 mechanical
nm with a pore properties with
size of ~4.7 μm adding PCL and
nanohydroxyapatite
to gelatin
PLAGA-hydroxyapatite Particle size in the Compressive Microsphere [251]
range of 53 μm to strength: 120 MPa; matrices
150 μm along with an elastic (cylindrical and
with pore modulus: 300 MPa tubular)
diameter of
~152–183 μm for
scaffold
Chitosan/PEG4000 -Silk Pore diameters in Increase in 3D scaffolds [8]
fibroin(3%) the range of mechanical
2.3–5.9 μm properties (4.5 MPa
elastic modulus
along with 95%
elongation at
break %)
PCL and hydroxyapatite Group1: Group 1: 3D composite [252]
Pore size: 750 μm compressive scaffold with
Porosity: 70% modulus: 76 MPa internal pores and
Group 2: Group 2: pore
Pore size: 450 μm compressive interconnectivity
Porosity: 60% modulus: 84 MPa to regenerate
bones
Chitosan-based scaffold Pore diameter Water absorption%: 3D hybrid [12]
containing silk fibroin, range of 8–52μm 75.3%; tensile scaffold
collagen, and hyaluronan and porosity of strength: 38.1MPa;
97.5%, modulus: 0.32 MPa
PCL and octa-calcium Fiber diameter Average ultimate Nano-fibrous [253]
phosphate (OCP) distribution: tensile strength: scaffold
0.1–1.7 μm ~4.34 MPa
Average fiber Maximum tensile
diameter: ~0.52 strain: ~182.01%
μm Young’s modulus:
~5.59 MPa
Collagen/chitosan 53.7 μm pore Average elastic Hybrid scaffold [15]
modified by PEG/PCL size, a porosity of modulus of ~18
composite ~92, and water MPa and Average
absorption of tensile
fourfold strength of 13.8
MPa
(continued)
184 A. Izadyari Aghmiuni et al.
Table 5 (continued)
Bio-matrices Pore size, porosity Mechanical Morphology Refs.
properties
Nanohydroxyapatite with Average pore Tensile strength of Composite [254]
PCL and size: 20 μm; fiber 12 MPa, ~269% scaffolds
PCL–Polyethyleneglycol diameter: 0.63 elongation at
(PEG)–PCL μm; porosity: breaking point, and
92% Young’s modulus of
~17.2 MPa along
with 10.41 MPa
storage modulus at
37 °C
quince seed mucilage Pore diameter: 22.8 MPa Elastic 3D hybrid [16]
along with PCL/PEG 16–57 μm; modulus, 13.8 MPa scaffold
copolymer porosity: 92.3% Tensile strength, and
~3 mm Elongation
at the breakpoint
PCL and PLGA along Pore size: 50–100 – Porous scaffolds [255]
with biphasic tricalcium μm
phosphate (BCP)
PCL and cuttlefish Pore dimension: The compressive Composite porous [256]
bone-derived 200–300 μm; modulus of 0.46 scaffold
hydroxyapatite porosity: ~85% PMa
not only can develop novel drugs or therapeutic methods with easier availability and
least/no side effect but also decreases the therapeutic economic burden and health
care problems.
References
1. Izadyari Aghmiuni, A., Heidari Keshel, S.: The Rol of the Extracellular Matrix (ECM)
and ECM-Like Polymeric Substrates in Health and Disease. In: Advances in Medicine and
Biology. pp. 145–175 (2021)
2. Aghmiuni, A.I., Ghadi, A., Azmoun. E., et al.: Electrospun Polymeric Substrates for Tissue
Engineering: Viewpoints on Fabrication, Application, and Challenges. In: Electrospinning -
Material Technology of the Future. InTech, pp. 1–26 (2022)
3. Yang, R., Hou, M., Gao, Y., et al.: Biomineralization-inspired Crystallization of Manganese
Oxide on Silk Fibroin Nanoparticles for in vivo MR/fluorescence Imaging-assisted Tri-modal
Therapy of Cancer. Theranostics 9, 6314–6333 (2019)
4. Pilehvar-Soltanahmadi, Y., Dadashpour, M., Mohajeri, A., et al.: An Overview on Application
of Natural Substances Incorporated with Electrospun Nanofibrous Scaffolds to Development
of Innovative Wound Dressings. Mini Rev. Med. Chem. 18, 414–427 (2018)
5. Cheng, Y., Deng, S., Chen, P., et al.: Polylactic acid (PLA) synthesis and modifications: a
review. Front. Chem. China 4, 259–264 (2009)
6. Shefa, A.A., Sultana, T., Park, M.K., et al.: Curcumin incorporation into an oxidized cellu-
lose nanofiber-polyvinyl alcohol hydrogel system promotes wound healing. Mater. Des. 186,
108313 (2020)
Plant-Based Biomaterials in Tissue Engineering and Drug Delivery … 185
7. Huang, G., Li, F., Zhao, X., et al.: Functional and Biomimetic Materials for Engineering of
the Three-Dimensional Cell Microenvironment. Chem. Rev. 117, 12764–12850 (2017)
8. Aghmiuni, A.I., Keshel, S.H., Rezaei-tavirani, M., et al.: Effect of PEG Molecular Weight and
Volume Ratio of Chitosan/PEG and Silk Fibroin on Physicomechanical Properties of Chitosan/
PEG-SF Scaffold as a Bio-mimetic Substrate in Skin-tissue Engineering Applications. Fibers
Polym 23, 3358–3368 (2022)
9. Sengupta, P., Prasad, B.L.V.: Surface Modification of Polymeric Scaffolds for Tissue
Engineering Applications. Regenerative Engineering and Translational Medicine 4, 75–91
(2018)
10. Türkkan, S., Atila, D., Akdağ, A., et al.: Fabrication of functionalized citrus pectin/silk fibroin
scaffolds for skin tissue engineering. J Biomed Mater Res Part B Appl Biomater 106, 2625–
2635 (2018)
11. Izadyari Aghmiuni A, Heidari Keshel S. The Role of the Extracellular Matrix (ECM) and
ECM-Like Polymeric Substrates in Health and Disease. In: Berhardt L V. (ed) Advances in
Medicine and Biology. NOVA Medicine and Health, 2022, pp. 145–175.
12. Izadyari Aghmiuni, A., Heidari Keshel, S., Sefat, F., et al.: Fabrication of 3D hybrid scaffold
by combination technique of electrospinning-like and freeze-drying to create mechanotrans-
duction signals and mimic extracellular matrix function of skin. Mater. Sci. Eng. C 120,
111752 (2021)
13. Farhadihosseinabadi B, Farahani M, Tayebi T, et al. Amniotic membrane and its epithelial and
mesenchymal stem cells as an appropriate source for skin tissue engineering and regenerative
medicine. Artif Cells, Nanomedicine, Biotechnol 2018; 0: 1–10.
14. Ghiasi, M., Kalhor, N., Tabatabaei Qomi, R., et al.: The effects of synthetic and natural
scaffolds on viability and proliferation of adipose-derived stem cells. Front Life Sci 9, 32–43
(2016)
15. Aghmiuni, A.I., Baei, M.S., Keshel, S.H., et al.: Design of Novel 3D-Scaffold as a Potential
Material to Induct Epidermal-Dermal Keratinocytes of Human-Adipose-Derived Stem Cells
and Promote Fibroblast Cells Proliferation for Skin Regeneration. Fibers Polym 21, 33–44
(2020)
16. Izadyari Aghmiuni A, Heidari Keshel S, Sefat F, et al. Quince seed mucilage-based scaffold
as a smart biological substrate to mimic mechanobiological behavior of skin and promote
fibroblasts proliferation and h-ASCs differentiation into keratinocytes. Int J Biol Macromol.
Epub ahead of print October 2019. https://doi.org/10.1016/j.ijbiomac.2019.10.008.
17. Lin K, Zhang D, Macedo MH, et al. Advanced Collagen-Based Biomaterials for Regenerative
Biomedicine. Adv Funct Mater 2019; 29: 1804943 (1–16).
18. Kumar Giri, T., Thakur, D., Alexander, A., et al.: Alginate based hydrogel as a poten-
tial biopolymeric carrier for drug delivery and cell delivery systems: present status and
applications. Curr. Drug Deliv. 9, 539–555 (2012)
19. Brovold M, Almeida JI, Pla-Palacín I, et al. Naturally-Derived Biomaterials for Tissue Engi-
neering Applications. 2020. Epub ahead of print 2020. https://doi.org/10.1007/978-981-13-
0947-2_23.
20. Manoukian OS, Ahmad A, Marin C, et al. Bioactive nanofiber dressings for wound healing.
In: Wound healing biomaterials. Elsevier, 2016, pp. 451–481.
21. Debele, T.A., Mekuria, S.L., Tsai, H.-C.: Polysaccharide based nanogels in the drug delivery
system: Application as the carrier of pharmaceutical agents. Mater. Sci. Eng. C 68, 964–981
(2016)
22. Ye, G., Li, G., Wang, C., et al.: Extraction and characterization of dextran from Leuconostoc
pseudomesenteroides YB-2 isolated from mango juice. Carbohydr. Polym. 207, 218–223
(2019)
23. Kothari, D., Tingirikari, J.M.R., Goyal, A.: In vitro analysis of dextran from Leuconostoc
mesenteroides NRRL B-1426 for functional food application. Bioact carbohydrates Diet fibre
6, 55–61 (2015)
24. Baruah R, Maina NH, Katina K, et al. Functional food applications of dextran from Weissella
cibaria RBA12 from pummelo (Citrus maxima). Int J Food Microbiol. Epub ahead of print
2016. https://doi.org/10.1016/j.ijfoodmicro.2016.11.012.
186 A. Izadyari Aghmiuni et al.
25. Chen, F., Huang, G., Huang, H.: Preparation and application of dextran and its derivatives as
carriers. Int. J. Biol. Macromol. 145, 827–834 (2020)
26. Sood, A., Gupta, A., Agrawal, G.: Recent advances in polysaccharides based biomaterials for
drug delivery and tissue engineering applications. Carbohydr Polym Technol Appl 2, 100067
(2021)
27. Ha TLB, Quan TM, Vu DN, et al. Naturally Derived Biomaterials: Preparation and
Application. In: Regenerative Medicine and Tissue Engineering. 2013, pp. 247–274.
28. Park, B.K., Kim, M.-M.: Applications of chitin and its derivatives in biological medicine. Int.
J. Mol. Sci. 11, 5152–5164 (2010)
29. Li, Z., Ramay, H.R., Hauch, K.D., et al.: Chitosan–alginate hybrid scaffolds for bone tissue
engineering. Biomaterials 26, 3919–3928 (2005)
30. Venkatesan, J., Kim, S.-K.: Chitosan composites for bone tissue engineering—an overview.
Mar. Drugs 8, 2252–2266 (2010)
31. Eo, M.Y., Fan, H., Cho, Y.J., et al.: Cellulose membrane as a biomaterial: from hydrolysis to
depolymerization with electron beam. Biomater Res 20, 1–13 (2016)
32. Sawatjui, N., Limpaiboon, T., Schrobback, K., et al.: Biomimetic scaffolds and dynamic
compression enhance the properties of chondrocyte-and MSC-based tissue-engineered
cartilage. J. Tissue Eng. Regen. Med. 12, 1220–1229 (2018)
33. Kim, D.K., Kim, J.I., Sim, B.R., et al.: Bioengineered porous composite curcumin/silk
scaffolds for cartilage regeneration. Mater. Sci. Eng. C 78, 571–578 (2017)
34. Fu, L., Zhang, J., Yang, G.: Present status and applications of bacterial cellulose-based
materials for skin tissue repair. Carbohydr. Polym. 92, 1432–1442 (2013)
35. Mano, J.F., Silva, G.A., Azevedo, H.S., et al.: Natural origin biodegradable systems in tissue
engineering and regenerative medicine: present status and some moving trends. J. R. Soc.
Interface. 4, 999–1030 (2007)
36. Kowalska-Ludwicka, K., Cala, J., Grobelski, B., et al.: Special paper–new methods modified
bacterial cellulose tubes for regeneration of damaged peripheral nerves. Arch. Med. Sci. 9,
527–534 (2013)
37. Awad, H.A., Wickham, M.Q., Leddy, H.A., et al.: Chondrogenic differentiation of adipose-
derived adult stem cells in agarose, alginate, and gelatin scaffolds. Biomaterials 25, 3211–3222
(2004)
38. Gao, M., Lu, P., Bednark, B., et al.: Templated agarose scaffolds for the support of motor
axon regeneration into sites of complete spinal cord transection. Biomaterials 34, 1529–1536
(2013)
39. Lynam, D.A., Shahriari, D., Wolf, K.J., et al.: Brain derived neurotrophic factor release from
layer-by-layer coated agarose nerve guidance scaffolds. Acta Biomater. 18, 128–131 (2015)
40. Zarrintaj, P., Bakhshandeh, B., Rezaeian, I., et al.: A novel electroactive agarose-aniline
pentamer platform as a potential candidate for neural tissue engineering. Sci. Rep. 7, 17187
(2017)
41. Han, S., Lee, J.Y., Heo, E.Y., et al.: Implantation of a Matrigel-loaded agarose scaffold
promotes functional regeneration of axons after spinal cord injury in rat. Biochem. Biophys.
Res. Commun. 496, 785–791 (2018)
42. Park, S.-B., Lih, E., Park, K.-S., et al.: Biopolymer-based functional composites for medical
applications. Prog. Polym. Sci. 68, 77–105 (2017)
43. Stanisz, M., Klapiszewski, Ł, Jesionowski, T.: Recent advances in the fabrication and appli-
cation of biopolymer-based micro-and nanostructures: A comprehensive review. Chem. Eng.
J. 397, 125409 (2020)
44. Pires PC, Mascarenhas-melo F, Pedrosa K, et al. Polymer-based biomaterials for pharamaceu-
tical and biomedical applications : A focus on topical drug administration. Eur Polym J; 187.
Epub ahead of print 2023. https://doi.org/10.1016/j.eurpolymj.2023.111868.
45. Niknejad H, Peirovi H, Jorjani M, et al. PROPERTIES OF THE AMNIOTIC MEMBRANE
FOR POTENTIAL USE IN TISSUE.
46. C Echave M, S Burgo L, L Pedraz J, et al. Gelatin as biomaterial for tissue engineering. Curr
Pharm Des 2017; 23: 3567–3584.
Plant-Based Biomaterials in Tissue Engineering and Drug Delivery … 187
47. Chen, J., Liu, W., Zhao, J., et al.: Gelatin microspheres containing calcitonin gene-related
peptide or substance P repair bone defects in osteoporotic rabbits. Biotechnol. Lett. 39, 465–
472 (2017)
48. Gonzalez, D., Ragusa, J., Angeletti, P.C., et al.: Preparation and characterization of func-
tionalized heparin-loaded poly-1-caprolactone fibrous mats to prevent infection with human
papillomaviruses. PLoS ONE 13, e0199925 (2018)
49. Park, J., Kim, J., Hwang, S.R., et al.: Chemical conjugate of low molecular weight heparin
and suramin fragment inhibits tumor growth possibly by blocking VEGF165. Mol. Pharm.
12, 3935–3942 (2015)
50. Wan, X., Li, P., Jin, X., et al.: Poly (ε-caprolactone)/keratin/heparin/VEGF biocomposite mats
for vascular tissue engineering. J Biomed Mater Res Part A 108, 292–300 (2020)
51. Wang, W., Liu, S., Huang, Y., et al.: Biodegradable dextran vesicles for effective haemoglobin
encapsulation. J Mater Chem B 3, 5753–5759 (2015)
52. Anak, D., Davis, S., Parish, C.R.: Heparan sulfate : a ubiquitous glycosaminoglycan with
multiple roles in immunity. Front. Immunol. 4, 1–7 (2013)
53. Lee, C.-T., Kung, P.-H., Lee, Y.-D.: Preparation of poly (vinyl alcohol)-chondroitin sulfate
hydrogel as matrices in tissue engineering. Carbohydr. Polym. 61, 348–354 (2005)
54. Henson, F.M.D., Getgood, A.M.J., Caborn, D.M., et al.: Effect of a solution of hyaluronic acid–
chondroitin sulfate–N-acetyl glucosamine on the repair response of cartilage to single-impact
load damage. Am. J. Vet. Res. 73, 306–312 (2012)
55. Xu L, Tang L, Zhang L. Proteoglycans as miscommunication biomarkers for cancer diagnosis.
1st ed. Elsevier Inc. Epub ahead of print 2019. https://doi.org/10.1016/bs.pmbts.2018.12.003.
56. Weyers, A., Linhardt, R.J.: Neoproteoglycans in tissue engineering. FEBS J. 280, 2511–2522
(2013)
57. Yang, J., Shen, M., Wen, H., et al.: Recent advance in delivery system and tissue engineering
applications of chondroitin sulfate. Carbohydr. Polym. 230, 115650 (2020)
58. Muzzarelli, R.A.A., Greco, F., Busilacchi, A., et al.: Chitosan, hyaluronan and chondroitin
sulfate in tissue engineering for cartilage regeneration: a review. Carbohydr. Polym. 89, 723–
739 (2012)
59. Lafuente-Merchan, M., Ruiz-Alonso, S., Zabala, A., et al.: Chondroitin and dermatan sulfate
bioinks for 3D bioprinting and cartilage regeneration. Macromol. Biosci. 22, 2100435 (2022)
60. Kim, S.S., Kang, M.S., Lee, K.Y., et al.: Therapeutic effects of mesenchymal stem cells and
hyaluronic acid injection on osteochondral defects in rabbits’ knees. Knee Surg Relat Res 24,
164–172 (2012)
61. Migliore, A., Procopio, S.: Effectiveness and utility of hyaluronic acid in osteoarthritis. Clin.
Cases Miner. Bone Metab. 12, 31 (2015)
62. Park, S.H., Seo, J.Y., Park, J.Y., et al.: An injectable, click-crosslinked, cytomodulin-modified
hyaluronic acid hydrogel for cartilage tissue engineering. NPG Asia Mater 11, 30 (2019)
63. Macadam, S.A., Lennox, P.A.: Acellular dermal matrices: use in reconstructive and aesthetic
breast surgery. Can. J. Plast. Surg. 20, 75–89 (2012)
64. Entcheva, E., Bien, H., Yin, L., et al.: Functional cardiac cell constructs on cellulose-based
scaffolding. Biomaterials 25, 5753–5762 (2004)
65. Robb, K.P., Shridhar, A., Flynn, L.E.: Decellularized matrices as cell-instructive scaffolds to
guide tissue-specific regeneration. ACS Biomater. Sci. Eng. 4, 3627–3643 (2017)
66. Granados, M., Morticelli, L., Andriopoulou, S., et al.: Development and characterization of a
porcine mitral valve scaffold for tissue engineering. J. Cardiovasc. Transl. Res. 10, 374–390
(2017)
67. Rana, D., Zreiqat, H., Benkirane-Jessel, N., et al.: Development of decellularized scaffolds
for stem cell-driven tissue engineering. J. Tissue Eng. Regen. Med. 11, 942–965 (2017)
68. Bozuk, M.I., Fearing, N.M., Leggett, P.L.: Use of decellularized human skin to repair
esophageal anastomotic leak in humans. JSLS J Soc Laparoendosc Surg 10, 83 (2006)
69. Lin LM, Lin CC, Chen CL, et al. Effects of an education program on intensive care unit
nurses’ attitudes and behavioral intentions to advocate deceased donor organ donation. In:
Transplantation proceedings. Elsevier, 2014, pp. 1036–1040.
188 A. Izadyari Aghmiuni et al.
70. Moroni, F., Mirabella, T.: Decellularized matrices for cardiovascular tissue engineering. Am
J Stem Cells 3, 1 (2014)
71. Methe, K., Bäckdahl, H., Johansson, B.R., et al.: An alternative approach to decellularize
whole porcine heart. Biores Open Access 3, 327–338 (2014)
72. Taylor, D.A., Sampaio, L.C., Gobin, A.: Building new hearts: a review of trends in cardiac
tissue engineering. Am. J. Transplant. 14, 2448–2459 (2014)
73. Manji, R.A., Menkis, A.H., Ekser, B., et al.: Porcine bioprosthetic heart valves: The next
generation. Am. Heart J. 164, 177–185 (2012)
74. Baptista, P.M., Siddiqui, M.M., Lozier, G., et al.: The use of whole organ decellularization
for the generation of a vascularized liver organoid. Hepatology 53, 604–617 (2011)
75. Jaramillo, M., Yeh, H., Yarmush, M.L., et al.: Decellularized human liver extracellular matrix
(hDLM)-mediated hepatic differentiation of human induced pluripotent stem cells (hIPSCs).
J. Tissue Eng. Regen. Med. 12, e1962–e1973 (2018)
76. Kakabadze, Z., Kakabadze, A., Chakhunashvili, D., et al.: Decellularized human placenta
supports hepatic tissue and allows rescue in acute liver failure. Hepatology 67, 1956–1969
(2018)
77. Baptista PM, Vyas D, Moran E, et al. Human liver bioengineering using a whole liver
decellularized bioscaffold. Organ Regen Methods Protoc 2013; 289–298.
78. Chang, J., DeLillo, N., Jr., Khan, M., et al.: Review of small intestine submucosa extracellular
matrix technology in multiple difficult-to-treat wound types. Wounds 25, 113–120 (2013)
79. Chun, S.Y., Lim, G.J., Kwon, T.G., et al.: Identification and characterization of bioactive
factors in bladder submucosa matrix. Biomaterials 28, 4251–4256 (2007)
80. Xiao, H.H., Gao, Q.G., Zhang, Y., et al.: Vanillic acid exerts oestrogen-like activities in
osteoblast-like UMR 106 cells through MAP kinase (MEK/ERK)-mediated ER signaling
pathway. J. Steroid Biochem. Mol. Biol. 144, 382–391 (2014)
81. Don, M.J., Lin, L.C., Chiou, W.F.: Neobavaisoflavone stimulates osteogenesis via p38-
mediated up-regulation of transcription factors and osteoid genes expression in MC3T3-E1
cells. Phytomedicine 19, 551–561 (2012)
82. Lee, C.H., Huang, Y.L., Liao, J.F., et al.: Ugonin K-stimulated osteogenesis involves estrogen
receptor-dependent activation of non-classical Src signaling pathway and classical pathway.
Eur. J. Pharmacol. 676, 26–33 (2012)
83. Yoon HY, Yun S Il, Kim BY, et al. Poncirin promotes osteoblast differentiation but inhibits
adipocyte differentiation in mesenchymal stem cells. Eur J Pharmacol 2011; 664: 54–59.
84. Pang, W.Y., Wang, X.L., Mok, S.K., et al.: Naringin improves bone properties in ovariec-
tomized mice and exerts oestrogen-like activities in rat osteoblast-like (UMR-106) cells. Br.
J. Pharmacol. 159, 1693–1703 (2010)
85. Chen KY, Lin KC, Chen YS, et al. A novel porous gelatin composite containing naringin
for bone repair. Evidence-based Complement Altern Med; 2013. Epub ahead of print 2013.
https://doi.org/10.1155/2013/283941.
86. Tsuchiya, S., Sugimoto, K., Kamio, H., et al.: Kaempferol-immobilized titanium dioxide
promotes formation of new bone: Effects of loading methods on bone marrow stromal cell
differentiation in vivo and in vitro. Int. J. Nanomedicine 13, 1665–1676 (2018)
87. Carvalho, M.J.N., Ferreira, K.B., Augusti, R., et al.: LEISHMANICIDAL ACTIVITY OF
FLAVONOIDS NATURAL AND SYNTHETIC: A MINIREVIEW. OPEN Sci Res II(2),
266–282 (2022)
88. Zhao, L., Yuan, X., Wang, J., et al.: A review on flavones targeting serine/threonine protein
kinases for potential anticancer drugs. Bioorg. Med. Chem. 27, 677–685 (2019)
89. Zhao, K., Yuan, Y., Lin, B., et al.: LW-215, a newly synthesized flavonoid, exhibits potent
anti-angiogenic activity in vitro and in vivo. Gene 642, 533–541 (2018)
90. Camero, C.M., Germanò, M.P., Rapisarda, A., et al.: Anti-angiogenic activity of iridoids from
Galium tunetanum. Rev Bras Farmacogn 28, 374–377 (2018)
91. Mazidi, M., Katsiki, N., Banach, M.: A higher flavonoid intake is associated with less likeli-
hood of nonalcoholic fatty liver disease: results from a multiethnic study. J. Nutr. Biochem.
65, 66–71 (2019)
Plant-Based Biomaterials in Tissue Engineering and Drug Delivery … 189
92. Khalifa, I., Zhu, W., Li, K., et al.: Polyphenols of mulberry fruits as multifaceted compounds:
Compositions, metabolism, health benefits, and stability—A structural review. J Funct Foods
40, 28–43 (2018)
93. Liao, M.H., Tai, Y.T., Cherng, Y.G., et al.: Genistein induces oestrogen receptor-α gene
expression in osteoblasts through the activation of mitogen-activated protein kinases/NF-κB/
activator protein-1 and promotes cell mineralisation. Br. J. Nutr. 111, 55–63 (2014)
94. Sirtori, C.R., Arnoldi, A., Johnson, S.K.: Phytoestrogens: end of a tale? Ann. Med. 37, 423–438
(2005)
95. Sirotkin, A.V., Harrath, A.H.: Phytoestrogens and their effects. Eur. J. Pharmacol. 741, 230–
236 (2014)
96. Poluzzi, E., Piccinni, C., Raschi, E., et al.: Phytoestrogens in postmenopause: the state of
the art from a chemical, pharmacological and regulatory perspective. Curr. Med. Chem. 21,
417–436 (2014)
97. Sun, H., Li, L., Zhang, A., et al.: Protective effects of sweroside on human MG-63 cells and
rat osteoblasts. Fitoterapia 84, 174–179 (2013)
98. Kim, C., Choi, K.: Potential Roles of Iridoid Glycosides and Their Underlying Mechanisms
against Diverse Cancer Growth and Metastasis : Do They Have an Inhibitory Effect on Cancer
Progression ? Nutrients 13, 2–14 (2021)
99. Kouda, R., Yakushiji, F.: Recent advances in Iridoid chemistry: biosynthesis and chemical
synthesis. Chem. Asian J. 15, 3771–3783 (2020)
100. Castejón ML, Montoya T, Alarcón-de-la-Lastra C, et al. Potential protective role exerted
by secoiridoids from Olea europaea L. in cancer, cardiovascular, neurodegenerative, aging-
related, and immunoinflammatory diseases. Antioxidants 2020; 9: 149.
101. Huang, Q., Shi, J., Gao, B., et al.: Gastrodin: An ancient Chinese herbal medicine as a source
for anti-osteoporosis agents via reducing reactive oxygen species. Bone 73, 132–144 (2015)
102. Yan R, Liu H, Zhang J, et al. Phenolic glycosides and other constituents from the bark of
Magnolia officinalis. J Asian Nat Prod Res 2013; 37–41.
103. Huang, Q., Gao, B., Wang, L., et al.: Ophiopogonin D: A new herbal agent against
osteoporosis. Bone 74, 18–28 (2015)
104. Ivanchina, N.V., Malyarenko, T.V., Kicha, A.A., et al.: A New Steroidal Glycoside Gran-
ulatoside C from the Starfish Choriaster granulatus, Unexpectedly Combining Structural
Features of Polar Steroids from Several Different Marine Invertebrate Phyla Natalia. Nat.
Prod. Commun. 12, 1585–1588 (2017)
105. Liu, M., Kong, J.: The enzymatic biosynthesis of acylated steroidal glycosides and their
cytotoxic activity. Acta Pharm Sin B 8, 981–994 (2018)
106. Ito, Y., Nakashima, Y., Matsuoka, S.: Rice bran extract containing acylated steryl glucoside
fraction decreases elevated blood LDL cholesterol level in obese Japanese men. J Med Investig
62, 80–84 (2015)
107. Suh, K.S., Choi, E.M., Lee, Y.S., et al.: Protective effect of albiflorin against oxidative-stress-
mediated toxicity in osteoblast-like MC3T3-E1 cells. Fitoterapia 89, 33–41 (2013)
108. Zieli, M.: Monoterpenes and Their Derivatives—Recent Development in Biological and
Medical Applications. Int. J. Mol. Sci. 21, 1–38 (2020)
109. Lei, D., Qiu, Z., Qiao, J., et al.: Biotechnology for Biofuels Plasticity engineering of
plant monoterpene synthases and application for microbial production of monoterpenoids.
Biotechnol. Biofuels 14, 1–15 (2021)
110. Muthukumar, T., Aravinthan, A., Sharmila, J., et al.: Collagen/chitosan porous bone tissue
engineering composite scaffold incorporated with Ginseng compound K. Carbohydr. Polym.
152, 566–574 (2016)
111. Wang Z, Jiang R, Wang L, et al. Ginsenoside Rg1 Improves Differentiation by Inhibiting
Senescence of Human Bone Marrow Mesenchymal Stem Cell via GSK-3 β and β-Catenin.
Stem Cells Int; 2020. Epub ahead of print 2020. https://doi.org/10.1155/2020/2365814.
112. Chen, W., Balan, P.: Review of Ginseng Anti-Diabetic Studies. Molecules 24, 1–16 (2019)
113. Borzym-kluczyk, J.N.M.: The role of triterpenes in the management of diabetes mellitus and
its complications. Phytochem. Rev. 14, 675–690 (2015)
190 A. Izadyari Aghmiuni et al.
114. Darshani P, Sen S, Amit S, et al. Anti-viral triterpenes : a review. Springer Netherlands. Epub
ahead of print 2022. https://doi.org/10.1007/s11101-022-09808-1.
115. Ríos, J.: Effects of triterpenes on the immune system. J Ethnopharmacol J 128, 1–14 (2010)
116. Lee, S.U., Park, S.J., Kwak, H.B., et al.: Anabolic activity of ursolic acid in bone: Stimulating
osteoblast differentiation in vitro and inducing new bone formation in vivo. Pharmacol. Res.
58, 290–296 (2008)
117. Ge, Y.W., Lu, J.W., Sun, Z.Y., et al.: Ursolic acid loaded-mesoporous bioglass/chitosan porous
scaffolds as drug delivery system for bone regeneration. Nanomedicine Nanotechnology, Biol
Med 18, 336–346 (2019)
118. Manuscript A. Triterpenoids as potential agents for the chemoprevention and therapy of breast
cancer. NIH Public Access 2011; 980–996.
119. Schneider, C., Langer, R., Loveday, D., et al.: Applications of ethylene vinyl acetate
copolymers (EVA) in drug delivery systems. J. Control. Release 262, 284–295 (2017)
120. Park, K.: Controlled drug delivery systems: past forward and future back. J. Control. Release
190, 3–8 (2014)
121. Yun, Y.H., Lee, B.K., Park, K.: Controlled Drug Delivery: Historical perspective for the next
generation. J. Control. Release 219, 2–7 (2015)
122. Aghmiuni, A.I., Keshel, S.H., Rahmani, A., et al.: Retinal Tissue Engineering: Regenerative
and Drug Delivery Approaches. Curr. Stem Cell Res. Ther. 18, 608–640 (2023)
123. Bertrand, N., Wu, J., Xu, X., et al.: Cancer nanotechnology: the impact of passive and active
targeting in the era of modern cancer biology. Adv. Drug Deliv. Rev. 66, 2–25 (2014)
124. Adeosun SO, Ilomuanya MO, Gbenebor OP, et al. Biomaterials for Drug Delivery: Sources,
Classification, Synthesis, Processing, and Applications. In: Advanced Functional Materials
have. 2020, pp. 1–25.
125. Kamaly, N., Xiao, Z., Valencia, P.M., et al.: Targeted polymeric therapeutic nanoparticles:
design, development and clinical translation. Chem. Soc. Rev. 41, 2971–3010 (2012)
126. Kilicarslan, M., Ilhan, M., Inal, O., et al.: Preparation and evaluation of clindamycin phosphate
loaded chitosan/alginate polyelectrolyte complex film as mucoadhesive drug delivery system
for periodontal therapy. Eur. J. Pharm. Sci. 123, 441–451 (2018)
127. Islan, G.A., Castro, G.R., Islan, G.A., et al.: Tailoring of alginate–gelatin microspheres prop-
erties for oral Ciprofloxacin-controlled release against Pseudomonas aeruginosa. Drug Deliv.
21, 615–625 (2014)
128. Carriers, N., Chen, T., Li, S., et al.: Self-assembly pH-sensitive Chitosan/Alginate Coated
Polyelectrolyte Complexes for Oral Delivery of Insulin. J Microencapsul Micro Nano Carriers
2048, 1–30 (2019)
129. Kumar S, Bhanjana G, Kumar R, et al. Metformin-loaded alginate nanoparticles as an effective
antidiabetic agent for controlled drug release. J Pharm Pharmacol 2016; 1–8.
130. Varadharaj, V., Ramaswamy, A., Sakthivel, R., et al.: Antidiabetic and Antioxidant Activity
of Green Synthesized Starch Nanoparticles : An In Vitro Study. J. Clust. Sci. 3, 1–10 (2019)
131. Rehman A, Mahdi S, Tong Q, et al. International Journal of Biological Macromolecules Role
of peppermint oil in improving the oxidative stability and antioxidant capacity of borage seed
oil-loaded nanoemulsions fabricated by modi fi ed starch. Int J Biol Macromol 2020; 153:
697–707.
132. Usman, M., Mahmood, K., Sajid, M., et al.: In-vivo anti-diabetic and wound healing potential
of chitosan /alginate /maltodextrin /pluronic-based mixed polymeric micelles : Curcumin
therapeutic potential. Int. J. Biol. Macromol. 120, 2418–2430 (2018)
133. Gannon, S., Chu, A.F.: SUDDEN UNEXPECTED DEATH IN EPILEPSY AND LONG QT
SYNDROME. J. Am. Coll. Cardiol. 69, 2137 (2017)
134. Qi X, Yuan Y, Zhang J, et al. Oral administration of salecan-based hydrogels for controlled
insulin delivery. J Agric Food Chem. Epub ahead of print 2018. https://doi.org/10.1021/acs.
jafc.8b02879.
135. Singla R, Soni S, Patial V, et al. In vivo diabetic wound healing potential of nanobiocomposites
containing bamboo cellulose nanocrystals impregnated with silver nanoparticles. Int J Biol
Macromol 2017; 1–40.
Plant-Based Biomaterials in Tissue Engineering and Drug Delivery … 191
136. Abaee, A., Mohammadian, M., Mahdi, S.: Whey and soy protein-based hydrogels and nano-
hydrogels as bioactive delivery systems. Trends Food Sci. Technol. 70, 69–81 (2017)
137. Hou, K., Xu, Y., Cen, K., et al.: Nanoemulsion of cinnamon essential oil Co-emulsified
with hydroxypropyl- β -cyclodextrin and Tween-80: Antibacterial activity, stability and slow
release performance. Food Biosci. 43, 101232 (2021)
138. Cai, S., Thati, S., Bagby, T.R., et al.: Localized doxorubicin chemotherapy with a biopolymeric
nanocarrier improves survival and reduces toxicity in xenografts of human breast cancer. J.
Control. Release 146, 212–218 (2010)
139. Zheng AS, Han J, Jin Z, et al. Dual tumor-targeted multifunctional magnetic hyaluronic
acid micelles for enhanced MR imaging and combined photothermal-chemotherapy. Colloids
Surfaces B Biointerfaces 2018; 1–29.
140. Zhang Y, Ma Y, Xia Y, et al. Synthesis of Silk Fibroin-Insulin Bioconjugates and Their
Characterization and Activities In Vivo. J Biomed Mater Res Part B Appl Biomater 2006;
275–283.
141. Gangrade, A., Mandal, B.B.: Injectable Carbon Nanotube Impregnated Silk Based Multi-
functional Hydrogel for Localized Targeted and On-Demand Anticancer Drug Delivery. ACS
Biomater. Sci. Eng. 5, 2365–2381 (2019)
142. Zhang Y, Xiong GM, Ali Y, et al. Layer-by-layer coated nanoliposomes for oral delivery of
insulin. R cociety Chem 2020; 1–14.
143. Wang, S., Meng, S., Zhou, X., et al.: PH-Responsive and Mucoadhesive Nanoparticles for
Enhanced Oral Insulin Delivery: The Effect of Hyaluronic Acid with Different Molecular
Weights. Pharmaceutics 15, 820 (2023)
144. Acevedo-Guevara, L., Nieto-Suaza, L., Sanchez, L.T., et al.: Development of native and
modified banana starch nanoparticles as vehicles for curcumin. Int. J. Biol. Macromol. 111,
498–504 (2018)
145. Mukhopadhyay, P., Maity, S., Mandal, S., et al.: Preparation, characterization and in vivo
evaluation of pH sensitive, safe quercetin-succinylated chitosan-alginate core-shell-corona
nanoparticle for diabetes treatment. Carbohydr. Polym. 182, 42–51 (2018)
146. Lin YY, Lu SH, Gao R, et al. A Novel Biocompatible Herbal Extract-Loaded Hydrogel for
Acne Treatment and Repair. Oxid Med Cell Longev; 2021. Epub ahead of print 2021. https://
doi.org/10.1155/2021/5598291.
147. Abbas, M.N., Khan, S.A., Sadozai, S.K., et al.: Nanoparticles Loaded Thermoresponsive In
Situ Gel for Ocular Antibiotic Delivery against Bacterial Keratitis. Polymers (Basel) 14, 1–19
(2022)
148. Cheng, Y.H., Ko, Y.C., Chang, Y.F., et al.: Thermosensitive chitosan-gelatin-based hydrogel
containing curcumin-loaded nanoparticles and latanoprost as a dual-drug delivery system for
glaucoma treatment. Exp. Eye Res. 179, 179–187 (2019)
149. Osman, R., Fetih, G., Habib, F.: KETOROLAC TROMETHAMINE LOADED NANOPAR-
TICLES FOR OCULAR DELIVERY: FORMULATION, IN-VITRO AND EX-VIVO
EVALUATION. Bull Pharm Sci Assiut 43, 79–94 (2020)
150. Luo, L.J., Lin, T.Y., Yao, C.H., et al.: Dual-functional gelatin-capped silver nanoparticles for
antibacterial and antiangiogenic treatment of bacterial keratitis. J. Colloid Interface Sci. 536,
112–126 (2019)
151. Natarajan, J.V., Ang, M., Darwitan, A., et al.: Nanomedicine for glaucoma: Liposomes provide
sustained release of latanoprost in the eye. Int. J. Nanomedicine 7, 123–131 (2012)
152. Ahmad N, Khalid MS, Khan MF, et al. Beneficial effects of topical 6-gingerol loaded
nanoemulsion gel for wound and inflammation management with their comparative derma-
tokinetic. J Drug Deliv Sci Technol; 80. Epub ahead of print 2023. https://doi.org/10.1016/j.
jddst.2022.104094.
153. Donsì, F., Ferrari, G.: Essential oil nanoemulsions as antimicrobial agents in food. J.
Biotechnol. 233, 106–120 (2016)
154. Abd-Elsalam KA, Khokhlov AR. Eugenol oil nanoemulsion: antifungal activity against
Fusarium oxysporum f. sp. vasinfectum and phytotoxicity on cottonseeds. Appl Nanosci 2015;
5: 255–265.
192 A. Izadyari Aghmiuni et al.
155. Bhargava, K., Conti, D.S., da Rocha, S.R.P., et al.: Application of an oregano oil nanoemulsion
to the control of foodborne bacteria on fresh lettuce. Food Microbiol. 47, 69–73 (2015)
156. Wu, J.E., Lin, J., Zhong, Q.: Physical and antimicrobial characteristics of thyme oil emulsified
with soluble soybean polysaccharide. Food Hydrocoll 39, 144–150 (2014)
157. Li, B., Ge, Z.Q.: Nanostructured lipid carriers improve skin permeation and chemical stability
of idebenone. AAPS PharmSciTech 13, 276–283 (2012)
158. Wang, J., Wang, H., Zhou, X., et al.: Physicochemical characterization, photo-stability and
cytotoxicity of coenzyme Q10-loading nanostructured lipid carrier. J. Nanosci. Nanotechnol.
12, 2136–2148 (2012)
159. Gönüllü, Ü., Üner, M., Yener, G., et al.: Formulation and characterization of solid lipid
nanoparticles, nanostructured lipid carriers and nanoemulsion of lornoxicam for transdermal
delivery. Acta Pharm. 65, 1–13 (2015)
160. Wang, J., Tang, J., Zhou, X., et al.: Physicochemical characterization, identification and
improved photo-stability of alpha-lipoic acid-loaded nanostructured lipid carrier. Drug Dev.
Ind. Pharm. 40, 201–210 (2014)
161. Fan, H., Liu, G., Huang, Y., et al.: Development of a nanostructured lipid carrier formulation
for increasing photo-stability and water solubility of Phenylethyl Resorcinol. Appl. Surf. Sci.
288, 193–200 (2014)
162. Zhao J, Piao X, Shi X, et al. Podophyllotoxin-loaded nanostructured lipid carriers for skin
targeting: In vitro and in vivo studies. Molecules; 21. Epub ahead of print 2016. https://doi.
org/10.3390/molecules21111549.
163. Yu, Q., Chang, J., Wu, C.: Silicate bioceramics: from soft tissue regeneration to tumor therapy.
J Mater Chem B 7, 5449–5460 (2019)
164. Chaudhari, A.A., Vig, K., Baganizi, D.R., et al.: Future Prospects for Scaffolding Methods
and Biomaterials in Skin Tissue Engineering : A Review. Int. J. Mol. Sci. 17, 1974 (2016)
165. Shafei, S., Foroughi, J., Stevens, L., et al.: Electroactive nanostructured scaffold produced by
controlled deposition of PPy on electrospun PCL fibres. Res. Chem. Intermed. 43, 1235–1251
(2017)
166. Ahmed, L.A.: Stem cells and cardiac repair: alternative and multifactorial approaches. J Regen
Med Tissue Eng 2, 10–7243 (2013)
167. Talebian, S., Mehrali, M., Taebnia, N., et al.: Self-healing hydrogels: the next paradigm shift
in tissue engineering? Adv. Sci. 6, 1801664 (2019)
168. Zhao, Z., Vizetto-Duarte, C., Moay, Z.K., et al.: Composite hydrogels in three-dimensional
in vitro models. Front Bioeng Biotechnol 8, 611 (2020)
169. Wang, Y., Adokoh, C.K., Narain, R.: Recent development and biomedical applications of
self-healing hydrogels. Expert Opin. Drug Deliv. 15, 77–91 (2018)
170. Ramalingam, V., Raja, S., Sundaramahalingam, S., et al.: Chemical fabrication of graphene
oxide nanosheets attenuates biofilm formation of human clinical pathogens. Bioorg. Chem.
83, 326–335 (2019)
171. Izadyari Aghmiuni, A., Heidari Keshel, S., Sefat, F., et al.: Quince seed mucilage-based
scaffold as a smart biological substrate to mimic mechanobiological behavior of skin and
promote fibroblasts proliferation and h-ASCs differentiation into keratinocytes. Int. J. Biol.
Macromol. 142, 668–679 (2020)
172. Dey, S.K., Banerjee, D., Chattapadhyay, S., et al.: Antimicrobial activities of some medicinal
plants of West Bengal. Int J Pharma Bio Sci 1, 1–10 (2010)
173. Omenetto, F.G., Kaplan, D.L.: New opportunities for an ancient material. Science 329, 528–
531 (2010)
174. Kogan, G., Šoltés, L., Stern, R., et al.: Hyaluronic acid: a natural biopolymer with a broad
range of biomedical and industrial applications. Biotechnol. Lett. 29, 17–25 (2006)
175. Volpi, N., Schiller, J., Stern, R., et al.: Role, Metabolism, Chemical Modifications and
Applications of Hyaluronan. Curr. Med. Chem. 16, 1718–1745 (2009)
176. Petrey, A.C., de la Motte, C.A.: Hyaluronan, a crucial regulator of inflammation. Front.
Immunol. 5, 1–14 (2014)
Plant-Based Biomaterials in Tissue Engineering and Drug Delivery … 193
177. Lam, J., Truong, N.F., Segura, T.: Design of cell-matrix interactions in hyaluronic acid
hydrogel scaffolds. Acta Biomater. 10, 1571–1580 (2014)
178. Dicker, K.T., Gurski, L.A., Pradhan-Bhatt, S., et al.: Hyaluronan: A simple polysaccharide
with diverse biological functions. Acta Biomater. 10, 1558–1570 (2014)
179. Darzi, S., Paul, K., Leitan, S., et al.: Immunobiology and application of aloe vera-based
scaffolds in tissue engineering. Int. J. Mol. Sci. 22, 1–19 (2021)
180. Rahman, S., Carter, P., Bhattarai, N.: Aloe vera for tissue engineering applications. J Funct
Biomater 8, 6 (2017)
181. Paul, K., Darzi, S., Del Borgo, M.P., et al.: Vaginal delivery of tissue engineered endome-
trial mesenchymal stem/stromal cells in an aloe vera-alginate hydrogel alleviates maternal
simulated birth injury. Appl. Mater. Today 22, 100890 (2021)
182. Suganya, S., Venugopal, J., Agnes Mary, S., et al.: Aloe vera incorporated biomimetic nanofi-
brous scaffold: a regenerative approach for skin tissue engineering. Iran. Polym. J. 23, 237–248
(2014)
183. Tahmasebi, A., Shapouri Moghadam, A., Enderami, S.E., et al.: Aloe Vera-Derived Gel-
Blended PHBV Nanofibrous Scaffold for Bone Tissue Engineering. ASAIO J. 66, 966–973
(2020)
184. Ezhilarasu, H., Ramalingam, R., Dhand, C., et al.: Biocompatible Aloe vera and Tetracycline
Hydrochloride Loaded Hybrid Nanofibrous Scaffolds for Skin Tissue Engineering. Int. J. Mol.
Sci. 20, 5174 (2019)
185. Zadegan S, Nourmohammadi J, Vahidi B, et al. An investigation into osteogenic differentiation
effects of silk fibroin-nettle (Urtica dioica L.) nanofibers. Int J Biol Macromol 2019; 133:
795–803.
186. Ghiyasi, Y., Salahi, E., Esfahani, H.: Synergy effect of Urtica dioica and ZnO NPs on
microstructure, antibacterial activity and cytotoxicity of electrospun PCL scaffold for wound
dressing application. Mater Today Commun 26, 102163 (2021)
187. Hajiali, H., Summa, M., Russo, D., et al.: Alginate–lavender nanofibers with antibacterial and
anti-inflammatory activity to effectively promote burn healing. J Mater Chem B 4, 1686–1695
(2016)
188. Pilehvar-Soltanahmadi, Y., Nouri, M., Martino, M.M., et al.: Cytoprotection, proliferation and
epidermal differentiation of adipose tissue-derived stem cells on emu oil based electrospun
nanofibrous mat. Exp. Cell Res. 357, 192–201 (2017)
189. Fu, S.Z., Meng, X.H., Fan, J., et al.: Acceleration of dermal wound healing by using elec-
trospun curcumin-loaded poly(ε-caprolactone)-poly(ethylene glycol)-poly(ε-caprolactone)
fibrous mats. J Biomed Mater Res - Part B Appl Biomater 102, 533–542 (2014)
190. Gong, C.Y., Wu, Q.J., Wang, Y.J., et al.: A biodegradable hydrogel system containing curcumin
encapsulated in micelles for cutaneous wound healing. Biomaterials 34, 6377–6387 (2013)
191. Merrell, J.G., Mclaughlin, S.W., Tie, L., et al.: Curcumin Loaded Poly(ε-Caprolactone)
Nanofibers: Diabetic Wound Dressing with Antioxidant and Anti-inflammatory Properties
NIH Public Access Author Manuscript. Clin. Exp. Pharmacol. Physiol. 36, 1149–1156 (2009)
192. Nguyen VC, Nguyen VB, Hsieh MF. Curcumin-loaded chitosan/gelatin composite sponge
for wound healing application. Int J Polym Sci; 2013. Epub ahead of print 2013. https://doi.
org/10.1155/2013/106570.
193. Mitra, T., Manna, P.J., Raja, S.T.K., et al.: Curcumin loaded nano graphene oxide reinforced
fish scale collagen-a 3D scaffold biomaterial for wound healing applications. RSC Adv. 5,
98653–98665 (2015)
194. Venkata V, Reddy S, Kuppusamy G, et al. Curcumin loaded chitosan nanoparticles impreg-
nated into collagen- alginate scaffolds for diabetic wound healing. Int J Biol Macromol. Epub
ahead of print 2016. https://doi.org/10.1016/j.ijbiomac.2016.05.038.
195. Jin, G., Prabhakaran, M.P., Kai, D., et al.: Tissue engineered plant extracts as nanofibrous
wound dressing. Biomaterials 34, 724–734 (2013)
196. Krishnan, R., Rajeswari, R., Venugopal, J., et al.: Polysaccharide nanofibrous scaffolds as a
model for in vitro skin tissue regeneration. J. Mater. Sci. Mater. Med. 23, 1511–1519 (2012)
194 A. Izadyari Aghmiuni et al.
217. Gyawali, D., Nair, P., Zhang, Y., et al.: Citric acid-derived in situ crosslinkable biodegradable
polymers for cell delivery. Biomaterials 31, 9092–9105 (2010)
218. Reddy, N., Reddy, R., Jiang, Q.: Crosslinking biopolymers for biomedical applications. Trends
Biotechnol. 33, 362–369 (2015)
219. Reddy, N., Yang, Y.: Potential of plant proteins for medical applications. Trends Biotechnol.
29, 490–498 (2011)
220. Cao, H., Xu, S.-Y.: EDC/NHS-crosslinked type II collagen-chondroitin sulfate scaffold:
characterization and in vitro evaluation. J. Mater. Sci. Mater. Med. 19, 567–575 (2008)
221. Mitra, T., Sailakshmi, G., Gnanamani, A., et al.: Cross-linking with acid chlorides improves
thermal and mechanical properties of collagen based biopolymer material. Thermochim. Acta
525, 50–55 (2011)
222. Grover, C.N., Cameron, R.E., Best, S.M.: Investigating the morphological, mechanical and
degradation properties of scaffolds comprising collagen, gelatin and elastin for use in soft
tissue engineering. J. Mech. Behav. Biomed. Mater. 10, 62–74 (2012)
223. Sionkowska, A.: Current research on the blends of natural and synthetic polymers as new
biomaterials. Prog. Polym. Sci. 36, 1254–1276 (2011)
224. Bi, L., Cao, Z., Hu, Y., et al.: Effects of different cross-linking conditions on the properties
of genipin-cross-linked chitosan/collagen scaffolds for cartilage tissue engineering. J. Mater.
Sci. Mater. Med. 22, 51–62 (2011)
225. David, G., Cristea, M., Balhui, C., et al.: Effect of cross-linking methods on structure and prop-
erties of poly (ε-caprolactone) stabilized hydrogels containing biopolymers. Biomacromol 13,
2263–2272 (2012)
226. Kuo, C.K., Ma, P.X.: Maintaining dimensions and mechanical properties of ionically
crosslinked alginate hydrogel scaffolds in vitro. J Biomed Mater Res Part A An Off J Soc
Biomater Japanese Soc Biomater Aust Soc Biomater Korean Soc Biomater 84, 899–907
(2008)
227. Duan, X., Sheardown, H.: Crosslinking of collagen with dendrimers. J Biomed Mater Res Part
A An Off J Soc Biomater Japanese Soc Biomater Aust Soc Biomater Korean Soc Biomater
75, 510–518 (2005)
228. Saito, H., Murabayashi, S., Mitamura, Y., et al.: Characterization of alkali-treated collagen
gels prepared by different crosslinkers. J. Mater. Sci. Mater. Med. 19, 1297–1305 (2008)
229. Wang, L., Stegemann, J.P.: Glyoxal crosslinking of cell-seeded chitosan/collagen hydrogels
for bone regeneration. Acta Biomater. 7, 2410–2417 (2011)
230. Reddy, N., Li, Y., Yang, Y.: Alkali-catalyzed low temperature wet crosslinking of plant proteins
using carboxylic acids. Biotechnol. Prog. 25, 139–146 (2009)
231. Reddy, N., Li, Y., Yang, Y.: Wet cross-linking gliadin fibers with citric acid and a quantita-
tive relationship between cross-linking conditions and mechanical properties. J. Agric. Food
Chem. 57, 90–98 (2009)
232. Li, Y., Reddy, N., Yang, Y.: A new crosslinked protein fiber from gliadin and the effect
of crosslinking parameters on its mechanical properties and water stability. Polym. Int. 57,
1174–1181 (2008)
233. Lu, W., Sun, J., Jiang, X.: Recent advances in electrospinning technology and biomedical
applications of electrospun fibers. J Mater Chem B 2, 2369–2380 (2014)
234. Mekhail, M., Wong, K.K.H., Padavan, D.T., et al.: Genipin-cross-linked electrospun collagen
fibers. J. Biomater. Sci. Polym. Ed. 22, 2241–2259 (2011)
235. Kiechel, M.A., Schauer, C.L.: Non-covalent crosslinkers for electrospun chitosan fibers.
Carbohydr. Polym. 95, 123–133 (2013)
236. Shi, L., Le Visage, C., Chew, S.Y.: Long-term stabilization of polysaccharide electrospun
fibres by in situ cross-linking. J. Biomater. Sci. Polym. Ed. 22, 1459–1472 (2011)
237. Jiang, Q., Xu, H., Cai, S., et al.: Ultrafine fibrous gelatin scaffolds with deep cell infiltration
mimicking 3D ECMs for soft tissue repair. J. Mater. Sci. Mater. Med. 25, 1789–1800 (2014)
238. Hauert, S., Bhatia, S.N.: Mechanisms of cooperation in cancer nanomedicine: towards systems
nanotechnology. Trends Biotechnol. 32, 448–455 (2014)
196 A. Izadyari Aghmiuni et al.
239. Reddy, N., Shi, Z., Xu, H., et al.: Development of wheat glutenin nanoparticles and their
biodistribution in mice. J Biomed Mater Res Part A 103, 1653–1658 (2015)
240. Avvakumova, S., Colombo, M., Tortora, P., et al.: Biotechnological approaches toward
nanoparticle biofunctionalization. Trends Biotechnol. 32, 11–20 (2014)
241. Al-Remawi, M.M.A.: Properties of chitosan nanoparticles formed using sulfate anions as
crosslinking bridges. Am. J. Appl. Sci. 9, 1091 (2012)
242. Pichayakorn, W., Boonme, P.: Evaluation of cross-linked chitosan microparticles containing
metronidazole for periodontitis treatment. Mater. Sci. Eng. C 33, 1197–1202 (2013)
243. Hu, J., Ma, P.X.: Nano-fibrous tissue engineering scaffolds capable of growth factor delivery.
Pharm. Res. 28, 1273–1281 (2011)
244. Yin, Y., Ye, F., Cui, J., et al.: Preparation and characterization of macroporous chitosan-gelatin/
β -tricalcium phosphate composite scaffolds for bone tissue engineering. J Biomed Mater Res
- Part A 67, 844–855 (2003)
245. Chen, S., Zhu, L., Wen, W., et al.: Fabrication and Evaluation of 3D Printed Poly(l -lactide)
Scaffold Functionalized with Quercetin-Polydopamine for Bone Tissue Engineering. ACS
Biomater. Sci. Eng. 5, 2506–2518 (2019)
246. Zhu, L, Chen S, Liu K, et al. 3D poly (L-lactide)/chitosan micro/nano fibrous scaffolds
functionalized with quercetin-polydopamine for enhanced osteogenic and anti-inflammatory
activities. Chem Eng J; 391. Epub ahead of print 2020. https://doi.org/10.1016/j.cej.2019.
123524
247. Khan, Y.M., Katti, D.S., Laurencin, C.T.: Novel polymer-synthesized ceramic composite-
based system for bone repair: An in vitro evaluation. J Biomed Mater Res - Part A 69,
728–737 (2004)
248. Kuo, Y.C., Ku, H.F., Rajesh, R.: Chitosan/γ-poly(glutamic acid) scaffolds with surface-
modified albumin, elastin and poly-L-lysine for cartilage tissue engineering. Mater. Sci. Eng.
C 78, 265–277 (2017)
249. Wang, H., Li, Y., Zuo, Y., et al.: Biocompatibility and osteogenesis of biomimetic nano-
hydroxyapatite/polyamide composite scaffolds for bone tissue engineering. Biomaterials 28,
3338–3348 (2007)
250. Gautam, S., Sharma, C., Purohit, S.D., et al.: Gelatin-polycaprolactone-nanohydroxyapatite
electrospun nanocomposite scaffold for bone tissue engineering. Mater. Sci. Eng. C 119,
111588 (2021)
251. Li, M., Mondrinos, M.J., Chen, X., et al.: Elastin Blends for Tissue Engineering Scaffolds. J
Biomed Mater Res Part A 79, 963–973 (2006)
252. Shor, L., Güçeri, S., Wen, X., et al.: Fabrication of three-dimensional polycaprolactone/
hydroxyapatite tissue scaffolds and osteoblast-scaffold interactions in vitro. Biomaterials 28,
5291–5297 (2007)
253. Heydari, Z., Mohebbi-Kalhori, D., Afarani, M.S.: Engineered electrospun polycaprolactone
(PCL)/octacalcium phosphate (OCP) scaffold for bone tissue engineering. Mater. Sci. Eng. C
81, 127–132 (2017)
254. Remya, K.R., Joseph, J., Mani, S., et al.: Nanohydroxyapatite incorporated electrospun poly-
caprolactone/polycaprolactone-polyethyleneglycol-polycaprolactone blend scaffold for bone
tissue engineering applications. J. Biomed. Nanotechnol. 9, 1483–1494 (2013)
255. Thi Hiep, N., Chan Khon, H., Dai Hai, N., et al.: Biocompatibility of PCL/PLGA-BCP porous
scaffold for bone tissue engineering applications. J. Biomater. Sci. Polym. Ed. 28, 864–878
(2017)
256. Kim, B.S., Yang, S.S., Lee, J.: A polycaprolactone/cuttlefish bone-derived hydroxyapatite
composite porous scaffold for bone tissue engineering. J Biomed Mater Res - Part B Appl
Biomater 102, 943–951 (2014)
Plant-Based Biomaterials in Tissue Engineering and Drug Delivery … 197
A. Thakur
University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, India
S. Thakur
Department of Pharmacy, Guru Nanak Dev University, Amritsar, India
S. Sharma (B)
Microbial Type Culture Collection Centre and Gene Bank, CSIR-Institute of Microbial
Technology, Chandigarh, India
e-mail: soniabt918@gmail.com
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 199
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_7
200 A. Thakur et al.
1 Introduction
Nanotechnology is a known branch of science and technology since the last century. A
famous lecture “There’s Plenty of Room at the Bottom” presented by Nobel laureate
Richard P. Feynman, in 1959, first introduced concept of “nanotechnology” [1].
Long before this conceptualization, nano-sized particle were in use in ancient times.
People from ancient Egypt used black hair dye which was believed to be plant based
product, henna but recent research of Egyptian burial sites revealed the hair dye was
not henna instead it was paste of lime, leadoxide and water. They used hair dye paste
in a way that reacts with keratin sulphur to produce lead-sulfide nanoparticles, which
give an even and steady colour to hair [2]. This field of research generally plays at
atomic, molecular scale to create and synthesize novel nano-scale structures, products
and devices. The structures of range 1–100 nm scale are referred to as nanomaterials
and these nanomaterials have some unique properties as compared to their larger
dimensions. Surface effect and quantum effect are two major factors that provide
unique and significant properties to the nanomaterial over their bulk counterparts and
lead to enhanced mechanical, magnetic, catalytical, optical, electronic properties [3,
4]. Nanomaterial due to its small size, has high number of particles per mass unit and
therefore have large surface area as compared to bulk material. Moreover the nano-
materials not only have an increased fraction of atom exposed on the surface but also
have fewer direct neighbours and these differences impact the physiochemical prop-
erties of nanomaterials to a level that the melting point for gold nanoparticles could be
brought to a significantly lower level as compared to large scale bulk gold material [3,
5]. Nanomaterials also display pronounced quantum effect with the decrease in size
of material and these quantum structures contain all the charge carriers (electrons)
confined within a specific physical dimension [6]. With this quantum effect, some
non-magnetic materials in its large dimension would develop magnetic properties
when brought to nanoscale [5]. The quantum effect of nanomaterial brings significant
changes in its electron affinity, which in return directly dictates catalytic properties of
the material. Due to such significant properties of nanomaterials, nanotechnology is
considered multidisciplinary research area that is being explored for diverse purpose;
agriculture, health, environment, catalysis, sensing, electronics, photonics [7, 8].
2 Dimensions of Nanotechnology
There are three dimensions of nanotechnology (Fig. 1); wet, dry, and computa-
tional. Wet nanotechnology, involves live organisms or biological components
like whole cell, secreted enzymes, secreted compounds, to develop nanoproducts.
Dry nanotechnology unlike wet nanotechnology, is related to the production of
nanostructures using inorganic compounds like silicon and carbon. In contrast to
both, computational nanotechnologies do not deal with nanostructure production
but are associated with a very important aspect of development of nanostructure and
Gold Nanoparticles from a Microorganism: A Synthetic Approach 201
3 Types of Nanomaterial
4 Nanoparticles (NPs)
Nanoparticles (NPs) are nano-objects that have all the three dimensions of the struc-
ture along the three axes, measures in nanoscale range, with no significant differences
in the measure of the longest axis and the shortest axis. NPs can vary in their shapes,
sizes, and structures and range from spherical, hollow core, spiral to irregular shape,
cylindrical, tubular and conical [14]. NPs can measure from 1 to 100 nm in size and
NPs lower than 1 nm in dimension, is termed as atom clusters. NPs can be crystalline
or amorphous, uniform or multilayered and either loose or agglomerated [15]. NP is
not just simple molecules but is formed of three or sometimes two layers. The three
layered structure is composed of the outermost layer, i.e., surface layer, a middle shell
layer and the innermost core. Surface layer is generally fabricated with a wide variety
of molecules viz., metal ions, amino acid, peptides, polymers etc. The shell layer is
a protective layer of core that is made of different material as compared to the core,
and the core, which is the central most structure of nanoparticle is referred to as the
NP itself which dictates the property of nanoparticle and thus the application [16].
5 Classification of NPs
NPs can be classified into three major classes, based on the components involved in
synthesis of nanoparticles. These are organic, carbon-based, and inorganic (Fig. 3).
Gold Nanoparticles from a Microorganism: A Synthetic Approach 203
Organic nanoparticles are fabricated with organic compounds like, lipids, carbohy-
drates, proteins and peptides, and other organic compounds. They are non-toxic,
biodegradable and biocompatible in nature and therefore are increasingly being
used in medicine and food [17]. The main types of ONPs are lipid, protein and
polysaccharide-based ONPs (Fig. 3).
Lipid-based systems mostly involve the use of phospholipids and are advantageous
over other encapsulation methods as it uses natural ingredients which can entrap
compounds of different solubility and can be targeted [17]. They are an attraction
in pharmaceutical industry due to its favourable properties like biocompatibility
and biodegradability. Liposomes, lipid nanoparticles, lipid nanotubes and lipid nano
sphere, are some of the lipid-based nanostructures whereas the main lipid-based nano-
encapsulation system includes nanoliposomes, nanoco-chelates, and archaeosomes
[17–19].
204 A. Thakur et al.
CNPs are class of NPs that are made of carbon atoms and it includes carbon
quantum dots, fullerenes, carbon black NPs, etc. [3]. Fullerenes are symmetrical
closed-cage structure of carbon molecules and are formed with an arrangement of
different number of carbon molecules, for instance C60 fullerenes is 60 carbon atoms
arrangement that looks like soccer ball and few other examples include, C70 and
C540 fullerenes. On the other hand, carbon black is arranged in grape-like struc-
ture with bunch of spherical particles fused together whereas carbon quantum dots
are quasi-spherical carbon NPs [3, 23, 24]. Carbon-based NPs is the technology of
putting together physio-chemical properties of sp2 -hybridized carbon bonds and
nano-sized material. Carbon NPs have unique properties like electrical conductivity,
electron affinity, high strength, thermal resistance, and adsorption which lead to its
Gold Nanoparticles from a Microorganism: A Synthetic Approach 205
usage in a wide range of applications viz., bioimaging, delivery carrier for drugs and
biomolecules, energy storage, photovoltaic devices and sensors for monitor microbial
pathogens [25, 26].
Nanoparticles, synthesized using metal (silver, gold, platinum) and metal oxide
(oxide of iron and zinc) are known as inorganic nanoparticles. Inorganic nanoparticles
posses localized surface plasmon resonance, unique thermal, magnetic, optical and
electrical properties which makes them desirable material for development of nanode-
vices with wide variety of application in field of electronics, biomedicines physics and
chemistry [3, 14, 16, 27]. In comparison amongst all metal NPs, nanoparticles fabri-
cated with copper, mercury, silver, platinum, and gold, gold nanoparticles have gained
researchers’ attention all around the globe for its unique optical and physiochemical
properties to investigate and probe gold NPs for different field applications [28].
7 Properties of GNPs
Plasmon is oscillation of electron cloud under the exposure of light [34]. Plasmon
resonance (PR) is an absorption phenomenon of material, which occurs with the
oscillation of electrons produced by metal, on exposure to light [35]. The effect
of incoming electromagnetic wave on the metal depends on the depth of the metal
and generally decreases with increase in depth of the metal. This resonance on the
metal surface is called surface plasmon resonance (SPR) [36]. SPR in small volume of
nanoparticle leads to phenomenon of Localized Surface Plasmon Resonance (LSPR).
LSPR is responsible for increased electromagnetic field and extinction coefficient
and these two effects are further exploited for various biophysical and biochemical
techniques like FRET, SEF, SERS, colorimetric methods, PTT and PDT [37].
Monochromatic incoming light on particle scatters with the same frequency in the
same direction as the incident light. But when there is any adsorbed molecule on
the surface of particles a deviation in the frequency of the scattered light is observed
because of the additional vibration of the adsorbed molecule. This difference in the
incident and scattered photon energies is called Raman scattering (RS) [38]. The
change in energy is due to the energy of the vibrating adsorbed molecule. Raman
image is known to provide a chemical fingerprint that is highly resolved (≈0.1 nm)
vibrational information but with the limitation of low signal sensitivity [39, 40].
Advancement in RS with surface-enhanced Raman Spectroscopy (SERS) is benefi-
cial, which enhances the Raman signal up to 1015 from 106 . This improved signal
has led to its application in bio-sensing, diagnosis and imaging [41]. GNPs are being
used as contrast agents of SERS, due to their optical properties for imaging tumors,
distinguishing tumor cells and normal cells, tumor metabolism and tumor markers.
GNPs due to their local surface Plasmon resonance, show two effects on the signal
intensity of fluorescent molecules these are fluorescence quenching and amplifica-
tion which in turn is dependent on the distance between the fluorescence molecules
and GNPs. Fluorescence resonance energy transfer (FRET) is the main cause of the
GNP’s fluorescence quenching. If the distance is less than 5 nm, the excited fluores-
cent group transfers its energy to the GNPs resulting in fluorescence quenching [42].
Gold Nanoparticles from a Microorganism: A Synthetic Approach 207
The fluorescence quenching and enhancement can be used to study the interaction
among the particles and therefore can be exploited in the field of diagnostics.
7.5 Photosensitization
GNPs have a high molar absorption coefficient, which enhances their colorimeter
sensitivity to the nanomole level. GNPs-based colorimetric assays are thus advan-
tageous as it also takes into account the property of induced plasmon resonance
wherein the change in GNPs- fabricated biomolecules (cytokines, proteins, nucleic
acids, etc) aggregation corresponds to the change in endpoint colour from red to blue,
purple or gray [45].
208 A. Thakur et al.
Gold (Au, atomic number 79), with higher molecular weight shows an increase in
the X-ray absorption coefficient and thus creates minimum damage to normal tissue
with the minimal exposure time for X-ray and thus arose as a promising sensitizer
for radiotherapy [46].
7.8 Functionalization
GNPs can be linked with S-and N-containing groups with a stable interaction which
allows them to be fabricated with a wide variety of molecules and thereby bestow
properties like biocompatibility, target therapy and delivery of drug and biomolecules
(Fig. 4) [47].
The potential application of GNPs is depended upon their size, shape and functional
groups attached (Fig. 4). There is a dramatic difference in colour observed for rod
shaped GNPs than that for spheres. It is because the rod-shaped GNPs exhibit the
plasmon oscillation along two axes of nanorod, one along the short axis and another
along the long axis. In addition to this the resonance further depends strongly on
the nanorod length-to-width ratio [48, 49], thus due to their unique optical proper-
ties, GNPs are subjected to research in bio-imaging, diagnosis and materials science
[50]. To develop GNPs for specific applications, specific methodologies with repro-
ducibility in product formation have been employed and developed in the last couple
of years [51, 52].
9 Synthesis of Nanoparticles
The two major approaches of synthesis of GNPs are top-down and bottom-up
approach (Fig. 5). Top down approach is a subtractive process that involves slicing of
starting bulk material into nanoscale structure. The different strategies of top-down
approach include bulk laser ablation, UV-irradiation and IR-irradiation, ion sput-
tering and aerosol technology [53]. Large sized matter is subjected to these different
strategies to form small particles but in contrast to this, bottom-up approach synthe-
sizes GNPs starting from atomic level using gold of small scale. There are three
distinct ways to synthesizes GNPs in bottom-up approach; physical method, chemical
method and biological method.
This is the commonly used method of synthesis of GNPs and requires the use of chem-
ical entities for gold ions reduction to nanoparticles and for stabilization of nanopar-
ticles. The chemical agents mostly used are sodium citrate, sodium hydroxide,
hydrogen tetrachloroaurate hydrate (HAuCl4 ), SDS, sodium borohydride, cetyl-
trimethylammonium bromide, lithium aluminium hydride and ethyl glycol. The first
report of the chemical synthesis of GNPs by Turkevich et al. [58] showed the synthesis
of GNPs, on reduction of HAuCl4 by sodium citrate in the single-phase aqueous solu-
tion [58]. GNPs are formed by the reduction of Au+3 ions from HAuCl4 to Au0 . It
is formed in clusters which then aggregate into large, polycrystalline form. In other
reported methods, surfactant was used at varying concentrations of the gold-to-thiol
ratio to improve GNPs range of diameters from 1.5 to 20 [59]. Similarly, use of
NH2 OH, ascorbic acid (AA) and CTAB has shown the synthesis of monodisperse
GNPs [60, 61].
Synthesis of GNPs by above-mentioned methods produces a high yield and is
economically viable, but a few disadvantages associated with these methods, such as
the use of hazardous chemical agents, and contaminating chemical precursors, with
high toxicity, limit the use of these methods [62]. Moreover in physical methods,
a large amount of energy and low stability of GNPs, uncontrolled growth of the
crystal, and particle aggregation make the physical approach less advantageous as
well as expenses and thereby impact economic aspects of the production [63]. An
alternative to the physical and chemical method is the biological method which uses
non-toxic agents and does not require the additional use of any external stabilizing
agents and reducing agents. Moreover, it uses renewable materials with minimal
energy expenditure and thus considered ecologically. To overcome these difficulties
linked with physical and chemical approaches, enhanced interest in green synthesis
is observed among researchers and they have investigated the biological production
Gold Nanoparticles from a Microorganism: A Synthetic Approach 211
of GNPs through various biological means. Great diversity in plants and microorgan-
isms provides not only huge options but also allows the production of specific GNPs
using both whole organism and their products. The biomolecules not only synthesize
GNPs but has a positive effect on the application of synthesized GNPs [63].
There is a wide variety of plant species growing on earth amongst which there are
a lot of plants that are well known in use for their medicinal value since ancient
times. Therefore the potential of plants for GNPs has been explored for its econom-
ical as well as environment-friendly methodology. Different parts of plant, leaves,
stem, root and fruit extract have been used to explore the production and applica-
tion of GNPs [62]. Different constituents of plant extracts can synthesize GNPs like
phenolic acids, flavonoids, terpenoids, polyphenols, etc., by reducing metal ions to
corresponding nanoparticles [67]. Flavonoids and gold trivalent ion form an inter-
mediate complex through flavonoid free radical which then results in the oxidization
of flavonoid to Keto-forms and reducing trivalent gold to GNPs [68–71]. Salicin,
a phenolic compound through its hydroxyl group and glucoside bonds participates
in GNP synthesis and the stabilization, terpenoids can also synthesize nanoparticles
by oxidizing itself (aldehyde group’s oxidization to carboxylic acids) and reducing
metal ions [71, 72]. The hydroxyl groups in the polyphenolic compound from Mimosa
tenuiflora extract has been found to transfer an electron to trivalent gold ion, oxidizing
themselves in the process to synthesize GNPs [73]. The role of tannins, alkaloids,
polyols and plant extract sugar in reducing metal ions to nanoparticles is also observed
[74–77]. In addition to the compounds, higher molecular weight proteins can also be
considered important molecules for the synthesis of GNPs [78, 79]. Citrulline from
watermelon is one such example [80].
212 A. Thakur et al.
But the use of microorganisms is advantageous over plants as they are easily cultivable
and can be produced in large amounts in less time, in addition to this they do not
require anything more than nutrients to grow at the temperature, pH and pressure
in which they generally inhabit. The use of bacteria, fungi, yeast and algae has
been reported in the synthesis of metal nanoparticles [64]. Synthesis of nanomate-
rials using microbial extracts or whole culture is actually the merging of the fields
of microbiology, biotechnology, nanotechnology and giving rise to a new area of
research and nano-biotechnology. It is a greener route of synthesis in which varied
species of bacteria and fungi have been studied by researchers and have been reported
[81, 82]. Microorganisms can synthesize nanoparticles using either extracellular or
intracellular mechanisms. Extracellular synthesis basically involves adsorption of the
metal ions at the microbial cell surface, followed by their direct reduction whereas
in intracellular synthesis, metal ions are transported into the cell cytoplasmic region
of the micro-organisms followed by their subsequent reduction of metal ions. The
synthesized GNPs are then derived into the extract [64, 82].
Fungi can withstand high metal ion concentrations as compared to bacteria and
can secrete abundant of extracellular proteins, which can be instrumental in synthe-
size of GNPs. These secreted biomolecules have the ability to reduce soluble metal
ions to their insoluble form and eventually to nanoparticles. The huge diversity of
fungi and their potential to produce novel metal reductases for metal detoxifica-
tion and bioreduction are potential enzymes for nanoparticle synthesis [83]. GNPs
synthesis through fungi can take place in intracellular or extracellular space or by
using fungal extract containing reducing agents. The extracellular mechanism of
GNP formation in fungi relies on the entrapment of Au3+ ions by cell wall proteins,
followed by reduction. According to the report on Verticillium sp., the extracel-
lular interaction of AuCl4+ on the cell surface enzymes with groups having positive
charges, (e.g. lysine) leads to reduction of AuCl4+ to Au0 [84, 85]. In either mech-
anism, NADPH-dependent oxidoreductases are the key enzymes in GNPs biosyn-
thesis and NADH acts as a cofactor of the enzyme [86, 87]. In the intracellular GNP
synthesis mechanism, gold (Au3+ ) ions get diffused into the cell membrane where
the ions are reduced by cystolic redox enzymes [86]. In the intracellular mechanism,
reducing sugars, reductase enzymes such as, glyceraldehyde-3-phosphate dehydro-
genase, ATPase and 3-glucan-binding proteins involved in the energy metabolism of
fungal cells can catalyze the synthesis of nanoparticles [51, 84]. Au3+ diffuses into
the cell membrane through transporter protein and is reduced by cytosolic preva-
lent redox enzymes. Aspergillus fumigates and Aspergillus flavus were reported
to synthesize intracellular GNPs [83]. A glutathione-like compound, phytochelatin
from Candida albicans, has been shown to directly catalyze the GNPs synthesis [88].
Gold ions are reduced to GNP by phytochelatin in the presence of glutathione [88].
Gold Nanoparticles from a Microorganism: A Synthetic Approach 213
Bacteria can be considered a “biofactory” due to their potential for GNPs produc-
tion. The mechanism for the biosynthesis of GNPs reported in bacteria is both
extracellular and intracellular [82, 101]. The bacterial enzyme, NADH reductase
and nitrate reductase were found to play a vital role in the gold ions reduction
214 A. Thakur et al.
10 Applications
GNPs have high electron density which can be actively used for the identifi-
cation and visualization of biological specimens through Transmission Electron
microscopy (TEM), and high-resolution transmission electron microscope (HRTEM)
with advanced systems of digital recording and the processing of images. The prac-
tical uses of GNPs in immune electron spectroscopy are in use for the identifica-
tion of infectious agents and their surface antigens. Scanning probe microscopy,
scanning electron microscopy, and fluorescence microscopy are also used for the
same purpose. All types of GNPs ranging from conventional spherical GNPs to non-
spherical particles, such as nanorods, nanoshells, nanocages, nanostars, and other
types of particles are in use in diagnostics [113]. The biggest challenge to surgeons
in tumor removal is to locate precise boundaries of tumor among healthy tissue.
In this scenario, the removal of any less or more tissue can lead to leaving tumor
tissue behind or removing healthy tissue that can be vital. Magnetic Resonance
Imaging (MRIs) and Computed Tomography (CT) have a limitation of ability to
detect tumors in millimeters or approximately 10 million cells. To overcome this
limitation, photoimaging has emerged as a novel approach where functionalized
GNPs are injected into the tumor which then binds to the cancer cells only and
Gold Nanoparticles from a Microorganism: A Synthetic Approach 215
shines, thereby distinguishing the tumor and healthy cells [114]. Qian et al., [115],
reported the enhanced Raman signal and detection by conjugating GNPs to antibodies
specific to epidermal growth factor receptors (EGFR), present on cancer cells and in
xenograft tumor models and thereby help in efficient diagnosis [115]. Hossain and
co-workers also used GNP-conjugated ITO substrate to enhance the Raman signals
which can be used to differentiate characteristics sub-types of breast cancer cells
whether from the same organ origin or different organs [116]. Lin et al., used GNP-
based SERS for the diagnosis of colorectal tumor markers with enhanced sensitivity
and specificity [117].
216 A. Thakur et al.
High resistance or escape pathogens are one of the serious problems in medicine
as these microorganisms are ever evolving and thus very efficiently develop mech-
anisms to resistance to even the most advanced antibiotics. To overcome the issue
newer technology is always employed to tackle such pathogens. Nanoparticles from
various metals and especially gold are being explored. GNPs have been reported by
many to show antimicrobial activities but antimicrobial activity depends on GNPs
dimension and concentration [64, 118]. Any antimicrobial agent with a novel target
and a broad range of activity both on gram positive and gram negative could be advan-
tageous to tackle a broad range of pathogenic bacteria. The cell wall of bacteria is
negatively charged which has a high affinity for positively charged GNPs. The differ-
ence in GNPs activity towards Gram negative and Gram positive is because of the
cell wall structure difference. GNPs show better activity in Gram-negative bacteria
as they have thinner cell walls which ease GNPs entry into the cell as compared to
thick rigid layers of peptidoglycan in Gram-positive bacteria. Considerable difference
in antibacterial effect has been observed between biosynthesized GNPs and chemi-
cally synthesized GNPs. This enhanced activity in biosynthesized antibacterial GNPs
could be due to the activity of the capping agents, which showed an enhanced syner-
gistic effect [28]. There are many reports showing the antimicrobial activity of GNPs
against bacteria (E. coli, Enterobacter sp., B. subtilis, etc.) and bacterial biofilms.
GNPs showed inhibition of virulence factors i.e. exopolysaccharide production and
thereby weakening the biofilm formation of Proteus sp. [119]. GNPs conjugated with
tetracycline demonstrated greater antibacterial activity as compared to tetracycline
alone against both Gram-positive and Gram-negative bacteria [120].
Like bacteria, pathogenic fungi also pose a serious threat to clinical research, due to
the emerging antibiotic resistance. This new approach to combat these pathogenic
fungi and their associated diseases is a need of the time. Nanotechnology is a
promising area of research that is being explored for antifungal potential against
pathogenic fungi like Aspergillus spp., C. albicans, Trichoderma spp., Penicil-
lium spp., etc [121]. Nanoparticles of gold are being exploited as a potential as anti-
fungal agent and have been observed as a potent agent. GNPs damage cell wall
macromolecules and inhibit β-glucan synthase affecting the cell wall structure and
its integrity and ultimately resulting in cell wall damage. Besides this antifungal
activity of GNPs is also attributed to increased ROS in C. albicans [122].
Gold Nanoparticles from a Microorganism: A Synthetic Approach 217
Viral diseases have gained a lot more attention these days as these are one of the
greatest threats to humans and a great challenge to medical research. This is a very
known fact that for many viral diseases for which there is no cure short drugs or
vaccines developed. Therefore new approach like the use of metal nanoparticles has
come up as a promising technology to fight these dangerous viruses [64]. GNPs are
expected to show antiviral activity as it shows the potential to able to bind a viral
particle and can block the viral receptors that lead to inhibition of the viral cycle.
Moreover, GNPs have a charge density that on binding to viral particles’ surface
brings on a significant change in the membrane potential of the virus particle and
thereby affects viral penetration into the cell [123]. GNPs have been reported as
an effective agent against the measles virus (MeV) [124]. El-Sheikh et al. reported
significant inhibition of Herpes Simplex (HSV-1) using GNPs in a dose-dependent
manner [125].
The GNPs have been found to offer antidiabetic activity in diabetic animals that were
administered with oral GNPs injection and were found to regulate the metabolic
process in addition to the restoration of cholesterol and triglycerides levels [149].
Another study in which GNPs treated rats was found to improve the body weight of
rats by regulating insulin and glycemic levels [150]. Antidiabetic activity of GNPs
synthesized from Fritillaria cirrohosa was observed in preclinical models [151].
GNPs from Ziziphus jujuba showed a decline in lipid peroxidation and oxidative
stress which was fruitful in controlling diabetes [152].
Gold Nanoparticles from a Microorganism: A Synthetic Approach 219
GNPs from Jasminum nervosum leaf extract showed potent larvicidal activity against
filarial and arbovirus vector Culex quinquefasciatus [142, 157]. Larvae and pupae of
the malaria vector A. stephensi and the dengue vector A. aegypti were also reported
to be managed by GNPs [154].
GNPs can be functionalized by binding molecules with various functional groups and
this property provides GNPs with the ability to deliver various therapeutic agents. The
bound molecules attribute to a wide variety of delivery applications of GNPs. GNPs
can be linked through covalently and non-covalently linkages with other materials
[101]. GNPs can be fabricated with compounds that have healing effects, molecules
like PEG and BSA as a coating to provide a surface for attachment of specific cells
[14] for instance PEGylated GNPs selectively minimize uptake of macrophages and
monocytes and thereby prolong their availability of tumor tissue in concentration
[155]. In addition to this, GNPs also offer delivery of biomolecules like DNA, RNA,
peptides, and proteins [156]. Anticancer drugs, doxorubicin and 5-Fluorouracil were
targeted by GNPs for delivery [157, 158].
Tissue Engineering is a field of study that aims to standardize tissue growth and differ-
entiation, mimicking normal environment. Bone Tissue Engineering (BTE) aims at
providing appropriate conditions for osteoblast differentiation and matrix mineraliza-
tion of osteoblastic precursor cell lines [159–161]. Mesenchymal stem cells (MSCs)
are precursor cells of bone tissue that further differentiate into multiple lineages
which are osteoblasts, chondrocytes, and adipocytes [162]. GNPs have been shown
to induce BTE by stimulating osteogenic differentiation. Yi et al. showed the inter-
action of GNPs with cytosolic protein and extracellular matrix, leading to the activa-
tion of the p38 MAPK signaling pathway [163]. Activation of this pathway causes
up-regulation of osteogenic genes and simultaneous downregulation of adipogenic
genes. Zhang et al. showed that GNPs activate the ERK/ MAPK pathway by increased
ERK phosphorylation which is important for osteoblast differentiation and mineral-
ization [164]. The studies showed the significant ability of GNPs and G / HA-NPs
for differentiation of osteogenic cells and the ability to induce differentiation is very
much dependent on the concentration, size and shape of the GNPs [165–167]. GNPs
have been found to promoting not only cell proliferation and differentiation but also
add mechanical strength.
220 A. Thakur et al.
11 Conclusion
Nanoparticles have attracted all spheres of research due to their unique physical
and chemical properties. Nanoparticles can be subdivided on the basis of their
composition into organic nanoparticles, carbon nanoparticles and inorganic nanopar-
ticles. Amongst inorganic nanoparticles, gold is highly chosen metal for nanoparticle
synthesis and characterization these days. Gold nanoparticles (GNPs) can be synthe-
sized by physical methods, chemical methods and biological methods. These two
methods are conventionally used methods but these methods have many disadvan-
tages to the environment and health, like the efflux of hazardous chemicals which
is not only cancer-causing but polluting the environment. Moreover, there is a high
energy requirement in the physical method that renders it cost ineffective. An alterna-
tive method that could prove advantageous to the environment with minimal energy
requirement and no risk to health is biological methods. In biological methods, plants,
and microorganisms can be employed for the synthesis of GNPs. Plant extract from
different parts can be used for synthesis whereas in microorganisms there are two
different ways in which GNPs are synthesized; internal and external. Externally
the gold ions (Au+ ) are trapped by the membrane protein followed by membrane-
reducing enzymes while in the internal method, the gold ions are firstly transported
inside the cell through transported protein and then the cytosolic proteins reduced the
gold ion into GNPs. There is a wide range of microorganisms employed to synthesize
GNPs and researchers have explored a variety of applications of biogenic GNPs.
GNPs have been reported to exhibit antimicrobial, anticancer, antioxidant, calori-
metric detection of Hg, bioimaging and diagnosis, degradation of contaminants,
etc.
References
1. Feynman, R.: There’s plenty of room at the bottom. Engineering and science and science. Sci.
Res. 3(23), 22–36, 1960
2. Walter, P., Welcomme, E., Hallégot, P., Zaluzec, N.J., Deeb, C., Castaing, J., Veyssière, P.,
Bréniaux, R., Lévêque, J.L., Tsoucaris, G.: Early use of PbS nanotechnology for an ancient
hair dyeing formula. Nano Lett. 6(10), 2215–2219 (2006)
3. Joudeh, N., Linke, D.: Nanoparticle classification, physicochemical properties, characteriza-
tion, and applications: a comprehensive review for biologists. J. Nanobiotechnology. 20(1),
262 (2022)
4. Lines, M.G.: Nanomaterials for practical functional uses. J. Alloy. Compd. 449(1–2), 242–245
(2008)
5. Roduner, E.: Size matters: why nanomaterials are different. Chem. Soc. Rev. 35(7), 583–592
(2006)
6. Geoffrion, L.D., Guisbiers, G.: Quantum confinement: size on the grill! J. Phys. Chem. Solids
1(140), 109320 (2020)
7. Bratovcic, A.: Diferent applications of nanomaterials and their impact on the environment.
Int J Mater Sci Eng. 5, 1–7 (2019)
8. Gajanan, K., Tijare, S.N.: Applications of nanomaterials. Materials Today: Proceedings. 5(1),
1093–1096 (2018)
Gold Nanoparticles from a Microorganism: A Synthetic Approach 221
9. Khan, F., Shariq, M., Asif, M., Siddiqui, M.A., Malan, P., Ahmad, F.: Green nanotechnology:
plant-mediated nanoparticle synthesis and application. Nanomaterials 12(4), 673 (2022)
10. Chung, I.M., Park, I., Seung-Hyun, K., Thiruvengadam, M., Rajakumar, G.: Plant-mediated
synthesis of silver nanoparticles: their characteristic properties and therapeutic applications.
Nanoscale Res. Lett. 11(1), 1–4 (2016)
11. Dreaden, E.C., Alkilany, A.M., Huang, X., Murphy, C.J., El-Sayed, M.A.: The golden age:
gold nanoparticles for biomedicine. Chem. Soc. Rev. 41(7), 2740–2779 (2012)
12. Bhatia, S., Bhatia, S.: Nanoparticles types, classification, characterization, fabrication
methods and drug delivery applications. Nat. Polym. Drug Deliv. Syst.: Nanoparticles, Plants,
Algae, 33–93 (2016)
13. Alex, S., Tiwari, A.: Functionalized gold nanoparticles: synthesis, properties and applica-
tions—a review. J. Nanosci. Nanotechnol. 15(3), 1869–1894 (2015)
14. Ealia, S.A., Saravanakumar, M.P.: A review on the classification, characterisation, synthesis
of nanoparticles and their application. In: IOP conference series: materials science and
engineering, vol. 263, no. 3, p. 032019. IOP Publishing, (2017)
15. Machado, S., Pacheco, J.G., Nouws, H.P., Albergaria, J.T., Delerue-Matos, C.: Characteri-
zation of green zero-valent iron nanoparticles produced with tree leaf extracts. Sci. Total.
Environ. 15(533), 76–81 (2015)
16. Khan, I., Saeed, K., Khan, I.: Nanoparticles: Properties, applications and toxicities. Arab. J.
Chem. 12(7), 908–931 (2019)
17. Kumar, R., Lal, S.: Synthesis of organic nanoparticles and their applications in drug delivery
and food nanotechnology: a review. J Nanomater Mol Nanotechnol 3: 4. of. 2014, 11:2.Verma
et al., (2010)
18. Hughes, G.A.: Nanostructure-mediated drug delivery. Nanomedicine: Nanotechnol., Biol.
Med. 1(1), 22–30 (2005)
19. Mozafari, M.R.: Nanoliposomes: preparation and analysis. Liposomes: methods and proto-
cols, vol. 1: Pharmaceutical Nanocarriers., 29–50 (2010)
20. Hong, S., Choi, D.W., Kim, H.N., Park, C.G., Lee, W., Park, H.H.: Protein-based nanoparticles
as drug delivery systems. Pharmaceutics. 12(7), 604 (2020)
21. Swierczewska, M., Han, H.S., Kim, K., Park, J.H., Lee, S.: Polysaccharide-based nanoparticles
for theranostic nanomedicine. Adv. Drug Deliv. Reviews. 1(99), 70–84 (2016)
22. Plucinski, A., Lyu, Z., Schmidt, B.V.: Polysaccharide nanoparticles: From fabrication to
applications. J. Mater. Chem. B. 9(35), 7030–7062 (2021)
23. Yuan, X., Zhang, X., Sun, L., Wei, Y., Wei, X.: Cellular toxicity and immunological effects
of carbon-based nanomaterials. Part. Fibre Toxicol. 16(1), 1–27 (2019)
24. Lu, K.Q., Quan, Q., Zhang, N., Xu, Y.J.: Multifarious roles of carbon quantum dots in
heterogeneous photocatalysis. J. Energy Chem. 25(6), 927–935 (2016)
25. Fang, F., Li, M., Zhang, J., Lee, C.S.: Different strategies for organic nanoparticle preparation
in biomedicine. ACS Materials Letters. 2(5), 531–549 (2020)
26. Mauter, M.S., Elimelech, M.: Environmental applications of carbon-based nanomaterials.
Environ. Sci. & Technology. 42(16), 5843–5859 (2008)
27. Mody, V.V., Siwale, R., Singh, A., Mody, H.R.: Introduction to metallic nanoparticles. J.
Pharm. Bioallied Sci. 2(4), 282 (2010)
28. Nagalingam, M., Kalpana, V.N., Panneerselvam, A.: Biosynthesis, characterization, and
evaluation of bioactivities of leaf extract-mediated biocompatible gold nanoparticles from
Alternanthera bettzickiana. Biotechnology Reports. 1(19), e00268 (2018)
29. Giljohann, D.A., Seferos, D.S., Daniel, W.L., Massich, M.D., Patel, P.C., Mirkin, C.A.: Gold
nanoparticles for biology and medicine. Angew. Chem. Int. Ed. 49(19), 3280–3294 (2010)
30. Hammami, I., Alabdallah, N.M.: Gold nanoparticles: Synthesis properties and applications.
J. King Saud Univ.-Sci. 33(7), 101560 (2021)
31. Barber, D.J., Freestone, I.C.: An investigation of the origin of the colour of the Lycurgus Cup
by analytical transmission electron microscopy. Archaeometry 32(1), 33–45 (1990)
32. Das, M., Shim, K.H., An, S.S., Yi, D.K.: Review on gold nanoparticles and their applications.
Toxicol. Environ. Health Sci. 3, 193–205 (2011)
222 A. Thakur et al.
33. Zhang, J., Mou, L., Jiang, X.: Surface chemistry of gold nanoparticles for health-related
applications. Chem. Sci. 11(4), 923–936 (2020)
34. Maier, S.A., Atwater, H.A.: Plasmonics: Localization and guiding of electromagnetic energy
in metal/dielectric structures. J. Appl. Phys. 98(1), 10 (2005)
35. Jauffred, L., Samadi, A., Klingberg, H., Bendix, P.M., Oddershede, L.B.: Plasmonic heating
of nanostructures. Chem. Rev. 119(13), 8087–8130 (2019)
36. Schuller, J.A., Barnard, E.S., Cai, W., Jun, Y.C., White, J.S., Brongersma, M.L.: Plasmonics
for extreme light concentration and manipulation. Nat. Mater. 9(3), 193–204 (2010)
37. Bai, X., Wang, Y., Song, Z., Feng, Y., Chen, Y., Zhang, D., Feng, L.: The basic properties of
gold nanoparticles and their applications in tumor diagnosis and treatment. Int. J. Mol. Sci.
21(7), 2480 (2020)
38. Shuker, R., Gammon, R.W.: Raman-scattering selection-rule breaking and the density of states
in amorphous materials. Phys. Rev. Lett. 25(4), 222 (1970)
39. Zumbusch, A., Holtom, G.R., Xie, X.S.: Three-dimensional vibrational imaging by coherent
anti-Stokes Raman scattering. Phys. Rev. Lett. 82(20), 4142 (1999)
40. Nie, S., Emory, S.R.: Probing single molecules and single nanoparticles by surface-enhanced
Raman scattering. Science 275(5303), 1102–1106 (1997)
41. Lee, M., Lee, S., Lee, J.H., Lim, H.W., Seong, G.H., Lee, E.K., Chang, S.I., Oh, C.H., Choo,
J.: Highly reproducible immunoassay of cancer markers on a gold-patterned microarray chip
using surface-enhanced Raman scattering imaging. Biosens. Bioelectron. 26(5), 2135–2141
(2011)
42. Yun, C.S., Javier, A., Jennings, T., Fisher, M., Hira, S., Peterson, S., Hopkins, B., Reich, N.O.,
Strouse, G.F.: Nanometal surface energy transfer in optical rulers, breaking the FRET barrier.
J. Am. Chem. Soc. 127(9), 3115–3119 (2005)
43. Huang, X., El-Sayed, I.H., Qian, W., El-Sayed, M.A.: Cancer cell imaging and photothermal
therapy in the near-infrared region by using gold nanorods. J. Am. Chem. Soc. 128(6), 2115–
2120 (2006)
44. Lucky, S.S., Soo, K.C., Zhang, Y.: Nanoparticles in photodynamic therapy. Chem. Rev. 115(4),
1990–2042 (2015)
45. Zhou, W., Gao, X., Liu, D., Chen, X.: Gold nanoparticles for in vitro diagnostics. Chem. Rev.
115(19), 10575–10636 (2015)
46. Seltzer, S.M.: Tables of X-Ray mass attenuation coefficients and mass energy-absorption
coefficients NISTIR 5632. Pramana 72(2), 375–387 (1995)
47. Jans, H., Huo, Q.: Gold nanoparticle-enabled biological and chemical detection and analysis.
Chem. Soc. Rev. 41(7), 2849–2866 (2012)
48. Kuo, W.S., Chang, Y.T., Cho, K.C., Chiu, K.C., Lien, C.H., Yeh, C.S., Chen, S.J.: Gold
nanomaterials conjugated with indocyanine green for dual-modality photodynamic and
photothermal therapy. Biomaterials 33(11), 3270–3278 (2012)
49. Wang, B., Wang, J.H., Liu, Q., Huang, H., Chen, M., Li, K., Li, C., Yu, X.F., Chu, P.K.: Rose-
bengal-conjugated gold nanorods for in vivo photodynamic and photothermal oral cancer
therapies. Biomaterials 35(6), 1954–1966 (2014)
50. Terentyuk, G., Panfilova, E., Khanadeev, V., Chumakov, D., Genina, E., Bashkatov, A., Tuchin,
V., Bucharskaya, A., Maslyakova, G., Khlebtsov, N., Khlebtsov, B.: Gold nanorods with
a hematoporphyrin-loaded silica shell for dual-modality photodynamic and photothermal
treatment of tumors in vivo. Nano Res. 7, 325–337 (2014)
51. Wang, N., Zhao, Z., Lv, Y., Fan, H., Bai, H., Meng, H., Long, Y., Fu, T., Zhang, X., Tan, W.:
Gold nanorod-photosensitizer conjugate with extracellular pH-driven tumor targeting ability
for photothermal/photodynamic therapy. Nano Res. 7, 1291–1301 (2014)
52. Bucharskaya, A., Maslyakova, G., Terentyuk, G., Yakunin, A., Avetisyan, Y., Bibikova,
O., Tuchina, E., Khlebtsov, B., Khlebtsov, N., Tuchin, V.: Towards effective photothermal/
photodynamic treatment using plasmonic gold nanoparticles. Int. J. Mol. Sci. 17(8), 1295
(2016)
53. Khanna, P., Kaur, A., Goyal, D.: Algae-based metallic nanoparticles: Synthesis, characteri-
zation and applications. J. Microbiol. Methods 1(163), 105656 (2019)
Gold Nanoparticles from a Microorganism: A Synthetic Approach 223
54. Correard, F., Maximova, K., Estève, M.A., Villard, C., Roy, M., Al-Kattan, A., Sentis,
M., Gingras, M., Kabashin, A.V., Braguer, D.: Gold nanoparticles prepared by laser abla-
tion in aqueous biocompatible solutions: assessment of safety and biological identity for
nanomedicine applications. Int. J. Nanomedicine 9, 5415 (2014)
55. González-Rubio, G., Guerrero-Martínez, A., Liz-Marzán, L.M.: Reshaping, fragmentation,
and assembly of gold nanoparticles assisted by pulse lasers. Acc. Chem. Res. 49(4), 678–686
(2016)
56. Vinod, M., Jayasree, R.S., Gopchandran, K.G.: Synthesis of pure and biocompatible gold
nanoparticles using laser ablation method for SERS and photothermal applications. Curr.
Appl. Phys. 17(11), 1430–1438 (2017)
57. Riedel, R., Mahr, N., Yao, C., Wu, A., Yang, F., Hampp, N.: Synthesis of gold–silica core–
shell nanoparticles by pulsed laser ablation in liquid and their physico-chemical properties
towards photothermal cancer therapy. Nanoscale 12(5), 3007–3018 (2020)
58. Turkevich, J., Stevenson, P.C., Hillier, J.: A study of the nucleation and growth processes in
the synthesis of colloidal gold. Discuss. Faraday Soc. 11, 55–75 (1951)
59. Leff, D.V., Ohara, P.C., Heath, J.R., Gelbart, W.M.: Thermodynamic control of gold
nanocrystal size: experiment and theory. J. Phys. Chem. 99(18), 7036–7041 (1995)
60. Khan, Z., Singh, T., Hussain, J.I., Hashmi, A.A.: Au (III)–CTAB reduction by ascorbic acid:
Preparation and characterization of gold nanoparticles. Colloids Surf., B 1(104), 11–17 (2013)
61. Ma, X., Wang, M.C., Feng, J., Zhao, X.: Aspect ratio control of Au nanorods via covariation
of the total amount of HAuCl4 and ascorbic acid. J. Alloy. Compd. 15(637), 36–43 (2015)
62. Ramalingam, V.: Multifunctionality of gold nanoparticles: Plausible and convincing proper-
ties. Adv. Colloid Interface Sci. 1(271), 101989 (2019)
63. Iravani, S.: Green synthesis of metal nanoparticles using plants. Green Chem. 13(10), 2638–
2650 (2011)
64. Mikhailova, E.O.: Gold nanoparticles: biosynthesis and potential of biomedical application.
J. Funct. Biomater. 12(4), 70 (2021)
65. Gu, X., Xu, Z., Gu, L., Xu, H., Han, F., Chen, B., Pan, X.: Preparation and antibacterial
properties of gold nanoparticles: A review. Environ. Chem. Lett. 19, 167–187 (2021)
66. Sheny, D.S., Mathew, J., Philip, D.: Phytosynthesis of Au, Ag and Au–Ag bimetallic nanopar-
ticles using aqueous extract and dried leaf of Anacardium occidentale. Spectrochim. Acta Part
A Mol. Biomol. Spectrosc. 79(1), 254–262 (2011)
67. Tamuly, C., Hazarika, M., Bordoloi, M.: Biosynthesis of Au nanoparticles by Gymnocladus
assamicus and its catalytic activity. Mater. Lett. 1(108), 276–279 (2013)
68. Rajan, A., Vilas, V., Philip, D.: Studies on catalytic, antioxidant, antibacterial and anticancer
activities of biogenic gold nanoparticles. J. Mol. Liq. 1(212), 331–339 (2015)
69. Kumar, P.V., Kala, S.M., Prakash, K.S.: Green synthesis of gold nanoparticles using Croton
Caudatus Geisel leaf extract and their biological studies. Mater. Lett. 1(236), 19–22 (2019)
70. El-Borady, O.M., Ayat, M.S., Shabrawy, M.A., Millet, P.: Green synthesis of gold nanoparti-
cles using Parsley leaves extract and their applications as an alternative catalytic, antioxidant,
anticancer, and antibacterial agents. Adv. Powder Technol. 31(10), 4390–4400 (2020)
71. Islam, N.U., Jalil, K., Shahid, M., Rauf, A., Muhammad, N., Khan, A., Shah, M.R., Khan,
M.A.: Green synthesis and biological activities of gold nanoparticles functionalized with Salix
alba. Arab. J. Chem. 12(8), 2914–2925 (2019)
72. Shankar, S.S., Ahmad, A., Pasricha, R., Sastry, M.: Bioreduction of chloroaurate ions by
geranium leaves and its endophytic fungus yields gold nanoparticles of different shapes. J.
Mater. Chem. 13(7), 1822–1826 (2003)
73. Rodríguez-León, E., Rodríguez-Vázquez, B.E., Martínez-Higuera, A., Rodríguez-Beas, C.,
Larios-Rodríguez, E., Navarro, R.E., López-Esparza, R., Iñiguez-Palomares, R.A.: Synthesis
of gold nanoparticles using Mimosa tenuiflora extract, assessments of cytotoxicity, cellular
uptake, and catalysis. Nanoscale Res. Lett. 14(1), 1–6 (2019)
74. Kumar, K.M., Mandal, B.K., Sinha, M., Krishnakumar, V.: Terminalia chebula mediated green
and rapid synthesis of gold nanoparticles. Spectrochim. Acta Part A Mol. Biomol. Spectrosc.
1(86), 490–494 (2012)
224 A. Thakur et al.
75. Balasubramanian, S., Kala, S.M., Pushparaj, T.L.: Biogenic synthesis of gold nanoparticles
using Jasminum auriculatum leaf extract and their catalytic, antimicrobial and anticancer
activities. Journal of Drug Delivery Science and Technology. 1(57), 101620 (2020)
76. Muthuvel, A., Adavallan, K., Balamurugan, K., Krishnakumar, N.: Biosynthesis of gold
nanoparticles using Solanum nigrum leaf extract and screening their free radical scavenging
and antibacterial properties. Biomed. Prev. Nutr. 4(2), 325–332 (2014)
77. Shankar, S.S., Rai, A., Ahmad, A., Sastry, M.: Rapid synthesis of Au, Ag, and bimetallic Au
core–Ag shell nanoparticles using Neem (Azadirachta indica) leaf broth. J. Colloid Interface
Sci. 275(2), 496–502 (2004)
78. Ovais, M., Raza, A., Naz, S., Islam, N.U., Khalil, A.T., Ali, S., Khan, M.A., Shinwari, Z.K.:
Current state and prospects of the phytosynthesized colloidal gold nanoparticles and their
applications in cancer theranostics. Appl. Microbiol. Biotechnol. 101, 3551–3565 (2017)
79. Mukherjee, S., Sushma, V., Patra, S., Barui, A.K., Bhadra, M.P., Sreedhar, B., Patra, C.R.:
Green chemistry approach for the synthesis and stabilization of biocompatible gold nanopar-
ticles and their potential applications in cancer therapy. Nanotechnology 23(45), 455103
(2012)
80. Patra, J.K., Baek, K.H.: Novel green synthesis of gold nanoparticles using Citrullus lanatus
rind and investigation of proteasome inhibitory activity, antibacterial, and antioxidant
potential. Int. J. Nanomed. 10, 7253 (2015)
81. Hosseini, M., Mashreghi, M., Eshghi, H.: Biosynthesis and antibacterial activity of gold
nanoparticles coated with reductase enzymes. Micro & Nano Letters. 11(9), 484–489 (2016)
82. Ghosh, S., Ahmad, R., Zeyaullah, M., Khare, S.K.: Microbial nano-factories: synthesis and
biomedical applications. Front. Chem. 16(9), 626834 (2021)
83. Zhang, X., He, X., Wang, K., Yang, X.: Different active biomolecules involved in biosynthesis
of gold nanoparticles by three fungus species. J. Biomed. Nanotechnol. 7(2), 245–254 (2011)
84. Menon, S., Rajeshkumar, S., Kumar, V.: A review on biogenic synthesis of gold nanoparticles,
characterization, and its applications. Resour.-Effic. Technol. 3(4), 516–527 (2017)
85. Durán, N., Marcato, P.D., Durán, M., Yadav, A., Gade, A., Rai, M.: Mechanistic aspects in
the biogenic synthesis of extracellular metal nanoparticles by peptides, bacteria, fungi, and
plants. Appl. Microbiol. Biotechnol. 90, 1609–1624 (2011)
86. Das, S.K., Dickinson, C., Lafir, F., Brougham, D.F., Marsili, E.: Synthesis, characterization and
catalytic activity of gold nanoparticles biosynthesized with Rhizopus oryzae protein extract.
Green Chem. 14(5), 1322–1334 (2012)
87. Scott, D., Toney, M., Muzikár, M.: Harnessing the mechanism of glutathione reductase for
synthesis of active site bound metallic nanoparticles and electrical connection to electrodes.
J. Am. Chem. Soc. 130(3), 865–874 (2008)
88. Chauhan, A., Zubair, S., Tufail, S., Sherwani, A., Sajid, M., Raman, S.C., Azam, A., Owais,
M.: Fungus-mediated biological synthesis of gold nanoparticles: potential in detection of liver
cancer. Int. J. Nanomed. 12, 2305–2319 (2011)
89. Vetchinkina, E., Loshchinina, E., Kupryashina, M., Burov, A., Pylaev, T., Nikitina, V.: Green
synthesis of nanoparticles with extracellular and intracellular extracts of basidiomycetes.
PeerJ 20(6), e5237 (2018)
90. Apte, M., Girme, G., Bankar, A., RaviKumar, A., Zinjarde, S.: 3, 4-dihydroxy-L-
phenylalanine-derived melanin from Yarrowia lipolytica mediates the synthesis of silver and
gold nanostructures. J. Nanobiotechnology. 11(1), 1–9 (2013)
91. Shi, C., Zhu, N., Cao, Y., Wu, P.: Biosynthesis of gold nanoparticles assisted by the intracellular
protein extract of Pycnoporus sanguineus and its catalysis in degradation of 4-nitroaniline.
Nanoscale Res. Lett. 10, 1–8 (2015)
92. Liu, H., Zhu, N., Luo, D., Li, M., Wu, P., Mo, Y.: Spherical gold nanoparticles (AuNPs)
formation by selected intracellular protein fraction of fungi. Beilstein. Arch. 31(1), 34 (2019)
93. Zhang, X., Qu, Y., Shen, W., Wang, J., Li, H., Zhang, Z., Li, S., Zhou, J.: Biogenic synthesis
of gold nanoparticles by yeast Magnusiomyces ingens LH-F1 for catalytic reduction of
nitrophenols. Colloids Surf., A 20(497), 280–285 (2016)
Gold Nanoparticles from a Microorganism: A Synthetic Approach 225
94. Qu, Y., You, S., Zhang, X., Pei, X., Shen, W., Li, Z., Li, S., Zhang, Z.: Biosynthesis of
gold nanoparticles using cell-free extracts of Magnusiomyces ingens LH-F1 for nitrophenols
reduction. Bioprocess Biosyst. Eng. 41, 359–367 (2018)
95. Zhang, X., Qu, Y., Shen, W., You, S., Pei, X., Li, S., Wang, J., Zhou, J.: Colorimetric response
of biogenetic gold nanoparticles to mercury (II) ions. Colloids Surf., A 5(508), 360–365
(2016)
96. Abdel-Kareem, M.M., Zohri, A.A.: Extracellular mycosynthesis of gold nanoparticles using
Trichoderma hamatum: optimization, characterization and antimicrobial activity. Lett. Appl.
Microbiol. 67(5), 465–475 (2018)
97. Saravanakumar, K., Sathiyaseelan, A., Mariadoss, A.V., Hu, X., Venkatachalam, K., Wang,
M.H.: Nucleolin targeted delivery of aptamer tagged Trichoderma derived crude protein coated
gold nanoparticles for improved cytotoxicity in cancer cells. Process Biochem. 1(102), 325–
332 (2021)
98. Abu-Tahon, M.A., Ghareib, M., Abdallah, W.E.: Environmentally benign rapid biosynthesis
of extracellular gold nanoparticles using Aspergillus flavus and their cytotoxic and catalytic
activities. Process Biochem. 1(95), 1–1 (2020)
99. Clarance, P., Luvankar, B., Sales, J., Khusro, A., Agastian, P., Tack, J.C., Al Khulaifi, M.M., Al-
Shwaiman, H.A., Elgorban, A.M., Syed, A., Kim, H.J.: Green synthesis and characterization
of gold nanoparticles using endophytic fungi Fusarium solani and its in-vitro anticancer and
biomedical applications. Saudi J. Biol. Sciences. 27(2), 706–712 (2020)
100. Hemashekhar, B., Chandrappa, C.P., Govindappa, M., Chandrashekar, N.: Endophytic fungus
Alternaria spp isolated from Rauvolfia tetraphylla root arbitrate synthesis of gold nanopar-
ticles and evaluation of their antibacterial, antioxidant and antimitotic activities. Adv. Nat.
Sci.: Nanosci. Nanotechnol. 10(3), 035010 (2019)
101. Lee, K.X., Shameli, K., Yew, Y.P., Teow, S.Y., Jahangirian, H., Rafiee-Moghaddam, R.,
Webster, T.J.: Recent developments in the facile bio-synthesis of gold nanoparticles (AuNPs)
and their biomedical applications. Int. J. Nanomed. 16, 275–300 (2020)
102. Nangia, Y., Wangoo, N., Goyal, N., Shekhawat, G., Suri, C.R.: A novel bacterial isolate
Stenotrophomonas maltophilia as living factory for synthesis of gold nanoparticles. Microb.
Cell Fact. 8, 1–7 (2009)
103. Khandel, P., Shahi, S.K.: Microbes mediated synthesis of metal nanoparticles: current status
and future prospects. Int J Nanomater Biostruct. 6(1), 1–24 (2016)
104. Issazadeh, K., Jahanpour, N., Pourghorbanali, F., Raeisi, G., Faekhondeh, J.: Heavy metals
resistance by bacterial strains. Ann. Biol. Res. 4(2), 60–63 (2013)
105. Li, J., Webster, T.J., Tian, B.: Functionalized nanomaterial assembling and biosynthesis using
the extremophile Deinococcus radiodurans for multifunctional applications. Small 15(20),
1900600 (2019)
106. Gupta, R., Padmanabhan, P.: Biogenic synthesis and characterization of gold nanoparticles
by a novel marine bacteria Marinobacter algicola: progression from nanospheres to various
geometrical shapes. J. Microbiol. Biotechnol. Food Sci. 6(2021), 732–737 (2021)
107. Sharma, N., Pinnaka, A.K., Raje, M., Fnu, A., Bhattacharyya, M.S., Choudhury, A.R.:
Exploitation of marine bacteria for production of gold nanoparticles. Microb. Cell Fact. 11,
1–6 (2012)
108. Reddy, A.S., Chen, C.Y., Chen, C.C., Jean, J.S., Chen, H.R., Tseng, M.J., Fan, C.W., Wang,
J.C.: Biological synthesis of gold and silver nanoparticles mediated by the bacteria Bacillus
subtilis. J. Nanosci. Nanotechnol. 10(10), 6567–6574 (2010)
109. Rajeshkumar, S.: Anticancer activity of eco-friendly gold nanoparticles against lung and liver
cancer cells. J. Genet. Eng. Biotechnology. 14(1), 195–202 (2016)
110. Patil, M.P., Kang, M.J., Niyonizigiye, I., Singh, A., Kim, J.O., Seo, Y.B., Kim, G.D.: Extra-
cellular synthesis of gold nanoparticles using the marine bacterium Paracoccus haeundaensis
BC74171T and evaluation of their antioxidant activity and antiproliferative effect on normal
and cancer cell lines. Colloids Surf., B 1(183), 110455 (2019)
111. Nandhini, J.T., Ezhilarasan, D., Rajeshkumar, S.: An ecofriendly synthesized gold nanopar-
ticles induces cytotoxicity via apoptosis in HepG2 cells. Environ. Toxicol. 36(1), 24–32
(2021)
226 A. Thakur et al.
112. Shunmugam, R., Balusamy, S.R., Kumar, V., Menon, S., Lakshmi, T., Perumalsamy, H.:
Biosynthesis of gold nanoparticles using marine microbe (Vibrio alginolyticus) and its
anticancer and antioxidant analysis. J. King Saud Univ.-Sci. 33(1), 101260 (2021)
113. Khlebtsov, N., Dykman, L.: Biodistribution and toxicity of engineered gold nanoparticles: a
review of in vitro and in vivo studies. Chem. Soc. Rev. 40(3), 1647–1671 (2011)
114. Singh, P., Pandit, S., Mokkapati, V.R., Garg, A., Ravikumar, V., Mijakovic, I.: Gold nanopar-
ticles in diagnostics and therapeutics for human cancer. Int. J. Mol. Sci. 19(7), 1979
(2018)
115. Qian, X., Nie, S.: Surface-Enhanced raman nanoparticles for in-vivo tumor targeting and spec-
troscopic detection. In: AIP Conference Proceedings, vol. 1267, no. 1, pp. 81–81. American
Institute of Physics. (2010)
116. Hossain, M., Cho, H.Y., Choi, J.W.: Gold nanosphere-deposited substrate for distinguishing of
breast cancer subtypes using surface-enhanced raman spectroscopy. J. Nanosci. Nanotechnol.
16(6), 6299–6303 (2016)
117. Lin, D., Feng, S., Pan, J., Chen, Y., Lin, J., Chen, G., Xie, S., Zeng, H., Chen, R.: Colorectal
cancer detection by gold nanoparticle based surface-enhanced Raman spectroscopy of blood
serum and statistical analysis. Opt. Express 19(14), 13565–13577 (2011)
118. Ahmad, T., Wani, I.A., Manzoor, N., Ahmed, J., Asiri, A.M.: Biosynthesis, structural charac-
terization and antimicrobial activity of gold and silver nanoparticles. Colloids Surf., B 1(107),
227–234 (2013)
119. Samanta, A., Gangopadhyay, R., Ghosh, C.K., Ray, M.: Enhanced photoluminescence from
gold nanoparticle decorated polyaniline nanowire bundles. RSC Adv. 7(44), 27473–27479
(2017)
120. Naimi-Shamel, N., Pourali, P., Dolatabadi, S.: Green synthesis of gold nanoparticles using
Fusarium oxysporum and antibacterial activity of its tetracycline conjugant. J. Mycol. Med.
29(1), 7–13 (2019)
121. Yu, Q., Li, J., Zhang, Y., Wang, Y., Liu, L., Li, M.: Inhibition of gold nanoparticles (AuNPs)
on pathogenic biofilm formation and invasion to host cells. Sci. Rep. 6(1), 26667 (2016)
122. Wani, I.A., Ahmad, T.: Size and shape dependant antifungal activity of gold nanoparticles: a
case study of Candida. Colloids Surf., B 1(101), 162–170 (2013)
123. Lysenko, V., Lozovski, V., Lokshyn, M., Gomeniuk, Y.V., Dorovskih, A., Rusinchuk, N.,
Pankivska, Y., Povnitsa, O., Zagorodnya, S., Tertykh, V., Bolbukh, Y.: Nanoparticles as
antiviral agents against adenoviruses. Adv. Nat. Sci.: Nanosci. Nanotechnology. 9(2), 025021
(2018)
124. Meléndez-Villanueva, M.A., Morán-Santibañez, K., Martínez-Sanmiguel, J.J., Rangel-López,
R., Garza-Navarro, M.A., Rodríguez-Padilla, C., Zarate-Triviño, D.G., Trejo-Ávila, L.M.:
Virucidal activity of gold nanoparticles synthesized by green chemistry using garlic extract.
Viruses 11(12), 1111 (2019)
125. El-Sheekh, M.M., Shabaan, M.T., Hassan, L., Morsi, H.H.: Antiviral activity of algae biosyn-
thesized silver and gold nanoparticles against Herps Simplex (HSV-1) virus in vitro using
cell-line culture technique. Int. J. Environ. Health Res. 32(3), 616–627 (2022)
126. Li, H., Ma, X., Dong, J., Qian, W.: Development of methodology based on the formation
process of gold nanoshells for detecting hydrogen peroxide scavenging activity. Anal. Chem.
81(21), 8916–8922 (2009)
127. Lü, J.M., Lin, P.H., Yao, Q., Chen, C.: Chemical and molecular mechanisms of antioxidants:
experimental approaches and model systems. J. Cell Mol. Med. 14(4), 840–860 (2010)
128. Ahn, S., Singh, P., Jang, M., Kim, Y.J., Castro-Aceituno, V., Simu, S.Y., Kim, Y.J., Yang,
D.C.: Gold nanoflowers synthesized using Acanthopanacis cortex extract inhibit inflammatory
mediators in LPS-induced RAW264. 7 macrophages via NF-κB and AP-1 pathways. Colloids
Surf. B: Biointerfaces. 162, 398–404 (2018)
129. Wang, L., Xu, J., Yan, Y., Liu, H., Karunakaran, T., Li, F.: Green synthesis of gold nanoparticles
from Scutellaria barbata and its anticancer activity in pancreatic cancer cell (PANC-1). Artif.
Cells, Nanomedicine, Biotechnol. 47(1), 1617–1627 (2019)
Gold Nanoparticles from a Microorganism: A Synthetic Approach 227
130. Jafari, M., Rokhbakhsh-Zamin, F., Shakibaie, M., Moshafi, M.H., Ameri, A., Rahimi,
H.R., Forootanfar, H.: Cytotoxic and antibacterial activities of biologically synthesized gold
nanoparticles assisted by Micrococcus yunnanensis strain J2. Biocatal. Agric. Biotechnol.
1(15), 245–253 (2018)
131. Ismail, E.H., Saqer, A.M., Assirey, E., Naqvi, A., Okasha, R.M.: Successful green synthesis
of gold nanoparticles using a Corchorus olitorius extract and their antiproliferative effect in
cancer cells. Int. J. Mol. Sci. 19(9), 2612 (2018)
132. Mmola, M., Roes-Hill, M.L., Durrell, K., Bolton, J.J., Sibuyi, N., Meyer, M.E., Beukes, D.R.,
Antunes, E.: Enhanced antimicrobial and anticancer activity of silver and gold nanoparticles
synthesised using Sargassum incisifolium aqueous extracts. Molecules 21(12), 1633 (2016)
133. Kajani, A.A., Bordbar, A.K., Esfahani, S.H., Razmjou, A.: Gold nanoparticles as potent
anticancer agent: green synthesis, characterization, and in vitro study. RSC Adv. 6(68), 63973–
63983 (2016)
134. Rajkuberan, C., Sudha, K., Sathishkumar, G., Sivaramakrishnan, S.: Antibacterial and cyto-
toxic potential of silver nanoparticles synthesized using latex of Calotropis gigantea L.
Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 5(136), 924–930 (2015)
135. Dhas, T.S., Kumar, V.G., Karthick, V., Govindaraju, K., Narayana, T.S.: Biosynthesis of gold
nanoparticles using Sargassum swartzii and its cytotoxicity effect on HeLa cells. Spectrochim.
Acta Part A Mol. Biomol. Spectrosc. 10(133), 102–106 (2014)
136. Boomi, P., Ganesan, R.M., Poorani, G., Prabu, H.G., Ravikumar, S., Jeyakanthan, J.: Biolog-
ical synergy of greener gold nanoparticles by using Coleus aromaticus leaf extract. Mater.
Sci. Eng., C 1(99), 202–210 (2019)
137. Jeyaraj, M., Arun, R., Sathishkumar, G., MubarakAli, D., Rajesh, M., Sivanandhan, G.,
Kapildev, G., Manickavasagam, M., Thajuddin, N., Ganapathi, A.: An evidence on G2/
M arrest, DNA damage and caspase mediated apoptotic effect of biosynthesized gold
nanoparticles on human cervical carcinoma cells (HeLa). Mater. Res. Bull. 1(52), 15–24
(2014)
138. Alkilany, A.M., Murphy, C.J.: Toxicity and cellular uptake of gold nanoparticles: what we
have learned so far? J. Nanopart. Res. 12, 2313–2333 (2010)
139. Li, L., Zhang, W., Desikan Seshadri, V.D., Cao, G.: Synthesis and characterization of gold
nanoparticles from Marsdenia tenacissima and its anticancer activity of liver cancer HepG2
cells. Artificial cells, nanomedicine, and biotechnology. 47(1), 3029–3036 (2019)
140. Jennifer, M., Maciej, W.: Nanoparticle technology as a double-edged sword: cytotoxic,
genotoxic and epigenetic effects on living cells. J. Biomater. Nanobiotechnol. 4, 53–63 (2013)
141. Ganeshkumar, M., Sathishkumar, M., Ponrasu, T., Dinesh, M.G., Suguna, L.: Spontaneous
ultra fast synthesis of gold nanoparticles using Punica granatum for cancer targeted drug
delivery. Colloids Surf., B 1(106), 208–216 (2013)
142. Patra, S., Mukherjee, S., Barui, A.K., Ganguly, A., Sreedhar, B., Patra, C.R.: Green synthesis,
characterization of gold and silver nanoparticles and their potential application for cancer
therapeutics. Mater. Sci. Eng., C 1(53), 298–309 (2015)
143. Vemuri, S.K., Banala, R.R., Mukherjee, S., Uppula, P., Subbaiah, G.P., AV, G.R., Malarvilli,
T.: Novel biosynthesized gold nanoparticles as anti-cancer agents against breast cancer:
Synthesis, biological evaluation, molecular modelling studies. Mater. Sci. Eng.: C. 99,
417–429 (2019)
144. Kuno, T., Tsukamoto, T., Hara, A., Tanaka, T.: Cancer chemoprevention through the induction
of apoptosis by natural compounds. J. Biophys. Checm. 3(02), 156–173 (2012)
145. Qian, L., Su, W., Wang, Y., Dang, M., Zhang, W., Wang, C.: Synthesis and characterization
of gold nanoparticles from aqueous leaf extract of Alternanthera sessilis and its anticancer
activity on cervical cancer cells (HeLa). Artif. Cells, Nanomedicine, Biotechnol. 47(1), 1173–
1180 (2019)
146. Ramachandran, R., Krishnaraj, C., Sivakumar, A.S., Prasannakumar, P., Kumar, V.A., Shim,
K.S., Song, C.G., Yun, S.I.: Anticancer activity of biologically synthesized silver and gold
nanoparticles on mouse myoblast cancer cells and their toxicity against embryonic zebrafish.
Mater. Sci. Eng., C 1(73), 674–683 (2017)
228 A. Thakur et al.
147. Selim, M.E., Hendi, A.A.: Gold nanoparticles induce apoptosis in MCF-7 human breast cancer
cells. Asian Pac. J. Cancer Prev. 13(4), 1617–1620 (2012)
148. Baharara, J., Ramezani, T., Divsalar, A., Mousavi, M., Seyedarabi, A.: Induction of apoptosis
by green synthesized gold nanoparticles through activation of caspase-3 and 9 in human
cervical cancer cells. Avicenna J. Med. Biotechnol. 8(2), 75 (2016)
149. Venkatachalam, M., Govindaraju, K., Sadiq, A.M., Tamilselvan, S., Kumar, V.G., Singaravelu,
G.: Functionalization of gold nanoparticles as antidiabetic nanomaterial. Spectrochim. Acta
Part A Mol. Biomol. Spectrosc. 1(116), 331–338 (2013)
150. Daisy, P., Saipriya, K.: Biochemical analysis of Cassia fistula aqueous extract and phytochem-
ically synthesized gold nanoparticles as hypoglycemic treatment for diabetes mellitus. Int. J.
Nanomed. 7, 1189–1202 (2012)
151. Guo, Y., Jiang, N., Zhang, L., Yin, M.: Green synthesis of gold nanoparticles from Fritillaria
cirrhosa and its anti-diabetic activity on Streptozotocin induced rats. Arab. J. Chem. 13(4),
5096–5106 (2020)
152. Javanshir, R., Honarmand, M., Hosseini, M., Hemmati, M.: Anti-dyslipidemic properties
of green gold nanoparticle: Improvement in oxidative antioxidative balance and associated
atherogenicity and insulin resistance. Clinical Phytoscience. 6(1), 1 (2020)
153. Lallawmawma, H., Sathishkumar, G., Sarathbabu, S., Ghatak, S., Sivaramakrishnan, S.,
Gurusubramanian, G., Kumar, N.S.: Synthesis of silver and gold nanoparticles using
Jasminum nervosum leaf extract and its larvicidal activity against filarial and arboviral vector
Culex quinquefasciatus Say (Diptera: Culicidae). Environ. Sci. Pollut. Res. 22, 17753–17768
(2015)
154. Murugan, K., Benelli, G., Panneerselvam, C., Subramaniam, J., Jeyalalitha, T., Dinesh, D.,
Nicoletti, M., Hwang, J.S., Suresh, U., Madhiyazhagan, P.: Cymbopogon citratus-synthesized
gold nanoparticles boost the predation efficiency of copepod Mesocyclops aspericornis against
malaria and dengue mosquitoes. Exp. Parasitol. 1(153), 129–138 (2015)
155. Tiwari, P.M., Vig, K., Dennis, V.A., Singh, S.R.: Functionalized gold nanoparticles and their
biomedical applications. Nanomaterials 1(1), 31–63 (2011)
156. Kah, J.C., Wong, K.Y., Neoh, K.G., Song, J.H., Fu, J.W., Mhaisalkar, S., Olivo, M., Sheppard,
C.J.: Critical parameters in the pegylation of gold nanoshells for biomedical applications: an
in vitro macrophage study. J. Drug Target. 17(3), 181–193 (2009)
157. Mukherjee, S., Sau, S., Madhuri, D., Bollu, V.S., Madhusudana, K., Sreedhar, B., Banerjee,
R., Patra, C.R.: Green synthesis and characterization of monodispersed gold nanoparticles:
toxicity study, delivery of doxorubicin and its bio-distribution in mouse model. J. Biomed.
Nanotechnol. 12(1), 165–181 (2016)
158. Thirumurugan, A., Blessy, V., Karthikeyan, M.: Comparative study on doxorubicin loaded
metallic nanoparticles in drug delivery against MCF-7 cell line. In: Applications of
nanomaterials, pp. 303–313. Woodhead Publishing, (2018)
159. Li, H., Pan, S., Xia, P., Chang, Y., Fu, C., Kong, W., Yu, Z., Wang, K., Yang, X., Qi, Z.:
Advances in the application of gold nanoparticles in bone tissue engineering. J. Biol. Eng.
14, 1–5 (2020)
160. He, Y., Li, Y., Chen, G., Wei, C., Zhang, X., Zeng, B., Yi, C., Wang, C., Yu, D.: Concentration-
dependent cellular behavior and osteogenic differentiation effect induced in bone marrow
mesenchymal stem cells treated with magnetic graphene oxide. J. Biomed. Mater. Res., Part
A 108(1), 50–60 (2020)
161. Ouchi, T., Nakagawa, T.: Mesenchymal stem cell-based tissue regeneration therapies for
periodontitis. Regenerative Therapy. 1(14), 72–78 (2020)
162. Dong, Y., Hong, M., Dai, R., Wu, H., Zhu, P.: Engineered bioactive nanoparticles incorporated
biofunctionalized ECM/silk proteins based cardiac patches combined with MSCs for the repair
of myocardial infarction: In vitro and in vivo evaluations. Sci. Total Environ. 10(707), 135976
(2020)
163. Yi, C., Liu, D., Fong, C.-C., Zhang, J., Yang, M.: Gold nanoparticles promote Osteogenic
differentiation of Mesenchymal stem cells through p38 MAPK pathway. ACS Nano 4, 6439–
6448 (2010)
Gold Nanoparticles from a Microorganism: A Synthetic Approach 229
164. Zhang, D., Liu, D., Zhang, J., Fong, C., Yang, M.: Gold nanoparticles stimulate differentiation
and mineralization of primary osteoblasts through the ERK/ MAPK signaling pathway. Mater.
Sci. Eng. 42, 70–77 (2014)
165. Heo, D.N., et al.: Inhibition of osteoclast differentiation by gold nanoparticles functionalized
with Cyclodextrin Curcumin complexes. ACS Nano 8, 12049–12062 (2014)
166. Conners, C.M., Bhethanabotla, V.R., Gupta, V.K.: Concentration-dependent effects of alen-
dronate and pamidronate functionalized gold nanoparticles on osteoclast and osteoblast
viability. J. Biomed. Mater. Res. B Appl. Biomater. 105, 21–29 (2017)
167. Li, J., Chen, Y., Yang, Y., Kawazoe, N., Chen, G.: Sub-10 nm gold nanoparticles promote
adipogenesis and inhibit osteogenesis of mesenchymal stem cells. J Mater Chem B. 5, 1353–
1362 (2017)
Sonia Sharma did her Ph.D. from Guru Nana Dev University
Amritsar in 2021. She studied chitinases and chitin extracting
enzymes and explored these enzymes as an alternate to chemical
methods. She is currently working as Senior Project Associate
at MTTC, CSIR Institute of Microbial Technology, Chandigarh.
She is experienced in microbiology and intends to explore the
rich Indian microbial diversity for its abundant bio-molecules
reserves, targeting therapeutics as well as industrial important
enzymes. She has published seven research articles and one book
chapter in SCOPUS indexed journals.
Applications of Biomaterials
Nanostructured Biomaterials in Drug
Delivery
Abstract In recent years, there has been a significant advancement in the manu-
facturing of biomaterial-based nanoagents with distinctive characteristics such as
biodegradability, biocompatibility, high efficacy, safety, and physicochemical prop-
erties. In particular, research on the performance and drug delivery mechanisms based
on polymeric nanostructures is gaining interest in the field of health nanotechnolo-
gies. This chapter discusses all aspects of biomaterials-based nanoagent preparation
methods, characterizations, and drug delivery mechanisms. Furthermore, there has
been great attention to the preparation and performance of thermo-sensitive nanogels,
biopolymeric nanofibers, green nanoparticles, and nanocomposites for health care
systems. The unique advantages, present constraints, and potential future of green
nanocarriers and target delivery of chemotherapeutic nanodrugs were highlighted,
along with some novel approaches for nanoplatforms.
Abbreviations
AA Acrylic acid
AC Acetyl curcumin
CAP Cellulose acetate phthalate
CMC Carboxymethylcellulose
İ. M. Kahyaoğlu
Faculty of Science, Department of Chemistry, Ondokuz Mayıs University, 55139 Samsun, Turkey
E. C. Aytar · A. Durmaz
Faculty of Science, Department of Biology, Ondokuz Mayıs University, 55139 Samsun, Turkey
e-mail: alper.durmaz@windowslive.com
S. Karakuş (B)
Faculty of Engineering, Department of Chemistry, Istanbul University-Cerrahpaşa, 34320
Istanbul, Turkey
e-mail: selcan@iuc.edu.tr
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 233
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_8
234 İ. M. Kahyaoğlu et al.
1 Bio-Based Nanostructures
efficiency in crop production fields. This is especially important in the case of heavy
metal pollution, which can cause harm to both the ecosystem and crop production
[3, 4].
Biopolymers are utilized in bio-nanostructures due to the fact that they are natu-
rally occurring macromolecules that originate from renewable sources such as lignin,
flowers, plants, leaves, cellulose, animals, shells, algeas, and microorganisms. The
functional characteristics of the polysaccharide vary depending on its composition,
structure, and type. For instance, carbohydrates are made up of simple glucose
molecules and can have a wide range of shapes, such as linear, branching, or cyclical,
but proteins are formed up of amino acids with a complicated three-dimensional
architecture that affects their functionality. Moreover, biopolymers’ characteristics
can be altered chemically or by processing techniques to improve their compatibility
for a number of different uses. Recently, there has been growing interest among
researchers in these biostructures, mainly due to their abundant availability from
natural resources. For example, lignin is a byproduct of paper production and is one
of the polymeric materials found in plants. These materials, which make up approx-
imately 70–80% of a plant’s cell walls, are ubiquitous in the environment and are
integral components of biomaterials. Lignin has been the subject of numerous studies
investigating its potential benefits in bioscience. In a 2020 study, Lee et al. demon-
strated the pharmaceutical applications of lignin by creating nanomaterials with a
size of approximately 225 nm from rice-derived lignin. The study also identified for
the first time lignin’s inherent ability to provide UV protection, resulting in it being
a viable option for usage as sunscreen [5]. In another study, Adil et al. demonstrated
238 İ. M. Kahyaoğlu et al.
the antibacterial effect of the proposed nanostructure by mixing lignin with inor-
ganic additives to produce silver nanoparticles with a diameter ranging from 100 to
300 nm [6]. Cellulose, the most abundant material in nature, has many -OH groups
that make it a promising source for the production of beneficial nanomaterials. In
their study, Zhang et al. [7] synthesized nanostructures using cellulose fibers from
economically bleached wood cellulose pulp. The resulting spherical shapes were 50–
150 nm in particle size and were evaluated for temperature changes and in vitro drug
(5-fluorouracil, 5-FU) release effectivenes. The researchers found that the nanos-
tructures exhibited a rapid 5-FU release pattern, with up to 60% of the drug released
within the first hour [7]. An organic linear biopolymer consisting of 1,4-glycosidic
linkages connecting N-acetylglucosamine units is termed chitin. It is typically found
in the skeletons of insects, and fungus and also in their cell membranes. Chitin
has a broad spectrum of potential benefits in a number of sectors, including health-
care, agricultural, and environmental engineering because of its special qualities,
including biological compatibility and biodegradability. In their research, Maskur
et al. [8] reinforced chitin derived from crab shells using polylactic acid to develop
nanomaterials with a size distribution of 90 nm. They carried out the experiment using
varying chitin ratios to determine durability and revealed that the durability reduced
with increasing chitin concentration [8]. Because of chitin-chitin aggregating, which
results in poor distribution of the nanostructures within the matrix phase, additional
chitin nanofiber adding reduces the structural rigidity performance of the nanos-
tructure. Chitin derived from shrimp shells is a polymer with antibacterial and anti-
oxidant capabilities that are consistent with epithelial cells, suggesting potential
therapeutic potential, according to Coltelli et al. [9]. A natural, renewable alkaline
carbohydrate with high hydrating and adsorb characteristics is chitosan, a deacety-
lated derivative of chitin. Chitin is a naturally occurring substance that is plentiful but
has less favorable best possible way than cellulose. Chitosan is also non-toxic, has
really no known negative impacts, and is thought to be acceptable for use in a wide
range of fields. Because to its advantageous characteristics, including bioavailability,
nontoxicity, and chemical stability, chitosan has received considerable attention in
a number of sectors. Chitosan’s usage in both food and drugs has been authorized
by the United States Food and Drug Administration (FDA), which has confirmed
the nano/micromaterial’s safety. Its certification enables the use of chitosan as a
thickening agent, food stabilizer, component of nutritional supplements, and phar-
maceutical delivery mechanism. Chitosan has the potential to be a major potential
in a multitude of sectors, including agriculture, drugs, and nanotechnology, thanks
to its broad application possibilities [10]. While chitosan offers numerous benefits,
its ability to mitigate environmental contaminants is particularly noteworthy and has
garnered significant research attention.
In 2022, Ansari et al. synthesized novel chitosan/Fe2 O3 /NiFe2 O4 nanostructures
and investigated their antibacterial effects. Their results showed that the adsorbent
effectively eliminated 96.8% of the methylene green (MG) after 120 min of stirring
the solution, which decreased to 77.22% after 240 min. These findings highlight
the impressive ability of these nanoparticles to remove pollutants using methylene
green adsorption [11]. Since the initially developed polymers were developed for
Nanostructured Biomaterials in Drug Delivery 239
The two most used methods for delivering pharmaceuticals specifically targeted
to cancer patients are systemic chemotherapy administered in nanoagents and the
designed release of treatment drugs to the patient’s tissues and organs. Although
chemotherapy drugs are an effective approach to cancer treatment, they can also be
potentially dangerous to healthy cells in the body. Encapsulation is a method used to
modify how these drugs are absorbed and employed by the human body. Surrounding
them in nanocarriers can improve the bio-distribution, permeability, toxic effects,
tolerability, and acceptance of the pharmaceuticals, increasing their therapeutic value
and reducing any adverse reactions that may occur. By minimizing damage to healthy
tissues, this strategy can make it easier to provide greater doses of chemotherapy treat-
ments directly to cancerous cells. The structural affinities of nanostructures to organs
and pathogens are crucial for effective interactions between nanomaterials and living
organisms. When nanomaterials mimic the morphology and form of cellular organ-
isms, they are more likely to be recognized and interacted with, leading to enhanced
effectiveness and personalized solutions. The targeted administration and combina-
tion of pharmaceuticals within a person’s cells and tissues, as well as the monitoring of
illness concentrations in problematic fluids such as blood, saliva, and organs like the
240 İ. M. Kahyaoğlu et al.
kidneys and liver, can all be accomplished through the application of nanomaterials.
Pharmaceuticals efficiency can be improved while generating fewer risks to health
when they are encapsulated in nano-based compounds. Furthermore, the application
of nanotechnology can facilitate the development of pharmaceutical nanoformula-
tions that function as local analgesics by evading the human body’s immune system,
including the phagocytic system, due to their unique morphology. This prevents
potential side effects that could diminish positive health outcomes and results in
a long-lasting effect. Various factors, such as poor colloidal stability, unfavorable
hydrodynamic and pharmacodynamic properties, slow dissolution rate, and inade-
quate distribution, along with concerns about toxicity and stability, can contribute to
an inadequate pharmacological response and minimal drug release at the receptor site.
Despite the numerous neurobiological, pharmacological, and therapeutic challenges
associated with intravenous administration, healthcare professionals and scientists
are turning to nano-based carriers as a viable solution. Currently, various methods
are utilized for manufacturing nanostructured biomaterials, including chemical vapor
deposition (CVD), biological method, physical/mechanical method, self-assembly
method, sol–gel method, ultrasound waves-based method, sono-solvo method, and
hydrothermal method. These methods use different approaches to produce the desired
nanomaterials, which can vary in terms of effectiveness, complexity, and cost. The
synthesis of nanostructured biomaterials cannot be accomplished with a single tech-
nique. The optimum strategy is chosen by filtering the alternatives according to
factors such as the final product, the starting raw material, the cost of the procedure,
the application, and the quantity being manufactured. The procedures for nanopar-
ticle synthesis are constantly evolving as new tactics and raw materials are discovered
due to the advancement of innovation.
The ultrasonic method is a technique that uses high-frequency sound waves to create
mechanical vibrations in a liquid solution. These vibrations can induce a range of
physical and chemical effects, such as generating cavitation microstreaming, acoustic
cavitation, and acoustic flowing which can lead to the formation of nanostructures
of various shapes and sizes. The ultrasonic method uses a high-frequency ultra-
sonic instrument to generate sound waves that propagate through a liquid solu-
tion. The sound waves create alternating high/low pressure regions, causing liquid
molecules to vibrate and collide with each other. This leads to the creation of cavita-
tion bubbles, that can collapse violently, generating high temperatures and pressures.
These extreme conditions can trigger a range of chemical reactions, including the
formation of, nanowires, nanospheres, nanotubes, metal/metal oxide nanoparticles,
polymeric nanoparticles, quantum dots, and nanocomposites. The ultrasonic sound
methodology has been used for many kinds of applications, including cleaning, food
processing, agriculture, medical imaging nanosystems, nanomaterial processing,
nanoelectronic, nanocatalysis, nanodrug, chemical synthesis of nanostructures. It is
an adaptable and effective way to produce nanomaterials. The acoustic method gener-
ates localized hot spots in the liquid, facilitating chemical reactions that would other-
wise be difficult to perform under normal conditions due to the extreme temperatures
and pressures within these hot spots. The hotspots produced by the method affect
extremely high-pressure air bubbles, which rapidly reach temperatures of around
5000 K in nanoseconds. The size and shape of the final materials generated through
the ultrasound process are significantly affected by the process’s duration duration,
ambient temperature, concentration, and other parameters [15]. In 2021, Dai et al.
synthesized novel nanocarbon structures by modifying the conditions of ultrasonic
irradiation on a KOH/ethanol mixture. By subjecting a KOH/ethanol medium with
an amount of 210 mg mL−1 to ultrasonic irradiation for an hour at room tempera-
ture, followed by allowing it to rest at the same temperature for five days, they were
able to generate carbon nanotubes, nano-sized graphene polyhedra, and single/core
nanoonions. The ultrasonic synthesis method was used to synthesize different carbon
242 İ. M. Kahyaoğlu et al.
while minimizing its release in other areas of the body. This is important because it
can help to maximize the drug’s therapeutic effects while minimizing potential side
effects. Targeted drug delivery can be achieved through various strategies, including
encapsulation or coating of drugs with nanomaterials, use of nanoparticles or lipo-
somes, localized injection of the drug, or drug implantation. By controlling the release
and distribution of drugs within the body, nanodrugs can be tailored to effectively
manage a range of conditions. In recent years, the field of bio-nanostructured drug
delivery systems has become increasingly fascinating. Compared to conventional
biofilms, these systems have the potential to significantly improve the therapeutic
effect of drugs by delivering them directly to the targeted region. Various nanoma-
terials, such as thermo-sensitive nanogels, biopolymeric nanofibers, nanocompos-
ites, green-synthesized nanoparticles, nanotubes, and multilayered structures such
as MXenes, have been extensively researched as drug delivery systems. Scientists
aim to develop safer and more effective treatments for a wide range of diseases using
these innovative approaches to drug delivery, ultimately improving patient health
[21]. It is possible to deliver numerous dosages of drugs in a short amount of time
without changing the amount of the pharmaceuticals in the bloodstream. Patients
may fail to adhere to their drug plan as a result, though. To deal with this problem, it
is necessary for researchers to create small vehicles (nanocarriers) that provide drugs
continuously over an extended period, hence requiring minimal dosages. To achieve
this, the drug should be released at a constant rate, which is called a zero-order drug
release kinetic model. If a drug delivery system doesn’t release the drug effectively,
its potency may be reduced. Depending on the intended use of the system, drugs
can be delivered in various ways. These delivery mechanisms can occur simultane-
ously or at different stages. To develop controlled-release systems, it’s important to
understand these kinetic mechanisms. Table 1 displays various kinetic models.
where t: time, Mt: cumulative release of the drug in time t, and M∞: the theoretical maximum
release of the drug. K: model release constant, and n: the diffusion exponent
Nanostructured Biomaterials in Drug Delivery 245
that was effectively captured within the spherical nanostructure (encapsulation effi-
ciency), was extremely high for the PTX-loaded nanostructure. The PTX-loaded
nanostructure exhibited an encapsulation efficiency of 90.17 ± 0.52%. Moreover,
the capacity of the nanostructure for drug loading, was also found to be high at
15.28 ± 0.10%. These findings indicate that the PTX-loaded nanostructure has a
high drug loading capacity, high entrapment efficiency, and dual targeting, making it
a potential therapeutic delivery system for chemotherapy in breast cancer. The scien-
tists discovered that in the presence of hyaluronidase and/or lipase, the PTX-loaded
nanostructure they created rapidly released the drug. Based on research conducted
on cell lines, they found that 4T1 cells absorbed the nanostructure through the CD44
receptor and bio-specific proteins. Additionally, they observed that the PTX-loaded
nanostructure was more cytotoxic to 4T1 cells than PTX-loaded biotin-free nanos-
tructure. During their in vivo studies using mice with 4T1 tumors, they observed
a remarkable therapeutic efficacy of the PTX-loaded nanostructure. They claimed
that the nanostructure exhibited better pharmacological properties than Taxol and the
combined action of hyaluronic acid and biotin in the nanostructure produced an excel-
lent tumor targeting effect. Based on these findings, the researchers concluded that
PTX-loaded nanostructure could be an excellent candidate for breast cancer treat-
ment [26]. To encourage healing process in skin problems, Zhang and colleagues
designed a biocompatible nanomaterial with redox-sensitive functionalities. The
radical emulsion polymerization strategy was employed by the researchers to synthe-
size redox-sensitive PNA-based nanostructure. Scientists found that the biocompat-
ible membranes containing PNA enhanced cell adhesion as well as proliferation in
compared to the natural PLLA membrane through in vitro biological assessments.
They showed that the PNA-loaded nanomembranes’ bonds are formed controlled
the redox potential and adjusted the Reactive Oxygen Species (ROS) level in the
damaged epidermis, speeding the process of healing in in vivo experiments using
a mouse skin injury model. Consequently, the research showed that PNA-loaded
nanomembranes could be an effective treatment for skin diseases by balancing ROS
levels and promoting skin regeneration [27]. In 2017, Wang et al. synthesized novel
hyaluronic acid-coated chitosan nanoparticles (NPs) to enhance the chemothera-
peutic efficacy of the cancer drug 5-FU. The goal was to target therapeutic agents
to cancerous cells using CD44. The researchers observed a stable release profile of
5-FU from the nanoformulation with 75% of the total 5-FU released in the first 48
h. They found that the interaction between HA and CD44 facilitated more effective
drug delivery to cancerous cells when CD44 was overexpressed, resulting in greater
drug accumulation in these cells compared to free pharmaceuticals and uncoated
NPs. Based on these findings, the researchers concluded that HA-coated chitosan
NPs could represent a promising approach for inducing apoptosis in tumor cells
and treating cancer effectively. Finally, the study demonstrated the potential of HA-
coated chitosan NPs for CD44-mediated drug delivery to cancer and improving the
therapeutic efficacy of 5-FU and other anticancer agents [28]. In their 2022 study, Tak
et al. developed a synthesis way to produce graphene quantum dots from of Polygala
tenuifolia extracts for the inhibition of acetylcholine esterase. They then functional-
ized the graphene quantum dots with donepezil hydrochloride to create DO-loaded
Nanostructured Biomaterials in Drug Delivery 249
graphene quantum dots, which were compared to the conventional Alzheimer’s drug.
The scientists found that the novel DO-loaded graphene quantum dots demonstrated
greater inhibitory activity on acetylcholine esterase compared to graphene quantum
dots alone or the conventional medicine. Moreover, the study revealed a synergistic
effect between the pharmaceutical delivery mechanism and the Polygala tenuifolia
root extract in the Alzheimer’s disease treatment process. The research concluded
that the prepared DO-loaded graphene quantum dots represent a safe and effective
drug delivery system for Alzheimer’s treatment, with potential for future medical
applications [29]. Reddy et al. developed a novel drug delivery system in 2019
by incorporating acetyl curcumin (AC) into PLGA liposome nanostructures. The
researchers found that at its highest rate, 48% of the drug was released from the
nanocarriers within approximately 4 days. The study also showed that the prepared
nanostructure released more AC than the free drug due to functional group interac-
tions between the lipid head groups and AC. Importantly, the researchers observed
no cytotoxicity on the HDFa cell line, suggesting that the nanostructures are safe for
normal tissues. Based on these results, the researchers suggested that the proposed
nanostructure could serve as a promising drug delivery system for clinical applica-
tions [30]. In order to look into the possibility of employing exosomes loaded with
lapatinib as a drug delivery vehicle, Deirmenci et al. conducted an investigation in
2022. Exosomes were extracted from normal breast epithelial cells, and it was found
that their good biocompatibility, and low toxicity make them more appropriate as
carriers than synthesized nanostructures. In HER2 (+) SKBR 3 cell line, and MCF10
cell line, the pharmacological effects of Lapatinib/exosomes, free Lapatinib, and
uncharged exosomes were tested. In contrast to free Lapatinib, they observed that
exosomes carrying Lapatinib were more effective at preventing division of cells and
maintaining apoptosis in different types of cancer cells. The researchers hypothe-
sized that using nanocarriers loaded with lapatinib would allow the use of drugs at
smaller doses. Overall, the study suggests that exosomes loaded with Lapatinib have
the potential to be used as a drug delivery tool in cancer treatment [31].
Highly reactive chemicals known as ROS have the potential to harm cells if improp-
erly controlled. Cells have created a number of pathways that can react to variations
in ROS levels in order to protect themselves against damage that ROS causes. Super-
oxide dismutase, glutathione peroxidase, and catalase are a few antioxidant enzymes
that cells contain that can reduce ROS by converting them into less dangerous
molecules. Heat shock proteins (HSPs): HSPs have a role in preventing oxidative
cellular damage and can be generated in response to ROS and other cellular stres-
sors. HSP70, for instance, can inhibit particle agglomeration brought on by ROS.
A few transcription factors, such NRF2, can activate the production of enzymatic
antioxidants and other chemoprotective proteins in response to ROS. During the
250 İ. M. Kahyaoğlu et al.
process of autophagy, cells can recycle undesirable or damaged parts of their struc-
ture, including ROS-affected parts. Together, these processes defend cell lines from
ROS-caused harm and preserve cell balance. In 2023, Gandomi et al. developed a
novel nano-sonosensitizer by coating metal/metal oxide hybrid-based nanocarriers
with l-cysteine and graphene quantum dots. When exposed to internal and external
factors such as sonic and pH modifications, the bio-nanoformulation was able to
alter its surface characteristics in tumor cells and enhance its sonocatalytic prop-
erties. In contrast to the free form of curcumin, they observed that using the bio-
nanoformulation as a drug delivery vehicle for curcumin in chemo-sonodynamic
treatment resulted in a significant suppression of A549 cells. The l-cysteine/hybrid-
based nanocarriers and bio-nanoformulation demonstrated high drug loading effi-
ciencies of 61.3% and 72.2%, respectively [32]. Overall, the research indicates that
the novel bio-nanoformulation is an effective and safe approach to treating cancer.
use has been authorized. By preventing the STAT3 protein from functioning, napabu-
casin is used to treat pancreatic and stomach cancer. Advanced basal cell carci-
noma is treated with vismodegib, an inhibitor of the Hedgehog signaling system.
Relapsed acute myelocytic leukemia is treated with the IDH1 inhibitor ivosidenib.
Small lymphocytic lymphoma and chronic lymphocytic leukemia are both treated
with venetoclax, a Bcl-2 inhibitor [36]. With the development of nanomedicine,
studies on improving cancer chemotherapy are increasing day by day [37]. Table 3
lists some nanodrugs and cancer treatment.
By conjugating nano-drugs with ligands that target tumors and encourage their
intracellular uptake, anti-cancer medications can be enhanced in their efficacy. More-
over, negatively charged nano-drugs often stay in the bloodstream for longer periods
of time and are less likely to be metabolized by cancerous cells, which decreases
intracellular concentration of the drug. For this reason, researchers are focused on
overcoming the limitations associated with negatively charged surfaces to improve
the efficacy of nano-drugs. To increase the amount of drug inside cancer cells, posi-
tively charged nano-drugs are preferred, as they have a higher chance of being taken
up by these cells. There are several methods of producing positively charged nano-
drugs, including conjugating them with ligands that target specific receptors. In 2022,
Huang et al. developed a positively charged nanodrug to improve cancer intracellular
uptake. They used carbodiimide, hydroxysuccinimide, and platinum. They synthe-
sized metal-polymer-chlorin by reacting polyethyleneimine. In order to obtain the
bio-nanostructure, with an average size of 160 nm and a spherical structure, the
resulting polymer was dissolved in deionized water and filtered. Hyaluronic acid
was used to coat the bio-nanostructure in order to give them a negatively charged
surface potential and enhance the targeted delivery of bio-nanostructure to melanoma
cells that overexpress CD44. The hyaluronic acid was degraded by the positively
charged bio-nanostructure which increased the absorption into tumor cells. In their
in vivo study, they showed that bio-nanostructure has anti-cancer properties against
melanoma cells in mice [38]. In another study, Mo et al. designed a nano drug
system that combines drug delivery with cooperative chemo-photothermal anticancer
potential. They used a hydrothermal method to produce molybdenum disulfide nano-
layers and dispersed 50 mg of lipoic acid functionalized polymer (PEG) in 20 mL
of molybdenum disulfide dispersion. They produced the molybdenum disulfide/PEG
composite after 24 h of continuous stirring. To develop the nanocomposite, they added
3 integrin binding peptide to the composite. The anticancer drug (DOX) was used to
manufacture the nano-drug in the final stage after the required centrifugation proce-
dures. The resulting 200–300 nm-sized flower-like nanomaterials. Using the MTT
assay, the produced nanocarrier MPRS’s cytotoxicity against MCF-7 cells, VSMC
cells, Hela cells, and H9c2 cells was assessed. The produced nano drug’s release
in the tumor microenvironment was boosted, increasing its ability to destroy tumor
cells while decreasing its toxicity against healthy cells [39]. In another study, Skoll
et al. investigated into the development of proteinaceous serum albumin nanocar-
riers with plant oil that do not contain hazardous cross-linkers or potentially harmful
non-aqueous solutions. They used a sonochemical method with different plant oils
to produce these nanomaterials, which produced particles with limited size distri-
bution and stable suspensions that can be frozen to keep for a long period. Using
bio-nanocarriers (molar ratio: 7/1), they were also succeeded in integrating wheat
germ agglutinin as a target molecule into the shell. Researchers found that nanocar-
riers had up to a 55% higher cell binding potential than those without a targeter after
conducting urothelial cell binding assays [40]. It shows that targeted protein nanocar-
riers could be an innovative pharmaceutical delivery approach. Recent studies have
254 İ. M. Kahyaoğlu et al.
shown promising results in the field of nanomedicine for treating difficult condi-
tions, particularly in the area of neurodegenerative diseases such as Parkinson’s,
Alzheimer’s, and Huntington’s. These problems are often challenging to treat effec-
tively, and nanomedicine offers new approaches to combat them. Central nervous
system disorders (CNS) are a group of diseases that also lack effective treatment
options. Among these, gliomas and glioblastomas, two types of brain cancer, are the
most common tumors in the central nervous system of adults. Systems for treating
these disorders with nanomedicine should have no trouble crossing the blood–brain
barrier. Peng et al. conducted a study in 2018 to assess the effectiveness of a combi-
nation therapy approach on glioma apoptosis and cancer progression inhibition. The
therapy consisted of temozolomide and a folate-targeted nanocarrier that delivered
anti-BCL-2 siRNA. The nanocarrier was designed to improve the permeability of the
blood–brain barrier. In their in vitro cell study, the researchers observed a significant
apoptotic response in cancer cells, which was attributed to the suppression of the
BCL-2 gene and activation of the proapoptotic Bax gene [41]. In the last few years,
the advancement of intelligent nano-drugs has led to a rise in research on reducing
the insufficiency and toxic impacts of drugs used in tumor cells with a complex
structure. As nano-based drugs advance to clinical trial stages, it is expected that
more efficient mechanisms will be uncovered in the following years. The escalation
in productive in vitro and in vivo studies demonstrates these outcomes.
References
1. Stanisz, M., Klapiszewski, Ł, Jesionowski, T.: Recent advances in the fabrication and applica-
tion of biopolymer-based micro- and nanostructures: a comprehensive review. Chem. Eng. J.
397, 125409 (2020). https://doi.org/10.1016/J.CEJ.2020.125409
2. Rashidinejad, A., Birch, E.J., Sun-Waterhouse, D., Everett, D.W.: Effect of liposomal encap-
sulation on the recovery and antioxidant properties of green tea catechins incorporated into
a hard low-fat cheese following in vitro simulated gastrointestinal digestion. Food Bioprod.
Process. 100, 238–245 (2016). https://doi.org/10.1016/J.FBP.2016.07.005
3. Li, Y., Qiu, S., Sun, J., Ren, Y., Wang, S., Wang, X., Wang, W., Li, H., Fei, B., Gu, X., Zhang,
S.: A new strategy to prepare fully bio-based poly(lactic acid) composite with high flame
retardancy, UV resistance, and rapid degradation in soil. Chem. Eng. J. 428, 131979 (2022).
https://doi.org/10.1016/J.CEJ.2021.131979
4. Santhiago, M., Garcia, P.S., Strauss, M.: Bio-based nanostructured carbons toward sustainable
technologies. Curr. Opin. Green Sustain. Chem. 12, 22–26 (2018). https://doi.org/10.1016/J.
COGSC.2018.04.009
5. Lee, S.C., Yoo, E., Lee, S.H., Won, K.: Preparation and application of light-colored lignin
nanoparticles for broad-spectrum sunscreens. Polymers 12, 699 (2020). https://doi.org/10.3390/
POLYM12030699
6. Aadil, K.R., Mussatto, S.I., Jha, H.: Synthesis and characterization of silver nanoparticles
loaded poly(vinyl alcohol)-lignin electrospun nanofibers and their antimicrobial activity. Int.
J. Biol. Macromol. 120, 763–767 (2018). https://doi.org/10.1016/J.IJBIOMAC.2018.08.109
7. Zhang, F., Wu, W., Zhang, X., Meng, X., Tong, G., Deng, Y.: Temperature-sensitive poly-
NIPAm modified cellulose nanofibril cryogel microspheres for controlled drug release.
Cellulose 23, 415–425 (2016). https://doi.org/10.1007/S10570-015-0799-4/TABLES/1
8. Maskur, M.H., Wan Nawawi, W.M.F., Ali, F., Alkhatib, M.F.R.: Crustacean chitin nanomaterial
as reinforcement for bio-based polymer 1–12 (2020). https://doi.org/10.1080/2374068X.2020.
1793638
9. Coltelli, M.B., Morganti, P., Castelvetro, V., Lazzeri, A., Danti, S., Benjelloun-Mlayah, B.,
Gagliardini, A., Fusco, A., Donnarumma, G.: Chitin nanofibril-nanolignin complexes as
carriers of functional molecules for skin contact applications. Nanomaterials 12, 1295 (2022).
https://doi.org/10.3390/NANO12081295
10. Wang, W., Meng, Q., Li, Q., Liu, J., Zhou, M., Jin, Z., Zhao, K.: Chitosan derivatives and their
application in biomedicine. Int. J. Mol. Sci. 21, 487 (2020). https://doi.org/10.3390/IJMS21
020487
11. Ansari, M.J., Jasim, S.A., Bokov, D.O., Thangavelu, L., Yasin, G., Khalaji, A.D.: Preparation of
new bio-based chitosan/Fe2O3/NiFe2O4 as an efficient removal of methyl green from aqueous
solution. Int. J. Biol. Macromol. 198, 128–134 (2022). https://doi.org/10.1016/J.IJBIOMAC.
2021.12.082
12. Aranaz, I., Alcántara, A.R., Civera, M.C., Arias, C., Elorza, B., Caballero, A.H., Acosta, N.,
Velasco, H., Mecerreyes, D., Antonio, R., Gimeno, B., María Díez-Pascual, A., Moreno, V.C.,
Serra, A.: Chitosan: an overview of ıts properties and applications. Polymers 13, 3256 (2021).
https://doi.org/10.3390/POLYM13193256
13. Gao, X., Gong, J., Cai, Y., Wang, J., Wen, J., Peng, L., Ji, H., Jiang, S., Guo, D.: Chitosan modi-
fied squalene nanostructured lipid carriers as a promising adjuvant for freeze-dried ovalbumin
vaccine. Int. J. Biol. Macromol. 188, 855–862 (2021). https://doi.org/10.1016/J.IJBIOMAC.
2021.08.074
256 İ. M. Kahyaoğlu et al.
14. Sathiskumar, S., Vanaraj, S., Sabarinathan, D., Bharath, S., Sivarasan, G., Arulmani, S., Preethi,
K., Ponnusamy, V.K.: Green synthesis of biocompatible nanostructured hydroxyapatite from
Cirrhinus mrigala fish scale—A biowaste to biomaterial. Ceram. Int. 45, 7804–7810 (2019).
https://doi.org/10.1016/j.ceramint.2019.01.086
15. Kim, K.H., Kim, K.B.: Ultrasound assisted synthesis of nano-sized lithium cobalt oxide.
Ultrason. Sonochem. 15, 1019–1025 (2008). https://doi.org/10.1016/J.ULTSONCH.2007.
11.004
16. Dai, D., Li, Y., Fan, J.: Room-temperature synthesis of various allotropes of carbon nanostruc-
tures (graphene, graphene polyhedra, carbon nanotubes and nano-onions, n-diamond nanocrys-
tals) with aid of ultrasonic shock using ethanol and potassium hydroxide. Carbon N Y. 179,
133–141 (2021). https://doi.org/10.1016/J.CARBON.2021.04.038
17. Wang, C., Yang, M., Shi, H., Yao, Z., Liu, E., Hu, X., Guo, P., Xue, W., Fan, J.: Carbon quantum
dots prepared by pyrolysis: Investigation of the luminescence mechanism and application as
fluorescent probes. Dyes Pigm. 204, 110431 (2022). https://doi.org/10.1016/j.dyepig.2022.
110431
18. Agnol, L.D., Neves, R.M., Maraschin, M., Moura, S., Ornaghi, H.L., Dias, F.T.G., Bianchi,
O.: Green synthesis of Spirulina-based carbon dots for stimulating agricultural plant growth.
Sustain. Mater. Technol. 30 (2021). https://doi.org/10.1016/j.susmat.2021.e00347
19. Nurfazianawatie, M.Z., Omar, H., Rosman, N.F., Malek, N.S.A., Afaah, A.N., Buniyamin,
I., Salifairus, M.J., Malek, M.F., Mahat, M.M., Rusop, M., Asli, N.A.: Forming multilayer
graphene at different precursor temperature using cooking oil waste from AYAMAS via chem-
ical vapour deposition. Mater Today Proc. 75, 127–132 (2022). https://doi.org/10.1016/j.matpr.
2022.10.169
20. Irfan, M.F., Arami-Niya, A.: Temperature effect on the synthesis of iron–cobalt nano-particles
using catalytic chemical vapor deposition of CO2 in thermo-gravimetric analyzer: analytical
and thermodynamic studies. Nano-Struct. Nano-Objects 18, 100261 (2019). https://doi.org/10.
1016/j.nanoso.2019.100261
21. Wang, T., Cornel, E.J., Li, C., Du, J.: Drug delivery approaches for enhanced antibiofilm
therapy. J. Control. Release 353, 350–365 (2023). https://doi.org/10.1016/J.JCONREL.2022.
12.002
22. Mariz, M., Murta, J., Gil, M.H., Ferreira, P.: An ocular insert with zero-order extended delivery:
release kinetics and mathematical models. Eur. J. Pharm. Biopharm. 181, 79–87 (2022). https://
doi.org/10.1016/J.EJPB.2022.10.023
23. Yang, M., Abdalkarim, S.Y.H., Yu, H.Y., Asad, R.A.M., Ge, D., Zhou, Y.: Thermo-sensitive
composite microspheres incorporating cellulose nanocrystals for regulated drug release
kinetics. Carbohydr. Polym. 301, 120350 (2023). https://doi.org/10.1016/J.CARBPOL.2022.
120350
24. Zirak, N., Bolandparvaz Jahromi, A., Salahinejad, E.: Vancomycin release kinetics from Mg–
Ca silicate porous microspheres developed for controlled drug delivery. Ceram Int. 46, 508–512
(2020). https://doi.org/10.1016/J.CERAMINT.2019.08.290
25. Bardajee, G.R., Hooshyar, Z., Farsi, M., Mobini, A., Sang, G.: Synthesis of a novel thermo/pH
sensitive nanogel based on salep modified graphene oxide for drug release. Mater. Sci. Eng. C
72, 558–565 (2017). https://doi.org/10.1016/J.MSEC.2016.11.109
26. Gao, D., Asghar, S., Ye, J., Zhang, M., Hu, R., Wang, Y., Huang, L., Yuan, C., Chen, Z., Xiao,
Y.: Dual-targeted enzyme-sensitive hyaluronic acid nanogels loading paclitaxel for the therapy
of breast cancer. Carbohydr. Polym. 294, 119785 (2022). https://doi.org/10.1016/J.CARBPOL.
2022.119785
27. Zhang, S., Li, Y., Qiu, X., Jiao, A., Luo, W., Lin, X., Zhang, X., Zhang, Z., Hong, J., Cai,
P., Zhang, Y., Wu, Y., Gao, J., Liu, C., Li, Y.: Incorporating redox-sensitive nanogels into
bioabsorbable nanofibrous membrane to acquire ROS-balance capacity for skin regeneration.
Bioact Mater. 6, 3461–3472 (2021). https://doi.org/10.1016/J.BIOACTMAT.2021.03.009
28. Wang, T., Hou, J., Su, C., Zhao, L., Shi, Y.: Hyaluronic acid-coated chitosan nanoparticles
induce ROS-mediated tumor cell apoptosis and enhance antitumor efficiency by targeted
drug delivery via CD44. J. Nanobiotechnol. 15, 7 (2017). https://doi.org/10.1186/s12951-016-
0245-2
Nanostructured Biomaterials in Drug Delivery 257
29. Tak, K., Sharma, P., Sharma, R., Dave, V., Jain, S., Sharma, S.: One-pot hydrothermal green
synthesis of Polygala tenuifolia mediated graphene quantum dots for acetylcholine esterase
inhibitory activity. J. Drug Deliv. Sci. Technol. 73, 103486 (2022). https://doi.org/10.1016/J.
JDDST.2022.103486
30. Reddy, A.S., Lakshmi, B.A., Kim, S., Kim, J.: Synthesis and characterization of acetyl
curcumin-loaded core/shell liposome nanoparticles via an electrospray process for drug
delivery, and theranostic applications. Eur. J. Pharm. Biopharm. 142, 518–530 (2019). https://
doi.org/10.1016/J.EJPB.2019.07.024
31. Değirmenci, N.S., Uslu, M., Kırbaş, O.K., Şahin, F., Önay Uçar, E.: Lapatinib loaded exosomes
as a drug delivery system in breast cancer. J. Drug Deliv. Sci. Technol. 75, 103584 (2022).
https://doi.org/10.1016/J.JDDST.2022.103584
32. Gandomi, F., Rostami, M., Ahmadi, F., Mohammad Sorouri, A., Badiei, A., Fasihi-Ramandi,
M., Reza Ganjali, M., Ehrlich, H., Rahimi-Nasrabadi, M.: ROS, pH, and magnetically respon-
sive ZnFe2O4@l-Cysteine@NGQDs nanocarriers as charge-reversal drug delivery system for
controlled and targeted cancer chemo-sonodynamic therapy. Inorg. Chem. Commun. 150,
110544 (2023). https://doi.org/10.1016/J.INOCHE.2023.110544
33. Zhang, Y., Qian, J., Gu, C., Yang, Y.: Alternative splicing and cancer: a systematic review.
Signal Transduct Target Ther. 6 (2021). https://doi.org/10.1038/S41392-021-00486-7
34. Qin, S.-Y., Zhang, A.-Q., Zhang, X.-Z., Qin, S.-Y., Q. A. Zhang, Zhang, Z. X.: Recent advances
in targeted tumor chemotherapy based on smart nanomedicines. Small 14, 1802417 (2018).
https://doi.org/10.1002/SMLL.201802417
35. Wei, G., Wang, Y., Yang, G., Wang, Y., Ju, R.: Recent progress in nanomedicine for enhanced
cancer chemotherapy. Theranostics 11, 6370 (2021). https://doi.org/10.7150/THNO.57828
36. Yan, J., Long, X., Liang, Y., Li, F., Yu, H., Li, Y., Li, Z., Tian, Y., He, B., Sun, Y.: Nanodrug
delivery systems and cancer stem cells: from delivery carriers to treatment. Colloids Surf. B
Biointerf. 217, 112701 (2022). https://doi.org/10.1016/J.COLSURFB.2022.112701
37. Khot, V.M., Salunkhe, A.B., Pricl, S., Bauer, J., Thorat, N.D., Townley, H.: Nanomedicine-
driven molecular targeting, drug delivery, and therapeutic approaches to cancer chemore-
sistance. Drug Discov. Today 26, 724–739 (2021). https://doi.org/10.1016/J.DRUDIS.2020.
12.016
38. Huang, X., Mu, N., Ding, Y., Lam, H.W., Yue, L., Gao, C., Chen, T., Yuan, Z., Wang, R.: Targeted
delivery and enhanced uptake of chemo-photodynamic nanomedicine for melanoma treatment.
Acta Biomater. 147, 356–365 (2022). https://doi.org/10.1016/J.ACTBIO.2022.05.015
39. Mo, C., Wang, Z., Yang, J., Ouyang, Y., Mo, Q., Li, S., He, P., Chen, L., Li, X.: Rational assembly
of RGD/MoS2/Doxorubicin nanodrug for targeted drug delivery, GSH-stimulus release and
chemo-photothermal synergistic antitumor activity. J. Photochem. Photobiol. B. 233, 112487
(2022). https://doi.org/10.1016/J.JPHOTOBIOL.2022.112487
40. Skoll, K., Ritschka, M., Fuchs, S., Wirth, M., Gabor, F.: Characterization of sonochemically
prepared human serum albumin nanocapsules using different plant oils as core component for
targeted drug delivery. Ultrason. Sonochem. 76, 105617 (2021). https://doi.org/10.1016/J.ULT
SONCH.2021.105617
41. Peng, Y., Huang, J., Xiao, H., Wu, T., Shuai, X.: Codelivery of temozolomide and siRNA with
polymeric nanocarrier for effective glioma treatment. Int. J. Nanomed. 13, 3467 (2018). https://
doi.org/10.2147/IJN.S164611
258 İ. M. Kahyaoğlu et al.
Pankaj Sharma
Abstract Materials in the nanoscale category are used for diagnostic purpose or to
administer therapeutic material to specialized targeted places in a regulated manner in
the relatively young but rapidly emerging field of nano-biomedicine. In the treatment
of chronic human illnesses, nanotechnology has a number of advantages, including
the targeted and site-specific administration of precise medications. The use of nano-
biomedicine (chemotherapeutic substances, biologic substances, immunotherapeutic
substances, etc.) in the therapy of many illnesses has recently resulted in an array of
notable developments. By doing careful examination of the development and appli-
cation of nano-biomaterials to enhance the effectiveness of both new and old drugs
(such as natural therapeutics) and particular detection by means of illness marker
molecules, this chapter gives a revised overview of current developments in the
discipline of nano-biomedicines and nano centred systems for drug delivery. The
benefits and drawbacks of employing nanomedicines for the therapeutic administra-
tion of pharmaceuticals derived from synthetic or natural sources are also discussed.
Additionally, we provided details on the developments and aspirations in the field of
nanomedicine.
Abbreviations
5-FU 5-fluorouracil
CMC Carboxymethylcellulose
XG Xanthan gum
PEG Polyethylene glycol
QDs Quantum Dots
P. Sharma (B)
Department of Pharmaceutics, ShriRam College of Pharmacy, Banmore, Morena, Madhya
Pradesh, India
e-mail: pankajsharma223@gmail.com
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 261
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_9
262 P. Sharma
1 Introduction
Humans have long employed natural remedies derived from plants to treat a variety
of illnesses. The majority of modern medications are made from herbs using conven-
tional wisdom and methods. On the market today, the most significant pharmaceutical
compounds and their derivatives make up around 25% of the total [1, 2]. A starting
point for the creation of novel pharmaceuticals is natural compounds with varied
molecular ancestries. Designing lead compounds that are synthetically accessible
and resemble their counterparts’ chemistry is the latest fad in natural product-based
pharmaceutical research [3]. Only a few of the astounding properties that biomate-
rials exhibit include remarkable chemical variation, biological and chemical potential
including macromolecular accuracy, and decreased noxiousness. They do so because
they see them as good prospects for developing new drugs [4]. Additionally, target-
based drug deliveries, drug prediction at the cellular level, and other next-generation
pharmaceutical breakthroughs have all been made possible thanks to computational
techniques.
Despite the benefits, healthcare industries [5] are disinclined to pay-out additional
funds on biomaterial-based medicaments development and medicaments delivery
setup, attempt to uncover new medicines by searching over databases of chem-
ical substances. However, natural moieties are now being used to treat a number
of grave diseases, such as cancer, inflammatory, microbiological, and cardiovas-
cular issues. The specific advantages that natural medicines offer like less toxicity and
side responses, cheap cost, and potent therapeutic value are largely to blame for this.
Nevertheless, it is more challenging to use natural compounds as medications because
of concerns regarding their bioactivity and security. Numerous natural compounds are
stalling in the clinical study stages as a result of these problems [6–8]. It is extremely
difficult to use large-scale biomaterials for drug delivery because of their in vivo
instability, poor body absorption, decreased solubility and bioavailability, difficulties
with site-specific delivery, problems with tonic performance, and possibly even nega-
tive pharmacological effects. Therefore, a different approach to solving these urgent
issues may be to employ cutting-edge drug delivery techniques to target treatments
at specific body regions [9, 10]. Therefore, nanotechnology has a significant influ-
ence on the formulations of contemporary medications, as well as their successful
delivery and regulated disintegration. In a diversity of medical domains, involving
nano-medicaments and nano-scaled drug delivery networks, where such substances
are of particular significance, the use of nanomaterials and nanophases has shown the
ability of nanotechnology to establish a connection between the physical and biolog-
ical sciences. Nano-biomaterials, which may be defined as substances with a diameter
of 1–100 nm, have a tremendous influence on the future of nanomedicine, impacting
everything from tissue engineering to microfluidics to drug delivery and biosensors
Nanostructured Biomaterials in Drug Delivery: Current Trends … 263
Fig. 1 Examples of nano-biomaterials and their delivery methods for usage in humans
delivery of organic remedies for the treatment of malignancy along with numerous
other diseases, it offers a number of interests. In light of their numerous unique
properties, including their ability to induce tumor-suppressing senescence and func-
tion as antimicrobials, biomaterials have been extensively explored in the context
of treating illnesses. Caffeine and curcumin and have been linked to apoptosis [15],
whilst cinnamaldehyde, curcumin, carvacrol, and eugenol have been linked to antimi-
crobial properties. Using nanotechnology allowed for the enhancement of various
qualities, including targeting, bioavailability, and controlled release. For example, the
bioactive ingredient in Nigella sativa called thymoquinone is examined after already
being enclosed in a lipid nanomaterial. Its bioavailability increased six fold following
encapsulation compared to free thymoquinone, protecting the gastrointestinal tract
Nanostructured Biomaterials in Drug Delivery: Current Trends … 265
The inherent issues with traditional dosage distribution techniques lead to the require-
ment of substances for controlled medication release. Administration of drugs often
requires numerous, repetitive dosages, resulting in highly variable medication levels
in the body during the treatment course. After delivery, medication levels rise to
therapeutic doses, but occasionally hazardous side effects develop whenever the
quantity exceeds the upper limit of what is safe [17]. As a consequence of degrada-
tion, metabolism, and transportation far from the targeted therapy, these approaches
also quickly reduce drug levels to levels that are no more helpful [17]. Together,
this causes materials and medicine waste and raises patient risk owing to probable
serious adverse reactions and decreased treatment efficacy. Strategies for reducing
the rate of release were created to solve these problems. These “sustained release”
methods included the required treatment in capsule form that was typically taken
orally, however some of them were also designed for parenteral use [18]. Drug
release was slowed down by using cellulose coverings that slowly dissolve, adding
chemicals that make drugs less soluble, compressing tablets, and using emulsion and
suspensions [18] that are all contained in capsules. Nevertheless patient to patient
variation, environmental influences, and the need for repeated dosing continued to
have a significant impact on controlled release preparations [18].
The optimum controlled medication release method offers a number of bene-
fits over extended release. These delivery substances preserve drug release within
the therapeutic range and prevent the inefficiency of the drug concentration fluctua-
tions of conventional formulations by releasing pharmaceuticals at rates that do not
fluctuate over duration (zero-order release). Controlled release substances have the
advantage of lowering the total quantity of medication required to attain treatment
effectiveness since they prevent “peaks and valleys” and stay within the therapeutic
range. These substances would help increase patient compliance, which would in
developed countries is just 50%, by lowering the total amount of dosages neces-
sary [19]. Such substances can also be administered or implanted precisely into a
particular sick tissue, decreasing off-target adverse effects and boosting efficacy.
Release of drugs can be regulated across extended therapeutic windows (i.e., days to
years). In order to prevent off-target consequences, sustained release methods must
improve the delivery of medications to particular tissues and cells inside the system
[20]. Targeted delivery techniques are used for precise preservation and absorption
by sick tissues and cells in order to increase tissue specificity [21]. In this method,
delivery components are coupled to ligands which attach to surface proteins or recep-
tors that are abundantly expressed in sick tissues and cells. Optimal controlled release
polymers should also shield pharmaceuticals against quick bodily clearance and/or
deterioration.
Engineering, physical science, biology, and clinical professionals must all work
together in a comprehensive approach to create suitable nano-biomaterials for
controlled drug release [22]. Design criteria involve: I including enough drug in
Nanostructured Biomaterials in Drug Delivery: Current Trends … 267
active or passive and uses nanostructures as the drug delivery networks, is another
crucial aspect of medicine administration. In targeted delivery, drugs are delivered by
means of peptides, antibodies, and other compounds that bind to receptor sites made
in the target area. Through preferences, which are influenced by alterable similar as
pH, molecular site, temperature, and structure as they circulate through the circula-
tion, the generated drug delivery vehicle molecule is carried to the target region in
passive targeting. The organism’s main targets are receptors on cellular membranes,
lipids in cellular membranes, and proteins or antigens on certain substrates [26].
Today, the bulk of drug delivery methods made available by nanotechnology are
focused on curing and treating cancer.
worse by inadequate lymph drainage in the vicinity of the tumour [30]. The distances
among blood vessels vary depending on the kind of tumour and are typically around
100 and 800 nm. Nano-biomaterials of a diameter range of 30–200 nm had the highest
tumour survival potential, according to experimental studies using several tumour
models [31]. Also, it was shown that round nano-biomaterilas, as opposed to rod- or
bar-shaped ones, might conformance into the tumour interstitium better successfully
due to their ability to retain laminar flow characteristics [32].
Active targeting, particularly includes the nano-biomaterials molecularly recog-
nizing the tumour, can solve passive targeting’s several disadvantages, including
unequal distribution throughout the solid tumour and trapping within the tumour
[33]. It is possible to attach tumor-specific ligands to an exterior of the nanomaterial,
allowing them to establish highly-affine interactions with receptors expressed on the
tumour surface. These ligands can include antibodies or other compounds. In order to
effectively internalize the nanomaterial into the cell and deliver the medicine, this also
promotes receptor-mediated cellular uptake. Multipurpose nano-biomaterials are
defined as nano-biomaterial that possess certain chemical identification signals; this
topic will be covered in more detail in subsequent chapters [34].
Therefore, active and passive targeting cooperate to deliver the drug to the cancer.
Therefore, active targeting increases the drug’s retention and absorption by the cancer
cells, whilst passive targeting ensures the concentration of the nano-particles from
the bloodstream into the tumour milieu.
5.1 Alginate
rate of cell mortality in the H460-lung tumor cell when compared to the unbound
drug.
5.2 Chitosan
5.3 Cellulose
The main purpose of using cellulose and its variants in pharmaceutical drug admin-
istration is to vary the medications’ gelation and solubility, which in turn affects how
quickly they are released from the body [39]. By conjugating calcium alginate pellets
with 5-fluoroacyl (5-FU) enriched carboxymethylcellulose (CMC), Agarwal et al.
[40] successfully created a medication-targeted method that is specific to the colon.
In the setting that mimicked a colonic milieu, the pellets with reduced CMC ratios
displayed more edema and muco-adhesiveness. The 5-FU contained in the beads was
released by the intestinal enzymes to 90% of its original concentration.
Amphiphilic block copolymers are used to create polymeric micelles, which coalesce
into a centred-shell configuration in watery solutions. The aquaphillic shell may stabi-
lize the hydrophobic core, which may be loaded with hydrophobic medicines (such as
docetaxel, camptothecin, and paclitaxel), making the entire system soluble in water.
The Enhanced permeability and retention (EPR) effect allows polymeric micelles,
which typically involve a size of less than 100 nm and a limited dissemination to
slow rapid renal clearance, to accumulate in cancer tissues. Their polymeric shell
also protects them from accidental interaction with biological components. Since
their inner core design allows for the absorption of this type of medication, these
nanostructures (Fig. 2) hold great potential for the delivery of hydrophobic drugs,
which will boost durability and bioavailability [43, 44].
6.2 Liposomes
Alec Bangham discovered them for the first time in 1960. Liposomes, one of the
most thoroughly researched medicament delivery carrier systems, are employed in
the health and beauty industries to transport a range of chemicals. Drug delivery
272 P. Sharma
has long been enhanced by the creation of liposomes. They typically consist of
phospholipids and steroids and are spherical vesicles between 50 and 450 nm in
diameter [45]. They are considered as superior vehicles for drug administration since
it is easy to put drugs into them and because their membrane topology identical to
that of membranes of cells [45]. They have also demonstrated the ability to stabilize
therapeutic molecules, enhance biodistribution, and permit the use of both aqueous
and hydrophobic pharmaceuticals. They are also biodegradable and biocompatible.
For liposomes, there are four groups: (1) Ordinary liposomes: they feature a
water-containing core wrapped in a cholesterol/phospholipid which can be neutral,
cationic/anionic. In that condition, the lipid bilayer or the water-filled gap can be
filled, respectively, with hydrophilic or hydrophobic molecules. (2) PEGylated types:
The surfaces of the liposomes are coated with polyethylene glycol (PEG) to create
a steric equilibrium. (3) Type of ligands that are intended for attachment to the
outermost layer of the liposome or to the ends of already connected PEG chains
include proteins, carbohydrates, and antibodies and (4) theranostic liposome form:
this type of liposome combines the preceding three and frequently consists of a
nanoparticle as well as elements for treatment, imaging, and targeting [46].
6.3 Dendrimers
6.4 Nanocrystals
Pure solid pharmaceutical nanocrystals have a size between 1000 and 2000 nm. The
majority of the time, they are stabilized by polymeric steric stabilizers or surfac-
tants and are entirely medication lacking any carriers molecules connected to it.
Typically, adding a surfactant agent referred to as nano-suspension will improve a
nanocrystals dispersion in a problematic liquid media. The predominant medium
for dispersion in this scenario is water, but it might also be any other aquatic or
non-aqueous medium-sized, such as liquefied polyethylene glycol and oils [51].
Nanocrystals display unique properties by overcoming problems such as increased
maximum solubility, increased dissolving rate, and enhanced glueyness to surface/
cell membranes. Chitosan microparticles and cinaciguat nanocrystals were combined
by Ni et al. [52] to transport the hydrophobic medication to the lungs. The swelling
and muco-adhesive properties of the polymer were used in the creation of the nanopar-
ticles to maintain the release of drugs. They found that inhalation efficiency could
be diminished in illness-related conditions, thus more study is needed to show the
technique’s more substantial advantages.
6.5 Nanoparticles
them to link to antibodies, medicines, and additional ligands, giving these structures
enhanced viability for use in healthcare research [57]. While gold, iron, silver, and
copper are the most commonly investigated metallic nanoparticles, growing curiosity
has been shown in other types of metallic nanoparticles, including, gadolinium, tita-
nium oxide, platinum, zinc oxide, selenium, cerium dioxide, and palladium as well
[57].
significantly in the years addressed by the present research. Future research into the
utilization of such metals, having gold and silver, for both therapeutics and diag-
nostics may increase the usage of nano-biomedicines. Gold nanoparticles, which
seem to be readily absorbed in delicate cancer cells and render the tumour suscep-
tible to radiation-carried thermal treatment (for instance, in the near-infrared range),
are among the primary sources of fascination in this research. The true influence of
nano-biomedicine and nano-drug methods of administration on the medical field,
especially in the treatment and detection of cancer, is still quite limited, notwith-
standing the widespread knowledge of their future potential. This is due to the topic
being a young one in scientific findings, with just two decades of actual study, and
the fact that many important, basic characteristics are still unexplained. The primary
focus of the upcoming study will be on the fundamental molecular indicators of
diseased tissues, especially the ones that allow for precision targeting despite compro-
mising regular functioning of cells. Eventually, the utilization of nano-biomedicine
will proceed alongside our growing comprehension of illness at the atomic level or
those that represent a nanomaterial-subcellular scale corresponding biomarker detec-
tion thereby opening up new avenues for therapy and diagnosis. Therefore, devel-
oping nanomedicine uses in the years to come will require knowledge of the molec-
ular fingerprints of illness. For more widespread applications of nano-biomedicine
beyond the ones we have discussed in this chapter using popular nanostructures,
more research would be required.
Pharmaceutical activity in tissues/cellular erect, technical advancements for
analysing these events, and the idea of regulated administration of specific medi-
cations at the afflicted regions are still distant from its full potential. The majority
of investigation in the subject of nano-biomedicine is concentrated on biomaterial
and compositional studies, which appear to represent the infancy of biomedicine
applications. Valuable knowledge that might be employed in medical treatment and
diagnostic studies will be obtained through time- and money-consuming transdis-
ciplinary research and experiments on animals. A growing worldwide movement is
the pursuit of more precise medical diagnosis and therapies, and the potential of
nano-biomedicine and nano-drug delivery systems seems bright.
The creation of nanostructures that serve as tissue diagnostic and repair mecha-
nisms with complete control from outside characteristics has garnered a lot of atten-
tion. This is still hypothetical research that hasn’t yet materialized but that humans
could accomplish in a few years. However, just as for their benefits, any drawbacks
of nanomedicine need to be carefully examined for both people and the environment
in its entirety. As a result, a careful analysis of the possible short-term or long-term
negative impacts of new nanomaterials on humans and the natural world is necessary.
Nano-biomedicines’ affordable cost adds another area for research that needs more
contributions as they gain in popularity. The regulation of nano-biomedicines, which
was covered in greater detail in the chapter above, will advance along with novel
applications for these substances.
276 P. Sharma
8 Conclusion
The most recent advances in nano-biomedicine are highlighted in the present chapter,
including innovative methods for diagnosis as well as technological advances in the
delivery of both new and old drugs. There are several reported nano-dimensional enti-
ties, particularly nanostructures. These molecules can be utilized in practical systems
for pharmacological activation, sensing, precise delivery to targets, and diagnostics.
Initially, the main application of nanotechnology was to enhance the bioavailability,
controlled administration, and dissolution of pharmaceuticals. The application of
nanotechnology to boost the therapeutic value of by now-known natural bioactive
chemicals has become common practise, yet the creation of nanopharmaceuticals is
plagued in ambiguity and searching for compounds with pharmacological activity
naturally occurring is not as common compared to it had been fifty years ago. The
medical efficacy of these organic compounds has been greatly enhanced by the
use of nanocarriers created with solid lipid nanoparticles, micelles, liposomes as
crystal nanoparticles, and dendrimers along with gold, cadmium sulphide, silver,
and titanium dioxide polymeric nanoparticles.
Natural, new biomaterials continue to be in demand due to their biodegradability,
biocompatibility, accessibility, potential to regenerate, and low toxicity. Beyond
merely acknowledging such proteins and polysaccharides as organic biopolymers,
one of the most cutting-edge research topics at the moment is on enhancing their
resilience in their presence of an alive platform and production circumstances. Also
widely used are polymer nanoparticles (nanocapsules and nanospheres) produced
via emulsion polymerization, solvent evaporation, and surfactant-free emulsion poly-
merization. In the past several years, as nano-biomedicine has expanded, a lot of focus
has been placed on the interconnected of therapy and identification (theranostic), that
employs tumours as a disease model.
Examples include the employing of cathepsin B to identify propagate, fluorogenic
protein devices connected to glycol-based chitosan the nanoparticles, photodynamic
colon cancer recognition via folic and alginate acid-mixed chitosan nanoparticles, the
use of iron oxide covered biopolymeric (hyaluronic acid) substances in carcinoma
treatment, and dextran. Since the 1990s, a staggering number of nanotechnology-
based products after approval of FDA as study projects have been developed, which
include polymer synthetic particles, liposome compositions, nanocrystals, micellar
particles, protein’s nanostructures, and many more. When paired with medications or
biological agents, these things are frequently used. Even though nano-biomedicine
has already significantly changed how we locate and use medications in biological
systems, the main focus of subsequent studies will be on evaluating the safety/
toxicity of nano-biomedicines and their approval procedures. We can now identify
illnesses and, in certain cases, connect a diagnosis to a treatment. Advances in nano-
biomedicine are to blame for this.
Funding There was no funding for this project from any source.
References
1. Swamy, M.K., Sinniah, U.R.: Patchouli (Pogostemon cablin Benth.): botany, agrotechnology
and biotechnological aspects. Ind Crops Prod. 87, 161–76 (2016)
2. Mohanty, S.K., Swamy, M.K., Sinniah, U.R., Anuradha, M.: Leptadenia reticulata (Retz.)
Wight & Arn. (Jivanti): botanical, agronomical, phytochemical, pharmacological, and biotech-
nological aspects. Molecules 22, 1019 (2017)
3. Rodrigues, T., Reker, D., Schneider, P., Schneider, G.: Counting on natural products for drug
design. Nat. Chem. 8, 531 (2016)
4. Siddiqui, A.A., Iram, F., Siddiqui, S., Sahu, K.: Role of natural products in drug discovery
process. Int. J. Drug Dev. Res. 6(2), 172–204 (2014)
5. Beutler, J.A.: Natural products as a foundation for drug discovery. Curr. Prot. Pharmacol. 46(1),
9–11 (2009)
6. Thilakarathna, S.H., Rupasinghe, H.: Flavonoid bioavailability and attempts for bioavailability
enhancement. Nutrients 5, 3367–3387 (2013)
7. Bonifacio, B.V., da Silva, P.B., dos Santos Ramos, M.A., Negri, K.M.S., Bauab, T.M., Chorilli,
M.: Nanotechnology-based drug delivery systems and herbal medicines: a review. Int. J.
Nanomed 9, 1 (2014)
8. Watkins, R., Wu, L., Zhang, C., Davis, R.M., Xu, B.: Natural product-based nanomedicine:
recent advances and issues. Int. J. Nanomed. 10, 6055 (2015)
9. Martinho, N., Damgé, C., Reis, C.P.: Recent advances in drug delivery systems. J. Biomater.
Nanobiotechnol. 2, 510 (2011)
10. Jahangirian, H., Lemraski, E.G., Webster, T.J., Rafiee-Moghaddam, R., Abdollahi, Y.A.P.:
Review of drug delivery systems based on nanotechnology and green chemistry: green
nanomedicine. Int. J. Nanomed. 12, 2957 (2017)
11. Sharma, P., Tailang, M.: In vivo study of orodispersible tablet of primaquine. Int. J. Pharm.
Sci. Res. 9(8), 3506–3510 (2018)
12. Sharma, P., Bhargava, S., Parashar, D., Mangal, A.: Formulation and evaluation of nanoparticles
containing cyclophosphamide. WJPR 7(1), 785–798 (2017)
13. Park, S.H., Oh, S.G., Mun, J.Y., Han, S.S.: Loading of gold nanoparticles inside the DPPC
bilayers of liposome and their effects on membrane fluidities. Colloid Surf. B 48, 112–118
(2006)
14. Mirza, A.Z., Siddiqui, F.A.: Nanomedicine and drug delivery: a mini review. Int. Nano Lett. 4,
94 (2014)
15. Wang, N., Feng, Y.: Elaborating the role of natural products-induced autophagy in cancer
treatment: achievements and artifacts in the state of the art. BioMed. Res. Int. 2015, 934207
(2015)
16. Abdelwahab, S.I., Sheikh, B.Y., Taha, M.M.E., How, C.W., Abdullah, R., Yagoub, U., El-
Sunousi, R., Eid, E.E.: Thymoquinone-loaded nanostructured lipid carriers: preparation, gastro-
protection, in vitro toxicity, and pharmacokinetic properties after extravascular administration.
Int. J. Nanomed. 8, 2163 (2013)
17. Schneider, C., Langer, R., Loveday, D., Hair, D.: Applications of ethylene vinyl acetate
copolymers (EVA) in drug delivery systems. J. Controlled Release 262, 284 (2017)
18. Langer, R.S., Peppas, N.A.: Present and future applications of biomaterials in controlled drug
delivery systems. Biomaterials 2(4), 201–214 (1981)
19. Sharma, P., Jain, V., Tailang, M.: Selection and role of polymers for designing of a drug carrier.
Drug Carriers. IntechOpen, London (2022). https://doi.org/10.5772/intechopen.103125
20. Sharma, P., Jain, V., Tailang, M.: Advancement of nanocarrier-based engineering for specific
drug delivery for cancer therapy. In: Targeted cancer therapy in biomedical engineering.
Singapore: Springer Nature Singapore, pp. 465–486 (2023)
278 P. Sharma
21. Kamaly, N., Xiao, Z., Valencia, P.M., Radovic-Moreno, A.F., Farokhzad, O.C.: Targeted poly-
meric therapeutic nanoparticles: design, development and clinical translation. Chem. Soc. Rev.
41(7), 2971–3010 (2012)
22. Joshi, R., Raje, S., Akram, W., Garud, N.: Particle engineering of fenofibrate for advanced drug
delivery system. FJPS 5, 1–1 (2019)
23. Saadeh, Y., Vyas, D.: Nanorobotic applications in medicine: current proposals and designs.
Am. J. Robot. Surg. 1, 4–11 (2014)
24. Sharma, P., Tailang, M.: Opportunities and challenges to develop transdermal patch with
nanocarrier. WJPR 6(15), 392–400 (2017)
25. Lu, H., Wang, J., Wang, T., Zhong, J., Bao, Y., Hao, H.: Recent progress on nanostructures for
drug delivery applications. J. Nanomater. 2016, 20 (2016)
26. Kumari, A., Kumar, V., Yadav, S.: Nanotechnology: a tool to enhance therapeutic values of
natural plant products. Trends Med. Res. 7, 34–42 (2012)
27. Pankaj, S., Mukul, T.: PH dependent release potential of natural polymers in sustained release
of ornidazole from colon targeted delivery system. Magnesium 10(10), 10 (2019)
28. Nie, S.: Understanding and overcoming major barriers in cancer nanomedicine. Nanomedicine
(Lond.) 5, 523–528 (2010)
29. Sharma, P., Tailang, M.: Design, optimization and evaluation of buccal drug delivery system
of propranolol for hypertension treatment. Int. J. Pharm. Sci. Res. 11(1), 301–311 (2020)
30. Maeda, H., Nakamura, H., Fang, J.: The EPR effect for macromolecular drug delivery to
solid tumors: improvement of tumor uptake, lowering of systemic toxicity, and distinct tumor
imaging in vivo. Adv. Drug Deliv. Rev. 65, 71–79 (2013)
31. Jain, R.K., Stylianopoulos, T.: Delivering nanomedicine to solid tumors. Nat. Rev. Clin. Oncol.
7, 653–664 (2010)
32. Sharma, P., Tailang, M.: Design, optimization, and evaluation of hydrogel of primaquine loaded
nanoemulsion for malaria therapy. FJPS 6, 1–1 (2020)
33. Bertrand, N., Wu, J., Xu, X., Kamaly, N., Farokhzad, O.C.: Cancer nanotechnology: the impact
of passive and active targeting in the era of modern cancer biology. Adv. Drug Deliv. Rev. 66,
2–25 (2014)
34. Bao, G., Mitragotri, S., Tong, S.: Multifunctional nanoparticles for drug delivery and molecular
imaging. Annu. Rev. Biomed. Eng. 15, 253–282 (2013)
35. Sosnik, A.: Alginate particles as platform for drug delivery by the oral route: state-of-the-art.
ISRN Pharm. 2014, 926157 (2014)
36. Patil, N.H., Devarajan, P.V.: Insulin-loaded alginic acid nanoparticles for sublingual delivery.
Drug Deliv. 23, 429–436 (2016)
37. Roman, J.V., Galan, M.A., del Valle, E.M.M.: Preparation and preliminary evaluation of algi-
nate crosslinked microcapsules as potential drug delivery system (DDS) for human lung cancer
therapy. Biomed. Phys. Eng. Expr. 22, 035015 (2016)
38. Jain, A., Jain, S.K.: Optimization of chitosan nanoparticles for colon tumors using experimental
design methodology. Artif. Cells Nanomed. Biotechnol. 44, 1917–1926 (2016)
39. Sun, B., Zhang, M., Shen, J., He, Z., Fatehi, P., Ni, Y.: Applications of cellulosebased materials
in sustained drug delivery systems. Curr. Med. Chem. 26(14), 2485–2501 (2017). https://doi.
org/10.2174/0929867324666170705143308
40. Agarwal, T., Narayana, S.G.H., Pal, K., Pramanik, K., Giri, S., Banerjee, I.: Calcium alginate-
carboxymethyl cellulose beads for colon-targeted drug delivery. Int. J. Biol. Macromol. 75,
409–417 (2015)
41. Goswami, S., Naik, S.: Natural gums and its pharmaceutical application. J. Sci. Innov. Res. 3,
112–121 (2014)
42. Huang, J., Deng, Y., Ren, J., Chen, G., Wang, G., Wang, F., Wu, X.: Novel in situ forming
hydrogel based on xanthan and chitosan re-gelifying in liquids for local drug delivery.
Carbohydr. Polym. 186, 54–63 (2018)
43. Miyata, K., Christie, R.J., Kataoka, K.: Polymeric micelles for nano-scale drug delivery. React.
Funct. Polym. 71, 227–234 (2011)
Nanostructured Biomaterials in Drug Delivery: Current Trends … 279
44. Xu, W., Ling, P., Zhang, T.: Polymeric micelles, a promising drug delivery system to enhance
bioavailability of poorly water-soluble drugs. J Drug Deliv. 2013, 340315 (2013)
45. Bozzuto, G., Molinari, A.: Liposomes as nanomedical devices. Int. J. Nanomed. 10, 975 (2015)
46. Sercombe, L., Veerati, T., Moheimani, F., Wu, S.Y., Sood, A.K., Hua, S.: Advances and
challenges of liposome assisted drug delivery. Front Pharm 6, 286 (2015)
47. Madaan, K., Kumar, S., Poonia, N., Lather, V., Pandita, D.: Dendrimers in drug delivery and
targeting: drug-dendrimer interactions and toxicity issues. J. Pharm. Bioallied Sci. 6, 139 (2014)
48. Tripathy, S., Das, M.: Dendrimers and their applications as novel drug delivery carriers. J.
Appl. Pharm. Sci. 3, 142–149 (2013)
49. Kesharwani, P., Jain, K., Jain, N.K.: Dendrimer as nanocarrier for drug delivery. Progr. Polym.
Sci. 39, 268–307 (2014)
50. Jain, K., Gupta, U., Jain, N.K.: Dendronized nanoconjugates of lysine and folate for treatment
of cancer. Eur. J. Pharm. Biopharm. 87, 500–509 (2014)
51. Du, J., Li, X., Zhao, H., Zhou, Y., Wang, L., Tian, S., Wang, Y.: Nanosuspensions of poorly
water-soluble drugs prepared by bottom-up technologies. Int. J. Pharm. 495, 738–749 (2015)
52. Ni, R., Zhao, J., Liu, Q., Liang, Z., Muenster, U., Mao, S.: Nanocrystals embedded in chitosan-
based respirable swellable microparticles as dry powder for sustained pulmonary drug delivery.
Eur. J. Pharm. Sci. 99, 137–146 (2017)
53. Misra, R., Sahoo, S.K.: Intracellular trafficking of nuclear localization signal conjugated
nanoparticles for cancer therapy. Eur. J. Pharm. Sci. 39, 152–163 (2010)
54. Lanjhiyana, S.K.: Polysaccharides based novel and controlled released multiparticulate systems
for colon-specific delivery: Contemporary scenario and future prospects. AJP 14(4) (2020)
55. Acharya, S., et al.: Targeted epidermal growth factor receptor nanoparticle bioconjugates for
breast cancer therapy. Biomaterials 30, 5737–5750 (2009)
56. McNamara, K., Tofail, S.A.: Nanoparticles in biomedical applications. Adv. Phys. 2, 54–88
(2017)
57. Kudr, J., et al.: Magnetic nanoparticles: from design and synthesis to real world applications.
Nanomaterials 7, 243 (2017)
58. Cai, W., Chen, X.: Nanoplatforms for targeted molecular imaging in living subjects. Small
3(11), 1840–1854 (2007)
59. Kim, K.Y.: Nanotechnology platforms and physiological challenges for cancer therapeutics.
Nanomed.: Nanotechnol. Biol. Med. 3(2), 103–110 (2007)
60. Morrow Jr.K.J., Bawa, R., Wei, C.: Recent advances in basic and clinical nanomedicine. Med.
Clin. North Am. 91(5), 805–843 (2007)
61. Li, D., Li, G.P., Guo, W., Li, P., Wang, E., Wang, J.: Glutathione-mediated release of functional
plasmid DNA from positively charged quantum dots. Biomaterials 29(18), 2776–2782 (2008)
62. Pandit, A., Zeugolis, D.I.: Twenty-five years of nano-bio-materials: have we revolutionized
healthcare? Future Med. 11(9), 985–987 (2016)
280 P. Sharma
Abstract Biomaterials are materials utilized to replace a human body part that
serves the same function as the original or restore the function of damaged or degen-
erated tissues and organs. Implants such as sutures, bone plates, joint replacements,
ligaments, vascular grafts, heart valves, intraocular lenses, and dental implants have
overtaken alternative therapy approaches and entered the mainstream of dental care
over the past ten years. One of the primary requirements for a biomaterial to be suit-
able for medical applications or implantation is that it should be nontoxic, not cause
any immune response, and be chemically stable, biocompatible, and well-tolerated
by the human body. Different materials like metals, ceramics, and polymers, are used
for the manufacturing of implants, depending on whether a permanent or temporary
implant is needed. However, they exhibit a number of disadvantages, including toxi-
city, a lack of mechanical stability, and processing complexity. Numerous bioma-
terials have been discovered, but most implanted biomaterials now in use cause
either acute or chronic inflammatory reactions inside the body. Nanotechnology has
aided in developing an entirely novel implant material with enhanced efficacy, low
cost, and a significant surface-to-volume ratio. Modifying the surfaces of present
implants to reduce the body’s reaction and enhance the natural healing of wounds is
also one strategy for developing such healing biomaterials. This chapter covers the
advancements in biomaterials used for implants, including various techniques for
physically and chemically modifying the surfaces, as well as the use of 3D printing
and nanotechnology.
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 281
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_10
282 R. Shivgotra et al.
1 Introduction
Contributions from scientists and engineers are resulting in significant new solutions
for critical medical issues in a scientific world that is changing rapidly. Diabetes,
osteoporosis, asthma, cardiac issues, cancer, and other critical disorders are no longer
solely treated with conventional pharmaceutical formulations [1]. Novel possibilities
for treating and curing disease are constantly being opened up and it is now under-
stood that a drug’s therapeutic value and potency are not directly linked together;
rather, they are linked with the process of developing drug formulation and absorp-
tion into the body. As the biotechnology sector advances with the emergence of
novel categories of biopharmaceuticals, there is a crucial need to enhance our funda-
mental understanding of the influence of drug delivery methods on safety and effi-
cacy. Additionally, exploring innovative delivery methods is essential to address the
unique challenges presented by these new biopharmaceuticals. Despite advances in
the field, delivering drugs effectively still presents a significant challenge, owing to
our incomplete knowledge of the biological obstacles that impede the delivery of
drugs. Because of these unresolved necessities and constraints, significant research
efforts have been directed toward designing, implementing, and translating biomate-
rials for drug delivery [2]. The utility and significance of metallic materials and their
composites have been considered since the early nineteenth century. Over the last 2–3
centuries, a lot of advancement has been made in the synthetic materials field to gain
insight into how biomaterial interacts with the human body and their effective use
[3]. Surgical repair is frequently required as a consequence of trauma, degradation,
and diseases. Biomaterials are synthetic substitutes that can replace various skeletal
components, including the joints of the knees, hips, fingers, and elbows, vertebrae,
teeth. These substances are designed to perform the same functions as the living
materials they replace [4].
A biomaterial is generally defined as “a nonviable material used in a medical
device, intended to interact with biological systems.” If we remove the phrase “used
in a medical device,” this definition becomes more comprehensive of the extensive
range of applications that involve the integration of biology with synthetic and altered
natural materials. The different biological functions of human tissue are replaced or
repaired using biomaterials, developed by humans and constantly exposed to bodily
fluids that facilitate daily human activities [3]. Biomaterials are materials, whether
natural or synthetic, that come into contact with biological fluids, blood, and tissue.
Their design aims to ensure that they do not have any negative impact on the living
organism or its components while serving various purposes such as prosthetic, ther-
apeutic, and diagnostic applications. Distinguishing between the terms “Biomate-
rial” and “Biological material” is crucial. A biological material refers to a material
that has been created by a biological system, such as the epidermis or an artery.
In contrast, biomaterials are materials that have been engineered to interact with
biological systems, often used in medical or scientific applications [5].
The term “bioimplants” refers to engineered medical devices developed to replace
biological structural human body parts that are malfunctioning or damaged and
Advancement in Biomaterials in the Form of Implants 283
support the given host [6]. In the past ten years, biomaterials have attracted greater
attention as an option for improving and even saving the lives of numerous patients. In
the United States alone, over 13 million medical devices are implanted each year [7].
The need for mechanical implantable devices is growing daily as a result of abrupt
shifts in the global population’s age distribution. The proportion of people over the
age of 60 in the population is dramatically growing, which has increased consumer
interest in artificial implants. By 2050, there is expected to be a 39% increase in the
demand for medical implants [4]. The medical industry has extensively explored and
created a diverse range of implants and devices for numerous applications within the
human body. The main objective of these devices is to safeguard human lives. This
encompasses both the technologies that augment the performance of natural human
organs, like pacemakers, as well as artificial devices that offer physical assistance,
such as knee replacements and synthetic blood vessels [8]. Figure 1 depicts the ideal
properties of the biomaterials that biomaterials should possess for their effective use
in various medical implant applications within the human body.
Considering all the progress made so far in material and biological research, we
have come a long way since the early Egyptians and South-Central American cultures
made use of implants. In the modern era, the quality and as well as availability of
implantable material have significantly improved, leading to the development and
widespread use of this promising therapeutic modality. Tooth implants have a long
Fig. 1 Desirable properties of biomaterials for effective use in medical implants within the human
body
284 R. Shivgotra et al.
history, dating back to ancient China and Egypt where they were made of stone and
ivory. During the sixteenth and seventeenth centuries, dental implants frequently
utilized gold and ivory as their primary materials. In the early twentieth century,
implants made up of metals such as gold, lead, tantalum, stainless steel, and cobalt
alloy were introduced as options for dental implant materials [9].
Biomedical implants, in general, involve a wide range of medical therapies for
various health issues, such as medical devices that can be implanted in the cardiovas-
cular system including stents, vascular grafts, heart valves, defibrillators, pacemakers,
and other related devices. Neural implants are another type of implantable medical
device, which can be used in different parts of the nervous system [10]. The “foreign-
ness” of these biomaterials is a major obstacle for the current implant technology
which should be overcome to enhance host tolerance, achieve the required release
of drugs either in a specific location or over a particular time period, and minimize
reactivity to foreign bodies [11]. As immunologists continue to gain insight into
immune and foreign body reactions, the production of biocompatible materials with
excellent performance will become progressively significant for the manufacturing of
devices that are implanted for the controlled release of drugs, cell-based treatments,
implantable detectors, and tissue engineering [2].
In this chapter, the biological characteristics of different biomaterials utilized in
implant manufacturing—which involves metals, polymeric materials, and titanium
alloys—are reviewed. With the objective to enhance the properties of metal implants,
the chapter also discusses the essential novel advancement in techniques like
surface modifications, 3D printing, and nanotechnology, utilized in their fabrication.
Applications of medical implants in daily life have also been discussed.
To create a biomaterial that is both safe and dependable for use in implants, it is
necessary to identify an appropriate blend of mechanical, chemical, physical, and
biological features that can withstand the rigors of long-term use without requiring
further revision surgeries. Table 1 outlines key properties that a biomaterial must
possess in order to function effectively within the human body [9, 12, 13].
Table 1 (continued)
Bulk properties
Surface roughness The surface roughness of a biomaterial can impact its
biocompatibility, osseointegration, and wear resistance, among
other factors. By enlarging the implant’s surface area in contact
with the bone and modifying the surface’s roughness, the
adherence of cells to the bone is improved, thereby influencing the
reaction of cells and tissues. Different elements, including
roughness, texture, and the orientation of abnormalities, have been
used to categorize implant surfaces
Chemical properties
Pitting corrosion When an implant containing small holes or pits on the material’s
surface makes contact with a solution, the metal ions in the area
dissolve, eliminating their positive charges in combination with
chlorine ions resulting in pitting corrosion causing tiny pits or holes
to develop on the material’s surface., which can compromise its
mechanical characteristics and increase the risk of implant failure
Crevices Corrosion This happens at the junction connecting an implant screw with the
bone. Crevice corrosion takes place by the positively charged
environment produced when metallic ions dissolve
Galvanic corrosion When two metals that are not compatible with one another come
into touch with an electrolyte, such as physiological fluids,
galvanic corrosion occurs
Electrochemical corrosion It occurs when a material is exposed to an electrolyte (such as
body fluids) and undergoes oxidation and reduction reactions that
result in the discharge of metal ions and the deterioration of the
material over a period of time
Additionally, it must be resistant to wear and corrosion in the human body’s environ-
ment. The efficiency of materials employed for implants will be determined by these
characteristics [9]. Several materials that can be employed as biomaterials for implant
applications have been produced over the past few years considering the criteria
outlined above. However, it might be hard for a single metallic substance to have all
the required characteristics. The three main categories of biomaterials employed in
biomedical purposes are ceramics, polymers, and metallic systems (Fig. 2) [6].
3.1 Ceramics
Among the materials used to design biomaterials, ceramics are one of the types.
Examples of such materials include zirconium oxide, aluminum oxide, and bioac-
tive glass which have higher yield strength, greater bioactivity, and osteointegration
with bone, hydroxyapatite (HAp), along with different ceramics consisting of silica
and calcium [5]. The inertness of ceramics within the body, their effortless molding
into a range of forms and porosities, significant strength during compression, and
Advancement in Biomaterials in the Form of Implants 287
superior wear properties all served as the rationale for their usage. Because of their
superior robustness, resistance against biodegradation, and low electrical and thermal
conductivity with a variety of diverse elastic properties, ceramics were first used for
surgical implant devices [9]. Ceramics are also utilized for orbital and middle ear
implants, bone grafts, dental and orthopedic implants, heart valves, coatings to make
metallic implants more biocompatible, artificial knees, hip prostheses, bone trans-
plants, and parts of the musculoskeletal system. For instance, higher Ca/P ratios in
hydroxyapatite-based ceramics are preferred because of their chemical similarities
between the hard tissues of teeth and bone. Ceramics are being used for bioim-
plant applications more frequently as a result of these desirable characteristics [6].
Ceramics are chemically inert, but because of their poor ductility and underlying brit-
tleness, they must be handled carefully and replaced and their use has been restricted
as a result [14].
3.1.1 Bioceramics
Ceramics are used to formulate biomaterials but are less frequently used than either
metals or polymers. But because of their ideal biocompatibility and ability to integrate
with bone, in addition to the fact that these materials are highly comparable to the
mineral constituents of bones, bioceramics of phosphates are frequently employed
for developing optimal biomaterials [5]. Bio-ceramics are biomaterials utilized as
288 R. Shivgotra et al.
a cementing agent, supportive layer, and implant for repairing and replacing weak-
ened skeletal system components. Bioceramics comprise about 50% of all biomate-
rial implants currently in use [15]. Bio-ceramics are a filler material used in orthopedic
surgery to detect bone abnormalities. According to reports, “around 2 million bone
grafting procedures, 280000 fractures of the hips, 700000 spinal tissues, and 250000
wrist fractures bone replacements have been performed throughout all over the world
every year.” Consequently, the demand for bio-ceramics is rising. Depending on
the way they interact with the host bone, bio-ceramics may be divided into three
groups: bio-inert ceramics, like alumina and zirconia, bioactive ceramics, including
hydroxyapatite, and bioresorbable or biodegradable ceramics [16].
Bioinert Ceramics
Bioinert ceramics are a type of bioceramic materials that are characterized by their
inertness and biocompatibility with living tissues. These ceramics are composed of
materials such as alumina, zirconia, and other metal oxides, which do not react with
biological systems and are not easily degraded by the body. Bioinert ceramics have
numerous potential uses in the medical field because of their biocompatibility along
with their ability to resist corrosion and wear and are commonly used in orthopedic
implants as well as in bone grafts and other medical devices [17].
Aluminum oxide, commonly referred to as alumina, is a bioinert ceramic mate-
rial extensively used for the production of orthopedic implants including hip and
knee replacements [18]. Alumina is the best suitable material that can be used for
implants that will be exposed to continuous stress and movement because of its
superior mechanical properties, biocompatibility, and exceptionally high level of
toughness and hardness. The ability of alumina to facilitate bone growth and attach-
ment, which plays a crucial role in the implant’s persistent efficacy, is the material’s
primary advantages when used in implants. Alumina implants also have a low rate
of wear, which reduces the number of debris discharged into the body and lowers the
chance of inflammation, implant loosening, and other challenges [6, 19]. Zirconia,
commonly called zirconium oxide, is the other bioinert ceramic material widely
employed in orthopedic implants, particularly dental implants. it can tolerate both
higher stresses and high temperatures. Zirconia ceramics offer several advantages,
such as high mechanical durability, biocompatibility, aesthetic appeal, and resistance
to wear and chemical degradation. However, there are also some limitations associ-
ated with zirconia implants. They may be more prone to fractures since they are more
brittle than conventional metal implants. Zirconia implants are also more expensive
and require a more sophisticated manufacturing procedure in comparison to metal
implants [17, 20]. Carbon in various forms, including diamond, glass oil, and graphite
is biocompatible and does not trigger an immunological reaction. Regarding dura-
bility and elastic modulus, carbon is fragile and very similar to bones. As a result,
it is biocompatible and appropriate to be used in coating medical equipment. Inves-
tigations and reports have been made on the usage of carbon in the manufacture of
orthopedic and cardiovascular implants. As a bone covering for implants, pyrolyzed
Advancement in Biomaterials in the Form of Implants 289
carbons are employed. Although it was initially employed for dental implants, the
material’s inadequate strength eventually led to its discontinuation. To enhance the
rigidity and longevity of implants of knee joints, they tend to be reinforced with
carbon fibers, while the implants that are subjected to bear heavy loads may utilize
coatings of diamond to improve their lubricating properties [17, 21].
Bio-active Ceramics
Ceramics that are capable of interacting with living tissues and contribute to the
process of healing and regeneration are referred to as bioactive ceramics. Because
of their biocompatibility and capacity to adhere to bone tissue, these ceramics are
frequently employed in medical applications such as bone grafts and dental implants.
Materials like Hap, tricalcium phosphate, and bioactive glass are examples of bioac-
tive ceramics. Bioactive ceramics may promote new bone formation and integra-
tion when used in medical implants, improving results and lowering the chance of
implant failure [17]. Due to its remarkable Osseointegration and biocompatibility,
HA attracted considerable amounts of interest [22]. Bioactive glass is composed
of a silicone oxide network, along with certain network-modifying oxides such
as magnesium oxide, and potassium oxides. The behavior of bio-glass ceramics,
which contain approximately 60% silica is similar to that of inert ceramics [23]. It is
possible to develop bioactive glasses in a variety of shapes, including microspheres
and porous implants. Animals with bioactive glass have been found to have various
bone formations. These possess advantageous properties, such as the tendency to
chemically connect to the tissue, promote osseointegration, and exhibit resistance
to bio-fouling. In maxillofacial surgery, dentistry, and orthopedics, bioactive sili-
cate glasses are frequently utilized. While such materials are unsuitable for implants
that are exposed to heavy loads due to their poor mechanical strength, they are
often utilized to coat such implants for added benefits [24]. Glass ceramics possess
acceptable thermal, mechanical, scratch, and abrasion resistance qualities. which
makes them well-suited for use in orthopedic applications. The production process
involves the use of metal alkoxides gel, which is heated at high temperatures to
create amorphous glass. This glass is then subjected to further heating to induce
crystal formation, and the resulting crystals are used in implants [6, 17].
Bioresorbable Ceramics
Bioresorbable ceramics are a type of material that can be absorbed by the body
over time, eventually breaking down into natural components that can be safely
metabolized or excreted. Bioresorbable ceramics demonstrate minimal reactivity
with the tissues of the host body. Owing to their superior biocompatibility and chem-
ical interactions, bioresorbable ceramics are frequently utilized in the fields of dental
and orthopedic applications. Compared to conventional non-bioresorbable materials,
these types of materials provide various advantages, including lower risk of long-term
290 R. Shivgotra et al.
issues, better healing results, and the opportunity to avoid the need to have another
surgery to remove the implant. However, bioresorbable ceramics have drawbacks as
well, including decreased mechanical strength when compared to non-bioresorbable
materials and possible health issues brought on by their degradation products [25].
In the nineteenth century, during the Industrial Revolution, metal implants became
very popular. There was an enormous demand for body tissue and bone regeneration
during World War 1, which triggered a surge in the utilization of metal implants
[26]. Several polymers along with composites are now been developed in order
to meet the need for implants and advancements in technology, several different
polymers and composites have now been created. need for implants and technological
advancement. Due to their mechanical strength and biocompatibility, metals are still
predominantly used as implantable materials. The inherent properties of these metals
have been further enhanced via alloys. Only a few metals and metal alloys have been
used in medical implants, including stainless steel (316L), titanium and alloys (Cp-
Ti, Ti6Al4V), aluminum alloys, zirconium-niobium, and tungsten heavy alloys [5,
27]. Despite the development of several metals and alloys, some of them are harmful
to humans and do have not much strength, making them incompatible with their
use. Metal implants are used most frequently in the medical industry and are often
employed as substitutes for hip, knee, plate, and dental implants as well as pins, dental
materials, and screws. Also, numerous medical products made from metal, including
craniofacial plates, and screws, components for artificial hearts, pacemakers, clips,
valves, balloon catheters, bone fixation devices, and dental implants have been made
possible by the field of biomaterials’ rapid growth and development [28].
Biological conditions and implant functions determine the kind of metal that
is employed in biomedical implants. The majority of implants, from cardiovas-
cular to otorhinology, are composed of 316L-type stainless steel. Cobalt-Chromium-
Molybdenum alloys are employed when the implant necessitates a high level of resis-
tance to wear, such as in prosthetic joints [5]. Compared to ceramics and polymeric
materials, the superior tensile strength and degradation limit of metals give manufac-
turers the ability for developing implants that can withstand high mechanical stress.
Metals, however, possess weaker strengths and modulus of elasticity than ceramics
and are more susceptible to failure with strain [13].
When a biomaterial of metal-based gets implanted in the biological medium,
the biomaterial’s surface may alter and disintegrate, generating certain by-products
like ions and debris. Interactions within the surface of the metallic implant and
cells/tissues take place as a result of this releasing process. The majority of alloys
allow metal ions to enter the plasma. The chance of ion accumulation in organs like
the liver and spleen, which subsequently form particulates and interfere with these
organs’ normal function, is increased when there is an excessive discharge of ions
in the blood [29]. When this condition continues to occur, it causes cytotoxicity
Advancement in Biomaterials in the Form of Implants 291
and eventually organ failure. Also, the human body does not fully accept metal-
based materials, and inadequate implant attachment hinders tissue growth, leading
to discomfort or significant pain at the implant location. Metallic implants have a
larger risk of infection than ceramic implants, and they heal more slowly as well.
These properties have encouraged current researchers to place the greatest emphasis
on recognizing the characteristics of metallic surfaces with the objective to produce
materials that are biocompatible [5]. Despite several shortcomings, metallic implants
should be preferred because of their superior mechanical durability, low cost, and
corrosion resistance. Platinum and gold, which are chemically inert, can be utilized as
bioimplants and do not show corrosion in situ but they are very costly. Consequently,
owing to their superior mechanical strength and improved biocompatibility, Titanium
and Magnesium-based alloys are now frequently utilized in biomedical industries
[30, 31].
The major disadvantage of Mg and Mg-based alloys was the speed at which they
corrode under physiological conditions. Since it has a high electrode potential (2.3 V),
and because of this Mg is extremely prone to corrosion in both aqueous environments
and bodily fluids. The reason for the high rate of erosion is that certain impurities,
such as iron, copper, and nickel, accumulate in Mg during casting along with refining
and speed up corrosion while they exist in concentrations over the permissible level.
For Iron, Nickel, and Copper in Mg, the impurity standards were 35–50 ppm, 20–
50 ppm, and 100–300 ppm, respectively. No electrochemically active cathodic sites
have developed If the above elements exist within these limits, which would accel-
erate corrosion [36]. Because of the rapid in vivo deterioration, speedy corrosion
leads to the frequent emission of byproducts like as hydrogen gases which indicates
that there is a need for surface modifications [6]. It has been explored to alloy with
different elements to combat rapid corrosion. The corrosion-resistant and mechan-
ical characteristics of Mg, for instance, have been improved by the inclusion of
elements like calcium, zinc, silver, aluminum, zirconium, yttrium, and neodymium.
Ca–Mg, Zn–Mg, and Ca–Zn–Mg are common examples and because of this, they
become perfect for replacing bone. The microstructure can be modified in accor-
dance with mechanical properties similar to the types of bone, by carefully selecting
an appropriate element and its composition [37].
3.2.2 Titanium
pitting corrosion. It was determined that corrosion leads Ti to have an adverse impact
on the body as hazardous metal ions were discharged into the rat’s bodies, hence
pure titanium shouldn’t be used when developing durable medical implants. Wear
debris that accumulates between implant surfaces can lead to bone degeneration in
addition to the compromise stability of implants in long term. Because of this, it
is used primarily as femoral stems and tibial trays in hip joint replacements, not as
an articulating component [42]. Thus, Titanium represents one of those commonly
utilized metals for the fabrication of medical implants used in the medical field,
although extensive investigations are required to improve its durability against wear
and tear. Further investigations may concentrate to enhance Ti’s wear resistance via
metal coupling. Considering metals can be hazardous, polymeric coating material is
a more effective solution in order to enhancing their properties [4].
The most common titanium alloy types used in biomedicine are alpha (Ti–
6Al–4V), near-α, α–β, and metastable β (Ti–6Al–7Nb). As a consequence of their
exceptionally high modulus of elasticity values, these alloys, despite being widely
employed as biometallic implants, have issues with shielding stress across the
implant-tissue junction. Vanadium and aluminum compounds present in the phys-
iological environment induce the release of poisonous aluminum and vanadium,
which can have detrimental effects on health. In order to substitute Vanadium and
aluminum in the alloy, a lot of attention has been given to alloys that comprise
Zirconium, Niobium, Tantalum, or Molybdenum [6].
The group of ferrous alloys referred to as stainless steel (SS) is composed of iron
(70%), nickel (10–14%), chromium (16–18%), and carbon (1%) [4]. Stainless steel
is typically used for the medical implant used to replace hips, knees, or shoulders
temporarily. After cardiac surgery, SS is typically used to make guidewires and
sensor wires implanted into our bodies [43]. Chromium provides a thin, passive oxide
coating that shields the implant surface from corrosion. Its mechanical characteris-
tics are enhanced by the inclusion of carbon, primarily the toughness for fractures,
corrosion resistance toward the corrosion, and implants’ tribological function [44,
45]. In India, SS grades 304 and 316L are the most preferred implantable mate-
rials for the medical field because they are more affordable, have a wider range of
resources available to them, are reliable, and are simpler to fabricate than Ti- and
Cobalt-based alloys. AISI type 316L stainless steel is the most prevalent SS grade
that is suitable for implant applications. They are a suitable material for orthopedic
implants due to their load-bearing potential. However, because of a pitting corrosion
attack with the ejection of ions (nickel and chromium), resulting in allergic reactions
near the implant area, about 90% of SS implants of 316L grade lose their properties.
As a result, adding a small percentage of molybdenum (2–4 weight %) increases the
316L stainless steel grade’s ability to resist corrosion and also strengthens it [6, 46].
Due to its better characteristics and reduced toxicity, nickel (Ni)-based SS proved
to be a superior alloy. Implants made of SS are transparent to X-rays; therefore,
294 R. Shivgotra et al.
noble metals (gold or platinum) coatings, are used to augment their radiopacity.
The mechanical properties of the implant shouldn’t be compromised throughout this
procedure, though [47]. Salahinejad et al. [48] evaluated and developed SS free of
Ni considering it is hazardous to humans. With nitrogen in place of Ni, he increased
steel strength and structural stability. In comparison to conventional Ni-based steel,
the results showed excellent strength, superior wear, corrosion, and biocompatibility
resistance.
Regardless of possessing numerous advantages, SS is less frequently used in
biomedical implants because of the reason of corrosion in an environment that
contains chloride, which can lead to metal ions like Ni and chromium to be released
that are hazardous to humans. Furthermore, because it is prone to deformation, its
ductility (flexibility) is restricted. Researchers in the future can therefore concentrate
on boosting the corrosion resistance against corrosion while simultaneously working
to increase the ductility of stainless steel.
In the first decade of the twentieth century, this alloy was first utilized as a hip replace-
ment implant material [26, 49]. Cobalt (Co) based alloys with chromium (Cr) (27–
30%), molybdenum (Mo) (5–7%), and a small amount of manganese, silicon (1%),
iron (0.75%), and Ni (0.5%) are regarded as among the best materials for implant
applications. The other components include nitrogen, carbon, tungsten, phosphorus,
and sulfur boron [50]. Co-based alloys are employed in applications for bioimplants,
especially for developing surgical implants for the hip, knee, shoulder, and broken
bone surfaces, in accordance with in vivo as well as in vitro studies because they
exhibit greater biocompatibility, excellent mechanical, corrosion, and wear proper-
ties [51]. Due to their remarkable combination of strength and flexibility, Co–Cr–Mo
alloys are among the most prevalent types of Co alloy. The alloy mentioned above
has better elastic modulus, density, and hardness when compared to other metallic
implants, making it an optimal choice for the implant process. Also, as these alloys
retain their original qualities for a relatively long period after implantation, this alloy
is typically employed in permanent implant fixation operations. Artificial ankles and
knees are made of a Co–Cr–Mo alloy and ultra-high molecular weight polyethylene
[52].
Implantation of Co–Cr alloy and Ti discs was performed in the subcutaneous layer
of mice. in an in vivo study executed by one of the studies. Following skin closure, the
implanted discs were immediately injected with a suspension comprising S. aureus.
In accordance with the results, Co–Cr alloy discs are more bacterially resistant than
Ti-made discs because they had fewer viable S. aureus compared to Ti discs. Ni,
Mo, and Cr are a few other significant alloying components for Co-based alloys
[53]. When these elements leak from the metal surface of Co alloys into bodily
fluids during corrosion, it has been shown that they are harmful to humans as they
can cause skin diseases as well as damage to the liver, blood cells, kidneys, and
lungs [54]. The mechanical strength and resistance to corrosion of Co–Cr–Mo are
Advancement in Biomaterials in the Form of Implants 295
improved through the inclusion of Ni, but the utilization of this alloy in bioimplants
is constrained due to nickel’s cytotoxicity. The tissue/implant interface experiences a
stress-shielding phenomenon due to the substantially greater elastic modulus (200–
250 GPa) and ultimate tensile strength (400–1000 GPa) of Co-based alloys, which
are ten times greater than those of human bone. Surface modifications of Co–Cr–
Mo alloys during plasma treatment with plasma increase their rigidity, wear, and
corrosion resistance but due to their inferior tensile and frictional characteristics,
they are still not suggested for joint fixtures [55, 56].
Because of their inferior frictional properties and corrosion behavior, Co-based
alloys are not preferred materials for load bearing and joint surfaces. In comparison
with the other Ti or other alloys, manufacturing Co-Cr alloys is more expensive and
tedious as well. By employing titanium or zirconium-based coatings or bioceramics
coatings on a material’s surface, future studies can concentrate on improving the
osteointegration of Co–Cr alloys. The development of Ni-free Co–Cr alloy should
be the main area of study to lessen the toxicity while making them safe for people
to use [4].
Other metals and alloys (Titanium, iridium, gold, platinum, and palladium have
also been employed for developing dental implant devices. Zirconium, hafnium,
and tungsten-based devices have recently been put through testing. These reactive
metal groups and their alloys have been claimed to offer certain valuable benefits.
Metals having relatively low strength values, such as gold, platinum, and palladium,
have a limited range of applications. Due to their nobility and availability, these
metals—especially gold—remain employed as implant materials in surgery [6].
3.3 Polymers
Since the 1980s, medical implants’ properties have been modified by coating
synthetic polymeric materials on them or using them as implantable materials [4].
Small repeating unit monomers (homopolymers and copolymers) are the building
blocks of polymers, which are divided into two distinct categories referred to
as biodegradable and non-biodegradable. Chitosan, Polyacetal, polycaprolactone,
alginate, and polylactide are typical examples of biodegradable polymers, while
polyethylene terephthalate, polymethylmethacrylate, polypropylene, polytetrafluo-
roethylene, etc., are examples of nonbiodegradable polymers [6].
The primary benefit of employing polymers is that they can conveniently have
their composition modified, allowing for the modification of their physical char-
acteristics in accordance with the application. In addition to pacemakers, implants
made of polymers are commonly utilized to replace kidneys, skin, heart valves, bone,
contact lenses, and artificial blood arteries. They can also be utilized to serve as a
binding agent for sealing off the junction within the two materials, or as a protec-
tive covering on its own. In comparison to metals and ceramics, polymers exhibit
lower strengths and elastic moduli. They lack adherence to biological tissue and are
less mechanically and physically stable than metals. Due to biological factors, the
296 R. Shivgotra et al.
like bisphenol, which are mostly present in other polycarbonates, it also exhibits
less adverse impacts on human health PMMA, a non-biodegradable polymer that is
typically used in applications requiring long-lasting durable implants. As a result,
PMMA is among the best biomaterials for covering implants in the body. PMMA
cement’s main drawback is that it doesn’t break down. Additionally, the material’s
high temperature of curing can result in necrosis of the tissue around it, which is a
major concern [61, 62].
3.3.3 Hydroxyapatite
One of the primary minerals found in human bone is HAp of which 70% is made up
of bones; the remaining 30% is collagen and water. The physical and chemical char-
acteristics of synthetic HAp are comparable with bone because HAp constitutes the
majority of bone [63]. It is referred as the most biocompatible material employed in
bone replacement currently on the market, with great bioactivity, osteoconductivity,
and less solubility within fluids. In order to improve their biocompatibility and facil-
itate bone regeneration, HAp is primarily applied as a surface coating to a variety of
titanium or stainless steel-based implants. Additionally, it can be utilized as cement
between permanent implants or for making small, less loaded medical implants. Its
limited use in long-term medical implants is due to its poor mechanical strength and
weak tensile strength [64].
3.3.5 Polyethylene
Polyethylene (PE) can be divided into groups based on their molecular weight, such as
low-density polyethylene and high-density polyethylene, which have various appli-
cations depending on their properties. When PE was used successfully for total
298 R. Shivgotra et al.
3.3.6 Polyurethane
Several implants have made use of Polyurethane (PU), which is also simple to modify
in accordance with various biomedical uses. On the other hand, PU may be degraded
with chemical exposures in vivo, which causes the material to deteriorate. If managed
properly, such type of degradation can be employed to aid in the development of new
tissues [68]. Additionally, it was discovered that PU had lesser water permeability,
which could be reduced even more by adding isopropyl myristate in a very concen-
tration. Capsular contracture is quite infrequent with PU breast implants. Due to its
excellent surface, thermomechanical, and biocompatible capabilities, thermoplastic
PU also exhibits significant potential if combined with polydimethylsiloxane for
application in implants; however, the material is still quite new, and only a few
studies have examined its characteristics [69].
3.3.7 Polypropylene
3.3.8 Polyamide
In both their natural and synthetic forms, polyamides (PA) are macromolecules that
include repeating units connected with amide bonds. Nylon is the usually widely
used type of PA in medical implants along with devices [73]. Although it’s commonly
used as a fiber material in composites to enhance their mechanical strength and in
the production of sutures and dentures, its usage as a material for packaging films is
infrequent [74]. In a recent study on microbiological contamination, nylon had been
evaluated among several other materials and emerged with the lowest contamination
level. A 3D printing technique may be used to efficiently develop nylon and certain
of its composites, such as glass fiber nylon [8].
Silicones are inert materials that are employed in a number of forms and applica-
tions. Silicone implants have found applications in laryngeal surgeries to address
various problems, including incomplete glottis closure and vocal impairment caused
by unilateral vocal fold paralysis. The vocal function of patients has improved,
according to studies that analyze these implants Additionally, silicone serves as
an encapsulant material in implants [75]. Silicone has been discovered to be more
appropriate for extended encapsulation within the body because of its superiority to
epoxy resin and PU coatings smoother topography and lower surface energy [76].
Additionally, the polymer itself is shielded from cells and molecules by these prop-
erties. The two silicone derivatives parylene and polydimethylsiloxane (PDMS) are
frequently used in biomedical implants. In implanted neurological prostheses, pary-
lene is commonly employed as a packaging material; parylene C is the most widely
used of its various forms. In addition to pacemakers, blood pumps, breast prostheses,
shunts, cochlear implants, esophagus replacements, and packaging for implantable
electrical devices and sensors, PDMS is another widely recognized silicone deriva-
tive [68, 77]. For the treatment of ocular posterior segment disorders such as diabetic
retinopathy, an implanted MEMS device that has been designed utilizing PDMS for
direct on-demand drug administration to a human eye was recently developed [8].
Despite their widespread use concerns regarding silicone implant failure, in general,
have been addressed.
Liquid crystal polymers (LCPs) are frequently utilized in microwave frequency elec-
tronics because of their high impact strength and Young’s modulus. In recent studies,
the potential of LCPs as biomaterials for a wide range of implants and devices,
including retinal and brain prosthetic implants, has garnered significant interest. In
one study, it was discovered the usage of LCP packaging for retinal implants had
an impact on pixel density and that the device’s elevated pixel resolution allowed
300 R. Shivgotra et al.
individuals to regain their ability to read and recognize faces. Additionally, LCPs
have been employed to develop a flexible electrode array in rats for in vivo research
on brain functioning and neurological disorders [8, 78]. In their study, Kim et al. [79]
showcased the remarkable MRI compatibility of the material in cochlear implantable
devices. Their findings suggest that the use of LCP packages resulted in reduced
cochlear device size and that they hold promise for future exploration of the auditory
perception mechanism.
Polylactic acid (PLA) is a type of thermoplastic polymer that has the unique properties
of being both biodegradable and compostable. It is made from renewable resources
like corn starch, cassava roots, or sugarcane. It belongs to the family of polyesters
and has various applications, including implants, medical devices, 3D printing, pack-
aging, and textiles. As a widely used bioresorbable polymer, PLA has been studied
for nearly 50 years [17]. The first reports of PLA’s use as sutures and rods for treating
dog jaw fractures date back to the 1960s, however, a Swedish scientist by the name
of Scheele made the discovery of PLA in 1780. PLA implants are particularly useful
because they can be designed to degrade over time, allowing the patient’s tissues to
take over the function of the implant. The two types of PLA that are most frequently
used in the medical field are L-PLA which is hydrolysis resistant (mainly crystalline)
and DL-PLA which is hydrolysis sensitive (primarily amorphous) [80].
Polygalactic acid (PGA) is a synthetic polymer synthesized from glycolic acid, which
is produced by the cyclic acid diester’s opening ring. Due to its strong crystallinity,
poor solubility, and high melting point, PGA was initially utilized in the medical field
for manufacturing biodegradable sutures. The primary advantage of PGA is that it is
non-toxic, can be aggregated, and degrades naturally. Because PGA is hydrophilic,
its breakdown rate is high. Usually, the strength of the implant drops to 50% after
implantation and to 90% within 28 days. Because of this, therapies application of
PLGA, the copolymer of PGA and PLA, is more common [17, 81].
Various biomaterials utilized in the production of implants with their applications,
advantages, and disadvantages are shown in Table 2.
Table 2 An overview of biomaterials in the implant industry: exploring advantages, disadvantages and their applications
Biomaterial Applications Advantages Disadvantages References
Ceramics
Bioinert ceramics (Alumina, Orthopedic implants, such as Durable, long-lasting implants Brittleness, difficulty in [17–19]
zirconia, and titanium dioxide) joint replacements, dental manufacturing, Implant
implants, bone grafts loosening
cardiovascular implants, and
orthodontic implants
Bio-active ceramics Dental implants, Orthopedic Bioactivity, biocompatibility, Not be appropriate for [17, 22, 23]
(Hydroxyapatite, tricalcium implants, Spinal implants, and osteoconductivity, resorbability load-bearing implants due
phosphate, and bioactive glass) maxillofacial implants to their weak mechanical
durability, slow
degradation, the potential
for implant rejection, and
variability in properties
Bioresorbable ceramics Bone grafts, dental implants, Biocompatibility, osteoconductivity, gradual Limited strength and [25]
Advancement in Biomaterials in the Form of Implants
orthopedic implants, drug resorption, design flexibility, and reduced availability, slow
delivery stress shielding, better healing results degradation rate, potential
for foreign body reaction,
and difficulty in removal
Metals and alloys
Magnesium and its alloys Orthopedic implants, Biodegradable, comparable to bones, light Susceptibility to [33, 36, 37]
cardiovascular implants, dental weight, biocompatibility, and superior corrosion, rapid
implants, drug delivery mechanical properties degradation, as well lower
mechanical strength
(continued)
301
Table 2 (continued)
302
The enormous requirement for biomedical implants in the biomedical industry will
likely continue to grow in the future. Bioimplants have emerged as a potentially
revolutionary therapy option for diseases such as blindness, neurological disorders,
orthopedic issues, cardiovascular disease, deformity, and dental deformities [82]. In
recent years, many implant biomaterials have evolved. There are usually two types of
implant failure: mechanical and biological. The strain on the implant is influenced by
our body position while walking, and activities such as jogging, leaping, or cycling
can increase the strain. If the implant is subjected to excessive load, it may experience
mechanical failure. Additionally, biological failure can occur when the body reacts to
the foreign material, and its defense mechanisms interfere with the implant’s function.
The “foreign-ness” of these biomaterials must be overcome for the currently available
implant technology to improve host tolerance, maintain the required temporal and/or
spatial drug release profile, and decrease foreign body reactions. If the treatment fails,
the patient may undergo further procedures and experience significant trauma, along
with the discomfort of their body rejecting the treatment. One of the most important
another aspect among each of the characteristics of a permanent implant is corrosion
resistance. Due to the presence of water, sodium, chlorine, proteins, and amino acids
in the human body, they produce an exceptionally corrosive environment. However,
if the material is not sufficiently adaptable, it will gradually decay, inducing an
emission of hazardous ions, that cause infection and dysfunction in the tissues of the
body [4]. Nanotechnology, 3D printing, novel types of biomaterials, surface coatings,
and modifications are being employed to address issues with current biomaterials
used for implant technology (Fig. 3). The development of a new class of implant
materials with enhanced effectiveness, cost-effectiveness, and high surface area-to-
volume ratio owes much to the crucial role played by nanotechnology. Moreover,
implant surface coating and modification using various techniques are desirable to
improve their mechanical integrity.
A biomaterial’s surface chemistry and topography are crucial factors that affect
protein adsorption, cell interaction, and host reaction [83]. The effective integra-
tion of bioimplants with human body tissues is the main necessity for the implant
process. The surface morphology and chemistry of biomedical implants impart more
command of the biological responses to lifespan and performance [6]. The modifi-
cation of the surface of biometallic materials is being proposed to attain the needed
qualities with the aim to enhance implant success rates, bioperformance, biocompat-
ibility, and osteoconductivity. When the surface is adequately modified, the general
308 R. Shivgotra et al.
Fig. 3 Shows the advancement of biomaterials to address the challenges associated with existing
biomaterials used in implant technology
functionality and attributes of the biomedical implant device won’t be impacted for
a while. The success rate for bioimplants can be considerably improved with the
benefit of bio-integration and the load-bearing capacity of biomaterials [84, 85].
A similar response can be produced in vivo by materials comprised of polymeric,
ceramic, or metallic bases that have various surface properties, ranging from being
hard to soft or hydrophilic to hydrophobic. Non-specific protein adsorption is thought
to be the reason for this observable phenomenon. On the other hand, every process in
nature depends on unique molecular interactions, such as those between proteins and
saccharides. The complex surfaces linked to biological recognition are effectively
regulating biological responses, in contrast to relatively simple surface chemistries
that organisms seem unresponsive to [83].
Nonfouling surfaces, also known as surfaces resistant to protein adsorption, and
more recently surfaces that regulate protein adsorption, have both been the subject
of extensive research [83]. Researchers have made efforts to improve implant bio-
integration by altering the implant surface that eventually comes in contact with the
body. The surface of the implants is being modified utilizing two different methods.
Covering the metallic surface with organic or inorganic materials without altering the
implant substrate is the first method [86]. The second method involves the employ-
ment of conversion coatings or surface-modified layers, in which a substrate’s surface
is chemically altered and a minor thickness increase is a consequence. The substrate
components in this instance play a role in the development of conversion coatings
[87].
Surface preparation through grinding and polishing is essential for conversion
coating in order to increase surface roughness for improved mechanical interlocking
of coatings. Surface modification through the application of an overlay coating is
Advancement in Biomaterials in the Form of Implants 309
advised because this method is critical. In order to achieve the synergy of both char-
acteristics, surface modification, and thin film deposition have recently been coupled
[44]. For enhanced biocompatibility, corrosion resistance, antibacterial activity, and
mechanical qualities, the surface of metallic implants is modified with a suitable
coating material in the evolving biomedical implant industry. Although there are
numerous ways to deposit bioactive surface coatings, a coating approach that is ideal
for biomedical purposes has not yet been established. The coatings on implant mate-
rials are currently applied using one of several deposition techniques, including elec-
trodeposition, electrophoretic deposition, sol-gel procedures, physical and chemical
vapor deposition, and sol-gel procedures [6]. Physical processes involve exposing
the surface to energetic charges or other physical resources such as flame or plasma.
On the other hand, chemical methods entail immersing the surface in chemically
active solutions, which results in the creation of a coating. The Physical method is
regarded as the best among them because it offers precise stoichiometry, high density,
excellent adhesion, and outstanding homogeneity when depositing metal or ceramic
materials on the surface of implants [88].
Among the physical coating techniques, the Physical vapor deposition technique
(PVD) is the most commonly used approach for coating the biomaterials over the
metal implant surfaces. Using a number of physical processes, the material is first
evaporated in this method. When the substance that has evaporated condenses over
the surface of another metallic substrate, the presence of gases like nitrogen and argon
in the vacuum chamber facilitates the formation of a coating [89]. The metal’s surface
topography is unaffected by the PVD method, which also exhibits good tribological
properties. Because of its rapid deposition rate, PVD is capable of producing coatings
with little processing time and basic equipment. Coatings having a thickness of less
than 10 nm can also be produced using PVD [90].
Several studies came to the conclusion that PVD may be an effective and inno-
vative method for coating the surface of biomaterials and is thus widely used in
industries for producing medical implants [91]. Tantalum carbide was coated with
Ti–6Al–4V using the PVD method in one study by Esmaeili et al. [92] and the results
showed that the resulting film was uniform, smooth, and homogeneous. The layer
was both physically and chemically stable, and it strengthened the material’s hard-
ness and resistance to corrosion. Despite this, PVD has several shortcomings. For
instance, the difference in the thermal expansion of the coating and substrate causes
the bonding force between them to weaken. Both the substrate’s mechanical strength
and wear resistance are not greatly increased by PVD. Undercuts are challenging
to coat with PVD, and the process is also costly [93, 94]. Consequently, PVD is an
innovative method with industrial uses for producing medical implants. Its use in
310 R. Shivgotra et al.
the medical field is constrained by the unstable bond in between the substrate and
coating surface.
In the biomedical industry, a plasma arc that has been generated by heating a gas to
high temperatures is used as a heat source in the thermal spray technique known as
“plasma spray technology” for producing surface coatings. Ionized states of matter,
such as plasma, contain neutral particles like neutrons as well as charged particles
like electrons and protons [95]. For coating biomaterials, the plasma spray process
has multiple advantages. With good adhesion to the substrate, it may produce coat-
ings that are homogeneous, and dense, and also enhance the performance and dura-
bility of biomaterials used for implants. Additionally, the coatings can be modi-
fied to have specific features including bioactivity, biocompatibility, and corrosion
resistance A high-energy heat source is utilized during the coating process to melt
and expedite the adherence of minute particles onto a substrate surface. Due to
the process of heat transmission, these molten particles rapidly solidify and cool
down on applying impact. Therefore, the buildup of these tiny particles produces a
coating to form on the surface [96]. Producing coatings for implant surfaces using
this method is among the most practical, simple, and reliable techniques available.
Due to its high operating temperature, it is typically used for developing implants
made of metal with coatings formed of apatite and its derivatives, including bioglass
coating, calcium silicate coating, zirconia, and titanium coating, etc. [97]. Overall,
the development of biomaterials for implants can be aided by plasma spray tech-
nology. This method has several disadvantages, including bioceramic disintegration
during elevated plasma spraying temperatures, significant thermal residual stresses
within the protective coating, and the existence of flaws that include gaps, cracks,
and unmelted particles. The high temperature and gas environment have an impact
on the substrate’s thermal characteristics and crystallinity, which eventually have an
impact on the osteogenic activity of the implant [4].
within the substrate and the film which progressively enhances the structural density
and substrate adhesion. This process enhances the mechanical strength of implants
and provides them with superior corrosion protection. These characteristics make
PIII&D significantly better compared to PVD and other fabrication methods [99].
Zinc ions were applied to the titanium implant’s surface using the PIII&D process
by Liang et al. [100] and the results describe that the coating was thick and uniform
throughout the implant’s surface. The technique enhanced a material’s resistance to
corrosion, fatigue, and abrasion. The PIII&D technique was shown to be effective for
processing materials of any shape and getting around various material restrictions.
The PIII&D method was also explored by Sun et al. [101] who utilized it to deposit
a titanium and oxygen film on the Ni and titanium alloy surface. The study results
showed that the coating is uniform and dense and that PIII&D had no effect on the
Ti-Ni’s shape memory. This method enormously enhanced the coating’s strength
and resistance to corrosion. It was determined that layers produced utilizing the
PIII&D process exhibit excellent wear properties and adhesion strength, making
them suitable for coating metal surfaces. In order to coat biomaterials over the metal
surface, PIII&D is one of the most popular methods and it is a desirable technique
from an industrial standpoint. It needs a cheap setup, has the ability to alter various
shapes, and enables large-scale mass production of medical implants. Future studies
can concentrate on the deposition of various metal elements which further improve
the characteristics of implants [4].
A thin film layer can be formed over the surface of a metal substrate using the coating
technique known as chemical vapor deposition (CVD). A non-volatile substance is
deposited onto the substrate surface in this procedure as a result of a chemical reaction
that occurs within chemicals in the gaseous phase and the sample surface. The coating
produced via this method is of high quality and has adjustable purity and density
[102]. Inorganic compounds like carbon nanotubes, graphene, TiO2 , and other such
materials are synthesized using this method in synthetic inorganic chemistry because
the end product can be precisely controlled in terms of both quantity and quality [103].
The benefits of CVD include avoiding the line of sight, producing thick coating layers
on substrate surfaces, and simultaneously co-depositing several materials. Several
studies have found that CVD is a more advantageous choice than PVD and can be
further improved to increase its resistance to corrosion [4]. Despite this, CVD use is
not as popular as other techniques since it requires a costly and expensive to set up on
a big scale, vapor deposition equipment, high reaction temperature, and hence has a
low deposition rate. The exhaust gas and gas source employed in the procedure could
have toxic consequences that would make the implant more toxic and dangerous for
human health [104].
312 R. Shivgotra et al.
A method for modifying the surface known as Micro Arc Oxidation (MAO) employs
the anodizing process to modify the surface of metals and their alloys in the imme-
diate extreme conditions of pressure and temperature produced by an arc discharge.
Metal oxides and electrolyte components constitute the modified ceramic coating
formed via MAO. The positive outcomes provided by MAO include its ease of use,
minimal space requirements, high processing capacity, and environmental safety.
Additionally, it has advantages for industry, such as cost-effectiveness and suitability
for very large-scale production. High hydrophilicity in the coating produced through
the MAO technique can resemble the process by which an implant interacts with
its surrounding biological environment. The presence of metal ions enhances the
implant’s antibacterial properties as well [105].
According to the outcomes of a study by Cao et al. [106] the MAO
method improved the adhesive strength of coating as the uniformity of PLC coating
increased. Another study by Wang et al. [107] examined the characteristics of an
MAO coating that was applied to magnesium in a two-step, current-decreasing mode.
The results indicate that the two steps current decreasing mode improved the charac-
teristics, contributed to energy conservation by lowering current consumption, and
displayed the best corrosion resistance, maximum adhesion, and highest hardness.
The MAO technique’s main disadvantage is that the MAO electrolyzer has very
high-power consumption and size limitations.
Sol-Gel Technique
It is the common technique to fabricate several oxide films is Sol-Gel. By using this
process, hybrid organic-inorganic materials and inorganic materials can be synthe-
sized at low temperatures. A gel is composed of a continuous solid phase that
encloses a liquid phase, while a sol is a colloidal suspension comprising solid parti-
cles dispersed in a liquid. With this technique, the chemical reaction takes place in the
solution rather than at the sample-to-gel interface [108]. Sol-gel technology offers
several advantages, including the ease of fabrication, exceptional film uniformity,
the ability to cover substrates of any size and over large areas, and low processing
temperatures. To coat implants made of metal with a coating of hydroxyapatite, TiO2 ,
SiO2 , and various other films in order to form a 3D rigid network on the substrate
surface, hydrolysis, and condensation reactions are used in this method. This method
has several industrial uses for the reason that it is more rapid than other methods and
uses a significantly lesser coating material [109]. The sol-gel technique, however,
comes with some drawbacks, including the possibility of large volume shrinkage and
disc cracking; the potential cost of the precursor used, which enhances the overall
process cost; and the sensitivity of the membrane formed to heat treatment, resulting
in film layer cracking. Due to these and other benefits, sol-gel technology has a
Advancement in Biomaterials in the Form of Implants 313
promising future in the medical field. Further applications are restricted by its sensi-
tivity to heat treatment. In the future, researchers may investigate its effect on thermal
effects and further develop the method to improve its effectiveness and uses [4, 110].
Several medical devices and implants have been developed with metal oxide nanopar-
ticles. Iron oxide’s magnetic properties have been used for therapeutic and diag-
nostic reasons as contrast materials to provide magnetic resonance imaging, magnetic
particle imaging, ultrasonic methods photoacoustic imaging, and magnetic particle
hyperthermia. Zinc oxide (ZnO) has a valuable electrical structure for biological
applications; for instance, cancer cells have been visualized using ZnO nanowires’
inherent fluorescence [112]. Numerous biological uses exist for (TiO2 ). For example,
in bone-substituting materials, a thin layer of TiO2 spontaneously forms on the top
surface of metallic titanium, which encouraged the utilization of TiO2 nanoparticles
for the regeneration of bones. Due to its similar compatibility to titanium with hard
tissues, zirconium oxide is currently utilized for dental implants [113].
314 R. Shivgotra et al.
The absorption and scattering in the visible and near-infrared ranges have led to the
usage of materials containing metal nanoparticles in scientific domains of sensing
and diagnostics. To improve luminescence, gold nanoparticles can be added to the
formulation of substrates or deposited on suitable substrates. The particle size and
morphology affect their absorption and scattering capabilities, which in turn deter-
mines how this technology is used. Until the development of technologies based on
nanomaterials, there weren’t many effective detection approaches [114]. However,
modern technologies based on nanotechnology facilitate the diagnosis of osteo-
porosis using a portable device. For instance, research has led to the development of a
novel biochip that uses gold nanoparticles to identify an osteoporosis-related protein.
It has been demonstrated that it can efficiently assess bone quality and precisely detect
and characterize the rate of bone degradation. The assessment of the effectiveness
of cancer treatment may also be aided by the use of fluorescent probes to identify
nanoparticles [113, 114].
The use of carbon nanotubes (CNTs) in various scientific fields has been prompted by
their physical and chemical characteristics. Their application in nanobiotechnology
has expanded as a result of surface modification studies performed on these particles
and their molecular functionalization with biological molecules [115, 116]. Recently,
researchers suggested employing a new type of tubular structure for bone regenera-
tion called collagen-modified calcium carbonate nanotubes. The emphasis on using
nanotechnology to increase the mechanical stability of ultra-high molecular weight
polyethylene—the most often used polymer for packaging orthopedic implants—
has developed primarily because of its wear resistance and higher biocompatibility
[113]. The incorporation of carbon nanotubes into this polymer has demonstrated
translational efficacy and can sometimes be employed as a liner for the acetabulum
or an element of the tibia [82].
In recent years, the use of 3D printable biomaterials has significantly increased in the
production of orthopedic implants. This is primarily due to the lightweight nature of
the materials, minimal waste generated during production, porous structure that facil-
itates tissue growth, and the convenience of making patient-specific implants with
complex topologies [17]. The biodegradable implants, which are 3D-printed and
customized for each patient, are sustainable and dissolve naturally within the body.
They offer superior healing properties compared to metal implants [117]. Challenges
Advancement in Biomaterials in the Form of Implants 315
for modifying the structure of the production system are made possible by the devel-
opment of 3D printing techniques. The current state of the art demonstrates a trend
toward 3D printing in the production of complicated implants such as heart valves,
blood arteries, tracheas, etc. [118]. The utilization of 3D printing for implants offers
several benefits, such as rapid production, cost-effectiveness, precision in creating
intricate porous structures, absorption of human bone cells, and the ability to facilitate
bone integration, while eliminating the risk of surface coating detachment. Currently,
several 3D-printed implants are available in the market, including knee joints, poste-
rior lumbar interbody fusion devices, meniscus tissues, spinal implants, hip joints,
and knee braces, among others. Implants can be 3D printed using different materials,
such as metals, ceramics, and polymers [119].
The three categories of 3D printed biomaterials are similar to those used for tradi-
tional biomaterials: 3D printable metals, 3D printable polymers, and 3D printable
ceramics. When the strength of the implant is an essential factor, such as in dental
implants, orthopedic implants, and the repair of long bone fractures, metallic bioma-
terials that are 3D printable are used [17]. The most common 3D printing polymers
utilized for manufacturing orthoses, implants, drug delivery systems, and ortho-
pedic implants include PLA, PEEK, Collagen, Polyether, and Polyesters. Because
Young’s modulus of these materials is lower than or equivalent to that of bones
and other body parts, implants made of these biomaterials offer superior dura-
bility and are biocompatible in comparison to those made of metal. The cortical
long bones implants are made with 3D printable bioceramics. Orthodontic and
orthopedic implants are developed from 3D printable biomaterials including HAp,
bioglass, glass ceramics, zirconia, etc. Low fatigue life is a major drawback of the
3D-printed implant. The number of alternative stress cycles that a part can with-
stand before failing is measured as fatigue life. Materials can fracture at lower stress
levels, even below their ultimate strength, if they are repeatedly subjected to alternate
stresses over a long period of time. Every metal has been affected by this; hence the
fatigue strength of 3D-printed implants must be examined [17, 119].
adopting these biomaterials since, in the current context, permanent implants and
bone replacement require biocompatibility along with the mechanical and biological
properties of the biomaterial used. Many biomaterials have been identified until now,
and because of their biocompatibility and biodegradability, these materials are widely
used in biotherapy and medical research [3]. Different biomaterials are used for manu-
facturing of implants, including metals, ceramics, and polymers, depending on the
requirement for either permanent or temporary implants. This article examines the use
of several metals and their alloys, polymeric materials, and other materials employed
for medical implants. To ensure biocompatibility and prevent corrosion in the body,
metals are chosen with care. Because of its biocompatibility and surface hardness,
titanium metal has multiple uses in the medical industry. The chapter discusses
various metals and their alloys. Although these alloys were able to perform well
as medical implants, there remains room for improvement in their nanotoxicity and
manufacturing complexity. Polymeric materials are also employed as materials used
for implantation typically as a surface coating material of metal implants to enhance
their properties. The surfaces of implants can be fabricated and modified using a
variety of methods, such as surface coatings and alterations to boost the mechanical
strength of the materials. Even though these methods have been widely employed
and have produced acceptable outcomes, there has yet opportunity for advancement
in addressing their drawbacks. Nanotechnology and 3D printing have further facili-
tated the development of a novel type of material for implants with improved efficacy,
affordability, and a substantial surface area-to-volume ratio. Designing materials that
are biocompatible, biodegradable, and possess appropriate osseointegration proper-
ties, while simultaneously avoiding adverse effects on biological structures, is a
significant challenge. Thus, focused and careful research is necessary in order to
develop the most effective and ideal materials used for the production of implants.
In conclusion, despite these challenges, future prospects of biomaterials used in
implants are promising, and ongoing research and development will continue to
improve their properties and performance. These advancements will lead to better
outcomes for patients and enable the development of more advanced implantable
devices.
References
1. Langer, R., Peppas, N.A.: Advances in biomaterials, drug delivery, and bionanotechnology.
AIChE J. 49(12), 2990–3006 (2003)
2. Fenton, O.S., Olafson, K.N., Pillai, P.S., Mitchell, M.J., Langer, R.: Advances in biomaterials
for drug delivery. Adv. Mater. 30(29), 1705328 (2018)
3. Bharadwaj, A.: An overview on biomaterials and its applications in medical science. In:
IOP Conference Series: materials Science and Engineering, vol. 1116, no. 1, p. 012178. IOP
Publishing
4. Gautam, S., Bhatnagar, D., Bansal, D., Batra, H., Goyal, N.: Recent advancements in
nanomaterials for biomedical implants. Biomed. Eng. Adv. 13, 100029 (2022)
Advancement in Biomaterials in the Form of Implants 317
5. Raghavendra, G.M., Varaprasad, K., Jayaramudu, T.: Biomaterials: design, development and
biomedical applications. In: Nanotechnology Applications for Tissue Engineering, pp. 21–44.
William Andrew Publishing (2015)
6. Amirtharaj Mosas, K.K., Chandrasekar, A.R., Dasan, A., Pakseresht, A., Galusek, D.: Recent
advancements in materials and coatings for biomedical implants. Gels 8(5), 323 (2022)
7. Wang, X.: Overview on biocompatibilities of implantable biomaterials. Lazinica, R. (ed.)
Adv. Biomater. Sci. Biomed. Appl. Biomed. 111–155 (2013)
8. Teo, A.J., Mishra, A., Park, I., Kim, Y.J., Park, W.T., Yoon, Y.J.: Polymeric biomaterials for
medical implants and devices. ACS Biomater. Sci. Eng. 2(4), 454–472 (2016)
9. Saini, M., Singh, Y., Arora, P., Arora, V., Jain, K.: Implant biomaterials: a comprehensive
review. World J. Clin. Cases: WJCC 3(1), 52 (2015)
10. Arsiwala, A., Desai, P., Patravale, V.: Recent advances in micro/nanoscale biomedical
implants. J. Control. Release 10(189), 25–45 (2014)
11. Franz, S., Rammelt, S., Scharnweber, D., Simon, J.C.: Immune responses to implants—A
review of the implications for the design of immunomodulatory biomaterials. Biomaterials
32(28), 6692–6709 (2011)
12. Misch, C.E.: Prosthetic options in implant dentistry. In: Contemporary Implant Dentistry, vol.
3, pp. 105–126 (1999)
13. Deepashree, R., Devaki, V., Balu Kandhasamy, R.A.: Evolution of implant biomaterials: a
literature review. J. Indian Acad. Dental Spec. Res. 4(2) (2017)
14. Amarnath, G.S., Muddugangadhar, B.C., Tripathi, S., Dikshit, S., MS, D.: Biomaterials for
dental implants: an overview. Int. J. Oral Implantol. Clin. Res. 2(1), 13–24 (2011)
15. Yamamuro, T.: Bioceramics. In: Poitout, D.G. (ed.) Biomechanics and Biomaterials in
Orthopedics, pp. 22–33. Springer, London (2004)
16. Elbadawi, M., Meredith, J., Hopkins, L., Reaney, I.: Progress in bioactive metal and ceramic
implants for load-bearing application. Adv. Tech. Bone Regen. [Internet] 31, 195–219 (2016)
17. Yadav, D., Garg, R.K., Ahlawat, A., Chhabra, D.: 3D printable biomaterials for orthopedic
implants: solution for sustainable and circular economy. Resour. Policy 1(68), 101767 (2020)
18. Raucci, M.G., Giugliano, D., Ambrosio, L.: Fundamental properties of bioceramics and
biocomposites. In: Handbook of Bioceramics and Biocomposites, pp. 35–58. Springer, Cham
(2016)
19. Thamaraiselvi, T., Rajeswari, S.: Biological evaluation of bioceramic materials-a review.
Carbon 24(31), 172 (2004)
20. Chevalier, J., Liens, A., Reveron, H., Zhang, F., Reynaud, P., Douillard, T., Preiss, L., Sergo,
V., Lughi, V., Swain, M., Courtois, N.: Forty years after the promise of “ceramic steel?”:
zirconia-based composites with a metal-like mechanical behavior. J. Am. Ceram. Soc. 103(3),
1482–1513 (2020)
21. Larose, J., Timms, D.: Hemocompatibility in mechanical circulatory support. In: Mechanical
Circulatory Support: a Companion to Braunwald’s Heart Disease, pp. 83–89. Elsevier (2020)
22. Dubok, V.A.: Bioceramics-yesterday, today, tomorrow. Powder Metall. Met. Ceram. 39, 381–
394 (2000)
23. Gupta, R., Kumar, A.: Bioactive materials for biomedical applications using sol-gel tech-
nology. Biomed. Mater. 3(3), 034005 (2008)
24. Baino, F., Verné, E.: Glass-based coatings on biomedical implants: a state-of-the-art review.
Biomed. Glasses 3(1), 1–7 (2017)
25. Salinas, A.J., Esbrit, P., Vallet-Regí, M.: A tissue engineering approach based on the use of
bioceramics for bone repair. Biomater. Sci. 1(1), 40–51 (2013)
26. Gotman, I.: Characteristics of metals used in implants. J. Endourol. 11(6), 383–389 (1997)
27. Sykaras, N., Iacopino, A.M., Marker, V.A., Triplett, R.G., Woody, R.D.: Implant materials,
designs, and surface topographies: their effect on osseointegration. A literature review. Int. J.
Oral Maxillofac. Implants 15(5) (2000)
28. Niinomi, M.: Recent metallic materials for biomedical applications. Metall. Mater. Trans. A
33, 477–486 (2002)
318 R. Shivgotra et al.
29. Cadosch, D., Chan, E., Gautschi, O.P., Filgueira, L.: Metal is not inert: role of metal ions
released by biocorrosion in aseptic loosening—Current concepts. J. Biomed. Mater. Res. Part
A: Off. J. Soc. Biomater. Jpn. Soc. Biomater. Austral. Soc. Biomater. Korean Soc. Biomater.
91(4), 1252–1262 (2009)
30. Hermawan, H., Ramdan, D., Djuansjah, J.R.: Metals for biomedical applications. Biomed.
Eng. Theory Appl. 29(1), 411–430 (2011)
31. Hanawa, T.: Research and development of metals for medical devices based on clinical needs.
Sci. Technol. Adv. Mater. (2012)
32. Bommala, V.K., Krishna, M.G., Rao, C.T.: Magnesium matrix composites for biomedical
applications: a review. J. Magn. Alloys 7(1), 72–79 (2019)
33. Harrison, R., Maradze, D., Lyons, S., Zheng, Y., Liu, Y.: Corrosion of magnesium and magne-
sium–calcium alloy in biologically-simulated environment. Progr. Nat. Sci.: Mater. Int. 24(5),
539–546 (2014)
34. Kraus, T., Fischerauer, S., Treichler, S., Martinelli, E., Eichler, J., Myrissa, A., Zötsch, S.,
Uggowitzer, P.J., Löffler, J.F., Weinberg, A.M.: The influence of biodegradable magnesium
implants on the growth plate. Acta Biomater. 15(66), 109–117 (2018)
35. Naujokat, H., Seitz, J.M., Açil, Y., Damm, T., Möller, I., Gülses, A., Wiltfang, J.: Osteosyn-
thesis of a cranio-osteoplasty with a biodegradable magnesium plate system in miniature pigs.
Acta Biomater. 15(62), 434–445 (2017)
36. Chakraborty Banerjee, P., Al-Saadi, S., Choudhary, L., Harandi, S.E., Singh, R.: Magnesium
implants: prospects and challenges. Materials 12(1), 136 (2019)
37. Jamel, M.M., Jamel, M.M., Lopez, H.F.: Designing advanced biomedical biodegradable Mg
alloys: a review. Metals 12(1), 85 (2022)
38. Sarraf, M., Rezvani Ghomi, E., Alipour, S., Ramakrishna, S., Liana, S.N.: A state-of-the-
art review of the fabrication and characteristics of titanium and its alloys for biomedical
applications. Bio-Des. Manuf. 26, 1–25 (2021)
39. Khadija, G., Saleem, A., Akhtar, Z., Naqvi, Z., Gull, M., Masood, M., Mukhtar, S., Batool,
M., Saleem, N., Rasheed, T., Nizam, N.: Short term exposure to titanium, aluminum and
vanadium (Ti 6Al 4V) alloy powder drastically affects behavior and antioxidant metabolites
in vital organs of male albino mice. Toxicol. Rep. 1(5), 765–770 (2018)
40. Fellah, M., Labaïz, M., Assala, O., Dekhil, L., Taleb, A., Rezag, H., Iost, A.: Tribological
behavior of Ti–6Al–4V and Ti–6Al–7Nb alloys for total hip prosthesis. Adv. Tribol. 21, 2014
(2014)
41. Olmedo, D.G., Duffó, G., Cabrini, R.L., Guglielmotti, M.B.: Local effect of titanium implant
corrosion: an experimental study in rats. Int. J. Oral Maxillofac. Surg. 37(11), 1032–1038
(2008)
42. Kirmanidou, Y., Sidira, M., Drosou, M.E., Bennani, V., Bakopoulou, A., Tsouknidas, A.,
Michailidis, N., Michalakis, K.: New Ti-alloys and surface modifications to improve the
mechanical properties and the biological response to orthopedic and dental implants: a review.
Biomed. Res. Int. 14, 2016 (2016)
43. Reclaru, L., Lerf, R., Eschler, P.Y., Meyer, J.M.: Corrosion behavior of a welded stainless-steel
orthopedic implant. Biomaterials 22(3), 269–279 (2001)
44. Bekmurzayeva, A., Duncanson, W.J., Azevedo, H.S., Kanayeva, D.: Surface modification of
stainless steel for biomedical applications: revisiting a century-old material. Mater. Sci. Eng.
C 1(93), 1073–1089 (2018)
45. Godbole, N., Yadav, S., Ramachandran, M., Belemkar, S.: A review on surface treatment of
stainless steel orthopedic implants. Int. J. Pharm. Sci. Rev. Res. 36(1), 190–194 (2016)
46. Ma, L., Wiame, F., Maurice, V., Marcus, P.: Origin of nanoscale heterogeneity in the surface
oxide film protecting stainless steel against corrosion. NPJ Mater. Degrad. 3(1), 29 (2019)
47. Mahmoudi Hashemi, P., Borhani, E., Nourbakhsh, M.S.: A review on nanostructured stainless
steel implants for biomedical application. Nanomed. J. 3(4), 202–216 (2016)
48. Salahinejad, E., Hadianfard, M.J., Macdonald, D.D., Sharifi-Asl, S., Mozafari, M., Walker,
K.J., Rad, A.T., Madihally, S.V., Tayebi, L.: In vitro electrochemical corrosion and cell
viability studies on nickel-free stainless steel orthopedic implants. Plos One 8(4), e61633
(2013)
Advancement in Biomaterials in the Form of Implants 319
49. Natiella, J.R., Armitage, J.E., Greene, Jr., G.W., Meenaghan, M.A.: Council on dental
materials and devices. Current evaluation of dental implants. J. Am. Dental Assoc. 84(6),
1358–1372 (1972)
50. Marti, A.: Cobalt-base alloys used in bone surgery. Injury 1(31), D18-21 (2000)
51. Garcia-Mendez, M.C., Urrutia-Baca, V.H., Cuao-Moreu, C.A., Lorenzo-Bonet, E., Alvarez-
Vera, M., Ortiz-Martinez, D.M., de la Garza-Ramos, M.A.: In vitro biocompatibility eval-
uation of a new Co–Cr–B alloy with potential biomedical application. Metals 11(8), 1267
(2021)
52. González-Mora, V.A., Hoffmann, M., Stroosnijder, R., Espinar Escalona, E., Llamas Carrera,
J.M., Fernández-Fairén, M., Gil Mur, F.J.: Influence of different CoCrMo counterfaces on
wear in UHMWPE for artificial joints. J. Biomed. Sci. Eng. (2011)
53. Watanabe, K., Fukuzaki, S., Sugino, A., Benson, N., Metcalf, N., Nakamura, M., Matsumoto,
M.: Cobalt–chromium alloy has superior antibacterial effect than titanium alloy: in vitro and
in vivo studies. Spine 46(17), E911 (2021)
54. Jaishankar, M., Tseten, T., Anbalagan, N., Mathew, B.B., Beeregowda, K.N.: Toxicity,
mechanism and health effects of some heavy metals. Interdiscip. Toxicol. 7(2), 60 (2014)
55. Guo, Z., Pang, X., Yan, Y., Gao, K., Volinsky, A.A., Zhang, T.Y.: CoCrMo alloy for ortho-
pedic implant application enhanced corrosion and tribocorrosion properties by nitrogen ion
implantation. Appl. Surf. Sci. 30(347), 23–34 (2015)
56. Lourenço, M.L., Cardoso, G.C., Sousa, K.D., Donato, T.A., Pontes, F.M., Grandini, C.R.:
Development of novel Ti–Mo–Mn alloys for biomedical applications. Sci. Rep. 10(1), 6298
(2020)
57. Maddock, N.A., James, N.L., McKenzie, D.R., Patrick, J.F.: Technological advances for
polymers in active implantable medical devices. Des. Manuf. Med. Dev. 1, 239–272 (2012)
58. Kurtz, S.M., Devine, J.N.: PEEK biomaterials in trauma, orthopedic, and spinal implants.
Biomaterials 28(32), 4845–4869 (2007)
59. Costa, M.M., Dantas, T.A., Bartolomeu, F., Alves, N., Silva, F.S., Miranda, G., Toptan, F.:
Corrosion behaviour of PEEK or β-TCP-impregnated Ti6Al4V SLM structures targeting
biomedical applications. Trans. Nonferrous Metals Soc. China 29(12), 2523–2533 (2019)
60. Panayotov, I.V., Orti, V., Cuisinier, F., Yachouh, J.: Polyetheretherketone (PEEK) for medical
applications. J. Mater. Sci. Mater. Med. 27, 1–1 (2016)
61. Li, C., Mason, J., Yakimicki, D.: Thermal characterization of PMMA-based bone cement
curing. J. Mater. Sci. Mater. Med. 15(1), 85–89 (2004)
62. Hacker, M.C., Krieghoff, J., Mikos, A.G.: Synthetic polymers. In: Principles of Regenerative
Medicine, pp. 559–590. Academic Press (2019)
63. Mazumder, S., Nayak, A.K., Ara, T.J., Hasnain, M.S.: Hydroxyapatite composites for
dentistry. Appl. Nanocompos. Mater. Dent. 1, 123–143 (2019)
64. Kannan, M.B.: Hydroxyapatite coating on biodegradable magnesium and magnesium-based
alloys. In: Hydroxyapatite (HAp) For Biomedical Applications, pp. 289–306. Woodhead
Publishing (2015)
65. Klinge, U., Klosterhalfen, B., Öttinger, A.P., Junge, K., Schumpelick, V.: PVDF as a new
polymer for the construction of surgical meshes. Biomaterials 23(16), 3487–3493 (2002)
66. Zhou, J., Huang, X., Zheng, D., Li, H., Herrler, T., Li, Q.: Oriental nose elongation using an
L-shaped polyethylene sheet implant for combined septal spreading and extension. Aesthetic
Plast. Surg. 38, 295–302 (2014)
67. Cools, P., Van Vrekhem, S., De Geyter, N., Morent, R.: The use of DBD plasma treatment
and polymerization for the enhancement of biomedical UHMWPE. Thin Solid Films 1(572),
251–259 (2014)
68. Rahimi, A., Mashak, A.: Review on rubbers in medicine: Natural, silicone and polyurethane
rubbers. Plast. Rubber Compos. 42(6), 223–230 (2013)
-
69. Pergal, M.V., Nestorov, J., Tovilović, G., Ostojić, S., Godevac, D., Vasiljević-Radović,
D., Djonlagić, J.: Structure and properties of thermoplastic polyurethanes based on poly
(dimethylsiloxane): assessment of biocompatibility. J. Biomed. Mater. Res. Part A 102(11),
3951–3964 (2014)
320 R. Shivgotra et al.
70. Zheng, F., Xu, L., Verbiest, L., Verbeken, E., De Ridder, D., Deprest, J.: Cytokine production
following experimental implantation of xenogenic dermal collagen and polypropylene grafts
in mice. Neurourol. Urodyn.: Off. J. Int. Cont. Soc. 26(2), 280–289 (2007)
71. Moalli, P., Brown, B., Reitman, M.T., Nager, C.W.: Polypropylene mesh: evidence for lack
of carcinogenicity. Int. Urogynecol. J. 25, 573–576 (2014)
72. Bergmann, P.A., Becker, B., Mauss, K.L., Liodaki, M.E., Knobloch, J., Mailänder, P.,
Siemers, F.: Titanium-coated polypropylene mesh (TiLoop Bra®)—An effective prevention
for capsular contracture? Eur. J. Plast. Surg. 37, 339–346 (2014)
73. Nanni, F., Lamastra, F.R., Pisa, F., Gusmano, G.: Synthesis and characterization of poly (ε-
caprolactone) reinforced with aligned hybrid electrospun PMMA/nano-Al2O3 fibre mats by
film stacking. J. Mater. Sci. 46, 6124–6130 (2011)
74. McMahon, R.E., Wang, L., Skoracki, R., Mathur, A.B.: Development of nanomaterials for
bone repair and regeneration. J. Biomed. Mater. Res. B Appl. Biomater. 101(2), 387–397
(2013)
75. Van Ardenne, N., Vanderwegen, J., Van Nuffelen, G., De Bodt, M., Van de Heyning,
P.: Medialization thyroplasty: vocal outcome of silicone and titanium implant. Eur. Arch.
Otorhinolaryngol. 268, 101–107 (2011)
76. Kirsten, S., Uhlemann, J., Braunschweig, M., Wolter, K.J.: Packaging of electronic devices
for long-term implantation. In: 2012 35th International Spring Seminar on Electronics
Technology, pp. 123–127. IEEE (2012)
77. Qin, Y., Howlader, M.M., Deen, M.J., Haddara, Y.M., Selvaganapathy, P.R.: Polymer inte-
gration for packaging of implantable sensors. Sens. Actuat. B Chem. 31(202), 758–778
(2014)
78. Sundaram, V., Sukumaran, V., Cato, M.E., Liu, F., Tummala, R., Weiland, J.D., Nasiatka,
P.J., Tanguay, A.R.: High density electrical interconnections in liquid crystal polymer (LCP)
substrates for retinal and neural prosthesis applications. In: 2011 IEEE 61st Electronic
Components and Technology Conference (ECTC), pp. 1308–1313. IEEE (2011)
79. Kim, J.H., Min, K.S., An, S.K., Jeong, J.S., Jun, S.B., Cho, M.H., Son, Y.D., Cho, Z.H., Kim,
S.J.: Magnetic resonance imaging compatibility of the polymer-based cochlear implant. Clin.
Exp. Otorhinolaryngol. 5(Suppl 1), S19–S23
80. Maurus, P.B., Kaeding, C.C.: Bioabsorbable implant material review. Oper. Tech. Sports Med.
12(3), 158–160 (2004)
81. Suh, J.K., Matthew, H.W.: Application of chitosan-based polysaccharide biomaterials in
cartilage tissue engineering: a review. Biomaterials 21(24), 2589–2598 (2000)
82. Chen, M.Q.: Recent advances and perspective of nanotechnology-based implants for
orthopedic applications. Front. Bioeng. Biotechnol. 10 (2022)
83. Ratner, B.D., Bryant, S.J.: Biomaterials: where we have been and where we are going. Annu.
Rev. Biomed. Eng. 15(6), 41–75 (2004)
84. Lam, M., Migonney, V., Falentin-Daudre, C.: Review of silicone surface modification tech-
niques and coatings for antibacterial/antimicrobial applications to improve breast implant
surfaces. Acta Biomater. 1(121), 68–88 (2021)
85. Accioni, F., Vázquez, J., Merinero, M., Begines, B., Alcudia, A.: Latest trends in surface
modification for dental implantology: innovative developments and analytical applications.
Pharmaceutics 14(2), 455 (2022)
86. Sikder, P., Ren, Y., Bhaduri, S.B.: Synthesis and evaluation of protective poly (lactic acid)
and fluorine-doped hydroxyapatite—Based composite coatings on AZ31 magnesium alloy. J.
Mater. Res. 34(22), 3766–3776 (2019)
87. Metroke, T.L., Parkhill, R.L., Knobbe, E.T.: Passivation of metal alloys using sol-gel-derived
materials—A review. Prog. Org. Coat. 41(4), 233–238 (2001)
88. Campbell, A.A.: Bioceramics for implant coatings. Mater. Today 6(11), 26–30 (2003)
89. Helmersson, U., Lattemann, M., Bohlmark, J., Ehiasarian, A.P., Gudmundsson, J.T.: Ionized
physical vapor deposition (IPVD): a review of technology and applications. Thin Solid Films
513(1–2), 1–24 (2006)
Advancement in Biomaterials in the Form of Implants 321
90. Liu, M.J., Zhang, M., Zhang, X.F., Li, G.R., Zhang, Q., Li, C.X., Li, C.J., Yang, G.J.: Transport
and deposition behaviors of vapor coating materials in plasma spray-physical vapor deposition.
Appl. Surf. Sci. 30(486), 80–92 (2019)
91. Ozkucur, N., Wetzel, C., Hollstein, F., Richter, E., Funk, R.H., Monsees, T.K.: Physical vapor
deposition of zirconium or titanium thin films on flexible polyurethane highly support adhe-
sion and physiology of human endothelial cells. J. Biomed. Mater. Res. Part A: Off. J. Soc.
Biomater. Jpn. Soc. Biomater. Austral. Soc. Biomater. Korean Soc. Biomater. 89(1), 57–67
(2009)
92. Esmaeili, M.M., Mahmoodi, M., Imani, R.: Tantalum carbide coating on Ti–6Al–4V by
electron beam physical vapor deposition method: study of corrosion and biocompatibility
behavior. Int. J. Appl. Ceram. Technol. 14(3), 374–382 (2017)
93. Qadir, M., Li, Y., Wen, C.: Ion-substituted calcium phosphate coatings by physical vapor
deposition magnetron sputtering for biomedical applications: a review. Acta Biomater. 15(89),
14–32 (2019)
94. Taran, A.V., Garkusha, I.E., Taran, V.S., Pyvovar, N.V., Muratov, R.M., Leonovych, A.V.,
Nikolaychuk, G.P., Baturin, A.A.: Structure of biocompatible nanocoatings obtained by phys-
ical vapor deposition on flexible polyurethane for medical applications. J. Adv. Microsc. Res.
13(3), 313–319 (2018)
95. Vardelle, A., Moreau, C., Themelis, N.J., Chazelas, C.: A perspective on plasma spray
technology. Plasma Chem. Plasma Process. 35, 491–509 (2015)
96. Singh, H., Sidhu, B.S., Puri, D., Prakash, S.: Use of plasma spray technology for deposition
of high temperature oxidation/corrosion resistant coatings—A review. Mater. Corros. 58(2),
92–102 (2007)
97. Xue, T., Attarilar, S., Liu, S., Liu, J., Song, X., Li, L., Zhao, B., Tang, Y.: Surface modification
techniques of titanium and its alloys to functionally optimize their biomedical properties:
thematic review. Front. Bioeng. Biotechnol. 11(8), 603072 (2020)
98. Anders, A.: Metal plasma immersion ion implantation and deposition: a review. Surf. Coat.
Technol. 93(2–3), 158–167 (1997)
99. Mändl, S., Manova, D.: Modification of metals by plasma immersion ion implantation. Surf.
Coat. Technol. 15(365), 83–93 (2019)
100. Liang, Y., Xu, J., Chen, J., Qi, M., Xie, X., Hu, M.: Zinc ion implantation-deposition technique
improves the osteoblast biocompatibility of titanium surfaces. Mol. Med. Rep. 11(6), 4225–
4231 (2015)
101. Sun, T., Wang, L.P., Wang, M.: (Ti, O)/Ti and (Ti, O, N)/Ti composite coatings fabricated
via PIIID for the medical application of NiTi shape memory alloy. J. Biomed. Mater. Res. B
Appl. Biomater. 96(2), 249–260 (2011)
102. Carlsson, J.O., Martin, P.M.: Chemical vapor deposition. In: Handbook of Deposition
Technologies for Films and Coatings, pp. 314–363. William Andrew Publishing (2010)
103. Brunette, D.M., Tengvall, P., Textor, M., Thomsen, P., Thull, R., Grant, D.: Physical and
chemical vapor deposition and plasma-assisted techniques for coating titanium. In: Titanium in
Medicine: material Science, Surface Science, Engineering, Biological Responses and Medical
Applications, pp. 283–341 (2001)
104. Stewart, C., Akhavan, B., Wise, S.G., Bilek, M.M.: A review of biomimetic surface func-
tionalization for bone-integrating orthopedic implants: mechanisms, current approaches, and
future directions. Prog. Mater Sci. 1(106), 100588 (2019)
105. Li, L.H., Kong, Y.M., Kim, H.W., Kim, Y.W., Kim, H.E., Heo, S.J., Koak, J.Y.: Improved
biological performance of Ti implants due to surface modification by micro-arc oxidation.
Biomaterials 25(14), 2867–2875 (2004)
106. Cao, X.Y., Tian, N., Dong, X., Cheng, C.K.: Implant coating manufactured by micro-arc oxida-
tion and dip coating in resorbable polylactide for antimicrobial applications in orthopedics.
Coatings 9(5), 284 (2019)
107. Wang, Z.X., Zhang, J.W., Ye, F., Lv, W.G., Lu, S., Sun, L., Jiang, X.Z.: Properties of micro-arc
oxidation coating fabricated on magnesium under two steps current-decreasing mode. Front.
Mater. 18(7), 261 (2020)
322 R. Shivgotra et al.
108. Feng, W., Mu-Sen, L., Yu-Peng, L., Yong-Xin, Q.: A simple sol-gel technique for preparing
hydroxyapatite nanopowders. Mater. Lett. 59(8–9), 916–919 (2005)
109. Castro, Y., Durán, A.: Control of degradation rate of Mg alloys using silica sol-gel coatings
for biodegradable implant materials. J. Sol-Gel. Sci. Technol. 15(90), 198–208 (2019)
110. Bollino, F., Catauro, M.: Sol-gel technology to prepare advanced coatings. Photoenergy Thin
Film Mater. 25, 321–378 (2019)
111. Zhang, L., Webster, T.J.: Nanotechnology and nanomaterials: promises for improved tissue
regeneration. Nano Today 4(1), 66–80 (2009)
112. Liu, T.Y., Liao, H.C., Lin, C.C., Hu, S.H., Chen, S.Y.: Biofunctional ZnO nanorod arrays
grown on flexible substrates. Langmuir 22(13), 5804–5809 (2006)
113. Ramos, A.P., Cruz, M.A., Tovani, C.B., Ciancaglini, P.: Biomedical applications of nanotech-
nology. Biophys. Rev. 9(2), 79–89 (2017)
114. Bhattacharya, R., Mukherjee, P.: Biological properties of “naked” metal nanoparticles. Adv.
Drug Deliv. Rev. 60(11), 1289–1306 (2008)
115. Sharma, P., Kumar Mehra, N., Jain, K., Jain, N.K.: Biomedical applications of carbon
nanotubes: a critical review. Curr. Drug Deliv. 13(6), 796–817 (2016)
116. Yang, W., Thordarson, P., Gooding, J.J., Ringer, S.P., Braet, F.: Carbon nanotubes for
biological and biomedical applications. Nanotechnology 18(41), 412001 (2007)
117. Chia, H.N., Wu, B.M.: Recent advances in 3D printing of biomaterials. J. Biol. Eng. 9(1), 1–4
(2015)
118. Javaid, M., Haleem, A.: Current status and challenges of additive manufacturing in
orthopaedics: an overview. J. Clin. Orthop. Trauma 10(2), 380–386 (2019)
119. Chunhua, S., Guangqing, S.: Application and development of 3D printing in medical field.
Modern Mech. Eng. 10(03), 25 (2020)
Smart Biomaterials in Drug Delivery
Applications
1 Introduction
The conventional approaches to treating deadly diseases like cancer, coronary artery
disease, and respiratory and abdominal infections face challenges such as invasive
procedures for solid tumours, poor solubility of drugs, short drug circulation, drug
resistance, non-specific targeting, and harmful side effects, both locally and system-
ically [1]. Millions of fresh incidents and fatalities are recorded worldwide due to
cancer, among the primary causes of death [2, 3]. The incidence and mortality rates
for 36 types of cancer in 185 countries are estimated by GLOBOCAN [3]. Hence,
there is an urgent need for new and innovative treatment methods that can target
S. G. Reddy (B)
Department of Chemistry, Amrita School of Engineering, Amrita Vishwa Vidyapeetham,
Bengaluru 560035, India
e-mail: s_giri@blr.amrita.edu
H. C. A. Murthy
Department of Applied Chemistry, School of Natural Science, Adama Science and Technology
University, Adama 1888, Ethiopia
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 323
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_11
324 S. G. Reddy and H. C. A. Murthy
cancer cells precisely without harming healthy tissues and organs surrounding the
tumour [4].
Recently drug delivery through nanotechnology has gained popularity due to the
unique properties of nanoscale materials. These properties, including enhanced intra-
cellular drug delivery and targeted subcellular structures, make it a promising field.
Nanoscale materials can also overcome physiological barriers in living organisms,
allowing them to reach previously inaccessible body regions [5].
Another emerging field is Supramolecular hydrogels which are versatile and
rapidly developing nanostructures with potential applications in diverse fields,
including nanofabrication, biosensing, catalysis, tissue engineering, and controlled
drug delivery [6]. These hydrogels are formed by low-molecular-weight supramolec-
ular hydrogelators that respond to a stimulus by forming a network of intertwined
fibres, resulting in a solid-like material with a 3D structure and high-water content
[7, 8].
Polymer micelles have emerged as a new area for drug delivery that can enhance
treatment efficacy while minimising negative effects. These functional biomaterials
rely on the self-assembly of amphiphilic polymers to create polymeric micelles,
which can enclose hydrophobic drugs in their core via hydrophobic interactions
[4, 8–12].
The release of medication can be managed by stimuli in the tumour microenvi-
ronment, such as enzymes, temperature, and pH [13, 14].
Another form of drug delivery system is transdermal drug delivery can bypass
first-pass metabolism and achieve continuous medication release, but the protective
skin layer (stratum corneum) poses a challenge [15]. Microneedles offer a user-
friendly solution for effective transdermal drug delivery with high bioavailability
[16]. These patches penetrate the stratum corneum and can deliver drugs painlessly
and minimally invasively for skin vaccination and drug delivery [17–19].
Overall, controlled release is a critical aspect of drug delivery, where drugs
are released at a controlled rate over time, ensuring maximum therapeutic efficacy
while minimising unwanted side effects. Stimuli-responsive materials have gained
significant attention recently as a promising tool for developing controlled-release
drug delivery systems [20]. Such substances can react to environmental variations,
including temperature changes, pH, magnetic or electric fields, and light, allowing
them to dispense medication in a controlled manner [21]. The development of stimuli-
responsive materials has revolutionised the field of drug delivery, as these materials
can overcome many limitations of conventional drug delivery systems [22].
This chapter will briefly overview the recent advances in stimuli-responsive
materials, their properties, and their potential applications in drug delivery [23].
Overall, stimuli-responsive materials have great potential to improve the effi-
cacy and safety of drug delivery, and their continued development is expected to
impact the field of medicine significantly. The various investigators, with their respec-
tive research areas on CDD, are influenced by different parameters such as pH,
temperature, light, and magnetic or electric fields as shown in Fig. 1.
Smart Biomaterials in Drug Delivery Applications 325
Fig. 1 Schematic
representation of controlled
release of drug delivery for
stimuli-responsive for pH,
temperature, light sensitivity
(NIR) and magnetic
pH is a widely studied environmental stimulus that can trigger drug release from
responsive materials. These materials can swell or collapse in response to pH changes,
leading to drug release. pH-responsive materials are particularly useful for targeted
drug delivery [24], as different tissues and organs in the body can have varying pH
levels. For instance, the stomach has a highly acidic pH (1–3), while the blood has
a slightly alkaline pH (7.35–7.45).
Hydrogels and liposomes are two types of extensively studied pH-responsive
materials. Hydrogels are 3D polymer networks that can swell or collapse in response
to pH changes, allowing drug encapsulation and release. Liposomes are spherical
structures made of a phospholipid bilayer that can encapsulate drugs and respond to
pH changes for drug release [25].
Some latest articles published and their outcomes for pH-responsive stimuli are
highlighted in Fig. 2.
A new drug delivery system was developed and evaluated by Pilch et al. [26] for
targeted cancer therapy. The system uses pH-responsive nanoparticles as carriers for
an unsymmetrical bisacridine drug designed to target cancer cells selectively. It is
demonstrated that the drug-loaded nanoparticles have a high drug-loading capacity.
326 S. G. Reddy and H. C. A. Murthy
Fig. 2 Application of
pH-responsive drug delivery
in fields
system had high drug loading content and encapsulation efficiency and did not nega-
tively affect healthy cells but reduced the survival rate of cancer cells. The nanocarrier
system showed excellent blood compatibility, making it a promising candidate for
anticancer nanocarrier systems.
Gou et al. [36] discuss a drug delivery system that utilises chiral mesoporous silica
nanoparticles (CMSN) that have been modified with chitosan (CS) to deliver drugs
to tumours in a targeted manner. The CS-CMSN system demonstrated the ability
to effectively contain and release doxorubicin (DOX) in response to changes in pH
levels. Both in vitro and in vivo experiments showed that the DOX@CS-CMSN
system had better uptake in tumour cells and was more efficient at killing them
while remaining safe for healthy cells. These findings suggest that the CS-CMSN
nanocarrier system is a promising approach for targeted therapy in treating tumours.
The drug delivery system that responds to changes in pH levels for photodynamic
therapy can treat tumours through chemotherapy reported by Zhang et al. [37]. The
system is created using water-soluble conjugated polymers attached to the anticancer
drug doxorubicin through acid-labile imine and acyl hydrazone bonds. The resulting
PFE-DOX-2 system can release drugs in response to pH changes and produce reactive
oxygen species when exposed to light, thus enhancing its therapeutic effectiveness.
The system also permits the monitoring of drug release by observing changes in
the fluorescence of the conjugated polymers. This research presents a promising
approach to treating cancer in vivo using a multifunctional drug delivery system that
responds to stimuli.
The research explored by Rao et al. [38] how palmyra palm kernel (PPK) behaves
as a gel and its possible usage as a carrier for delivering drugs to treat colon cancer.
The PPK hydrogels displayed impressive swelling properties when exposed to pH,
and they effectively encapsulated 5-Fluorouracil (5-FU) with an efficiency rate of up
to 62%. Molecularly dispersed 5-FU was found in the PPK hydrogel matrix, and in
laboratory tests, the drug was gradually released over 12 h. The study indicates that
PPK hydrogels carrying chemotherapeutic drugs can be a treatment alternative for
colon cancer.
Efficient chemotherapy was reported by Heragh et al. [39] for synthesising
cross-linked chitosan/laponite RD nanoparticles that co-encapsulate doxorubicin and
saffron extract. The nanocarriers had a high level of encapsulation for both DOX and
SAF and displayed a release behaviour sensitive to pH. The release of DOX and SAF
was regulated by the cross-linked CS content, with the release rate increasing in acidic
conditions. Additionally, the nanocarriers demonstrated antibacterial properties and
high toxicity against cancer cells. The research indicates that using nanotechnology
to combine natural antioxidants with synthetic anticancer agents can enhance the
effectiveness of chemotherapy while minimising side effects.
The drug delivery by nanoassemblies that release doxorubicin in acidic conditions
is typically found in cancerous tissues while exhibiting a lower release rate in normal
physiological conditions, as reported by Teixeira et al. [40] discuss the creation of.
The nanoassemblies’ hybrid composition allowed for high encapsulation efficiency
and drug-loading content, and they maintained their colloidal stability for at least four
weeks. The DOX-loaded nanoassemblies were shown to have higher cytotoxicity in
Smart Biomaterials in Drug Delivery Applications 329
cancer cell lines compared to free DOX and lower cytotoxicity in normal cell lines.
The results suggest that the nanoassemblies can be used in cancer therapy.
Li [41] describes the fabrication of pH-responsive nanoparticles using Polyamine-
modified β-Cyclodextrin and Sodium dodecyl benzene sulfonate loaded with
Doxorubicin and Celastrol. The nanoparticles were characterised using various tech-
niques and demonstrated minimal release in acidic environments and effective release
in basic environments. The release kinetics were studied using different models, and
the nanoparticles were found to be highly toxic to tumour cells and low in cytotoxicity
to normal epithelial cells. Flow cytometry analysis indicated that the nanoparticles
could induce cell apoptosis and had great potential as an oral administration drug
delivery system with sustained release and target specificity.
The process of creating hydrogels Batool et al. [42] that can sense pH, biodegrade
and are compatible with living organisms. The hydrogels are made using Na-CMC/
pectin Poly(methacrylic acid) and are loaded with the anti-cancer drug Cytarabine.
Their properties, such as texture, morphology, loading efficiency, and drug release
profile, were studied, and they were found to have effective drug encapsulation and
continuous release for 24 h. In toxicity studies with rabbits, the hydrogels were safe
and compatible with living organisms. In pharmacokinetic evaluation, they showed
a significant increase in plasma half-life and AUC. The study’s results suggest these
hydrogels could be used as a controlled drug delivery system.
The hydrogels that can hold two drugs discussed by Du et al. [43] have pH-responsive
and antibacterial characteristics, which are suitable for dressing skin wounds. The
hydrogels were loaded with two drugs (Gentamicin sulfate, and Lysozyme), an
antibacterial agent and a growth factor. They were designed to be pH-responsive,
releasing the drugs more rapidly in acidic environments like those found in infected
wounds. The hydrogels were found to have good antibacterial activity and could
promote wound healing in vitro. It was suggested that these hydrogels could be used
in developing advanced wound dressings with improved efficacy.
Another hydrogel reported by Xie et al. [44] describes the development of a
dressing loaded with microcarriers that release antibacterial and pro-vascularizing
agents for wound healing. The dressing is pH-responsive and promotes the controlled
release of vascular endothelial growth factor (VEGF) to aid in angiogenesis. The
dressing effectively inhibits bacterial formation and accelerates wound healing,
making it a promising treatment for infected wounds.
Pan et al. [45] focused on silica nanoparticles that are pH-sensitive and loaded
with Chlorhexidine for the specific release of the drug on alkaline wounds. These
nanoparticles were found to have antibacterial effects against both Gram-positive
and -negative bacterial pathogens and exhibited no cytotoxicity. The nanoparticles’
330 S. G. Reddy and H. C. A. Murthy
administration decreased the number of viable bacterial cells, suggesting they may
be useful in treating chronic wound infections.
A pH-responsive drug carrier for treating bone infections and promoting bone
tissue regeneration was developed by Zhang et al. [46]. The carrier comprises a
borate glass core and a mesoporous hydroxyapatite HA shell (BG-HA), coated with
glutaraldehyde-crosslinked chitosan (CS). Drug release is minimal at neutral pH, but
in acidic environments, the CS swells and opens the pores on the HA shell, resulting
in faster drug release. The carrier effectively eliminates bacteria like S. aureus and
E. coli and has the potential to gain self-regulated drug release, control the acidic
environment at bone infection sites, and promote the regeneration of bone tissue.
Patil [47] the study examined how permeable a pH-sensitive membrane made from
polyimide and polyacrylic acid (PI-PAAc) was and how it could be turned into a
glucose-sensitive membrane by attaching glucose oxidase (GOD) enzymes to it.
The PI membranes were initially created using a photolithographic process, with
PAAc being grafted onto the pores using a 248 nm KrF laser. The attached GOD
molecules were responsive to glucose and could transform it into gluconic acid. The
research suggests that these membranes can recognise and release corresponding
solute amounts, making them a potential candidate for glucose-sensing purposes.
Aashli et al. [48] the authors prepared transdermal films of Montelukast Sodium using
the solvent casting method. The films were prepared using different concentrations of
Polyvinyl alcohol (PVA), Polyethylene glycol (PEG), and Montelukast Sodium. The
prepared films were characterised using FTIR, DSC, SEM, and XRD techniques. The
drug release kinetics of Montelukast Sodium from the films were studied using the
in-vitro release method. The prepared transdermal films showed improved chemical
stability and extended drug release of Montelukast Sodium, making them an effective
drug delivery system for transdermal administration.
Smart Biomaterials in Drug Delivery Applications 331
The hydrogels that are pH-responsive using Carboxymethyl chitosan (CC), Sodium
alginate (SA), and Carvacrol (CA) synthesised by Cheng et al. [49]. The hydrogels
were formed through electrostatic interactions and hydrogen bonding, and increased
CC content resulted in better thermostability. The hydrogels increased in size with
higher pH, with CA release mainly controlled by Fickian diffusion. CA release
increased with temperature and pH, allowing for pH stimulation and on-demand
release. The hydrogels were stable for storage and biocompatible, providing a new
solution for environmentally responsive preservatives and intelligent preservation.
The production of halloysite nanotubes for a drug delivery system was reported by Lei
et al. [52] for responsiveness to pH. The drug release can be regulated based on the pH
of the environment surrounding the nanotubes by attaching pH-responsive polymers
to their surface. They discovered that halloysite nanotubes’ anisotropic structure
enhances their potential to hold and release drugs compared to other nanoparticle
types. The research suggests that this approach could have the potential for specific
drug delivery and other biomedical applications.
Elham Saleh et al. reported a nanocomposite material that can act as a carrier
for the controlled release of Piperine responsive to pH [53]. This material is made
by combining bovine serum albumin and oxidised gum Arabic. The material was
synthesised using a green approach and was stable and non-toxic. The controlled
release of Piperine was demonstrated using in vitro experiments, and the results
showed that the release rate was dependent on the pH of the solution. Also performed a
molecular docking study to investigate the interactions between the nanocomposite
material and Piperine. The study provides a promising approach for developing
pH-responsive drug delivery systems using natural and biocompatible materials.
de Jesus Oliveira et al. [54] conducted a study where they developed pH-responsive
phthalate cashew gum nanoparticles to enhance the delivery and effectiveness of
anti-Trypanosoma cruzi drugs. The study emphasised the importance of utilising
biodegradable and biocompatible materials for drug delivery systems. The nanopar-
ticles were spherical, stable, and responsive to pH levels, allowing for sustained drug
release. In vitro experiments indicated that drug-loaded nanoparticles had superior
efficacy against Trypanosoma cruzi compared to free drugs. The results suggest
that pH-responsive phthalate cashew gum nanoparticles could be a promising drug
delivery system for treating Chagas disease.
Lee et al. [55], describes a novel approach for treating renal fibrosis, a condi-
tion characterised by the excessive accumulation of extracellular matrix proteins in
the kidneys, leading to impaired kidney function. The approach involves using a pH-
responsive nanocarrier to deliver nitric oxide (NO), a signalling molecule with potent
anti-fibrotic effects, specifically to the fibrotic tissue in the kidneys. The nanocarrier
is designed to release NO in response to the acidic environment characteristic of
fibrotic tissue. The authors demonstrate that the pH-responsive nanocarrier effec-
tively delivers NO to fibrotic kidney tissue in a mouse model of renal fibrosis,
reducing fibrosis and improving kidney function. This approach has the potential
to be a promising therapeutic strategy for the treatment of renal fibrosis.
The pH-responsive Sodium alginate (SA) and Lignosulphonic (LS) biodegradable
polymeric blends with different proportions (80:20) were prepared by the solution
casting technique to achieve the desired drug release rate. The prepared polymeric
blends were characterised using FTIR. The drug release kinetics of Hydroxychloro-
quine sulphate (HCQ) from the polymeric blends were studied using the in-vitro
release method. The SA/LS blends showed a controlled release of HCQ over an
Smart Biomaterials in Drug Delivery Applications 333
extended period [56]. Crosslinking affects the control drug release of Hydroxychloro-
quine sulphate (HCQ) drug using alginate beads. The ionotropic gelation method
prepared the alginate beads and crosslinked them using different calcium chloride
concentrations. The drug release kinetics of HCQ from the beads were studied using
the in-vitro release method [57]. The authors concluded that the crosslinked alginate
beads showed a controlled release of HCQ over an extended period.
Temperature is another environmental stimulus that can be used to trigger the release
of drugs from stimuli-responsive materials. Temperature-responsive materials can
change their structure or solubility in response to temperature changes, resulting
in drug release [22]. Temperature-responsive materials are particularly useful for
localised drug delivery, as they can be triggered by changes in temperature at a
specific site in the body.
One class of temperature-responsive materials that have been extensively studied
is thermoresponsive hydrogels. These hydrogels can undergo a reversible phase tran-
sition from a swollen to a collapsed state in response to changes in temperature. Below
a certain temperature, the hydrogel is in a swollen state and can encapsulate the drug.
When the temperature is increased above a critical value, the hydrogel collapses,
resulting in the release of the drug. Another class of temperature-responsive materials
is liposomes. These materials can be designed to respond to changes in temperature,
causing the release of the encapsulated drug.
In summary, temperature-responsive materials have emerged as a promising tool
for developing controlled-release drug delivery systems. These materials can respond
to changes in temperature to release drugs in a controlled manner and can be used
for localised drug delivery. The development of temperature-responsive materials
has opened up new possibilities for drug delivery. Further research in this area is
expected to lead to the development of more effective and efficient drug delivery
systems.
Some of the well-known polymers that release drugs with temperature application
are furnished in Table 1.
These are a few examples of polymers commonly used in temperature-dependent
drug delivery systems. Depending on the specific drug delivery application and
desired temperature-responsive behaviour published by various researchers are
provided below.
334 S. G. Reddy and H. C. A. Murthy
the hydrophobic core of cyclodextrin and the azobenzene component. At low temper-
atures, the hydrogel can be injected through a syringe and become a gel after injection
due to its LCST-based properties. The researchers explored the drug release behaviour
under various light and temperature conditions. They also tested the efficacy of the
Paclitaxel-loaded hydrogel in vitro against cancerous fibroblastic cells (A-431) and
conducted in vivo studies using staining techniques.
The gold nanorods nanocomposite for synergistic breast cancer treatment was
developed by Rawand et al. [61]. The nanocomposites are coated with a pH-
temperature responsive polymer and Doxorubicin-loaded mesoporous silica for
drug delivery. Upon NIR radiation, the nanocomposite generates heat to induce
hyperthermia and trigger the release of Doxorubicin in response to high tempera-
ture/low pH at the tumour site. The nanocomposite demonstrated high efficiency
in suppressing MDA-MB-231 cell proliferation in vitro and good capability in
suppressing tumour growth in mice in vivo while exhibiting good biosafety.
The potential thermosensitive gels as carriers for tumour treatment in interven-
tional therapy developed by Hu et al. [62] can extend the lives of cancer patients,
especially those with primary liver cancer. These gels have biocompatibility and
drug retention ability and can turn gel-like in response to temperature changes,
allowing for rapid embolisation without the risk of ectopic embolisation. The authors
reviewed recent advancements in embolisation materials, including chitosan, poly-
acrylamide and poloxamers, to provide insights into developing and implementing
thermosensitive gels for cancer treatment.
Ruan et al. [63] address the issue of reversing drug resistance in cancer patients
undergoing chemotherapy and suggest that delivering drugs to the mitochondria
could be a solution. They developed a temperature-responsive drug delivery system
that targets the mitochondria and overcomes doxorubicin resistance in lung cancer.
The nanocarrier prevents drug efflux and allows drug accumulation in resistant
tumours, enhancing cytotoxicity and reversal of drug resistance in mice. This is
the first study to demonstrate the use of temperature-responsive drug delivery to
target the mitochondria and reverse drug resistance in cancer.
The drug delivery system was designed for treating lung cancer using temperature-
responsive micelles and hyaluronic acid biopolymers reported by Xu et al. [64]. The
system is created using layer-by-layer self-assembly technology, which enables effi-
cient drug loading and a controllable release mode based on environmental stimuli.
The study investigates the responsive behaviour of the films to temperature and
ionic strength and examines the influence of each component on the drug release
profile’s response to environmental triggers. The morphology integrity of the films
is retained after several temperature-triggered cycles. Additionally, the study exam-
ines the system’s intermolecular interaction and molecular motions to determine its
delivery efficiency. The development of these LBL films offers innovative ideas for
modifying traditional drug delivery materials and future functional material design.
The drug delivery system that responds to temperature and pH using a poly(N-
isopropyl acrylamide)-co-poly(acrylamide) copolymer and a melamine cross-linker
discussed by Thirupathi et al. [65]. The hydrogel system they created had phase tran-
sition properties sensitive to temperature. The drug release efficiency was sensitive
336 S. G. Reddy and H. C. A. Murthy
Mesoporous silica drug delivery method reported by Thirupathi et al. [69]. They
utilised a copolymer (PNIPAm-PAAm) that acted as a gatekeeper, reacting to changes
in temperature and pH, to cover the surface. This system regulates drug delivery under
various pH and temperature conditions. The system’s biocompatibility was confirmed
using MTT assay and cellular internalisation research. The research implies that the
Smart Biomaterials in Drug Delivery Applications 337
The drug delivery for stem cells in regenerative medicine is developed by Huang et al.
[72]. A temperature-sensitive hydrogel using a combination of two materials, methyl-
cellulose (MC) and polyvinyl alcohol (PVA) synthesised, to address the problem of
low cell survival and retention in cell delivery therapy. Creating physical hydrogen
bonds between MC and PVA chains produced porous hydrogels with a high water
retention capacity, stable modulus, fast gelation, and injectability. These hydrogels
are highly transparent to light. The team found that the hydrogels were non-toxic to
rabbit adipose-derived stem cells and could be utilised as a delivery method for stem
cells in regenerative medicine.
338 S. G. Reddy and H. C. A. Murthy
In their research, Ow et al. [75] examine the impact of ocular diseases on patients
and the healthcare system, and stress that nearly half of all vision problems can be
prevented or treated. One possible solution could be to use temperature-sensitive
hydrogels that are safe for the body and can be transformed into a gel by changing
the temperature. Recent advancements in temperature-responsive polymers show
potential for treating various eye conditions such as cataracts, dry eyes, glaucoma,
retinal detachment, and age-related macular degeneration. These developments may
have applications in various ocular treatments, such as vitreous substitutes for retinal
surgery, topical eye drops, and lenses that can reduce inflammation and discomfort.
A review article highlights the potential of these advances in treating ocular diseases.
Smart Biomaterials in Drug Delivery Applications 339
Liu et al. [79] have developed a new drug delivery system, which uses temperature-
responsive aerogels made from Hydroxypropyl methylcellulose (HPMC)-grafted N-
isopropyl acrylamide (NIPAM). The researchers conducted several analyses to deter-
mine the structure and morphology of the aerogels and tested the release behaviour of
a model drug, 5-Fluorouracil, from the aerogels. The study found that the release of
340 S. G. Reddy and H. C. A. Murthy
the drug was controlled and sustained and dependent on temperature. The researchers
also used models such as first-order kinetic, Higuchi, and Korsmyer-Peppas to
analyse the sustained-release mechanism. The results suggest that HPMC-NIPAM
aerogels could be a promising drug delivery system.
Hajebi et al. [80] describe the preparation of hybrid nanoparticles that are sensitive
to temperature. The nanoparticles were created using N-Isopropyl acrylamide and
vinyl-modified silica nanoparticles, with varying cross-linking densities achieved by
altering the N, N-Methylene bisacrylamide percentage. The silica cores were then
hydrolysed to form hollow poly(N-isopropyl acrylamide) nanogels. The researchers
studied the sensitivity of these nanogels to temperature using UV-vis spectroscopy
and dynamic light scattering. They also examined the release of the drug doxorubicin
from the nanogels at different temperatures and found that the Korsmeyer-Peppas
model provided the best fit for the release data.
Zhao and colleagues [81] developed a novel hydrogel loaded with micelles that
respond to changes in pH and temperature and can release hydrophobic drugs like
indomethacin in a controlled manner. The hydrogel was created using tremella
polysaccharide, carboxymethyl cellulose, and a nonionic surfactant called decyl
polyglucoside, and was prepared by crosslinking N-isopropyl acrylamide through
radical polymerisation. The hydrogel was subjected to various tests to characterise
its properties, and the researchers studied its drug release behaviour, which was
found to be influenced by changes in pH and temperature. The study suggests that
hydrogel has potential applications in biomedical fields such as drug delivery and
tissue engineering.
Light is another environmental stimulus that can be used to trigger the release of
drugs from stimuli-responsive materials. Light-responsive materials can be designed
to respond to specific wavelengths of light, resulting in the release of drugs. Light-
responsive materials are particularly useful for localised drug delivery, as they can
be triggered by light at a specific site in the body.
One class of light-responsive materials that have been extensively studied is
photoresponsive hydrogels. These hydrogels can undergo a reversible structural
Smart Biomaterials in Drug Delivery Applications 341
change in response to light, releasing the drug. The release mechanism can vary
depending on the type of photoresponsive material used. For example, some
photoresponsive materials undergo a photochemical reaction, while others undergo
a photothermal or photoisomerisation reaction. Another class of light-responsive
materials is nanoparticles, which can be designed to release drugs in response to
light.
Light-responsive materials are an efficient tool for developing controlled-release
drug delivery systems. These materials can respond to specific wavelengths of light
to release drugs in a controlled manner and can be used for localised drug delivery.
The development of light-responsive materials has opened up new possibilities for
drug delivery. Further research in this area is expected to lead to the development of
more promising and efficient drug delivery systems.
A small device made of DNA developed by Liu et al. [82] can deliver drugs in a
controlled manner to treat tumours. They modified GC-rich DNA duplexes with a
light-sensitive molecule to construct the device. They placed them on the surface
of nanoparticles that can convert near-infrared light into ultraviolet light. They then
added the chemotherapy drug doxorubicin to the DNA duplexes, where its fluorescent
signal was suppressed. When the nanoparticles were exposed to near-infrared light,
they emitted UV light, which caused the light-sensitive molecule to break and release
the doxorubicin in a controlled manner while restoring the fluorescent signal. The
nanodevice can selectively induce apoptosis in tumour cells through light activation
and allows in situ drug release monitoring. This approach has the potential for remote-
controlled drug delivery.
Lipid nanocapsules reported by Brion et al. [83] are sensitive to light and can
release an anti-tumour drug using photolysis. The researchers used photon upcon-
verting nanoparticles (LNC-UCs) connected to a photocleavable linker based on
coumarin. This linker can efficiently and quantitatively release the drug by upcon-
version luminescence-assisted photolysis when excited by a deep-red wavelength of
light. The nanocapsules are stable without light and do not release the drug, making
them suitable for nanomedicine applications. This technology allows for the precise
control of drug release or activation by light, making it ideal for the targeted treatment
of cancer and other diseases.
A hydrogel-based system for chemodynamic therapy (CDT) and photothermal
therapy (PTT) to treat tumours was designed by Liu et al. [84]. The system utilises a
composite nanomaterial called Cu-Hemin-Au loaded into agarose hydrogels, which
are injected intratumorally and stay in the tumour for an extended period. When
exposed to near-infrared light, Cu-Hemin-Au acts as a photothermal agent, gener-
ating heat, which causes the hydrogel to soften and release the Cu-Hemin-Au to
achieve photothermal therapy (PTT). The Au nanoparticles in Cu-Hemin-Au also
generate hydrogen peroxide, which reacts with Cu-Hemin-Au to produce reactive
342 S. G. Reddy and H. C. A. Murthy
oxygen species for CDT. The hydrogel system achieves good PTT/CDT synergy and
shows promising results in inhibiting tumour growth in mice. This hydrogel system
may inspire the development of new hydrogels for antitumor therapy.
The drug delivery system that responds to near-infrared (NIR-II) light using
Polyethylene glycol (PEG)-modified hollow Cux S nanoparticles developed by Zhou
et al. [85]. The nanoparticles were designed to release drugs at a high penetra-
tion depth, making them ideal for chemo-photothermal therapy. They demonstrated
high drug loading capacity and stimuli-responsive drug release triggered by NIR-II
laser irradiation. The study found that the nanoparticles achieved a 98.5% reduc-
tion in tumour size through synergistic chemo-photothermal therapy. These findings
suggest that CuxS-PEG nanoparticles have the potential as a comprehensive NIR-
II light-responsive nanocarrier-based drug delivery system for various biomedical
applications.
A gold nanorods (AuNRs) coated with mesoporous silica nanoshells (MSNs)
and two gatekeepers, Lauric acid (LA) and Tannic acid (TA), for pH- and near-
infrared (NIR) light-controlled release of drugs reported by Park et al. [86]. The pH-
sensitive TA layer swells or shrinks based on pH changes, enabling pH-responsive
drug release. In contrast, the thermosensitive LA layer undergoes a solid–liquid phase
transition due to heat generated by NIR light in AuNRs, enabling NIR light-controlled
drug release. The combination of these gatekeepers allows for on–off drug release
switching in acidic environments under NIR light without premature drug leakage.
The system has demonstrated effective anticancer performance.
In their study, Ray et al. [87] suggested that a combination of therapies could
be an effective approach to enhance the effectiveness of treatment and decrease
negative side effects. They created a new technology, which involved a dual-arm
acridine photocage that releases two amino and carboxylic acids at the same time.
This technology created a dual-drug delivery system that responds to visible light and
generates single-component fluorescent organic nanoparticles. These nanoparticles
were used to carry two anticancer drugs (chlorambucil and valproic acid) and were
tested on HeLa cell lines in vitro. The results showed that the nanoparticles had
better anticancer effectiveness and allowed for real-time fluorescence tracking of
drug release. The flat structure of the photocage facilitated its insertion into DNA,
thereby increasing the drugs’ cancer-killing potential.
Another gold nanostructure as a drug carrier for tunable optical properties against
cancer treatments was reported by Zafar et al. [88]. Stimuli-assisted drug delivery
systems, particularly those that are externally responsive, have received attention
from researchers as they provide more controlled drug delivery. Light-responsive
DDSs, particularly those that respond to near-infrared light, are potential candi-
dates due to their proficiency and spatiotemporal control. The article summarises
various shapes and structures of gold nanoparticles and how they can enhance the
effectiveness of drug delivery systems by exploiting the surface plasmon resonance
phenomenon.
Folic acid-functionalized polydopamine nanoparticles (NPs) for the combined
photothermal and chemotherapy treatment of breast cancer reported by Fan et al.
[89]. These NPs respond to tumour acidity and near-infrared light (NIR), allowing
Smart Biomaterials in Drug Delivery Applications 343
targeted drug delivery and enhanced cellular uptake. In vitro and in vivo studies
demonstrate that these NPs improve drug accumulation, penetration, and retention,
leading to higher therapeutic efficacy and better survival rate in breast cancer-bearing
mice. The article concludes that these NPs have the potential as a useful nanoscale
vector for enhanced cancer therapy.
Xue et al. [96] discuss the importance of exploring new materials for diagnosing and
treating brain diseases. Near-infrared (NIR) light-responsive materials are particu-
larly interesting due to their high spatial resolution and strong penetration into the
skull. The article summarises recent advances in developing NIR light-responsive
materials for diagnosing and treating various brain diseases, including approaches
targeting the blood–brain barrier (BBB), imaging-guided therapies, and challenges
and prospects for future research.
Xing et al. [97] examine the potential benefits of using light-sensitive hydrogels
for drug delivery. These hydrogels contain photosensitive components that enable
drug release through photoisomerisation, photochemical, or photothermal reactions.
Recent advances in materials science have created a wide range of photosensitisers
that can respond to various light sources, including ultraviolet and near-infrared
light, as well as up-conversion nanoparticles. Light-sensitive drug delivery systems
have been employed in various applications, such as chemotherapy, immunotherapy,
photodynamic therapy, gene therapy, and wound healing. The article provides an
overview of current research, challenges, and prospects in light-responsive hydrogels
for controlled drug delivery, emphasising the need to improve effectiveness, safety,
patient compliance, and convenience.
The article by Shan et al. [99] aims to enhance the efficacy of pesticide use while
reducing negative impacts on both the environment and human health. The authors
developed a biodegradable amphiphilic polymer via a simple and mild process. This
346 S. G. Reddy and H. C. A. Murthy
polymer was then used to create a nanosized pesticide system that responds to light
for controlled release of 2,4-dichloro phenoxy acetic acid (2,4-D). The system was
found to release the pesticide stably in the absence of UV light and showed increasing
release in the presence of UV light. The study demonstrated the system’s efficacy in
controlling weeds while reducing toxicity to non-target organisms.
Liu et al. [100] address the difficulties of targeted drug delivery and explore how light-
responsive drug delivery systems can help overcome these challenges. They explain
how photoremovable protecting groups, which are light-sensitive molecules that can
be attached to a drug molecule to control its function and structure with light, have
been used in recent applications of nanoparticle-based drug delivery. The authors
also discuss various approaches for achieving long-wavelength light excitation and
emphasise the importance of understanding these mechanisms for designing more
effective and precise photoresponsive drug delivery systems.
Khalifa et al. [101] have written about the benefits of pulsatile drug delivery
systems compared to conventional dosage forms. Pulsatile systems can administer
precise amounts of medication to targeted locations at specific times, enhancing
patient adherence and permitting customised treatment. The article concentrates
on recent breakthroughs in externally controlled pulsatile release systems, such as
electro-responsive, light-responsive, ultrasound-responsive, magnetically triggered,
and wirelessly managed implantable systems. The present state of these technologies
is explored, along with their possible future uses.
A review article was reported by Tang et al. [102] on the development and production
of nanocarriers for releasing drugs activated by near-infrared (NIR) light. The article
concentrates on NIR light with a 780–1700 nm wavelength range. It can penetrate
deep into tissues with minimal damage to cells, making it an ideal option for in vivo
light-activated delivery systems. The review highlights three nanocarrier triggering
mechanisms: chromophores’ photoreactions, photothermal effects caused by inor-
ganic or organic photothermal conversion agents, and photo-oxidation generated by
photosensitisers. Additionally, the article discusses the challenges and prospects for
creating NIR light-responsive nanocarriers.
Smart Biomaterials in Drug Delivery Applications 347
Magnetic fields are another environmental stimulus that can be used to trigger the
release of drugs from stimuli-responsive materials. Magnetic-responsive materials
can be designed to respond to an external magnetic field, releasing drugs [105].
Magnetic-responsive materials are particularly useful for targeted drug delivery, as
the magnetic field can guide the materials to a specific site in the body.
One class of magnetic-responsive materials that have been extensively studied in
magnetic nanoparticles. A temperature-responsive material can be used to coat these
nanoparticles, which react to temperature changes and cause the drug to be released
[106]. Additionally, a pH-responsive material can be used to coat the nanoparticles,
which react to pH changes and cause the drug to be released. Furthermore, an external
magnetic field can trigger the release of the drug, allowing the nanoparticles to be
guided to a particular location in the body [23].
Using magnetic-responsive materials has shown great potential in developing
drug delivery systems that can be controlled and released in a targeted manner. By
responding to an external magnetic field, these materials can be designed to release
drugs in a controlled way. The emergence of magnetic-responsive materials has
brought about new opportunities for drug delivery. As further studies are conducted
in this area, more efficient and effective drug delivery systems are expected to be
developed [107].
This chapter proposes the development of a soft, magnetically-responsive carrier
for drug delivery. The researchers used a moulding process to create prototypes from
a shape memory polymer called DiAPLEX MP-3510, which has a low transition
348 S. G. Reddy and H. C. A. Murthy
temperature. The design was validated through simulations, and the thermal respon-
siveness of the prototypes was tested both ex-vivo and in a phantom. The results show
that the design is viable and capable of delivering drugs to a targeted area in the large
phantom intestine, utilising the rubbery modulus of the polymer and enhancing the
recovery force through magnetic actuation [107].
Some of the articles published and their outcomes are furnished below.
Wound dressings are explored could effectively solve the problem of quick drug loss
and weak healing effects in patients who have undergone bladder tumour resection
[116]. To tackle this problem, they developed cellulose nanofibers wound dressing
that reacts to pH and near-infrared light, magnetic switching Fe3 O4 nanoparticles,
and temperature switching Pluronic®F-127. This dressing can be attached to the
350 S. G. Reddy and H. C. A. Murthy
tissue site, release drugs using an external magnetic field, and maintain a 3D network
for an extended time. Furthermore, the dressing exhibited strong antibacterial prop-
erties, eliminated biofilms, and killed T24 tumour cells. It also stimulated wound
healing through photothermal, photodynamic, and chemotherapy mechanisms. This
wound dressing is appropriate for bladder postoperative infected wound healing and
overcomes the issue of fast drug loss due to cyclical urination.
Li et al. [117] discuss how wound healing is a complicated process that requires
suitable microenvironments to be successful. These microenvironments consist of
various biological, chemical, and physical factors that are influenced by internal and
external factors. However, delivering drugs to wounds is challenging due to the need
for blood supply, persistent inflammation and other factors. Using stimuli-responsive
biomaterials can provide accurate drug delivery and release by responding to various
factors from wound microenvironments or external sources. The article discusses
recent developments in stimuli-responsive biomaterials to regulate microenviron-
ments during wound healing. Various biomaterials that respond to wound and
physical microenvironments and new drug carriers are highlighted.
A hydrogel was developed based on 2D MXene that uses photo and magnetic-
responsive drug delivery to treat chronic wounds [118]. This system includes
magnetic colloids wrapped with MXene and dual-network hydrogels, which enable
multiple response capabilities and controlled drug delivery. They tested this system
on infected wounds in a rat model and found it has potential for clinical wound
healing and other biomedical applications.
As a review reported by Armenia et al. [125], smart nanomedicine can enhance drug
delivery using smart nanomaterials that respond to internal and external stimuli. To
increase responsiveness to external stimuli, photonic and magnetic nanoparticles are
utilised. The authors highlight recent progress in integrating these materials within
352 S. G. Reddy and H. C. A. Murthy
different carriers to improve efficacy, stability, and toxicity. They also discuss the
current regulatory hurdles and the importance of standardising these materials.
A new hydrogel composite of ferrogel, responsive to magnetic fields by adding
Fe3 O4 nanoparticles to a pre-existing hydrogel, was temperature-responsive [126].
The study then investigated how the composition of the ferrogel affected its response
to alternating magnetic fields. The researchers also used artificial neural networks
to predict how the ferrogel would behave in different conditions accurately. These
findings have important implications for developing smart materials for use in the
biomedical field.
The device to treat bone infections called osteomyelitis was reported by Shademani
et al. [127], which includes a magnetic sponge capable of on-demand drug delivery.
This device employs a combination of Silver nitrate and Gentamicin to combat drug-
resistant pathogens effectively. The drug delivery system can release drugs for seven
days and is easy to fabricate, allowing for control of the release profile.
release achieved only under magnetic stimulation. The study suggests these nanopar-
ticles have good in vitro performance and are expected to have suitable in vivo
behaviour.
Magnetic/pH dual-sensitive hydrogels are synthesised by physically incorporating
superparamagnetic iron oxide nanoparticles (Fe3 O4 ) into Dextran hydrogels through
Schiff base reactions [130]. The hydrogels displayed magnetic and pH-responsive
behaviour and could release Doxorubicin in a controlled manner through a combina-
tion of diffusion, swelling, and erosion processes. The hydrogel/Fe3 O4 composites
have potential applications in various fields, including drug delivery.
6 Conclusion
References
1. Niculescu, A.G., Grumezescu, A.M.: Novel tumor-targeting nanoparticles for cancer treat-
ment—A review. Int. J. Mol. Sci. 23, 5253 (2022)
2. Sung, H., Ferlay, J., Siegel, R.L., Laversanne, M., Soerjomataram, I., Jemal, A., Bray, F.:
Glob. Cancer Stat. (2020)
3. GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185
Countries. CA Cancer J. Clin. 71, 209–249 (2021)
4. Wang, S., Chen, Y., Guo, J., Huang, Q.: Liposomes for tumor targeted therapy: a review. Int.
J. Mol. Sci. 24, 2643 (2023)
5. Banerjee, R.: Nanotechnology in drug delivery: present status and a glimpse into the future.
Ther. Deliv. 9(4), 231 (2018)
6. Fleming, S., Ulijn, R.V.: Design of nanostructures based on aromatic peptide amphiphiles.
Chem. Soc. Rev. 43, 8150–8177 (2014)
7. Lin, G.G., Scott, J.G.: Peptide-directed self-assembly of hydrogels. Acta Biomater. 100, 130–
134 (2012)
8. Jonker, A.M., Löwik, D.W.P.M., Van Hest, J.C.M.: Peptide- and protein-based hydrogels.
Chem. Mater. 24, 759–773 (2012)
9. Luk, B.T., Zhang, L.: Current advances in polymer-based nanotheranostics for cancer
treatment and diagnosis. ACS Appl. Mat. Interf. 6, 21859–21873 (2014)
354 S. G. Reddy and H. C. A. Murthy
10. Zhang, L., Zhu, D., Dong, X., Sun, H., Song, C., Wang, C., Kong, D.: Folate-modified lipid–
polymer hybrid nanoparticles for targeted paclitaxel delivery. Int. J. Nanomed. 10, 2101–2114
(2015)
11. Ahmad, Z., Shah, A., Siddiq, M., Kraatz, H.-B.: Polymeric micelles as drug delivery vehicles.
RSC Adv. 4, 17028–17038 (2014)
12. Lin, Y.-K., Yu, Y.-C., Wang, S.-W., Lee, R.-S.: Temperature, ultrasound and redox triple-
responsive poly(N-isopropylacrylamide) block copolymer: synthesis, characterization and
controlled release. RSC Adv. 7(43), 212–226 (2017)
13. Huang, X., Liao, W., Zhang, G., Kang, S., Zhang, C.Y.: PH-sensitive micelles self-assembled
from polymer brush (PAE-g-cholesterol)-b-PEG-b-(PAE-g-cholesterol) for anticancer drug
delivery and controlled release. Int. J. Nanomed. 12, 2215–2226 (2017)
14. Zhang, C.Y., Wu, W.S., Yao, N., Zhao, B., Zhang, L.J.: PH-sensitive amphiphilic copolymer
brush Chol-g-P(HEMA-co-DEAEMA)-b-PPEGMA: synthesis and self-assembled micelles
for controlled anti-cancer drug release. RSC Adv. 4(40), 232–240 (2014)
15. Dharadhar, S., Majumdar, A., Dhoble, S., Patravale, V.: Microneedles for transdermal drug
delivery: a systematic review. Drug Dev. Ind. Pharm. 45, 188–201 (2019)
16. Prausnitz, M.R.: Engineering microneedle patches for vaccination and drug delivery to skin.
Annu. Rev. Chem. Biomol. Eng. 8, 177–200 (2017)
17. Ye, Y., Yu, J., Wen, D., Kahkoska, A.R., Gu, Z.: Polymeric microneedles for transdermal
protein delivery. Adv. Drug Deliv. Rev. 127, 106–118 (2018)
18. Kim, J.Y., Han, M.R., Kim, Y.H., Shin, S.W., Nam, S.Y., Park, J.H.: Tip-loaded
dissolving microneedles for transdermal delivery of donepezil hydrochloride for treatment
of Alzheimer’s disease. Eur. J. Pharm. Biopharm. 105, 148–155 (2016)
19. Ma, G., Wu, C.: Microneedle, bio-microneedle and bio-inspired microneedle: a review. J.
Control Release 251, 11–23 (2017); Prausnitz, M., Langer, R.: Transdermal drug delivery.
Nat. Biotechnol. 26, 1261–1268 (2008)
20. Yao, J., Feng, J., Chen, J.: External-stimuli responsive systems for cancer theranostic. Asian
J. Pharm. Sci. 11(5), 585–595 (2016)
21. Yang, G., Liu, J., Wu, Y., Feng, L., Liu, Z.: Near-infrared-light responsive nanoscale drug
delivery systems for cancer treatment. Coord. Chem. Rev. 320–321, 100–117 (2016)
22. Kamaly, N., Yameen, B., Wu, J., Farokhzad, O.C.: Degradable controlled-release polymers
and polymeric nanoparticles: mechanisms of controlling drug release. Chem. Rev. (2016)
23. www.ncbi.nlm.nih.gov
24. Li, L., Yang, W.-W., Xu, D.-G.: Stimuli-responsive nanoscale drug delivery systems for cancer
therapy. J. Drug Target. (2019)
25. Gutiérrez, L., Stepien, G., Gutiérrez, L., Pérez-Hernández, M., Pardo, J., Pardo, J., Grazú, V.,
de la Fuente, J.M.: Nanotechnology in Drug Discovery and Development. Elsevier (2017)
26. Pilch, J., Pot˛ega, A., Kowalczyk, A., Kasprzak, A., Kowalik, P., Bujak, P., Paluszkiewicz, E.,
Augustin, E., Nowicka, A.M.: PH-responsive drug delivery nanoplatforms as smart carriers
of unsymmetrical bisacridines for targeted cancer therapy. Pharmaceutics 15(1), 201 (2023)
27. Altinbasak, I., Kocak, S., Colby, A.H., Alp, Y., Sanyal, R., Grinstaff, M.W., Sanyal, A.:
Biomater. Sci. 11, 813–821 (2023)
28. Gao, J., Si, F., Wang, F., Li, Y., Wang, G., Zhao, J., Ma, Y., Yu, R., Li, Y., Jin, C., Li, D.:
Hollow mesoporous structured MnFe2O4 nanospheres: a biocompatible drug delivery system
with pH-responsive release for potential application in cancer treatment. Solid State Sci. 135,
107066 (2023)
29. Son, M.J., Kim, T., Lee, S.-W.: Facile synthesis of fluorescent mesoporous nanocarriers with
pH-sensitive controlled release of naturally derived dieckol. In: Colloids and Surfaces A:
physicochemical and Engineering Aspects, Part A, vol. 657, p. 130535 (2023)
30. Anggraini, S.A., Prasetija, K.A., Yuliana, M., Wijaya, C.J., Bundjaja, V., Angkawijaya, A.E.,
Jiang, Y.-F., Putro, J.N., Hartono, S.B., Ayucitra, A., Santoso, S.P., Ismadji, S., Soetaredjo,
F.E.: PH-responsive hollow core zeolitic-imidazolate framework-8 as an effective drug carrier
of 5-fluorouracil. Mater. Today Chem. 27, 101277 (2023)
Smart Biomaterials in Drug Delivery Applications 355
31. Ghazimoradi, M., Tarlani, A., Alemi, A., Hamishehkar, H., Ghorbani, M.: “pH-responsive,
magnetic-luminescent core/shell carriers for co-delivery of anticancer drugs (MTX & DOX)
for breast cancer treatment. J. Alloy. Compd. 936, 168257 (2023)
32. Zhang, G., Han, W., Zhao, P., Wang, Z., Li, M., Sui, X., Liu, Y., Tian, B., He, Z., Fu, Q.:
Nanoscale 15, 1937–1946 (2023)
33. Akkaya, B., Akkaya, R.: Sümeyye Idil Celikkaya, Nurgül Sarıaydin, Kayode Yomi Raheem,
Doxorubucin loaded pH-responsive chitosan-poly(acrylamide-maleic acid) composite
hydrogel for anticancer targeting. J. Mol. Struct. 1274(1), 134536 (2023)
34. Truong-Thi, N.-H., Nguyen, N.H., Nguyen, D.T.D., Tang, T.N., Nguyen, T.H., Nguyen, D.H.:
pH-responsive delivery of Platinum-based drugs through the surface modification of heparin
on mesoporous silica nanoparticles. Euro. Polym. J. 185, 111818 (2023)
35. Sheykhisarem, R., Dehghani, H.: In vitro biocompatibility evaluations of pH-sensitive
Bi2MoO6/NH2-GO conjugated polyethylene glycol for release of daunorubicin in cancer
therapy. Colloids Surf. B 221, 113006 (2023)
36. Gou, K., Xin, W., Lv, J., Ma, Z., Yang, J., Zhao, L., Cheng, Y., Chen, X., Zeng, R., Li,
H.: A pH-responsive chiral mesoporous silica nanoparticles for delivery of doxorubicin in
tumour-targeted therapy. Colloids Surf. B 221, 113027 (2023)
37. Zhang, C., Yuan, Q., Zhang, Z., Tang, Y.: A pH-responsive drug delivery system based on
conjugated polymer for effective synergistic chemo-/photodynamic therapy. Molecules 28(1),
399 (2023)
38. Rao, K.-M., Rao, K.S.V.K., Palem, R.-R., Uthappa, U.-T., Ha, C.-S., Han, S.-S.: PH sensitive
drug delivery behavior of palmyra palm kernel hydrogel of chemotherapeutic agent. Gels
9(1), 38 (2023)
39. Heragh, B.K., Taherinezhad, H., Mahdavinia, G.R., Javanshir, S., Labib, P., Ghasemsolb, S.:
pH-responsive co-delivery of doxorubicin and saffron via cross-linked chitosan/laponite RD
nanoparticles for enhanced-chemotherapy. Mater. Today Commun. 34, 104956 (2023)
40. Teixeira, P.V., Adega, F., Martins-Lopes, P., Machado, R., Lopes, C.M., Lúcio, M.: PH-
responsive hybrid nanoassemblies for cancer treatment: formulation development, optimiza-
tion, and in vitro therapeutic performance. Pharmaceutics 15(2), 326 (2023)
41. Li, B.-L., Zhang, J., Jin, W., Chen, X.-Y., Yang, J.-M., Chi, S.-M., Ruan, Q., Zhao, Y.:
Oral administration of pH-responsive polyamine-modified cyclodextrin nanoparticles for
controlled release of anti-tumour drugs. React. Funct. Polym. 172, 105175 (2022)
42. Batool, N., Sarfraz, R.M., Mahmood, A., Rehman, U., Zaman, M., Akbar, S., Almasri, D.M.,
Gad, H.A.: Development and evaluation of cellulose derivative and pectin based swellable
pH responsive hydrogel network for controlled delivery of cytarabine. Gels 9(1), 60 (2023)
43. Du, M., Jin, J., Zhou, F., Chen, J., Jiang, W.: Dual drug-loaded hydrogels with pH-responsive
and antibacterial activity for skin wound dressing. Colloids Surf. B: Biointerfaces 222, 113063
(2023)
44. Xie, X., Lei, H., Fan, D.: Antibacterial hydrogel with pH-responsive microcarriers of slow-
release VEGF for bacterial infected wounds repair. J. Mater. Sci. Technol. 144, 198–212
(2023)
45. Pan, F., Giovannini, G., Zhang, S., Altenried, S., Zuber, F., Chen, Q., Boesel, L.F., Ren, Q.:
PH-responsive silica nanoparticles for the treatment of skin wound infections. Acta Biomater.
145, 172–184 (2022)
46. Zhang, R., Ding, J., Lu, X., Yao, A., Wang, D.: pH-responsive drug release and antibacterial
activity of chitosan-coated core/shell borate glass-hydroxyapatite microspheres. Ceramics Int.
49(3), 5161–5168 (2023)
47. Patil, R.S., Narayanan, A., Tantisuwanno, C., Sancaktar, E.: Immobilization of glucose oxidase
on pH-responsive polyimide-polyacrylic acid smart membranes fabricated using 248 nm KrF
excimer laser for drug delivery, 13(1), 11 (2023)
48. Reddy, S.G., Kumar, B.S., Prashanthi, K., Murthy, H.A.: Fabricating transdermal film formu-
lations of montelukast sodium with improved chemical stability and extended drug release.
Heliyon 9(3), e14469 (2023)
356 S. G. Reddy and H. C. A. Murthy
49. Cheng, M., Cui, Y., Guo, Y., Zhao, P., Wang, J., Zhang, R., Wang, X.: Design of carboxymethyl
chitosan-reinforced pH-responsive hydrogels for on-demand release of carvacrol and simu-
lation of release kinetics. Food Chem. 405(Part A), 134856 (2023)
50. Wang, Y., Peng, Z., Yang, Y., Li, Z., Wen, Y., Liu, M., Li, S., Su, L., Zhou, Z., Zhu, Y., Zhou, N.:
Auricularia auricula biochar supported γ-FeOOH nanoarrays for electrostatic self-assembly
and pH-responsive controlled release of herbicide and fertilizer. Chem. Eng. J. 437(Part 1),
134984 (2022)
51. Banihashemian, A.R., Kowsari-Esfahan, S., Ebrahimzadeh, M., Jafari, M., Akbari, A., Karimi-
Maleh, H.: PH-responsive drug delivery using graphene-based materials. Nanoscale 13(3),
1801–1823 (2021). https://doi.org/10.1039/D0NR06675J
52. Lei, X., Zhou, Y., Liu, X., Kong, L., Liao, L., Li, Y., Liu, M., Tian, L., Rao, W., Lv, G.:
Effective pH-responsive nanocarrier based on the anisotropic surfaces of halloysite nanotubes
for controlled drug release. Appl. Clay Sci. 232, 106799 (2023). ISSN:0169-1317
53. Jalali, E.S., Shojaosadati, S.A., Hamedi, S.: Green synthesis of bovine serum albumin/oxidized
gum Arabic nanocomposite as pH-responsive carrier for controlled release of piperine and
the molecular docking study. Int. J. Biol. Macromol. 225, 51–62 (2023). ISSN:0141-8130
54. de Jesus Oliveira, A.C., Silva, E.B., de Oliveira, T.C., Ribeiro, F.D.O.S., Nadvorny, D., de
Freitas Oliveira, J.W., Borrego-Sánchez, A., da Franca Rodrigues, K.A., Silva, M.S., Rolim-
Neto, P.J., Viseras, C.: pH-responsive phthalate cashew gum nanoparticles for improving
drugs delivery and anti-Trypanosoma cruzi efficacy. Int. J. Bio. Mol. 230, 123272 (2023)
55. Lee, T.-Y., Lu, H.-H., Cheng, H.-T., Huang, H.-C., Tsai, Y.-J., Chang, I.-H., Tu, C.-P.,
Chung, C.-W., Lu, T.-T., Peng, C.-H., Chen, Y.: Delivery of nitric oxide with a pH-responsive
nanocarrier for the treatment of renal fibrosis. J. Controll. Release 354, 417–428 (2023)
56. Giridhar Reddy, S.: Controlled release studies of Hydroxychloroquine sulphate (HCQ) drug-
using Biodegradable polymeric Sodium alginate and Lignosulphonic acid Blends. Rasayan
J. Chem. 4, 2209–2215 (2021)
57. Giridhar Reddy, S.: Effect of crosslinking on control drug release of hydroxychloroquine
sulphate drug-using alginate beads. Iran. J. Mater. Sci. Eng. 19(2) (2022)
58. Hemmatpour, H., Haddadi-Asl, V., Burgers, T.C., Yan, F., Stuart, M.C., Reker-Smit, C., Vlijm,
R., Salvati, A., Rudolf, P.: Nanoscale 15, 2402–2416 (2023)
59. Lin, Z., Ding, J., Chen, X., He, C.: pH- and temperature-responsive hydrogels based on tertiary
amine-modified polypeptides for stimuli-responsive drug delivery (2023)
60. Pourbadiei, B., Adlsadabad, S.Y., Rahbariasr, N., Pourjavadi, A.: Synthesis and characteriza-
tion of dual light/temperature-responsive supramolecular injectable hydrogel based on host-
guest interaction between azobenzene and starch-grafted β-cyclodextrin: Melanoma therapy
with paclitaxel. Carbohydr. Polym. 120667 (2023)
61. Mustafa, R.A., Ran, M., Wang, Y., Yan, J., Zhang, Y., Rosenholm, J.M., Zhang, H.: A pH/
temperature responsive nanocomposite for chemo-photothermal synergistic cancer therapy.
Smart Mater. Med. 4, 199–211 (2023)
62. Hu, X.-E., Shi, Y.-R., Zhu, X., Tian, K.-W., Xu, X.-L.: Temperature-responsive hydrogel for
tumor embolization therapy. J. Drug Deliv. Sci. Technol. 80, 104107 (2023)
63. Ruan, L., Chen, J., Du, C., Lu, H., Zhang, J., Cai, X., Dou, R., Lin, W., Chai, Z., Nie, G.,
Hu, Y. Mitochondrial temperature-responsive drug delivery reverses drug resistance in lung
cancer. Bioact. Mater. 13, 191–199 (2022)
64. Xu, L., Wang, H., Chu, Z., Cai, L., Shi, H., Zhu, C., Pan, D., Pan, J., Fei, X., Lei, Y.:
Temperature-responsive multilayer films of micelle-based composites for controlled release
of a third-generation EGFR inhibitor. ACS Appl. Polym. Mater. 2(2), 741–750 (2020)
65. Thirupathi, K., Phan, T.T.V., Santhamoorthy, M., Ramkumar, V., Kim, S.-C.: PH and ther-
moresponsive PNIPAm-co-polyacrylamide hydrogel for dual stimuli-responsive controlled
drug delivery. Polymers 15(1), 167 (2023)
66. Balan, K.E., Boztepe, C., Künkül, A.: Modeling the effect of physical crosslinking degree
of pH and temperature responsive poly(NIPAAm-co-VSA)/alginate IPN hydrogels on drug
release behavior. J. Drug Deliv. Sci. Technol. 75, 103671 (2022)
Smart Biomaterials in Drug Delivery Applications 357
67. Boztepe, C., Künkül, A., Yüceer, M.: Application of artificial intelligence in modeling of the
doxorubicin release behavior of pH and temperature responsive poly(NIPAAm-co-AAc)-PEG
IPN hydrogel. J. Drug Deliv. Sci. Technol. 57, 101603 (2020)
68. Sakai, S., Hirano, Y., Kobayashi, Y., Arai, N.: Effect of temperature on the structure and
drug-release behaviour of inclusion complex of β-cyclodextrin with cyclophosphamide: a
molecular dynamics study. Soft Matter. (2023)
69. Thirupathi, K., Santhamoorthy, M., Radhakrishnan, S., Ulagesan, S., Nam, T.-J., Phan, T.T.V.,
Kim, S.-C.: Thermosensitive polymer-modified mesoporous silica for pH and temperature-
responsive drug delivery. Pharmaceutics 15(3), 795 (2023)
70. Yu, S., Xing, J.: Preparation of temperature-responsive PMMA-based microspheres encap-
sulating erythromycin in situ by emulsion photopolymerization. J. Drug Deliv. Sci. Technol.
81, 104256 (2023)
71. Işıklan, N., Erol, Ü.H.: Design and evaluation of temperature-responsive chitosan/
hydroxypropyl cellulose blend nanospheres for sustainable flurbiprofen release. Int. J. Biol.
Macromol. 159, 751–762 (2020)
72. Huang, Z., Xiao, X., Jiang, X., Yang, S., Niu, C., Yang, Y., Yang, L., Li, C., Feng, L.: Prepara-
tion and evaluation of a temperature-responsive methylcellulose/polyvinyl alcohol hydrogel
for stem cell encapsulation. Polym. Test. 119, 107936 (2023)
73. Emam, H.E., Shaheen, T.I.: Design of a dual pH and temperature responsive hydrogel based
on esterified cellulose nanocrystals for potential drug release. Carbohydr. Polym. 278, 118925
(2022)
74. Zhu, Y., Zhang, M., Wei, S., Wang, B., He, J., Qiu, X.: Temperature-responsive P(NIPAM-
co-NHMA)-grafted organic-inorganic hybrid hollow mesoporous silica nanoparticles for
controlled drug delivery. J. Drug Deliv. Sci. Technol. 70, 103197 (2022)
75. Ow, V., Loh, X.J.: Recent developments of temperature-responsive polymers for ophthalmic
applications. J. Polym. Sci. (2022)
76. Hemmatpour, H., Haddadi-Asl, V., Burgers, T.Q., Yan, F., Stuart, M.C.A., Reker-Smit, C.,
Vlijm, R., Salvati, A., Rudolf, P.: Temperature-responsive and biocompatible nanocarriers
based on clay nanotubes for controlled anti-cancer drug release. Nanoscale 15, 2402–2416
(2023)
77. Choi, Y., Kim, J., Yu, S., Hong, S.: pH- and temperature-responsive radially porous silica
nanoparticles with high-capacity drug loading for controlled drug delivery. Nanotech-
nology 31, 335103 (2020)
78. Kotsuchibashi, Y.: Recent advances in multi-temperature-responsive polymeric materials.
Polym. J. 52, 681–689 (2020)
79. Liu, Z., Zhang, S., He, B.: Temperature-responsive hydroxypropyl methylcellulose-N-
isopropylacrylamide aerogels for drug delivery systems. Cellulose 27, 9493–9504
80. Hajebi, S., Abdollahi, A., Roghani-Mamaqani, H., Salami-Kalajahi, M.: Temperature-
responsive poly(N-Isopropylacrylamide) Nanogels: the role of hollow cavities and different
shell cross-linking densities on doxorubicin loading and release. Langmuir 36(10), 2683–2694
(2020)
81. Zhao, H., Li, Y.: A novel pH/temperature-responsive hydrogel based on tremella polysaccha-
ride and poly(N-isopropylacrylamide). Colloids Surf. A 586, 124270 (2020)
82. Liu, Q., Cheng, H.-B., Ma, R., Yu, M., Huang, Y., Li, L., Zhao, J.: A DNA-based nanodevice
for near-infrared light-controlled drug release and bioimaging. Nano Today 48, 101747 (2023)
83. Brion, A., Chaud, J., Léonard, J., Bolze, F., Chassaing, S., Frisch, B., Heurtault, B., Kichler, A.,
Specht, A.: Red light-responsive upconverting nanoparticles for quantitative and controlled
release of a coumarin-based prodrug. Adv. Healthc. Mater. 12, 2201474 (2023)
84. Liu, Z., Chen, H., Huang, C., Huang, Q.: A light-responsive injectable hydrogel with remod-
eling tumor microenvironment for light-activated chemodynamic therapy. Macromol. Biosci.
23, 2200329 (2023)
85. Zhou, T., Xie, S., Zhou, C., Chen, Y., Li, H., Liu, P., Jiang, R., Hang, L., Jiang, G.: All-
in-one second near-infrared light-responsive drug delivery system for synergistic chemo-
photothermal therapy. ACS Appl. Bio Mater. 5(8), 3841–3849 (2022)
358 S. G. Reddy and H. C. A. Murthy
86. Park, J.H., Sung, K.E., Kim, K.H., Kim, J.R., Kim, J., Moon, G.D., Hyun, D.C.: Dual gate-
keeping and reversible on-off switching drug release for anti-cancer therapy with pH- and NIR
light-responsive mesoporous silica-coated gold nanorods. J. Ind. Eng. Chem. 106, 233–242
(2002)
87. Ray, S., Banerjee, S., Singh, A.K., Ojha, M., Mondal, A., Singh, N.D.P.: Visible light-
responsive delivery of two anticancer drugs using single-component fluorescent organic
nanoparticles. ACS Appl. Nano Mater. 5(5), 7512–7520 (2022)
88. Zafar, M., Ijaz, M., Iqbal, T.: Efficient Au nanostructures for NIR-responsive controlled drug
delivery systems. Chem. Pap. 75, 2277–2293 (2021)
89. Fan, R., Chen, C., Hou, H., Chuan, D., Mu, M., Liu, Z., Liang, R., Guo, G., Xu, J.:
Tumor acidity and near-infrared light responsive dual drug delivery polydopamine-based
nanoparticles for chemo-photothermal therapy. Adv. Funct. Mater. 31, 2009733 (2021)
90. Rafiee, Z., Bodaghi, A., Omidi, S.: Fabrication of a photo- and pH-sensitive micelle by self-
assembly of azobenzene polyglycerol for anticancer drug delivery. Monatsh. Chem. 154,
259–265 (2023)
91. Patil, T.V., Dutta, S.D., Patel, D.K., Ganguly, K., Lim, K.-T.: Electrospinning near infra-red
light-responsive unzipped CNT/PDA nanofibrous membrane for enhanced antibacterial effect
and rapid drug release. Appl. Surf. Sci. 612, 155949 (2023)
92. Luo, J., Ma, Z., Yang, F., Wu, T., Wen, S., Zhang, J., Huang, L., Deng, S., Tan, S.: Fabrication
of laponite-reinforced dextran-based hydrogels for NIR-responsive controlled drug release.
ACS Biomater. Sci. Eng. 8(4), 1554–1565 (2022)
93. Hu, D., Zhang, C., Sun, C., et al.: Carvacrol combined with NIR light-responsive nano-drug
delivery system with specific anti-bacteria, anti-inflammation, and immunomodulation for
periodontitis. Nano Res. (2023)
94. Zhu, W., Zhao, Y., Wu, Z., Lv, F., Zhang, Y., Guo, S.: Application of UV responsive SiO2/
PVP composite hydrogels as intelligent controlled drug release patches. Polymer 264, 125535
(2023)
95. Huang, X., Xu, L., Yu, X., Li, Y., Huang, Z., Xu, R., Zeng, W., Zhang, Z., Li, W., Deng,
F.: Near-infrared light-responsive multifunctional hydrogel releasing peptide-functionalized
gold nanorods sequentially for diabetic wound healing. J. Colloid Interface Sci. 639 (2023)
96. Xue, D., Wang, Y., Zhang, H.: Advances of NIR light responsive materials for diagnosis and
treatment of brain diseases. Adv. Opt. Mater. 2202888 (2023)
97. Xing, Y., Zeng, B., Yang, W.: Light responsive hydrogels for controlled drug delivery. Front
Bioeng Biotechnol. 16(10), 1075670 (2022)
98. Abdelmohsen, H.A.M., Copeland, N.A., Hardy, J.G.: Light-responsive biomaterials for ocular
drug delivery. Drug Deliv. Transl. Res. (2022)
99. Shan, P., Lu, Y., Lu, W., Yin, X., Liu, H., Li, D., Lian, X., Wang, W., Li, Z., Li, Z.: Biodegrad-
able and light-responsive polymeric nanoparticles for environmentally safe herbicide delivery.
ACS Appl. Mater. Interfaces 14(38), 43759–43770 (2022)
100. Liu, J., Kang, W., Wang, W.: Photocleavage-based photoresponsive drug delivery. Photochem.
Photobiol. 98, 288–302 (2022)
101. Khalifa, A.Z., Zyad, H., Mohammed, H., Ihsan, K., Alrawi, L., Abdullah, M., Akram, O.:
Recent advances in remotely controlled pulsatile drug delivery systems. J. Adv. Pharm.
Technol. Res. 13(2), 77–82 (2022)
102. Tang, Y., Wang, G.: NIR light-responsive nanocarriers for controlled release. J. Photochem.
Photobiol. C: Photochem. Rev. 47, 100420 (2021)
103. Giménez, V.M.M., Arya, G., Zucchi, I.A., Galante, M.J., Manucha, W.: Soft Matter. (38),
2021 (2021)
104. Huang, R., Lan, R., Shen, C., Zhang, Z., Wang, Z., Bao, J., Wang, Z., Zhang, L., Hu, W.,
Yu, Z., Zhu, S.: Remotely controlling drug release by light-responsive cholesteric liquid
crystal microcapsules triggered by molecular motors. ACS Appl. Mater. Interfaces 13(49),
59221–59230 (2021)
105. Yang, L., Guo, C., Jia, L., Xie, K., Shou, Q., Liu, H.: Fabrication of biocompatible temperature-
and pH-responsive magnetic nanoparticles and their reversible agglomeration in aqueous
Milieu. Ind. Eng. Chem. Res. (2010)
Smart Biomaterials in Drug Delivery Applications 359
106. Rakshit, S., Sivasankar, S.: Cross-linking of a charged polysaccharide using polyions as
electrostatic staples. Soft Matter. (2011)
107. Heunis, C.M., Wang, Z., de Vente, G., Misra, S., Venkiteswaran, V.K.: A magnetic bio-inspired
soft carrier as a temperature-controlled gastrointestinal drug delivery system. Macromol.
Biosci. 2200559 (2023)
108. Zhu, J., Wang, J., Li, Y.: Recent advances in magnetic nanocarriers for tumor treatment.
Biomed. Pharmacother. 159, 114227 (2023)
109. Dagdelen, S., Mackiewicz, M., Osial, M., et al.: Redox-responsive degradable microgel modi-
fied with superparamagnetic nanoparticles exhibiting controlled, hyperthermia-enhanced drug
release. J. Mater. Sci. 58, 4094–4114 (2023)
110. Durkut, S.: Fe3 O4 magnetic nanoparticles-loaded thermoresponsive poly(N-
vinylcaprolactam)-g-galactosylated chitosan microparticles: investigation of physico-
chemical, morphological and magnetic properties. J. Macromol. Sci. Part A, 181–191
(2023)
111. Gonçalves, A., Simões, B.T., Almeida, F.V., Fernandes, S.N., Valente, M., Vieira, T.,
Henriques, C., Borges, J.P., Soares, P.I.P.: Engineering dual-stimuli responsive poly(vinyl
alcohol) nanofibrous membranes for cancer treatment by magnetic hyperthermia. Biomater.
Adv. 145, 213275 (2023)
112. Wang, X., Qi, Y., Hu, Z., et al.: Fe3 O4 @PVP@DOX magnetic vortex hybrid nanostructures
with magnetic-responsive heating and controlled drug delivery functions for precise medicine
of cancers. Adv. Compos. Hybrid Mater. 5, 1786–1798 (2022)
113. Mazidi, Z., Javanmardi, S., Naghib, S.M., Mohammadpour, Z.: Smart stimuli-responsive
implantable drug delivery systems for programmed and on-demand cancer treatment: an
overview on the emerging materials. Chem. Eng. J. 433(Part 1), 134569 (2022)
114. Demin, A.M., Vakhrushev, A.V., Pershina, A.G., Valova, M.S., Efimova, L.V., Syomchina,
A.A., Uimin, M.A., Minin, A.S., Levit, G.L., Krasnov, V.P., Charushin, V.N.: Magnetic-
responsive doxorubicin-containing materials based on Fe3 O4 nanoparticles with a SiO2 /PEG
shell and study of their effects on cancer cell lines. Int. J. Mol. Sci. 23(16), 9093 (2022)
115. Chen, Z., Song, S., Ma, J., Da Ling, S., Wang, Y.D., Kong, T.T., Xu, J.H.: Fabrication of
magnetic core/shell hydrogels via microfluidics for controlled drug delivery. Chem. Eng. Sci.
248(Part B), 117216 (2022)
116. Dong, D., Chen, R., Jia, J., Zhao, C., Chen, Z., Lu, Q., Sun, Y., Huang, W., Wang, C., Li, Y., He,
H.: Tailoring and application of a multi-responsive cellulose nanofibre-based 3D nanonetwork
wound dressing. Carbohydr. Polym. 305, 120542 (2023)
117. Li, H., Li, B., Lv, D., Li, W., Lu, Y., Luo, G.: Biomaterials releasing drug responsively to
promote wound healing via regulation of pathological microenvironment. Adv. Drug Deliv.
Rev. 114778 (2023)
118. Yang, X., Zhang, C.Q., Deng, D.W., Gu, Y.Q., Wang, H., Zhong, Q.F.: Multiple stimuli-
responsive MXene-based hydrogel as intelligent drug delivery carriers for deep chronic wound
healing. Small 18, 2104368 (2022)
119. Zeyni, V., Karimi, S., Namazi, H.: Surface PEGylation of ZIF-8 metal-organic framework
based on magnetic hydroxyapatite as a pH/magnetic targeting responsive system for anticancer
drug delivery. Microporous Mesoporous Mater. 354, 112544 (2023)
120. Cai, W., Zhang, W., Chen, Z.: Magnetic Fe3O4@ZIF-8 nanoparticles as a drug release vehicle:
pH-sensitive release of norfloxacin and its antibacterial activity. Colloids Surf. B 223, 113170
(2023)
121. Shrivastava, P., Vishwakarma, N., Gautam, L., Vyas, S.P.: Chapter 5—Magnetically respon-
sive polymeric gels and elastomeric system(s) for drug delivery. In: Smart Polymeric
Nano-Constructs in Drug Delivery, pp. 129–150. Academic Press (2023)
122. Nguyen, T.P.T., Ménager, C., Rieger, J., Coumes, F.: Rational design of stimuli-responsive
magnetic polymer hybrid (nano)materials. Polym. Int. (2023)
123. Mohanan, S., Guan, X., Liang, M., Karakoti, A., Vinu, A.: Stimuli-responsive Silica Silanol
conjugates: strategic nanoarchitectonics in targeted drug delivery. Small 2301113 (2023)
360 S. G. Reddy and H. C. A. Murthy
124. Fragal, E.H., Fragal, V.H., Silva, E.P., Paulino, A.T., da Silva Filho, E.C., Mauricio, M.R.,
Silva, R., Rubira, A.F., Muniz, E.C.: Magnetic-responsive polysaccharide hydrogels as smart
biomaterials: synthesis, properties, and biomedical applications. Carbohydr. Polym. 292,
119665 (2022)
125. Armenia, I., Ayllón, C.C., Herrero, B.T., Bussolari, F., Alfranca, G., Grazú, V., de la Fuente,
J.M.: Photonic and magnetic materials for on-demand local drug delivery. Adv. Drug Deliv.
Rev. 191, 114584 (2022)
126. Boztepe, C., Daskin, M., Erdogan, A.: Synthesis of magnetic responsive poly(NIPAAm-co-
VSA)/Fe3O4 IPN ferrogels and modeling their deswelling and heating behaviors under AMF
by using artificial neural networks. React. Funct. Polym. 173, 105219 (2022)
127. Shademani, A., Jackson, J.K., Thompson, C.J., Chiao, M.: Controlled and localized antibiotics
delivery using magnetic-responsive beads for synergistic treatment of orthopedic infection.
J. Biomed. Mater. Res. 110(5), 1036–1051 (2022)
128. Bag, N., Bardhan, S., Roy, S., Roy, J.: D Mondal nanoparticle-mediated stimulus-responsive
antibacterial therapy. Biomater. Sci. 11, 1994–2019 (2023)
129. Casillas-Popova, N., Bernad-Bernad, M.J., Gracia-Mora, J.: Modeling of adsorption and
release kinetics of methotrexate from thermo/magnetic responsive CoFe2O4–BaTiO3,
CoFe2O4–Bi4Ti3O12 and Fe3O4–BaTiO3 core-shell magnetoelectric nanoparticles func-
tionalized with PNIPAm. J. Drug Deliv. Sci. Technol. 68, 103121 (2022)
130. Zeng, N., He, L., Jiang, L., Shan, S., Su, H.: Synthesis of magnetic/pH dual responsive dextran
hydrogels as stimuli-sensitive drug carriers, Carbohydr. Res. 520, 108632 (2022)
Advanced Tissue Engineering with Novel
Engineered Biomaterials
Abstract Nowadays, the concept of regenerative medicine is known via matrix prop-
erties that can mimic natural tissue functions. In this field, the therapeutic successes in
implantation and tissue regeneration depend on the type of biomaterials applied in the
fabrication of polymer substrates or substitutes. Such materials lead to the design of
intelligent/smart substrates, the promotion of specific bioactive signals, the improve-
ment of cell-to-cell interaction, and control of the designed micro-environment func-
tions to respond to the cellular behavior during the process of tissue repair. These
scaffolds, alone or incorporated with bioactive drugs, cells, and biomolecules, can be
promising in the medical and pharmaceutical fields. Accordingly, this chapter aims
to introduce novel biomaterials applied in tissue engineering and focuses on their
main advance and limitations.
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 361
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_12
362 A. Izadyari Aghmiuni and A. Gholami
1 Introduction
Tissue engineering involves biological principles and approaches along with polymer
and materials sciences, and engineering techniques to design tissue substitutes for
mimicking the structural and physiological functions of native tissues and conse-
quently regenerating injured or diseased tissues [1]. However, this field is contin-
uously developing through the input of different scientific areas such as nanotech-
nology, cell therapy, phytochemistry, electronics, etc., which can lead to the precise
control of cell functions (proliferation, differentiation, migration). Although, nowa-
days, tissue engineering has evident growing impacts in therapeutic and research
fields, known as a novel and improved therapy; however, it is still struggling with the
issues of in-vivo transfer. In this field, novel methods like 3D and 4D bio-printing,
microstructures (organs on a chip), and/or a combination of several methods can
provide promising perspectives [2–5].
To fully understand the diagnostic therapeutic potential of this technique, it is
crucial to possess a rationally designed network based on biomaterials. On this
matter, there are research advances related to new biomaterials and their copoly-
mers or composites that can control the interactions of cell/biomaterial networks and
cell/cell and improve cell signaling pathways [6–8]. Hence, this chapter focuses on
reviewing novel engineered biomaterials, biomimetic materials, and some of the most
exciting studies regarding the use of these materials to apply in regenerative medicine
and tissue engineering, along with their technological approaches. In the following,
some of the new topics on tissue engineering have been reviewed including personal-
ized regenerative medicine, 3D and 4D bio-printing, and engineered microstructure
models for diagnostic or therapeutic strategies.
Recently, many studies have been made on the use of engineered biomaterials/
biomimetic materials and their interactions with biological environments. Indeed,
these materials can play a crucial role in cellular infiltration within an implanted
matrix at the local implant sites and then form new tissues on the biomaterial-based
matrix. In this matter, biomaterial itself can act as a mechanical substrate or pharma-
cological/therapeutic agent to improve biological signals and transfer these signals to
cells for tissue regeneration [9–12]. Generally, biomimetic materials can be applied
to increase tissue regeneration, however, having the engineered scaffold is essential
for supplying mechanical support, preventing scar formation, and successful regen-
eration of tissue. These biomaterials can provide cell adhesion factors, proliferation,
induction, differentiation, and migration and lead to the modification of cellular
responses during tissue regeneration, via the incorporation of survival factors and
creating covalently or non-covalently bonds with cells [13, 14].
Advanced Tissue Engineering with Novel Engineered Biomaterials 363
In this regard, the recognition of biomaterials and their functions for the improve-
ment of cellular adhesion sites, the spatial distribution of ligands, selection of suitable
cells and their spatial distribution can play a pivotal role in designing biomimetic
materials and bio-matrices [15–17]. The transformation of active materials from one
state to another state is one of the attractive features of biomaterials that occurs in
the presence of biological systems or response to some of the external stimuli like
temperature, chemical composition, light, etc., and leads to new noninvasive surgical
procedures [2, 18, 19]. Moreover, the incorporation of DNAs into biomaterials to
control cellular responses and stimulate in situ production of growth factors, as well
as the design of micro-fabricated bio-matrices by micro-fabrication techniques to
improve cellular adhesion behavior, delivery of oxygen and nutrients, etc., are other
applications of these materials [2, 19–23].
Based on the studies, the modification of biomaterial surface can also be a suitable
way to fabricate a biomimetic matrix, and the use of bioactive molecules, especially
ECM biomolecules can be a suitable option to achieve this aim. In this field, proteins
like heparin, fibronectin, elastin, etc., are considered as the protein covers on these
materials which can promote adhesion and growth of cells; and increase elastic
modulus of artificial ECMs, and improve osteoblastic mineralization and osteoin-
duction [24–26]. The use of synthetic peptide sequences and short-chain peptides are
other methods for surface modification of biomaterials that can be led to improved
adsorption to the biomaterial surface and increased cell access to all surfaces. Such
modified biomaterials can be used in the network of polymer matrices as potentially
artificial ECMs to guide the formation of new tissues [24, 27].
Although, the common peptides in this field are Arg-Gly-Asp (RGDs) derived
from fibronectin and laminin, however, other peptides like Tyr-Ile-Gly-Ser-Arg, Arg-
Glu-Asp-Val, and Ile-Lys-Val-Ala-Val have also developed for modifying engineered
matrices [24, 28, 29].
There are well-known techniques such as coupling with functional groups or
bi-functional crosslinking that are used for ensuring the covalent binding between
the peptide and the surfaces of the biomaterial and its immobilization and provide
flexible movement in biological environments [24] Other techniques in this matter
include the following [24, 30–33]:
1. Photochemical immobilization: when the polymer matrices lack functional
groups to form the coupling reactions.
2. Limiting non-specific adsorption of protein and the reactive chains.
3. Bulk modification of biomaterial: in this technique, cell-signaling peptide is
incorporated into the bio-matrix structure to exist the recognition sites in the
bulk of the biomaterials.
Some of the suitable techniques for modifying the surface of biomaterials or
biomimetic materials to mimic biological environments and achieve microenviron-
ments similar to native ECMs have been indicated in Fig. 1.
Generally, biomimetic means the study and assessment of naturally made models,
systems, processes, and elements, and then mimicking and copying their structures
and functions. Rose petals, lotus leaves, beehives, butterfly wings, spider silk, water
364 A. Izadyari Aghmiuni and A. Gholami
strider foot, etc., are examples in this field that possess unique micro/nanostructures
with hydrophobic nature. Common techniques for designing such structures include
chemical deposition, electro-deposition, and hydrothermal reaction which are used
for designing biomimetic matrices or biomaterial structures in various industries.
Some studies in this field have been listed in Table 1.
Based on the reports, the photo-functionalization method by ultraviolet radiation
is known as another method in the design of biomaterial/biomimetic matrices to treat
diseases and regenerate tissue. This technique provides the physicochemical modifi-
cations of the bio-matrix surface and leads to the improvement of its biological prop-
erties [46]. Notably, given that direct exposure to ultraviolet light, after implantation
of biomatrix, can be very harmful, hence photo-functionalization with this radiation
should carry out before implantation [47]. Such a process can increase cellular adhe-
sion and the adsorption of proteins, and help in-vitro osteogenic differentiation and
mineralization [48, 49]. In this field, Dini et al. indicated that the UV-mediated photo-
functionalization process can improve the adsorption of proteins in titanium-treated
discs [50]. The authors stated that photo-functionalization of the PEO coating leads to
the improvement of the biological activities of dental and bone implants by changing
the physicochemical properties of titanium (Ti). Assessment of the human gingival
fibroblast cells confirmed the non-cytotoxic effects of these discs. Hence, it seems
Advanced Tissue Engineering with Novel Engineered Biomaterials 365
Table 1 Some of the common methods for designing biomimetic matrices and biomaterial
structures
The aim Techniques Description Refs.
Development of Ti-based Electrochemical Creation of Zr–Nb [34]
alloys in dentistry technique alloys-based microstructures
for decreasing corrosion
behavior and without any
toxic effects
Electrochemical Improvement of binary Ti-Ag [35]
technique alloys-based surface
characterization and cell
responses
Spark plasma Creation of microstructures to [36]
sintering (SPS) improve mechanical
technique properties and decrease Ti-Mn
alloys-related cytotoxicity
Casting technique Improvement of mechanical [37]
property and deformation
behavior of Ti–Cr-based
alloys
Lotus-leaf-like surface Simple Superhydrophobic metal [38]
electrochemical microstructure surface
reaction obtained via the non-electric
chemical plating of the Cu
onto the steel sheet
Superelastic alloy Thermal process Ti–Zr-based biomedical alloy [39]
containing Mo–Sn to improve
superelastic properties via
regulating Mo and Sn contents
Oil–water separation, Electrodeposition The design of the Cu-coated [40]
improvement of the Cu superhydrophobic copper foil
corrosion resistance, stain in a zigzag manner
repellency, sound
absorption, and
mechanochemical
robustness
Oil–water separation, and Electrochemical The design of biomimetic [41]
the creation of technique graphene on the stainless steel
superhydrophobic and surfaces inspired by rose
superoleophilic surfaces petals, butterfly wings, nacre,
and honeycomb
The control of the corrosion Melting method Design of [42]
behavior of the alloy Ti-6Al-4V-xZr-based
microstructures and the
passivation of Ti-6Al-4V-xZr
alloy via increasing Zr content
(continued)
366 A. Izadyari Aghmiuni and A. Gholami
Table 1 (continued)
The aim Techniques Description Refs.
Superhydrophobic films Electrodeposition The design of [43]
process corrosion-resistant
superhydrophobic surfaces
along with excellent chemical
stability and self-cleaning
effects
Superhydrophobic structures Electrochemical The design of graphene [44]
co-deposition oxide-myristic acid-based
superhydrophobic structure
(nickel-reduced) to increase
corrosion resistance
Sheet-like superhydrophobic Etching-heat The design of [45]
surfaces combination superhydrophobic sheets
treatment based on Cu to decrease
corrosion
that such a bio-functional coating can act as a promising approach for biomedical
applications.
Nagay et al. demonstrated that non-thermal plasma can improve surface and
physicochemical properties of the soft liner surfaces based on the hexamethyl
disiloxane, and lead to the inhabitation of C. albicans biofilm [51]. Based on their
reports, these treated surfaces can provide candida-related stomatitis protection, and
antimicrobial effects (decrease in fungal colonization) and act as a suitable candidate
for medical applications. Cordeiro et al. also indicated that Ti-Nb-Zr-Ta alloys treated
with plasma electrolytic oxidation (PEO) can create porous surfaces and crystalline
structure along with an increase in electrochemical stability, hardness, roughness,
and surface free energy [52]. Finally, the authors stated that such alloys can be suit-
able alternatives for fabricating dental implants, due to their nontoxic and elastic
modulus similar to the bone.
In the following, Prosdent et al. indicated that some specimens of tissue condi-
tioners such as visco-gels possess high water sorption and solubility [53]. Moreover,
some other tissue conditioners such as dentu-soft and coe-soft (as novel biomimetic
materials) can provide suitable functions to fabricate denture relining. Furthermore,
the effects of the coating through the biomimetic technique on titanium substrates
were assessed by Coelho et al. [54]. This technique consists of the immersion of tita-
nium substrate into the solution of simulated body fluid with conditions similar to the
blood plasma, to form the slow and well-organized nucleation and promote apatite to
be deposited. Based on their results, the proposed biomimetic coating in this research
can be effective to achieve hydroxyapatite and carbonated hydroxyapatite, as well
as, enhance the osseointegration process of the implanted bones. Moreover, titanium
treated with the method possesses a higher resistance to corrosion than pure titanium.
Hence, they stated that titanium substrates coated with biomimetic materials can be
a suitable candidate for bone implantation applications.
Advanced Tissue Engineering with Novel Engineered Biomaterials 367
Given that the macroscopic structure of biomimetic materials has led to the limi-
tation in the mechanical functions of the product biomaterial or matrix, the use of
hierarchical architecture consisting of a microscopic cellular structure can remove
the mentioned limitation. Several studies in this field illustrate that hierarchical struc-
tures of bone improve mechanical properties. Barth et al. stated that X-ray irradiation
on the hierarchical structures of bone can progressively degrade the mechanical func-
tions of human cortical bone (strength, fracture resistance, ductility, and toughness)
[55, 56]. This degradation increases with the promotion of X-ray irradiation. More-
over, the characterization of hierarchical structures of bone in the conditions of aging,
disease, and treatment was assessed by Milovanovic et al. for understanding the archi-
tectural modifications and mechanical integrity [57]. Based on the results, young and
healthy bones possess the highest resistance to mechanical loading. These bones also
indicated better mineralization along with preservation of osteocyte-lacunar proper-
ties, but aging bones and osteoporosis lead to alterations in bone material proper-
ties like increased osteocyte apoptosis and decreased mechanical competence. The
authors stated that structural or compositional change in the bone material leads to
the maintenance or decay of bones in disease and health conditions.
Furthermore, deformation and fracture in bones were assessed by Zimmermann
et al., at the strain rate of 10−5 –10−1 (1/s) [58]. The results indicated that bone
toughness decreased with increasing strain rate; so that higher strain rate led to
lower intrinsic bone matrix toughness and consequently fracture induced by a loss in
toughness in the matrix. In continuing, Peng et al. designed an ultra-light biomimetic
material via the 3D printing technique [59]. Based on their reports, the designed
biomimetic possessed an ultrahigh elasticity and stability, at the compressive strain
of 95%, along with remarkable stiffness and resilience.
Chitosan-based flexible robust structures are considered another promising candi-
date in bio-friendly catalyst applications. Based on the reports of Tseng et al., TiO2 -
attached biomimetic chitosan films can be effective in the photo-catalytic reduc-
tion of carbon dioxide [60]. Notably, the sol–gel process along with heat treatment
was used for preparing these films. Moreover, the use of the nano-casting method
provided the duplication of structures of natural leaves on these films. It was also
found that hydroxyl and amine groups can fasten the TiO2 nanoparticles on the
chitosan films. Adding glutaraldehyde leads to also the improvement of the film
denseness, the adsorption of carbon dioxide, stability, and hydrophobicity of the
surfaces. The authors stated such films as eco-friendly photocatalysts can be suitable
candidates to produce pure chemical fuels via decreasing CO2 .
Generally, modified biomimetic surfaces can act as transport barriers for
decreasing water/molecules/ions loss inside the cell. Likewise, the uptake of liquids/
molecules can also decrease via these surfaces [61]. Selenium is another material that
led to the inactivation of carcinogens and inhibition of cell damage from free radicals.
Hence the use of this material in the modified biomimetic structures such as micro-
gels and matrices, etc. not only can control and remove free radicals but also prevents
dysfunctions of the cellular metabolism caused by conventional selenium-containing
compounds [62].
368 A. Izadyari Aghmiuni and A. Gholami
Given that the uncontrolled differentiation of cells can increase the risk of tumori-
genicity, the use of therapeutic strategies for the complete and irreversible differenti-
ation of these cells can play a crucial role in targeted differentiation. These strategies
consist of the use of bio-materials and/or herbal bioactive materials for designing
polymer matrices and providing suitable physicochemical/mechanical properties to
differentiate stem cells and regenerate tissue [90, 91]. Such biomaterials play a key
role in providing the microenvironments for the differentiation of cells and controlling
cellular functions and interactions by creating an ideal matrix.
The studies of Izadyari Aghmiuni et al. indicate that herbal bioactive materials
possess high therapeutic and regenerative effects [92, 93]. However, some of their
forms such as mucilage, the extract/essential oil, gel, etc. cannot be applied to
damaged tissues directly. This research team demonstrated that the use of these
biomaterials in the structure of engineered matrices can turn the matrix into a smart
biological substrate [92]. They stated that some of the herbal materials such as
quince seed mucilage are known as polysaccharides that are composed of xylose
and glucuronic acid. These polysaccharides can swell in water owing to the exis-
tence of hydrophilic groups, unlike chitosan which is suspended in an acidic solvent.
Based on their reports, quince seed mucilage-based substrates can provide a better
porous network than chitosan-based matrices. These substrates can also support the
proliferation of fibroblast cells and provide higher water absorption capacity for
applications of skin tissue engineering. Moreover, the authors indicated that the
combination of this mucilage with PEG increased the transduction of mechanical
signals and improved the biological signals for inducting stem cell differentiation
into dermal tissue.
According to the reports of Tahmasebi et al., nano-fibrous scaffolds of poly(3-
hydroxybutyrate-co-3-hydroxy valerate blended with aloe vera gel can be also effec-
tive in bone tissue engineering applications [94]. Indeed, an increase in the biocom-
patibility of these scaffolds is related to aloe vera contents. Likewise, they indicated
that these hybrid scaffolds possess better alkaline phosphatase activities and mineral-
ization, and high expression of bone-related genes/proteins compared with aloe vera-
free nano-fibrous. It is also known that aloe vera gel possesses osteoinductive poten-
tial and can be used in the network of bio-implants. Moreover, Suganya et al. indi-
cated that electrospun mesh of aloe vera can be an effective transdermal therapeutic
strategy. Accordingly, the fibers of polycaprolactone containing aloe vera powder
(10 wt %) fabricated and stated these meshes can provide better hydrophilic proper-
ties, more suitable mechanical properties (tensile strength:6.28 MPa, elastic modulus
similar to dermal tissue) than polycaprolactone-collagen fibers. These electrospun
fibers also lead to more desirable cell proliferation, better secretion of collagen, and
a high level of expression of F-actin [95].
Another study is about an aloe vera-based scaffold that possesses wide appli-
cations in biomedicine. The popularity of this herbal polysaccharide is inducted
by its bioactive components such as anthraquinones, anthrones, vitamins, etc., and
Advanced Tissue Engineering with Novel Engineered Biomaterials 375
The studies of Yu et al. also show that naringin in 50μg/ml concentration activates
the Notch signaling pathways and stimulates osteogenic differentiation. While higher
concentrations than 100μg/ml suppress the rate of cell proliferation [104]. Based
on the reports of Zhang, et al., naringin can also induce osteogenic activities and
differentiation of canine bone marrow stromal cells at 10–6 mol/l concentrations
[105]. Based on their reports, this range of concentration can lead to an increase in
cellular proliferation and calcium nodules. In this regard, Beom Su Kim et al. also
showed that the extract of brown algae Laminaria japonica (fucoidan) in 0.1–10μg/
ml concentrations led to JNK- and ERK-dependent BMP2 –Smad 1/5/8 signaling
and induction of osteoblast differentiation [106].
Fei Li et al. also stated that some of the phenylethanoid glycosides such as Echi-
nacoside isolated from Cistanches Herba stem (as a novel biomaterial) can increase
cell bioactivities and mineralization of osteoblastic [107]. To this end, the level of
osteoprotegerin, osteocalcin, and collagen secretion on the cell culture was assessed.
The results indicated that concentrations of 0.01–10 nmol/l can increase cell prolif-
eration, osteocalcin levels, and collagen I content, as well as, lead to the promotion of
the mineralization of osteoblastic. They reported that such glycosides possess a stim-
ulatory effect on the formation of osteoblastic bone and can potentially be effective
against osteoporosis.
Moreover, MaríaSatué et al. reported that flavonoids not only can stimulate stem
cell differentiation into osteoblast but inhibit osteoclastogenesis in RAW 264.7 cells,
as well [108]. Based on the results, doses greater than 100 μM of diosmetin, galangin,
and chrysin flavonoids as well as 500 μM doses of taxifolin flavonoid possess toxic
effects on cells. While, quercitrin with safe doses of 200 and 500 μM, and taxifolin in
safe doses of 100 and 200 μM can induce osteocalcin mRNAs and bone sialoprotein
expression and lead to higher osteocalcin levels.
The study of Kim et al. indicates Herba Siegesbeckia-extracted kirenol can
promote osteoblast differentiation by activating the BMPs and signaling pathway in
cells [109]. This natural diterpenoid can also promote mineralization, ALP activities,
osteopontin, collagen content, and expression of OPG/RANKL. Choi et al. also stated
that Magnolia Officinalis-isolated Honokiol can lead to the stimulation of osteoblast
MC3T3-E1 cell functions and reduce/inhibit the production of bone-resorbing medi-
ators [110]. They reported that such phenolic compounds can be effective in natural
therapies for osteoporosis. Likewise, Hui-HuiXiao et al. indicated that Vanillic acid
derived by Sambucus williamsii Hance can increase estrogen-like activities of cells
[111]. The results of this study illustrate that these phenolic acids can stimulate
cellular proliferation and ALP activity and promotes Runx2, osteocalcin, and the
ratio of OPG-RANKL2 mRNA expression.
Phytoestrogens and puerarin derived from Pueraria Mirifica are considered the
other herbal biomaterials that can increase the growth of cells and the expression of
osteoblastic differentiation [112]. On this matter, Tiyasatkulkovit et al. demonstrated
that puerarin can improve the mRNA expression of ALP, decrease the expression
level of RANKL, and induce bone gain via increasing osteoblast differentiation in
Tissue engineering develops alternative approaches for decreasing the risks of disease
transfer of allografts and the lack of availability of auto-grafts [113, 114]. The strategy
of this method includes the regeneration of the guided tissues via modeling human
diseases, pharmaceutical screening tools, designed nano/micro-tissues, etc. In this
field, natural polymers as scaffolding biomaterials play an important role; so that
they can provide organ physiology-like structures and lead to the imitation of tissue
functions.
Generally, tissue engineering creates a personalized medicine concept to a new
level that can be used for tuning the suitable approaches to meeting the patient’s
requirement. Integration of technological advances like micro/nano-fabrications and
technologies of cell therapy can be also effective in this regard. Personalized regen-
erative medicine, 3D and 4D bio-printing, and engineered microstructure models
are considered as new topics in diagnostic or therapeutic strategies. On this matter,
personalized regenerative medicine not only benefits patients due to the following
treatment but also provides lower healthcare and social costs. Moreover, the use of
3D or 4D printing technology along with an imaging approach can play an impact
on assembling and directing the 3D/4D systems for the design of clinical sizes, and
mechanical and structural integrity of tissue-mimicking structures (Fig. 2).
Some of the biomaterials used in bio-printing techniques along with their
applications include the following [115]:
12.4.1 Gold, titanium, platinum, steel, etc.: for applications of orthopedic
implants, screws, pin
12.4.2 Ceramics and carbon compounds: For applications of bioactive orthopedic
implant
12.4.3 Calcium phosphate salt, titanium oxide, aluminum oxide, and, glass: For
applications of dental implants and artificial hearing aids
12.4.4 Poly (methyl methacrylate), polycarbonates, polyurethanes,
poly(caprolactone): for applications of tissue-engineered scaffolds, drug delivery
systems, and Prostheses
12.4.5 Carbon fibers, dental cement composites, types of bone cement, polyethy-
lene with ultra-high molecular weight: for applications of dental fillings, ortho-
pedic, rubber catheters, gloves, and implants
378 A. Izadyari Aghmiuni and A. Gholami
4 Transforming 3D to 4D Printing
(continued)
379
Table 3 (continued)
380
(continued)
Advanced Tissue Engineering with Novel Engineered Biomaterials
381
Table 3 (continued)
382
(continued)
A. Izadyari Aghmiuni and A. Gholami
Table 3 (continued)
Method Description Figures Refs.
Stereolithography The device utilized a moving [125]
helium-cadmium laser for producing an
ultraviolet light spot on top of the polymer
vat and an optical scanning system is applied
to manage the movement of this spot
Table 4 (continued)
Type of 3D Materials The type of cell or Fabrication process/ Refs.
printing test model Function
Extrusion Polycaprolactone-printed Hormones-derived PCL thermoplastic [139]
3D constructs laden with induced pluripotent pellets applied to
estrogen or progesterone stem cells (estrogen load hormones,
and/or progesterone) hormones-coated
pellets extruded as
filaments/For
obstetric and
gynecologic
applications such as
intrauterine devices
Hot melt Oleo-gum-resins disks Staphylococcus 3D-printed disks [140]
extrusion treated by nano-oxides of aureus, into model
TiO2 , P25, Cu2 O, and pseudomonas geometries and the
MoO3 aeruginosa, disk-diffusion
escherichia coli, and methodology was
candida albicans applied to assess
antimicrobial and
antifungal activities
of studied materials
against clinical
isolates/To study the
potential of
3D-printed
bio-actives
Inkjet Phosphoric Rabbit bone marrow Scaffold printed by [141]
acid-polyvinyl alcohol stromal cells ZPrinter at 0.1 mm
scaffold with a powder thickness/
hydroxyapatite/ Bone tissue
β-tricalcium phosphate engineering and
medical implants
UV Methacrylated hyaluronic Human bone Methacrylated [142]
cross-linked acid hydrogel marrow-derived hyaluronic acid
mesenchymal dissolved in an alpha
stromal cells minimum essential
medium
concentration,
scaffold bio-printed
by the dispensing
system, scaffold UV
cross-linked at
1800 mJ/Bone
regenerative
medicine
(continued)
386 A. Izadyari Aghmiuni and A. Gholami
Table 4 (continued)
Type of 3D Materials The type of cell or Fabrication process/ Refs.
printing test model Function
Fused Biphasic calcium Human Fabrication of the [143]
deposition phosphate scaffolds mesenchymal stem 3D slurry foam with
modeling reinforced with zirconia cells blending agents, the
removal of blending
materials at high
temperature,
extruding at 600kPa
pressure and 100
mm/min printing
speed/Bone tissue
engineering
Polyjet Rigid acrylic resin Preoperative Patient-specific [144]
planning in intrahepatic vessel
anatomical resection model/To
of hepatocellular understand the
carcinoma positional
relationships
between tumors and
vessel, as well as
operator and
assistant during
operation
Inkjet-based Calcium Sulfate Scaffolds Osteoblast-like The scaffolds treated [145]
3D printing sarcoma cells with heat,
heat-treated
scaffolds at 300 °C
possessed almost
adequate strength
but severe toxicity,
by contrast, the
modified scaffold at
500–1000°C was
non-toxic but
possessed
insufficient
mechanical strength/
hard tissue
engineering
inorganic particles, etc. can act as suitable supports to improve mechanical properties,
increase alkaline enzymatic activities, and form calcium in osteoblast cells [146, 153,
154]. Moreover, the formation of composite scaffolds can increase rates of shape
recovery potential and be applied for regenerating small bones.
References
1. Gomes, M.E., Rodrigues, M.T., Domingues, R.M.A., et al.: Tissue engineering and regen-
erative medicine: new trends and directions—a year in review. Tissue Eng. Part B Rev. 23,
211–224 (2017)
2. Izadyari Aghmiuni, A., Heidari Keshel, S.: Eye-on-a-chip. In: Principles of Human Organs-
on-Chips, pp. 315–370 (2023)
3. Chen, L.-J., Raut, B., Kaji, H.: On-Chip Disease Models of the Human Retina, 2nd edn.
Elsevier Inc. Epub ahead of print (2019). https://doi.org/10.1016/b978-0-12-813671-3.000
12-8
4. Izadyari Aghmiuni, A., Heidari Keshel, S., Sefat, F., et al.: Fabrication of 3D hybrid scaffold
by combination technique of electrospinning-like and freeze-drying to create mechanotrans-
duction signals and mimic extracellular matrix function of skin. Mater. Sci. Eng. C 120,
111752 (2021)
5. Ashammakhi, N., Ahadian, S., Zengjie, F., et al.: Advances and future perspectives in 4D
bioprinting. Biotechnol. J. 13, 1–12 (2018)
6. Darzi, S., Paul, K., Leitan, S., et al.: Immunobiology and application of aloe vera-based
scaffolds in tissue engineering. Int. J. Mol. Sci. 22, 1–19 (2021)
388 A. Izadyari Aghmiuni and A. Gholami
7. Wang, S., Meng, S., Zhou, X., et al.: PH-responsive and mucoadhesive nanoparticles for
enhanced oral insulin delivery: the effect of hyaluronic acid with different molecular weights.
Pharmaceutics 15, 820 (2023)
8. Aghmiuni, A.I., Baei, M.S., Keshel, S.H., et al.: Design of novel 3D-scaffold as a potential
material to induct epidermal-dermal keratinocytes of human-adipose-derived stem cells and
promote fibroblast cells proliferation for skin regeneration. Fibers Polym. 21, 33–44 (2020)
9. Brovold, M., Almeida, J.I., Pla-Palacín, I., et al.: Naturally-derived biomaterials for tissue
engineering applications (2020). Epub ahead of print. https://doi.org/10.1007/978-981-13-
0947-2_23
10. Ghorbani, M., Ramezani, S., Rashidi, M.: Fabrication of honey-loaded ethylcellulose/gum
tragacanth nanofibers as an effective antibacterial wound dressing. Colloids Surfaces A
Physicochem. Eng. Asp. 621, 126615 (2021)
11. Zhong, S.P., Zhang, Y.Z., Lim, C.T.: Tissue scaffolds for skin wound healing and dermal
reconstruction. Wiley Interdiscip. Rev.: Nanomedicine Nanobiotechnology 2, 510–525 (2010)
12. Li, Q., Niu, Y., Xing, P., et al.: Bioactive polysaccharides from natural resources including
Chinese medicinal herbs on tissue repair. Chinese Med. (United Kingdom) 13, 1–11 (2018)
13. Wang, X., Ali, M.S., Lacerda, C.M.R.: A three-dimensional collagen-elastin scaffold for
heart valve tissue engineering. Bioengineering 5. Epub ahead of print (2018). https://doi.org/
10.3390/bioengineering5030069
14. Theocharis, A.D., Skandalis, S.S., Gialeli, C., et al.: Extracellular matrix structure. Adv. Drug
Deliv. Rev. 1–76 (2015)
15. Ghiasi, M., Kalhor, N., Tabatabaei Qomi, R., et al.: The effects of synthetic and natural
scaffolds on viability and proliferation of adipose-derived stem cells. Front Life Sci. 9, 32–43
(2016)
16. Zahedi, E., Esmaeili, A., Eslahi, N., et al.: Fabrication and Characterization of Core-Shell
Electrospun Fibrous Mats Containing Medicinal Herbs for Wound Healing and Skin Tissue
Engineering. Mar. Drugs 17, 27 (2019)
17. Stratton, S., Shelke, N.B., Hoshino, K., et al.: Bioactive polymeric scaffolds for tissue
engineering. Bioact Mater 1, 93–108 (2016)
18. Pires, P.C., Mascarenhas-melo F, Pedrosa K, et al. Polymer-based biomaterials for phara-
maceutical and biomedical applications : a focus on topical drug administration. Eur. Polym.
J. 187. Epub ahead of print 2023. https://doi.org/10.1016/j.eurpolymj.2023.111868
19. Huang, G., Li, F., Zhao, X., et al.: Functional and biomimetic materials for engineering of the
three-dimensional cell microenvironment. Chem. Rev. 117, 12764–12850 (2017)
20. Howard, D., Buttery, L.D., Shakesheff, K.M., et al.: Tissue engineering: strategies, stem cells
and scaffolds. J. Anat. 213, 66–72 (2008)
21. Ezhilarasu, H., Ramalingam, R., Dhand, C., et al.: Biocompatible aloe vera and tetracycline
hydrochloride loaded hybrid nanofibrous scaffolds for skin tissue engineering. Int. J. Mol.
Sci. 20, 5174 (2019)
22. Thi Hiep, N., Chan Khon, H., Dai Hai, N., et al.: Biocompatibility of PCL/PLGA-BCP porous
scaffold for bone tissue engineering applications. J. Biomater. Sci. Polym. Ed. 28, 864–878
(2017)
23. Loh, Q.L., Choong, C.: Three-dimensional scaffolds for tissue engineering applications: role
of porosity and pore size. Tissue Eng. Part B Rev. 19, 485–502 (2013)
24. Shin, H., Jo, S., Mikos, A.G.: Biomimetic materials for tissue engineering. Biomaterials 24,
4353–4364 (2003)
25. Yan, S., Zhang, Q., Wang, J., et al.: Silk fibroin/chondroitin sulfate/hyaluronic acid ternary
scaffolds for dermal tissue reconstruction. Acta Biomater. 9, 6771–6782 (2013)
26. Sengupta, P., Prasad, B.L.V.: Surface modification of polymeric scaffolds for tissue engi-
neering applications. Regen. Eng. Transl. Med. 4, 75–91 (2018)
27. Daamen, W.F., Veerkamp, J.H., Van Hest, J.C.M., et al.: Elastin as a biomaterial for tissue
engineering. Biomaterials 28, 4378–4398 (2007)
28. Hern, D.L., Hubbell, J.A.: Incorporation of adhesion peptides into nonadhesive hydrogels
useful for tissue resurfacing. J. Biomed. Mater. Res. 39, 266–276 (1998)
Advanced Tissue Engineering with Novel Engineered Biomaterials 389
29. Tang X, Thankappan, S.K., Lee P, et al. Polymeric biomaterials in tissue engineering and
regenerative medicine. In: Natural and Synthetic Biomedical Polymers. Elsevier, pp. 351–371
(2014)
30. Zhang, J., Sheng, X., Jiang, L.: The dewetting properties of lotus leaves. Langmuir 25, 1371–
1376 (2009)
31. Bixler, G.D., Bhushan, B.: Bioinspired rice leaf and butterfly wing surface structures
combining shark skin and lotus effects. Soft Matter 8, 11271–11284 (2012)
32. Panchanathan, D., Rajappan, A., Varanasi, K.K., et al.: Plastron regeneration on submerged
superhydrophobic surfaces using in situ gas generation by chemical reaction. ACS Appl.
Mater. Interfaces 10, 33684–33692 (2018)
33. Feng, L., Zhang, Y., Xi, J., et al.: Petal effect: a superhydrophobic state with high adhesive
force. Langmuir 24, 4114–4119 (2008)
34. Zhou, F.Y., Wang, B.L., Qiu, K.J., et al.: Microstructure, corrosion behavior and cytotoxicity
of Zr-Nb alloys for biomedical application. Mater. Sci. Eng. C 32, 851–857 (2012)
35. Zhang, B.B., Qiu, K.J., Wang, B.L., et al.: Surface characterization and cell response of binary
Ti-Ag alloys with CP Ti as material control. J. Mater. Sci. Technol. 28, 779–784 (2012)
36. Zhang, F., Weidmann, A., Nebe, B.J., et al. Preparation of TiMn alloy by mechanical alloying
and spark plasma sintering for biomedical applications. J. Phys. Conf. Ser. 144. Epub ahead
of print 2009. https://doi.org/10.1088/1742-6596/144/1/012007
37. Ho, W.F., Chiang, T.Y., Wu, S.C., et al.: Mechanical properties and deformation behavior of
cast binary Ti-Cr alloys. J. Alloys Compd. 468, 533–538 (2009)
38. Han, J.T., Jang, Y., Lee, D.Y., et al.: Fabrication of a bionic superhydrophobic metal surface
by sulfur-induced morphological development. J. Mater. Chem. 15, 3089–3092 (2005)
39. Ijaz, M.F., Kim, H.Y., Hosoda, H., et al.: Superelastic properties of biomedical (Ti-Zr)-Mo-Sn
alloys. Mater. Sci. Eng. C 48, 11–20 (2015)
40. Huang, Z., Quan, Y., Mao, J., et al.: Multifunctional superhydrophobic composite materials
with remarkable mechanochemical, r.obustness., stain, r.epellency., oil-water separation and
sound-absorption properties. Chem. Eng. J. 358, 1610–1619 (2019)
41. Tang, W., Sun, D., Liu, S., et al.: One step electrochemical fabricating of the biomimetic
graphene skins with superhydrophobicity and superoleophilicity for highly efficient oil-water
separation. Sep. Purif. Technol. 236, 116293 (2020)
42. Xia, C., Zhang, Z., Feng, Z., et al.: Effect of zirconium content on the microstructure and
corrosion behavior of Ti-6Al-4V-xZr alloys. Corros. Sci. 112, 687–695 (2016)
43. Fan, Y., He, Y., Luo, P., et al.: A facile electrodeposition process to fabricate corrosion-resistant
superhydrophobic surface on carbon steel. Appl. Surf. Sci. 368, 435–442 (2016)
44. Jena, G., Thinaharan, C., George, R.P., et al.: Robust nickel-reduced graphene oxide-myristic
acid superhydrophobic coating on carbon steel using electrochemical codeposition and its
corrosion resistance. Surf. Coatings Technol. 397, 125942 (2020)
45. Zeng, Y., Qin, Z., Hua, Q., et al.: Sheet-like superhydrophobic surfaces fabricated on copper
as a barrier to corrosion in a simulated marine system. Surf. Coatings Technol. 362, 62–71
(2019)
46. Ogawa, T.: Ultraviolet photofunctionalization of titanium implants. Oral Craniofacial Tissue
Eng. 2
47. Cortat, B., Garcia, C.C.M., Quinet, A., et al.: The relative roles of DNA damage induced by
UVA irradiation in human cells. Photochem. Photobiol. Sci. 12, 1483–1495 (2013)
48. Aita, H., Hori, N., Takeuchi, M., et al.: The effect of ultraviolet functionalization of titanium
on integration with bone. Biomaterials 30, 1015–1025 (2009)
49. Att, W., Hori, N., Iwasa, F., et al.: The effect of UV-photofunctionalization on the time-related
bioactivity of titanium and chromium–cobalt alloys. Biomaterials 30, 4268–4276 (2009)
50. Dini, C., Nagay, B.E., Cordeiro, J.M., et al.: UV-photofunctionalization of a biomimetic
coating for dental implants application. Mater. Sci. Eng. C 110, 110657 (2020)
51. Nagay, B.E., Bitencourt, S.B., Commar, B.C., et al.: Antimicrobial and protective effects of
non-thermal plasma treatments on the performance of a resinous liner. Arch. Oral Biol. 117,
104822 (2020)
390 A. Izadyari Aghmiuni and A. Gholami
52. Cordeiro, J.M., Nagay, B.E., Ribeiro, A.L.R., et al.: Functionalization of an experimental
Ti-Nb-Zr-Ta alloy with a biomimetic coating produced by plasma electrolytic oxidation. J.
Alloys Compd. 770, 1038–1048 (2019)
53. Maciel, J.G., Sugio, C.Y.C., de, C.ampos., C.G., et al.: Determining acceptable limits for water
sorption and solubility of interim denture resilient liners. J. Prosthet. Dent. 121, 311–316
(2019)
54. Coelho, M.FC., De Sousa, L.L., Ferreira, C.C., et al.: Biomimetic coating on titanium: evalu-
ation of bioactivity and corrosion. Mater. Res. Express 6. Epub ahead of print (2019). https://
doi.org/10.1088/2053-1591/ab67f1
55. Barth, H.D., Zimmermann, E.A., Schaible, E., et al.: Characterization of the effects of x-ray
irradiation on the hierarchical structure and mechanical properties of human cortical bone.
Biomaterials 32, 8892–8904 (2011)
56. Barth, H.D., Launey, M.E., MacDowell, A.A., et al.: On the effect of X-ray irradiation on the
deformation and fracture behavior of human cortical bone. Bone 46, 1475–1485 (2010)
57. Milovanovic, P., Zimmermann, E.A., Riedel, C., et al.: Multi-level characterization of human
femoral cortices and their underlying osteocyte network reveal trends in quality of young,
aged, osteoporotic and antiresorptive-treated bone. Biomaterials 45, 46–55 (2015)
58. Zimmermann, E.A., Gludovatz, B., Schaible, E., et al.: Fracture resistance of human cortical
bone across multiple length-scales at physiological strain rates. Biomaterials 35, 5472–5481
(2014)
59. Peng, M., Wen, Z., Xie, L., et al.: 3D printing of ultralight biomimetic hierarchical graphene
materials with exceptional stiffness and resilience. Adv. Mater. 31, 1902930 (2019)
60. Tseng, I.-H., Liu, Z.-C., Chang, P.-Y.: Bio-friendly titania-grafted chitosan film with
biomimetic surface structure for photocatalytic application. Carbohydr. Polym. 230, 115584
(2020)
61. Barthlott, W., Mail, M., Bhushan, B., et al.: Plant surfaces: structures and functions for
biomimetic innovations. Nano-Micro Lett. 9, 1–40 (2017)
62. Xu, Q., He, C., Xiao, C., et al.: Reactive oxygen species (ROS) responsive polymers for
biomedical applications. Macromol. Biosci. 16, 635–646 (2016)
63. Xia, J., Li, T., Lu, C., et al.: Selenium-containing polymers: perspectives toward diverse
applications in both adaptive and biomedical materials. Macromolecules 51, 7435–7455
(2018)
64. Tian, J., Liu, Q., Asiri, A.M., et al.: Ultrathin graphitic carbon nitride nanosheets: a novel
peroxidase mimetic, Fe doping-mediated catalytic performance enhancement and application
to rapid, highly sensitive optical detection of glucose. Nanoscale 5, 11604–11609 (2013)
65. Cao, X.-N., Lian, S., Tong, Y., et al.: Fluorescent Se-modified carbon nitride nanosheets as
biomimetic catalases for free-radical scavenging. Chem. Commun. 56, 916–919 (2020)
66. Zhao, X., Chen, Z., Liu, Y., et al.: Silk fibroin microparticles with hollow mesoporous silica
nanocarriers encapsulation for abdominal wall repair. Adv. Healthc. Mater. 7, 1801005 (2018)
67. Zhang, W., Li, Y., Jiang, D., et al.: Promotion of hernia repair with high-strength, flexible,
and bioresorbable silk fibroin mesh in a large abdominal hernia model. ACS Biomater. Sci.
Eng. 4, 2067–2080 (2017)
68. Skrobot, J., Zair, L., Ostrowski, M., et al.: New injectable elastomeric biomaterials for hernia
repair and their biocompatibility. Biomaterials 75, 182–192 (2016)
69. Song, Z., Peng, Z., Liu, Z., et al.: Reconstruction of abdominal wall musculofascial defects
with small intestinal submucosa scaffolds seeded with tenocytes in rats. Tissue Eng. Part A
19, 1543–1553 (2013)
70. Izadyari Aghmiuni, A., Heidari Keshel, S., Sefat, F., et al.: Quince seed mucilage-based scaf-
fold as a smart biological substrate to mimic mechanobiological behavior of skin and promote
fibroblasts proliferation and h-ASCs differentiation into keratinocytes. Int. J. Biol. Macromol..
Epub ahead of print October (2019). https://doi.org/10.1016/j.ijbiomac.2019.10.008
71. Da, L., Gong, M., Chen, A., et al.: Composite elastomeric polyurethane scaffolds incorporating
small intestinal submucosa for soft tissue engineering. Acta Biomater. 59, 45–57 (2017)
Advanced Tissue Engineering with Novel Engineered Biomaterials 391
72. Jiang, W., Zhang, J., Lv, X., et al.: Use of small intestinal submucosal and acellular dermal
matrix grafts in giant omphaloceles in neonates and a rabbit abdominal wall defect model. J.
Pediatr. Surg. 51, 368–373 (2016)
73. Cao, G., Wang, C., Fan, Y., et al.: Biomimetic SIS-based biocomposites with improved
biodegradability, antibacterial activity and angiogenesis for abdominal wall repair. Mater.
Sci. Eng. C 109, 110538 (2020)
74. Kim, T.G., Shin, H., Lim, D.W.: Biomimetic scaffolds for tissue engineering. Adv. Funct.
Mater. 22, 2446–2468 (2012)
75. Kharaziha, M., Fathi, M.H., Edris, H.: Development of novel aligned nanofibrous composite
membranes for guided bone regeneration. J. Mech. Behav. Biomed. Mater. 24, 9–20 (2013)
76. Naghieh, S., Foroozmehr, E., Badrossamay, M., et al.: Combinational processing of 3D
printing and electrospinning of hierarchical poly (lactic acid)/gelatin-forsterite scaffolds as a
biocomposite: Mechanical and biological assessment. Mater. Des. 133, 128–135 (2017)
77. Kharaziha, M., Nikkhah, M.: Spatial patterning of stem cells to engineer microvascular
networks. Microscale Technol. Cell Eng. 143–166 (2016)
78. Li, S., Abdel-Wahab, A., Silberschmidt, V.V.: Analysis of fracture processes in cortical bone
tissue. Eng. Fract. Mech. 110, 448–458 (2013)
79. Hardy, J.G., Ghezzi, C.E., Saballos, R.J., et al.: Supracolloidal assemblies as sacrificial
templates for porous silk-based biomaterials. Int. J. Mol. Sci. 16, 20511–20522 (2015)
80. Liu, Y., Kim, J.H., Young, D., et al.: Novel template-casting technique for fabricating β-
tricalcium phosphate scaffolds with high interconnectivity and mechanical strength and
in vitro cell responses. J. Biomed. Mater. Res. Part A 92, 997–1006 (2010)
81. Luo, H., Zhang, Y., Li, G., et al.: Sacrificial template method for the synthesis of three-
dimensional nanofibrous 58S bioglass scaffold and its in vitro bioactivity and cell responses.
J. Biomater. Appl. 32, 265–275 (2017)
82. Cadman, J., Zhou, S., Chen, Y., et al. Characterization of cuttlebone for a biomimetic design
of cellular structures. In: Acta Mechanica Sinica/Lixue Xuebao, pp. 27–35 (2010)
83. Saravanan, S., Selvamurugan, N.: Bioactive mesoporous wollastonite particles for bone tissue
engineering. J. Tissue Eng. 7. Epub ahead of print (2016). https://doi.org/10.1177/204173141
6680319
84. Eilbagi, M., Emadi, R., Raeissi, K., et al.: Mechanical and cytotoxicity evaluation of nanos-
tructured hydroxyapatite-bredigite scaffolds for bone regeneration. Mater. Sci. Eng. C 68,
603–612 (2016)
85. Williams, J.M., Adewunmi, A., Schek, R.M., et al.: Bone tissue engineering using poly-
caprolactone scaffolds fabricated via selective laser sintering. Biomaterials 26, 4817–4827
(2005)
86. Schumacher, T.C., Volkmann, E., Yilmaz, R., et al.: Mechanical evaluation of calcium-
zirconium-silicate (baghdadite) obtained by a direct solid-state synthesis route. J. Mech.
Behav. Biomed. Mater. 34, 294–301 (2014)
87. Abbasian, V., Emadi, R., Kharaziha, M.: Biomimetic nylon 6-baghdadite nanocomposite
scaffold for bone tissue engineering. Mater. Sci. Eng. C 109, 110549 (2020)
88. Wang, R., Li, J., Chen, W., et al.: A biomimetic mussel-inspired ε-poly-l-lysine hydrogel with
robust tissue-anchor and anti-infection capacity. Adv. Funct. Mater. 27. Epub ahead of print
(2017). https://doi.org/10.1002/adfm.201604894
89. Yuan, H., Zhan, Y., Rowan, A.E., et al.: Biomimetic networks with enhanced photodynamic
antimicrobial activity from conjugated polythiophene/polyisocyanide hybrid hydrogels.
Angew. Chemie Int. Ed. 59, 2720–2724 (2020)
90. Izadyari Aghmiuni, A., Heidari Keshel, S.: The role of the extracellular matrix (ECM) and
ECM-like polymeric substrates in health and disease. In: Berhardt, L.V. (ed.) Advances in
Medicine and Biology. NOVA Medicine and Health, pp. 145–175 (2022)
91. Aghmiuni, A.I., Ghadi, A., Azmoun, E., et al.: Electrospun polymeric substrates for tissue
engineering: viewpoints on fabrication, application, and challenges. In: Electrospinning—
Material Technology of the Future. InTech, pp. 1–26 (2022)
392 A. Izadyari Aghmiuni and A. Gholami
92. Izadyari Aghmiuni, A., Heidari Keshel, S., Sefat, F., et al.: Quince seed mucilage-based
scaffold as a smart biological substrate to mimic mechanobiological behavior of skin and
promote fibroblasts proliferation and h-ASCs differentiation into keratinocytes. Int. J. Biol.
Macromol. 142, 668–679 (2020)
93. Izadyari Aghmiuni, A., Akbarzadeh Khiavi, A.: Medicinal plants to calm and treat psoriasis
disease. In: El-Shemy, H.A. (ed.) Aromatic and Medicinal Plants—Back to Nature, pp. 1–28.
InTech, Rijeka
94. Tahmasebi, A., Shapouri Moghadam, A., Enderami, S.E., et al.: Aloe vera-derived gel-blended
PHBV nanofibrous scaffold for bone tissue engineering. ASAIO J. 66, 966–973 (2020)
95. Suganya, S., Venugopal, J., Agnes Mary, S., et al.: Aloe vera incorporated biomimetic nanofi-
brous scaffold: a regenerative approach for skin tissue engineering. Iran. Polym. J. 23, 237–248
(2014)
96. Rahman, S., Carter, P., Bhattarai, N.: Aloe vera for tissue engineering applications. J. Funct.
Biomater. 8, 6 (2017)
97. Paul, K., Darzi, S., Del Borgo, M.P., et al.: Vaginal delivery of tissue engineered endome-
trial mesenchymal stem/stromal cells in an aloe vera-alginate hydrogel alleviates maternal
simulated birth injury. Appl. Mater. Today 22, 100890 (2021)
98. Zadegan, S., Nourmohammadi, J., Vahidi, B., et al.: An investigation into osteogenic differ-
entiation effects of silk fibroin-nettle (Urtica dioica L.) nanofibers. Int. J. Biol. Macromol.
133, 795–803 (2019)
99. Ghiyasi, Y., Salahi, E., Esfahani, H.: Synergy effect of Urtica dioica and ZnO NPs on
microstructure, antibacterial activity and cytotoxicity of electrospun PCL scaffold for wound
dressing application. Mater. Today Commun. 26, 102163 (2021)
100. Martins, R.M., Alves, G.D.A.D., Martins, S.D.S., et al. Apple extract (Malus sp.) and rutin as
photochemopreventive agents: evaluation of ultraviolet B-induced alterations on skin biopsies
and tissue-engineered skin. Rejuvenation Res. 23, 465–475 (2020)
101. Hajiali, H., Summa, M., Russo, D., et al.: Alginate–lavender nanofibers with antibacterial
and anti-inflammatory activity to effectively promote burn healing. J. Mater. Chem. B 4,
1686–1695 (2016)
102. Pilehvar-Soltanahmadi, Y., Nouri, M., Martino, M.M., et al.: Cytoprotection, proliferation and
epidermal differentiation of adipose tissue-derived stem cells on emu oil based electrospun
nanofibrous mat. Exp. Cell Res. 357, 192–201 (2017)
103. Peng-Zhang, Dai, K.R., Yan, S.G., et al.: Effects of naringin on the proliferation and osteogenic
differentiation of human bone mesenchymal stem cell. Eur. J. Pharmacol. 607, 1–5 (2009)
104. Yu, G.Y., Zheng, G.Z., Chang, B., et al.: Naringin stimulates osteogenic differentiation of rat
bone marrow stromal cells via activation of the notch signaling pathway. Stem Cells Int. Epub
ahead of print (2016). https://doi.org/10.1155/2016/7130653
105. Zhang, J., Xu, D.P., Shang, J., et al.: Effects of naringin on the proliferation and osteogenic
differentiation of canine bone marrow stromal cells in vitro. J. Hard Tissue Biol. 30, 73–78
(2021)
106. Kim, B.S., Kang, H.J., Park, J.Y., et al.: Fucoidan promotes osteoblast differentiation via JNK-
and ERK-dependent BMP2-Smad 1/5/8 signaling in human mesenchymal stem cells. Exp.
Mol. Med. 47, 1–9 (2015)
107. Li, F., Yang, Y., Zhu, P., et al.: Echinacoside promotes bone regeneration by increasing OPG/
RANKL ratio in MC3T3-E1 cells. Fitoterapia 83, 1443–1450 (2012)
108. Satué, M., Arriero, M.D.M., Monjo, M., et al.: Quercitrin and taxifolin stimulate osteoblast
differentiation in MC3T3-E1 cells and inhibit osteoclastogenesis in RAW 264.7 cells.
Biochem. Pharmacol. 86, 1476–1486 (2013)
109. Kim, M.B., Song, Y., Hwang, J.K.: Kirenol stimulates osteoblast differentiation through acti-
vation of the BMP and Wnt/β-catenin signaling pathways in MC3T3-E1 cells. Fitoterapia 98,
59–65 (2014)
110. Choi, E.M.: Honokiol isolated from Magnolia officinalis stimulates osteoblast function and
inhibits the release of bone-resorbing mediators. Int. Immunopharmacol. 11, 1541–1545
(2011)
Advanced Tissue Engineering with Novel Engineered Biomaterials 393
111. Xiao, H.H., Gao, Q.G., Zhang, Y., et al.: Vanillic acid exerts oestrogen-like activities in
osteoblast-like UMR 106 cells through MAP kinase (MEK/ERK)-mediated ER signaling
pathway. J. Steroid Biochem. Mol. Biol. 144, 382–391 (2014)
112. Tiyasatkulkovit, W., Charoenphandhu, N., Wongdee, K., et al.: Upregulation of osteoblastic
differentiation marker mRNA expression in osteoblast-like UMR106 cells by puerarin and
phytoestrogens from Pueraria mirifica. Phytomedicine 19, 1147–1155 (2012)
113. Delloye, C., Cornu, O., Druez, V., et al.: Bone allografts: what they can offer and what they
cannot. J. Bone Joint Surg. Br. 89, 574–580 (2007)
114. Seiler, J.G., 3rd., Johnson, J.: Iliac crest autogenous bone grafting: donor site complications.
J. South. Orthop. Assoc. 9, 91–97 (2000)
115. Tappa K, Jammalamadaka U. Novel biomaterials used in medical 3D printing techniques. J.
Funct. Biomater. 9. Epub ahead of print (2018). https://doi.org/10.3390/jfb9010017
116. Xu, T., Jin, J., Gregory, C., et al.: Inkjet printing of viable mammalian cells. Biomaterials 26,
93–99 (2005)
117. Seyedmahmoud, R., Messler, M.J., Loboa, E.G.: 3D bioprinting technologies for tissue
engineering: a mini review. HSOA J. Stem Cells Res. Dev. Ther. 6
118. Ong, C.S., Yesantharao, P., Huang, C.Y., et al.: 3D bioprinting using stem cells. Pediatr. Res.
83, 223–231 (2018)
119. Akkuş, H., Günaydin, B., Ustundag, C.: DOku mühendisliğinde 3 boyutlu biyo-baski için
biyofonksiyonel mürekkepler. Int. J. 3D Print Technol. Digit. Ind. 4 285–294 (2020)
120. Hacıoglu, A., Yılmazer, H., Ustundag, C.B.: 3D printing for tissue engineering applications.
Politek. Derg. 21, 221–227 (2018)
121. Tasnim, N., De la Vega, L., Anil Kumar, S., et al.: 3D bioprinting stem cell derived tissues.
Cell. Mol. Bioeng. 11, 219–240 (2018)
122. Vurat, M.T.: Üç-boyutlu biyobasım için yeni kompozit biyomürekkep geliştirilmesi ve
karakterizasyonu
123. Cui, X., Boland, T., DD’Lima, D., et al.: Thermal inkjet printing in tissue engineering and
regenerative medicine. Recent Pat. Drug Deliv. Formul. 6, 149–155 (2012)
124. Murphy, S.V., Atala, A.: 3D bioprinting of tissues and organs. Nat. Biotechnol. 32, 773–785
(2014)
125. Yilmaz, B., Tahmasebifar, A., Baran, E.T.: Bioprinting technologies in tissue engineering.
Curr. Appl. Pharm. Biotechnol. 279–319 (2020)
126. Liu, T., Guessasma, S., Zhu, J., et al.: Microstructural defects induced by stereolithography
and related compressive behaviour of polymers. J. Mater. Process. Technol. 251, 37–46 (2018)
127. Ligon, S.C., Liska, R., Stampfl, J., et al.: Polymers for 3D printing and customized additive
manufacturing. Chem. Rev. 117, 10212–10290 (2017)
128. Rhee, S., Puetzer, J.L., Mason, B.N., et al.: 3D bioprinting of spatially heterogeneous collagen
constructs for cartilage tissue engineering. ACS Biomater. Sci. Eng. 2, 1800–1805 (2016)
129. Laronda, M.M., Rutz, A.L., Xiao, S., et al.: A bioprosthetic ovary created using 3D printed
microporous scaffolds restores ovarian function in sterilized mice. Nat. Commun. 8, 15261
(2017)
130. Markstedt, K., Mantas, A., Tournier, I., et al.: 3D bioprinting human chondrocytes with
nanocellulose–alginate bioink for cartilage tissue engineering applications. Biomacromol 16,
1489–1496 (2015)
131. Nguyen, D., Hägg, D.A., Forsman, A., et al.: Cartilage tissue engineering by the 3D bioprinting
of iPS cells in a nanocellulose/alginate bioink. Sci. Rep. 7, 1–10 (2017)
132. Tan, Z., Parisi, C., Di Silvio, L., et al.: Cryogenic 3D printing of super soft hydrogels. Sci.
Rep. 7, 16293 (2017)
133. Bee, S.-L., Hamid, Z.A.A., Mariatti, M., et al.: Approaches to improve therapeutic efficacy
of biodegradable PLA/PLGA microspheres: a review. Polym. Rev. 58, 495–536 (2018)
134. Fontes, A.B., Marcomini, R.F.: 3D bioprinting: a review of materials, processes and bioink
properties. J. Eng. Exact Sci. 6, 617–639 (2020)
135. Yu, J., Park, S.A., Kim, W.D., et al.: Current advances in 3D bioprinting technology and its
applications for tissue engineering. Polymers (Basel) 12, 2958 (2020)
394 A. Izadyari Aghmiuni and A. Gholami
136. Aduba, D.C., Jr., Margaretta, E.D., Marnot, A.E.C., et al.: Vat photopolymerization 3D
printing of acid-cleavable PEG-methacrylate networks for biomaterial applications. Mater.
Today Commun. 19, 204–211 (2019)
137. Lee, J.S., Park, T.H., Ryu, J.Y., et al.: Osteogenesis of 3d-printed pcl/tcp/bdecm scaffold using
adipose-derived stem cells aggregates; an experimental study in the canine mandible. Int. J.
Mol. Sci. 22. Epub ahead of print (2021). https://doi.org/10.3390/ijms22115409
138. Shim, J.H., Won, J.Y., Park, J.H., et al.: Effects of 3D-printed polycaprolactone/β-tricalcium
phosphate membranes on guided bone regeneration. Int. J. Mol. Sci. 18. Epub ahead of print
(2017). https://doi.org/10.3390/ijms18050899
139. Tappa, K., Jammalamadaka, U., Ballard, D.H., et al.: Medication eluting devices for the field
of OBGYN (MEDOBGYN): 3D printed biodegradable hormone eluting constructs, a proof
of concept study. PLoS ONE 12, 1–17 (2017)
140. Horst, D.J., Tebcherani, S.M., Kubaski, E.T., et al.: Bioactive potential of 3D-printed oleo-
gum-resin disks: B. papyrifera, C. myrrha, and S. benzoin loading Nanooxides—TiO2 , P25,
Cu2 O, and MoO3 . Bioinorg. Chem. Appl. (2017). Epub ahead of print. https://doi.org/10.
1155/2017/6398167
141. Wang, Y., Wang, K., Li, X., et al.: 3D fabrication and characterization of phosphoric acid
scaffold with a HA/β-TCP weight ratio of 60:40 for bone tissue engineering applications.
PLoS ONE 12, 1–17 (2017)
142. Poldervaart, M.T., Goversen, B., De Ruijter, M., et al.: 3D bioprinting of methacrylated
hyaluronic acid (MeHA) hydrogel with intrinsic osteogenicity. PLoS ONE 12, 1–15 (2017)
143. Sa, M.W., Nguyen, B.N.B., Moriarty, R.A., et al.: Fabrication and evaluation of 3D printed
BCP scaffolds reinforced with ZrO2 for bone tissue applications. Biotechnol. Bioeng. 115,
989–999 (2018)
144. Kuroda, S., Kobayashi, T., Ohdan, H.: 3D printing model of the intrahepatic vessels for
navigation during anatomical resection of hepatocellular carcinoma. Int. J. Surg. Case Rep.
41, 219–222 (2017)
145. Asadi-Eydivand, M., Solati-Hashjin, M., Shafiei, S.S., et al.: Structure, properties, and in vitro
behavior of heat-treated calcium sulfate scaffolds fabricated by 3D printing. PLoS ONE 11,
e0151216 (2016)
146. Esworthy, T.J., Miao, S., Lee, S.-J., et al.: Advanced 4D-bioprinting technologies for brain
tissue modeling and study. Int. J. Smart Nano Mater. 10, 177–204 (2019)
147. Arslan-Yildiz, A., El, A.R., Chen, P., et al.: Towards artificial tissue models: past, present,
and future of 3D bioprinting. Biofabrication 8, 14103 (2016)
148. Gao, B., Yang, Q., Zhao, X., et al.: 4D bioprinting for biomedical applications. Trends
Biotechnol. 34, 746–756 (2016)
149. Li, Y.-C., Zhang, Y.S., Akpek, A., et al.: 4D bioprinting: the next-generation technology for
biofabrication enabled by stimuli-responsive materials. Biofabrication 9, 12001 (2016)
150. Saravanan, S., Vimalraj, S., Thanikaivelan, P., et al.: A review on injectable chitosan/beta
glycerophosphate hydrogels for bone tissue regeneration. Int. J. Biol. Macromol. 121, 38–54
(2019)
151. Graham, S., Marina, P.F., Blencowe, A.: Thermoresponsive polysaccharides and their
thermoreversible physical hydrogel networks. Carbohydr. Polym. 207, 143–159 (2019)
152. Suo, H., Zhang, D., Yin, J., et al.: Interpenetrating polymer network hydrogels composed
of chitosan and photocrosslinkable gelatin with enhanced mechanical properties for tissue
engineering. Mater. Sci. Eng. C 92, 612–620 (2018)
153. Yang, J., van Lith, R., Baler, K., et al.: A thermoresponsive biodegradable polymer with
intrinsic antioxidant properties. Biomacromol 15, 3942–3952 (2014)
154. Chen, J.-P., Tsai, M.-J., Liao, H.-T.: Incorporation of biphasic calcium phosphate microparti-
cles in injectable thermoresponsive hydrogel modulates bone cell proliferation and differen-
tiation. Colloids Surfaces B Biointerfaces 110, 120–129 (2013)
Advanced Tissue Engineering with Novel Engineered Biomaterials 395
Abstract In modern medicine, drug delivery is a broad field of research for the
evolution of novel materials or carrier systems for effective therapeutic delivery
of drugs as the controlled drug delivery is a challenge on the basis of solubility,
bioavailability, cytotoxicity along with pharmacokinetic parameters.
Protein-based drug delivery systems (DDS) have shown promising results due to
structural support, cell-targeted delivery, bioavailability, biocompatibility and non-
immunogenicity, etc. Collagen as an important extracellular matrix component has
attracted drug delivery-based research in recent years. Collagen based-hydrogels/
composites/biofilms are excellent objects for drug delivery, tissue engineering,
wound dressings and gene therapeutics etc. due to high encapsulating capacity,
mechanically strong swollen structural network and efficient mass transfer prop-
erties. Some of the applications of collagen are the formation of microspheres and
microneedles for drug delivery, formulation of nanoparticles (NPs) for gene delivery,
development of pellets and tablets for protein delivery, formation of gels and combi-
nation with liposomes for sustained drug delivery, cancer treatment and collagen
shields in ophthalmology.
DDS based on NPs display enhanced efficacy of drugs and improve the drug’s half-
life, hydrophobic drug solubility and controlled/sustained drug release in the infected
body regions. Stimuli-responsive NPs regulate drug biodistribution and reduce drug
toxicity. Protein-based nanocomposites can be prepared through various physical
and chemical methods like desolvation, emulsification, phase separation, electro-
spray, electrospinning and milling, etc. These methods have their operating ease and
difficulties for the production of the desired quality of nanomaterials/composites.
Current Polymeric NPs systems are sensitive to stimuli such as temperature,
light, pH, oxidizing/reducing agents, magnetic fields and enzymes which increases
efficiency and specificity for various applications. Collagen with NPs results in
stabilization of the nanoparticles and helps with entrapment of the drug, to attain
steady and regulated drug release for ideal therapeutic reactions. Collagen NPs have
A. K. Verma (B)
Department of Biosciences, Srinivasa Ramanujan Block, Jamia Millia Islamia, Jamia Nagar, New
Delhi 110 025, India
e-mail: averma@jmi.ac.in
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 397
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_13
398 A. K. Verma
advantage over other natural and synthetic polymeric NPs due to biocompatibility,
biodegradability, low antigenicity, high contact surface and reduced toxicity.
Significant advancements have been achieved using collagen-based nano-DDS to
deliver biomolecules with better efficacy at targeted sites. In spite of the substan-
tial progress, collagen is still affected by low mechanical strength and high rate of
degradability, which is a serious concern during clinical trials of targeting intracel-
lular molecules like genes, drugs and growth factors, etc. In future, collagen-based
nano-DDS will be the key player for the delivery of desired drugs/biomolecules at
specific target for different medical conditions.
1 Introduction
Drug delivery systems (DDS) are the important, impressive and effective tools for
disease treatment, management and prevention in the field of modern medicine. The
DDS comprises the introduction and delivery of active biomolecule or pharmaceu-
tical at targeted location in the body with improvised efficiency, biocompatibility and
safety. Further, DDS involves the augmented therapeutic performance via increased
absorption, prolonged or sustained release at specific target site with minimal cyto-
toxicity and effectively removal from the body without causing adverse physiolog-
ical conditions [1–3]. Guided drug delivery is the challenge, because the bioactive
molecule has to survive through various cellular or tissue level barriers during circula-
tion before binding to a particular target in body. In recent years, the use of nanoscale
protein-based polymers has been sufficiently progressed for delivery of drugs and
vaccines due to the ability to cross biological barriers and to reach at the target site
at molecular level [4].
Nanoparticles (NPs) are microscopic structures with typical size range of 1–100
nm in diameter have gained the attention of scientists due to their small size, high
surface-to-volume ratio, stability, self-assembly behaviour and mutual interactions at
fluid interfaces, make them the suitable candidate for delivering the variety of drugs
of active biomolecules in biomedicine [5, 6].
The crucial biological molecules- proteins and peptides are the most important
objects of research in the areas of nanomedicine. These biomolecules based nanocon-
structs are the centre of attraction in the field of nanomedicine, nanobiotechnology,
toxicology and immunology to understand the interactions with other molecules in
body for therapeutic and diagnostic purposes [7, 8]. The nature and organic compo-
sition of protein-based nanocomposites have the instinctive affinity to interact with
living cells or tissues and they are often not considered as foreign entity and are readily
eliminated from the body through macrophages. The typical property of protein nano-
material is inherent uniform size and homogenous size distribution throughout the
An Insight into Collagen-Based Nano Biomaterials for Drug Delivery … 399
Nanotechnology is the modern branch of science and engineering deals with the
technological developments of materials at the nanoscale for various physical, chem-
ical, industrial and medical applications [17, 18]. This technology envisages the
construction and characterization of engineered materials by rearranging at atomic,
molecular and supramolecular scales at 1–100 nm size in one or more dimensions.
Based on shapes, the nanomaterials could be of zero dimension in NPs, one dimen-
sion in nanorods, nanosheets in two dimension and three dimensions in nanotubes,
nanowires or multi-nanolayers and dendrimers [19, 20]. In present scenario, nano
biomaterials are utilized in different biomedical applications such as drug delivery,
biosensing, bioimaging, antibacterial and anti-cancer activity, medical diagnosis,
tissue engineering/regeneration, wound healing, medical equipment, cosmetics, food
packaging industry and environmental remedy, etc. [21].
Nanocollagen (NC) is the typical regular collagen downsized at the nanoscale
dimension of 1–100 nm, having desirable qualities of NPs like high surface area to
volume ratio and biomimetic, bioavailability, low antigenicity, optimal penetration
potential at particular location of collagen, making the collagen as very effective
biomolecule for drug delivery and wound healing applications in medical sciences
[21]. Collagen nanofibers are cylindrical network of fibres with less than 1000 nm
of external diameter and more than 50 ratio of length and width [22]. Wang et al.
[23] designed the NC fibre with uniform networking and structure with attributes of
elasticity and resistance to break, making it suitable for tissue engineering applica-
tion. The NC fibres could withstand 500 mN tensile load with 50 nN load resolution,
which later augmented the load transfer efficiency.
Protein-based NPs preparation and drug encapsulation are performed in optimal
environment to avoid the toxic solvents or organic chemicals as residue after the
synthesis. NC can be generally produced by chemical, physical and self-assembly
methods [18, 24]. The chemical methods are emulsification and coacervation or
400 A. K. Verma
Table 1 Pros and cons of protein/collagen-based NPs formulation preparations. (Table adapted
from open source, https://doi.org/10.3390/app112311369)
Process Advantages Disadvantages References
Emulsification/ Easy method, equipment required Large sized NPs [63, 64]
solvent extraction simple, high flexibility, stability, produced,
selectivity and encapsulation thermodynamic
potential of NPs, controllable instability needs more
shape, size of NPs and secondary amount of surfactant
structure of protein, zeta potential and stabilizers or
in limit organic solvents may
lead to toxicity
Complex highly stable and small sized NPs, Challenge for scale up [65]
coacervation or NPs shape and size controllable
polyelectrolyte with experiment conditions, protein
complexation or peptide like sensitive drug can be
mixed
Phase separation Homogenous NPs formed, specific Particle size restricted [37, 66]
instrument not involved, particle to 50–500 nm, small
size controlled by altering polymer scale production,
amount involved organic
solvents may be toxic
Electrospinning Flexibility and easy insertion of Use of organic solvents, [41, 67]
drug, NPs etc., high porosity, large limited control of pore,
surface-to-volume ratio, swelling structure
capacity, controllable, gaseous
permeation, economical and simple
Nanospray Experimentally easy method, Difficult incorporation [68, 69]
drying cost-effective, easy encapsulation of hydrophobic drugs,
of hydrophilic drugs, reduced confined to small scale
degradation of heat-sensitive production, decreased
samples, NPs size can be restricted encapsulation potential,
high energy required
Electrospray Formulation of dry and highly Parameters like drying, [47, 70]
deposition stable NPS, convenient synthesis sheer stress of nozzle
due to one-step method, efficient produce thermal
drug loading capacity, scalable for degradation of
industrial purposes, high yield and macromolecules
reproducibility, versatile method
Milling Simple and cost-effective process Cooling required [50, 71]
for large scale fabrication of NPs during procedure due to
heat generation.
Coarse NPs produced,
reduced control for NPs
shape
Self assembly Small NPs with enhanced Uncontrollable NPs [52, 53]
encapsulating efficiency can be size and shape, high
formed through this highly stable possibility of protein
process, simplicity, versatility strain
(continued)
402 A. K. Verma
Table 1 (continued)
Process Advantages Disadvantages References
Desolvation Controllable size and shape of NPs, Process applicable with [17, 51]
better encapsulating potential and proteins affected by
highly stable process desoluble process or
diluted by transporter
proteins
Interfacial Simple process, undesirable Long duration of the [56, 58]
polymerization monomer purity process
Polymer chain NPs attributes controllable, great Restricted NPs [72, 73]
collapse stability and improved spherical diameter (5–20 nm) and
shaped NPs challenging control for
side reactions
In the emulsion solvent evaporation for production of NPs, aqueous and organic
phase plays crucial role. The advantage of the phase separation method is that it
is a relatively simple procedure and needs minimum apparatus. In this method, the
collagen polymer is dissolved in solution and phase separation is triggered, though
404 A. K. Verma
Fig. 3 Phase separation—By altering pH, temperature and ionic strength, collagen in aqueous
solution form aggregates followed by addition of crosslinker, form the crosslinked collagen NPs
through ultrasonication technique. (Image modified from open source, https://doi.org/10.3390/app
112311369)
addition of surfactant to this protein solution. The resulting mixture is then ultra-
sonicated for proper homogenization followed by polymer droplets formation and
isolation of solvent. The formulated NPs by the phase separation are having 50–500
nm of diameter (Fig. 3) [24]. Yoon et al. [37] fabricated the NPs of conjugated poly-
mers through phase separation technique forming the lipid-incorporated NPs having
the potential to form composites with inorganic/organic nanosubstances, cell specific
ligands with polyethylene glycol for in vivo applications.
2.4 Electrospinning
Fig. 4 Electrospinning—Collagen solution under influence of high electric field is passed through
syringe, form Taylor cone followed by drying of solvent and generation of interconnected collagen
nanofibers
[41]. Among all the methods for nanofibers formulations, electrospinning is mostly
applied process due to economical and simplicity [42].
This technique is the physical NPs fabrication process to produce the collagen NPs in
liquid medium. The collagen solution along with hot CO2 and nitrogen gas is sprayed
through nozzle into the heated chamber to generate the hollow NPs, which are later
collected at the bottom of the chamber under the influence of charged electrodes.
Finally, the created NPs are frozen, lyophilized and crosslinked (Fig. 5). Liquid N2
is used to avoid the denaturation of collagen molecules during the spraying in highly
heated chamber [43]. This method is fast and economical to fabricate the collagen
NPs at small scale and to encapsulate the hydrophilic drugs for delivery purposes.
The addition of surfactant to the protein solution makes the spherical NPs and also
stabilize them. The NPs size can be modified by restricting the nozzle size and rate
of spraying [44, 45].
406 A. K. Verma
Fig. 5 Nanospray drying—at high temperature, stream of N2 , CO2 along with cooled collagen is
sprayed through nozzle in chamber and dried NPs are collected at the electrode. (Image modified
from open source, https://doi.org/10.3390/app112311369)
This technique is the process of liquid atomization, where the high voltage is applied
to the collagen solution and liquid jet stream is passed through nozzle to form the
aerosolized drops of collagen NPs (Fig. 6). Drugs and nucleic acids can be efficiently
incorporated into these generated NPs with high productivity [24, 46]. The operating
conditions like applied voltage, operating distance, gauge diameter of needle and flow
rate fluctuate according to different types of drug delivery modules. In the advanced
technique of electrospray, coaxial spray of collagen in salt solution and drug is
performed simultaneously to form the solid collagen NPs with encapsulated drug for
delivery objectives [47]. Electrospray deposition method is economical, convenient
to handle with efficient encapsulating capability. Solid and stable collagen NPs can
be formulated to overcome the complications of reduced encapsulation potential and
low rate of biocompatibility during drug delivery [48].
An Insight into Collagen-Based Nano Biomaterials for Drug Delivery … 407
Fig. 6 Electrospray deposition—Liquid jet stream of drug and protein solution are passed through
nozzle to generate the aerosolized droplets of drug encapsulated NPs at high temperature. (Image
modified from open source, https://doi.org/10.3390/app112311369)
2.7 Milling
Milling is the mechanical process to fabricate the collagen NPs under the influence
of high mechanical energy. The milling balls are utilized to generate mechanical
collisions to produce disintegrated polymeric material into finer NPs (Fig. 7). During
the milling, excess mechanical and kinetic energy generate the heat in the vessel;
to avoid the heat milling vessel is cooled to reduce the breakdown of polymeric
substance. Heat responsive protein or collagen material is milled under the control
of liquid N2 and cryogenic temperatures to counter the protein denaturation. Milling
is the cost-effective technique to scaledown the particle size at large scale production
of NPs [49, 50].
408 A. K. Verma
Fig. 7 Milling—Polymeric solution is mixed with milling balls under high mechanical and kinetic
energy to produce the finer collagen NPs at cryogenic temperature. (Image modified from open
source, https://doi.org/10.3390/app112311369)
When altered hydrophobic protein molecules are dissolved in aqueous solution, form
the micelle NPs through self-assembly process. The hydrophobic pockets in these
micelle NPs are the suitable place for encapsulation of active ingredients. In the
self-assembly process, when the fibres of protein in solution at a critical solution
temperature surpass the critical micelle concentration leads to the development of
nanoscale composites. Later, through the process of solidification these nanoscale
micelles become stable due to crosslinking between chains or fibres or sheets of
protein molecules. By varying the self-assembly parameters of the collagen fabri-
cates, sustained release of active molecules in the collagen-based wound dressings
could be obtained to enhance the fibroblast production and expedited wound healing
[51, 52]. Layer-by-layer (LbL) self-assembly process is conventional technique for
fabricating multi-layer films and indulges the sequential adsorption of two or more
building block that can complementarily combine with specific substrate at molec-
ular level through electrostatic, hydrophobic interactions and hydrogen bonding.
These interactions facilitate the deposition of alternate layers with opposite charged
biomaterials and control the nanoscale characters like thickness, surface attributes
and composition to the film [53, 54]. The main advantages of LbL self-assembly
An Insight into Collagen-Based Nano Biomaterials for Drug Delivery … 409
Polymeric membranes and particles can be fabricated through the technique of inter-
facial polymerization (Fig. 9). This method is a type of step-growth polymerization,
where two immiscible solutions form the polymeric substances with particular topo-
logical and chemical attributes, anisotropic shapes, hollow structures or alternative
surface chemistry at the interface region [56]. Here, the polymer is confined to inter-
face region. Through this technique large scale production of ultrathin layers or
membranes, hollow nanospheres and nanofibers is possible [57]. The category of
synthesized polymeric materials is zero-dimension NPs, 1D nanofibers, 2D films
and 3D composite membranes [58].
410 A. K. Verma
Fig. 9 Interfacial polymerization—two immiscible solutions like polymer and oil, form the
polymeric material at the phase interface after polymerization. (Adapted from open source
paper (https://www.researchgate.net/publication/267901674_A_Multifunctional_Smart_Coating_
for_Automous_Corrosion_Control)
Polymer chain collapse technique can produce the single-chain polymer NPs (SCNP)
with excellent stability in size range of 5–20 nm (Fig. 10). The NPs size can be
controlled by polymer molecular weight and the extent of crosslinking [59]. The
intramolecular polymer chain collapse method can generate the extremely small
polymeric NPs of approximately 10nm in diameter, not possible by other methods
of synthesis. Under the control of polymer precursor chain, the typical molecules of
NPs can be generated [60]. Various types of SCNP fabrication processes are available
on the basis of involved functional groups in the reactions [61]. Effective folding of
single-chain polymer through single-chain nano objects can mimic and control the
functionality and morphology of natural biomacromolecules such as enzymes, drug
delivery modules and catalysts. NPs with systematically smaller and spherical than
their homofunctional analogues can be produced by increasing the various types of
chemical linker molecules through orthogonal chemistry methods [62].
Protein-based nanomaterials are the emerging materials of research for drug delivery,
medical devices, therapeutics, tissue engineering and other biomedicine areas due
to small size, significant absorption, distribution in the human body, ability to cross
the biological barriers and exhibiting negligible cytotoxicity [20]. When the size of
the particle reaches at the nano level, most of the physical attributes of NPs are quite
distinct form microparticles and bulk materials, providing more advanced options
An Insight into Collagen-Based Nano Biomaterials for Drug Delivery … 411
wounded site exhibited the exception wound-healing capacity of collagen NPs [78].
In contrast to collagen microfibers, silver NPs (AgNPs) impregnated nanocollagen
had better potential for rapid regeneration of wounded tissue [81]. In a report, Cardoso
et al. [82], fabricated the AgNPs-type 1 collagen composite (AgNPcol) for antibac-
terial and cell viability potentials. All the formulations were analyzed on the basis
of particle size, zeta potential and polydispersity index (PDI). The particle diam-
eter and PDI were 64–81 nm and 0.40–0.77 of range, respectively. PDI value is the
selecting parameter for low-polydispersity solution related to cell viability test. The
positive zeta potential of AgNPcol expressed effective antibacterial activity against
Escherichia coli and Staphylococcus aureus. The opposite surface charge and small
size of NPs promote antibacterial activity.
Roshanbinfar et al. [83] synthesized the electrically conductive hydrogel of type
I collagen and branched polyethyleneimine (bPEI)-caoted AuNPs for cardiac tissue
generation by adding neonatal rat ventricular or human embryonic or induced
pluripotent stem cell (hiPSC) derived cardiomyocytes. Along the collagen matrix,
bPEI-AuNPs were uniformly distributed with distance in the magnitude of few
hundred nanometers. A wide range of negatively charged biomedical drug, active
ingredients and growth factors can be effectively loaded on the positively charged
coating of AuNPs. During the cardiac cycle, high amount of forces are produced,
that should be acceptable by engineered material to assure the no drug or cell content
leakage. Production of hydrogel-based cardiac tissue material for large animal’s
mechanical stress is a tough task. The AuNPs escalated the hydrogel stiffness from 91
to 146 kPa and also expanded the electrical conductivity from 40 to 49–69 mS cm−1
for the slow and uniform release of incorporated drug. These hydrogels-based engi-
neered tissues have the potential to reduce cardiac arrest and treatment of ailments
of electrically sensitive tissues.
Terzopoulou et al. [84] designed the curcumin (Cur)-loaded chitosan (CS) NPs
and incorporated into the hydrophilic and biocompatible collagen-based patch as
topical preparation for psoriatic skin. Size of the NPs is very crucial parameter influ-
encing the drug release profile and biological performance of NPs. NPs less than 200
nm in diameter are supposed to manage the drug discharge rate and penetrate the cells
more adequately, while the zeta potential of greater than +30 mV contributed better
stability and capacity to adhere the negatively charged biomembranes. The fabri-
cated Cur-CS-NPs were expected to penetrate the upper layers of skin through injec-
tion. The collagen patches expressed the significant swelling ratio of upto ~1500%
and after crosslinking with EDC (1-ethyl-3-(3-dimethylaminopropyl) carbodiimide
hydrochloride)/ NHS (N-hydroxysuccinimide) the pore size decreased which leads
to decreased hydrolysis rate. Release of curcumin within collagen patches generated
the proliferative inhibitory effect on psoriatic human keratinocytes in vitro.
NP’s introduction into the body may lead to rapid degradation and instability of
NPs followed by elimination from the body through phagocytic processes [85]. The
stability of NPs is depended on the composition of the composite and its interaction
with surroundings. Lipid and polymer-based NPs are most sensitive to aggregation
and instability [20]. In body, circulatory system factors like blood flow, excretion and
phagocytic cells may decrease the NPs stability and delivery. NPs with diameter of
An Insight into Collagen-Based Nano Biomaterials for Drug Delivery … 413
<10 nm usually removed through kidney in fast manner while NPs with >200 nm may
initiate the complement system [86]. Several fabricated NPs utilize the coating of
polyethylene glycol (PEG) to avert the fast excretion of NPs from body. PEGylation
enhanced the circulation time though modification of NPs size and solubility and
provide protection to NPs surface from degradation, secretion and clearance caused
by enzymes and antibodies. At high concentration, PEG coated NPs may lead to
anti-PEG antibodies could activate the clearance of PEGylated NPs [87].
Polymeric NPs for drug administration should have high drug loading capacity and
the reduced volume of media. Active ingredients or pharmaceuticals can be incorpo-
rated in NPs through two approaches—first, encapsulate the drug during the synthesis
of NPs and second, on the surface of NPs attach or insert drug through adsorption after
the synthesis of NPs [88]. These alterations make the NPs quintessential for delivery
applications and are suitable for delivery of various payloads of hydrophobic and
hydrophilic materials along with cargos of different molecular weights like small
molecules, bio-macromolecules, proteins and vaccines [89–92]. The loading effi-
ciency and release kinetics of loaded pharmaceuticals on NPs can be accurately
governed through modification of properties like composition, stability, response
and surface charge of NPs [66, 93]. Nano-carriers are considered as promising
therapeutics formulations due to high loading ability, significant intracellular drug
delivery, effective cellular absorption and controlled release in cytosols with adequate
biodegradation [78]. For the increment of biomolecule loading capability and cellular
absorption potential, surface roughening of NPs has been formulated as another
method [94].
Awad et al. [95] assessed the cytotoxicity effect of three combinations- collagen
NPs, theophylline loaded with collagen NPs and free theophylline alone using MTT
assay on NHSF cell line at the concentration of 180 μg/ml with double fold serial
dilutions. At the lower doses, collagen NPs, theophylline loaded with collagen NPs
and free theophylline had more than 97% cell viability. The cytotoxicity enhanced
significantly by increasing the concentration of theophylline with 55.59% of viability
at 180 μg/ml. Theophylline loaded with collagen NPs had higher cell viability of
78.53% and lower toxicity than at 180 μg/ml of theophylline alone. The order of cyto-
toxicity was collagen NPs < theophylline loaded with collagen NPs < theophylline
for all the concentrations. Theophylline loaded with collagen NPs showed lower
toxicity than free theophylline and slow release due to efficient encapsulation inside
collagen NPs. The collagen NPs were least cytotoxic at all the tested concentrations
due to natural biodegradable collagen and additives utilized for fabrication [96, 97].
The toxicity findings also expressed the biocompatibility, harmlessness of collagen
NPs with normal cells making them suitable as drug carriers for medicine.
414 A. K. Verma
For the optimal delivery of active ingredients, drug release and polymer biodegrad-
ability are the crucial criteria. The release rates of NPs is based on desorption of
the surface attached or adsorbed molecules, diffusion through NPs matrix, diffu-
sion through the polymer wall, erosion of NPs matrix and diffusion process. The
solubility, diffusion and biodegradation of NPs affect the discharge of drug in the
system. The releasee of drug through protein-based polymeric NPs is accomplished
via protein degradation, through pores, from polymer surfaces and electrical pulse-
based delivery through magnetic field etc. [98]. The drug release potential of protein-
based NPs can be controlled by modifying the geometry, coating thickness, porosity
and crystallinity of the protein drug vehicles [99]. Spherical-shaped NPs with less
than 200 nm diameter could cross the spleen to avoid the contact forces along the
blood vessel walls and other flow barriers in body [100]. To obtain the high drug
loading and emission, delivery vehicles can also be altered to attain the large pore
volume and surface areas. A drug carrier’s morphology, shape, diameter and thick-
ness should be considered for effective tailored drug release potential [3]. Porosity
of the NPs material is the preferred property to control the delivery of drug such as
surface area, permeability, loading capacity, release rate and pharmacokinetics and is
directly interconnected with pore size and pore interconnectivity. The increased pore
percentage can accelerate the degradation and augmented drug release in polymer
matrix [101].
At the high concentration or dose of pharmaceutical drug or active ingredients
after systemic introduction in body may generate undesirable side effects. To achieve
the similar efficiency or exceptional results, local delivery of therapeutic molecules
at low dosage has been researched [102]. By regulating the degree of crosslinking in
porous collagen-based scaffold, therapeutic substances can be encapsulated and their
retention and release can also be controlled [103]. Instead of administering the growth
factor directly, incorporation of growth factor loaded NPs in the crosslinked collagen
scaffolds, the target molecules may be delivered via composite delivery system.
For the controlled release of therapeutics, crosslinked collagen scaffold act as the
vehicle for lodging the NP carriers. NPs create more opportunities for interaction with
collagen, when they are directly or indirectly crosslinked with collagen. Metal oxide
NPs utilized for crosslinking with collagen by directly binding to its side chain. The
mechanical attributes of collagen-based scaffolds may be augments through metal
NPs like ZnO, iron oxide, alumina oxide, etc. Through UV irradiation, radical oxygen
species, promoted collagen crosslinking by metal oxide NPs and is incredible for
formulation of collagen shields for controlled drug delivery for glaucoma treatment
[104]. Choi et al. [105] developed the collagen constructs as inserts for wounded
human gingival for optimization of the release profile of growth factor TGF-β1 and
NPs of Au, TaO, dextran and ferritin. The released amount of biomolecule NPs
was quantified through fluorescence intensity and X-ray scattering. The collagen
hydrogel may be the promising system for sustained release of growth factor and
imaging agents in human gingival to promote the regeneration of oral tissues.
An Insight into Collagen-Based Nano Biomaterials for Drug Delivery … 415
In the field of medicine, traditional drugs are the pivotal strategies to treat various
ailments. The administration of classical drug in body at high dosage leads to mild or
severe side effects, sometimes leads to life-threatening condition. The formulation of
effective DDS for the transportation of natural or synthetic desired drug at target site
with optimal amount has been a demanding clinical investigation and research for
several years [106]. In the last decades, the growth of nanotechnology has unblocked
the different innovative possibilities in biomedicine, specifically in the area of drug
delivery. Different novel drug delivery vehicles at the micro or nano scale have been
formulated and number of such delivery modules has been increasing tremendously
to compensate for the need for targeted DDS [72].
Collagen is the most abundant extracellular protein found in connective tissues
in mammals and can be used for DDS through various architectural types such as
protein cages, nanoparticles, nanofilms, hydrogels and minirods with combination of
different natural or synthetic polymers and crosslinking chemicals. Several methods
such as emulsification, desolvation, electrospray, electrospinning, self-assembly,
milling and phase separation, etc., are applied for the fabrication of collagen-based
nanocomposites. Each preparatory process has various advantages and limitations.
In Table 2, list of different collagen-based nanocomposites or nanoformulations,
their method of preparation and loaded active ingredients or pharmaceuticals are
mentioned.
Lin et al. [107] fabricated the biocompatible nanofiber membrane of protein
collagen and zein by co-electrospinning method. Zein improvised the spinnability of
collagen and generated nanofiber properties like fibre diameter, surface wettability,
mechanical strength degradation potential and cell adhesiveness could be modi-
fied through alteration of collagen/zein ratio. The incorporated berberine drug in
nanofiber membrane was assessed for controlled release and antibacterial activity
and the berberine exhibited little effect on fibre morphology and cell viability. The
wound healing process and histological experiments were conducted in vivo on the
female Sprague–Dawley rats for the fabricated nanomembrane.
In a report by Karri et al. [108], a novel nanohybrid scaffold was fabricated through
emulsification method using curcumin (CUR) impregnated in chitosan (CS) NPs and
collagen scaffold for tissue regeneration application. The formulated CUR-CS-NPs
were assessed on the basis of size, zeta potential, SEM, DSC, X-ray analyses and the
novel nanohybird of CUR-CS-NPs-collagen scaffold was evaluated for water uptake,
biocompatibility and sustained release parameters. The in vivo wound closure anal-
ysis exhibited the faster and significant contraction of wound treated by nanohybrid
scaffold in contrast to control and placebo groups. The wound showed the complete
epithelialization with thick tissue formed in nanohybrid treated scaffold than control
and other group. The nanohybrid scaffold showed the promising potential for better
wound healing capacity for diabetic wounds.
416 A. K. Verma
Table 2 List of various collagen-based nanocomposites, their method of preparation and loaded
active ingredients. EDC-Hcl—3-ethyl carbodiimide-hydrochloride; MDA—malondialdehyde;
chitosan—CS; NHS—N-hydroxysuccinimide; NLC—nanostructured lipid carrier; PCL—poly(ε-
caprolactone); bPEI—branched polyethyleneimine; TGF-β1—transforming growth factor; FGF-
2—fibroblast growth factor; PLGA—poly(lactic-co-glycolic acid); BMP-2—bone morphogenic
protein; PLA—poly lactic acid; VEGF—vascular endothelial growth factor; PDGF—platelet
derived growth factor; EGF—endothelial growth factor; PHA—polyhydroxy butyrate
Nanocomposite Method of preparation Loaded References
pharmaceutical
Collagen NPs Nanoprecipitation Theophylline [95]
method using
non-solvent (ethanol)
Collagen NPs crosslinked with Emulsification Silymarin [96]
EDC-Hcl & MDA
Collagen patches-CS NPs Ion gelation method Curcumin [84]
crosslinked with Ion gelation
method EDC/NHS/heparin
Collagen gel-CS NLC Emulsification SiRNA [115]
Collagen hydrogel Layer-by-layer Doxorubicin [53]
self-assembly
Hydrolyzed collagen-PCL Radical Hydrocortisone [114]
co-polymerization
Collagen peptide-CS NPs Ion gelation method Doxorubicin [116]
Collagen-bPEI-AuNP hydrogel Desolvation Phenylephrine, [83]
isoproterenol,
doxorubicin
Collagen-CS dressing Freeze drying Ag NPs [117]
Collagen type 1 Chemical synthesis Ag NPs [82]
Collagen-iron oxide NPs Freeze drying, Fluorescein [118]
hydrogel coprecipitation
Collagen-ferrtin NPs-TaO NPs Microemulsion TGF-β1 [105]
hydrogel
Zein-collagen-PCl nanofiber Electrospinning Aloe vera [110]
scaffold
Collagen-graphene oxide Layer-by-layer FGF-2 [119]
sheet-hydrogel self-assembly
Collagen-hydroxyapatite-PLGA Emulsification (W/O/ BMP-2, alendronate [113]
scaffold W method)
Atelocollagen Mixing siRNA [120]
Type 1 collagen hydrogel Mixing Royal jelly [121]
extracellular vesicles
Collagen peptide-chito Electrospinning Antibacterial activity [122]
oligosaccharides membrane
PLGA/collagen nanofibrous Electrospinning Wound healing [123]
membrane
(continued)
An Insight into Collagen-Based Nano Biomaterials for Drug Delivery … 417
Table 2 (continued)
Nanocomposite Method of preparation Loaded References
pharmaceutical
Au-hydroxyapatite-collagen Wet precipitation/ Doxorubicin [124]
nanomaterial microwave assisted
synthesis
Collagen-based 3D culture Self assembly Coumarin [109]
system-conjugated NPs
Collagen-hydrogel-nano/ Electrospray Rat [125]
microfibers pheochromocytoma
cell cuture
PCL-collagen-nanobioglass Electrospinning Peripheral nerve [126]
nanocomposite regeneration
Collagen-chitosan Ion gelation, phase Minocycline [112]
membrane-chitosan NPs separation
Collagen-PLGA NPs asymmetric Electrospinning Aspirin, curcumin [127]
nanomembrane
Collagen-chitosan NPs Emulsification Curcumin [108]
Ethyl cellulose-PLA-collagen Electrospinning Silver sulfadiazine [128]
nanofiber mat
Collagen-zein nanofiber Co-electrospinning Berberine [107]
membrane
PLGA-collagen scaffold Electrospinning Glucophage [129]
nanomembrane
Collagen-hyaluronic acid-gelatin Electrospinning VEGF, PDGF, bFGF, [130]
NPs nanocomposite EGF
PHA-gelatin-collagen nanofibre Electrospinning Ostholamide [131]
nanofiber composite
Collagen-polyamide nanofiber Electrospinning N-acetly cysteine [132]
composite
Gelatin-fish collagen-chitosan Electrospinning Lithospermi radix [133]
nanofiber bilayer scaffold extract
PCL-collagen-polyethylene oxide Electrospinning Doxycycline [134]
PCL-collagen nanofibers Electrospinning Gentamicin sulphate [135]
Targeted drug delivery and positive antitumor outcomes are the main challenges
in cancer therapy. In view of this, Le et al. [109] developed the collagen-based 3D
multicellular culture module with coumarin conjugated NPs by self-assembly method
for the selection of new nanocarriers for drug delivery in preclinical research. The
findings showed that 3-D cell colonies were successfully developed from 95-D, U87
and HTC116 cell lines after seven day culture in collagen matrix. The coumrarin-
NPs were easily penetrated the matrix gel and targeted the tumor cells and the drug
module was good for evaluating the therapeutic outcomes of drug transport in vivo
or investigation in tumor biology for better DDS for cancer treatment.
418 A. K. Verma
upto 4 weeks and 2 weeks, respectively in contrast to BMP-2 and ALN without
PLGA microspheres. Evaluation of accumulation and maturation of bone-like tissue
in calvarial bone defect was performed by micro-computed tomography imaging
at 2, 4 and 8 weeks after implantation in rat models. Enhanced bone regeneration
was observed after 8 weeks post-implantation in rat with 8 mm critical-sized defect.
The fabricated nano-composite could be the future biomaterial for cell-free tissue
engineering.
Recently, conductive composites have been developed for in vitro and in vivo
biomedical applications. The conductive composites like current-stimuli polymeric
structures have been successfully introduced extra controlling DDSs equipped with
switchable release profile. Nowadays, drug release from conductive polymeric mate-
rials by applying the required voltage at a precise duration time is available. The
conductive polymers have broad applications in microelectronic industry, electrode
production, photovoltaic devices, biotechnological systems such as nerve regen-
eration and drug delivery systems. Pourjavadi and Doroudian [114], developed a
semi-conductive nanocomposite for electrically controlled drug delivery. Hydrolyzed
collagen was altered with administration of polycaprolactone. The hydrogel was
fabricated by radical co-polymerization of acrylic acid in the presence of crosslinker.
The reaction parameters influencing the water absorbance of hydrogel were opti-
mized through Taguchi method. In situ polymerization of aniline has been incorpo-
rated conductive nanofiber pathways throughout the hydrogel matrix. The nanocom-
posites were characterized through techniques such as 1 H NMR, TGA, AFM, SEM,
FTIR, UV–vis cyclic voltametry and conductivity estimations. In vitro conductive
stimuli drug release of hydrocortisone as model drug was evaluated. Any cytotox-
icity was not observed for conductive and non-conductive hydrogels. According to
findings, the fabricated nanoconstruct acts as precise externally controlled DDS that
may be customized to meet the physiological processes requirement (Table 2).
5 Conclusion
References
1. Rosen, H., Abribat, T.: The rise and rise of drug delivery. Nat. Rev. Drug Discov. 4, 381–385
(2005). https://doi.org/10.1038/nrd1721
2. Verma, D., Gulati, N., Kaul, S., Mukherjee, S., Nagaich, U.: Protein based nanostructures for
drug delivery. J. Pharm. 9285854 (2018). https://doi.org/10.1155/2018/9285854
3. Jao, D., Xue, Y., Medina, J., Hu, X.: Protein-based drug-delivery materials. Materials (Basel)
10(5), 517 (2017). https://doi.org/10.3390/ma100505517
4. Singh, R., Lillard, J.W.: Nanoparticle-based targeted drug delivery. Exp. Mol. Pathol. 86(3),
215–223 (2009). https://doi.org/10.1016/j.yexmp.2008.12.004
5. Guo, D., Xie, G., Luo, J.: Mechanical properties of nanoparticles: basics and applications. J.
Phys. D: Appl. Phys. 47(1) (2014). https://doi.org/10.1088/0022-3727/47/1/013001
6. DeFrates, K., Markiewicz, T., Gallo, P., Rack, A., Weyhmiller, A., Jarmusik, B., et al.: Protein
polymer-based nanoparticles: fabrication and medical applications. Int. J. Mol. Sci. 19(6),
1717 (2018). https://doi.org/10.3390/ijms19061717
7. Cai, R., Chen, C.: The crown and the scepter: roles of the protein corona in nanomedicine.
Adv. Mater. 31(45), 1805740 (2019). https://doi.org/10.1002/adma.201805740
8. Ovais, M., Guo, M., Chen, C.: Tailoring nanomaterials for targeting tumor associated
macrophages. Adv. Mater. 31(19), 1808303 (2019). https://doi.org/10.1002/adma.201808303
9. Aljabali, A.A.A., Barclay, J.E., Lomonossoff, G.P., Evans, D.J.: Virus templated metallic
nanoparticles. Nanoscale 2(12), 2596–2600 (2010). https://doi.org/10.1039/C0NR00525H
10. Sainsbury, F., Saunders, K., Aljabali, A.A.A., Evans, D.J., Lomonossoff, G.P.: Peptide-
controlled access to the interior surface of empty virus nanoparticles. ChemBioChem 12(16),
2435–2440 (2011). https://doi.org/10.1002/cbic.201100482
An Insight into Collagen-Based Nano Biomaterials for Drug Delivery … 421
11. Albu, M.G., Titorencu, I., Ghica, M.V.: Collagen-based drug delivery systems for tissue
engineering. In: Pignatello, R. (ed.) Biomaterials Applications for Nanomedicine. Inte-
chOpen 333–358 (2011). http://www.intechopen.com/books/biomaterialsapplications-for-
anomedicine/collagen-based-drug-delivery-systems-for-tissue-engineering
12. Zhang, Y., Sun, T., Jiang, C.: Biomacromolecules as carriers in drug delivery and tissue
engineering. Acta Pharm. Sin. B 8(1), 34–50 (2018). https://doi.org/10.1016/j.apsb.2017.
11.005
13. Ahmed, M., Verma, A.K., Patel, R.: Collagen extraction and recent biological activities of
collagen peptides derived from sea-food waste: a review. Sustain. Chem. Pharm. 18, 100315
(2020). https://doi.org/10.1016/j.scp.2020.100315
14. Daza, J.H.U., Righetto, G.M., Chaud, M.V., Martins, V.C.A., Camargo, I.L.B.C., Plepis,
A.M.G.: PVA/anionic collagen membranes as drug carriers of ciprofloxacin hydrochloride
with sustained antibacterial activity and potential use in the treatment of ulcerative keratitis.
J. Biomater. Appl. 35(3), 301–312 (2020). https://doi.org/10.1177/0885328220931733
15. Geanaliu-Nicolae, R.-E., Andronescu, E.: Blended natural support materials-collagen based
hydrogels used in biomedicine. Materials 13(24), 5641 (2020). https://doi.org/10.3390/ma1
3245641
16. Maham, A., Tang, Z., Wu, H., Wang, J., Lin, Y.: Protein-based nanomedicine platforms for
drug delivery. Small 5, 1706–1721 (2009). https://doi.org/10.1002/smll.200801602
17. Lo, S., Fauzi, M.B.: Current update of collagen nanomatrials-fabrication, characterisation and
its applications: a review. Pharmaceutics 13(3), 316 (2021). https://doi.org/10.3390/pharma
ceutics13030316
18. Makkithaya, K.N., Nadumane, S., Zhuo, G.Y., Chakrabarty, S., Mazumdar N. Nanoparticle
based collagen biomaterials for wound healing. In: Mazumdar, N. (ed.) Collagen biomaterials.
IntechOpen (2022). https://doi.org/10.5772/intechopen.104851
19. Bhushan, B. (ed.): Springer Handbook of Nanotechnology. Springer, Berlin (2007)
20. Mitchell, M.J., Billingsley, M.M., Haley, R.M., Wechsler, M.E., Peppas, N.A., Langer, R.:
Engineering precision nanoparticles for drug delivery. Nat. Rev. Drug Discov. 20(2), 101–124
(2021). https://doi.org/10.1038/s41573-020-0090-8
21. Naskar, A., Kim, K.S.: Recent advances in nanomaterial-based wound-healing therapeutics.
Pharmaceutics 12(6), 499, 1–20 (2020). https://doi.org/10.3390/pharmaceutics12060499
22. Wilson, A.: The formation of dry, wet, spunlaid and other types of nonwovens. In: Applications
of Nonwovens in Technical Textiles, pp. 3–17 (2010). https://doi.org/10.1533/978184569974
1.1.3
23. Wang, X., Yan, Y., Yost, M.J., Fann, S.A., Dong, S., Li, X.: Nanomechanical characterization
of micro/nanofiber reinforced type 1 collagens. J. Biomed. Mater. Res. Part A 83A, 130–135
(2007). https://doi.org/10.1002/jbm.a.31207
24. Arun, A., Malrautu, P., Laha, A., Luo, H., Ramakrishna, S.: Collagen nanoparticles in drug
delivery systems and tissue engineering. Appl. Sci. 11(23), 11369 (2021). https://doi.org/10.
3390/app112311369
25. Chak, V., Kumar, D., Visht, S.: A review on collagen based drug delivery systems. Int. J.
Pharm. Teach. Pract. 4, 811–820 (2013)
26. Musazzi, U.M., Franzè, S., Minghetti, P., Casiraghi, A.: Emulsion versus nanoemulsion: how
much is the formulative shift critical for a cosmetic product? Drug Deliv. Transl. Res. 8,
414–421 (2017). https://doi.org/10.1007/s13346-017-0390-7
27. Singh, A.N., Yethiraj, A.: Liquid–liquid phase separation as the second step of complex
coacervation. J. Phys. Chem. B 125(12), 3023–3031 (2021). https://doi.org/10.1021/acs.jpcb.
0c07349
28. Singh, Y., Meher, J.G., Raval, K., Khan, F.A., Chaurasia, M., Jain, N.K., et al.: Nanoemulsion:
concepts, development and applications in drug delivery. J. Control. Release 252, 28–49
(2017). https://doi.org/10.1016/j.jconrel.2017.03.008
29. Tan, S.F., Masoumi, H.R., Karjiban, R.A., Stanslas, J., Kirby, B.P., Basri, M., et al.: Ultra-
sonic emulsification of parenteral valproic acid-loaded nanoemulsion with response surface
methodology and evaluation of its stability. Ultrason. Sonochem. 29, 299–308 (2016). https://
doi.org/10.1016/j.ultsonch.2015.09.015
422 A. K. Verma
30. Dehghani, F., Farhadian, N., Golmohammadzadeh, S., Biriaee, A., Ebrahimi, A., Karimi, M.:
Preparation, characterization and in vivo evaluation of microemulsions containing tamoxifen
citrate anti-cancer drug. Eur. J. Pharm. Sci. 96, 479–489 (2017). https://doi.org/10.1016/j.
ejps.2016.09.033
31. Gurpreet, K., Singh, S.K.: Review of Nanoemulsion Formulation and Characterization Tech-
niques. Indian J. Pharm. Sci. 80, 781–789 (2018). https://doi.org/10.4172/pharmaceutical-sci
ences.1000422
32. Souto, E.B., Fernandes, A.R., Martin-Gomes, C., Coutino, T.E., Durazzo, A., Lucarini, M.,
et al.: Nanomaterials for skin delivery of cosmeceuticls and pharmaceuticals. Appl. Sci. 10(5),
1594 (2020). https://doi.org/10.3390/app10051594
33. Katz, L.M., Dewan, K., Bronaugh, R.L.: Nanotechnology in cosmetics. Appl. Sci. 10(5), 1594
(2020). https://doi.org/10.3390/app10051594
34. Feng, L., Zhu, C., Yuan, H., Liu, L., Lv, F., Wang, S.: Conjugated polymer nanoparticles:
Preparation, properties, functionalization and biological applications. Chem. Soc. Rev. 42,
6620–6633 (2013). https://doi.org/10.1039/C3CS60036J
35. Hong, S., Choi, D.W., Kim, H.N., Park, C.G., Lee, W.L., Park, H.H.: Protein-based nanopar-
ticles as drug delivery systems. Pharmaceutics 12(7), 604 (2020). https://doi.org/10.3390/pha
rmaceutics12070604
36. Truong-Le, V.L., August, J.T., Leong, K.W.: Controlled gene delivery by DNA–gelatin
nanospheres. Hum. Gene Ther. 9, 1709–1717 (1998). https://doi.org/10.1089/hum.1998.9.
12-1709
37. Yoon, J., Kwag, J., Shin, T.J., Park, J., Lee, Y.M., Lee, Y., et al.: Nanoparticles of conjugated
polymers prepared from phase-separated films of phospholipids and polymers for biomedical
applications. Adv. Mater. 26, 4559–4564 (2014). https://doi.org/10.1002/adma.201400906
38. Law, J.X., Liau, L.L., Saim, A., Yang, Y., Idrus, R.: Electrospun collagen nanofibers and their
applications in skin tissue engineering. Tissue Eng. Regen. Med. 14(6), 699–718 (2017).
https://doi.org/10.1007/s13770-017-0075-9
39. Xue, J., Wu, T., Dai, Y., Xia, Y.: Electrospinning and electrospun nanofibers: methods, mate-
rials, and applications. Chem. Rev. 119(8), 5298–5415 (2019). https://doi.org/10.1021/acs.
chemrev.8b00593
40. Azimi, B., Maleki, H., Zavagna, L., De la Ossa, J.G., Linari, S., Lazzeri, A., Danti, S.: Bio-
based electrospun fibers for wound healing. J. Funct. Biomater. 11(3), 67 (2020). https://doi.
org/10.3390/jfb11030067
41. Ghorbani, S., Eyni, H., Tiraihi, T., Asl, L.S., Soleimani, M., Atashi, A., et al.: Combined
effects of 3D bone marrow stem cell-seeded wet-electrospun poly lactic acid scaffolds on
full-thickness skin wound healing. Int. J. Polym. Mater. Polym. Biomater. 67(15), 905–912
(2018). https://doi.org/10.1080/00914037.2017.1393681
42. Mbese, Z., Alven, S., Aderibigbe, B.A.: Collagen-based nanofibers for skin regeneration and
wound dressing applications. Polymers (Basel) 13(24), 4368 (2021). https://doi.org/10.3390/
polym13244368
43. Haggag, Y.A., Faheem, A.M.: Evaluation of nano spray drying as a method for drying and
formulation of therapeutic peptides and proteins. Front. Pharmacol. 6, 140 (2015). https://doi.
org/10.389/fphar.2015.00140
44. Lee, S.H., Heng, D., Ng, W.K., Chan, H.K., Tan, R.B.: Nano spray drying: a novel method
for preparing protein nanoparticles for protein therapy. Int. J. Pharm. 403, 192–200 (2011).
https://doi.org/10.1016/j.ijpharm.2010.10.012
45. Gulfam, M., Kim, J.E., Lee, J.M., Ku, B., Chung, B.H.: Anticancer drug-loaded gliadin
nanoparticles induce apoptosis in breast cancer cells. Langmuir 28, 8216–8223 (2012). https://
doi.org/10.1021/la300691n
46. Boda, S.K., Li, X., Xie, J.: Electrospraying an enabling technology for pharmaceutical and
biomedical applications: a review. J. Aerosol Sci. 125, 164–181 (2018). https://doi.org/10.
1016/j.jaerosci.2018.04.002
47. Nagarajan, U., Kawakami, K., Zhang, S., Chandrasekaran, B., Nair, B.U.: Fabrication of
solid collagen nanoparticles using electrospray deposition. Chem. Pharm. Bull. 62, 422–428
(2014). https://doi.org/10.1248/cpb.c13-01004
An Insight into Collagen-Based Nano Biomaterials for Drug Delivery … 423
48. Kempf, M., Miyamura, Y., Liu, P.Y., Chen, A.C., Nakamura, H., Shimizu, H., et al.: A dena-
tured collagen microfiber scaffold seeded with human fibroblasts and keratinocytes for skin
grafting. Biomaterials 32, 4782–4792 (2011). https://doi.org/10.1016/j.biomaterials.2011.
03.023
49. Mhetar, S.K., Ashok, N.A., Patil, R.L., Pawar, R.A., Patil, M.M., Shinde, H.T.: Cost effective
ball milling machine for producing nanopowder. Int. Res. J. Eng. Technol. 4, 330–334 (2017)
50. Kumar, M., Xiong, X., Wan, Z., Sun, Y., Tsang, D.C., Gupta, J., et al.: Ball milling as a
mechanochemical technology for fabrication of novel biochar nanomaterials. Biores. Technol.
312, 123613 (2020). https://doi.org/10.1016/j.biortech.2020.123613
51. Weber, C., Coester, C., Kreuter, J., Langer, K.: Desolvation process and surface character-
isation of protein nanoparticles. Int. J. Pharm. 194, 91–102 (2000). https://doi.org/10.1016/
S0378-5173(99)00370-1
52. Kandamachira, A., Selvam, S., Marimuthu, N., Kalarical, S.J., Nishter, N.F.: Collagen-
nanoparticle interactions: type I collagen stabilization using functionalized nanoparticles.
Soft Mater. 13, 59–65 (2014). https://doi.org/10.1080/1539445X.2015.1009550
53. Choi, D., Heo, J., Park, J.H., Jo, Y., Jeong, H., Chang, M., et al.: Nano-film coatings onto
collagen hydrogels with desired drug release. J. Ind. Eng. Chem. 36925, 326–333 (2016).
https://doi.org/10.1016/j.jiec.2016.02.023
54. Liu, G., Li, L., Huo, D., Li, Y., Wu, Y., Zeng, L., et al.: A VEGF delivery system targeting
MI improves angiogenesis and cardiac function based on the tropism of MSCs and layer-
by-layer self-assembly. Biomaterials 127, 117–131 (2017). https://doi.org/10.1016/j.biomat
erials.2017.03.001
55. Jeong, H., Hep, J., Son, B., Choi, D., Park, T.H., Chang, M., et al.: Intrinsic hydrophobic
cairnlike multilayer films for antibacterial effect with enhanced durability. ACS Appl. Mater.
Interfaces 7(47), 26117–26123 (2015). https://doi.org/10.1021/acsami.5b07613
56. Morgan, P.W.: Interfacial polymerization. In: Encylopedia of Polymer Science and Tech-
nology. Wiley, New York (2011). https://doi.org/10.1002/0471440264.pst168
57. Raaijmakers, M.J.T., Benes, N.E.: Current trends in interfacial polymerization chemistry.
Prog. Polym. Sci. 63, 86–142 (2016). https://doi.org/10.1016/j.progpolymsci.2016.06.004
58. Song, Y., Fan, J.B., Wang, S.: Recent progress in interfacial polymerization. Mater. Chem.
Front. 1, 1028–1040 (2017). https://doi.org/10.1039/C6QM00325G
59. Prasher, A., Loynd, C.M., Tuten, B.T., Frank, P.G., Chao, D., Berda, E.B.: Efficient fabrication
of polymer nanoparticles via sonogashira cross-linking of linear polymers in dilute solution.
J. Polym. Sci. Part A: Polym. Chem. 54, 209–217 (2015). https://doi.org/10.1002/pola.27942
60. Kröger, A.P.P., Hamelmann, N.M., Juan, A., Lindhoud, S., Paulusse, J.M.J.: Biocompatible
single-chain polymer nanoparticles for drug delivery-a dual approach. ACS Appl. Mater.
Interfaces 10, 30946–30951 (2018). https://doi.org/10.1021/acsami.8b07450
61. Hanlon, A.M., Chen, R., Rodriguez, K., Willis, C., Dickinson, J.G., Cashman, M., et al.:
Scalable synthesis of single-chain nanoparticles under mild conditions. Macromolecules 50,
2996–3003 (2017). https://doi.org/10.1021/acs.macromol.7b00497
62. Verso, F.L., Pomposo, J.A., Colmenero, J., Moreno, A.J.: Multi-orthogonal folding of single
polymer chains into soft nanoparticles. Soft Matter 10, 4813–4821 (2014). https://doi.org/10.
1039/C4SM00459K
63. Wang, G., Uludag, H.: Recent developments in nanoparticle-based drug delivery and targeting
systems with emphasis on protein-based nanoparticles. Expert Opin. Drug Deliv. 5, 499–515
(2008). https://doi.org/10.1517/17425247.5.5.499
64. Komlosh, A., Weinstein, V., Loupe, P., Hasson, T., Timan, B., Konya, A., et al.: Physico-
chemical and biological examination of two glatiramer acetate products. Biomedicines 7, 49
(2019). https://doi.org/10.3390/biomedicines7030049
65. Posadas, I., Monteagudo, S., Ceña, V.: Nanoparticles for brain-specific drug and genetic
material delivery, imaging and diagnosis. Nanomedicine 11, 833–849 (2016). https://doi.org/
10.2217/nnm.16.15
66. Patra, J.K., Das, G., Fraceto, L.F., Campos, E.V.R., Rodriguez-Torres, M.D.P., Acosta-Torres,
L.S., et al.: Nano based drug delivery systems: recent developments and future prospects. J.
Nanobiotechnology 16, 71 (2018). https://doi.org/10.1186/s12951-018-0392-8
424 A. K. Verma
67. Akhmetova, A., Heinz, A.: Electrospinning proteins for wound healing purposes: opportuni-
ties and challenges. Pharmaceutics 13(1), 4 (2021). https://doi.org/10.3390/pharmaceutics13
010004
68. Oliveira, A.M., Guimarães, K.L., Cerize, N.N., Tunussi, A.S., Poço JG. Nano spray drying
as an innovative technology for encapsulating hydrophilic active pharmaceutical ingredients
(API). J. Nanomedicine Nanotechnol. 4 (2013). https://doi.org/10.4172/2157-7439.1000186
69. Kim, M.T., Chen, Y., Marhoul, J., Jacobson, F.: Statistical modeling of the drug load distribu-
tion on trastuzumab emtansine (Kadcyla), a lysine-linked antibody drug conjugate. Bioconjug.
Chem. 25, 1223–1232 (2014). https://doi.org/10.1021/bc5000109
70. Bock, N., Woodruff, M.A., Hutmacher, D.W., Dargaville, T.R.T.R.: Electrospraying, a repro-
ducible method for production of polymeric microspheres for biomedical applications.
Polymers 3, 131–149 (2011). https://doi.org/10.3390/polym3010131
71. Chelle, P., Yeung, C.H.T., Croteau, S.E., Lissick, J., Balasa, V., Ashburner, C., et al.: Devel-
opment and validation of a population-pharmacokinetic model for Rurioctacog Alfa Pegol
(Adynovate®): a report on behalf of the WAPPS-Hemo Investigators Ad Hoc Subgroup. Clin.
Pharmacokinet. 59, 245–256 (2020). https://doi.org/10.1007/s40262-019-00809-6
72. Lohcharoenkal, W., Wang, L., Chen, Y.C., Rojanasakul, Y.: Protein nanoparticles as drug
delivery carriers for cancer therapy. Biomed. Res. Int. 180549, 1–12 (2014). https://doi.org/
10.1155/2014/180549
73. Zhang, X., Wang, D., Zhou, Z., Ma, Y.: Robust low-rank tensor recovery with rectification
and alignment. IEEE Trans. Pattern Anal. Mach. Intell. 43(1), 238–255 (2021). 10.1109/
TPAMI.2019.29290
74. Garg, A., Visht, S., Sharma, P.K., Kumar, N.: Formulation, characterization and application
on nanoparticle: a review. Der Pharm. Sin. 2(2), 17–26 (2011)
75. Kaszuba, M., Corbett, J., Watson, F.M., Jones, A.: High-concentration zeta potential measure-
ments using light-scattering techniques. Philos. Trans. R. Soc. A 2010(368), 4439–4451
(1927). https://doi.org/10.1098/rsta.2010.0175
76. Mistry, A., Glud, S.Z., Kjems, J., Randel, J., Howard, K.A., Stolnik, S., et al.: Effect of
physicochemical properties on intranasal nanoparticle transit into murine olfactory epithelium.
J. Drug Target 17, 543–552 (2009). https://doi.org/10.1080/10611860903055470
77. Nitta, S.K., Numata, K.: Biopolymer-based nanoparticles for drug/gene delivery and tissue
engineering. Int. J. Mol. Sci. 14, 1629–1654 (2013). https://doi.org/10.3390/ijms14011629
78. Shalaby, M., Ghareeb, A.Z., Khedra, S.M., Mostafa, H.M., Saeed, H., Hamouda, D.: Nanopar-
ticles of bioactive natural collagen for wound healing: experimental approach (2023). https://
doi.org/10.1101/2023.02.21.529363
79. Benyettou, F., Rezgui, R., Ravaux, F., Jaber, T., Blumer, K., Jouiad, M.: Synthesis of silver
nanoparticles for the dual delivery of doxorubicin and alendronate to cancer cells. J. Mater.
Chem. B 2015(3), 7237–7245 (2015). https://doi.org/10.1039/C5TB00994D
80. Yildirimer, L., Thanh, N.T., Loizidou, M., Seifalian, A.M.: Toxicology and clinical potential of
nanoparticles. Nano Today 6, 585–607 (2011). https://doi.org/10.1016/j.nantod.2011.10.001
81. Subha, V., Kirubanandan, S., Ilangovan, R., Renganathan, S.: Silver nanoparticles impreg-
nated nanocollagen as scaffold for soft tissue repair-synthesis, characterization, and in vitro
investigation. Int. J. Med. Nano Res. 8(1), 1–9 (2021). https://doi.org/10.23937/2378-3664.
1410034
82. Cardoso, V.S., Quelemes, P.V., Amorin, A., Primo, F.L., Gobo, G.G., Tedesco A C. et al.:
Collagen-based silver nanoparticles for biological applications: synthesis and characteriza-
tion. J. Nanobiotechnology 12, 36 (2014). https://doi.org/10.1186/s12951-014-0036-6
83. Roshanbinfar, K., Kolesnik-Gray, M., Angeloni, M., Schruefer, S., Fiedler, M., Schubert, D.
et al.: Collagen hydrogel containing polyethylenimine-gold nanoparticles for drug release
and enhanced beating properties of engineered cardiac tissues. Adv. Healthc. Mater. 2202408
(2023). https://doi.org/10.1002/adhm.202202408
84. Terzopoulou, Z., Michopoulou, A., Palamidi, A., Koliakou, E., Bikiaris, D.: Preparation and
evaluation of collagen-based patches as curcumin carriers. Polymers (Basel) 12(10), 2393
(2020). https://doi.org/10.3390/polym12102393
An Insight into Collagen-Based Nano Biomaterials for Drug Delivery … 425
85. Mohanraj, V., Chen, Y.: Nanoparticles-a review. Trop. J. Pharm. Res. 5, 561–573 (2006).
https://doi.org/10.4314/tjpr.v5i1.14634
86. Hoshyar, N., Gray, S., Han, H., Bao, G.: The effect of nanoparticle size on in vivo pharma-
cokinetics and cellular interaction. Nanomedicine 11(6), 673–692 (2016). https://doi.org/10.
2217/nnm.16.5
87. McSweeney, M.D., Wessler, T., Price, L.S.L., Ciociola, E.C., Herity, L.B., Piscitelli, J.A.,
et al.: A minimal physiologically based pharmacokinetic model that predicts anti-PEG IgG-
mediated clearance of PEGylated drugs in human and mouse. J. Control. Release 284, 171–178
(2018). https://doi.org/10.1016/j.jconrel.2018.06.002
88. Breitenbach, M., Kamm, W., Hungere, K., Hund, H., Kissel, T.: Oral and nasal administration
of tetanus toxoid loaded nanoparticles consisting of novel charged biodegradable polyesters
for mucosal vaccination. In: Proceedings of the International Symposium on Controlled
Release of Bioactive Materials, vol. 26, pp. 348–349 (1999)
89. Zhang, L., Beatty, A., Lu, L., Abdalrahman, A., Makris, T.M., Wang, G., et al.: Microfluidic-
assisted polymer-protein assembly to fabricate homogeneous functional nanoparticles. Mater.
Sci. Eng. C 111, 110768 (2020). https://doi.org/10.1016/j.msec.2020.110768
90. Knight, F.C., Gilchuk, P., Kumar, A., Becker, K.W., Sevimli, S., Jacobson, M.E., et al.:
Mucosal immunization with a pH-responsive nanoparticle vaccine induces protective CD8+
lung-resident memory T cells. ACS Nano 13(10), 10939–11096 (2019). https://doi.org/10.
1021/acsnano.9b00326
91. Strand, M.S., Krasnick, B.A., Pan, H., Zhang, X., Bi, Y., Brooks, C., et al.: Precision delivery
of RAS-inhibiting siRNA to KRAS driven cancer via peptide-based nanoparticles. Oncotarget
10(46), 4761–4775 (2019). https://doi.org/10.18632/oncotarget.27109
92. Afsharzadeh, M., Hashemi, M., Mokhtarzadeh, A., Abnous, K., Ramezani, M.: Recent
advances in co-delivery systems based on polymeric nanoparticle for cancer treatment.
Artif. Cells Nanomedicine Biotechnol. 46(6), 1095–1110 (2018). https://doi.org/10.1080/
21691401.2017.1376675
93. Volpatti, L.R., Matranga, M.A., Cortinas, A.B., Delcassian, D., Daneil, K.B., Langer, R.,
Anderson, D.G. et al.: Glucose-responsive nanoparticles for rapid and extended self-regulated
insulin delivery. ACS Nano 14(1), 488–497 (2020). https://doi.org/10.1021/acsnano.9b06395
94. Niu, Y., Yu, M., Hartono, S.B., Yang, J., Xu, H., Zhang, H., et al.: Nanoparticles mimicking
viral surface topography for enhanced cellular delivery. Adv. Mater. 25, 6233–6237 (2013).
https://doi.org/10.1002/adma.201302737
95. Awad, A., Shalaby, M., Batiha, G., Mady, R., Al-kuraishy, H., Shaheen, H.M.: Cytotox-
icity effect assessment of theophylline loaded with collagen nanoparticles. Damanhour J.
Veterinary Sci. 8(2), 5–10 (2022). https://doi.org/10.21608/djvs.2022.140639.1073
96. Rathore, P., Arora, I., Rastogi, S., Akhtar, M., Singh, S., Samim, M.: Collagen–curcumin
nanocomposites showing an enhanced neuroprotective effect against short term focal cerebral
ischemia. RSC Adv. 10(4), 2241–2253 (2020). https://doi.org/10.1039/C9RA08508D
97. Anwar, M.M., Shalaby, M.A., Saeed, H., Mostafa, H.M., Hamouda, D.G., Nounou, H.:
Theophylline-encapsulated Nile Tilapia fish scale-based collagen nanoparticles effectively
target the lungs of male Sprague–Dawley rats. Sci. Rep. 12(1), 1–12 (2022).
98. Soppimath, K.S., Aminabhavi, T.M., Kulkarni, A.R., Rudzinski, W.E.: Biodegradable poly-
meric nanoparticles as drug delivery devices. J. Control. Release 70, 1–20 (2001). https://doi.
org/10.1016/S0168-3659(00)00339-4
99. Pritchard, E.M., Hu, X., Finley, V., Kuo, C.K., Kaplan, D.L.: Effect of silk protein processing
on drug delivery from silk films. Macromol. Biosci. 13, 311–320 (2013). https://doi.org/10.
1002/mabi.201200323
100. Champion, J.A., Katare, Y.K., Mitragotri, S.: Particle shape: a new design parameter for micro-
and nanoscale drug delivery carriers. J. Control. Release 121, 3–9 (2007). https://doi.org/10.
1016/j.jconrel.2007.03.022
101. Kundu, B., Soundrapandian, C., Nandi, S.K., Mukherjee, P., Dandapat, N., Roy, S.,
et al.: Development of new localized drug delivery system based on ceftriaxone-sulbactam
composite drug impregnated porous hydroxyapatite: a systematic approach for in vitro and
426 A. K. Verma
118. Bettini, S., Bonfrate, V., Syrgiannis, Z., Sannino, A., Salvatore, L., Madaghiele, M., et al.:
Biocompatible collagen paramagnetic scaffold for controlled drug release. Biomacromol
16(9), 2599–2608 (2015). https://doi.org/10.1021/acs.biomac.5b00829
119. Choi, M., Chung, J.H., Cho, Y., Hong, B.Y., Hong, J.: Nano-film modification of collagen
hydrogels for controlled growth factor release. Chem. Eng. Sci. 137, 626–630 (2015). https://
doi.org/10.1016/j.ces.2015.07.011
120. Minakuchi, Y., Takeshita, F., Kosaka, N., Sasaki, H., Yamamoto, Y., Kouno, M., et al.:
Atelocollagen-mediated synthetic small interfering RNA delivery for effective gene silencing
in vitro and in vivo. Nucleic Acids Res. 32(13), e109 (2004). https://doi.org/10.1093/nar/
gnh093
121. Ramírez, O.J., Alvarez, S., Contreras-Kallens, P., Barrera, N.P., Aguayo, S., Schuh, C.M.A.P.:
Type I collagen hydrogels as a delivery matrix for royal jelly derived extracellular vesicles.
Drug Deliv. 27(1), 1308–1318 (2020). https://doi.org/10.1080/10717544.2020.1818880
122. Wang, Y., Zhang, C.L., Zhang, Q., Li, P.: Composite electrospun nanomembranes of fish scale
collagen peptides/chito-oligosaccharides: antibacterial properties and potential for wound
dressing. Int. J. Nanomed. 6, 667–676 (2011). https://doi.org/10.2147/IJN.S17547
123. Liu, S.J., Kau, Y.C., Chou, C.Y., Chen, J.K., Wu, R.C., Yeh, W.L.: Electrospun PLGA/collagen
nanofibrous membrane as early-stage wound dressing. J. Membr. Sci. 355(1–2), 53–59 (2010).
https://doi.org/10.1016/j.memsci.2010.03.012
124. Mondal, S., Hoang, G., Manivasagan, P., Moorthy, M.S., Phan, T.T.V., Kim, H.H., et al.: Rapid
microwave-assisted synthesis of gold loaded hydroxyapatite collagen nano-bio materials for
Drug Deliv. and tissue engineering application. Ceram. Int. 45(3), 2977–2988 (2019). https://
doi.org/10.1016/j.ceramint.2018.10.016
125. Wei, D., Sun, J., Yang, Y., Wu, C., Chen, S., Guo, Z., et al.: Cell alignment guided by nano/
micro oriented collagen fibers and the synergistic vascularization for nervous cell functional
expression. Mater. Today Chem. 8, 85–95 (2018). https://doi.org/10.1016/j.mtchem.2018.
03.001
126. Mohamadi, F., Ebrahimi-Barough, S., Nourani, M.R., Derakhshan, M.A., Goodarzi, V.,
Nazockdast, M.S., et al.: Electrospun nerve guide scaffold of poly (ε-caprolactone)/collagen/
nanobioglass: an in vitro study in peripheral nerve tissue engineering. J. Biomed. Mater. Res.
Part A 105A, 1960–1972 (2017). https://doi.org/10.1002/jbm.a.36068
127. Ghavimi, M.A., Shahabadi, A.B., Jarolmasjed, S., Memar, M.Y., Dizaj, S.M., Sharifi, S.:
Nanofibrous asymmetric collagen/curcumin membrane containing aspirin-loaded PLGA
nanoparticles for guided bone regeneration. Sci. Rep. 10, 18200 (2020). https://doi.org/10.
1038/s41598-020-75454-2
128. Ahmadian, S., Ghorbani, M., Mahmoodzadeh, F.: Silver sulfadiazine-loaded electrospun ethyl
cellulose/polylactic acid/collagen nanofibrous mats with antibacterial properties for wound
healing. Int. J. Biol. Macromol. 162, 1555–1565 (2020). https://doi.org/10.1016/j.ijbiomac.
2020.08.059
129. Lee, C.H., Chang, S.H., Chen, W.J., Hung, K.C., Lin, Y.H., Liu, S.J., et al.: Augmentation of
diabetic wound healing and enhancement of collagen content using nanofibrous glucophage-
loaded collagen/PLGA scaffold membranes. J. Colloid Interface Sci. 439, 88–97 (2015).
https://doi.org/10.1016/j.jcis.2014.10.028
130. Lai, H.J., Kuan, C.h., Wu, H.C., Tsai, J.C., Chen, T.M., Hseish DJ et al. Tailored design of
electrospun composite nanofibers with staged release of multiple angiogenic growth factors
for chronic wound healing. Acta Biomater. 10(10), 4156-4166 (2014). https://doi.org/10.1016/
j.actbio.2014.05.001
131. Kandhasamy, S., Perumal, S., Madhan, B., Umamaheswari, N., Banday, J.A., Perumal, P.T.,
et al.: Synthesis and fabrication of collagen-coated ostholamide electrospun nanofiber scaffold
for wound healing. ACS Appl. Mater. Interfaces 9(10), 8556–8568 (2017). https://doi.org/10.
1021/acsami.6b16488
132. Hou, J., Chen, L., Zhou, M., Li, J., Liu, J., Fang, H., et al.: Multi-layered polyamide/collagen
scaffolds with topical sustained release of N-acetylcysteine for promoting wound healing. Int.
J. Nanomed. 15, 1349–1361 (2020). https://doi.org/10.2147/IJN.S232190
428 A. K. Verma
133. Yao, C.H., Chen, K.Y., Chen, Y.S., Li, S.J., Huang, C.H.: Lithospermi radix extract-containing
bilayer nanofiber scaffold for promoting wound healing in a rat model. Mater. Sci. Eng. C 96,
850–858 (2019). https://doi.org/10.1016/j.msec.2018.11.053
134. Tort, S., Acartürk, F., Beşikci, A.: Evaluation of three-layered doxycycline-collage loaded
nanofiber wound dressing. Int. J. Pharm. 529(1–2), 642–653 (2017). https://doi.org/10.1016/
j.ijpharm.2017.07.027
135. Khodir, W.K.W.A., Razak, A.H.A., Ng, M.H., Guarino, V., Susanti, D.: Encapsulation and
characterization of gentamicin sulfate in the collagen added electrospun nanofibers for skin
regeneration. J. Funct. Biomater. 9(2), 36 (2018). https://doi.org/10.3390/jfb9020036
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 429
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_14
430 Amisha et al.
Abbreviations
AM Additive Manufacturing
3D Three Dimensional
4D Four Dimensional
4DP Four Dimensional Printing
DLP Digital Light Processing
FDM Fused Deposition Modelling
SLS Selective Laser Sintering
IJP Inkjet Printing
SLA Stereolithography
DIW Direct Ink Writing
BTE Bone Tissue Engineering
SMs Stimuli-responsive Materials
SMMs Shape Memory Materials
SCMs Shape-Changing Materials
ABS Acrylonitrile Butadiene Styrene
PLA Polylactic Acid
PC Polycarbonate
PA Polyamide
PVB Polyvinyl Butyral
PCL Polycaprolactone
PS Polystyrenes
DOD Drop-On-Demand
NIR Near Infrared
IR Infrared
UV Ultraviolet
1 Introduction
matrix, that is vital for a biomaterial since it is used in the design and construction
of scaffolds [14]. In the recent era, photo or light has been employed as an alternate
treatment method for ailments like melanoma, depression, acne, skin problems or
tissue engineering. It is an especially enticing source of energy as compared to other
energy types, such as thermal or magnetic energy, which are frequently utilized as an
external push to govern stimuli-responsive biomaterials [15]. Firstly, modern optical
device enables light to be aimed at a particular region. Moreover, the duration or
degree of exposure to light might be employed to modulate the stimulation intensity.
Eventually, light might be segmented into a spectrum of wavelengths that can be
individually modulated via distinct biomaterials to inspire new bio-inductive stimuli
for upregulating interactions. Thus, contrasted to temperature or pH value fluctua-
tion, light is a substantially less detrimental alternative for altering biomaterials in
the physiological environment. As a corollary, light is a possible source of energy
for constructing biomaterials that respond to stimuli for tissue engineering [16, 17].
So far, 4D bioprinting methodologies are still in its inception, and experts are
continually learning about its mechanics and concepts. In this overview, we confer
several types of stimuli-responsive materials (SMs), their shape-transformation
mechanisms and their cell-inherent characteristics for 4D bioprinting and special
emphasis is provided on photo responsive biomaterials as described in the literature
at present [18]. A variety of approaches have additionally been explored to facilitate
the functionalization and maturation of tissues or cells in 3D-printed structures over
time. In addition, this chapter fuses the most recent research with potential thera-
peutic insights to prioritize on the development of tissue engineering 4D bioprinting
and incorporate its sophisticated applications in tissue regeneration. Lastly, we confer
about the foremost challenges that 4D bioprinting is facing as it develops and assess
its potential future possibilities [19, 20].
2.1 DLP
DLP printing has been demonstrated as a viable means of rapidly creating intri-
cate 3D and 4D constructions since it leverages light as a crosslinking agent. This
434 Amisha et al.
Fig. 2 Development of 4D printing year by year from the inception in various biomedical sector
approach provides an assortment of printing materials and could yield prints featuring
improved print quality, print clarity, and print contour accuracy. In contrast to SLA,
DLP cures liquid resin in a reservoir in approximately one second per layer adopting
a digital light pattern. Since a DLP printer tends to make use of a surface-projection-
based fabrication technique, it can print at high rates that are in the range of numerous
times greater than what is feasible with SLA [27, 28]. Zhang et al. (2021) fabricated
UV-curable and mechanically sturdy SMPs via DLP approach that has consider-
ably increased the mechanical characteristics of SMP-relied 4D printing scaffolds,
enabling their use in engineering applications like aerospace, intelligent furniture,
and soft robots [29].
2.2 FDM
The fundamental printing principles employed by the FDM approach are quite similar
to those used in SLA printing. The peculiarity in this process is that the substance
used to print the object is dispensed from the nozzle. Each layer cools and hardens
when it gets into the interface of the bottom layer. Generally defined, the FDM
Table 1 Current 4D printing technique for biomaterial
Printing Advantage Drawbacks Materials Applications
technique employed
Digital light High printing speed and resolution. Biocompatible with cells Inadequate tensile attributes. Shape Bone tissue
processing Time for processing is quite memory engineering,
high polymers, Cardiac tissue
photo curable engineering
resins,
Ceramics
Fused Deposition Needed no support framework. Fabricate an array of substances that are biocompatible. Designs porous Low spatial precision. High Shape Bone tissue
modelling scaffolds. High-density cells are transformed into tissues temperature demands memory engineering,
to reduced cell viability polymers, drug delivery
Polymer
composites
Selective laser Needed no support framework. Designs porous scaffolds Highly porous and extreme Metals, Bone
sintering temperature is required polymers, regeneration
ceramics and tissue
engineering
Inkjet printing Significant compatibility with bioactive compounds that have low viscosities. Producing bioink Low spatial precision. High Shape Osteochondral
materials containing organisms. Nozzle without contact temperature demands to memory tissue
reduced cell viability polymers, engineering,
hydrogel dental
implants
Photo Responsive Material for 4D Printing in Tissue Engineering
Stereolithography Smooth and fine surfaces are apparent in manufactured products. Incredibly precise method for Cytotoxic effect is induced Shape Dental
generating complex frameworks. The loading of cells and biomolecules benefited from moderate by an uncured photo memory implants and
conditions initiator. Biocompatibility polymers, bone defect
and inefficient photo-curable regeneration
decomposition of resins
photo-responsive materials.
Printing numerous material
layers is not possible
Demands enormously potent
UV rays to merge
(continued)
435
Table 1 (continued)
436
2.3 SLS
A pretty recent 3DP technique called selective laser sintering (SLS) presents novel
prospects for the manufacturing of pharmaceuticals. In order to develop incredibly
intricate scaffolds, this approach precisely sinters powder particles in conjunction
with a laser. Moreover, it prevents material waste and stimulates the recycling of
feedstock since the unsintered powder remains accessible and could be employed
again. Further, this demonstrates the technology’s affordability; in a technological
comparison, the expense of SLS was discovered to be less expensive in contrast
to SLA, FDM, and injection moulding [33]. Wu et al. (2021) manufactured the
ubiquitous robotics structure known as a gripper employing SLS-relied 4D printing
of magnetism-responsive substances, and its propensity to deform when subjected to
an exterior magnetic field was investigated. Further investigations into 4D printing
driven by magnets would elaborate on the insights. Additional exploration is vital to
enhance the gripper’s framework for better integration. The amount of energy used
while driving should also be considered [34].
One of the oldest frequently utilized 4DP techniques was inkjet printing, often
referred as drop-on-demand (DOD) printing. Since inkjet printing causes the printing
head’s ejection of photo resin droplets as a corollary of pressure and voltage pulses,
it is referred to as DOD printing. UV-curable acrylate liquid is frequently included
in inkjet printing fluids, since it quickly cures when exposed to UV light. On the
building platform, with a second inkjet printing process, an inkjet printhead that may
also move easily in 2D dispenses a liquid binder. The first pattern is lowered for the
438 Amisha et al.
application of the second powder-liquid binder layer. Each step of the powder-inkjet
printing process has to be repeated numerous times to produce the desired results [35].
The fabrication of 3D scaffolds continues to remain challenging as it’s complicated
to confine cells in 3D frameworks, and regulate cell–cell arrangement in 3D, so Cui
et al. (2020) built a framework for printing in 4D via inkjet. This technology generates
micropatterns that fold themselves into 3D. As a result, the standard 2D cell-seeding
approach is complemented by a user-friendly platform provided by 4D printing and
allows users to create a customized 3D cellularized scaffold. This research showed
that 4D printing holds great promise for tissue engineering applications [36].
2.5 SLA
Another prevalent 4DP method is SLA, which employs light (laser) emitted at various
frequencies to harden liquid resins and polymers. The technology utilizes lasers
to layer by layer cures a liquid resin to fabricate a 3D framework. Simply stated,
the reaction happens when a photo strikes the substance and the resin molecules
link up to form a solid framework. After being dried, photopolymers transform
into gels that cross-link to form solid polymers. In contrast to other methods, SLA
could start designing with an elongation of up to 80%. It is frequently utilized to
construct challenging and intricate buildings. Moreover, relevant materials for SLA
printing have been discovered [37, 38]. Zhuo et al. (2022) supported the idea of the
4D printing by the usage of poly (N-vinyl caprolactam) an adaptable polymer for
temperature, which is typically produced using radical-free polymerization. This led
to the successful preparation of 4D prints adopting SLA 3D printer. Under changing
temperatures, these prints demonstrated adaptive and reversible expansion/shrinkage
characteristics [39].
2.6 Micro-Extrusion
Among the most popular forms of additive manufacturing processes, known as micro
extrusion bioprinting, constructs layer-by-layer cell-laden frameworks by depositing
cell-laden bio-inks in forging threads, filaments, or droplets via a tip or syringe. In
order to dispense the biomaterial from the nozzle or syringe, micro extrusion uses
force. A broad spectrum of substances with varying viscosities could be printed
employing micro extrusion bioprinters. The viscosity and characteristics of the bio-
ink have a significant impact on the printing’s resolution and accuracy. However,
additional factors should be considered, namely dispensing pressure or mechanical
force, printing speed and distance. Micro extrusion bioprinting offers the benefit
of allowing for the regulated fabrication of architectures with high cell densities
in physiological circumstances [40, 41]. It is still challenging to execute 3D bio
printing of living cellular entities featuring variability in cell types and extracellular
Photo Responsive Material for 4D Printing in Tissue Engineering 439
matrices [ECMs) that are analogous to those of biological tissues. Thus, Wu et al.
(2020) demonstrated how micro extrusion-based (ME) bioprinting techniques could
substantially resolve this issue through the design of bio-ink materials. Ultimately,
these findings demonstrated that complex structures comprising various cell types
and ECMs might be bio-printed with potential applications in translational tissue
engineering alongside basic biomedical research [42].
2.7 DIW
Viscoelastic inks are dispensed via needles adopting this extrusion-based technique
at high pressure to generate dynamic, intelligent structures in DIW. Biodegradable
scaffolds have been constructed using this approach for tissue engineering purposes.
UV-assisted DIW was employed to print self-healing, highly flexible SMPs for wound
healing applications and biomedical repair devices [43]. Weng et al. (2021) offered an
approach for constructing self-morphing frameworks with high modulus (4.8 GPa)
and substantial deformation. Employing composite inks with a high-volume propor-
tion of fumed silica, solvent, short glass fibres, and photocurable polymer resin, the
frameworks are printed utilising multi-material DIW. This technique for generating
composite framework with programmable topologies and superior mechanical char-
acteristics suggests possible uses in the transformation of lightweight structures into
load-bearing structures [44].
A light cue might be employed to alter the dimension or morphology of the structures
utilizing photo-responsive materials, providing volume transformations, contrac-
tion, and bending. Externally transmitted optical signals could be captured by
photo-responsive materials and translated into mechanical responses. Near-infrared
(NIR), infrared (IR), and ultraviolet (UV) areas are among the light wavelength
ranges that could activate photo-responsive biomaterials, that are widely used in
biomedical applications including TE and controlled medication release [45, 46].
These substances encompass photosensitive nanomaterials, nitrobenzene, azoben-
zene, fulgide and stilbene-containing polymers, as well as their derivatives. Chro-
mophores are incorporated into polymer resins to develop photo-responsive poly-
meric products. The fabrication of these materials is also feasible by incorporating
photosensitive nanoparticles into polymers. Depending on the type of chromophore,
the system’s function might be either reversible or irreversible [47, 48].
The most prevalent reaction mechanisms for photo-responsive constituents, that
have been extensively used to develop active 4D shape-changing frameworks, are
440 Amisha et al.
chitosan exhibits a sol–gel phase transition. The pH-responsive swelling of the opti-
mized hydrogel efficiently delays shrinkage at a neutral pH. Moreover, the optimized
hydrogel exhibits outstanding biocompatibility and biodegradability. These findings
demonstrate the constructed hydrogel’s tremendous potential to be implemented in
an array of applications, encompassing delivery systems, wound healing, and tissue
engineering [55].
A novel technique to generate methacrylated carboxymethyl chitosan (CMCS-
MA) hydrogel, a shielding layer that can be agglomerated by visible light, was
investigated by Xing et al. (2022). This new optimized hydrogel demonstrated quick
light cure, high biocompatibility, and lysozyme degradation. Hence, the flexible
and convenient biocompatible prepared hydrogels might have a promising use in
the regeneration of periodontal tissue [56]. Using MHBC and soluble collagen,
a simplistic photo/thermo dual curing composite hydrogel layout was brought on
by Liu et al. (2021). Integration of temperature-induced sol–gel transition and
contraction by M/C composite hydrogel, the optimum microstructure, customiz-
able mechanical characteristics, enhanced cyto-compatibility, and biodegradability
for three-dimensional cell culture. M/C composite hydrogel is a viable option for
bio-ink that can be employed in in situ three-dimensional bioprinting [57]. Min
et al. (2021) proposed the first hydrogel comprised of platelet lysate from human
whole blood that could be methacrylate photopolymerized to produce a crosslinked
hydrogel layer by layer, and then employed to construct cell-filled hydrogel frame-
works. Efficient proliferation and cell dispersion at small concentrations of bio-ink
optimizes biocompatibility and moldability. The 3D-printable PLMA bioinks could
represent a viable method for designing tailored microenvironments incorporating
materials derived from live organisms for the repair of diverse tissues [58].
The substantial light attenuation attributed to biological tissues, however, causes
concern. Investigating the implementation of ultra-IR light, that penetrates more
deeply into the tissue than UV radiation and less phototoxicity and absorption in
live tissue might aid in resolving this challenge. Owing to their unanticipated cyto-
toxicity, several photo-initiators have limitations in tissue engineering applications.
It has become feasible to generate new photo-polymerized systems with substan-
tially reduced detrimental consequences on cellular metabolic activities and prolif-
erative potential, paving the way to activities like cell encapsulation and the bio-
fabrication of injectable hydrogels. Figure 3 further portrays the applicability of
photo-responsive materials with their penetrability and ubiquitous optical attributes.
The implementation of light responsive materials in biomedical field is represented
in Table 2.
Bone tissue In vitro (mesenchymal stem cells) In this investigation, they employed a bismuth sulfide/hydroxyapatite (BS/ [62]
engineering HAp) film to rapidly and repeatedly generate a photoelectric-responsive
microenvironment around a transplant. Via modulating the in vivo
photoelectric surroundings using NIR light, this research has developed a
biological therapeutic technique that can accomplish in vitro distantly,
reliably, and noninvasively directing cell differentiation behaviours
(continued)
443
Table 2 (continued)
444
The skin, the outermost and major organ of the human body, offers a physical barrier
to shield individuals from hazards including diseases and microorganisms. Skin that
has been wounded is consequently vulnerable to infection. While healing, uninfected
wounds have a pH of 5.5–6.5, but infected wounds have a pH greater than 6.5. As
a result, a condition brought on by bacteria is frequently considered prevalent when
a wound’s pH shifts from acidic to alkaline [71]. Because of fragile nature of skin
tissue, hydrogels are frequently utilized to print skin analogues in 3D with respect
to time. Skin constructs are constructed using a plethora of 3D printing techniques,
including laser-assisted bioprinting as well as extrusion-based printing. To treat burns
or persistent wounds, complete skin structures are also generated employing tissue
engineering. A substantial amount of printing research involves materials made of
collagen since it is a key component of natural skin. Nevertheless, collagen has a slow
crosslinking rate and poor printability. Due to its antibacterial qualities and capacity
to induce haemostasis, chitosan is favoured over collagen for applications involving
wound healing. Furthermore, skin constructions can be printed using alginate, gela-
tine, GelMA, and fibrin. Synthetic polymers like polycaprolactone (PCL), polyethy-
lene glycol, polyglycolic acid (PGA), polylactic acid (PLA), and related copolymers
like polylactic-co-glycolic acids (PLGA) due to their higher compatibility with bodily
446 Amisha et al.
tissues, are utilized for gathering matrices separately or as composites for the regen-
eration of tissues. Derived from rice and corn, PLA is a biodegradable polyester that
has received FDA approval. PLA has indeed been extensively adopted in the appli-
cation of tissue engineering scaffolds to transfer cells to the site of the wound in spite
of possessing drawbacks such slow degradation rate, limited elongation, poor cell
interaction, and hydrophobicity, which can cause an inflammatory response [72, 73].
With a slower rate of deterioration than PLA and PLGA, PCL seems to be another
popular biomaterial that has been utilized in various research for skin tissue regen-
eration. Originally employed to generate biodegradable films and moulds. Due to
the development of electrospinning technology, PCL has been utilized to make drug
delivery systems, scaffolds for tissue regeneration, and absorbable sutures. Consid-
ering its acidic by-products and relatively slow degradation rate, PCL could be a
potential replacement to strengthen the mechanical attributes and reducing the rate
of deterioration of natural biomaterials [74, 75]. PEG is a desirable synthetic bioma-
terial due to its chemical and structural characteristics, but it lacks interacting cell
qualities. Liu et al. (2022), employing gelatine, sodium alginate, and fibrinogen, they
developed a distinctive bio-ink. A dermal layer design including fibroblasts was orig-
inally developed, and then laminin and keratinocytes were subsequently added on top
of it by adjusting the ratio of the bio-components, inks the 3D model’s layout, and the
printing circumstances. Full-thickness skin tissue was generated through air–liquid
interface (ALI) culturing adopting sterilized wire mesh. In addition to being physi-
cally similar to human skin, HE and immunofluorescence labelling revealed that the
bio-printed skin also displayed particular markers relevant to epidermal transition
and stratum corneum development [76].
A tissue with high mechanical strength and mineralization is bone. To better imitate
bone tissue, it is crucial to strengthen the mechanical attributes of printed polymer.
Numerous researches have been published in the various literatures that high-
lighted the assembly of printed scaffolds for BTE [77]. The polymers are blended
with minerals, notably tricalcium phosphate (TCP) and hydroxyapatite (HAP), to
strengthen the printed products. For bone tissue scaffolds created via 3D printing,
the most prevalent polymer is PCL. And for several medicinal applications, PCL
is an FDA-approved, biodegradable and biocompatible polymer. The fact that this
polymer has a comparatively low Tm and Tg (60 °C) makes it a great product for fused
deposition modelling among the key reasons for adopting it. PCL, however, lacks any
cell-attractive moieties and is a hydrophobic polymer. To boost the hydrophilicity of
3D printed PCL scaffolds, it was incorporated with poly (propylene fumarate) (PPF)
in a study [78]. The biological functions and microstructures, mechanical attributes,
porosity, of 4D-printed SMPs scaffolds with collagen-dexamethasone (Col-Dex) and
bioactive fillers (alendronate and hydroxyapatite) covering were thoroughly inves-
tigated in this research by Zhou et al. (2022). Under an alternating magnetic field,
Photo Responsive Material for 4D Printing in Tissue Engineering 447
the SMP scaffolds developed in the study might be generated into temporary forms
of tiny sizes and recovered to functioning shapes and sizes to treat bone inade-
quacies. Additionally, the new 4D printed scaffolds with the Col-Dex coating and
bioactive fillers would offer a suitable method for enhanced bone tissue regeneration
and repair of customized bone tissue [79]. In the proposed investigation, Hwangbo
et al. (2021) successfully used 3D printing, leaching, and coating techniques to
create a hierarchical scaffold of biomimetic collagen/hydroxyapatite with multiple
microchannels in each framework (i.e., one-way shape morphing, 4D printing). In
a posterolateral lumbar spinal fusion mouse model, this biomimetic microchannel
framework significantly increased blood vessel ingrowth as well as novo bone forma-
tion. It also displayed remarkable promise for osteogenic activities in vitro. These
in vivo and in vitro results point to the possibility of using the microchannel collagen/
hydroxyapatite scaffold in place of bone grafts to facilitate effective fusion at high
rates [80].
of shape memory, customized vascular stents with negative Poisson’s ratio, and are
manufactured via 4D printing. The architecture is optimized adopting the genetic
technique. The mechanical characteristics of the stents are examined using axial
compression, three-point bending, and radial compression tests. The finite element
technique is additionally employed to explore the fluid–structure connection and
stress distribution during the form process of recovery. The stents exhibit remark-
able shape memory characteristics, and in vitro feasibility investigations show that
they can rapidly broaden a mimicked constricted blood artery. For the management
of vascular stenosis, 4D printed shape memory scaffolds with a negative Poisson’s
ratio framework are therefore very interesting [87]. Creating a T-shaped vascular
bifurcation using 3D printed shape-changing layers and a mathematical model by
Kitana et al. (2023) is made possible for the first time using a 4D bio fabrication-
based methodology. Tubular constructions with different diameters might be created
by carefully controlling the parameters (for example, crosslinking time). As a conse-
quence, when immersed in water that has a few centimetres in diameter, the 3D-
printed films show self-transformation into a T-junction. When the tubular T-junction
is perfused with an aqueous liquid simulating blood flow via capillaries, it displays
very little spillages with a peak flow velocity of 0.11 ms−1 . Moreover, human umbil-
ical vein endothelial cells that have been planted on the inside of a simple T-junction
exhibit exceptional growth characteristics and superb cell survival. The sizes that have
been reached are comparable to those of natural blood vessels, which is still prob-
lematic to achieve using 3D bio fabrication. The formation of completely automatic,
self-regulating vascular divergences as vascular transplants is facilitated through this
method [88].
muscle repair processes. Current research has focused on the utilisation of appro-
priate biomaterials that might be used as a prototype to direct tissue remodelling
and eventually give cells the ideal microenvironment. These methods include using
biomaterials solely as well as combining them with ex vivo-produced cells and exoge-
nous growth agents [93, 94]. Although various approaches for the management of
serious muscular injuries have been developed so far though, functional engineered
muscles remain a critical clinical issue that must be addressed. Skeletal muscle’s
(SM) ability to regenerate is, in fact, insufficient for repairing significant damage,
and SM reconstruction is still a challenging problem. In order to achieve this goal
and encourage muscle regeneration, tissue-engineered muscles ought to offer a suit-
able biomimetic extracellular matrix (ECM) substitute that is defined by a myogenic
milieu and an aligned/micro topographical structure. As a result, choosing the right
materials and fabrication methods is essential to developing a successful therapeutic
strategy. Tissue-specific decellularized ECM (dECM) is among the most potential
material for assisting muscle repair and regeneration [95]. Technologically advanced
bio fabrication strategies, like bioprinting, enables for the construction of frameworks
laden with cells that could greatly speed up the healing process of tissue. Bioprinted
cell-filled structures can imitate the extracellular matrix and offer a bioactive envi-
ronment for cells to grow, distribute, and distinguish, leading in the skeletal muscle
tissue’s regeneration at the site of injury. In the analyses, Sonaye et al. (2022) devel-
oped bio-ink-capable alginate-gelatine composite inks. Then, they employed the
inks inside an extrusion printing technique to create frameworks with customized
contours for a potential VML therapy. Although the gelatine concentration was held
constant at 6% w/v, the alginate concentration was changed between 4 and 12% w/
v. The alginate-gelatine inks with 12 and 6% w/v alginate and gelatine respectively,
according to rheological study, were the best for creating architecture with good struc-
tural fidelity and high resolution [96]. In this study, they investigated the function
of topographical variables on modulating behaviours of human mesenchymal stem
cells (hMSCs) onto skeletal muscle tissues via blending surface coating methodology
with FDM. This staircase defect will then be translated into an efficient bioengi-
neering strategy. This inclusive strategy provides to construct anisotropic scaffolds,
contour-specific, multi-dimensional, employing distinct biomaterials. hMSC align-
ment is efficaciously aimed by 2D anisotropic frameworks, that were initially exhib-
ited here with varying quantities of polycaprolactone. This is certainly relevant when
a support loop is employed to fix the framework. The polymer solution from within
FDM-printed sacrificial frameworks is interface encased to produce 3D anisotropic
frameworks featuring thin wall characteristics. Subsequently, using these frame-
works, seeded hMSCs are driven through a spinning bioreactor. Utilizing layer-by-
layer coating, including an SMP, smart constructs demonstrating contour stabiliza-
tion and recovery procedures are equipped, trying to bring this investigation into the
domain of 4D printing. The topographical indications produced by FCT, based on
immunofluorescence labelling and real-time quantitative polymerase chain reaction
evaluation, significantly improve the interpretation of myogenic genes like desmin
and myosin heavy chain-2. The fact that many organs and tissues, including skeletal
muscle, contain highly coordinated and anisotropic extracellular matrix components
450 Amisha et al.
leads researchers to the conclusion that this FCT method offers extensive application
potentials in tissue engineering [32].
The anisotropic alignment of the nerve fibres generates the directional (uniaxial)
architecture of nerve tissue. Since a lesion, a damaged nerve’s both distal and prox-
imal ends are brought together using nerve aides. Moreover, they could be constructed
employing 3D printing to create patient-specific constructions with a sophisticated
internal layout. Products for nerve tissue engineering are frequently made using an
AM approach, including ink jet printing, SLA, FDM, and bioprinting. To manu-
facture constructions for nerve applications in tissue engineering, polyurethane,
collagen, fibrin, gellan gum, PLA, carboxymethyl chitosan, GelMA, agarose, and
PEGDA were also employed [97, 98]. The constructs primarily contain neural stem
cells (NSCs), although investigations on nerve regeneration have also used glial
cells, astrocytes, bone marrow stem cells (BMSCs), retinal ganglion cells (RGCs),
Schwann cells (SCs), and primary cortical neurons. Since the nervous system is
comprised of various cell types, including neurons, SCs, and glial cells, numerous
researches have focused on printing these types of cells to construct a complete nerve
tissue. The direction of these cells is also crucial because of the anisotropic nature
of nerve tissue. This issue can be successfully handled by bio printing which makes
it possible to print cells on fibres or in tubes of hydrogel with a specified align-
ment. The 3D or 4D printed frameworks can also include biochemically significant
molecules [6, 99]. Apsite et al. (2020) highlight the employment of the 4D bio fabri-
cation approach to construct a synthetic nerve graft. Electrospinning was designed
to create bilayer scaffolds that included randomly aligned methacrylated hyaluronic
acid (HA-MA) fibres and uniaxially aligned polycaprolactone-poly (glycerol seba-
cate) (PCL-PGS) fibres meant for the incubation of PC-12 neuron cells. After a
fibrous bilayer is immersed in an aqueous buffer, tubular structures immediately
form. The width of these tubes might be altered by adjusting bilayer variables like
the thickness of each layer, the medium counterion concentration, and the overall
thickness. Over the course of four weeks of real-time deterioration, the scaffolds
were designed to reveal a self-folding scroll-like structure with remarkable stability
[100]. Low-level light treatment (LLLT) has been demonstrated to be effective in
treating neurological disorders and degenerative nerve diseases. In light of this, Zhu
et al. (2017) hypothesize that integrating LLLT with a 3D-printed neural scaffold
would culminate in an innovative approach to managing neural degeneration. To
accomplish this, they applied red laser light to stimulate neural stem cells on 3D
printed scaffolds, and afterwards we investigated at how the cells responded in terms
of differentiation and proliferation. Thus, they demonstrate that after 15 s of laser
stimulation and one day of culture, the rate of proliferation of cell and the generation
of intracellular reactive oxygen species (ROS) were both significantly elevated [99].
Photo Responsive Material for 4D Printing in Tissue Engineering 451
Applications for tissue engineering of the cornea, meniscus, cartilage, and urethra
have used 4D or 3D printing. A significant contributor to chronic impairment is
cartilage deterioration, which is typically treated with highly invasive treatments.
The emergence of 3D bioprinting as a prospective tissue engineering methodology
to address this problem. The structures created with this technique lack the dynamic
reactivity of native tissues and are stagnant. Moreover, it was still difficult to fabricate
curved or tubular structures, particularly while using soft tissues. The above issues
might be addressed by 4D bioprinting, a recently developed, next-generation bio-
fabrication technology [6]. The application of corneal transplantation is anticipated
to be a viable approach for a myriad of corneal disease requirements. Although
it has been used as an effective treatment for the majority of corneal problems,
patients still have a number of difficulties since rejection is one unidentified hazard
of corneal transplant material, and there aren’t sufficient healthy donor corneas attain-
able. A lot of emphasis has been put on the creation of tissue-engineered scaffolds
for corneal regeneration and healing utilizing corneal tissue engineering (CTE). The
development of a substrate with equivalent mechanical and transmittance qualities is
investigated by employing various methods in order to enhance corneal tissue regen-
eration. Due to their amazing spatial control, which results in a 3D corneal framework
filled with cells, bio-printed scaffolds have recently attracted enough attention in this
regard to simulate corneal structure [101]. In spite of the fact that tissue engineering is
frequently utilized to create intricate, three-dimensional biocompatible architectures,
researchers are currently working to expand the approach into the fourth dimension.
This 4D is the gradual metamorphosis of three-dimensional materials, specifically
the alteration of their composition, function, and/or shape in retort to certain external
cues. The study explores the creation of a 4D biomaterial with an intrinsic stimulating
function by Miotto et al. (2019). Contractile cells change the form and structure of
tissues by acting as bio-actuators. Namely, curving collagen-based hydrogels under
controlled, cell-driven conditions to produce cornea-shaped, curved stromal tissue
analogues. The following is accomplished by leveraging a contraction-inhibiting
peptide amphiphile to modify the activity of the bio-actuators in particular regions
of the gels. The ability of the self-curved structures to promote the in vitro devel-
opment of a corneal epithelium is then assessed, along with the gel stiffness, cell
phenotypic, and cell and collagen fibril rearrangement. The results show that self-
curved gels made using a 4D engineering technique have structural and mechanical
properties that are much better than those of planar 3D scaffolds and more like native
tissue. From this aspect, the study shows how versatile cell bio-actuators are for 4D
tissue engineering applications [102].
SLS, ink-jet bioprinting, and extrusion-based bioprinting, for instance, have
all been employed to cartilage tissue engineering. The printing polymers include
polyurethane (PU), nanocellulose, collagen, alginate and agarose. The most often
used cells in cartilage regeneration are chondrocytes [103]. The innovative 4D
452 Amisha et al.
gel network equilibrium water content, mechanical and physical characteristics, and
cytotoxicity were investigated, and an attempt was made to 3D print the hydrogel.
Then, the viability of urethral tissue engineering with the hydrogel was assessed.
The results demonstrated that the hydrogel made of poly (2-hydroxyethyl methacry-
late) (p-HEMA) underwent visible-light curing at 405 nm and it was accomplished
using the composite photo initiator system. The p-HEMA hydrogel’s mechanical
characteristics, biocompatibility, and other qualities were enhanced by the addition
of sodium alginate [108]. Zhang et al. (2017) have constructed in vitro urethras with
various polymer varieties and structural features adopting 3D bioprinting technology.
The usage of PCL and PLCL polymers with a spiral scaffold design may be used
to simulate the structure and mechanical properties of a rabbit’s natural urethra, and
a fibrin hydrogel containing cells may offer a better environment for cell devel-
opment. Adopting an integrated bioprinting methodology, tubular constructs were
fabricated, and smooth muscle and urothelial cells were uniformly distributed into
the interior and exterior regions of the framework within the cell-laden hydrogel. The
native urethra of the rabbit was mechanically comparable to the spiral PCL/PLCL
(50:50) structure. When the bio-printed urethra’s cell bioactivity was evaluated, it
was discovered that SMCs and UCs still had more than 80% of their original viability
7 days after printing [109].
The next era of tissue engineering techniques, referred as 4D bioprinting, that inte-
grates “time” as the fourth dimension to 3D bio printing, is anticipated to facilitate
the construction of complex entities featuring potentially controlled shapes and func-
tionalities on request. With the emergence of stimuli-responsive biomaterials such as
light responsive biomaterials and improved knowledge of tissue regeneration over the
past several years, the biomedical enterprise and clinical applications have garnered
4D bioprinting innovation a significant amount of attention [110]. To illustrate, the
domain of customized tissue regeneration offers tremendous application potential for
4D bioprinting innovation. For the 4D-printed implantation, the lesion sites would be
precisely geometrized with a specified size and form. In the post-printing phase, the
operational modification of the transplant would include characteristics that mirror
biological processes, promoting tissue development and remodelling. Personalized
tissue engineering now has new possibilities to design neo-tissue growth owing to
recent breakthroughs in computational model systems. By developing self-growing
frameworks, the transformational capabilities of 4D bioprinting could also help treat
adolescent patients [111].
A Hi-tech, innovative technique named 4D bioprinting generates tailored elements
of sophisticated component by interpreting digital medical imaging and demon-
strates remarkable automation control. Via constructing and creating 3D cell-laden
dynamical constructs, this technology is progressing and paving the way for novel
approaches in biological research [112]. Exorbitant researchers recently shown how
454 Amisha et al.
stimulation, and the large majority of these cues are exclusively temperature-related,
which confines the spectrum of biological applications they can be utilized for. There
might be the potential to adopt new devices for biomedical applications because
to developments in functionalizing current monomers and polymers to strengthen
their biocompatibility or render them printable [116]. The recently created and
then used 4D system known as soybean oil epoxidized acrylate is an example of
a new material. Rapid initiatives are required to investigate and create innovative
multi-responsive hydrogels and SMPs. Further, the fabrication of versatile func-
tional bioengineered tissues exhibiting remarkable biomechanical attributes can be
accelerated by the augmentation of MNP nanoparticles, ceramic, and graphene into
hydrogels and SMPs. The development of printed products is booming attributable
to 4D bioprinting technology, which also holds the potential to completely transform
TERM and DDS. However, it has a number of drawbacks, including the inability to
forecast and design structural resilience into 4D-printed structures [117]. To address
these difficulties, focused study on the empirical modelling of this technology is
required. Given that 4D bioprinting has been implemented in applications involving
tissue implantation, it is vital to create novel bio-ink substances that have dynamic
behaviour, improved multi-stimuli sensitivity, cellular adherence, and optimal stiff-
ening for in-vitro testing. Innovative bioprinters, structural designs and procedures
are anticipated for 4D bioprinting expertise in addition to the bio-ink materials listed
above [118]. The hurdles preventing the invention in 4DP from being commercial-
ized provide an opportunity to examine strengths, weaknesses, opportunities, and
risks. By overcoming each of these obstacles, 4DP of polymer-relied composites
will broaden the range of biomedical applications it may be used for, including
those for medical devices, cardiac, cartilaginous, bone, skin, neuro, DDSs, and indi-
vidualized medicine. Further, the tremendous improvements in the contemporary
iteration of 4D printing approach would help us better understand stimuli-response
substances with their characteristics, and prospective biological attributes. In addition
to the TERM applications, 4D bioprinting might potentially be employed to facilitate
organ replacement in chronic disorders or fatal accidents [114]. Hence, it remains
difficult to create printed things that can simultaneously respond to many stimuli and
go through intricate shape-transformation processes. To actualize the manufacture
of complicated self-transforming items, computer design advancements and intri-
cate adaptable stimuli-responsive approaches must be incorporated. This will help to
accelerate the scaling-up of fabrication and acknowledge the intricately foreordained
regulation of 4DP in tissue engineering. Platform designed by MIT called Project
Cyborg that allows for the simulation of self-assembling systems and the integration
of architectures with programmable substances. The tracking of bodily motion has
been done using biosensors and bio-actuators. However, more research is needed to
offer accurate, least invasive control of the stimuli delivered to bio-printed structures.
Yet, these programmable and stimuli-cues biomaterials could be expensive, and it
is often expensive to produce such materials using advanced computer design tools.
Production expansion should be feasible and affordable in the interim. As a result,
there is still a trade-off between the excellence of 4D bio-printed constructions and
their manufacturing viability [9].
456 Amisha et al.
6 Conclusion
The 4D printing industry has made great strides during the past ten years. Since
its inception, it has expanded its influence into a number of industrialized regions.
In 4D printing, a shape-changing mechanism is often induced over time employing
additive manufacturing processes and stimuli-responsive materials. Single-printed
object might be exploited for numerous procedures, improving flexibility and adapt-
ability. To put it another way, 4D printing represents a new advancement in 3D
printing. The utilization of stimuli-responsive components in this approach is crucial
for smart materials, and 3D printing technology enables dynamic printed frame-
work. This trait demonstrates the tremendous future potential of 4D printing. So far
it has been successfully applied to fields that seek for dynamic constructs, such as
tissue engineering and organ transplants. As a corollary, the combination of tissue
engineering and light-responsive biomaterials used in 4D printing led to significant
improvements in our understanding of therapeutic processes based on the interest in
how monochromatic light interacts with biological tissues. A comprehensive under-
standing of photodynamic mechanisms would be made possible by the science of
tissue engineering which is currently undergoing rapid expansion. The state of the
graphics in this domain of expertise could be directly improved with the help of this
review chapter. Retrieving and exhibiting crucial data for the oversight of endeav-
ours that seek to use these sophisticated protocols. There is no disputing about the
immense potential of combining these principles, which are at the forefront of tech-
nology of knowledge and can aid in the creation of new biological assays used
in a variety of clinical therapies. Moreover, 4D printing can be developed to create
specialized prosthetics, tailored implants, and anatomical models using imaging tech-
niques like CT scans and MRI. Although there have been numerous improvements,
4D printing is still in its initial stages. The aspects of 4D printing methodology,
such as imaging techniques, additive manufacturing methods, stimulus-responsive
materials and stimuli, thus need more exploration. Nevertheless, not all biomate-
rials are stimulus-responsive materials, and all stimulus-responsive materials might
not be utilized for printing equipment. Sensitive materials should be employed for
4D printing. Moreover, often these materials only adapt to a unique stimulation.
Thus, by searching into unique responsive materials and attempting to make the
Photo Responsive Material for 4D Printing in Tissue Engineering 457
References
1. Wan, Z., Zhang, P., Liu, Y., Lv, L., Zhou, Y.: Four-dimensional bioprinting: Current
developments and applications in bone tissue engineering. Acta Biomater. 101, 26–42 (2020)
2. Top, N., Sahin, I., Gokce, H., Gokce, H.: Computer-aided design and additive manufacturing
of bone scaffolds for tissue engineering: state of the art. J. Mater. Res. 1–21 (2021)
3. Jiang, D., Ning, F., Wang, Y.: Additive manufacturing of biodegradable iron-based particle
reinforced polylactic acid composite scaffolds for tissue engineering. J. Mater. Process.
Technol. 289, 116952 (2021)
4. Santoni, S., Gugliandolo, S.G., Sponchioni, M., Moscatelli, D., Colosimo, B.M.: 3D
bioprinting: current status and trends—a guide to the literature and industrial practice. Bio-Des
Manuf. 5(1), 14–42 (2022)
5. Chu, H., Yang, W., Sun, L., Cai, S., Yang, R., Liang, W, et al.: 4D printing: a review on recent
progresses. Micromachines (Basel) 11(9) (2020)
6. Tamay, D.G., Dursun Usal, T., Alagoz, A.S., Yucel, D., Hasirci, N., Hasirci, V.: 3D and 4D
printing of polymers for tissue engineering applications. Front. Bioeng. Biotechnol. 7, 164
(2019)
7. Khalid, M.Y., Arif, Z.U., Noroozi, R., Zolfagharian, A., Bodaghi, M.: 4D printing of shape
memory polymer composites: a review on fabrication techniques, applications, and future
perspectives. J. Manuf. Process. 81, 759–797 (2022)
8. Noroozi, R., Zolfagharian, A., Fotouhi, M., Bodaghi, M.: 4D-printed shape memory polymer:
modeling and fabrication. In: Smart Materials in Additive Manufacturing, Volume 2: 4D
Printing Mechanics, Modeling, and Advanced Engineering Applications: Elsevier, pp. 195–
228 (2022)
9. Arif, Z.U., Khalid, M.Y., Zolfagharian, A., Bodaghi, M.: 4D bioprinting of smart polymers
for biomedical applications: recent progress, challenges, and future perspectives. React Funct
Polym. 105374 (2022)
10. Wang, Y., Cui, H., Esworthy, T., Mei, D., Wang, Y., Zhang, L.G.: Emerging 4D printing
strategies for next-generation tissue regeneration and medical devices. Adv. Mater. 34(20),
e2109198 (2022)
11. Donderwinkel, I., Tuan, R.S., Cameron, N.R., Frith, J.E.: Tendon tissue engineering: current
progress towards an optimized tenogenic differentiation protocol for human stem cells. Acta
Biomater. (2022)
12. Dhandayuthapani, B., Yoshida, Y., Maekawa, T., Kumar, D.S.: Polymeric scaffolds in tissue
engineering application: a review. Int. J. Polym Sci. (2011)
458 Amisha et al.
13. Gao, J., Yu, X., Wang, X., He, Y., Ding, J.: Biomaterial-related cell microenvironment in
tissue engineering and regenerative medicine. Engineering (2022)
14. Eldeeb, A.E., Salah, S., Elkasabgy, N.A.: Biomaterials for tissue engineering applications
and current updates in the field: a comprehensive review. AAPS Pharm Sci. Tech. 23(7), 267
(2022)
15. Lee, H.P., Gaharwar, A.K.: Light-responsive inorganic biomaterials for biomedical applica-
tions. Adv. Sci. (Weinh) 7(17), 2000863 (2020)
16. Naskar, A., Kim, K.S.: Photo-stimuli-responsive CuS nanomaterials as cutting-edge platform
materials for antibacterial applications. Pharmaceutics 14(11) (2022).
17. Dolmans, D.E., Fukumura, D., Jain, R.K.: Photodynamic therapy for cancer. Nat. Rev. Cancer
3(5), 380–387 (2003)
18. Bajpai, A., Baigent, A., Raghav, S., Bradaigh, C.O., Koutsos, V., Radacsi, N.: 4D printing:
materials, technologies, and future applications in the biomedical field. Sustainabilit 12(24),
10628 (2020)
19. Yang, G.H., Yeo, M., Koo, Y.W., Kim, G.H.: 4D bioprinting: technological advances in
biofabrication. Macromol. Biosci. 19(5), e1800441 (2019)
20. Amukarimi, S., Rezvani, Z., Eghtesadi, N., Mozafari, M.: Smart biomaterials: from 3D
printing to 4D bioprinting. Methods 205, 191–199 (2022)
21. Yang, Q., Gao, B., Xu, F.: Recent advances in 4D bioprinting. Biotechnol. J. 15(1), e1900086
(2020)
22. Agarwal, T., Hann, S.Y., Chiesa, I., Cui, H., Celikkin, N., Micalizzi, S., et al.: 4D printing in
biomedical applications: emerging trends and technologies. J. Mater Chem. B 9(37), 7608–
7632 (2021)
23. Haleem, A., Javaid, M., Singh, R.P., Suman, R.: Significant roles of 4D printing using smart
materials in the field of manufacturing. Adv. Ind. Eng. Polym. Res. 4(4), 301–311 (2021)
24. McLellan, K., Sun, Y.C., Naguib, H.: A review of 4D printing: materials, structures, and
designs towards the printing of biomedical wearable devices. Bioprinting e00217 (2022)
25. Zhou, H., Dong, G., Gao, G., Du, R., Tang, X., Ma, Y., et al.: Hydrogel-based stimuli-
responsive micromotors for biomedicine. Cyborg Bionic Syst. 2022, 9852853 (2022)
26. Shen, Z., Chen, F., Zhu, X., Yong, K.T., Gu, G.: Stimuli-responsive functional materials for
soft robotics. J. Mater. Chem. B (2020)
27. He, Y., Yu, R., Li, X., Zhang, M., Zhang, Y., Yang, X., et al.: Digital light processing 4D
printing of transparent, strong, highly conductive hydrogels. ACS Appl. Mater. Interfaces
13(30), 36286–36294 (2021)
28. Kang, X., Zhang, X.B., Gao, X.D., Hao, D.J., Li, T., Xu, Z.W.: Bioprinting for bone tissue
engineering. Front. Bioeng. Biotechnol. 10, 1036375 (2022)
29. Zhang, B., Li, H., Cheng, J., Ye, H., Sakhaei, A.H., Yuan, C., et al.: Mechanically robust
and UV-curable shape-memory polymers for digital light processing based 4D printing. Adv.
Mater. 33(27), e2101298 (2021)
30. Gao, Q., Lee, J.S., Kim, B.S., Gao, G.: Three-dimensional printing of smart constructs using
stimuli-responsive biomaterials: a future direction of precision medicine. Int. J. Bioprinting
9(1), 638 (2023)
31. Miao, S., Cui, H., Nowicki, M., Lee, S.J., Almeida, J., Zhou, X., et al.: Photolithographic-
stereolithographic-tandem fabrication of 4D smart scaffolds for improved stem cell
cardiomyogenic differentiation. Biofabrication 10(3), 035007 (2018)
32. Miao, S., Nowicki, M., Cui, H., Lee, S.J., Zhou, X., Mills, D.K., et al.: 4D anisotropic skeletal
muscle tissue constructs fabricated by staircase effect strategy. Biofabrication 11(3), 035030
(2019)
33. Fina, F., Goyanes, A., Madla, C.M., Awad, A., Trenfield, S.J., Kuek, J.M., et al.: 3D printing
of drug-loaded gyroid lattices using selective laser sintering. Int. J. Pharm. 547(1–2), 44–52
(2018)
34. Wu, H., Wang, O., Tian, Y., Wang, M., Su, B., Yan, C., et al.: Selective laser sintering-
based 4D printing of magnetism-responsive grippers. ACS Appl. Mater. Interfaces 13(11),
12679–12688 (2021)
Photo Responsive Material for 4D Printing in Tissue Engineering 459
35. Murthy, H., Thakur, N., Shankhwar, N.: Nickel-based inks for flexible electronics—a review
on recent trends. J. Adv. Manuf. Syst. 21(03), 591–624 (2022)
36. Cui, C., Kim, D.O., Pack, M.Y., Han, B., Han, L., Sun, Y., et al.: 4D printing of self-folding
and cell-encapsulating 3D microstructures as scaffolds for tissue-engineering applications.
Biofabrication 12(4), 045018 (2020)
37. Quan, H., Zhang, T., Xu, H., Luo, S., Nie, J., Zhu, X.: Photo-curing 3D printing technique
and its challenges. Bioact Mater. 5(1), 110–115 (2020)
38. Deshmane, S., Kendre, P., Mahajan, H., Jain, S.: Stereolithography 3D printing technology
in pharmaceuticals: a review. Drug Dev. Ind. Pharm. 47(9), 1362–1372 (2021)
39. Zhuo, S., Geever, L.M., Halligan, E., Tie, B.S.H., Breheny, C.: A development of new mate-
rial for 4D printing and the material properties comparison between the conventional and
stereolithography polymerised NVCL hydrogels. J. Funct. Biomater. 13(4) (2022)
40. Davoodi, E., Sarikhani, E., Montazerian, H., Ahadian, S., Costantini, M., Swieszkowski, W.,
et al.: Extrusion and microfluidic-based bioprinting to fabricate biomimetic tissues and organs.
Adv. Mater. Technol. 5(8) (2020)
41. Colosi, C., Costantini, M., Barbetta, A., Dentini, M.: Microfluidic bioprinting of heteroge-
neous 3D tissue constructs. Methods Mol. Biol. 1612, 369–380 (2017)
42. Wu, Y., Wenger, A., Golzar, H., Tang, X.S.: 3D bioprinting of bicellular liver lobule-mimetic
structures via microextrusion of cellulose nanocrystal-incorporated shear-thinning bioink.
Sci. Rep. 10(1), 20648 (2020)
43. Ahmed, A., Arya, S., Gupta, V., Furukawa, H., Khosla, A.: 4D printing: fundamentals,
materials, applications and challenges. Polymer 228, 123926 (2021)
44. Weng, S., Kuang, X., Zhang, Q., Hamel, C.M., Roach, D.J., Hu, N., et al.: 4D printing of
glass fiber-regulated shape shifting structures with high stiffness. ACS Appl. Mater. Interfaces
13(11), 12797–12804 (2021)
45. Luo, Y., Lin, X., Chen, B., Wei, X.: Cell-laden four-dimensional bioprinting using near-
infrared-triggered shape-morphing alginate/polydopamine bioinks. Biofabrication 11(4),
045019 (2019)
46. Cui, H., Miao, S., Esworthy, T., Lee, S.-j., Zhou, X., Hann, S.Y., et al.: A novel near-infrared
light responsive 4D printed nanoarchitecture with dynamically and remotely controllable
transformation 12, 1381–1388 (2019)
47. Patdiya, J., Kandasubramanian, B.: Progress in 4D printing of stimuli responsive materials.
Polym.-Plast. Technol. Mater. 60(17), 1845–1883 (2021)
48. Shi, H., Wang, C., Ma, Z.: Stimuli-responsive biomaterials for cardiac tissue engineering and
dynamic mechanobiology. APL Bioeng. 5(1), 011506 (2021)
49. Brown, T.E., Anseth, K.S.: Spatiotemporal hydrogel biomaterials for regenerative medicine.
Chem. Soc. Rev. 46(21), 6532–6552 (2017)
50. Lim, K.S., Klotz, B.J., Lindberg, G.C., Melchels, F.P., Hooper, G.J., Malda, J., et al.: Visible
light cross-linking of gelatin hydrogels offers an enhanced cell microenvironment with
improved light penetration depth 19(6), 1900098 (2019)
51. Rosenfeld, A., Gockler, T., Kuzina, M., Reischl, M., Schepers, U., Levkin, P.A.: Designing
inherently photodegradable cell-adhesive hydrogels for 3D cell culture. Adv. Healthc. Mater.
10(16), e2100632 (2021)
52. Lv, W., Long, K., Yang, Y., Chen, S., Zhan, C., Wang, W.: A red light-triggered drug
release system based on one-photon upconversion-like photolysis. Adv. Healthc. Mater. 9(21),
e2001118 (2020)
53. Watanabe, U., Sugiura, S., Kakehata, M., Yanagawa, F., Takagi, T., Sumaru, K., et al.: Fabri-
cation of hollow structures in photodegradable hydrogels using a multi-photon excitation
process for blood vessel tissue engineering. Micromachines (Basel) 11(7) (2020)
54. Wu, K., Wu, X., Zhang, Y., Chen, S., Qiao, Z., Wei, D., et al.: Semiconvertible hyaluronic
hydrogel enabled red-light-responsive reversible mechanics, adhesion, and self-healing.
Biomacromol 23(3), 1030–1040 (2022)
55. Che, Q.T., Charoensri, K., Seo, J.W., Nguyen, M.H., Jang, G., Bae, H., et al.: Triple-conjugated
photo-/temperature-/pH-sensitive chitosan with an intelligent response for bioengineering
applications. Carbohydr. Polym. 298, 120066 (2022)
460 Amisha et al.
56. Xing, X., Su, J., Liu, Y., Lin, H., Wang, Y., Cheng, H.: A novel visible light-curing chitosan-
based hydrogel membrane for guided tissue regeneration. Colloids Surf. B Biointerfaces 218,
112760 (2022)
57. Liu, Y., Luo, X., Wu, W., Zhang, A., Lu, B., Zhang, T., et al.: Dual cure (thermal/photo)
composite hydrogel derived from chitosan/collagen for in situ 3D bioprinting. Int. J. Biol.
Macromol. 182, 689–700 (2021)
58. Min, S.J., Lee, J.S., Nah, H., Kim, S.H., Moon, H.J., Reis, R.L., et al.: Development of
photo-crosslinkable platelet lysate-based hydrogels for 3D printing and tissue engineering.
Biofabrication 13(4) (2021)
59. Chua, C.J., Han, J.L., Li, W., Liu, W., Entcheva, E.: Integration of engineered “spark-cell”
spheroids for optical pacing of cardiac tissue. Front. Bioeng. Biotechnol. 9, 658594 (2021)
60. Wang, Y., Cui, H., Wang, Y., Xu, C., Esworthy, T.J., Hann, S.Y., et al.: 4D printed cardiac
construct with aligned myofibers and adjustable curvature for myocardial regeneration. ACS
Appl. Mater. Interfaces 13(11), 12746–12758 (2021)
61. Cui, H., Miao, S., Esworthy, T., Lee, S.J., Zhou, X., Hann, S.Y., et al.: A novel near-infrared
light responsive 4D printed nanoarchitecture with dynamically and remotely controllable
transformation. Nano Res. 12, 1381–1388 (2019)
62. Fu, J., Liu, X., Tan, L., Cui, Z., Zheng, Y., Liang, Y., et al.: Photoelectric-responsive
extracellular matrix for bone engineering. ACS Nano 13(11), 13581–13594 (2019)
63. Zhao, W., Wang, H., Wang, H., Han, Y., Zheng, Z., Liu, X., et al.: Light-responsive dual-
functional biodegradable mesoporous silica nanoparticles with drug delivery and lubrication
enhancement for the treatment of osteoarthritis. Nanoscale 13(13), 6394–6399 (2021)
64. Akhavan, O., Ghaderi, E., Shirazian, S.A.: Near infrared laser stimulation of human neural
stem cells into neurons on graphene nanomesh semiconductors. Colloids Surf. B Biointerfaces
126, 313–321 (2015)
65. Wu, C., Su, B., Xin, N., Tang, J., Xiao, J., Luo, H., et al.: An upconversion nanoparticle-
integrated fibrillar scaffold combined with a NIR-optogenetic strategy to regulate neural cell
performance. J. Mater Chem. B 11(2), 430–440 (2023)
66. Griffin, M., Castro, N., Bas, O., Saifzadeh, S., Butler, P., Hutmacher, D.W.: The current
versatility of polyurethane three-dimensional printing for biomedical applications. Tissue
Eng. Part B Rev. 26(3), 272–283 (2020)
67. Vanaei, S., Parizi, M.S., Salemizadehparizi, F., Vanaei, H.R.: An overview on materials and
techniques in 3D bioprinting toward biomedical application. Eng. Regen. 2, 1–8 (2021)
68. Varma, M.V., Kandasubramanian, B., Ibrahim, S.M.: 3D printed scaffolds for biomedical
applications. Mater. Chem. Phys. 255, 123642 (2020)
69. Norotte, C., Marga, F.S., Niklason, L.E., Forgacs, G.: Scaffold-free vascular tissue engineering
using bioprinting. Biomaterials 30(30), 5910–5917 (2009)
70. Miri, A.K., Khalilpour, A., Cecen, B., Maharjan, S., Shin, S.R., Khademhosseini, A.:
Multiscale bioprinting of vascularized models. Biomaterials 198, 204–216 (2019)
71. Battisti, M., Vecchione, R., Casale, C., Pennacchio, F.A., Lettera, V., Jamaledin, R., et al.:
Non-invasive production of multi-compartmental biodegradable polymer microneedles for
controlled intradermal drug release of labile molecules. Front. Bioeng. Biotechnol. 7, 296
(2019)
72. Sheikholeslam, M., Wright, M.E., Jeschke, M.G., Amini-Nik, S.: Biomaterials for skin
substitutes. Adv. Healthc. Mater. 7(5), 1700897 (2018)
73. Ferri, J.M., Jorda, J., Montanes, N., Fenollar, O., Balart, R.: Manufacturing and characteri-
zation of poly (lactic acid) composites with hydroxyapatite. J. Thermoplast. Compos. Mater.
31(7), 865–881 (2018)
74. Mondal, D., Griffith, M., Venkatraman, S.S.: Polycaprolactone-based biomaterials for tissue
engineering and drug delivery: Current scenario and challenges. Int. J. Polym. Mater. Polym.
Biomater. 65(5), 255–265 (2016)
75. Li, M., Chen, J., Shi, M., Zhang, H., Ma, P.X., Guo, B.: Electroactive anti-oxidant polyurethane
elastomers with shape memory property as non-adherent wound dressing to enhance wound
healing. Chem. Eng. J. 1(375), 121999 (2019)
Photo Responsive Material for 4D Printing in Tissue Engineering 461
76. Liu, J., Zhou, Z., Zhang, M., Song, F., Feng, C., Liu, H.: Simple and robust 3D bioprinting of
full-thickness human skin tissue. Bioengineered 13(4), 10087–10097 (2022)
77. Wang, W., Caetano, G., Ambler, W.S., Blaker, J.J., Frade, M.A., Mandal, P., et al.: Enhancing
the hydrophilicity and cell attachment of 3D printed PCL/graphene scaffolds for bone tissue
engineering. Materials (Basel) 9(12) (2016)
78. Lee, S.J., Lee, D., Yoon, T.R., Kim, H.K., Jo, H.H., Park, J.S., et al.: Surface modification of
3D-printed porous scaffolds via mussel-inspired polydopamine and effective immobilization
of rhBMP-2 to promote osteogenic differentiation for bone tissue engineering. Acta Biomater.
40, 182–191 (2016)
79. Zhou, W., Dong, X., He, Y., Zheng, W., Leng, J.: In-vitro and in-vivo studies of 4D
printed shape memory scaffolds with bioactive fillers and coating for enhanced bone tissue
regeneration. Smart Mater. Struct. 31(10), 105002 (2022)
80. Hwangbo, H., Lee, H., Roh, E.J., Kim, W., Joshi, H.P., Kwon, S.Y., et al.: Bone tissue engi-
neering via application of a collagen/hydroxyapatite 4D-printed biomimetic scaffold for spinal
fusion. 8(2), 021403 (2021)
81. Nurden, A.T.: Platelets, inflammation and tissue regeneration. Thromb. Haemost. 105(Suppl
1), S13-33 (2011)
82. Gu, W., Hong, X., Potter, C., Qu, A., Xu, Q.: Mesenchymal stem cells and vascular
regeneration. Microcirculation 24(1) (2017)
83. Arslan-Yildiz, A., El Assal, R., Chen, P., Guven, S., Inci, F., Demirci, U.: Towards artificial
tissue models: past, present, and future of 3D bioprinting. Biofabrication 8(1), 014103 (2016)
84. Carrabba, M., Madeddu, P.: Current strategies for the manufacture of small size tissue
engineering vascular grafts. Front. Bioeng. Biotechnol. 6, 41 (2018)
85. Zhu, J., Wang, Y., Zhong, L., Pan, F., Wang, J.: Advances in tissue engineering of vasculature
through three-dimensional bioprinting. Dev. Dyn. 250(12), 1717–1738 (2021)
86. Zhang, J., Wehrle, E., Rubert, M., Muller, R.: 3D bioprinting of human tissues: biofabrication,
bioinks, and bioreactors. Int. J. Mol. Sci. 22(8) (2021)
87. Lin, C., Zhang, L., Liu, Y., Liu, L., Leng, J.: 4D printing of personalized shape memory
polymer vascular stents with negative Poisson’s ratio structure: a preliminary study. Sci.
China Technol. Sci. 63(4), 578–588 (2020Apr)
88. Kitana, W., Apsite, I., Hazur, J., Boccaccini, A.R., Ionov, L.J.A.M.T.: 4D biofabrication of
T-shaped vascular bifurcation 8(1), 2200429 (2023)
89. Wang, H.D., Lough, D.M., Kurlander, D.E., Lopez, J., Quan, A., Kumar, A.R.: Muscle-derived
stem cell-enriched scaffolds are capable of enhanced healing of a murine volumetric muscle
loss defect. Plast. Reconstr. Surg. 143(2), 329e-e339 (2019)
90. Mase, V.J., Jr., Hsu, J.R., Wolf, S.E., Wenke, J.C., Baer, D.G., Owens, J., et al.: Clinical
application of an acellular biologic scaffold for surgical repair of a large, traumatic quadriceps
femoris muscle defect. Orthopedics 33(7), 511 (2010)
91. Kwee, B.J., Mooney, D.J.: Biomaterials for skeletal muscle tissue engineering. Curr. Opin.
Biotechnol. 47, 16–22 (2017)
92. Mulbauer, G.D., Matthew, H.W.: Biomimetic scaffolds for skeletal muscle regeneration.
Discoveries 7, e90 (2019)
93. Downing, K., Prisby, R., Varanasi, V., Zhou, J., Pan, Z., Brotto, M.: Old and new biomarkers
for volumetric muscle loss. Curr. Opin. Pharmacol. 59, 61–69 (2021)
94. Gilbert-Honick, J., Grayson, W.: Vascularized and innervated skeletal muscle tissue engi-
neering. Adv. Healthc. Mater. 9(1), e1900626 (2020)
95. Baiguera, S., Del Gaudio, C., Di Nardo, P., Manzari, V., Carotenuto, F., Teodori, L.: 3D printing
decellularized extracellular matrix to design biomimetic scaffolds for skeletal muscle tissue
engineering. Biomed. Res. Int. 2020, 2689701 (2020)
96. Sonaye, S.Y., Ertugral, E.G., Kothapalli, C.R., Sikder, P.: Extrusion 3D (bio)printing of
alginate-gelatin-based composite scaffolds for skeletal muscle tissue engineering. Materials
(Basel) 15(22) (2022)
97. Layrolle, P., Payoux, P., Chavanas, S.: Message in a scaffold: natural biomaterials for three-
dimensional (3D) bioprinting of human brain organoids. Biomolecules 13(1) (2022)
462 Amisha et al.
98. Esworthy, T.J., Miao, S., Lee, S.J., Zhou, X., Cui, H., Zuo, Y.Y., et al.: Advanced 4D bioprinting
technologies for brain tissue modeling and study. Int. J. Smart Nano Mater. 10(3), 177–204
(2019)
99. Zhu, W., George, J.K., Sorger, V.J., Grace, Z.L.: 3D printing scaffold coupled with low level
light therapy for neural tissue regeneration. Biofabrication 9(2), 025002 (2017)
100. Apsite, I., Constante, G., Dulle, M., Vogt, L., Caspari, A., Boccaccini, A.R., et al.: 4D biofab-
rication of fibrous artificial nerve graft for neuron regeneration. Biofabrication 12(3), 035027
(2020)
101. Orash Mahmoud Salehi, A., Heidari-Keshel, S., Poursamar, S.A., Zarrabi, A., Sefat,
F., Mamidi, N., et al.: Bioprinted membranes for corneal tissue engineering: a review.
Pharmaceutics 14(12) (2022)
102. Miotto, M., Gouveia, R.M., Ionescu, A.M., Figueiredo, F., Hamley, I.W., Connon, C.J.: 4D
corneal tissue engineering: achieving time-dependent tissue self-Curvature through localized
control of cell actuators. Adv. Funct. Mater. 29(8), 1807334 (2019)
103. Avila, H.M., Schwarz, S., Rotter, N., Gatenholm, P.: 3D bioprinting of human
chondrocyte-laden nanocellulose hydrogels for patient-specific auricular cartilage regener-
ation. Bioprinting 1, 22–35 (2016)
104. Kalogeropoulou, M.: 4D bioprinting for cartilage tissue engineering: a controlled shape
transformation approach (2021)
105. Singh, R., Gupta, N., Vanathi, M., Tandon, R.: Corneal transplantation in the modern era.
Indian J. Med. Res. 150(1), 7–22 (2019)
106. Li, H., Liao, Z., Yang, Z., Gao, C., Fu, L., Li, P., et al.: 3d printed poly(epsilon-caprolactone)/
meniscus extracellular matrix composite scaffold functionalized with kartogenin-releasing
PLGA microspheres for meniscus tissue engineering. Front. Bioeng. Biotechnol. 9, 662381
(2021)
107. Pastorek, D., Culenova, M., Csobonyeiova, M., Skuciova, V., Danisovic, L., Ziaran S.: Tissue
engineering of the urethra: from bench to bedside. Biomedicines 9(12) (2021)
108. Gu, X., Xu, Y., Li, S., Wang, Z., Meng, Q., Yu, J.: Preparation of a photocured biocompatible
hydrogel for urethral tissue engineering. ACS Appl. Polym. Mater. 3(7), 3519–3527 (2021)
109. Zhang, K., Fu, Q., Yoo, J., Chen, X., Chandra, P., Mo, X., et al.: 3D bioprinting of urethra
with PCL/PLCL blend and dual autologous cells in fibrin hydrogel: An in vitro evaluation of
biomimetic mechanical property and cell growth environment. Acta Biomater. 50, 154–164
(2017)
110. Li, Y.C., Zhang, Y.S., Akpek, A., Shin, S.R., Khademhosseini, A.: 4D bioprinting: the
next-generation technology for biofabrication enabled by stimuli-responsive materials.
Biofabrication 9(1), 012001 (2016)
111. Bryant, S.J., Vernerey, F.J.: Programmable hydrogels for cell encapsulation and neo-tissue
growth to enable personalized tissue engineering. Adv. Healthc. Mater. 7(1), 1700605 (2018)
112. Taylor, J.M., Luan, H., Lewis, J.A., Rogers, J.A., Nuzzo, R.G., Braun, P.V.: Biomimetic and
biologically compliant soft architectures via 3D and 4D assembly methods: a perspective.
Adv. Mater. 34(16), e2108391 (2022)
113. Kamolz, L.P., Kotzbeck, P., Schintler, M., Spendel, S.: Skin regeneration, repair, and
reconstruction: present and future. Eur. Surg. 54(3), 163–169 (2022)
114. Guan, J., Jia, Y., Zhang, B., Zhao, G.: Application of 4D bioprinting in tissue engineering.
Chin J. Tissue Eng. Res. 26(3), 446 (2022)
115. Chiu, D.T., Demello, A.J., Di Carlo, D., Doyle, P.S., Hansen, C., Maceiczyk, R.M., et al.:
Small but perfectly formed? Successes Challenges Oppor. Microfluid. Chem. Biol. Sci. 2(2),
201–223 (2017)
116. Lee, J.Y., An, J., Chua, C.K.: Fundamentals and applications of 3D printing for novel materials.
Appl. Mater. Today 7, 120–133 (2017)
117. Miao, S., Zhu, W., Castro, N.J., Nowicki, M., Zhou, X., Cui, H., et al.: 4D printing smart
biomedical scaffolds with novel soybean oil epoxidized acrylate. Sci. Rep. 6, 27226 (2016)
118. Chan, B.Q.Y., Chong YT, Wang, S., Lee, C.J.J., Owh, C., Wang, F., et al.: Synergistic combi-
nation of 4D printing and electroless metallic plating for the fabrication of a highly conductive
electrical device 430, 132513 (2022)
Photo Responsive Material for 4D Printing in Tissue Engineering 463
119. Zhou, W., Qiao, Z., Nazarzadeh Zare, E., Huang, J., Zheng, X., Sun, X., et al.: 4D-
printed dynamic materials in biomedical applications: chemistry, challenges, and their future
perspectives in the clinical sector. J. Med. Chem. 63(15), 8003–8024 (2020)
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 465
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_15
466 B. Soni et al.
Abbreviations
1 Introduction
In various fields of medicine, there has been extensive use of biomaterials throughout
a significant duration, and their perception has varied over time. Williams gave the
first comprehensive explanation of biomaterials in 1987, describing them as “Non-
living materials utilized in healthcare devices with a focus on interacting with biolog-
ically systems”. These substances had to be risk-free, non-carcinogenic, chemically
and physiologically stable, and mechanically robust enough to withstand repeated
loads during their existence [1]. As advancements in the applications and composi-
tions of biomaterials progressed, these definitions were modified to incorporate these
developments [2, 3]. The following examples illustrate the evolution of biomaterial
definitions.
• “Synthetic substance utilizes to modify a component of a living system or to
interact closely with living tissue.”
• “A pharmacologically and systemically inactive material intended for integration
with or implantation among biological systems.”
Surface-Modified Biomaterials in Medical Device Development 467
Due to their higher affinity for a wider range of proteins than others, albumin,
fibrinogen, IgG, fibronectin, and Von Willebrand factor make up the majority of
plasma proteins found in utilized biomaterials. The process of protein adsorption
Surface-Modified Biomaterials in Medical Device Development 469
and interaction can change the shape of the biomaterial that is produced. Espe-
cially fibrinogen, which becomes more adhesive when in contact with biomaterial or
implant surfaces, can cause denaturation of biomaterials. In order to prevent neoepi-
tope exposure after cell and tissue interactions and preserve the biomaterial for the
duration of its intended duration of storage, it is crucial to change the biomaterial’s
surface.
Protein adhesion, cell disruption, and inflammatory response are all significantly
influenced by the surface chemistry and structure of biomaterials. For instance,
in vitro cell adsorption can be produced by changing the surface chemistry of a
biomaterial. However, traditional chemistry is not a requirement for the in vivo mech-
anisms of externally applied biomaterials. Typically made of polymers, ceramics,
metals (cobalt-chrome, titanium), and other materials, biomaterials have a variety
of surface properties that affect how they react in vivo, ranging from hydrophilic to
hydrophobic and tough to soft. In order to construct highly friendly biomaterial archi-
tectures, a variety of different techniques, including physical modifications, chemical
alterations, and radiation, are employed to alter the surface of biomaterials.
Surface modification methods that significantly affect protein adsorption and
biological responses in vitro include those that modify polymer chemistry, wetting
capacity, and domain. Additionally, it has been demonstrated that protein adsorption
and cellular responses are significantly influenced by the structure and design of
biomaterials. Several topical experiments have made use of methods for changing
surfaces that have been mentioned in the literature, such as plasma therapy, chemical
graft modification, and self-assembled monolayer methods [14]. This instance of a
foreign body reaction is typical of how cells in higher organisms react to artificial
biomaterials that have been inserted.
The method that tissues and the implant surface interact is always varying.
Within the initial several seconds following implantation, dissolved ions, unbound
biomolecules, and water are everywhere over the implant surface. Figure 1 illus-
trates how the bio fluid surrounding the wound changes in composition and a layer
of biomolecules is adsorbing to the surface to begin the healing process. The adsorbed
layer then controls how the cells behave once they reach the surface. Eventually, a
tissue integration or fibrous capsule will form as the types of cells and their activity
on the surface alter. The atomic, molecular, and higher-level physical textural prop-
erties operate as contact points for biological elements like proteins, cells, tissues,
etc. At the same time, it’s crucial to understand that the biomolecules react to the
chemical activity of the implant surfaces.
The various bonding modalities connected to each of these biological units have
an impact on how a surface is integrated hierarchically into its bonding environment.
Chemical species that start distinct levels of bonding have an impact on adhesion
qualities. A component experiences various chemical processes depending on the
470 B. Soni et al.
surroundings, which makes it more difficult to understand the specific nature of the
interactions [15, 16].
5.1.1 Blasting
The expected electrical grinding system and the grinding’s electrochemical opera-
tions. In this process, an electrode is positioned above a metallic bond grinding wheel
that is conducting, with a gap of around 0.3 mm, or nearly 1/6 the size of the surface
of the wheel. The electrode is given a negative potential, while the grinding wheel is
given a positive potential. It employs an expert pulse generator. The potential during
the process electrolytically breaks down the conductive alkaline machining fluid used
in the grinding procedure, producing hydroxide ions. When the work piece receives
the proper amount of positive potential, free hydroxide ions (OH-) are attracted to
the surface being processed by substances, which are present in the machining fluid.
This emphasizes the ions to the surface, which results in the formation of a stable
oxidized layer. For our investigation, we used test pieces made of type 316 stainless
steel and the Ti6AI-4V alloy, each of which is frequently used as metallic biomate-
rials. A grinding wheel with a metal-resin-hybrid bond (#8000, diamond abrasives,
with a diameter of approximately 2 microns) was used. [21].
For the purpose of developing biomaterials for drugs, the sol-gel method is a straight-
forward, wet chemical process that doesn’t necessitate a high pH or high sintering
temperature. The term “sol-gel coating” refers to the colloidal dispersion of solid
particles (1–500 nm) in a liquid solution. The techniques of spraying, dipping, spin-
ning, and doctor-blading can be used to apply a sol to a substrate. To create the top
layer, the substrate gel is dried or calcined. By immersing a metal sample in a calcium
and phosphorus gel at lower temperatures, calcium phosphate layers are created on
the sample using this approach. The covering coating is calcined at 400–600 °C
depending upon the material since the layer produced is porous and less dense [22].
5.2.2 Anodization
Anodizing is a process that is widely used to treat titanium and its alloys. It is similar
to acid etching. In this procedure, the substance is dissolved in an ionic solution of
H2SO4 , H3PO4 , or acetic acid while being exposed to anodic voltage. The process
results in the surface of titanium and its alloys developing a thicker oxide layer,
increasing their corrosion resistance. However, because this oxide layer is porous,
sealing is frequently needed. On titanium substrates, titanium nanotubular structures
can develop when fluoride-containing electrolytes are employed. The voltage, elec-
trolyte concentration, and reaction time are some of the anodization parameters that
significantly affect the shape and structure of these nanotubular layers.
Surface-Modified Biomaterials in Medical Device Development 473
Researchers have looked into how these anodized structures affect bacterial adher-
ence, cell function, and bone formation. Studies have demonstrated that compared
to normal flat titanium substrates, anodized surfaces with titanium nanotubes of 40–
60 nm in diameter significantly increase adhesion and proliferation of bone marrow
cells. Additionally, calcium deposition on anodized substrates is significantly higher
than on conventional titanium substrates. S. aureus and S. epidermidis are two bacteria
that are impacted by the diameter of the nanotubes’ effects on bacterial adhesion to
nanotubular titanium surfaces. The lowest bacterial adherence is seen on surfaces
with 80 nm diameter nanotubes, which are less rigid than ordinary flat surfaces or
nanotubular surfaces with tube diameters of 20, 40, and 60 nm.
Table 2 Advantages and disadvantages of metals, ceramics, and polymers in medical devices
Different Metals Ceramics Polymers Composites
materials
Advantages Strength and Biocompatibility Biocompatibility Non-toxic
Durability Wear Resistance Flexibility Strength and
Biocompatibility Corrosion Lightweight durability
Corrosion Resistance Versatility Light weight
Resistance Strength and Durability Moldability
Ease of Hardness Radiolucency
Fabrication Radiolucency
Radiopacity
Disadvantages Metal Allergies Lack of Limited Limited material
Thermal Flexibility temperature range option
conductivity Brittleness Moisture Lack of
Costly Rough surface absorption standardization
finish Biocompatibility Limited shelf life
Difficulty in
manufacturing
Surface-Modified Biomaterials in Medical Device Development 475
6.1.1 Steel
Since stainless steel has a high level of corrosion resistance and strength, it is
frequently utilized for biomedical implants. To keep the nickel content low, nickel-
free stainless steel is used, and nitrogen is added to increase biocompatibility. Despite
stainless steel’s lower biocompatibility and corrosion resistance compared to tita-
nium, temporary bone fracture therapies and implant studies frequently employ it
since it is more affordable. Custom-sized stainless-steel implants can be created using
3D printing, and 3D dental implants can be created using liquid phase sintering.
Implants composed of pure metals, which had lesser corrosion resistance and
mechanical strength, had been developed before stainless steel entered the biomedical
business. The two types of stainless steel utilized in biomedical applications are
standard stainless steel and Ni-free stainless steel, which is devoid of nickel. Stainless
steel is widely used for bone fracture treatments, such as screws, nails, and fracture
plates, and for fabricating durable implant trials. Compared to titanium, stainless
steel has inferior biocompatibility, osseointegration, and corrosion resistance [27].
476 B. Soni et al.
The Titanium and Ti-based alloys are well suited for usage with a high pace of loading
than stainless steel for the reason that they have a higher strength-to-weight ratio.
In comparison to steel, titanium implants have a significantly stronger bond with
tissues because of the titanium dioxide layer’s high dielectric constant, which forms
quickly on the surface of bare titanium. It may increase the mechanical durability
of titanium alloy by annealing, quenching, and thermal aging, and it can be further
modified by alloying other elements like aluminum and niobium. For functionally
demanding anatomically complex locations like cranio-maxillofacial surgery, 3D-
printed titanium implants can be tailored.
Techniques for melting electron beams and directly sintering metal have been used
to create customized titanium prostheses. To reduce stress shielding, porosity might
be added to the implant’s structure. To reduce stress shielding and encourage tissue
regeneration or vascularization, porosity might be added to the implant’s structure.
The structure is rich in interconnected grooves and has 3D Ti-6Al-4V dental implants’
Young’s modulus gradient changes from 104 GPa at the metal inner core to 77 GPa
for the very porous outer shell when gradient porosity is generated utilizing the
selective remain sintering procedure [28, 29].
Since Co-Cr alloys have a higher wear resistance compared to Ti alloys, they
frequently used in prosthetic hip joints. Although they are less biocompatible and
less capable of osseointegration than Ti, their higher elastic modulus and stiffness
result in higher stress shielding compared to Mg, Ti, and Ti alloys. Ti is frequently
utilized in clinical settings for components higher stress shielding compared to Mg,
Ti, and Ti alloys, while Co-Cr is the preferred material for components that do not
come into contact with the bone [30]. At the point where Co-Cr and Ti come into
contact, metal corrosion and shredding remain serious problems.
Tantalum has been studied for usage in a range of applications where outstanding
durability against corrosion even under acidic conditions and biocompatibility in
biomaterials are required. Tantalum’s ability to resist corrosion is because a protective
coating of natural, stable Ta2 O5 forms over the implant surface. The functioning of
porous tantalum to connect with bone makes it a promising material for use in
artificial joints as a polymeric matrix or includes a coating on titanium and implants
are constructed from stainless steel to improve osseointegration and prevention from
corrosion [31]. Tantalum has a density of 16.6 g/cm3 and an elastic modulus of
Surface-Modified Biomaterials in Medical Device Development 477
more than 186 GPa Because they are significantly different from native cortical (12–
18 GPa) and cancellous bone (0.1–0.5 GPa) in these respects, these features are
detrimental when used in orthopedic implants.
It is challenging to produce this metal in large quantities due to its refractory prop-
erties, especially given its unusually high melting point of approximately 3017 °C.
Due to Ta’s high temperature conductivity, patients who undergo cranioplasty may
also experience temperature-dependent headaches. To meet the growing need for
orthopedic applications, metallic bioimplants have been developed using cutting-
edge manufacturing techniques. An efficient method to create implants of the proper
size may be discovered through a three-dimensional visualization of the patient and
printing of the modified implant. Laser sintering is a technique that can be used
to produce high-fidelity copies of pre-packaged implants using reasonably priced
316L stainless steel. Another use of liquid phase sintering is the fabrication of
stainless-steel 3D dental implants.
method for his 3D printing of Co-Cr alloys. After 26 weeks in adult sheep femurs,
the implants were shown to have satisfactory overall bone-implant connection. The
implant’s surroundings gradually underwent tissue ingrowth and densification, and
the generated bone’s mineral crystallinity, apatite to collagen ratio, and carbonate
to phosphate ratio differed between Co-Cr and Ti-6Al. It was in the range of 4 V
implants [35].
Printing implants, both solid and mesh, is possible using EBM. Co-29Cr-
6Mo alloy mesh-strut and foam-ligament microstructures both produced columnar-
directed Cr23C6 precipitate architectures that were spaced about 2 m apart in the build
direction to represent the directional solidification of solid cylindrical components
during the assembly process in the melt pool. While similarly manufactured solid Ti-
6Al-4V implants displayed acicular platelets in the - phase, mesh and foam implants
largely displayed residual 0-martensitic phase. It displayed a fine cellular microstruc-
ture with enriched Mo and depleted Co grain boundaries when the CoCrMo implant
was created using SLM. This provides the implant more corrosion resistance than a
cast alloy by reducing carbide precipitation and the creation of the Martensitic phase
on its surface. These characteristics, which also limit the discharge of metal ions
into the peri-implant environment, lower the possibility of developing metallosis.
Rates of ion release and corrosion were correlated with the number of laser melt
pool borders [26].
There are many technological obstacles that are keeping 3D printing from devel-
oping. The presence of a vascular network poses the toughest challenge from a
biological standpoint. An adequate supply of nutrients, correct gas exchange, and
effective waste disposal are necessary for the growth of 3D tissues or organs during
perfusion; these processes would not occur in the absence of such a network. As
a result, the artificial organ’s overall efficacy and cell longevity would be affected.
However, there is still a significant obstacle to overcome before a system can success-
fully deliver nutrients, growth hormones, and oxygen to the cells without interfering
with their metabolic activities [36].
Despite multiple attempts by researchers to develop vascular trees using computer
models, it has not yet been possible to produce fractured veins with branched chan-
nels that are advantageous mechanically. For instance, electro-hydrodynamic inkjet
printing (e-jetP) has the ability to generate prints with far higher resolution than
traditional inkjet printing techniques. By utilizing an electric field to overcome
surface tension, a metallic nanoparticle solution, such as Ag, Cu, Au, or Co, is
evacuated as droplets in e-jetP [37]. Rapid layer-by-layer assembly with fine control
over the form, dimension, and resolution of micro- and nano-scale features is made
possible by accelerated solvent evaporation. Although there are currently few macro-
scopic implants that utilize this technology, these forms may be ideal for implantable
electrodes and sensors [38].
Surface-Modified Biomaterials in Medical Device Development 479
For all metallic devices to perform their function efficiently and to endure for a long
time, AM-based surface changes of metallic biomaterials are essential [39]. Passi-
vating materials (Al, Cr, Ti) are added, as well as surface energy, topography, and
crystalline structure to increase osseointegration, biocompatibility, and prevent corro-
sion without affecting the bulk properties of implants [40, 41]. Although including
roughness to implant surfaces may reduce their mechanical properties, it can improve
tissue integration [42].
It has been demonstrated that porous materials with purposeful topology promote
adhesion and advantageous differentiation. Based on computer tomography (CT)
images, AM is used to build biomimetic surface structures that have substantially
identical surface characteristics to those found in living cells. The most prevalent
technique for preventing corrosion is the incorporation of self-passivating materials
(Ti, Cr, and AL). Implant of the lifespan is increased by the desirable wear qualities
of some alloys, such as CoCrMo, as opposed to softer metals like titanium. But
even these artificial limbs can become worn down gradually, releasing cobalt and
chromium ions that can lead to metallosis and osteolysis. Degradation of UHMWPE,
which leads to the creation of microparticles, might come from the incompatibility
of hardness between the metal and the polymer.
Ceramics are utilized in various medical fields such as dentistry, orthopedics, and
medication delivery. They are very helpful in the construction of bone scaffolds for
the vital repair of fractures and other injuries in which the normal healing capacity of
the bone is compromised. Ceramics are also used in dental procedures for enamel and
root replacement, bone cement for minor fractures, ceramic-on-ceramic articulating
surfaces for joints, and the hip, knee alignment, and shoulder area implants as coatings
healing, and other drug release applications [43]. Ninety-nine percent of the calcium
in the human body is found in bones and teeth, making it the fifth most common
element in the body. The fact that many biomedical ceramics are calcium-based is
therefore not surprising. Both bone and teeth are mostly made of ceramic materials,
which have the right chemical makeup and crystallography for these uses.
Due to the similarities in chemical and crystal structure, as well as in micro-
and nano topography, biomimetic devices are made that replicate the appearance
and functionality of biological tissue, thereby preventing inflammation or rejection
brought on by immune reactions to foreign objects [44]. The additional benefit of
biomimicry is that it breaks down into ions and calcium phosphates the fact that
the body is able to rapidly resorb or excrete. Whether used as a restorable material,
degradation has a minor impact, and the rate of product absorption can be modified in
accordance with the speed of healing of the tissue by altering the surface, the structure
480 B. Soni et al.
of the crystals, or the calcium phosphate phase. Calcium phosphate scaffolds have a
high degree of elemental resemblance to actual bone in applications that come into
touch with it, although they lack trace elements like Si4+ , Mg2+ , Sr2+ , Zn2+ , Cu2+ , and
Fe2+ /Fe3+ , among others. Recent studies have concentrated on ionic elements that
are naturally present in bone that can be added to bone scaffolds. Further improving
the biomimetic features of ceramics in biomedical applications are the ionic’ roles
in angiogenesis, osteogenesis, and osteo induction [45]. Different ceramic materials
used for surface modification of biomaterials are given below.
6.2.1 Alumina
6.2.2 Zirconia
With the use of proximal femur, zirconia was first examined as a biomaterial in the
late 1960s. The combination of zirconia and yttrium, known as Tetragonal zirconia
polycrystals, often known as TZP (tetragonal zirconia polycrystals), is the most
common widespread after testing various chemical composition possibilities and
is most frequently utilized in femoral heads [46]. It is one of the ceramic materials
used in prosthetic equipment most frequently because of its mechanical and chemical
qualities. It combines with oxygen and produces the biocompatible zirconia oxide
(ZrO2 ) as a result. Due to their great biocompatibility, good flexural strength, and
fracture resistance, ZrO2 implants are mostly employed in femoral heads for hip
arthroplasty and dental implants.
6.2.3 Hydroxyapatite
Apatite crystals constitute the minerals found in bone tissues. The chief inorganic
constituent responsible for the development of bones and teeth is a high hardness
calcium phosphate salt, known as HA. Due to its chemical makeup and structural
similarity with natural HA, HA ceramic exhibits exceptional biocompatibility.
Surface-Modified Biomaterials in Medical Device Development 481
6.2.4 Carbon
Carbon implants have an advantage over metallic implants in that they do not expe-
rience fatigue. However, because of their fragility and low tensile strength, they are
not suitable for situations that necessitate support for substantial loads. Although it
is legal to use it in blood-contact devices, it is still a good idea to double-check.
Because they perform better than metals and polymers in terms of compressive
strength and corrosion resistance, additively produced ceramics are mostly used in
dental applications. However, resorbable drug delivery systems are the main focus
of surface modification of ceramics through AM. The low breakdown temperature
of the additional material is one of the major difficulties, which makes it challenging
to sinter an AM ceramic loaded with pharmaceuticals or biomolecules. The ceramic
powder particles are held together by a binder, but they are nevertheless prone to
bulk disintegration and the early release of loaded molecules.
In order to achieve consistent release profiles and ensure implant stability in vivo,
surface modification techniques can be employed. One such approach is to introduce
layers of polymeric coatings to the surface to prevent the sudden release of medica-
tion. Another technique involves coating ceramics with polymers and dosing them
with desired medicine after calcination to regulate drug elution. Trace elements have
also been utilized as additives or dopants to accelerate bone development and vascu-
larization. When the drug delivery system (DDS) is subcutaneous or gastrointestinal,
several binder formulations have been employed to create components with a bulk
interior chemistry and a less soluble, polymer-enhanced outer surface chemistry. [47].
482 B. Soni et al.
Natural rubber latex is a colloidal system best recognized for its use in dental tools,
condoms, and other medical supplies. Extensive research and documentation have
been conducted on allergies to natural rubber latex. However, a recently developed
manufacturing technique that avoids the use of carbamates or sulfur has resulted in a
more biocompatible product. This development has opened up new possibilities for
using the product as a delivery system for drugs, proteins, and nanoparticles, as well
Surface-Modified Biomaterials in Medical Device Development 483
as for aiding in the controlled formation of bones and the healing of soft scar tissue
[26].
Polyolefins are plastic resins that are produced from either propylene or ethylene,
depending on the type of polyolefin. These plastics are hydrophobic and inert,
meaning that they do not break down in the body. Polyethylene, or PE, is a type of
polyolefin that comes in five different variants: high-density polyethylene (HDPE),
low-density polyethylene (LDPE), linear low-density polyethylene (LLDPE), ultra-
low-density polyethylene (ULDPE) and ultra-high-molecular weight polyethylene
(UHMWPE). PE can be processed by various methods such as blowing, extrusion,
and injection molding, utilized in sutures and netting and is physiologically inert.
UHMWPE, which is one variant of PE, is commonly used for artificial joint surfaces
because of its high impact strength, chemical stability, and good biocompatibility.
However, the release of particles due to the friction of the components can cause
reactions in the body that can lead to mechanical failure of the prostheses.
To resolve this issue, highly cross-linked polyethylene has been used, and the
material is treated to gamma radiation to improve its biocompatibility and wear
resistance. Antioxidants like vitamin E were added to the biomaterial to produce the
second generation of cross-linked UHMWPE. Numerous medical fields, including
orthopedics, cardiology, and neurology, use UHMWPE. Some examples of products
made of UHMWPE are surgical cables for bone fractures, high-strength orthopedic
sutures for soft tissue repair, catheters, stent grafts, heart valves, and disc replace-
ments for spinal repair. Similar to polyethylene, polypropylene is a type of polyolefin
that is biologically inert and used to manufacture nets and sutures.
6.3.3 PVC
6.3.4 Silicones
6.3.5 Acetal
It is a hard plastic substance made from formaldehyde that is highly durable and has
a low coefficient of friction. This makes it a useful material for machining to create
prototypes of medical devices and parts. Acetal is susceptible to the radiation used
in sterilization, so it should be kept in mind that this could make the material brittle
and easily break.
6.3.6 Polyamides
The most widely used synthetic polyamide used in medicinal uses is nylon. Due to
its great tensile strength, it is utilized in suture lines. A composite material of nylon
and PU with nylon strength and elasticity makes up the balloons of the catheters used
in angioplasty. Recent studies have concentrated on synthetic polymers with assur-
ance for use in soft and hard tissue regeneration, such as polycaprolactone (PCL),
polypropylene fumarate (PPF), polyether ether ketone (PEEK), and functionalized
polyurethanes [50, 51]. Furthermore, due to their attraction as naturally occurring
biopolymers or their degrading properties, natural polymers including collagen and
fibrin as well as polysaccharides like alginate, silk, hyaluronic acid, and chitosan
have been investigated for biomedical uses.
Collagen, the most common protein and naturally occurring polymer in the ECM,
is commonly used for surface modification. Cell mobility is supported and adhered
to by collagen. The less biodegradable and even permanent polymers are polyte-
trafluoroethylene (PTFE) and poly (methyl methacrylate) (PMMA). PMMA, with
its excellent optical properties, is used for contacts and intraocular lenses. However,
foreign body reactions can occur, leading to the need for surface modification [52].
PMMA is used frequently in bone cement to anchor and secure implants. In partic-
ular, the difficulties of enhancing ocular compatibility and maintaining the duration
of implantable ocular devices, the organ’s shape, and the primary physiological roles
of the component tissues influence the choice of biomaterials for ocular implants.
bioactivity may hinder cell adhesion, prolonging the healing process, and perhaps
causing the development of fibrotic tissue.
As comparison to HAP alone, biocomposites with a bioactive glass layer offer higher
adhesion and mechanical strength while retaining the bioactivity of HAP. The Ti-6Al-
4V with bioglass-apatite coatings have been the subject of numerous research, which
have revealed better mechanical properties. Particularly, ZrO2 and Y2O3 composites
on Ti-6Al-4V have shown enhancement on mechanical characteristics and increased
bioactivity [42, 43]. It has also been discovered that bioceramic-free composites, like
porcelain wollastonite, have improved mechanical characteristics. Also, an animal
investigation using composites of HAP and bio-inert alumina demonstrated excellent
osseointegration with bone [56, 57].
Metal oxides like ZnO, Al2O3 , TiO3 , and SiO2 are examples of inorganic materials
that can be coupled with novel, high-performance materials. Thanks to research on
polymer (organic)-inorganic composites. To enhance adhesion and microstructural
homogeneity, a homogeneous PEEK/bioactive glass composite coating was created
for NiTi. The PEEK polymer, which was present in the composite, provided the mate-
rial with good resistance to chemical and tribological effects as well as high strength,
according to the researchers [58]. The different materials used for manufacturing
biomaterials for medical devices are as shown in Fig. 3.
Additionally, these particles can pass through defenses like the blood-brain barrier
because they are small enough. Regulatory restrictions are required for AM materials
that come into touch with the human body in order to address these risks. The FDA
has recognized the value of personalized medicine and the potential of AM in the next
years. Devices developed using AM methods were to be subject to a set of regulatory
standards to be established by the FDA. In May 2016, the FDA released a drafting
guidance for 3D-printed medical devices that included its most recent opinions on
device design and testing [64, 65].
The primary obstacle the business is now facing is acquiring FDA certification
for the materials used in additive manufacturing. There needs to be minimal variance
in mechanical attributes from build to build and testing criteria need to be estab-
lished. There are now just three ASTM standards in use for testing materials made
via additive manufacturing. Additionally, the FDA is responsible for ensuring that
patient-specific devices, also known as objection-of-care products, adhere to regu-
latory standards. These devices, along with those used for pharmaceutical and cell
treatment produced via AM, are not yet covered by the drafted guidelines [25].
9 Future Prospectives
10 Conclusion
In conclusion, while implantable devices have come a long way through years of
research in biomaterial science, they still have drawbacks that limit their application
in comparison with non-surgical or grafting techniques. However, the emerging tech-
nology of additive manufacturing (AM) has the capacity to revolutionize the way we
think about design and develop novel devices. By allowing for direct control over both
bulk and surface characteristics in the three-dimensional space, AM enables the use
of materials that were previously unsuitable for certain applications. Additionally,
surface modification methods such as nanometer-scale electrochemical anodizing
have the potential to improve cellular interactions and protein adhesion in biomed-
ical applications. With AM, we can look forward to personalized implants being
created for each patient that are reliable, match their demands, and become the stan-
dard treatment for the many people throughout the world who are afflicted with
illness and strategy that leverages.
References
1. Patel, N.R., Gohil, P.P.: A review on biomaterials: scope, applications & human anatomy
significance. Int. J. Emerg. Technol. Adv. Eng. 2(4), 91–101 (2012)
2. Ratner, B.D., Hoffman, A.S., Schoen, F.J., Lemons, J.E.: Biomaterials Science: An Introduction
to Materials in Medicine. Elsevier (2004)
3. Kulinets, I.: Biomaterials and their applications in medicine. In: Regulatory Affairs for
Biomaterials and Medical Devices, pp. 1–10. Woodhead Publishing (2015)
4. Martin, J.L., Norris, B.J., Murphy, E., Crowe, J.A.: Medical device development: the challenge
for ergonomics. Appl. Ergon. 39(3), 271–283 (2008)
490 B. Soni et al.
5. Madalina Mihai, M., Maria Holban, A., Giurcaneanu, C., Gabriela Popa, L., Mihaela Oanea,
R., Lazar, V., Carmen Chifiriuc, M., Popa, M., Ioan, P.M.: Microbial biofilms: impact on
the pathogenesis of periodontitis, cystic fibrosis, chronic wounds and medical device-related
infections. Curr. Top. Med. Chem. 15(16), 1552–1576 (2015)
6. Agarwal, R., García, A.J.: Surface modification of biomaterials. In: Principles of Regenerative
Medicine, pp. 651–660). cademic Press (2019)
7. Fabbri, P., Messori, M.: Surface modification of polymers: chemical, physical, and biolog-
ical routes. In: Modification of Polymer Properties, pp. 109–130. William Andrew Publishing
(2017)
8. Merola, M., Affatato, S.: Materials for hip prostheses: a review of wear and loading
considerations. Materials 12(3), 495 (2019)
9. Abraham, A.M., Venkatesan, S.: A review on application of biomaterials for medical and dental
implants. Proc. Inst. Mech. Eng. Part L J. Mater. Des. Appl. 237(2), 249–273 (2023)
10. Li, S., Wang, S., Liu, W., Zhang, C., Song, J.: Current strategies for enhancement of the
bioactivity of artificial ligaments: a mini-review. J. Orthop. Transl. 1(36), 205–215 (2022)
11. Milinkovic, D.D., Zimmermann, F., Balcarek, P.: Medial patellofemoral ligament reconstruc-
tion using nonresorbable sutures yields comparable outcomes to reconstruction with a pedicled
quadriceps tendon autograft when performed in addition to bony risk factor correction. Knee
Surg. Sports Traumatol. Arthrosc. 31(1), 264–271 (2023)
12. Wang, Y., Li, G., Yang, L., Luo, R., Guo, G.: Development of innovative biomaterials and
devices for the treatment of cardiovascular diseases. Adv. Mater. 34(46), 2201971 (2022)
13. Bui, H.T., Khair, N., Yeats, B., Gooden, S., James, S.P., Dasi, L.P.: Transcatheter heart valves:
a biomaterials perspective. Adv. Healthcare Mater. 10(15), 2100115 (2021)
14. Raval, N., Kalyane, D., Maheshwari, R., Tekade, R.K.: Surface modifications of biomaterials
and their implication on biocompatibility. In: Biomaterials and Bionanotechnology, pp. 639–
674. Academic Press (2019)
15. Kurella, A., Dahotre, N.B.: Surface modification for bioimplants: the role of laser surface
engineering. J. Biomater. Appl. 20(1), 5 (2005)
16. Steckbeck, S.: Experimental Analysis of Interactions between Biomolecules and Inorganic
Surfaces. Doctoral dissertation, Bremen, Universität Bremen, Diss. (2014)
17. Amani, H., Arzaghi, H., Bayandori, M., Dezfuli, A.S., Pazoki-Toroudi, H., Shafiee, A., Moradi,
L.: Controlling cell behavior through the design of biomaterial surfaces: a focus on surface
modification techniques. Adv. Mater. Interfaces 6(13), 1900572 (2019)
18. Bose, S., Robertson, S.F., Bandyopadhyay, A.: Surface modification of biomaterials and
biomedical devices using additive manufacturing. Acta Biomater. 15(66), 6–22 (2018)
19. Brunette, D.M., Tengvall, P., Textor, M., Thomsen, P.: Titanium in Medicine: Material Science,
Surface Science, Engineering, Biological Responses and Medical Applications. Springer,
Berlin (2001)
20. Ahirwar, H., Zhou, Y., Mahapatra, C., Ramakrishna, S., Kumar, P., Nanda, H.S.: Materials
for orthopedic bioimplants: modulating degradation and surface modification using integrated
nanomaterials. Coatings 10(3), 264 (2020)
21. Ohmori, H., Katahira, K., Nagata, J., Mizutani, M., Komotori, J.: Improvement of corrosion
resistance in metallic biomaterials using a new electrical grinding technique. CIRP Ann. 51(1),
491–494 (2002)
22. Callister, W.D.: Fundamentals of Materials Science and Engineering. Wiley, London (2000)
23. Priyadarshini, B., Rama, M., Chetan, V.U.: Bioactive coating as a surface modification tech-
nique for biocompatible metallic implants: a review. J. Asian Ceram. Soc. 7(4), 397–406
(2019)
24. Gibson, I.R., Bonfield, W.: Preparation and characterization of magnesium/carbonate co-
substituted hydroxyapatites. J. Mater. Sci. Mater. Med. 13(7), 685–693 (2002)
25. Kulkarni, M., Mazare, A., Schmuki, P., Iglič, A., Seifalian, A.: Biomaterial surface modification
of titanium and titanium alloys for medical applications. Nanomedicine 111(615), 111 (2014)
26. Festas, A.J., Ramos, A., Davim, J.P.: Medical devices biomaterials—a review. Proc. Inst. Mech.
Eng. Part L J. Mater. Des. Appl. 234(1), 218–228 (2020)
Surface-Modified Biomaterials in Medical Device Development 491
27. Lara, N., Neto, A.F., da Mota, A.J., de Souza, A.E., Brito, A.G., Da Silva, A.C., Martinez,
A.C., Nozaki, A.P., da Silva, A.A., de Vasconcelos Ferraz, A., Moraes, Â.M.: Engenharia de
Materiais: materializando o futuro. Pimenta Cultural (2022)
28. Soni, R., Pande, S., Kumar, S., Salunkhe, S., Natu, H., Hussein, H.M.: Wear characterization
of laser cladded Ti-Nb-Ta alloy for biomedical applications. Crystals 12(12), 1716 (2022)
29. Su, Y., Luo, C., Zhang, Z., Hermawan, H., Zhu, D., Huang, J., Liang, Y., Li, G., Ren, L.:
Bioinspired surface functionalization of metallic biomaterials. J. Mech. Behav. Biomed. Mater.
1(77), 90–105 (2018)
30. Devgan, S., Sidhu, S.S.: Evolution of surface modification trends in bone related biomaterials:
a review. Mater. Chem. Phys. 15(233), 68–78 (2019)
31. Bandyopadhyay, A., Mitra, I., Shivaram, A., Dasgupta, N., Bose, S.: Direct comparison of addi-
tively manufactured porous titanium and tantalum implants towards in vivo osseointegration.
Addit. Manuf. 1(28), 259–266 (2019)
32. Hojjatzadeh, S.M., Parab, N.D., Yan, W., Guo, Q., Xiong, L., Zhao, C., Qu, M., Escano, L.I.,
Xiao, X., Fezzaa, K., Everhart, W.: Pore elimination mechanisms during 3D printing of metals.
Nat. Commun. 10(1), 3088 (2019)
33. Ni, J., Ling, H., Zhang, S., Wang, Z., Peng, Z., Benyshek, C., Zan, R., Miri, A.K., Li, Z., Zhang,
X., Lee, J.: Three-dimensional printing of metals for biomedical applications. Mater. Today
Bio. 1(3), 100024 (2019)
34. Sing, S.L., Tey, C.F., Tan, J.H., Huang, S., Yeong, W.Y.: 3D printing of metals in rapid
prototyping of biomaterials: techniques in additive manufacturing. In: Rapid Prototyping of
Biomaterials, pp. 17–40. Woodhead Publishing (2020)
35. Liu, X., Chu, P.K., Ding, C.: Surface modification of titanium, titanium alloys, and related
materials for biomedical applications. Mater. Sci. Eng. R. Rep. 47(3–4), 49–121 (2004)
36. Mondy, W.L., Cameron, D., Timmermans, J.P., De Clerck, N., Sasov, A., Casteleyn, C.,
Piegl, L.A.: Computer-aided design of microvasculature systems for use in vascular scaffold
production. Biofabrication 1(3), 035002 (2009)
37. Setti, L., Fraleoni-Morgera, A., Ballarin, B., Filippini, A., Frascaro, D., Piana, C.: An ampero-
metric glucose biosensor prototype fabricated by thermal inkjet printing. Biosens. Bioelectron.
20(10), 2019–2026 (2005)
38. Farahani, R.D., Dubé, M., Therriault, D.: Three-dimensional printing of multifunctional
nanocomposites: manufacturing techniques and applications. Adv. Mater. 28(28), 5794–5821
(2016)
39. Stevens, M.M., George, J.H.: Exploring and engineering the cell surface interface. Science
310(5751), 1135–1138 (2005)
40. Elfick, A.P., Green, S.M., Krikler, S., Unsworth, A.: The nature and dissemination of UHMWPE
wear debris retrieved from periprosthetic tissue of THR. J. Biomed. Mater. Res. Part A Off.
J. Soc. Biomater. Japan. Soc. Biomater. Aust. Soc. Biomater. Korean Soc. Biomater. 65(1),
95–108 (2003)
41. Roy, M., Balla, V.K., Bandyopadhyay, A., Bose, S.: MgO-doped tantalum coating on Ti:
microstructural study and biocompatibility evaluation. ACS Appl. Mater. Interfaces 4(2),
577–580 (2012)
42. Bai, L., Gong, C., Chen, X., Sun, Y., Zhang, J., Cai, L., Zhu, S., Xie, S.Q.: Additive
manufacturing of customized metallic orthopedic implants: materials, structures, and surface
modifications. Metals 9(9), 1004 (2019)
43. Darsell, J., Bose, S., Hosick, H.L., Bandyopadhyay, A.: From CT scan to ceramic bone graft.
J. Am. Ceram. Soc. 86(7), 1076–1080 (2003)
44. Bose, S., Fielding, G., Tarafder, S., Bandyopadhyay, A.: Understanding of dopant-induced
osteogenesis and angiogenesis in calcium phosphate ceramics. Trends Biotechnol. 31(10),
594–605 (2013)
45. Tarafder, S., Bose, S.: Polycaprolactone-coated 3D printed tricalcium phosphate scaffolds for
bone tissue engineering: in vitro alendronate release behavior and local delivery effect on in vivo
osteogenesis. ACS Appl. Mater. Interfaces 6(13), 9955–9965 (2014)
492 B. Soni et al.
46. Khaled, S.A., Burley, J.C., Alexander, M.R., Roberts, C.J.: Desktop 3D printing of controlled
release pharmaceutical bilayer tablets. Int. J. Pharm. 461(1–2), 105–111 (2014)
47. Song, S.W., Hidajat, K., Kawi, S.: Functionalized SBA-15 materials as carriers for controlled
drug delivery: influence of surface properties on matrix−drug interactions. Langmuir 21(21),
9568–9575 (2005)
48. Stallard, C.P., Solar, P., Biederman, H., Dowling, D.P.: Deposition of non-fouling PEO-like
coatings using a low temperature atmospheric pressure plasma jet. Plasma Processes Polym.
13(2), 241–252 (2016)
49. Gloria, A., Causa, F., Russo, T., Battista, E., Della Moglie, R., Zeppetelli, S., De Santis, R.,
Netti, P.A., Ambrosio, L.: Three-dimensional poly (ε-caprolactone) bioactive scaffolds with
controlled structural and surface properties. Biomacromol 13(11), 3510–3521 (2012)
50. Puppi, D., Chiellini, F., Piras, A.M., Chiellini, E.: Polymeric materials for bone and cartilage
repair. Prog. Polym. Sci. 35(4), 403–440 (2010)
51. Kurtz, S.M., Devine, J.N.: PEEK biomaterials in trauma, orthopedic, and spinal implants.
Biomaterials 28(32), 4845–4869 (2007)
52. Dawson, J.I., Wahl, D.A., Lanham, S.A., Kanczler, J.M., Czernuszka, J.T., Oreffo, R.O.: Devel-
opment of specific collagen scaffolds to support the osteogenic and chondrogenic differentiation
of human bone marrow stromal cells. Biomaterials 29(21), 3105–3116 (2008)
53. Inzana, J.A., Olvera, D., Fuller, S.M., Kelly, J.P., Graeve, O.A., Schwarz, E.M., Kates, S.L.,
Awad, H.A.: 3D printing of composite calcium phosphate and collagen scaffolds for bone
regeneration. Biomaterials 35(13), 4026–4034 (2014)
54. Yu, D.G., Branford-White, C., Ma, Z.H., Zhu, L.M., Li, X.Y., Yang, X.L.: Novel drug delivery
devices for providing linear release profiles fabricated by 3DP. Int. J. Pharm. 370(1–2), 160–166
(2009)
55. Lee, K.Y., Mooney, D.J.: Alginate: properties and biomedical applications. Prog. Polym. Sci.
37(1), 106–126 (2012)
56. Balamurugan, A., Balossier, G., Michel, J., Ferreira, J.M.: Electrochemical and structural eval-
uation of functionally graded bioglass-apatite composites electrophoretically deposited onto
Ti6Al4V alloy. Electrochim. Acta 54(4), 1192–1198 (2009)
57. Stojanovic, D., Jokic, B., Veljovic, D., Petrovic, R., Uskokovic, P.S., Janackovic, D.: Bioactive
glass–apatite composite coating for titanium implant synthesized by electrophoretic deposition.
J. Eur. Ceram. Soc. 27(2–3), 1595–1599 (2007)
58. Boccaccini, A.R., Peters, C., Roether, J.A., Eifler, D., Misra, S.K., Minay, E.J.: Electrophoretic
deposition of polyetheretherketone (PEEK) and PEEK/Bioglass® coatings on NiTi shape
memory alloy wires. J. Mater. Sci. 41, 8152–8159 (2006)
59. Lerouge, S., Guignot, C., Tabrizian, M., Ferrier, D., Yagoubi, N., Yahia, L.H.: Plasma-based
sterilization: effect on surface and bulk properties and hydrolytic stability of reprocessed
polyurethane electrophysiology catheters. J. Biomed. Mater. Res. 52(4), 774–782 (2000)
60. Hooper, K.A., Cox, J.D., Kohn, J.: Comparison of the effect of ethylene oxide and γ-irradiation
on selected tyrosine-derived polycarbonates and poly (L-lactic acid). J. Appl. Polym. Sci.
63(11), 1499–1510 (1997)
61. Simmons, A.: Future trends for the sterilisation of biomaterials and medical devices. In:
Sterilisation of Biomaterials and Medical Devices, pp. 310–320. Woodhead Publishing (2012)
62. Srivastava, M., Rathee, S., Maheshwari, S., Kundra, T.K.: Additive Manufacturing: Fundamen-
tals and Advancements. CRC Press (2019)
63. Guddati, S., Kiran, A.S., Leavy, M., Ramakrishna, S.: Recent advancements in additive manu-
facturing technologies for porous material applications. Int. J. Adv. Manuf. Technol. 105,
193–215 (2019)
64. Morrison, R.J., Kashlan, K.N., Flanangan, C.L., Wright, J.K., Green, G.E., Hollister,
S.J., Weatherwax, K.J.: Regulatory considerations in the design and manufacturing of
implantable 3D-printed medical devices. Clin. Transl. Sci. 8(5), 594–600 (2015)
65. Schuh, J.C., Funk, K.A.: Compilation of international standards and regulatory guidance
documents for evaluation of biomaterials, medical devices, and 3-D printed and regenerative
medicine products. Toxicol. Pathol. 47(3), 344–357 (2019)
Surface-Modified Biomaterials in Medical Device Development 493
66. Harawaza, K., Cousins, B., Roach, P., Fernandez, A.: Modification of the surface nanoto-
pography of implant devices: a translational perspective. Mater. Today Bio. 1(12), 100152
(2021)
67. Douglass, M., Garren, M., Devine, R., Mondal, A., Handa, H.: Bio-inspired hemocompatible
surface modifications for biomedical applications. Prog. Mater. Sci. 17, 100997 (2022)
68. Zhang, D., Chen, Q., Shi, C., Chen, M., Ma, K., Wan, J., Liu, R.: Dealing with the foreign-body
response to implanted biomaterials: strategies and applications of new materials. Adv. Func.
Mater. 31(6), 2007226 (2021)
69. Punj, S., Singh, J., Singh, K.: Ceramic biomaterials: properties, state of the art and future
prospectives. Ceram. Int. 47(20), 28059–28074 (2021)
70. Alghamdi, H.S., Jansen, J.A.: The development and future of dental implants. Dent. Mater. J.
39(2), 167–172 (2020)
R. Abdel-Karim
Abstract Nanoporous materials (NPMs) are a class of materials in the quest for
high-surface-area conductive and catalytic materials. Developing a broad spectrum of
nanoporous materials has enhanced their extensive applications in catalysis, sensing,
separation, and environmental, energy, and biomedical areas. Various types of pores
like open, closed, transport, and blind pores in the porous solid allow them to adsorb
drugs and release them in a more reproducible and predictable manner. According
to the composition of the materials, the nanomaterials used in nano drug delivery
systems (NDDSs) can be classified into organic, inorganic, and composite materials.
In this book chapter, we present an overview of newly developed synthetic strategies
for producing NPMs along with an in-depth discussion of the application of NPMs
for drug delivery. The remarkable characteristics of NPFs are highlighted throughout
this book chapter. Finally, challenges and future perspectives relating to nanoporous
materials foams are concluded.
Abbreviations
R. Abdel-Karim (B)
Department of Metallurgy, Cairo University, Giza, Egypt
e-mail: randaabdelkarim@gmail.com
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 495
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_16
496 R. Abdel-Karim
1 Introduction
As illustrated in Fig. 1, drugs have been loaded into mesoporous matrices using a
variety of approaches, including the process of physical mixing, techniques that are
solvent-based, melt operations, processes based on supercritical fluids, techniques
using microwave irradiation, the technique of co-spraying drying, and the surfactant-
assisted drug loading method [21].
In accordance with Budiman et al. [22], the solvent evaporation approach was used
to efficiently load drugs onto meso silica (MS) using chloroform as the solvent. Due
to its high efficiency and well-known unit activities like filtering and drying, this
approach is appealing.
Absorption
Physical mixing
Solvent evaporation
Incipient wetness Melting method
impregnation
Diffusion supported Co-milling
loading
Supercritical fluid technology Microwave -based
Co-spray drying
Covalent grafting
Fig. 1 Schematic illustration of different methods of drug loading into porous matrix
Nanoporous Materials as Versatile Nanoplatforms for Drug Delivery … 499
A review of the application of supercritical fluid technology for drug loading and
delivery systems was presented by Chakravarty et al. [24]. SCF is non-toxic, inert,
affordable, and ecologically benign. Many materials such as H2 O, N2 , Xe, SF6 , N2 O,
C2 H4 , CHF3 , ethylene, propylene, propane, ammonia, n-pentane, and ethanol have
been used for the SCF process, with CO2 exhibiting the best behavior.
It is one of the solvent-free techniques. The drug and mesoporous materials must
be physically mixed to achieve a homogeneous dispersion. The adsorbent is added
to the drug solution and stirred with a magnetic stirrer for an appropriate amount
of time, followed by drying at 60 ºC. This technique can be applied for preparing
multiple drugs [21].
It is one of the solvent-free techniques. Melting the drug and the carrier together at
high temperatures to create a physical mixture is the basis of the melting approach,
which is both simple and solvent-free [21]. However, drug stability and thermal
qualities are crucial and may restrict the process’s usefulness.
Table 1 Types and classifications of porous materials/carriers NPMs for drug delivery. Modified
after Ahuja et al. [26]
Pore category Pore Pore formation
aperture
1. Microporous <2 nm Mesoporous
2. Mesoporous 2> and <50 This type of pore is formed due to the presence of some
nm structural defects
3. Macroporous >50 nm The macroporous structure is a result of some structural defects
such as cracks, fissures, and etching grooves
As indicated in Table 1, and according to the International Union of Pure and Applied
Chemistry, micropores are defined as pores with a diameter of 2 nm or less, mesopores
span 2–50 nm, and macropores are defined as those greater than 50 nm in diameter
[26, 27].
As illustrated in Fig. 2, the pores in the solid particles could be irregularly shaped
and connected. The open pores are connected to the solid’s external surface and can
facilitate the drug’s diffusion through the solid. On the other hand, a closed pore is
an isolated vacancy within a solid and does not permit the diffusion of an adsorbate
through the solid materials. Blind pores do not lead to other pores or surfaces and
connect to the outside surface to the internal microporosity [26].
NPMs in DDS
Polymer Intelegent
Inorganic NPs
POPs NPs
Poloxamers
RP Polymers
CPS, Lectins
Transferrin
SELPs
Fig. 3 Classification of porous nanoparticles NPMs in drug delivery systems DDS. Abbreviations
SLPs: silk-like proteins type, ELPs: Elastin-like proteins type, PEG: Polyethylene glycol peloric
type, PLGA: Poly (lactic-co-glycolic acid), PNIPAA<: Poly (N-isoppproplylacryla-mide), PF127:
Pulronic F127, CPPs: Cell-penetrating peptides, SELPs: Silk-elestinlike protein type [28]
Porous organic substances are more frequently used in drug delivery systems due
to their higher biocompatibility, lower cost, lower cytotoxicity, and lower immuno-
genicity than inorganic nanoparticles. This section reviews the main porous organic
particles that are currently being explored and their most recent developments
[30]. The morphologies of porous organic polymeric carriers include microspheres,
porous fibers, porous microneedles, and hydrogel systems. As illustrated in Fig. 3,
several synthetic polymers as well as many natural polymers, including chitosan,
poly-sucrose, and alginate, have been used to create porous particles [31].
POPs are long-lasting porous substances produced by controlling the covalent
bonding of stiff, multi-dimensional, organic building blocks using π-conjugation.
POPs with interconnected gaps that are permeable to liquids, gases, and tiny
molecules are created by the stiff building blocks. By adjusting the linker lengths, one
502 R. Abdel-Karim
may precisely control the pore volume, while functional groups serve as a foundation
for further adjustments. POPs have an advantage over other materials because, like
metal-organic frameworks MOFs, their constituent parts can be assembled precisely
with enhanced atom economy. In addition, they can be strongly or weakly crys-
talline, and their crystallinity is dependent on how reversible the covalent connec-
tion that connects them is formed [32]. There are mainly four different types of
synthetic methods for the preparation of crystalline polymers, (i) Ionothermal, (ii)
Solvothermal, (iii) Microwave, and (iv) Mechanochemical grinding techniques [33].
Porous organic polymers (POPs) with low toxicity and high specific surface
area are ideal as drug delivery vehicles. However, their potential applications were
severely limited because POPs could not be metabolized and would accumulate in the
body. By condensing D-Mannitol (MA) and biodegradable hexa (p-formyl phenyl)
cyclotriphosphazene compounds, a well-designed and straightforwardly fabricated
acetal-linked porous organic polymer, HCTP-MA, was produced. The oxygen-rich
acetal links endowed the porous skeletons with strong polarity, which improved the
drug loading capacity. Hydrogen bonds and van der Waals’ forces helped the poly-
hydric drug loading into the porous skeleton. The pH could allow for the acetal
gatekeepers’ degradation and regulated release of cancer drugs [34]. According to
Liu et al. [35] materials with an organic composition can have either natural or
synthetic ingredients [35]. Sajid et al. [28] have investigated some natural as well
as synthetic types of polymers as efficient drug-delivery tools for protein. Natural
polymers were further subdivided into polysaccharide-based and protein-based poly-
mers. Meanwhile, synthetic polymers were subgrouped into polyesters, polyethers,
poloxamers, and recombinant protein-based polymers.
Chitosan, albumin, alginate, hydroxyapatite, and hyaluronic acid are popular natural
polymers that are currently used as drug delivery systems [36]. Additionally, some
natural polymers such as apoferritin DN-Apo [37], arginine, dextrin, polysac-
charides, poly (glycolic acid), and poly (lactic acid) are involved in polymeric
drug delivery systems [38]. Lv et al. [39] have encapsulated N-((2-hydroxy-3-
trimethylammonium) propyl) anticancer drug on chitosan chloride (HTCC), with
enhanced drug transportation across the intestinal barrier to the target site. Small
interfering RNA (siRNA)—a class of nucleic acid-based drugs—was applied as a
targeted therapy protocol in combination with polymeric, organic, and inorganic
nanoparticles.
According to Suresh et al. [40] functionalized antibodies were added to gelatin
nanoparticles. Collagen’s cousin, gelatin, is another natural molecule with a practical
application: lowering the body’s immune response to drugs being administered intra-
venously. Gelatin was synthesized via a two-step desolvation procedure. Tris-glycine
was used to quench excess glutaraldehyde. The obtained, porous gelatin hydrogels
can provide high delivery efficiency with low toxicity due to the presence of the
aldehyde groups.
Nanoporous Materials as Versatile Nanoplatforms for Drug Delivery … 503
POP morphologies, pore structures, and particular surface areas can be controlled
using template techniques, template-free techniques, mechanical techniques, and
control techniques such as electrospinning and 3D printing. The regulated release
provided by POPs’ dual action, molecular receptors, and stimuli-response to heat,
pH, and light can positively affect cancer treatment. There are many factors that
can affect the performance of polymeric DDS such as; the polymer assembly and
manufacturing with specific porous structures, hydrophilicity, functionalization, and
stimuli-responsiveness with the right choice of preparation techniques. Some poly-
mers such as sodium polystyrene sulfonate and poly (fluoro acrylic acid) can serve
as active drugs themselves [41].
According to Hao et al. [42], a triazine-based porous polymer containing thio-
phene units (2, 4, 6-tris (5-bromothiophene2-yl)-1, 3, 5-triazine [TBrTh]–1, 3, 5-
benzene-triyltriboronic acid pinacol ester [BTBPE]–CTF) was synthesized using
one-pot Suzuki cross-coupling reaction. This material exhibited good drug loading
and releasing behavior due to π-π stacking as a result of the highly conjugated struc-
ture and abundant nitrogen sites in triazine rings. Bialik et al. [43] presented a review
on the application of biodegradable synthetic polyesters as macromolecular carriers
of antihypertensive drugs.
This category of NPMPs for drug delivery includes carbon-based, metals, metal
oxides, calcium phosphates, and metal-organic frameworks (Fig. 3).
Fig. 4 Classification of carbon-based nanomaterials CBNs according to their shape and geometrical
structure
f = kH .t 1/2 (3)
f = kK. t n (4)
where f is the fraction of drug loading, kH is the constant used in the Higuchi method,
kK is called the constant used in Korsmeyer–Peppas mathematical relation, n is the
exponent, kF is called the rate constant in the first order equation, and t is the time
taken for drug release [48, 49].
Nanoporous Materials as Versatile Nanoplatforms for Drug Delivery … 505
NPSi is an excellent drug carrier based on its high porosity and surface area, biocom-
patibility, as well as biodegradability. For drug delivery systems, pore size is impor-
tant because drug molecules must be suitable in order to pass through the pores. The
quantity of medicine absorbed by pores is directly proportional to their porosity [52].
Their photoluminescent properties and simple silicon electrochemical anodization
manufacturing are two of their many benefits [53]. Some review articles can provide
significant studies about preparation technologies, properties, and application of
nanoporous silicon for drug delivery [54–56].
Yang et al. [57] synthesized highly porous Si by electrochemical etching tech-
nique. Porous Si nanoparticles are safe and have low toxicity because they degrade
into orthosilicic acid in the body. Colloidal instability, decreased blood circulation
retention times, and unpredictable behavior in vitro and in vivo systems are some
of the disadvantages of this biodegradable and biocompatible material. Hernández-
Montelongo et al. [58] synthesized nPSi layers by electrochemical etching. The
nanocomposites composed of oxidized nPSi (nPSi-Ox) microparticles cascade were
treated with chitosan (CHI) and β-cyclodextrin (βCD) biopolymers. The as-received
nPSi– βCD composites allowed a higher control in the drug release kinetic of flor-
fenicol (FF) compared to nPSi-Ox microparticles. Perrone Donnorso et al. [59]
fabricated water-soluble nanoporous silicon nanoparticles (NPS) with the anodizing
approach. Annealed nPSi can be used to load both hydrophilic and hydrophobic
drugs. They showed improved luminescent properties and can prevent aggregation
in a water solution with a higher therapeutic index compared to traditional drugs.
In 1992, liquid crystal shaping was first used to synthesize the M41S family of
silicate and alumina silicate-mesoporous materials MPS. Members of this family
506 R. Abdel-Karim
suitable polymers or functional groups, such as CN, SH, NH2 , and Cl. Multifunc-
tional DDS based on MSNs are currently under development to release anticancer
medications on demand and in a targeted way while limiting the drug’s premature
release.
Manzano et al. [73] presented an overview of synthesizing, characteristics, and
most recent applications of mesoporous silica for drug delivery. The siliation process,
as well as the co-condensation process, are the techniques applied for functionalizing
the surface of MPSs with the desired functional groups [74].
El-Ghannam et al. [75], investigated the efficiency of a porous silica–calcium
phosphate nanocomposite (SCPC) as a drug delivery vehicle for 5-Fluorouracil (5-
FU) anti-cancer in vitro and in vivo, with minimal side effects. Meihua et al. [76]
prepared superior core-shell mesoporous silica MPS coated with a CaP-hyaluronic
acid hybrid for the drug delivery of anticancer. An additional hyaluronic acid layer
was applied in order to target CD44 over-expressed cancer cells.
Chen et al. [77] provided a review on nano-encapsulated curcumin (CUR) and
its function in biomedical applications. It allows many pharmacological roles such
as anti-inflammatory, anti-bacterial, anti-cancer, anti-Alzheimer, and anti-fungal. It
has low bioavailability due to its limited absorption of water.
According to Ellahioui et al. [78] photosensitizers’ anti-cancer efficiency can be
improved through integration within MPSs, which decreases their tendency to aggre-
gate. Recently, Presumptive Drug Test (PDT) using MPSs modified with ruthenium
(III) compounds was attempted, along with the ensuing sensitization. The ruthenium
complex makes tumor cells more sensitive to light, which then destroys them.
Liu et al. [79] took a different tack, employing the MPS as a synergistic inorganic
nanohybrid instrument. Two distinct changes to the MPS surfaces turned them into
“Janus” nanoparticles. Lung cancer cells’ CD44 receptors were successfully targeted
by the HA ligand on the modified Janus MSNs, and the acidic pH of A549 cells
triggered charge reversal in the DMMA molecules- a psychoactive drug, leading to
increased absorption.
Mesoporous silica nanoparticles (MSNs) can contain pharmaceuticals and bioac-
tive drugs that can be released when a variety of stimulus-responsive molecular
“gatekeepers” or “nano valves” are activated by redux stimuli-, pH, temperature, and
magnetic field stimuli [80].
Many important references are recommended for reviewing the production, char-
acterization, and modification of NPG as a tool for drug delivery systems [81,
82]. As illustrated in Fig. 5, there are three common electrochemical techniques
for the production of NPG on a solid support. (1) electrochemical etching, (2)
electrodeposition, and (3) chemical and electrochemical dealloying [83].
Neupane et al. [84] developed NPG wires modified with thiolated β-cyclodextrin
HS-β-CD drug delivery system containing DOX- anti-tumor drugs. The drug release
from this implantable and biocompatible material was activated by pH- stimuli,
508 R. Abdel-Karim
showing a lower rate at the physiological condition and an accelerated release under
acidic conditions. NPG materials are characterized by ease of size regulation, ease of
manufacturing, mechanical and chemical stability, control of pore size, and ease of
surface modification and drug loading. On the other hand, NPG is not biodegradable
and should be removed after application.
The regulated porous architecture of np-Au is very important for the field of
nanomedicine. The stability and efficiency of drug distribution are enhanced by
capping groups on the surface of the porous gold carriers, and the mechanism of
action is determined by the size of the carriers [85].
Kang et al. [86] synthesized NPG using templated pulsed electrochemical deposi-
tion technique for producing Ag–AgAu nano-segments followed by selective etching
of the more active Ag phase. The dimercaptosuccinic acid (MSA) modified NPG
showed the best performance of light-stimulated drug-releasing response for doxoru-
bicin drug compared to those of the mercaptopropionic acid (MPA) modified NPG
and the bare NPG.
Nanoporous Materials as Versatile Nanoplatforms for Drug Delivery … 509
• Multilayered.
3)Stepwise pulsed anodozing
• Hierachical.
4) Assymetric two steps anodizing
Fig. 6 Different anodization approaches used for fabrication of NAA with different morphology
molecules and nanotube surface, the rate of drug solubility, the diffusion coefficient,
and finally the pH of the electrolyte [93].
The drug delivery system is a diffusion-controlled process, which can be studied
by Fick’s first law. The molar flux (J) of drug molecules is dependent on the concen-
tration gradient (dc/dx), as presented by Fick’s first law of diffusion presented in
Eq. (6). According to Fick’s second law presented in Eq. (7), the second derivative
of the concentration change with distance determines how fast the concentration of
a solution c is changed at a given position in space. It addresses how the gradient of
concentration can alter over time and at any distance. When a drug release complies
with Fick’s law, it is referred to as Fickian diffusion; when it does not, it is considered
as non-Fickian or anomalous diffusion [94].
dc dc
Jα orJ = D. (6)
dx dx
dc d 2c
= D. 2 (7)
dt dx
The drug release process from MOFs is characterized by controlled, delayed liber-
ation of the drugs via matrix breakdown, in contrast to other nanocarriers, which
usually release the drug molecules in a burst. BioMIL-1 MOFs containing iron
demonstrated up to 75% higher nicotinic acid loading and controlled drug delivery
compared to native MOF structures. MOF-based delivery devices have been used to
accomplish multimodal medicine administration and imaging by combining these
approaches. Notably, MOFs can carry different types of drugs utilizing the nonco-
valent approach, including drugs with controlled drug release patterns and no burst
effects, such as cisplatin and ibuprofen. Zeolite and mesoporous silica are useful for
constructing robust frameworks because they do not trigger a burst-release reaction
in MOF complexes [89].
Yang et al. [96] provided a review focusing on the stimuli and multi-stimuli-
responsive. MOFs applied for drug delivery systems. MOFs-based DDSs are char-
acterized by accurate pore size, adjustable composition and structure, controllable
size, variable functionality, high loading capacity, and improved biocompatibility.
Many metals such as Fe, Zn, Zr, Mn, Mg, and Cu are widely used for the manufac-
turing of MOFs, due to their reasonable toxicity as measured by oral lethal dose 50
(LD50). MOF-based materials are characterized by malleable structures that allow
for a wide range of morphologies, compositions, sizes, and chemical properties,
making them ideal as drug delivery systems. Finally, weak coordination connections
allow MOFs to be biodegradable [97].
The chemical characteristics of MOFs can be modified by adding some inorganic
clusters and/or organic ligands, which is not the case with other porous materials.
Some metal-organic frameworks (MOFs) have lanthanide elements, for instance,
fluoresce when exposed to ultraviolet light. Furthermore, the organic compounds
can be modified either before or after synthesis to incorporate the desired func-
tional groups [98]. Production techniques of MOFs include; microwave-assisted
solvothermal synthesis, reverse-phase microemulsion synthesis, electrochemical
synthesis, techniques of dry-gel conversion, preparation techniques using a microflu-
idics device, mechano-chemical or sonochemical synthesis, synthesis using step-by-
step, and high-throughput methods. Figure 7 summarizes the benefits and drawbacks
of the above-mentioned manufacturing techniques [99].
One of the most crucial and significant properties of MOFs’ is their capacity to
interact with biological systems in response to a variety of stimuli, including pH,
temperature, light, magnetic field, pressure, glucose level, and numerous stimuli-
responsive systems. This characteristic enhances the solubility of amorphous and
poorly soluble medicines and allows for precise and sustained drug release [100].
According to Al Haydar et al. [101], different types of biocompatible MOFs such
as Ca-MOF and Fe-MILs (53, 100, and 101) were synthesized using a solvothermal
process and applied for FBP delivery. The percentage of FBP drug release was
evaluated using the following Eq. (8);
512 R. Abdel-Karim
3) Microwave -Assisted
2) Solvothermal
Approach Solvothermal Approach
1) Nonsolvothermal Approach
- Finer and more uniform - Environmently-friendly, fast. -
- Working under normal Regulated structure and pore size
pressure, simple process. crystals than non-
solvothermal. with high yield.
- Long process duration, low - Recognized particles mono-
yield, large particles size. - Long duration process.
dispersity.
- Special device is needed.
5) Mechanochemical
4) Sonochemical Approach
Approach 6) Electrochemical
- Simple and environmently-
- No Solvent, low-cost and high- Approach
friendly with short duration.
throughput production. - Increased solids content.
- Recognized monodispersity.
-High energy and high materials - Special equipments are
- Excesive soni consumption, and remarkable required.
- Low yield process. structure change.
References
1. Hadden, M., Martinez-Martin, D., Yong, K.-T., Ramaswamy, Y., Singh, G.: Recent advance-
ments in the fabrication of functional nanoporous materials and their biomedical applications.
Materials 15, 2111 (2022)
2. Maksoud F J, Velázquez de la Paz M F, Hann A J, Thanarak J, Reilly G C, Claeyssens F,
Green N H and Zhang Y S. Porous Biomaterials for tissue engineering: a review. J. Mater.
Chem. B; 2022, 10: 8111–8165
514 R. Abdel-Karim
3. Benˇcin, M., Junkar, I., Vesel, A., Mozetiˇ, M., Igliˇ, A.: Nanoporous stainless steel materials
for body implants—review of synthesizing procedures. Nanomaterials 12, 2924 (2022)
4. Shen, S., Wu, Y., Liu, Y., Wu, D.: High drug-loading nanomedicines: progress, current status,
and prospects. Int. J. Nanomed. 12, 4085–4109 (2017)
5. Reisner, D E. Bionanotechnology II: Global Prospects. 1st ed: Taylor and Francis CRC Press;
2011. Chapter 11, Nanotechnology in biomaterials: nanoparticles as drug delivery systems;
p. 217–233
6. Zhuo, S., Zhang, F., Yu, J., Zhang, X., Yang, G., Liu, X.: PH-sensitive biomaterials for drug
delivery. Molecules 25, 5649 (2020)
7. Mazzotta, E., Tavano, L., Muzzalupo, R.: Thermo-sensitive vesicles in controlled drug delivery
for chemotherapy. Pharmaceutics 10(3), 150 (2018)
8. Jiang M, Liu Q, Zhang Y, Wang H, Zhang J, Chen M, Yue Z, Wang Z, Wei X, Shi S, Wang
M, Y Hou, Wang Z, Sheng F, Tian N, Wang Y . Construction of magnetic drug delivery system
and its potential application in tumor theranostics. Biomedicine & Pharmacotherapy 2022;
154:113545.
9. Entzian, K., Aigner, A.: Drug delivery by ultrasound-responsive nanocarriers for cancer
treatment. Pharmaceutics 13(8), 1135 (2021)
10. Xiang, W., Chen, L.: In light-sensitive Drug Delivery System Nanoparticles. Mediate
Oxidative Stress, Am J Transl Res. 12(5), 1469–1480 (2020)
11. G. Wang, Silica Nanoporous Materials Adsorption and Release Study, Syracuse University,
Dissertation, New York, USA, 2011.
12. Kharissova O V, Torres-Martínez L, Kharisov B I. Handbook of Nanomaterials and Nanocom-
posites for Energy and Environmental Applications. Springer International Publishing; 2021.
Chapter 4, Nanoporous Metallic Foams for Energy Applications: Electrochemical Approaches
for Synthesizing and Characterization; p.489–511
13. Abdelfatah A, Reda Y, Abdel-Karim R, El-Raghy S, Zohdy K M. Electrochemical characteri-
zation of electrodeposited Ni–Cu foams and their application as electrodes for supercapacitors.
Frontiers in Mechanical Engineering 2020, 6, paper number 36.
14. Ahmad I. Adv Mater Appl: Micro to Nano Scale. One Central Press, 2017. Chapter 4,
Electrochemical deposition of porous metallic foams for energy applications; p.78–91.
15. Ebothé J., Nanofabrication using Nanomaterials. One Central Press, 2016. Chapter 7,
Fabrication of Nanoporous Alumina; p. 197–218.
16. Lei, Q., Guo, J., Noureddine, A., Wang, A., Wuttke, S., Brinker, C.J., Zhu, W.: Sol–gel–based
advanced porous silica materials for biomedical applications. Adv. Funct. Mater. 30, 1909539
(2020)
17. Głowniak, S., Szcz˛eśniak, B., Choma, J., Jaroniec, M.: Advances in Microwave Synthesis of
Nanoporous Materials. Adv. Mater. 33, 2103477 (2021)
18. Aihara H, Zider J, Fanton G, and Duerig T. Combustion synthesis porous nitinol for biomedical
applications. International Journal of Biomaterials 2019; Article ID 4307461: 11 pages.
19. Bai, Z., Zhang, L., Li, H., Liu, L.: Nanopore creation in graphene by ion beam irradiation:
geometry, quality, and efficiency. ACS Appl. Mater. Interfaces 8(37), 24803–24809 (2016)
20. Wanga, C., Huang, H., Zhang, Z., Zhang, L., Yan, J., Ren, L.: Formation, evolution and
characterization of nanoporous structures on the Ti6 Al4 V surface induced by nanosecond
pulse laser irradiation. Mater. Des. 223, 111243 (2022)
21. Trzeciak, K., Chotera-Ouda, A., Bak-Sypien, I.I., Potrzebowski, M.J.: Mesoporous silica parti-
cles as drug delivery systems—The state of the art in loading methods and the recent progress
in analytical techniques for monitoring these processes. Pharmaceutics 13, 950 (2021)
22. Budiman, A., Aulifa, D.L.: Encapsulation of the drug into mesoporous silica by solvent
evaporation: A comparative study of drug characterization in mesoporous silica with various
molecular weights. Heliyon 7, e086272 (2021)
23. Jaime-Escalante, B., Melgoza-Contreras, L.M., Leyva-Gómez, G., Mendoza-Muño, N.:
Synthesis and drug loading improvements on mesoporous SBA-15 by spray drying. Drug
Dev. Ind. Pharm. 47(12), 1895–1903 (2021)
Nanoporous Materials as Versatile Nanoplatforms for Drug Delivery … 515
24. Chakravarty, P., Famili, A., Nagapudi, K., Al-Sayah, M.A.: Using supercritical fluid tech-
nology as a green alternative during the preparation of drug delivery systems. Pharmaceutics
11, 629 (2019)
25. Won TW. Use of microwave in the processing of drug delivery systems, current drug delivery
2008; 5(2):77–84.
26. Ahuja, G., Pathaka, K.: Porous carriers for controlled/modulated drug delivery. Indian J.
Pharm. Sci. 71(6), 599–607 (2009)
27. Jibowu, T.: A Review on nanoporous metals. Front Nanosci Nanotech 2(4), 165–168 (2016)
28. Sajid, M., Akash, H., Rehman, K., Chen, S.: Natural and synthetic polymers as drug carriers
for delivery of therapeutic proteins. Polym. Rev. 55(3), 371–406 (2015)
29. Ameen S, Akhtar M S and Shin H-S. Nanopores, IntechOpen; 2021. Chapter 2: Potential
Application of Nanoporous Materials in Biomedical Field; 17 pages.
30. Radhakrishnan, D., Mohanan, S., Choi, G., Choy, J.-H., Tiburcius, S., Trinh, H.T., Bolan, S.,
Verrills, N., Tanwar, P., Karakoti, A., Vinu, A.: The Emergence of nanoporous materials in
lung cancer therapy. Sci. Technol. Adv. Mater. 23(1), 225–274 (2022)
31. Sayed E, Haj-Ahmad R, Ruparelia K, Arshad M S, Chang M-w and Ahmad Z. Porous inorganic
drug delivery systems – a review. AAPS Pharm Sci Tech 2017; 18: 1507–1525.
32. Singh, N., Son, S., An, J., Kim, I., Choi, M., Kong, N., Tao, W., Kim, J.S.: Nanoscale porous
organic polymers for drug delivery and advanced cancer theranostics. Chem. Soc. Rev. 50,
12883–12896 (2021)
33. Zhang, S., Yang, Q., Wang, C., Luo, X., Kim, J., Wang, Z.: Porous organic frameworks:
advanced materials in analytical chemistry. Adv. Sci. 5, 1801116 (2018)
34. Xu, Z., Hu, L., Ming, J., Cui, X., Zhang, M., Dou, J., Zhang, W., Zhou, B.: Self-gated porous
organic polymer as drug delivery system for pH stimuli-responsive controlled quercetin.
Microporous Mesoporous Mater. 303, 110259 (2020)
35. Liu, T., Liu, G.: Porous organic materials offer vast future opportunities. Nat. Commun. 11,
4984 (2020)
36. Idrees H, Zohaib S, Zaidi J, Sabir A, Khan R U, Zhang X, and Hassan S-u. A review of
biodegradable natural polymer-based nanoparticles for drug delivery applications. Nanoma-
terials 2020; 10: 1970.
37. Luo, Y., Wang, X., Du, D., et al.: Hyaluronic acid-conjugated apoferritin nanocages for lung
cancer targeted drug delivery. Biomater Sci. 3(10), 1386–1394 (2015)
38. Sung YK and Kim S W. Recent advances in polymeric drug delivery systems. Biomaterials
Research. 2020; 24(12).
39. Lv, U.P.P., Wei, W., Yue, H., et al.: Porous quaternized chitosan nanoparticles containing
paclitaxel nanocrystals improved therapeutic efficiency in non-small-cell lung cancer after
oral administration. Biomacromol 12(12), 4230–4239 (2011)
40. Suresh, D., Zambre, A., Mukherjee, S., et al.: Silencing AXL by covalent siRNA-gelatin-
antibody nanoconjugate inactivates mTOR/EMT pathway and stimulates p53 for TKI
sensitization in NSCLC. Nanomedicine 20, 102007 (2019)
41. Tang Y, Varyambath A, Ding Y, Chen B, Huang X, Zhang Y, Yu D-g, Kim I and Song W. Porous
organic polymers for drug delivery: hierarchical pore structures, variable morphologies, and
biological properties. Biomater. Sci. 2022; 10: 5369.
42. Hao D, Wang D-Y, Dong B, Xi S-C, and Jiang G. Facile synthesis of a triazine-based
porous organic polymer containing thiophene units for effective loading and releasing of
temozolomide. e-Polymers 2022; 22: 664–675.
43. Bialik M, Kuras M, Sobczak M & Oledzka E. Biodegradable synthetic polyesters in the tech-
nology of controlled dosage forms of antihypertensive drugs – the overview, expert opinion
on drug delivery 2019; 16 (9): 953–967.
44. Xu, G., Liu, S., Niu, H., Lv, W., Wu, R.: Functionalized mesoporous carbon nanoparticles
for targeted chemo-photothermal therapy of cancer cells under near-infrared irradiation. RSC
Adv. 4(64), 33986–33997 (2014)
45. Yallappa, S., Abdul Mana, S.A., Hegde, G.: Synthesis of a biocompatible nanoporous carbon
and its conjugation with florescent dye for cellular imaging and targeted drug delivery to
cancer cells. New Carbon Mater. 33(2), 162–172 (2018)
516 R. Abdel-Karim
67. Narayan, R., Nayak, U.Y., Raichur, A.M., Garg, S.: Review mesoporous silica nanoparticles:
A comprehensive review on synthesis and recent advances. Pharmaceutics 10, 118 (2018)
68. Song, S., Hidajat, K.: Functionalized SBA-15 materials as carriers for controlled drug delivery:
influence of surface properties on matrix-drug interactions. Langmuir 21, 9568–9575 (2005)
69. Heikkilä, T., Salonen, J., Tuura, J., Kumar, N., Salmi, T., Murzin, D.Y., et al.: Evaluation of
mesoporous TCPSi, MCM-41, SBA-15, and TUD-1 materials as API carriers for oral drug
delivery. Drug Deliv. 14, 337–347 (2007)
70. Horcajada, P., Ramila, A., Perez-Pariente, J., Vallet-Regí, M.: Influence of pore size of MCM-
41 matrices on drug delivery rate. Microporous Mesoporous Mater. 68, 105–109 (2004)
71. Vallet-Regi, M., Ramila, A., Del Real, R., Pérez-Pariente, J.: A new property of MCM-41:
drug delivery system. Chem. Mater. 13, 308–311 (2001)
72. Wang, Y., Zhao, Q., Han, N., Bai, L., Li, J., Liu, J., Che, E., Hu, L., Zhang, Q., Jiang, T.,
Wang, S.: Mesoporous silica nanoparticles for drug delivery: current insights. Nanomedicine
11, 313–327 (2015)
73. Manzano, M., Vallet-Reg, M.: Mesoporous silica nanoparticles for drug delivery. Adv. Funct.
Mater. 30, 1902634 (2020)
74. Issa, A.A., Luyt, A.S.: Kinetics of alkoxysilanes and organoalkoxysilanes polymerization: A
review. Polym (Basel) 11(3), 537 (2019)
75. El Ghannam, A., Ricci, K., Malkawi, A., Jahed, K., Vedantham, K., Wyan, H., Allen, L.D.,
Dréau, D.: A ceramic-based anticancer drug delivery system to treat breast cancer. J. Mater.
Sci. Mater. Med. 21, 2701–2710 (2010)
76. Meihua, Y., Siddharth, J., Peter, T., Jiezhong, C., Wenyi, G., Chengzhong, Y.: Hyaluronic acid
modified mesoporous silica nanoparticles for targeted drug delivery to CD44-overexpressing
cancer cells. Nanomaterials 5, 178–183 (2013)
77. Chen, Y., Lu, Y., Lee, R.J., et al.: Nano encapsulated curcumin: and its potential for biomedical
applications. Int. J. Nanomedicine 15, 3099–3120 (2020)
78. Ellahioui, Y., Patra, M., Mari, C., et al.: Mesoporous silica nanoparticles functionalized
with a photoactive ruthenium (ii) complex: Exploring the formulation of a metal-based
photodynamic therapy photosensitizer. Dalton Trans. 48(18), 5940–5951 (2019)
79. Liu, Y., Dai, R., Wei, Q., et al.: Dual-functionalized janus mesoporous silica nanoparticles
with active targeting and charge reversal for synergistic tumor-targeting therapy. ACS Appl.
Mater. Interfaces 11(47), 44582–44592 (2019)
80. Karimi, M., Mirshekari, H., Aliakbari, M., Sahandi-Zangabad, P., Hamblin, M.R.: Smart
mesoporous silica nanoparticles for controlled-release drug delivery. Nanotechnol. Rev.
Nanotechnol. Rev. 5(2), 195–207 (2016)
81. Sondhi, P., Lingden, D., Bhattarai, J.K., Demchenko, A.V., Stine, K.J.: Applications of
nanoporous gold in therapy, drug delivery, and diagnostics. Metals 13, 78 (2023)
82. Shuangjue, X., Wang, W.X., Xu, P.: Nanoporous gold: A review and potentials in biotechno-
logical and biomedical applications. Nano Select. 2, 1437–1458 (2021)
83. Bhattarai, J.K., Neupane, D., Nepal, B., Mikhaylov, V., Demchenko, A.V., Stine, K.J.: Prepa-
ration, modification, characterization, and biosensing application of nanoporous gold using
electrochemical techniques. Nanomaterials 8, 171 (2018)
84. Neupane, D., Bhattarai, J.K., Demchenko, A.V., Stine, K.J.: A pH-ensitive thiolated β-
cyclodextrin-modified nanoporous gold for controlled release of doxorubicin. J Drug Deliv
Sci Technol. 60, 101985 (2020)
85. Xiao, S., Wang, S., Wang, X., Xu, P.: Nanoporous gold: A review and potentials in
biotechnological and biomedical applications. Nano Select. 2, 1437–1458 (2021)
86. Kang, T.Y., Park, K., Kwon, S.H., Chae, W.-S.: Surface-engineered nanoporous gold
nanoparticles for light-triggered drug release. Opt. Mater. 106, 109985 (2020)
87. Bae, I., Lim, K.-S., Park, J.-K., Song, J.H., Oh, S.-H., Kim, J.-W., Zhang, Z., Park, C., Koh,
J.-T.: Evaluation of cellular response and drug delivery efficiency of nanoporous stainless
steel. Material Biomaterials Research 25, 30 (2021)
88. Bao, L., Park, J., Bonfante, G., Kim, B.: Recent advances in porous microneedles: materials,
fabrication, and transdermal applications. Drug Deliv. Transl. Res. 12, 395–414 (2022)
518 R. Abdel-Karim
89. Sezer A D. IntechOpen; 2016. Chapter 3, Smart drug delivery strategies based on porous
nanostructure materials in smart drug delivery system; p. 63–90.
90. Pourmadadi, M., Farokh, A., Rahmani, E., Shamsabadipour, A., Eshaghi, M.M., Rahdar,
A., Fernando, L., Ferreira, R.: Porous alumina as potential nanostructures for drug delivery
applications, synthesis, and characteristics. Journal of Drug Delivery Science and Technology
77, 103877 (2022)
91. Kapruwan P, Ferré-Borrull J, and Marsal L F. Nanoporous anodic alumina platforms for drug
delivery applications: recent advances and perspective. Adv. Mater. Interfaces 2020; 2001133.
92. Jeon, G., Yang, S.Y., Byun, J., Kim, J.K.: Electrically actuatable smart nanoporous membrane
for pulsatile drug release. Nano Letter 11(3), 1284–1288 (2011)
93. Orosz, K.E., Gupta, S., Hassink, M., Abdel-Rahman, M., Moldovan, L., Davidoff, F.H.,
Moldovan, N.I.: Delivery of antiangiogenic and antioxidant drugs of ophthalmic interest
through a nanoporous inorganic filter. Molecule Vision 10(68), 555–565 (2004)
94. Losic, D., Aw, M.S., Santos, A., Gulati, K., Bariana, M.: Titania nanotube arrays for local
drug delivery: recent advances and perspectives. Expert Opin. Drug Deliv. 12(1), 103–127
(2015)
95. Aw, M.S., Kurian, M., Losic, D.: Non-eroding drug-releasing implants with ordered
nanoporous and nanotubular structures: concepts for controlling drug release. Biomaterials
Science 2, 10–34 (2014)
96. Yang, J., Wang, H., Liu, J., Ding, M., Xie, X., Yang, X., Peng, Y., Zhou, S., Ouyang, R.,
Miaoa, Y.: Recent advances in nanosized metal organic frameworks for drug delivery and
tumor therapy. RSC Adv. 11, 3241 (2021)
97. Wu, M.-X., Yan, Y.-W.: Metal–organic framework (MOF)-based drug/cargo delivery and
cancer therapy. Adv. Mater. 29, 1606134 (2017)
98. He, S., Wua, L., Li, X., Sun, H., Xiong, T., Liu, J., Huang, C., Xua, H., Sunf, H.,
Chene, W., Gref, R., Zhang, J.: Metal-organic frameworks for advanced drug delivery. Acta
Pharmaceutica Sinica B 11(8), 2362–2395 (2021)
99. Sun Y, Zheng L, Yang Y, Qian X, Fu T, Li X, Yang Z, Yan H, Cui C, Tan W. Metal–organic
framework nanocarriers for drug delivery in biomedical applications. Nano-Micro Lett. 2020;
12:103, 29 pages.
100. Maranescu, B., Visa, A.: Applications of metal-organic frameworks as drug delivery systems.
Int. J. Mol. Sci. 23, 4458 (2022)
101. Al Haydar, M., Abid, H.R., Sunderland, B., Wang, S.: Metal-organic frameworks as a drug
delivery system for flurbiprofen. Drug Des. Dev. Ther. 11, 2685–2695 (2017)
102. Zhang, Y.Y., Zhang, L., Lin, X.W., et al.: Dual-responsive nanosystem for precise molecular
subtyping and resistant reversal of EGFR targeted therapy. Chem. Eng. J. 372, 483–495 (2019)
103. Sun, Q., You, Q., Pang, X., et al.: A photoresponsive and rod-shape nanocarrier: single wave-
length of light-triggered photothermal and photodynamic therapy based on AuNRs-capped &
Ce6-doped mesoporous silica nanorods. Biomaterials 122, 188–200 (2017)
104. Zhao, S., Sun, S., Jiang, K., et al.: In situ synthesis of fluorescent mesoporous silica–carbon dot
nanohybrids featuring folate receptor-overexpressing cancer cell targeting and drug delivery.
Nano-Micro Lett. 11(1), 32 (2019)
Nanoporous Materials as Versatile Nanoplatforms for Drug Delivery … 519
Abbreviations
AF Angiogenic factor
BM-MSC Bone marrow derived mesenchymal stem cell
CABG Coronary Artery Bypass Surgery
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 521
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_17
522 G. Baradwaj et al.
CB Cardiovascular Biomaterials
CVD Cardiovascular disease
DDS Drug Delivery Systems
ECM Extracellular Matrix
GAGs Glycosaminoglycans
MI Myocardial Infarction
PCL Polycaprolactone
PEG Polyethylene glycol
PET Polyethylene terephthalate
PHEMA Poly (2-hydroxyethyl methacrylate)
PNIPAAM Poly (N-isopropyl acrylamide)
SM Stem cell
TE Tissue Engineering
VEGF Vascular Endothelial Growth Factor
3D 3 Dimensional
1 Introduction
A multitude of illnesses affecting the heart and blood vessels are referred to as “car-
diovascular diseases” (CVDs) [1, 2]. They consist of cerebrovascular disease, coro-
nary heart disease, congenital heart disease, pulmonary embolism, deep vein throm-
bosis, etc. [2–6]. Myocardial infractions and strokes commonly occur suddenly. A
blockage that halts the flow of blood to the heart or brain, respectively, is the main
cause of the acute episodes. The most prevalent cause of this is fatty buildup on
the inside surfaces of blood arteries that transport blood to the brain or heart [3, 4].
Strokes may be caused by blood clots or hemorrhages from a brain blood vessel. The
word “biomaterial” refers to any material that interacts with biological systems. They
can be created in the lab using metallic, polymer, ceramic, or composite materials, or
they can be natural or synthetic materials derived from nature [5–7]. To enhance or
replace a natural function, biomaterials are commonly used in medical devices [3–
5]. Examples include hip replacements, heart valves, and supplies frequently used in
surgery and dentistry. Biomaterials play a crucial part in modern medicine, helping
patients recover from illness or damage by restoring function [6]. In therapeutic
settings, biomaterials are employed to sustain, enhance, or replace damaged tissue
or bioactivity. Ancient Egyptians were the first to use biomaterials; they did it by
using sutures made of animal sinew [2, 5, 6]. The study of biomaterials nowadays is
influenced by a variety of scientific fields, including engineering, medical science,
biology, physics, and chemistry [5]. The growth of field has been significant over
the past decade as a consequence of developments in TE, regenerative medicine, and
other fields. Implants for hearing loss, dental implants, prosthetic joints, ligaments,
tendons, heart valves, stents, and grafts are all examples of medical implants [7–9].
Using dissolvable bandages, stitches, clamps, and staples to close wounds can all
Development of Cardiovascular Biomaterials From Collagenous Tissues 523
hasten the healing of human tissues [10]. Cells, bioactive substances, and biomate-
rial scaffolds have all been used to regenerate human tissues [3, 8, 10]. One example
is a lab-grown human bladder, and another is a hydrogel that regenerates bone [8–
11]. Additionally, biocompatible molecular probes and nanoparticles are employed
to help cancer molecular imaging and therapy [12]. Biosensors are used to quantify
and communicate information about the presence and amount of certain chemicals.
Sensors that gauge brain activity and blood glucose levels are two examples [10–12].
Researchers are looking towards drug delivery systems that either apply or transport
drugs to a medical goal [5–8]. Examples include vascular stents coated with drugs
and implanted chemotherapy wafers for cancer patients [8–10]. The cardiovascular
system is one of the most significant applications for biomaterials. Cardiovascular
biomaterials are anticipated to account for most of the market for biomaterials in
2014, with a value of around $20.7 billion [10–12]. Cardiovascular biomaterials
(CB) must mix with the surrounding environment and be compatible with blood
to be employed [5–8]. When utilizing CB, two important considerations should be
given equal weight: the material’s physical and mechanical qualities, including its
strength, deformation, fatigue, creep, resistance to friction and wear, flow character-
istics, pressure loss, and other designed features [7–9]. There should also be consid-
eration for biocompatibility, which describes how a substance interacts with tissue
[8]. To analyze and evaluate these features, various in vivo and in vitro experiments
must be carried out [7–10].
that it is challenging to give such hydrogels the essential stiffnesses, more advanced
molecular engineering techniques may be required [13–15]. The development of
“small molecular hydrogels” is dependent on peptides like D FEFKD FEFKYRGD,
which in turn has been proven to provide a scaffold for mesenchymal stem cells
altered with hepatocyte growth factor to maintain cardiac rhythm, post infarction
while reducing ventricular remodeling in an animal model [17, 18]. It is important
to investigate this approach even if these are obviously quite different from the
previously established cellular engineering scaffolds [15–18].
Collagen is perhaps the most widely utilized organic polymer for uses in tissue engi-
neering since it is present in the ECM of almost all human tissues [11–14]. Since the
first analysis of the relationship between cells and acquired collagen was conducted
in the early twentieth century, collagen has been employed as a biomaterial [15–18].
Collagen is used for biological applications because of a variety of characteristics
that make it a great material: Benefits include biodegradability, poor antigenicity, and
excellent biocompatibility. Cell receptors which can recognize a particular sequence
of peptide inside collagen molecules help to further increase cell adhesion [12, 13,
15, 16]. As an added bonus, collagen, one of the most common and well-preserved
proteins in vertebrates, may be extracted from a variety of sources. Bovine tendons
and skin are frequently used in extraction of collagen, which makes it an affordable
option for scaffold-based tissue engineering [19–22]. Since the 1970s, the applica-
tion of collagen as a framework for the construction of a DDS has piqued the interest
of numerous investigators throughout the world for multiple applications, like regen-
eration of bone, ocular, heart, and brain medicine [18–20]. Various role of collagen
in cardiovascular medicines is shown in Fig. 1.
As a result of complicated design of cardiac structure, such as heart valves, tissue
engineering solutions for heart disorders primarily rely on implantation and colo-
nization of the acellular matrix. However, the effectiveness of xenogenic cellular
heart valves remains a concern because of their high immunogenic potential and
proclivity to calcify [23–25]. This immunologic problem prompted the creation of
readily accessible human acellular scaffolding for heart and vascular reconstructive
surgery, such as Cryolife®, which provides tissue replacement spanning from valves
in the heart to vascular conduits [25, 26]. Collagen-based biomaterials are crucial for
regenerative medicine and transplantation. Collagen is still the protein of priority for
production of biomaterials due to its low immunogenicity and great biocompatibility
[23]. It can be taken from a variety of tissues and combined with other molecules like
stem cells, angiogenic factors, or grafted during CABG surgery [19, 23, 25]. In the
laboratory, it can be used to replace decellularized ECM for fundamental investiga-
tions or as a tissue substitution material for medical purposes [27–30]. The majority of
current research is directed at improving the mechanical strength, biodegradability,
Development of Cardiovascular Biomaterials From Collagenous Tissues 525
go through a range of cross-linking technologies and methods that are suitable for
different tissue sources and species of origin [35–37].
Collagen can be utilized as a vascular implant (or stent), a component of artificial
heart valves, and an outer coating for cardiovascular stents since it is “hypoaller-
genic,” or less prone to induce allergies [37–39]. Heydarkhan-Hagvall et al. devel-
oped a 3D electrospinning hybrid scaffold for cardiovascular tissue engineering that
utilizes collagen/elastin/polycaprolactone (PCL) [37, 38]. Recently, a functional,
bilayer PCL/silica nanofibrous and PCL/collagen vascular graft with a tiny diameter
was created. Its makeup encourages quick healing and delays blood vessel aging or
replacement by native blood vessels [38–40].
Although collagen has been widely used in the synthesis of biomaterials in recent
years, it comes with a lot of benefits and drawbacks. As the primary protein in
all connective tissues and a component of ECM, the collagen family is one of the
most prevalent protein families, and has been recognized for a long time as having the
potential to serve as the foundation for biomaterials, either as natural, unaltered tissue
grafts or as manufactured goods for cardiac repair [41]. It has been proposed that
therapy using an injectable collagen-based matrix might mediate apoptosis, vascular-
ization, and regeneration to offer functional recovery in models of ischemia, necrosis,
and/or infarction. The adhesion of cells to scaffold and grow has been facilitated by
the use of them as matrices in conjunction with various anchoring materials [42, 43].
Collagen matrix has been shown to mediate several healing pathways and stop the
progression of cardiac decompensation after MI. The healing of the myocardium is
also observed to be significantly impacted by the timing of the collagen hydrogel
application. Timing of collagen matrix hydrogel distribution impacts its ability to
inhibit adverse remodeling, minimize scar size, and enhance infarcted heart perfor-
mance [44]. Collagen is typically added to hydrogels formed of fibrin to boost their
mechanical strength and to attract endothelial cells for heart regeneration. A study
using hydrogels has demonstrated that the production of cardiac cells from embryonic
cells may be controlled by the addition of collagen to the ECM. Hydrogels with lower
concentrations of collagen in the ECM are less capable of differentiating hESC into
cardiac cells than those with higher collagen content [45, 46]. Another key advantage
of collagen as a biomaterial is that there is very little chance that an immune reaction
will arise. In addition to this collagen has the capability to interact with the host and
either support new host tissue or act as a scaffold for the production of new tissue
before reabsorption [47]. In order to reduce blood loss and improve biocompati-
bility with the surrounding tissues, collagen is also employed as a coating material
for PET grafts [48]. Another interesting characteristic of collagen is its composite
formation with other polymers. Increased angiogenesis and thickness of ventricular
wall were seen after a VEGF-collagen patch was put onto a heart with a damaged
right ventricular free wall. When given with a fibrin glue hydrogel rather than alone,
Development of Cardiovascular Biomaterials From Collagenous Tissues 527
Despite the enormous discoveries and improvements made in the field of cardiac
tissue engineering, a lot more research is still needed to effectively treat cardiovas-
cular illnesses, which are a significant global concern. One such field is the application
of biomaterials. Biomaterials are widely employed in the disciplines of regenerative
medicine and tissue engineering. While there are number of different biomaterials
accessible today that are utilized to treat different heart problems, these materials do
offer a risk to the body, and the rate of cardiac muscle regeneration is not always
100%. Much research is presently being conducted in this area to reduce problems
from the employment of these biomaterials and improve the effectiveness of the
therapy. Finding the appropriate biomaterial-cell type combination for tissue regen-
eration is the aim of biomaterials and tissue engineering. The danger of producing a
substrate for arrhythmia is the main worry with employing biomaterials to treat heart
illness. Although this topic hasn’t received much attention in the literature, a recent
study did discover that the biomaterials’ spreading characteristics have an impact on
how much the electrophysiological alterations are altered [67]. We need to identify
beneficial cells that stimulate myogenesis, reroute apoptotic mechanisms, and restart
the dormant-cell process in order to treat heart failure. More research is required
on the type of biomaterial, dose, delivery technique, and timing. It is crucial to get
through these obstacles and increase our knowledge of injectable cardiac tissue engi-
neering from every angle [68]. Another difficulty is to limit the immune response to
avoid the encapsulation of biomaterials or tissue constructions since doing so hinders
transplanted cells from completely integrating with their original environment. As
the heart is the body’s main source of bioelectricity, creating conductive polymers
that can help cardiomyocytes beat in unison will improve communication between
transplanted tissues and the native myocardium [69, 70]. This is crucial because
electrical field stimulation improves electrical signal propagation, cell polarization,
and protein structure [71, 72].
Despite these difficulties, research in biomaterials and tissue engineering has
achieved significant strides over the past two decades and holds out a great amount
of hope. The end goal is to partially or fully regenerate an organ using biomaterial
scaffolds in conjunction with the proper cell types. In the end, this will lessen the
need for organ transplants, treat present irreparable illnesses, and enhance quality
of life. More study is required in this field, but injectable cardiac tissue engineering
combined with previously existing medications may reduce mortality and enhance
quality of life in MI patients [72].
Development of Cardiovascular Biomaterials From Collagenous Tissues 529
2 Conclusion
The extraction and modification of collagen for the creation of biomaterials for
cardiac tissue engineering are the main topics of current research in this area. Bioma-
terials based on collagen are a great option for transplantation and regenerative
medicine. Collagen continues to be the protein of choice for creating biomaterials
due to its great biocompatibility and low immunogenicity. It may be taken out of
many tissues and mixed with other substances. It can be used to replace decellu-
larized ECM in the lab for basic research or as a tissue replacement for medic-
inal applications. Collagen can also be used in conjunction with other polymers to
improve cell adherence and proliferation of cardiac tissues. Other naturally occur-
ring polymers can also be used to treat cardiovascular diseases. In order to improve
collagen-based composites’ mechanical strength, biodegradability, or transport prop-
erties for medical applications, the majority of current research is concentrated in this
area. Further study attempts to incorporate natural polymers into more challenging
procedures and formulations of biodegradable, injectable, and mechanically stronger
components for restoring the performance of diverse organs.
References
11. Liu, Z., Wang, H., Wang, Y., et al.: The influence of chitosan hydrogel on stem cell engraftment,
survival and homing in the ischemic myocardial microenvironment. Biomaterials 33(11), 3093–
3106 (2012)
12. Spadaccio, C., Chello, M., Trombetta, M., Rainer, A., Toyoda, Y., Genovese, J.A.: Drug
releasing systems in cardiovascular tissue engineering. J. Cell. Mol. Med. 13(3), 422–439
(2009)
13. Bella, J., Hulmes, D.J.: Fibrillar collagens. Fibrous proteins: Structures and mechanisms. 457–
90 (2017)
14. Boccafoschi, F., Habermehl, J., Vesentini, S., Mantovani, D.: Biological performances of
collagen-based scaffolds for vascular tissue engineering. Biomaterials 26(35), 7410–7417
(2005)
15. Bou-Gharios, G., Ponticos, M., Rajkumar, V., Abraham, D.: Extra-cellular matrix in vascular
networks. Cell Prolif. 37(3), 207–220 (2004)
16. Furthmayr, H., Timpl, R.: Immunochemistry of collagens and procollagens. Int. Rev. Connect.
Tissue Res. 1(7), 61–99 (1976)
17. Copes, F., Pien, N., Van Vlierberghe, S., Boccafoschi, F., Mantovani, D.: Collagen-based tissue
engineering strategies for vascular medicine. Front. Bioeng. Biotechnol. 12(7), 166 (2019)
18. Williams, D.F.: Challenges with the development of biomaterials for sustainable tissue
engineering. Front. Bioeng. Biotechnol. 31(7), 127 (2019)
19. Hussey, G.S., Dziki, J.L., Badylak, S.F.: Extracellular matrix-based materials for regenerative
medicine. Nat. Rev. Mater. 3(7), 159–173 (2018)
20. Vunjak-Novakovic, G.: Tissue engineering of the heart: An evolving paradigm. J. Thorac.
Cardiovasc. Surg. 153(3), 593–595 (2017)
21. Saul, J.M., Williams, D.F.: Hydrogels in regenerative medicine. In: Handbook of Polymer
Applications in Medicine and Medical Devices, pp. 279–302. William Andrew Publishing
(2011)
22. Parenteau-Bareil, R., Gauvin, R., Berthod, F.: Collagen-based biomaterials for tissue engi-
neering applications. Materials 3(3), 1863–1887 (2010)
23. Prockop, D.J., Kivirikko, K.I.: Collagens: Molecular biology, diseases, and potentials for
therapy. Annu. Rev. Biochem. 64, 403–434 (1995)
24. Exposito, J.Y., Cluzel, C., Garrone, R., Lethias, C.: Evolution of collagens. Anat. Rec. 268,
302–316 (2002)
25. Miller, E.J.: Structural studies on cartilage collagen employing limited cleavage and solubi-
lization with pepsin. Biochemistry 11, 4903–4909 (1972)
26. Stachowiak, A.N., Irvine, D.J.: Inverse opal hydrogel-collagen composite scaffolds as a
supportive microenvironment for immune cell migration. J. Biomed. Mater. Res. A 85, 815–828
(2008)
27. Joyce, K., Fabra, G.T., Bozkurt, Y., Pandit, A.: Bioactive potential of natural biomaterials:
Identification, retention and assessment of biological properties. Signal Transduct. Target.
Ther. 6(1), 122 (2021)
28. Lin, K., Zhang, D., Macedo, M.H., Cui, W., Sarmento, B., Shen, G.: Advanced collagen-based
biomaterials for regenerative biomedicine. Adv. Funct. Mater. 29(3), 1804943 (2019)
29. Zhu, J., Li, Z., Zou, Y., Lu, G., Ronca, A., D’Amora, U., Liang, J., Fan, Y., Zhang, X., Sun, Y.:
Advanced application of collagen-based biomaterials in tissue repair and restoration. J. Leather
Sci. Eng. 4(1), 30 (2022)
30. Sorushanova, A., Delgado, L.M., Wu, Z., Shologu, N., Kshirsagar, A., Raghunath, R., Mullen,
A.M., Bayon, Y., Pandit, A., Raghunath, M., Zeugolis, D.I.: The collagen suprafamily: From
biosynthesis to advanced biomaterial development. Adv. Mater. 31(1), 1801651 (2019)
31. Chen, H., Xue, L., Gong, G.: Collagen-based materials in reproductive medicine and engineered
reproductive tissues. J. Leather Sci. Eng. 4(3), 1–15 (2022)
32. Ramshaw, J.A., Werkmeister, J.A., Glattauer, V.: Collagen-based biomaterials. Biotechnol.
Genet. Eng. Rev. 13, 335–382 (1996)
33. Kwon, H., Brown, W.E., Lee, C.A., Wang, D., Paschos, N., Hu, J.C., Athanasiou, K.A.:
Surgical and tissue engineering strategies for articular cartilage and meniscus repair. Nat. Rev.
Rheumatol. 15(9), 550–570 (2019)
Development of Cardiovascular Biomaterials From Collagenous Tissues 531
34. Salvatore, L., Gallo, N., Natali, M.L., Campa, L., Lunetti, P., Madaghiele, M., Blasi, F.S.,
Corallo, A., Capobianco, L., Sannino, A.: Marine collagen and its derivatives: Versatile and
sustainable bio-resources for healthcare. Mater. Sci. Eng. C 113, 110963 (2020)
35. Liu, W., Lin, H., Zhao, P., Xing, L., Li, J., Wang, Z., Ju, S., Shi, X., Liu, Y., Deng, G., Gao, G.,
Sun, L., Zhang, X.: A regulatory perspective on recombinant collagen-based medical devices.
Bioact Mater. 12, 198–202 (2022)
36. Yang, Y., Campbell Ritchie, A., Everitt, N.M.: Recombinant human collagen/chitosan-based
soft hydrogels as biomaterials for soft tissue engineering. Mater. Sci. Eng. C 121, 111846
(2021)
37. Gelse, K., Poschl, E., Aigner, T.: Collagens–structure, function, and biosynthesis. Adv. Drug
Deliv. Rev. 55(12), 1531–1546 (2003)
38. Church, R.L., Pfeiffer, S.E., Tanzer, M.L.: Collagen biosynthesis: Synthesis and secretion of a
high molecular weight collagen precursor (procollagen). Proc. Natl. Acad. Sci. U.S.A. 68(11),
2638–2642 (1971)
39. An, B., Lin, Y.S., Brodsky, B.: Collagen interactions: Drug design and delivery. Adv. Drug
Deliv. Rev. 97, 69–84 (2016)
40. Delgado, L.M., Fuller, K., Zeugolis, D.I.: Collagen cross-linking: Biophysical, biochemical,
and biological response analysis. Tissue Eng. Part A 23(19–20), 1064–1077 (2017)
41. R.H., A, N.T., P., R.G., G., S.E., R., H.G., T.: A collagen mesh prosthesis for wound repair and
hernia reinforcement. InSurgical Forum 13, 29–31 (1962)
42. Burdick, J.A., Mauck, R.L., Gorman III, J.H., Gorman R.C.: Acellular biomaterials: An
evolving alternative to cell-based therapies. Sci. Transl. Med. 5(176):176ps4 (2013)
43. Johnson, T.D., Christman, K.L.: Injectable hydrogel therapies and their delivery strategies for
treating myocardial infarction. Expert Opin. Drug Deliv. 10(1), 59–72 (2013)
44. Blackburn, N.J., Sofrenovic, T., Kuraitis, D., Ahmadi, A., McNeill, B., Deng, C., Rayner, K.J.,
Zhong, Z., Ruel, M., Suuronen, E.J.: Timing underpins the benefits associated with injectable
collagen biomaterial therapy for the treatment of myocardial infarction. Biomaterials 1(39),
182–192 (2015)
45. Chaudhuri, R., Ramachandran, M., Moharil, P., Harumalani, M., Jaiswal, A.K.: Biomaterials
and cells for cardiac tissue engineering: Current choices. Mater. Sci. Eng., C 1(79), 950–957
(2017)
46. Duan, Y., Liu, Z., O’Neill, J., Wan, L.Q., Freytes, D.O., Vunjak-Novakovic, G.: Hybrid gel
composed of native heart matrix and collagen induces cardiac differentiation of human embry-
onic stem cells without supplemental growth factors. J. Cardiovasc. Transl. Res. 4, 605–615
(2011)
47. Ramshaw, J.A., Werkmeister, J.A., Glattauer, V.: Collagen-based biomaterials. Biotechnol.
Genet. Eng. Rev. 13(1), 335–382 (1996)
48. Jaganathan, S.K., Supriyanto, E., Murugesan, S., Balaji, A., Asokan, M.K.: Biomaterials in
cardiovascular research: Applications and clinical implications. Biomed. Res. Int. 8, 2014
(2014)
49. Miyagi, Y., Chiu, L.L., Cimini, M., Weisel, R.D., Radisic, M., Li, R.K.: Biodegradable collagen
patch with covalently immobilized VEGF for myocardial repair. Biomaterials 32(5), 1280–
1290 (2011)
50. Frederick, J.R., Fitzpatrick III, J.R., McCormick, R.C., Harris, D.A., Kim, A.Y., Muenzer, J.R.,
Marotta, N., Smith, M.J., Cohen, J.E., Hiesinger, W., Atluri, P.: Stromal cell-derived factor-1α
activation of tissue-engineered endothelial progenitor cell matrix enhances ventricular func-
tion after myocardial infarction by inducing neovasculogenesis. Circulation 122(11_suppl_1),
S107–17 (2010)
51. Nair, L.S., Laurencin, C.T.: Biodegradable polymers as biomaterials. Prog. Polym. Sci. 32(8–9),
762–798 (2007)
52. Dong, C., Lv, Y.: Application of collagen scaffold in tissue engineering: Recent advances and
new perspectives. Polymers 8(2), 42 (2016)
53. Ullah, S., Chen, X.: Fabrication, applications and challenges of natural biomaterials in tissue
engineering. Appl. Mater. Today 1(20), 100656 (2020)
532 G. Baradwaj et al.
54. Ning, L.J., Zhang, Y.J., Zhang, Y., Qing, Q., Jiang, Y.L., Yang, J.L., Luo, J.C., Qin, T.W.: The
utilization of decellularized tendon slices to provide an inductive microenvironment for the
proliferation and tenogenic differentiation of stem cells. Biomaterials 1(52), 539–550 (2015)
55. Li, Q., Williams, C.G., Sun, D.D., Wang, J., Leong, K., Elisseeff, J.H.: Photocrosslinkable
polysaccharides based on chondroitin sulfate. J. Biomed. Mater. Res. Part A: Off. J. Soc.
Biomater., Jpn. Soc. Biomater., Aust. Soc. Biomater. Korean Soc. Biomater. 68(1), 28–33
(2004)
56. Nettles, D.L., Vail, T.P., Morgan, M.T., Grinstaff, M.W., Setton, L.A.: Photocrosslinkable
hyaluronan as a scaffold for articular cartilage repair. Ann. Biomed. Eng. 32, 391–397 (2004)
57. Johnson, K.A., Rogers, G.J., Roe, S.C., Howlett, C.R., Clayton, M.K., Milthorpe, B.K., Schind-
helm, K.: Nitrous acid pretreatment of tendon xenografts cross-linked with glutaraldehyde and
sterilized with gamma irradiation. Biomaterials 20(11), 1003–1015 (1999)
58. Roe, S.C., Milthorpe, B.K., True, K., Rogers, G.J., Schindhelm, K.: The effect of gamma
irradiation on a xenograft tendon bioprosthesis. Clin. Mater. 9(3–4), 149–154 (1992)
59. Cheung, D.T., Perelman, N., Tong, D., Nimni, M.E.: The effect of γ-irradiation on collagen
molecules, isolated α-chains, and crosslinked native fibers. J. Biomed. Mater. Res. 24(5), 581–
589 (1990)
60. Huang, N.F., Yu, J., Sievers, R., Li, S., Lee, R.J.: Injectable biopolymers enhance angiogenesis
after myocardial infarction. Tissue Eng. 11(11–12), 1860–1866 (2005)
61. S. Zaitseva, T., Yang, G., Dionyssiou, D., Zamani, M., Sawamura, S., Yakubov, E., Ferguson,
J., Hallett, R.L., Fleischmann, D., Paukshto, M.V., Huang, N.F.: Delivery of hepatocyte growth
factor mRNA from nanofibrillar scaffolds in a pig model of peripheral arterial disease. Regen.
Med. 15(6), 1761–73 (2020)
62. Khanna, A., Zamani, M., Huang, N.F.: Extracellular matrix-based biomaterials for cardiovas-
cular tissue engineering. J. Cardiovasc. Dev. Dis. 8(11), 137 (2021)
63. Neve, A., Cantatore, F.P., Maruotti, N., Corrado, A., Ribatti, D.: Extracellular matrix modulates
angiogenesis in physiological and pathological conditions. BioMed Res. Int. (2014)
64. Ahmed, M., Ffrench-Constant, C.: Extracellular matrix regulation of stem cell behavior. Curr.
Stem Cell Rep. 2, 197–206 (2016)
65. Ferrans, V.J., Boyce, S.W., Billingham, M.E., Jones, M., Ishihara, T., Roberts, W.C.: Calcific
deposits in porcine bioprostheses: Structure and pathogenesis. Am. J. Cardiol. 46(5), 721–734
(1980)
66. Bruck, S.D.: Problems and challenges of biomaterials in cardiovascular applications: A status
report. Biomater. Med. Devices Artif. Organs 11(4), 271–280 (1983)
67. Suarez, S.L., Rane, A.A., Muñoz, A., Wright, A.T., Zhang, S.X., Braden, R.L., Almutairi, A.,
McCulloch, A.D., Christman, K.L.: Intramyocardial injection of hydrogel with high interstitial
spread does not impact action potential propagation. Acta Biomater. 15(26), 13–22 (2015)
68. Chachques, J.C.: Cellular cardiac regenerative therapy in which patients? Expert Rev.
Cardiovasc. Ther. 7(8), 911–919 (2009)
69. Griffith, L.G., Naughton, G.: Tissue engineering—current challenges and expanding opportu-
nities. Science 295(5557), 1009–14 (2002)
70. Tandon, N., Cannizzaro, C., Chao, P.H., Maidhof, R., Marsano, A., Au, H.T., Radisic, M.,
Vunjak-Novakovic, G.: Electrical stimulation systems for cardiac tissue engineering. Nat.
Protoc. 4(2), 155–173 (2009)
71. Radisic, M., Park, H., Shing, H., Consi, T., Schoen, F.J., Langer, R., Freed, L.E., Vunjak-
Novakovic, G.: Functional assembly of engineered myocardium by electrical stimulation of
cardiac myocytes cultured on scaffolds. Proc. Natl. Acad. Sci. 101(52), 18129–18134 (2004)
72. Cui, Z., Yang, B., Li, R.K.: Application of biomaterials in cardiac repair and regeneration.
Engineering 2(1), 141–148 (2016)
Development of Cardiovascular Biomaterials From Collagenous Tissues 533
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 535
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_18
536 K. Trehan et al.
1 Introduction
The effective and precise delivery of drugs to diseased tissues is essential for treating
numerous disorders, such as cancer, cardiovascular, and inflammatory diseases [1].
However, traditional drug administration methods often lead to non-specific targeting
of different tissues and organs, requiring large amounts of drugs and causing poor
therapeutic efficacy and adverse effects to normal tissues [2]. To tackle these obsta-
cles, molecular prodrugs have been developed to increase the targeting of affected
cells and tissues and enhancement of treatment effectiveness [3]. However, designing
and preparing prodrugs can be a complex process, requiring special drug modifica-
tions for individual drugs, making it difficult to establish a standardized method
for prodrug development [4]. To tackle these challenges, an alternative approach
involves utilizing nanocarrier systems based on nanoscale materials to deliver medi-
cations to particularly affected tissues [5]. Nanoparticles used to transport drugs have
numerous advantages, including safeguarding therapeutic drugs against degradation,
allowing the administration of drugs at low dosages, prolonging the time they stay
in the body, and boosting their effectiveness in treating diseases while reducing their
impact on healthy tissues [6]. Applying specialized groups on the surface of nanopar-
ticles shields the therapeutic cargo from harsh conditions in the body, ensuring it
stays intact. Furthermore, these modified nanoparticles can be accurately directed
to affected cells and tissues, leading to better treatment outcomes with negligible
adverse effects [7].
Achieving spatiotemporal regulation over the targeted drug delivery is critical to
enhancing treatment efficiency once nanocarriers have accumulated in pathological
areas via passive or active targeting [8]. Due to the swift advances in nanobiotech-
nology, artificial biomaterials that react to stimuli have become increasingly popular
because of their possible application in medical and pharmaceutical fields, such
as medical transportation, biosensing, and tissue adhesion. These materials exhibit
quick fluctuations in structure, solubility, or polarity on utilizing external or internal
triggers, such as temperature, light, magnetic fields, pH, or enzymes [9]. By making
use of these materials as carriers for drugs on a nanoscale, it becomes possible to
transport medications with precision to specific locations in a safe and controlled
way. This method helps to prevent harmful side effects that may arise from harming
healthy tissues.
The idea of SRDD relies on the ability of biological systems to react to different
stimuli. For instance, drugs can be released as a reaction to changes in pH levels,
such as when the stomach environment changes from acidic to alkaline. Bacteria
also react to fluctuations in environmental factors, such as temperature and pH.
The development of DDSs that respond to stimuli has become feasible thanks to
advancements in nanotechnology, materials science, and chemistry. These systems
typically consist of a stimulus-sensitive component, a drug, and a carrier matrix. The
stimulus-sensitive component, which can be a polymer, liposome, or nanoparticle,
is designed to react to a specific stimulus to trigger drug release. The drug is usually
Stimuli-Responsive Material in Controlled Release of Drug 537
encapsulated or bound to the carrier matrix, which can be made from natural or
synthetic materials. Figure 1 illustrates the various forms of stimuli and stimuli-
responsive materials.
SRDDSs have several advantages, one of which is their ability for localized drug
administration, such as to tumors or inflamed tissues. For instance, pH-responsive
systems can target tumors since they have a lower pH compared to healthy tissues.
Enzyme-responsive systems, on the other hand, can target inflamed tissues where
there is a high concentration of enzymes [10]. Another benefit of stimuli-responsive
DDS is the potential to improve drug efficacy while reducing toxicity. By releasing
drugs in a controlled manner, these systems can maintain therapeutic drug levels
for longer periods, decreasing the need for frequent dosing. Moreover, they can
decrease drug toxicity by minimizing off-target effects and improving drug selec-
tivity [11]. Despite the promising potential of stimuli-responsive DDS, there are still
several challenges that need to be addressed. A significant obstacle is the insuffi-
cient perception of the complex interactions between the drug, carrier matrix, and
stimulus-sensitive component. Additionally, the stability and biocompatibility of
these systems need to be optimized to ensure their safe and effective use in vivo.
SRDDSs are categorized into two main types: endogenous and exogenous systems.
Endogenous SRDDSs utilize biological signals present in the target tissue or organ,
such as pH, enzymes, and redox reactions. These systems are highly specific and can
selectively target diseased tissues with higher levels of these biological signals. pH-
responsive and enzyme-responsive nanocarriers are instances of endogenous stimuli-
responsive systems [12]. Exogenous SRDDSs are triggered by external triggers such
as light, temperature, and magnetic fields. These systems are highly tuneable, as the
538 K. Trehan et al.
stimuli can be precisely controlled to achieve a desired drug release profile. Exam-
ples of exogenous stimuli-responsive systems include light-responsive nanoparticles,
temperature-responsive polymers, and magnetic nanoparticles [13].
feature a sulfonamide group attached to aniline. The sulfonyl group in the struc-
ture acts as an electron-withdrawing element from the nitrogen atom of the amine,
causing the hydrogen atom bound to it to become ionized [24].
Le et al. developed a hydrogel actuator with anisotropic properties by combining
pAAm and pAAc. They achieved this by immersing a pAAm hydrogel in a solu-
tion of AAc monomers and polymerizing them using photopolymerization. The
uneven distribution of pAAc within the hydrogel resulted from imperfect penetra-
tion of UV light, leading to asymmetrical swelling and shrinking responses to pH
changes [26]. Similarly, Shang et al. fabricated a composite actuator using patterned
pNIPAm/pAAc that could undergo controlled shape changes in response to pH,
ionic strength, and temperature variations. The actuator was created by immersing
a pAAc hydrogel in a NIPAm/water solution and exposing it to UV light through
a mask. At pH 11, the hydrogel bent towards the pNIPAm-rich side [27]. Huang
et al. conducted a study where a digital projector was utilized to dynamically and
spatially control light for triggering polymerization. This resulted in varying conver-
sion and crosslinking density within a p(HEA-co-HEMA-co-PSPMA) hydrogel. The
hydrogel was 3D printed and exhibited a non-uniform swelling ratio and mechanical
properties, leading to the transformation into a specific shape upon swelling [28].
Actuators based on cationic polymers have also been explored, albeit not as
extensively as anion-responsive actuators. Huang et al. demonstrated that a bilayer
hydrogel actuator made of pDMAEMA/p(DMAEMA-co-AAm) becomes increas-
ingly curved as the solution pH decreases [29]. This behavior is attributed to the
pDMAEMA layer swelling significantly at lower pH because of amine protona-
tion, in contrast to the p(DMAEMA-co-AAm) layer [30]. Furthermore, Lee et al.
demonstrated the utilization of varying crosslinking densities in the hydrogel network
to develop actuators responsive to pH changes. They employed the ionoprinting
technique along with catechol-metal coordination chemistry to manufacture a pH-
responsive actuator. By locally imprinting Fe3+ ions into poly (N-hydroxyethyl
acrylamide-co-dopamine methacrylamide) (p(HEA-co-DMA)) through the appli-
cation of an electrical potential via an iron electrode, they successfully created the
pH-responsive actuator [31].
Magnetic stimulation offers several advantages for drug delivery, such as precise
targeting of the delivery system, monitoring of concentration and distribution, and
control over the rate of drug release [32]. Iron oxide nanoparticles (IONPs) are
FDA-approved inorganic nanomaterials that respond to external magnetic fields.
They are biocompatible, low in toxicity, and easy to synthesize, making them highly
attractive for numerous studies. Due to their superparamagnetic properties, Iron oxide
nanoparticles can function as imaging enhancers for MRI and allow for drug release to
be controlled remotely using a magnetic field [33]. Permanent magnets are positioned
around the substance to make a magnetic field gradient, which holds or eliminates the
Stimuli-Responsive Material in Controlled Release of Drug 541
converted electrical energy into thermal energy (called the electrothermal effect),
causing them to expand and bend when exposed to an electrical current. Applying a
low voltage (less than 0.5 V) and an external magnetic field enabled the actuator to
swing up to 9000 times per minute, within the range of insect wing flapping speed
(between 1000 and 15,000 swings per minute) [8].
Ge et al. have reported on the initial successful in vivo application of drug delivery
activated by an external electric field. They utilized conductive polymer nanoparticles
that were encapsulated in thermo-responsive block copolymers, which can change
from a liquid to a hydrogel state. The release of negatively charged fluorescein and
positively charged daunorubicin from the conductive nanoparticles was ascribed
to redox reactions induced by the electric field. This finding suggests the possi-
bility of non-invasively initiating drug delivery by manipulating the characteristics
of the electric field and selectively releasing co-loaded drug molecules [46]. Yan
et al. have devised a system of voltage-sensitive vesicles that can undergo reversible
self-assembly. This system includes a cyclodextrin-capped polystyrene chain and a
ferrocenyl group-containing polyethylene oxide homopolymer. The self-assembled
structure can be dissociated by applying an oxidizing voltage to expel the charged
ferrocenyl cations from the cyclodextrin cavity.
On the other hand, a reducing voltage can revert the ferrocenyl functionality to
its neutral state and allow for self-assembly. Yan et al. have introduced a system
of electrical-responsive vesicles. This system consists of a polystyrene chain with
a cyclodextrin motif at the end and a polyethylene oxide homopolymer containing
a ferrocenyl group. When an oxidizing voltage is applied, the charged ferrocenyl
cations are ejected from the cyclodextrin cavity, resulting in the dissociation of the
self-assembled structure. The release of Rhodamine B was dependent on the applied
voltage [47]. Chang and colleagues have developed a novel system of voltage-
responsive reversible self-assembly using double hydrophilic block copolymers.
The self-organized polyethylene glycol-poly(acrylic acid) (PEG113-b-PAA30) block
copolymers into micelles by adding a cationic ferrocenyl surfactant. Oxidation of the
ferrocenyl group into ferrocenium cations disrupts the micelles because of enhanced
water solubility. The system demonstrated electrically induced disassembly, and the
release of rhodamine 6G increased with raising oxidative voltage via electrostatic
attraction [48].
Another investigation by Kim et al. introduced liposomes that can be triggered by
an electric field by attaching random copolymers onto the surface of phosphatidyl-
choline liposomes. The imposition of an electric field ionizes the carboxyl groups
of the attached chain, which in turn causes shear stress on the liposomal membrane,
resulting in the release of the entrapped molecules. The liberation of calcein depended
on the carboxyl content in the liposomes and the imposed electrical potential.
Moreover, H+ generated by water electrolysis was also able to cause membrane
compression, providing an alternative pathway to membrane disruption [49]. Qu
et al. developed an injectable hydrogel by combining chitosan-g-polyaniline with
oxidized dextran. They discovered that applying a voltage to the hydrogel resulted
in the pulsatile release of both hydrophilic and lipophilic agents, which returned to
a passive release rate when the voltage was switched off.
544 K. Trehan et al.
Using light as a trigger for drug delivery provides precise control over when and where
drugs are released [53]. Light-responsive materials have advantages over traditional
DDSs, one being their potential to focus on specific sites using different wavelengths
of light [54]. The characteristics of light, like frequency, charge, strength, and length
can be modified to attain precise drug discharge [55]. Therapeutic drugs loaded onto
nanocarriers can selectively home in diseased tissues which use either passive or
active binding and internalization pathways. Once the nanocarriers accumulate in
diseased tissues and cells, radiation using light can be applied to the affected area
with minimal impact on healthy tissues, and the light exposure can be managed in
both spatial and temporal aspects.
Light-responsive drug delivery nanocarriers can be divided into three categories
which depend on the wavelength of the light they respond to, namely ultraviolet (UV)
light (200–400 nm), visible (Vis) light (400–700 nm), and near-infrared (NIR) light
(700–1000 nm) [56]. Among these, UV light is preferred because of its ability to acti-
vate a broad range of light-responsive materials and trigger the release of therapeutic
drugs through various reactions such as cleavage, isomerization, rearrangement, or
cross-linking. Nevertheless, the utilization of UV radiation has certain disadvan-
tages, such as inadequate tissue penetration and elevated phototoxicity, which can
induce harm to the tissues, making it inefficient in treating deep-seated ailments.
Hence, drug-delivery nanosystems activated by UV light have restricted usefulness
in clinical applications [57].
In contrast to UV light, NIR and certain visible light wavelengths in the range of
600–1000 nm are more suitable for light-responsive materials since they can penetrate
deeper into tissues. This is because they are less affected by natural pigments like
Stimuli-Responsive Material in Controlled Release of Drug 545
Table 2 (continued)
Reference Method Materials Key findings
Kim et al. Electro-responsive The surface of 1. Carboxyl
[50] liposomes phosphatidylcholine liposomes functionalities of the
was coated with random anchored chain
copolymers made up of ionize and induce
poly(hydroxyethyl mechanical force on
acrylate-co-hexadecyl the liposomal
acrylate-co-carboxyethyl membrane upon
acrylate) utilization of an
electric field
2. Release of
encapsulated
molecules (calcein),
this effect is due to
both the voltage
applied and the
amount of carboxyl
groups present in the
liposomes
Liu et al. Electrically triggered A glass substrate featuring an 1. Low oxidative
[53] device for drug electrode made of indium tin voltage (≈1 V)
release controlled oxide (ITO), as well as layers results in the
remotely of polyvinyl alcohol and conductive polymer
poly(3-hexylthiophene) switching from being
hydrophobic to
hydrophilic
2. Release of
encapsulated
molecules through
increased
penetrability
resulting from this
shift voltage of 10 V
for 5 min
blood, water, melanin, and other biomolecules [58]. A study by Juzenas et al. has
shown that the depth of skin penetration of incident light in rats varies based on the
wavelength, with NIR light having a much deep penetration depth than UV light.
Specifically, at λ = 705 nm, the tissue permeation depth is 7.5 ± 0.5 mm, at λ =
633 nm it is 6.3 ± 0.5 mm, and at λ = 408 nm it is only 1.0 ± 0.02 mm [59].
As a result, NIR light has become a promising candidate for photo-responsive drug
delivery as it can penetrate deeper into tissues while minimizing tissue damage.
Numerous light-responsive nanocarriers have been evaluated for cancer theranos-
tics and drug delivery. These comprise a wide range of materials, such as polyplexes,
polyion complex vesicles (PICsomes), nanoparticles, upconverting nanoparticles
(UCNPs), nano gels, polymeric micelles, liposomes, nanorods [60], nano rattles [61].
Light-responsive nanocarriers exhibit various activities in response to light, including
Stimuli-Responsive Material in Controlled Release of Drug 547
linkers that use O-nitrobenzyl as a base are broken down at wavelengths between
300 nm and 365 nm. The speed of the breakdown can differ, lasting from a couple
of min. to numerous hours, depending on the wavelength to which it is exposed.
Coumarin-4-ylmethyl-based linkers, on the other hand, are generally cleaved with
UV light at 254 nm. These linkers have been incorporated into diverse DDSs, such
as nanoparticles and microspheres [70]. New research has indicated that photo-
switching nanocarriers have promise for loading a range of bioactive substances
and releasing them upon exposure to UV–Vis light, including drugs like paclitaxel,
docetaxel, and doxorubicin, and for treating cancer. Nonetheless, the limited wave-
length range of UV–Vis light might constrain their application. As a result, nanocar-
riers that respond to NIR light have been created to regulate drug delivery and enable
deeper tissue penetration [76]. For example, polymeric micelles incorporated with
IR-780 can respond to NIR for doxorubicin release [77].
Nanocarriers that are responsive to light can assist in the efficient delivery of
bioactive agents like genes, photosensitizers, & anticancer drugs into cells [60].
These carriers can be engineered to respond to light and release their cargo within
cells. For instance, PICsomes incorporating Al (III) phthalocyanine chloride disul-
fonic acid (AlPcS2a) can be activated by laser irradiation, leading to the escape of
the drug from the endosomes and resulting in higher photocytotoxicity compared to
AlPcS2a alone. A type of polyplex micelle with three layers that responds to light
has been created using polycationic polymers. These micelles can condense pDNA
and carry dendrimer phthalocyanine, which can enable successful gene transfection
throughout the body via photochemical internalization (PCI), which is activated by
light and allows for escape from the endosomal/lysosomal pathway [78]. Further-
more, light-responsive nanocarriers have the potential to be simulated for tumor
therapy and theranostics guided by imaging. By combining light-triggered genera-
tion of ROS and photothermal effects, these nanocarriers can lead to tumor ablation
or be used in conjunction with other bioactive agents for multimodal cancer thera-
nostics. Additionally, light-responsive nanocarriers have illustrated high efficacy in
treating cancers that are resistant to multiple drugs (MDR) [60].
apoptosis-inducing ligand (TRAIL) and DOX to the site of the tumor [83]. In
the tumor microenvironment, the HAase enzyme rapidly degraded the HA shell
surrounding the liposomes, leading to the quick release of TRAIL outside of the cells.
Afterwards, the liposomes were internalized, leading to the integration of TRAIL and
DOX. The co-delivery system of TRAIL and DOX showcased a remarkable IC50
value of 83 ng/mL against MDA-MB-231 cells. This demonstrated a potency that
was 5.9 times greater than that of DOX used alone in the therapy [85].
In the study Zhang et al., constructed a PEGylated system that responds to
Cathepsin B, an enzyme commonly found in the tumor microenvironment, releasing
gemcitabine in their study [84]. The quantity of gemcitabine discharged highly relied
on the existence or nonexistence of Cathepsin B [84]. Van Hove et al. created
a PEG hydrogel that included pro-angiogenic peptides that were responsive to
MMP2, an enzyme that can be utilized in cancer treatment or to reduce inflam-
mation [33]. The hydrogel successfully released the peptides at the desired sites
without any nonspecific degradation [33]. Lee et al. focused on the stabilization
of insulin at increased temperatures by preparing a trehalose-based hydrogel that
could trigger insulin release in response to glucose [85]. Their finding suggested that
the trehalose hydrogel, which responded to glucose, effectively combatted heating
stress, preserving over 50% of the cargo that was loaded at high temperatures [85].
The trehalose glycopolymers in the hydrogel served as active stabilizers for proteins,
such as insulin.
Enzyme-responsive systems offer various applications in a broad spectrum of
diseases and medical conditions. One of the key benefits of these systems is their
selectivity and natural capacity to react to internal triggers, leveraging the patholog-
ical or physiological microenvironment. This specificity can help minimize toxicity
in healthy tissues and cells. However, one major drawback is the potential for drug
release prior to arriving at the desired destination. To overcome this issue, enzyme-
triggered biomaterials often possess additional properties that respond to other trig-
gers, such as changes in pH, to protect the therapeutic cargo until it reaches the
intended location. However, it is crucial to consider that exposure to the enzyme
trigger or a similar enzyme can result in the early release of the drug.
Scientists have been highly interested in polymeric materials that respond to ultra-
sound for many years. These smart materials are more useful than other responsive
materials, like those that respond to UV, heat, or pH, because they can deliver drugs
more effectively and precisely target treatment without being too invasive. Mechan-
ical waves Having frequencies that are 20 kHz or higher are known as ultrasound
waves, which can be directed and transmitted through certain mediums. Ultrasound
waves have been employed in numerous clinical scenarios, providing a solution that
is both non-invasive and cost-efficient. These applications encompass a range of
fields, such as live imaging, physical therapy, beauty and skincare, and the culinary
sector [96]. Materials that are made up of polymers and are capable of reacting to
ultrasound comprise various forms [97]. These particular drug delivery mechanisms,
which react to ultrasound, have the capability to carry various types of medications,
like small molecule drugs, DNA, and proteins [98]. These vehicles that respond to
ultrasound have a wide range of potential uses, including, tumor treatment, disrupting
the blood–brain barrier, fighting infectious diseases, delivering drugs through the
skin, and thrombolysis [96]. It is thought that the mechanism is linked to the creation
of both thermal and non-thermal impacts through the energy released by ultrasound
radiation. The process of transforming sound energy into heat energy generates heat
within the tissue that comes into contact with the ultrasound waves, which is referred
to as the thermal effect. This can disrupt the cell membrane and lead to higher
permeability of blood vessels [99].
The use of hyperthermia as a treatment method has resulted in noteworthy
advancements in the therapy of tumors. The non-thermal effect is another mechanism
that could be responsible for ultrasound-mediated drug delivery, primarily linked to
cavitation. This phenomenon can arise in pre-existing microbubbles or cavitation
nuclei [96]. The creation of effective nanosystems/nanoplatforms that can respond to
ultrasound signals and have specific composition, structure, and features is essential
for developing theranostic applications that are activated by ultrasound. This typically
involves utilizing the underlying material and synthetic chemistry to design these
nanosystems. At present, nanoparticles being investigated for their responsiveness
to ultrasound fall into three categories: organic, inorganic, or a combination of both
known as organic–inorganic hybrid nanosystems. As a result, the methods used to
create these ultrasound-responsive nanosystems differ greatly from one another. This
indicates that the fundamental material chemistry also differs among these adaptable
nanosystems. To provide an example, when creating nanoparticles that can change
phases in response to ultrasound for therapeutic and diagnostic purposes, perfluo-
rocarbon (PFC) is commonly used. In contrast, porphyrin-based nanomedicine is
commonly employed in the treatment of tumors that exhibit a favorable response to
ultrasound [100].
554 K. Trehan et al.
Although Substantial headway has been achieved in the creation of DDSs that can
respond to stimuli, there are still several obstacles that must be overcome in order
to make them more practical for clinical use. A key challenge is the capability to
achieve precise control over the release of drugs. The release kinetics of the drug
should be precisely controlled to ensure that the therapeutic dose is delivered at the
right time and site. Several factors, such as the dimensions and configuration of the
nanoparticles, the choice of polymer, and the type of stimulus, can influence drug
release kinetics. Thus, further studies are needed to optimize these parameters and
develop efficient and controlled DDSs.
A further difficulty in DDSs is ensuring that the nanoparticles used are both
biocompatible and biodegradable. If the nanoparticles are toxic, they can lead to
undesired side effects that can limit their practical application in medicine. Therefore,
it is crucial to use materials that are safe for use in humans and that can break down
naturally. Numerous studies have investigated the use of natural polymers such as
chitosan, alginate, and cellulose for this purpose [101]. Additionally, ensuring the
reliability of SRDDS (Super Reliable Data Storage) during storage and transportation
poses an additional obstacle. Factors such as temperature, pH, and shear stress can
affect the stability of nanoparticles. Hence, it is essential to develop DDSs that can
keep their integrity and effectiveness during storage and transportation.
Although several in vitro and in vivo studies have proven the potential of these
systems, their clinical efficacy and safety remain to be established. Therefore, it is
necessary to conduct extensive preclinical and clinical trials to validate the efficacy,
safety, and feasibility of these systems for clinical use [102, 103]. Despite the chal-
lenges, SRDDS holds tremendous possibilities for the treatment of several diseases.
In the future, several developments are expected in this field, including developing
new stimuli-responsive materials, optimizing drug release kinetics, and validating
clinical efficacy and safety. Furthermore, the integration of advanced technologies,
such as artificial intelligence and machine learning, can significantly improve the
development and optimization of SRDDS.
4 Conclusion
Polymer blends have emerged as a popular approach due to their easy fabrication
and manipulation of device properties. Interpolymer complexes, interpenetrating
polymer networks, and block copolymers have also been recognized as effective
stimuli-responsive materials. The use of various synthetic routes and architectures has
further enhanced the performance and functionality of these materials. Temperature-
sensitive, pH-sensitive, and electric and magnetic field-responsive materials have
been extensively studied for biomedical applications. These materials respond to
various stimuli and exhibit a desired response, making them useful for controlled
drug release. Controlled DDSs offer unique advantages over conventional therapy,
including improved efficacy, reduced toxicity, and enhanced patient compliance.
Responsive materials that can react to specific stimuli have found extensive use in
diverse biomedical fields such as cancer therapy, gene therapy, tissue engineering,
and diagnosis. For instance, temperature-sensitive gels have been utilized to deliver
drugs precisely to tumors, while pH-sensitive systems have been employed for gene
delivery purposes. Additionally, materials responsive to electric and magnetic fields
have demonstrated potential in both diagnosing and treating various diseases.
Despite the considerable advancements achieved in the field of stimuli-responsive
materials, there remain several challenges that require attention. These challenges
encompass optimizing material properties, establishing scalable and cost-effective
synthesis methods, and identifying appropriate target diseases and patient popula-
tions. Future research endeavors in this area are anticipated to focus on advancing
state-of-the-art materials that exhibit intelligent responsiveness to multiple stimuli,
resulting in improved specificity and sensitivity. Furthermore, the integration of
advanced imaging techniques like magnetic resonance imaging and positron emis-
sion tomography with stimuli-responsive materials is expected to enable real-time
monitoring and visualization of drug release and treatment efficacy. To summa-
rize, stimuli-responsive materials hold great promise for the progress of advanced
DDSs and biomedical applications. By incorporating diverse scientific disciplines
and employing advanced synthetic methodologies and structures, scientists have
successfully developed materials capable of responding to various stimuli and elic-
iting desired reactions. Further investigation in this area is anticipated to produce
innovative materials that could significantly transform the diagnosis and treatment
of various diseases and disorders.
References
1. Rosenblum, D., Joshi, N., Tao, W., Karp, J.M., Peer, D.: Progress and challenges towards
targeted delivery of cancer therapeutics. Nat. Commun. 9(1), 1410 (2018)
2. Srinivasarao, M., Low, P.S.: Ligand-targeted drug delivery. Chem. Rev. 117(19), 12133–12164
(2017)
3. Xiao, M., Sun, W., Fan, J., Cao, J., Li, Y., Shao, K., Li, M., Li, X., Kang, Y., Zhang, W.,
Long, S.: Aminopeptidase-N-activated theranostic prodrug for NIR tracking of local tumor
chemotherapy. Adv. Func. Mater. 28(47), 1805128 (2018)
556 K. Trehan et al.
4. Liu, J., Zhang, R., Xu, Z.P.: Nanoparticle-based nanomedicines to promote cancer
immunotherapy: Recent advances and future directions. Small 15(32), 1900262 (2019)
5. Qiu, M., Singh, A., Wang, D., Qu, J., Swihart, M., Zhang, H., Prasad, P.N.: Biocompatible
and biodegradable inorganic nanostructures for nanomedicine: Silicon and black phosphorus.
Nano Today 1(25), 135–155 (2019)
6. Chamundeeswari, M., Jeslin, J., Verma, M.L.: Nanocarriers for drug delivery applications.
Environ. Chem. Lett. 17(2), 849–865 (2019)
7. Ahmed, A., Sarwar, S., Hu, Y., Munir, M.U., Nisar, M.F., Ikram, F., Asif, A., Rahman, S.U.,
Chaudhry, A.A., Rehman, I.U.: Surface-modified polymeric nanoparticles for drug delivery
to cancer cells. Expert Opin. Drug Deliv. 18(1), 1–24 (2021)
8. Hu, L., Zhang, Q., Li, X., Serpe, M.J.: Stimuli-responsive polymers for sensing and actuation.
Mater. Horiz. 6(9), 1774–1793 (2019)
9. Das, S.S., Bharadwaj, P., Bilal, M., Barani, M., Rahdar, A., Taboada, P., Bungau, S., Kyzas,
G.Z.: Stimuli-responsive polymeric nanocarriers for drug delivery, imaging, and theragnosis.
Polymers 12(6), 1397 (2020)
10. Ding, C., Tong, L., Feng, J., Fu, J.: Recent advances in stimuli-responsive release function
DDSs for tumor treatment. Molecules 21(12), 1715 (2016)
11. Li, L., Yang, W.W., Xu, D.G.: Stimuli-responsive nanoscale DDSs for cancer therapy. J. Drug
Target. 27(4), 423–433 (2019)
12. Raza, A., Rasheed, T., Nabeel, F., Hayat, U., Bilal, M., Iqbal, H.M.: Endogenous and
exogenous SRDDS for programmed site-specific release. Molecules 24(6), 1117 (2019)
13. Cook, A.B., Decuzzi, P.: Harnessing endogenous stimuli for responsive materials in
theranostics. ACS Nano 15(2), 2068–2098 (2021)
14. Li, S., Hu, K., Cao, W., Sun, Y., Sheng, W., Li, F., Wu, Y., Liang, X.J.: PH-responsive
biocompatible fluorescent polymer nanoparticles based on phenylboronic acid for intracellular
imaging and drug delivery. Nanoscale 6(22), 13701–13709 (2014)
15. Municoy, S., Álvarez Echazú, M.I., Antezana, P.E., Galdopórpora, J.M., Olivetti, C., Mebert,
A.M., Foglia, M.L., Tuttolomondo, M.V., Alvarez, G.S., Hardy, J.G., Desimone, M.F.: Stimuli-
responsive materials for tissue engineering and drug delivery. Int. J. Mol. Sci. 21(13), 4724
(2020)
16. Hu, Y.B., Dammer, E.B., Ren, R.J., Wang, G.: The endosomal-lysosomal system: From
acidification and cargo sorting to neurodegeneration. Transl. Neurodegener. 4, 1 (2015)
17. Wang, L., Neumann, M., Fu, T., Li, W., Cheng, X., Su, B.L.: Porous and responsive hydrogels
for cell therapy. Curr. Opin. Colloid Interface Sci. 1(38), 135–157 (2018)
18. Aycan, D., Alemdar, N.: Development of pH-responsive chitosan-based hydrogel modified
with bone ash for controlled release of amoxicillin. Carbohyd. Polym. 15(184), 401–407
(2018)
19. Ouyang, L., Sun, Z., Wang, D., Qiao, Y., Zhu, H., Ma, X., Liu, X.: Smart release of doxorubicin
loaded on polyetheretherketone (PEEK) surface with 3D porous structure. Colloids Surf., B
1(163), 175–183 (2018)
20. Bahadır, E.B., Sezgintürk, M.K.: Poly (amidoamine)(PAMAM): An emerging material for
electrochemical bio (sensing) applications. Talanta 1(148), 427–438 (2016)
21. Li, W., Feng, R., Wang, R., Li, D., Jiang, W., Liu, H., Guo, Z., Serpe, M.J., Hu, L.:
Polyelectrolyte-based physical adhesive hydrogels with excellent mechanical properties for
biomedical applications. J. Mater. Chem. B 6(29), 4799–4807 (2018)
22. Zhao, W., Quan, M., Cao, Z., Zhang, Y., Wen, J., Pan, D., Dong, Z., Yang, Z., Wang, D., Cao,
H., He, W.: Visual multi-triggered sensor based on inverse opal hydrogel. Colloids Surf., A
5(554), 93–99 (2018)
23. Xie, X., Doloff, J.C., Yesilyurt, V., Sadraei, A., McGarrigle, J.J., Omami, M., Veiseh, O.,
Farah, S., Isa, D., Ghani, S., Joshi, I.: Reduction of measurement noise in a continuous
glucose monitor by coating the sensor with a zwitterionic polymer. Nat. Biomed. Eng. 2(12),
894–906 (2018)
24. Aguilar, M.R., San Román, J., (eds.): Smart Polymers and Their Applications. Woodhead
Publishing (2019)
Stimuli-Responsive Material in Controlled Release of Drug 557
25. Bazban-Shotorbani, S., Hasani-Sadrabadi, M.M., Karkhaneh, A., Serpooshan, V., Jacob, K.I.,
Moshaverinia, A., Mahmoudi, M.: Revisiting structure-property relationship of pH-responsive
polymers for drug delivery applications. J. Control. Release 10(253), 46–63 (2017)
26. Le, X.X., Zhang, Y.C., Lu, W., Wang, L., Zheng, J., Ali, I., Zhang, J.W., Huang, Y.J., Serpe,
M.J., Yang, X.T., Fan, X.D.: A novel anisotropic hydrogel with integrated self-deformation
and controllable shape memory effect. Macromol. Rapid Commun. 39(9), 1800019 (2018)
27. Shang, J., Theato, P.: Smart composite hydrogel with pH-, ionic strength-and temperature-
induced actuation. Soft Matter. 14(41), 8401–8407 (2018)
28. Huang, L., Jiang, R., Wu, J., Song, J., Bai, H., Li, B., Zhao, Q., Xie, T.: Ultrafast digital
printing toward 4D shape changing materials. Adv. Mater. 29(7), 1605390 (2017)
29. Cheng, Y., Huang, C., Yang, D., Ren, K., Wei, J.: Bilayer hydrogel mixed composites that
respond to multiple stimuli for environmental sensing and underwater actuation. J. Mater.
Chem. B 6(48), 8170–8179 (2018)
30. Burillo, G., Bucio, E., Arenas, E., Lopez, G.P.: Macromol. Mater. Eng. 292, 214 (2007)
31. Lee, B.P., Konst, S.: Novel hydrogel actuator inspired by reversible mussel adhesive protein
chemistry. Adv. Mater. 26(21), 3415–3419 (2014)
32. Wells, C.M., Harris, M., Choi, L., Murali, V.P., Guerra, F.D., Jennings, J.A.: Stimuli-responsive
drug release from smart polymers. J. Funct. Biomater. 10(3), 34 (2019)
33. Wu, B.Y., Xu, Y.W., Le, X.X., Jian, Y.K., Lu, W., Zhang, J.W., Chen, T.: Smart hydrogel
actuators assembled via dynamic boronic ester bonds. Acta Polym. Sin. 1(50), 496–504 (2019)
34. Zhou, Q., Zhang, L., Yang, T., Wu, H.: Stimuli-responsive polymeric micelles for drug delivery
and cancer therapy. Int. J. Nanomed. 13, 2921 (2018)
35. Wei, H., Zhang, X., Cheng, C., Cheng, S.X., Zhuo, R.X.: Self-assembled, thermosensitive
micelles of a star block copolymer based on PMMA and PNIPAAm for controlled drug
delivery. Biomaterials 28(1), 99–107 (2007)
36. Li, H., Yan, K., Shang, Y., Shrestha, L., Liao, R., Liu, F., Li, P., Xu, H., Xu, Z., Chu, P.K.: Folate-
bovine serum albumin functionalized polymeric micelles loaded with superparamagnetic iron
oxide nanoparticles for tumor targeting and magnetic resonance imaging. Acta Biomater.
15(15), 117–126 (2015)
37. Hu, L., Wan, Y., Zhang, Q., Serpe, M.J.: Harnessing the power of stimuli-responsive polymers
for actuation. Adv. Func. Mater. 30(2), 1903471 (2020)
38. Qu, J., Zhao, X., Ma, P.X., Guo, B.: Injectable antibacterial conductive hydrogels with dual
response to an electric field and pH for localized “smart” drug release. Acta Biomater. 1(72),
55–69 (2018)
39. Kolosnjaj-Tabi, J., Gibot, L., Fourquaux, I., Golzio, M., Rols, M.P.: Electric field-responsive
nanoparticles and electric fields: Physical, chemical, biological mechanisms and therapeutic
prospects. Adv. Drug Deliv. Rev. 1(138), 56–67 (2019)
40. Deng, Z., Guo, Y., Zhao, X., Ma, P.X., Guo, B.: Multifunctional stimuli-responsive hydro-
gels with self-healing, high conductivity, and rapid recovery through host–guest interactions.
Chem. Mater. 30(5), 1729–1742 (2018)
41. Zhang, A., Jung, K., Li, A., Liu, J., Boyer, C.: Recent advances in stimuli-responsive polymer
systems for remotely controlled drug release. Prog. Polym. Sci. 1(99), 101164 (2019)
42. Gao, S., Tang, G., Hua, D., Xiong, R., Han, J., Jiang, S., Zhang, Q., Huang, C.: Stimuli-
responsive bio-based polymeric systems and their applications. J. Mater. Chem. B 7(5), 709–
729 (2019)
43. George, P.M., LaVan, D.A., Burdick, J.A., Chen, C.Y., Liang, E., Langer, R.: Electrically
controlled drug delivery from biotin-doped conductive polypyrrole. Adv. Mater. 18(5), 577–
581 (2006)
44. Baker, C.O., Huang, X., Nelson, W., Kaner, R.B.: Polyaniline nanofibers: Broadening
applications for conducting polymers. Chem. Soc. Rev. 46(5), 1510–1525 (2017)
45. Arteshi, Y., Aghanejad, A., Davaran, S., Omidi, Y.: Semi self-doped electroconductive and
biocompatible polyaniline/sulfonated β-cyclodextrin (PANI/SCD) inclusion complex with
potential use in regenerative medicine. Int. J. Polym. Mater. Polym. Biomater. (2019)
558 K. Trehan et al.
46. Ge, J., Neofytou, E., Cahill, T.J., III., Beygui, R.E., Zare, R.N.: Drug release from electric-
field-responsive nanoparticles. ACS Nano 6(1), 227–233 (2012)
47. Yan, Q., Yuan, J., Cai, Z., Xin, Y., Kang, Y., Yin, Y.: Voltage-responsive vesicles based on
orthogonal assembly of two homopolymers. J. Am. Chem. Soc. 132(27), 9268–9270 (2010)
48. Chang, X., Cheng, Z., Ren, B., Dong, R., Peng, J., Fu, S., Tong, Z.: Voltage-responsive
reversible self-assembly and controlled drug release of ferrocene-containing polymeric
superamphiphiles. Soft Matter. 11(38), 7494–7501 (2015)
49. Kim, J.A., Kim, J.C.: Temperature and electric field-triggerable liposomes incorporating poly
(hydroxyethyl acrylate-co-hexadecyl acrylate-co-carboxyethyl acrylate). J. Ind. Eng. Chem.
25(62), 383–391 (2018)
50. Liu, H.W., Hu, S.H., Chen, Y.W., Chen, S.Y.: Characterization and drug release behavior
of highly responsive chip-like electrically modulated reduced graphene oxide–poly (vinyl
alcohol) membranes. J. Mater. Chem. 22(33), 17311–17320 (2012)
51. Servant, A., Methven, L., Williams, R.P., Kostarelos, K.: Electroresponsive polymer-carbon
nanotube hydrogel hybrids for pulsatile drug delivery in vivo. Adv. Healthc. Mater. 2(6) (2013)
52. Liu, S., Chen, X., Wang, Y., Li, L., Wang, G., Li, X., Chen, H., Guo, J., Lin, H., Lian, Q.Q.,
Ge, R.S.: A role of KIT receptor signaling for proliferation and differentiation of rat stem
Leydig cells in vitro. Mol. Cell. Endocrinol. 15(444), 1–8 (2017)
53. Olejniczak, J., Carling, C.J., Almutairi, A.: Photocontrolled release using one-photon
absorption of visible or NIR light. J. Control. Release 10(219), 18–30 (2015)
54. Rapp, T.L., DeForest, C.A.: Targeting drug delivery with light: A highly focused approach.
Adv. Drug Deliv. Rev. 1(171), 94–107 (2021)
55. Cheng, L., Wang, C., Feng, L., Yang, K., Liu, Z.: Functional nanomaterials for phototherapies
of cancer. Chem. Rev. 114(21), 10869–10939 (2014)
56. Silva, J.M., Silva, E., Reis, R.L.: Light-triggered release of photocaged therapeutics-Where
are we now? J. Control. Release 28(298), 154–176 (2019)
57. Zhao, W., Zhao, Y., Wang, Q., Liu, T., Sun, J., Zhang, R.: Remote light-responsive nanocarriers
for controlled drug delivery: Advances and perspectives. Small 15(45), 1903060 (2019)
58. Juzenas, P., Juzeniene, A., Kaalhus, O., Iani, V., Moan, J.: Noninvasive fluorescence excitation
spectroscopy during application of 5-aminolevulinic acid in vivo. Photochem. Photobiol. Sci.
1(10), 745–748 (2002)
59. Mi, P.: Stimuli-responsive nanocarriers for drug delivery, tumor imaging, therapy and
theranostics. Theranostics. 10(10), 4557 (2020)
60. Li, C., Zhang, Y., Li, Z., Mei, E., Lin, J., Li, F., Chen, C., Qing, X., Hou, L., Xiong, L., Hao,
H.: Light-responsive biodegradable nanorattles for cancer theranostics. Adv. Mater. 30(8),
1706150 (2018)
61. Zhao, Y.: Light-responsive block copolymer micelles. Macromolecules 45(9), 3647–3657
(2012)
62. Yan, B., Boyer, J.C., Branda, N.R., Zhao, Y.: Near-infrared light-triggered dissociation of
block copolymer micelles using upconverting nanoparticles. J. Am. Chem. Soc. 133(49),
19714–19717 (2011)
63. Luo, D., Li, N., Carter, K.A., Lin, C., Geng, J., Shao, S., Huang, W.C., Qin, Y., Atilla-
Gokcumen, G.E., Lovell, J.F.: Rapid light-triggered drug release in liposomes containing
small amounts of unsaturated and porphyrin–phospholipids. Small 12(22), 3039–3047 (2016)
64. Wang, S., Lin, J., Wang, Z., Zhou, Z., Bai, R., Lu, N., Liu, Y., Fu, X., Jacobson, O., Fan,
W., Qu, J.: Core–satellite polydopamine–gadolinium-metallofullerene nanotheranostics for
multimodal imaging guided combination cancer therapy. Adv. Mater. 29(35), 1701013 (2017)
65. Fan, W., Lu, N., Xu, C., Liu, Y., Lin, J., Wang, S., Shen, Z., Yang, Z., Qu, J., Wang, T.,
Chen, S.: Enhanced afterglow performance of persistent luminescence implants for efficient
repeatable photodynamic therapy. ACS Nano 11(6), 5864–5872 (2017)
66. Wang, Z., Huang, P., Jacobson, O., Wang, Z., Liu, Y., Lin, L., Lin, J., Lu, N., Zhang, H.,
Tian, R., Niu, G.: Biomineralization-inspired synthesis of copper sulfide–ferritin nanocages
as cancer theranostics. ACS Nano 10(3), 3453–3460 (2016)
Stimuli-Responsive Material in Controlled Release of Drug 559
67. Zhu, F., Tan, S., Dhinakaran, M.K., Cheng, J., Li, H.: The light-driven macroscopic directional
motion of a water droplet on an azobenzene–calix [4] arene modified surface. Chem. Commun.
56(74), 10922–10925 (2020)
68. Goulet-Hanssens, A., Barrett, C.J.: Photo-control of biological systems with azobenzene
polymers. J. Polym. Sci., Part A: Polym. Chem. 51(14), 3058–3070 (2013)
69. Giménez, V.M., Arya, G., Zucchi, I.A., Galante, M.J., Manucha, W.: Photo-responsive poly-
meric nanocarriers for target-specific and controlled drug delivery. Soft Matter. 17(38),
8577–8584 (2021)
70. Zhang, D., Shah, P.K., Culver, H.R., David, S.N., Stansbury, J.W., Yin, X.,
Bowman, C.N.: Photo-responsive liposomes composed of spiropyran-containing triazole-
phosphatidylcholine: investigation of merocyanine-stacking effects on liposome–fiber
assembly-transition. Soft Matter. 15(18), 3740–3750 (2019)
71. Volarić, J., Szymanski, W., Simeth, N.A., Feringa, B.L.: Molecular photoswitches in aqueous
environments. Chem. Soc. Rev. 50(22), 12377–12449 (2021)
72. Movia, D., Prina-Mello, A., Volkov, Y., Giordani, S.: Determination of spiropyran cytotoxicity
by high content screening and analysis for safe application in bionanosensing. Chem. Res.
Toxicol. 23(9), 1459–1466 (2010)
73. Tao, Y., Chan, H.F., Shi, B., Li, M., Leong, K.W.: Light: A magical tool for controlled drug
delivery. Adv. Func. Mater. 30(49), 2005029 (2020)
74. Razavi, B., Abdollahi, A., Roghani-Mamaqani, H., Salami-Kalajahi, M.: Light-and
temperature-responsive micellar carriers prepared by spiropyran-initiated atom transfer poly-
merization: Investigation of photochromism kinetics, responsivities, and controlled release of
doxorubicin. Polymer 20(187), 122046 (2020)
75. Hansen, M.J., Velema, W.A., Lerch, M.M., Szymanski, W., Feringa, B.L.: Wavelength-
selective cleavage of photoprotecting groups: strategies and applications in dynamic systems.
Chem. Soc. Rev. 44(11), 3358–3377 (2015)
76. Wang, S., Lin, J., Wang, T., Chen, X., Huang, P.: Recent advances in photoacoustic imaging
for deep-tissue biomedical applications. Theranostics. 6(13), 2394 (2016)
77. Wang, J., Liu, Y., Ma, Y., Sun, C., Tao, W., Wang, Y., Yang, X., Wang, J.: NIR-activated
supersensitive drug release using nanoparticles with a flow core. Adv. Func. Mater. 26(41),
7516–7525 (2016)
78. Yen, H.C., Cabral, H., Mi, P., Toh, K., Matsumoto, Y., Liu, X., Koori, H., Kim, A.,
Miyazaki, K., Miura, Y., Nishiyama, N.: Light-induced cytosolic activation of reduction-
sensitive camptothecin-loaded polymeric micelles for spatiotemporally controlled in vivo
chemotherapy. ACS Nano 8(11), 11591–11602 (2014)
79. Pham, S.H., Choi, Y., Choi, J.: Stimuli-responsive nanomaterials for application in antitumor
therapy and drug delivery. Pharmaceutics 12(7), 630 (2020)
80. Mu, J., Lin, J., Huang, P., Chen, X.: Development of endogenous enzyme-responsive
nanomaterials for theranostics. Chem. Soc. Rev. 47(15), 5554–5573 (2018)
81. Yang, C., Wang, X., Yao, X., Zhang, Y., Wu, W., Jiang, X.: Hyaluronic acid nanogels with
enzyme-sensitive cross-linking group for drug delivery. J. Control. Release 10(205), 206–217
(2015)
82. Jiang, T., Mo, R., Bellotti, A., Zhou, J., Gu, Z.: Gel–liposome-mediated co-delivery of anti-
cancer membrane-associated proteins and small-molecule drugs for enhanced therapeutic
efficacy. Adv. Func. Mater. 24(16), 2295–2304 (2014)
83. Zhu, Q., Chen, X., Xu, X., Zhang, Y., Zhang, C., Mo, R.: Tumor-specific self-degradable
nanogels as potential carriers for systemic delivery of anticancer proteins. Adv. Func. Mater.
28(17), 1707371 (2018)
84. Van Hove, A.H., Beltejar, M.J., Benoit, D.S.: Development and in vitro assessment of
enzymatically-responsive poly (ethylene glycol) hydrogels for the delivery of therapeutic
peptides. Biomaterials 35(36), 9719–9730 (2014)
85. Lee, J., Ko, J.H., Mansfield, K.M., Nauka, P.C., Bat, E., Maynard, H.D.: Glucose-responsive
trehalose hydrogel for insulin stabilization and delivery. Macromol. Biosci. 18(5), 1700372
(2018)
560 K. Trehan et al.
86. Bordat, A., Boissenot, T., Nicolas, J., Tsapis, N.: Thermoresponsive polymer nanocarriers for
biomedical applications. Adv. Drug Deliv. Rev. 1(138), 167–192 (2019)
87. Qiao, S., Wang, H.: Temperature-responsive polymers: Synthesis, properties, and biomedical
applications. Nano Res. 11, 5400–5423 (2018)
88. Uchiyama, S., Gota, C., Tsuji, T., Inada, N.: Intracellular temperature measurements with
fluorescent polymeric thermometers. Chem. Commun. 53(80), 10976–10992 (2017)
89. Perera, M.M., Fischesser, D.M., Molkentin, J.D., Ayres, N.: Stiffness of thermoresponsive
gelatin-based dynamic hydrogels affects fibroblast activation. Polym. Chem. 10(46), 6360–
6367 (2019)
90. Chen, Y., Wang, Z., Harn, Y.W., Pan, S., Li, Z., Lin, S., Peng, J., Zhang, G., Lin, Z.: Resolving
optical and catalytic activities in thermoresponsive nanoparticles by permanent ligation with
temperature-sensitive polymers. Angew. Chem. Int. Ed. 58(34), 11910–11917 (2019)
91. Kotsuchibashi, Y.: Recent advances in multi-temperature-responsive polymeric materials.
Polym. J. 52(7), 681–689 (2020)
92. Palanisamy, A., Sukhishvili, S.A.: Swelling transitions in layer-by-layer assemblies of UCST
block copolymer micelles. Macromolecules 51(9), 3467–3476 (2018)
93. Jana, S., Anas, M., Maji, T., Banerjee, S., Mandal, T.K.: Tryptophan-based styryl
homopolymer and polyzwitterions with solvent-induced UCST, ion-induced LCST and
pH-induced UCST. Polym. Chem. 10(4), 526–538 (2019)
94. Zhang, H., Zhang, J., Dai, W., Zhao, Y.: Facile synthesis of thermo-, pH-, CO2 -and oxidation-
responsive poly (amido thioether) s with tunable LCST and UCST behaviors. Polym. Chem.
8(37), 5749–5760 (2017)
95. Khutoryanskiy, V.V., Georgiou, T.K., (eds.): Temperature-Responsive Polymers: Chemistry,
Properties, and Applications. John Wiley & Sons (2018)
96. Wei, P., Cornel, E.J., Du, J.: Ultrasound-responsive polymer-based DDSs. Drug Deliv. Transl.
Res. 11, 1323–1339 (2021)
97. Wei, P., Sun, M., Yang, B., Xiao, J., Du, J.: Ultrasound-responsive polymersomes capable of
endosomal escape for efficient cancer therapy. J. Control. Release 10(322), 81–94 (2020)
98. Kuo, J.H., Jan, M.S., Sung, K.C.: Evaluation of the stability of polymer-based plasmid DNA
delivery systems after ultrasound exposure. Int. J. Pharm. 257(1–2), 75–84 (2003)
99. Kamaly, N., Yameen, B., Wu, J., Farokhzad, O.C.: Degradable controlled-release polymers
and polymeric nanoparticles: Mechanisms of controlling drug release. Chem. Rev. 116(4),
2602–2663 (2016)
100. Cui, X., Han, X., Yu, L., Zhang, B., Chen, Y.: Intrinsic chemistry and design principle of
ultrasound-responsive nanomedicine. Nano Today 1(28), 100773 (2019)
101. Idrees, H., Zaidi, S.Z., Sabir, A., Khan, R.U., Zhang, X., Hassan, S.U.: A review of biodegrad-
able natural polymer-based nanoparticles for drug delivery applications. Nanomaterials
10(10), 1970 (2020)
102. Torchilin, V.P.: Multifunctional, stimuli-sensitive nanoparticulate systems for drug delivery.
Nat. Rev. Drug Discovery 13(11), 813–827 (2014)
103. Song, Y., Jing, H., Vong, L.B., Wang, J., Li, N.: Recent advances in targeted stimuli-responsive
nano-based DDSs combating atherosclerosis. Chin. Chem. Lett. 33(4), 1705–1717 (2022)
Stimuli-Responsive Material in Controlled Release of Drug 561
Abstract Global Cancer Therapy (GLOBOCAN) estimates that there are around
18 million new cancer cases each year. Various techniques have been used for cancer
therapy. Innovation of the nano approach will be signed in to locate one of the chief
concerns while using chemotherapy and radiation. Targeting the gold nanoparticle
(GNP)-based systems to the tumor can increase local radiation dosage while also
enhancing treatment through controlled releases of chemotherapeutics. In the last
20 years, more than 20 medicinal items based on nanotechnology have received
clinical use accreditation. This review article’s goal is to determine ways to employ
GNP-based therapy systems more quickly in clinical settings while still reducing
S. Parveen
Department of Chemistry, Dr. Mahalingam College of Engineering and Technology,
Pollachi 642003, India
T. Sathiyapriya
Department of Chemistry, Coimbatore Marine College, Coimbatore 641032, India
D. Tharani
Department of Science and Humanities, Nehru Institute of Engineering and Technology,
Coimbatore 641105, India
S. U. Mohammed Riyaz
PG & Research Department of Biotechnology, Islamiah College (Autonomous), Vaniyambadi,
Tamil Nadu 635752, India
R. A. Dinesh · J. Shanmugam
Department of Biotechnology, Sathyabama Institute of Science and Technology, Chennai 600119,
India
K. Rajakumar · D. Zherebtsov
Nanotechnology Research and Education Centre South, Ural State University,
Chelyabinsk 454080, Russia
M. Dhayalan (B)
Department of Prosthodontics, Saveetha Dental College & Hospitals, Saveetha Institute of
Medical and Technical Sciences (Saveetha University, TamilNadu, Chennai 600 077, India
e-mail: manikandandhayalan88@gmail.com
A. Stalin
Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology
of China, Chengdu 610064, China
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 563
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_19
564 S. Parveen et al.
normal tissue toxicity and improving treatment efficacy. Future cancer treatments will
be improved by nanomedicines and have fewer adverse effects. Gold nanoparticles
that have been altered and have regulated geometrical and optical characteristics have
undergone substantial research and are employed in biomedicine. The application of
gold nanoparticles in varied sizes, forms, and architectures in biomedicine will be
covered in this study’s summary of recent advancements and continuing research. The
major focus of the paper is how gold nanoparticle conjugates are used in biological
diagnostics and analytics.
1 Introduction
The development of chemistry via the use of novel nanoparticles (NPs) and their
notable applications in the nursing of certain diseases have captured the attention of
people all over the globe [1]. With various sluggish practices in medical investiga-
tion and medication, nanotechnology is dynamic for the transmission of medicinal
materials. Innumerable uses of NPs exist in the vast nanomedicine sector, including
cancer treatment, research techniques, and therapeutic solutions [2]. Their distinc-
tive physicochemical characteristics greatly facilitate the choice of NPs for cancer
treatment. Due to their higher size-to-volume ratio, the NP units utilized in nanotech-
nology with sizes that span between 1.0 nm and 100 nm completely incorporate
novel or inventive properties [3]. Metal NPs are the subject of investigation because
they provide a cutting-edge technique for treating cancer locally. Over recent years,
numerous distribution resources were proposed for the synthesizing of nanomaterials.
Numerous nanoparticles and nanomaterials have evolved from various metal
elements thanks to contemporary advancements in nanotechnology and medical
research. These materials are manufactured utilizing a variety of methods [4, 5].
The capability to influence nanoparticle structures unwraps up numerous opportu-
nities to reconnoiter these nanoparticles for drug delivery and diagnostic purposes
[6]. Due to their optical qualities, gold nanoparticles are used in imaging-based ther-
apeutics and ultrasensitive detection. In this study, we emphasize brand-new ideas
for investigating gold nanoparticles in cancer detection and therapy.
One of the key metals that have been disclosed is gold. Gold has been sought after
and studied for an extremely long time in several fields. In research studies employed
in numerous medical domains, effectuated Gold Nanoparticles (GNPs) with system-
atized optical and geometrical features are emphasized [7]. The recognition of GNPs
Gold Nanoparticles: Clinical Applications 565
Since the harmfulness and absorption of NPs are closely linked to their surface
composition, surface modification of nanoparticles is a crucial way to resolve
or minimize these difficulties. The impacts and biokinetics of nanoparticles after
systemic administration are dangerously influenced by the surface functionaliza-
tion of nanoparticles and their resulting physicochemical characteristics [10–12].
A top-down method is used to confine gold nanoparticles in cellular membranes
that resemble those of real red blood cells (RBCs). It is possible to observe in the
gold nanoparticles the robust membrane layer that covers the biological traits of the
donor cells [13]. GNP’s ability to quench fluorescence was lessened by the addi-
tion of coumarin to the PEG spacer’s surface [14]. Transferrin (TF), which can
be accurately detected and taken up by TF receptors dynamically distributed on
the surface of distinct tumor cells, is an ideal “ligand” for conjugating medicines
[15]. As a result, researchers have started using the transferrin-receptor interface
as a potentially effective method for the cellular absorption of medicines and
genes [16–19]. Gold nanoparticles (GHMP) stabilized by sodium hexametaphos-
phate (HMP) were described by the researcher in an aqueous media. To create
molecular probes with possible therapeutic applications, polyethylene glycol, and
transferrin were used to modify the surface of Au-HMP nanoparticles. In vitro, cell
survival assays employing naturally occurring G-HMP nanoparticles and their equiv-
alents with modified surfaces demonstrate the nanoparticles’ biocompatibility [20].
Gastric cancer is studied using a tailored fluorescent probe made of transferrin-linked
cysteine-capped GNP [21].
566 S. Parveen et al.
4 Cellular Detections
It may take several days to identify the cell pathogen, which eats up vital time for
those with severe bacterial infections. Innovative platform technology is needed to
enhance bacterial identification and detection in clinical practice. Aptamer-GNPs, a
non-polymerase chain reaction (PCR)-based method for recognizing Staphylococcus
aureus, one of the most important human pathogens that root more than 5 lakh
infections globally, helped to accomplish the target. This method’s quickness, great
sensitivity, and cost are its main advantages. Thus, within 1.5 h it finds a single
Staphylococcus aureus cell [32].
For more than a few decades, scientists have been enthralled by the potential
applications of nanoparticles and gold colloids in a variety of disciplines, including
biology, chemistry, engineering, and medicine. Using the Brust-Schiffrin method,
monodispersed gold nanoparticles with diameters ranging fluctuate 1 to 3 nm were
produced. Antibodies are now being developed in combination with gold nanorods
and nanoshells for photothermal treatment to target cancer cells. The important class
of materials known as golden nanoconjugates has already demonstrated promise
in applications related to fundamental cell biology. Using golden nanoconjugates
Gold Nanoparticles: Clinical Applications 567
The innovative materials known as gold nanoparticles have a broad range of uses in
both technology and medicinal fields. Several types of research are being in process
for coating DNA with gold particles and injecting them into plant embryos or plant
cells this will enable the association of some genetic material into the cells and ensure
some transformations in it. This will confirm that a portion of the genetic material
will penetrate the cells and transform them.
Colloidal gold nanoparticles have been used for a variety of unique purposes
throughout various fields [36]. JeongHoonByeon demonstrated the cytotoxicity and
transfection properties of thiol-capped gold nanoparticles that function as a potential
material for biomedical applications. Thiol-fused nanoparticles were cultured with
human embryonic kidneys for 24 h to assess the survival of the kidney cells. The
568 S. Parveen et al.
results demonstrated approximately 78% improved cell viability for fused particu-
late matter [37]. With a mortality rate of roughly 20%, lung cancer is one of the
noncommunicable illnesses that sees an increase in cases every year. To solve the
aforementioned issue lung cancer therapy uses an interfering RNA (siRNA) using
Mini Circle DNA Transfection (McD) via Gold Conjugation [38].
To deliver genes, peptide-modified gold nanoparticles (GNPs) have been created
using a simple and gentle technique. Four functional segments of a peptide with
the order (CRGDKGPDC) GPLGLAGIIIGRRRRRRRR-NH2 (CPIR28) have been
created. The GNPs with diameters of 10 nm are efficiently created by a moderate one-
pot reaction involving CPIR28, HAuCl4 , and NaBH4 in an aqueous solution, and they
contain CPIR28 molecules embedded on their surface. The R8 section is responsible
for the GNPs’ significant surface positive charge density. Electrostatic interactions
with negatively charged DNA may result in condensed complexes, which have little
cytotoxicity. In this system, CPIR28 serves as a superb stabilizer by grafting on
the surface of the GNPs to prevent their aggregation and precipitation in addition to
acting as a modifier to control the nucleation and growth of GNPs to control the shape
and size. Additionally, the surface-fused peptide molecules have positive charges that
allow them to connect with cells and aid in the effective delivery of genes [39].
Silicon nanowire arrays (SN), which reacted to reactive oxygen species (ROS)
safely, were used to provide a platform for secure gene and drug transfection. The
ROS-responsive SN is a novel, inventive, and secure platform for medicine and gene
transfection because of the platform’s moral biocompatibility.
Inorganic nanoparticles are being studied more and more as therapeutic and diag-
nostic tools in the oncology field in recent years. With some potential for clin-
ical trials, inorganic nanoparticles have shown efficiency in tumor photography
and treatment both internally and externally. Applications for gold nanoparticles
in the shapes of spheres, shells, rods, cages, etc., as well as photothermal treat-
ment, have all been studied. Other applications include carbon nanotubes, iron oxide
magnetic nanoparticles, ceramic nanoparticles, semiconductor fluorescent quantum
dots, and nanoscale ceramics. Because of their exceptional x-ray attenuation capabil-
ities, unique optical features, and surface modification properties, gold nanoparticle
development and applications have a lot of potential. According to reports, gold
nanoparticles show promise as CT contrast agents and can be used for medication
administration, photothermal and photodynamic treatments, and radiation. Due to
its high atomic number, gold has a significant X-ray absorption. Gold nanoparticles
can also benefit greatly from the easily changed surface when acting as target CT
contrast agents. Gold nanoparticles’ CT imaging capabilities are said to depend on
their size. The attenuation of X-rays in CT imaging increases when gold nanoparticle
size decreases. Gold nanoparticles are availed as disparity agents in computed tomog-
raphy imaging and ASX-ray-based radiotherapy (RT) agents because of their X-ray
Gold Nanoparticles: Clinical Applications 569
attenuation qualities. Gold nanoparticles also have the potential to be very effective
in photothermal therapy (PTT) and photodynamic therapy (PDT), which both aim
to kill cancerous tumor cells by converting light into heat or inducing the production
of free radicals. This is a result of their plasmonic and optical characteristics. When
employing PTT and PDT, the ability of gold nanoparticles to target tumor tissue
precisely is advantageous. This tumor selectivity prevents healthy tissue from being
amputated. This tumor-specificity may be affected by the increased permeability
and retention (EPR) effect of GNPs and surface modification with active targeting
ligands. Furthermore, gold nanoparticles are promising in the realm of drug adminis-
tration due to their easily adjustable surface. Multifunctional drug delivery platforms
can be created by embedding gold nanoparticles into big structures like liposomes
and polymeric particles that can transport medicines or imaging agents. Theranostic
delivery, which combines the payload of medications with diagnostic agents for
delivery at the same time, is currently receiving a lot of attention in the biomedical
sectors. Longer incubation times and larger concentrations of gold nanoparticles can
effectively accumulate inside pancreatic cancer cells. The outcomes of this study
will assist in the creation and improvement of therapeutic and diagnostic agents
based on gold nanoparticles for X-ray Drug Delivery System applications. Building
a therapeutic system out of functionalized gold nanoparticles that may particularly
respond to the tumor environment is a novel approach. The healing method included
employing a specific peptide substrate for the protease to bind doxorubicin to Gold
nanoparticles through a thiol-Au bond. The rapid release of the anti-tumor medication
(doxorubicin) from the functionalized gold nanoparticles, which may simultaneously
limit tumor growth and enable fluorescence imaging, has been shown to occur when
MMP-2 protease is overexpressed in tumor tissue and intracellular GSH.
7 Applications in Cells
is related to several major disorders. After entering the body and coming into contact
with blood-borne human cells, GNPs specifically target organs and the immune
system. Due to their stability and ease of decorating, GNPs are frequently employed
in the treatment of diseases, particularly in the administration of medications and
photothermal therapy with external stimulation. According to published research,
GNPs were also found to cause a variety of cell deaths, mostly through intrinsic routes
such as ROS generation, altered mitochondrial activity, and ER stress. The Surface
chemistry, dispersion state, and also size, and shape of the Gold nanoparticles play
important roles in determining cell death. Smaller GNPs have a higher propensity
to induce necrosis. In comparison to hexagonal and spherical gold nanoparticles,
gold nanorods are more likely to induce apoptosis. Apoptosis causes minimal to
no inflammation in the tissues around the cell, hence changing a cancer cell’s fate
to undergo apoptotic death through GNP intervention may be beneficial. Further-
more, ligands decorated on GNPs can be targeted to boost the efficiency of cell
death induced by targeted tissues and cells. Several various types of apoptosis might
happen simultaneously and interact with each other through the various cell death
pathways. Yet, the majority of research has solely addressed one particular form of
cell death. Therefore, further study should focus on the ability of GNP features to
predict various forms of cell death.
One of the main cancer treatment choices is chemotherapy [22]. Even though a range
of chemotherapy drugs are often used in medical settings, there are still substantial
challenges such as side effects and drug resistance [35]. There have been several
attempts to make cytotoxic medications stronger, including the use of very potent
substances like auristatin and maytansine as well as the mixing of many chemother-
apeutic agents [7–19]. However, systemic adverse effects and a narrow therapeutic
window limit their practical applicability. With the introduction of monoclonal anti-
bodies, it is now possible to administer medicine to specific areas of the body utilizing
their strong binding properties. Antibody—drug conjugates (ADCs), developed and
produced over many decades by combining antibodies with cytotoxic medications,
are based on this concept [10]. ADCs connect to tumor cell receptors with prefer-
ence [33]. The receptor-ADC complex is then typically absorbed by the endocytosis
pathway after that. Cytotoxic chemicals are released once the linker is split. As a
consequence, these drugs have the potential to be cytotoxic in several ways, including
by binding to DNA’s minor groove or reacting with tubulin.
Gold Nanoparticles: Clinical Applications 571
The number of ADCs being tested in clinical trials for the treatment of solid tumors
and hematologic malignancies has sharply increased during the last five years [2, 3].
A substantial category of anti-cancer drugs called ADCs has just emerged.
8.2 Gemtuzumabozogamicin
8.3 Brentuximabvedotin
8.4 Ado-Trastuzumabemtansine
NCT00829166)93,94 that T-DM1 was approved for were. The median progression-
free survival (PFS) time in the T-DM1 arm was 9.6 months, compared to just
6.4 months in the active comparator (P0.001) [36]. The overall response rate (ORR)
was 43.6% against 30.8% for the T-DM1, and overall survival (OS) was 30.9 months
versus 25.1 months for the comparator (P 0.001). (P 0.001). In terms of the percentage
of serious adverse events (15.5% vs 18.0%) 94 and the total incidence of adverse
events (40.8%), the T-DM1 arm outperformed the comparator arm (57.0%).
8.5 Inotuzumabozogamicin
8.6 Polatuzumabvedotin-Piiq
One of most latest ADC to enter the market (in June 2019) is polatuzumabvedotin-piiq
(Polivy®), which was produced via conjugating MMAE to an anti-CD79b antibody
utilizing a protease cleavable dipeptide linker [9]. The results of the GO29365 phase
Ib/II trial (NCT02257567) were used to decide the polatuzumabvedotin-expedited
piiq’s approval. In this trial, patients with large B-cell lymphoma do give the choice
of receiving polatuzumabvedotin with bendamustine and rituximab (BR).
ADCs with increased selectivity and potency are among the next directions, as are
ADCs that can deliver various payload classes [21].
8.9 Toxicity
It is generally known that the use of GNPs in the biomedical area has opened up new
possibilities for disease diagnosis and therapy. However, the extent to which they
are hazardous once given to patients, either by intravenous injection or oral dose,
determines how well they may be applied in the biomedical area, particularly in
therapeutic and diagnostic modalities. Cells can become poisonous to any molecular
compound at relatively large concentrations. Even therapeutic chemicals can cause
an imbalance in cellular activity when exposed to excessive concentrations, which
eventually results in harmful toxic consequences. What maximum concentration of
GNPs may be provided without causing detrimental toxicity at the cellular level
is therefore the correct issue to ask. How much GNP may be used for therapeutic
574 S. Parveen et al.
research. Numerous studies have been published that demonstrate how intracellular
ROS may be produced by GNPs, although controlled application can result in potent
antioxidants by limiting the formation of NF-B and the subsequent inflammatory
reactions. In the first phases of cancer treatment, when the therapeutic objective is
to accelerate cell death, the advantageous impact of GNPs’ toxic activity may be
helpful. Prior to using GNPs’ risky behavior in cancer research, issues with protein
corona formation and their efficient clearance from the body of treated patients must
be properly evaluated and resolved.
8.11 Conclusion
The unique qualities of GNPs make it possible to use them in medical applica-
tions. Several synthetic approaches for creating particles of different sizes and shapes
576 S. Parveen et al.
provide a broad toolbox for conjugates with enhanced affinity for specific cell recep-
tors, effective cell internalization, extended circulation half-life, tumor permeability,
and high biocompatibility. These characteristics of GNPs, along with their elec-
trical and optical responsiveness, have made it possible to create strong platforms
for developing new vaccines, effective gene therapy techniques, improved radiation
procedures, and novel cancer treatment approaches. Even if gold is less hazardous,
health complications might still occur due to the relatively slow rate of clearance from
tissues and circulation. For this reason, methods that improve the precise targeting
of sick cells must be standardized before GNPs are used regularly in humans.
References
1. Amiri-Kordestani, L., Blumenthal, G.M., Xu, Q.C., Zhang, L., Tang, S.W., Ha, L., Wein-
berg, W.C., Chi, B., Candau-Chacon, R., Hughes, P., Russell, A.M.: FDA Approval: Ado-
Trastuzumab Emtansine for the Treatment of Patients with HER2-Positive Metastatic Breast
CancerFDA Approval Summary for T-DM1 for HER2+ MBC. Clin. Cancer Res. 20(17),
4436–4441 (2014)
2. Beck, A., Wurch, T., Bailly, C., Corvaia, N.: Strategies and challenges for the next generation
of therapeutic antibodies. Nat. Rev. Immunol. 10(5), 345–352 (2010)
3. Birrer, M.J., Moore, K.N., Betella, I., Bates, R.C.: Antibody-drug conjugate-based therapeutics:
state of the science. JNCI: J. Natl. Cancer Inst. 111(6), 538–49 (2019)
4. Bross, P.F., Beitz, J., Chen, G., Chen, X.H., Duffy, E., Kieffer, L., Roy, S., Sridhara, R., Rahman,
A., Williams, G., Pazdur, R.: Approval summary: gemtuzumab ozogamicin in relapsed acute
myeloid leukemia. Clin. Cancer Res. 7(6), 1490–1496 (2001)
5. Canepa, E., Salassi, S., Simonelli, F., Ferrando, R., Rolandi, R., Lambruschini, C., Canepa, F.,
Dante, S., Relini, A., Rossi, G.: Non-disruptive uptake of anionic and cationic gold nanoparticles
in neutral zwitterionic membranes. Sci. Rep. 11(1), 1256 (2021)
6. Charbgoo, F., Nejabat, M., Abnous, K., Soltani, F., Taghdisi, S.M., Alibolandi, M., Shier, W.T.,
Steele, T.W., Ramezani, M.: Gold nanoparticle should understand protein corona for being a
clinical nanomaterial. J. Control. Release 28(272), 39–53 (2018)
7. Chari, R.V., Miller, M.L., Widdison, W.C.: Antibody–drug conjugates: an emerging concept
in cancer therapy. Angew. Chem. Int. Ed. 53(15), 3796–3827 (2014)
8. Chen, Y.S., Hung, Y.C., Liau, I., Huang, G.S.: Assessment of the in vivo toxicity of gold
nanoparticles. Nanoscale Res. Lett. 4, 858–864 (2009)
9. Deeks, E.D.: Polatuzumab vedotin: first global approval. Drugs 79(13), 1467–1475 (2019)
10. Diamantis, N., Banerji, U.: Antibody-drug conjugates—an emerging class of cancer treatment.
Br. J. Cancer 114(4), 362–367 (2016)
11. DiJoseph, J.F., Armellino, D.C., Boghaert, E.R., Khandke, K., Dougher, M.M., Sridharan, L.,
Kunz, A., Hamann, P.R., Gorovits, B., Udata, C., Moran, J.K.: Antibody-targeted chemotherapy
with CMC-544: a CD22-targeted immunoconjugate of calicheamicin for the treatment of B-
lymphoid malignancies. Blood 103(5), 1807–1814 (2004)
12. Doronina, S.O., Toki, B.E., Torgov, M.Y., Mendelsohn, B.A., Cerveny, C.G., Chace, D.F.,
DeBlanc, R.L., Gearing, R.P., Bovee, T.D., Siegall, C.B., Francisco, J.A.: Development of
potent monoclonal antibody auristatin conjugates for cancer therapy. Nat. Biotechnol. 21(7),
778–784 (2003)
13. Goodman, C.M., McCusker, C.D., Yilmaz, T., Rotello, V.M.: Toxicity of gold nanoparticles
functionalized with cationic and anionic side chains. Bioconjug. Chem. 15(4), 897–900 (2004)
14. Jen, E.Y., Ko, C.W., Lee, J.E., Del Valle, P.L., Aydanian, A., Jewell, C., Norsworthy, K.J.,
Przepiorka, D., Nie, L., Liu, J., Sheth, C.M.: FDA Approval: Gemtuzumab Ozogamicin for
Gold Nanoparticles: Clinical Applications 577
the Treatment of Adults with Newly Diagnosed CD33-Positive Acute Myeloid LeukemiaFDA
Approval Summary: Gemtuzumab Ozogamicin. Clin. Cancer Res. 24(14), 3242–3246 (2018)
15. Kalimuthu, K., Lubin, B.C., Bazylevich, A., Gellerman, G., Shpilberg, O., Luboshits, G., Firer,
M.A.: Gold nanoparticles stabilize peptide-drug-conjugates for sustained targeted drug delivery
to cancer cells. Journal of nanobiotechnology. 16(1), 1–3 (2018)
16. Kantarjian, H.M., DeAngelo, D.J., Stelljes, M., Martinelli, G., Liedtke, M., Stock, W.,
Gökbuget, N., O’Brien, S., Wang, K., Wang, T., Paccagnella, M.L.: Inotuzumab ozogamicin
versus standard therapy for acute lymphoblastic leukemia. N. Engl. J. Med. 375(8), 740–753
(2016)
17. Kovtun, Y.V., Audette, C.A., Mayo, M.F., Jones, G.E., Doherty, H., Maloney, E.K., Erickson,
H.K., Sun, X., Wilhelm, S., Ab, O., Lai, K.C.: Antibody-maytansinoid conjugates designed to
bypass multidrug resistance. Can. Res. 70(6), 2528–2537 (2010)
18. Lamb, Y.N.: Inotuzumab ozogamicin: first global approval. Drugs 77, 1603–1610 (2017)
19. Lambert, J.M., Berkenblit, A.: Antibody–drug conjugates for cancer treatment. Annu. Rev.
Med. 29(69), 191–207 (2018)
20. Lyon, R.P., Bovee, T.D., Doronina, S.O., Burke, P.J., Hunter, J.H., Neff-LaFord, H.D., Jonas, M.,
Anderson, M.E., Setter, J.R., Senter, P.D.: Reducing hydrophobicity of homogeneous antibody-
drug conjugates improves pharmacokinetics and therapeutic index. Nat. Biotechnol. 33(7),
733–735 (2015)
21. Maruani, A., Smith, M.E., Miranda, E., Chester, K.A., Chudasama, V., Caddick, S.: A plug-
and-play approach to antibody-based therapeutics via a chemoselective dual click strategy. Nat.
Commun. 6(1), 6645 (2015)
22. Miller, D.R.: A tribute to Sidney Farber–the father of modern chemotherapy. Br. J. Haematol.
134(1), 20–26 (2006)
23. Moskowitz, C.H., Nademanee, A., Masszi, T., Agura, E., Holowiecki, J., Abidi, M.H., Chen,
A.I., Stiff, P., Gianni, A.M., Carella, A., Osmanov, D.: Brentuximab vedotin as consolidation
therapy after autologous stem-cell transplantation in patients with Hodgkin’s lymphoma at risk
of relapse or progression (AETHERA): a randomised, double-blind, placebo-controlled, phase
3 trial. The Lancet. 385(9980), 1853–1862 (2015)
24. Pakravan, A., Salehi, R., Mahkam, M.: Comparison study on the effect of gold nanoparticles
shape in the forms of star, hallow, cage, rods, and Si-Au and Fe-Au core-shell on photothermal
cancer treatment. Photodiagn. Photodyn. Ther. 1(33), 102144 (2021)
25. Patterson, A.W., Peltier, H.M., Sasse, F., Ellman, J.A.: Design, synthesis, and biological prop-
erties of highly potent tubulysin D analogues. Chemistry–A Eur. Journal. 13(34), 9534–41
(2007)
26. Petersdorf, S.H., Kopecky, K.J., Slovak, M., Willman, C., Nevill, T., Brandwein, J., Larson,
R.A., Erba, H.P., Stiff, P.J., Stuart, R.K., Walter, R.B.: A phase 3 study of gemtuzumab ozogam-
icin during induction and postconsolidation therapy in younger patients with acute myeloid
leukemia. Blood, The Journal of the American Society of Hematology. 121(24), 4854–4860
(2013)
27. Quintieri, L., Geroni, C., Fantin, M., Battaglia, R., Rosato, A., Speed, W., Zanovello, P.,
Floreani, M.: Formation and antitumor activity of PNU-159682, a major metabolite of
nemorubicin in human liver microsomes. Clin. Cancer Res. 11(4), 1608–1617 (2005)
28. Saleh, H.M., Soliman, O.A., Elshazly, M.O., Raafat, A., Gohar, A.K., Salaheldin, T.A.: Acute
hematologic, hepatologic, and nephrologic changes after intraperitoneal injections of 18 nm
gold nanoparticles in hamsters. Int. J. Nanomed. 11, 2505 (2016)
29. Sasse, F., Sieinmetz, H., Heil, J., Hoefle, G., Reichenbach, H.: Tubulysins, new cytostatic
peptides from myxobacteria acting on microtubuli production, isolation, physico-chemical
and biological properties. J. Antibiot. 53(9), 879–85 (2000)
30. Senter, P.D.: Potent antibody drug conjugates for cancer therapy. Curr. Opin. Chem. Biol. 13(3),
235–244 (2009)
31. Senut, M.C., Zhang, Y., Liu, F., Sen, A., Ruden, D.M., Mao, G.: Size-dependent toxicity of
gold nanoparticles on human embryonic stem cells and their neural derivatives. Small 12(5),
631–646 (2016)
578 S. Parveen et al.
32. Sievers, E.L.: Efficacy and safety of gemtuzumab ozogamicin in patients with CD33-positive
acute myeloid leukaemia in first relapse. Expert Opin. Biol. Ther. 1(5), 893–901 (2001)
33. Sievers, E.L., Senter, P.D.: Antibody-drug conjugates in cancer therapy. Annu. Rev. Med. 17,
64 (2013)
34. Steckiewicz, K.P., Barcinska, E., Malankowska, A., Zauszkiewicz–Pawlak, A., Nowaczyk, G.,
Zaleska-Medynska, A., Inkielewicz-Stepniak, I.: Impact of gold nanoparticles shape on their
cytotoxicity against human osteoblast and osteosarcoma in in vitro model. Evaluation of the
safety of use and anti-cancer potential. J. Mater. Sci.: Mater. Medicine. 30,1–5 (2019)
35. Tsuchikama, K., An, Z.: Antibody-drug conjugates: recent advances in conjugation and linker
chemistries. Protein Cell 9(1), 33–46 (2018)
36. Verma, S., Miles, D., Gianni, L., Krop, I.E., Welslau, M., Baselga, J., Pegram, M., Oh, D.Y.,
Diéras, V., Guardino, E., Fang, L.: Trastuzumab emtansine for HER2-positive advanced breast
cancer. N. Engl. J. Med. 367(19), 1783–1791 (2012)
37. Vetter, J.: Toxins of Amanita phalloides. Toxicon 36(1), 13–24 (1998)
38. Widdison, W.C., Ponte, J.F., Coccia, J.A., Lanieri, L., Setiady, Y., Dong, L., Skaletskaya, A.,
Hong, E.E., Wu, R., Qiu, Q., Singh, R.: Development of anilino-maytansinoid ADCs that
efficiently release cytotoxic metabolites in cancer cells and induce high levels of bystander
killing. Bioconjug. Chem. 26(11), 2261–2278 (2015)
39. Younes, A., Gopal, A.K., Smith, S.E., Ansell, S.M., Rosenblatt, J.D., Savage, K.J., Ramchan-
dren, R., Bartlett, N.L., Cheson, B.D., De Vos, S., Forero-Torres, A.: Results of a pivotal phase
II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin’s lymphoma.
J. Clin. Oncol. 30(18), 2183 (2012)
Biocidal Effect of Copper Contained
in a Mineral Tailing on the Growth
of Shewanella Putrefaciens
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 579
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_20
580 H. J. Marín-García et al.
1 Introduction
strains isolated from shellfish only determined the maximum tolerable concentra-
tion (CMT) when cultivated in culture media added with copper sulfate (CuSO4),
obtaining bacteria that grew in 100, 3000, and >5000 mg of copper per liter; there-
fore, the purpose of this work is to analyze the biocidal effect of copper contained
in the tailings on the growth of Shewanella putrefaciens ATCC® 8071 for a specific
application in biomining.
2 Experimental Procedure
From the colony developed in the marine agar, an inoculum was taken with the
bacteriological loop and suspended in marine broth and incubated at 35 ± 0.3 °C
with 150 rpm of constant agitation for 72 h, aliquots were taken and inoculated into
tubes with marine broth enriched with magnetite PFD 00-007-0322 (FeO· Fe2 O3 ),
which were incubated under the same conditions mentioned above. The kinetics
were carried out at 35 ± 0.3 °C with 150 rpm of constant agitation for 240 h, then
the optical density of the biomass generated at 620 nm was read by the ultraviolet–
visible light spectroscopy technique, taking as standards the scale of McFarland [22,
23]. Regarding the working inoculum, the previous conditions are repeated, but the
incubation of the marine broths was for 48 h to ensure that the bacteria is in the
exponential growth phase.
Mixtures of marine broth with 5% solids (direct tailings and washed tailings) were
prepared separately, and sterilized, they were subjected to 35 ± 0.3 °C with 150 rpm
582 H. J. Marín-García et al.
of constant agitation for 500 h to observe if the culture broth reacts with the tailings,
later in the supernatant pH and copper in solution were determined by atomic absorp-
tion spectroscopy, while the solids were analyzed by scanning electron microscopy.
The aforementioned liquids were subjected to sterilization and previously prepared
inoculums were added to them, these samples were incubated aerobically at 35 ±
0.3 °C for 72 h, later, a 20 µL inoculum was taken and spread out with a glass rod. in
marine agar, they were incubated aerobically at 35 ± 0.3 °C for 48 to 72 h to observe
the development of CFU.
Culture media were prepared with marine agar enriched with copper sulfate hexahy-
drate with the following concentrations: 3.12, 6.35, 9.53, 12.71, 15.89, 19.06, 22.24,
25.42, 28.60, 31.77, 34.95, 38.13, 41.30, 44.48, 47.64., 76.26, 101.67, 127.10,
152.51, 177.93, 203.35 and 228.77 ppm of Cu to determine the Maximum Toler-
able Concentration (CMT) of copper to which the bacterium Shewanella putrefa-
ciens ATCC® 8071 can be subjected; in case of growth above 50.84 ppm of Cu, it
is considered a strain resistant to this metal, but if it grows below it is considered
sensitive [20, 24]. For the sowing of the culture media in an aerobic environment, a
20 µL inoculum was taken and sown in an extended way with a glass rod and after
72 h of incubation at 35 ± 0.3 °C they were observed in a colony counter.
The Shewanella putrefaciens ATCC® 8071 strain sown on marine agar showed abun-
dant growth under aerobic conditions (Fig. 1, section a), the cream-colored, slightly
viscous, and concave colony together with the Gram bacilli ( ), are characteristic of
this bacterium (Section b) [25, 26]. Regarding subsection c, the formation of a black
precipitate is observed in the TSI medium, indicating that the bacterium generates
hydrogen sulfide and does not ferment sugars, by maintaining the red color of the indi-
cator, which differentiates it from Enterobacteriaceae and the genus Pseudomonas
sp. [27].
Biocidal Effect of Copper Contained in a Mineral Tailing on the Growth … 583
Figure 3 shows the dissolution of copper caused by chemical leaching between the
tailings TH (Tailing Head), HTwas (Head Tailing washed), and the marine broth. The
measured pH is close to neutrality and the characteristic yellow color is observed;
after 500 h of agitation, the coloration of the marine broths turned green, being more
584 H. J. Marín-García et al.
Fig. 2 a Inoculum with sedimented magnetite and during shaking; b Growth kinetics of Shewanella
putrefaciens ATCC® 8071 in an anaerobic environment.
intense in the RSL than in the RL, mainly due to the excess of sulfuric acid that the
RSL still contained; at this point the pH decreased to 5.63 and dissolved 120.74 ppm
Cu, while the RL slightly decreased the pH to 6.05 and dissolved 67.36 ppm Cu. It is
also possible to observe that the greatest chemical leaching of copper occurs during
the first 250 h, since there is no significant difference with the longer leaching time,
and the pH remains without significant changes.
Regarding the analysis by scanning electron microscopy, Fig. 4 shows the micro-
graphs obtained with the electron backscattering technique, observing that in the TH
before the interaction with seawater (section a), the particles present a cloudy layer
on its surface associated with the remaining presence of sulfuric acid, since in the
HT was it disappears and the quality of the image of the particles with higher atomic
weight is improved (paragraph c) [7]. After 250 h of agitation with the marine broth,
both the RSL and the RL present agglomerated particles made up of low atomic
weight elements (from the marine broth), due to the homogeneity that the grays
present in the backscattered electron technique; it was possible to highlight some
particles or areas with brightness indicating elements of greater atomic weight (see
sections b and d); nevertheless, with this result it is shown that the high concentration
of salts, when dried on the tailings, generate agglomerates that can interfere with the
efficiency of the next gold extraction process if a washing stage is not carried out
that eliminates or considerably reduces said salts.
Figure 5, section a, shows the effect of the copper concentration on the growth
of Shewanella putrefaciens ATCC® 8071, after incubation of the TH and HT was
supernatants, plus the sterility control; the three presented null development of CFU,
indicating that the concentrations of 59.73 and 120.73 ppm of Cu in the supernatants
exceed the resistance mechanisms that the bacteria presents naturally, while the
Biocidal Effect of Copper Contained in a Mineral Tailing on the Growth … 585
Fig. 3 Copper concentration and pH in the marine broth, after 250 and 500 h of agitation of the
Tailing Head (TH), and the Head Tailing washed (HTwas)
Fig. 4 Micrographs applying the backscattered electron technique to the mineral samples after
500 h of leaching in marine broth. a and b TH; c and d HTwas
586 H. J. Marín-García et al.
Fig. 5 a Effect of dissolved copper from tailings on the growth of Shewanella putrefaciens ATCC®
8071 and sterility control; b Growth of the positive control in the marine agar.
sterility control indicated that aseptic conditions were maintained during the exper-
iment. Regarding the section b, the marine broth inoculated as a positive control is
shown, which presented abundant growth, showing that this medium is favorable for
the development of Shewanella putrefaciens ATCC® 8071.
Figure 6 section a shows the massive growth of the bacterium in the marine agar
without the presence of copper; however, as the concentration of this element
increases, the development of colonies decreases, Such is the case of the 19.06 ppm
Cu concentration (section b), where 782 ± 15 CFUs were counted, the Maximum
Tolerance Concentration (MTC) being 34.94 ppm Cu, in which 425 ± 9 CFUs were
counted (section c); from the concentration 38.13 to 228.77 ppm of Cu, no bacte-
rial growth was observed, as observed in section d. Therefore, when the Shewanella
putrefaciens ATCC® 8071 bacterium grows below 50.84 ppm Cu, it is considered to
be sensitive to this metal, confirming the inhibitory effect of copper solubilized by the
marine broth, where the lowest concentration obtained in the HTwas of 59.73 ppm
Cu is much higher than the value obtained in the MTC.
Biocidal Effect of Copper Contained in a Mineral Tailing on the Growth … 587
Fig. 6 Marine agar plates to determine the MTC to copper. a Control plate without addition of
cupric sulfate; b Plate with 19.06 ppm Cu and 782 CFU; c Plate with 34.95 ppm Cu and 425 ±
9 CFU; d Example of plates without growth for concentrations between 38.13 to 228.77 ppm of Cu
4 Conclusions
Acknowledgements The financial support for this work from the Consejo Nacional de Ciencia y
Tecnología (CONACyT) of Mexico. Hugo Jhonathan Marín-García deeply thanks to CONACyT
for the scholarship during his Ph.D. studies through the grant (444401).
References
1. Alcalde, J., Kelm, U., Vergara, D.: Historical assessment of metal recovery potential from old
mine tailings: A study case for porphyry copper tailings, Chile, Miner. Eng. 127, 334–338
(2018) https://doi.org/10.1016/j.mineng.2018.04.022
2. Sethurajan, M., van Hullebusch, E.D., Nancharaiah, Y.V.: Biotechnology in the management
and resource recovery from metal bearing solid wastes: Recent advances, J. Environ. Manage.
211, 138–153 (2018) https://doi.org/10.1016/j.jenvman.2018.01.035
3. Moreno, R., Monroy, M.G., Castañeda, E.P.: Evaluación geoquímica de reiduos mineros (jales
o colas) de mineralización tipo epitermal, Hidalgo, México. Rev. Geológica América Cent. 41,
79–98 (2009)
4. de Andrade Lima, L.R.P., Bernardez, L.A., Barbosa, L.A.D.: Characterization and treatment
of artisanal gold mine tailings. J. Hazard. Mater. 150, 747–753 (2008) https://doi.org/10.1016/
j.jhazmat.2007.05.028
5. Bingöl, D., Canbazoğlu, M.: Dissolution kinetics of malachite in sulphuric acid, Hydrometal-
lurgy. 72, 159–165 (2004) https://doi.org/10.1016/j.hydromet.2003.10.002
6. Nicol, M.J.: The kinetics of the dissolution of malachite in acid solutions. Hydrometallurgy.
177, 214–217 (2018) https://doi.org/10.1016/j.hydromet.2018.03.017
7. Marín-García, H.J., Escudero-García, R., Cortés-Penagos, C., Cholico-González, D.F.: Chem-
ical characterization and quantification of metallic gold from copper tailings in the western
region of Mexico, MRS Adv. (2022) https://doi.org/10.1557/s43580-022-00437-6
8. Mandigan, M.T., Martinko, J.M., Brock, J.P.: Biología de los microorganismos. 12va ed.,
Pearson-Prentice Hall, Madrid España, (2009)
9. Bird, L.J., Bonnefoy, V., Newman, D.K.: Bioenergetic challenges of microbial iron metabolims.
Trends Microbiol.Microbiol. 19, 330–340 (2011)
10. Bonneville, S., Behrends, T., Cappellan, P.V., Hyacinthe, C., Röling, W.F.M.: Reduction of
Fe(III) colloids by Shewanella putrefaciens: A kinetic model. Geochim. Cosmochim. Acta.
Cosmochim. Acta 70, 5842–5854 (2006)
11. Castro, L., Blázquez, M.L., González, F., Muñoz, J.A., Ballester, A.: Anaerobic bioleaching
of jarosites by Shewanella putrefaciens, influence of chelators and biofilm formation.
Hydrometallurgy. 168, 56–63 (2017) https://doi.org/10.1016/j.hydromet.2016.08.002
12. Zhang, S., Wang, Y., Song, H., Lu, J., Yuan, Z., Guo, J.: Copper nanoparticles and copper
ions promote horizontal transfer of plasmid-mediated multi-antibiotic resistance genes across
bacterial genera. Environ. Int. 129, 478–487 (2019) https://doi.org/10.1016/j.envint.2019.
05.054
13. Pal, C., Asiani, K., Arya, S., Rensing, C., Stekel, D.J., Larsson, D.G.J., Hobman, J.L.: Chapter
seven - metal resistance and its association with antibiotic resistance, in: R.K.B.T.-A. In: Poole,
M.P. (ed.) Microbiol. Met. Ions, pp. 261–313. Academic Press, (2017) https://doi.org/10.1016/
bs.ampbs.2017.02.001.
14. Kaksonen, A.H., Boxall, N.J., Gumulya, Y., Khaleque, H.N., Morris, C., Bohu, T., Cheng,
K.Y., Usher, K.M., Lakaniemi, A.-M.: Recent progress in biohydrometallurgy and microbial
characterisation. Hydrometallurgy. 180, 7–25 (2018) https://doi.org/10.1016/j.hydromet.2018.
06.018
Biocidal Effect of Copper Contained in a Mineral Tailing on the Growth … 589
Asra Hamidi
A. Hamidi (B)
Free Biotechnology Researcher, Erbil, Kurdistan, Iraq
e-mail: Asra.Mahmood.Reza@gmail.com
© The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd. 2023 591
R. Malviya and S. Sundram (eds.), Engineered Biomaterials, Engineering Materials,
https://doi.org/10.1007/978-981-99-6698-1_21
592 A. Hamidi
1 Introduction
“The adeno-associated virus (AAV) is a tiny virus that primarily affects mammals,
but there is currently no evidence to suggest that it causes any illnesses. The engi-
neered type of adeno-associated virus (AAV) that cannot replicate has emerged as a
frequently utilized carrier for genome editing and gene transfer projects, particularly
those utilizing in vivo methods” [1, 2]. “Treatments based on genome transfer, editing
and programming are basic and new treatments that generally try to repair congen-
ital or acquired genetic mutations. Newer studies show the direction and progress of
treatments based on genetic material, from the laboratory phase to the creation of
practical and real solutions. Meanwhile, AAV vectors, with high delivery efficiency,
specific target tissues, replication defects, and significant immunological success
rates, have been used repeatedly and have gained relatively good validity” [3]. “The
results of research have indicated that AAV is associated with fewer immunological
issues compared to most other viruses in the context of in vivo genetic engineering
when used as a vector. This has contributed to AAV becoming the most popular viral
vector for genetic engineering. However, there is still a possibility that the immune
system may eliminate the virus or that it may mutate, which raises concerns about
the safety of in vivo applications. Another potential issue is the size limit on genetic
material that can be packaged into AAV, which may limit its usefulness in certain
applications That is why some researchers prefer to use separate AAVs to deliver
sgRNA and Cas9 and co-inject them, while others have investigated the use of smaller
Cas proteins or target fragments for gene editing” [4].
“A suitable viral vector should not only have mass production capability but
also exhibit precise delivery efficiency, reasonable price, acceptable packaging
constraints, and low immunological challenges” [3].
“It is crucial to assess the levels of anti-adeno antibodies in individuals to ensure
the safety and effectiveness of genetic treatments utilizing AAV. This is particularly
important as a considerable fraction of humans already have pre-existing immunity to
adenoviruses. Screening tests have been developed to identify patients with signif-
icant antibody levels and prevent adverse outcomes during the treatment process.
These tests not only help prevent transduction inhibition but also allow for the
measurement of total antibody levels, which can be a valuable predictor of AAV
treatment safety” [5]. “In addition, the matters of large-scale production and the
expensive nature of using AAV vectors for private and commercial purposes are
significant factors to take into account when considering the practical utilization of
this carrier in genetic therapies” [6].
This chapter will begin by discussing the significance and applications of AAV
vectors in genome editing, programming, and gene transfer. While providing an
overview of the potential hurdles in utilizing this vector, we will delve into the specific
immunological obstacles that may arise during in vivo projects. We will then explore
various solutions that have been proposed to overcome these challenges. Finally, we
will discuss the future possibilities and progress in the utilization of AAV vectors,
specifically in gene transfers, DNA editing, and programming.
Examining the Problems and Possibility of Immunological Control … 593
2 Research Methodology
This study examines the immunological challenges associated with using AAV
vectors in genetic engineering and proposes solutions to overcome them. We
conducted a systematic review of 58 scientific articles published between 2018 and
2023, searching for information on AAV vectors, gene therapy, genome editing,
epigenome editing, and immunology. We synthesized the information and analyzed
it by categorizing the challenges and proposed solutions into themes. The study’s
limitations include potential bias and the need for further validation of proposed
solutions through experiments. However, the study provides a comprehensive and
systematic approach to understanding the immunological challenges of using AAV
vectors in genetic engineering.
in DNA sequences such as insertion, deletion, and base substitution. Prime editing
does not rely on double-stranded DNA cleavage or the use of donor DNA templates.
Instead, it uses an engineered sgRNA connected to reverse transcriptase and nCas9
to synthesize new DNA strands and introduce them to the targeted site. Since prime
editing can address a wide range of genetic mutations associated with human genetic
problems, it holds great potential as a clinical therapy tool” [10].
“The potential of large DNA fragment insertion for treating genetic diseases is
significant, but current gene editing technologies can only effectively insert short
fragments. However, a new method for gene editing, known as GRAND editing, has
been developed to enable the precise insertion of big DNA pieces devoid of the need
for DNA donors. Unlike prime editors, which require templates that can hybridize
with the target sequence, GRAND editing uses a pair of guide-RNAs with reverse
transcriptase templates that are not homologous to the intended region but instead
match each other. This method has achieved a success rate of up to 63.0% for a 150-
bp insertion with minimal by-products, and 28.4% for a 250-bp insertion. Although
it can accommodate insertions of up to ~1 kb, the potency decreases for fragments
larger than 400 bp” [11].
“Safe and effective delivery methods for gene editing agents are essential for
successful in vivo gene editing therapies aimed at treating genetic diseases. Various
delivery technologies have been studied, including viral vectors, lipid nanoparticles,
and virus-like particles, to achieve this goal. These methods play a crucial role in
enabling therapeutic in vivo gene editing” [4].
“Eukaryotic cells have a defense mechanism against foreign DNA different from
the CRISPR system used by prokaryotes. They use DNA sensors that add chem-
ical markers to exogenous DNA, inhibiting its transcription and reducing its impact
on the cell. However, this poses a challenge in gene therapy. The DNA-sensing
machinery of the host can detect certain elements of Recombinant AAV vectors,
which are frequently utilized in gene therapy, resulting in decreased expression of
the transgene. In addition, the HUSH complex can deposit repressive histone marks
on viral DNA, and the NP220 DNA sensor can bind to double-stranded DNA from
either host or foreign viral genomes. Following this interaction, the HUSH complex
is recruited, which in turn facilitates the recruitment of histone-modifying enzymes,
such as HDAC and SETDB1. These enzymes remove acetyl marks and add repres-
sive methyl marks, respectively. In summary, NP220-HUSH complex plays a role
in epigenetic silencing of DNA by modifying histones which should be taken into
consideration when planning genome editing and gene therapy projects based on
AAV” [12].
“Epigenome editing is a method used to manipulate the epigenome of specific
DNA to determine the role of the epigenome in a particular gene. This technique
involves using a catalytically inactive mutant of Cas9 (dCas9) to recruit epige-
netic modifying enzymes to specific target sequences. While forward genetics is
a phenotype-to-gene approach, reverse genetics analyzes the phenotype caused by
the mutation of specific genes. Epigenetic modifications are crucial for the activa-
tion of genes regulation in the course of embryogenesis and are linked to different
Examining the Problems and Possibility of Immunological Control … 595
maladies. DNA methylation and histone modifications are responsible for transmit-
ting information for controlling gene expression in a manner unrelated to alterations
in DNA sequence. Epigenome editing technology has the potential to expand the
use of epigenetic therapy in solid tumors” [13]. In the meantime, “AAV vectors are
considered one of the most interesting options for epigenome editing” [14].
Furthermore, “Epic Bio, a company established by Stanley Qi, a bioengineer from
Stanford, has created a technology known as Gene Expression Modulation System
(GEMS), which enables the modification of gene expression without making perma-
nent alterations to DNA. GEMS comprises DNA-binding proteins, guide RNAs,
and modulators, which can activate or repress gene expression and modify the gene
locus. They use the largest library of novel modulators, which can be customized
for specific effects upon a target gene. CasMINI is a modified Cas DNA-binding
protein that has been engineered to operate strongly within mammalian cells. Addi-
tionally, it is smaller in size than both Cas9 and Cas12a, measuring less than half
of their dimensions. Epic has licensed CasMINI for human use from Stanford and
can deliver it to target organs via adeno-associated virus (AAV), lipid nanoparticles
(LNPs), or lentiviruses. They want to de-risk its initial programs by using AAV as
it has more clinical experience, but it is not tied to AAV and aims to validate the
efficacy of the constructs with other delivery methods” [15].
“AAVs are tiny viruses that do not have an envelope and contain a genome consisting
of single-stranded DNA. They have a capsid structure that is composed of three
proteins arranged in a 1:1:10 ratio, which creates a symmetrical T = 1 icosahedron
structure. The AAV genome is compact and contains two open-reading frames that
are bordered by palindromic inverted terminal repeats arranged in a T-shape. These
repeats enable the genome to be replicated and packaged. The cap gene codes for
the three capsid proteins, while the rep gene encodes four replication proteins” [6]
(Fig. 1).
“AAV vectors are commonly used in genetic engineering as they are highly effec-
tive at transporting to specific cells. Also, they can sustain persistent transgene expres-
sion” [17]. “ AAV vectors can transduce cells that are either actively dividing or non-
dividing, and they can persist as an extrachromosomal element without integrating
into the host cell’s genome” [18]. However, “they still pose some immunological chal-
lenges and carry a slight possibility of causing mutagenesis by insertion” [19]. “AAV
vectors have demonstrated high delivery efficiency, replication defects, specificity
toward target tissues, and notable immunological success rates, and have therefore
gained considerable credibility in their repeated use. Compared to other viral vectors,
AAV poses fewer immunological challenges when utilized for in vivo gene transfer
and DNA edit, and has thus become the preferred vector in this field” [3]. “Despite
596 A. Hamidi
Fig. 1 The diagram shows the use of AAVs in gene replacement therapy, where the therapeutic
sequence takes the place of the rep and cap genes. The AAV capsid structure, which is highly
symmetrical, consists of three proteins and has been crafted to optimize non-covalent interactions
to accommodate the genetic material carried by the virus [6, 16]
this, there remain potential concerns regarding the safety of in vivo applications, such
as the possibility of the immune system eliminating or mutating the virus. AAV’s
effectiveness for specific applications is limited by the size constraints of the genetic
material it can accommodate, which requires to be addressed. Researchers have
proposed various solutions, including the use of separate AAVs to attach sgRNA and
Cas9, the exploration of smaller Cas proteins, and the targeting of smaller genetic
fragments for transfer” [4].
In addition to the advantages and challenges of using AAV vectors, there are some
other important aspects to consider.
“AAV vectors offer versatility in targeting different tissues due to their diverse
capsids. Researchers can modify the tropism of these vectors by combining natural
serotype capsids” [11]. “More than 13 naturally occurring AAV serotypes have been
identified, in addition to over 108 capsid variants that have been classified” [7]. “To
construct an AAV vector, the therapeutic gene, and genetic material must be placed
between two sets of inverted terminal repeats (ITRs) obtained from AAV serotype 2.
The size of the entire construct must not exceed 4.6 kb” [18]. For example, “AAV9
has been observed to be effective in systemically delivering therapeutic agents to the
nervous system. This includes the capacity to penetrate natural obstacles, such as the
cerebral vascular barrier, to reach its target” [6].
Examining the Problems and Possibility of Immunological Control … 597
“The production and purification of AAV vectors require careful handling and elim-
ination of any probable impurities. A commonly used method for AAV production
is transient transfection of mammalian cell lines, but it has limitations in terms of
scalability and downstream processing. An alternative method uses Sf9 insect cells
and baculovirus but this has limitations, so TIPS cells are utilized to overcome these
issues. AAV vectors are manufactured through a process that involves the use of
serum-free suspension culture, where TIPS cells and new Sf9 cells are co-cultured.
The resulting AAV particles are obtained by lysing the producer cells” [18] (Fig. 2).
“Presently, the preferred way of purifying AAV for clinical and research purposes
is through ultracentrifugation on a cesium chloride or iodixanol density. Nevertheless,
this process has certain disadvantages, such as being time-consuming and difficult to
expand, and it may also lead to impurities being co-purified. On the other hand, liquid
chromatography, particularly high-performance liquid chromatography (HPLC), is
Fig. 2 The process of purifying AAV through AAVX affinity chromatography [19]
598 A. Hamidi
a more effective and scalable method for purifying AAV. However, this technique
requires optimization for different AAV serotypes to work efficiently. Recently,
commercial AAV binding resins have been introduced that provide a more flexible
and efficient method for purifying AAV for research purposes than ultracentrifuga-
tion or traditional chromatographic methods. These resins require serotype-specific
optimization and may have lower binding efficiency for certain AAV serotypes, but
overall they offer promising results” [19].
“AAV-based genome editing may not work effectively in individuals with existing
immune responses to certain AAV variations” [5]. “The body’s exposure to natural
AAV serotypes can produce specific antibodies, such as neutralizing antibodies
(NAb), which can prevent AAV vectors from entering the objective cells, reducing
Delivery effectiveness and lowering the synthesis of therapeutic agents. Patient exclu-
sion from clinical trials is prevalent due to high titers of neutralizing antibodies
against AAV vectors, which can vary depending on demographic factors. It is essen-
tial to evaluate several AAV serotypes and determine the collective occurrence of the
primary serotypes employed in AAV-mediated treatment because cross-reactivity
can arise between distinct AAV serotypes” [20] (Table 1).
“The binding of anti-AAV AAV capsid antibodies can impact the delivery of these
vectors. This effect can occur in two ways: Neutralizing antibodies can block vector
transfer, whereas non-neutralizing antibodies have no effect. Both types of antibodies
can also cause the redirection of the vector to secondary lymphoid organs, as illus-
trated in Fig. 3. However, administering rAAV vectors directly into protected sites
might be less susceptible to the impact of non-systemic AAV-targeting antibodies”
[21].
Table 1 Comparing tests for transduction inhibition and total antibody detection [5]
Evaluation TI evaluation TAb evaluation
Fundamental A reporter gene encoding rAAV A capture-based method that uses
vector is used in a cell-based test immunochemical analysis (For example,
ELISA or ECLA)
Cell transduction inhibition tests Detects the existence of antibodies
can be performed using against AAV (with no regard to their
blood-derived samples containing neutralizing capacity)
neutralizing antibodies and
Non-immunoglobulin components
Benefits The method determines the extent This unique approach is less complex and
of cell transduction inhibition by more amenable to automation than TI
identifying neutralizing antibodies tests. Therefore, it holds promise aimed
and additional neutralizing agents at the progress of diagnostic tests that
that may impact transduction could be utilized in diverse medical
laboratory settings
A frequent design involves The detection of specific
selecting the same rAAV capsid as immunoglobulin isotypes is possible
gene therapy with options for cell using an antigen-capture format
lines and reporter genes
Possible Necessary specialized experts It is difficult to standardize due to the
drawbacks various available platforms and format
options
Can be difficult to develop into It is possible that low-affinity non-specific
readily available commercial test antibodies could be detected, but their
kits presence potentially does not impact the
clinical outcome of gene therapy
“AAV vectors are generally considered safe but require thorough pre-clinical testing
and patient monitoring to ensure their safety and effectiveness” [5]. “AAV vectors
have elements that trigger the activation of toll-like receptor (TLR)2 and TLR9,
leading to the discharge of pro-inflammatory signaling molecules and immune system
proteins that promote inflammation and antiviral responses. AAV vector suspensions
may contain impurities from the production process, which can also affect their
immunogenicity. The detection of AAV vectors is greatly influenced by the pres-
ence of plasmacytoid dendritic cells (pDCs) and can secrete and produce substantial
quantities of type I interferon and irritating cytokines in response to viral challenges.
Non-packaged viral DNA in AAV vector production preparations can influence their
immunogenic properties in human pDCs” [23]. “The prevalence of anti-AAV anti-
bodies raises safety concerns for gene therapy. Excluding positive patients from
clinical trials could hinder progress without sufficient evidence of safety risks” [24].
“AAV vectors are a powerful tool for genetic engineering with great potential in
various applications, but their use requires careful consideration of serotype selection,
vector production, pre-existing immunity, and safety concerns” [1].
600 A. Hamidi
Fig. 3 Non-inhibitory antibodies against AAV may Interact with the protein shell of the recombi-
nant adeno-associated virus, but they do not obstruct attachment of the vector or transduction. The
mechanism of neutralizing antibodies against AAV is depicted [22]
“AAV has been successful in delivering therapeutic genes for illnesses like
hemophilia, spinal muscular atrophy, and inherited retinal disorders during testing on
patients” [25]. “Genome editing tools like CRISPR-Cas9 can be delivered by adeno-
associated viruses (AAVs) to target cells, enabling accurate genome modifications”
[2]. Additionally, “ AAV vectors are frequently utilized in fundamental research to
investigate genetic mechanisms, control, create preclinical models of human illnesses
using animals” [26, 27], and “apply newer genome editing techniques such as prime
editing and base editing” [28].
“AAV vectors have been used in agriculture to deliver genetic modifications to
crops, improving resistance to pests and environmental stressors” [29].
“AAV vectors can target specific cells by incorporating cell-specific promoters
and can mediate extended gene function due to their potential to incorporate into the
recipient cell’s DNA” [30].
“AAV vectors are promising for treating neurological disorders in the central
nervous system like Alzheimer’s, Huntington’s, and Parkinson’s illness” [1]. “AAV
Examining the Problems and Possibility of Immunological Control … 601
vectors are capable of being used to regulate the body’s defense response” [31] and
“have potential in cancer therapy by delivering genes that regulate immune function,
express tumor suppressor genes, or induce apoptosis in cancer cells” [32].“AAV
vectors have limited cargo capacity” [13], They can serve as in vivo carriers, partic-
ularly for CRISPR-based systems such as Cas9, Cas12a, and Cas13a, which find
prominent application in the field of cancer therapy [33] and “pre-existing immunity
can reduce their effectiveness” [5], but “efforts to improve AAV vector technology
show promise for overcoming these challenges” [13]. “Despite these challenges,
AAV vectors offer a valuable tool for genetic engineering with various uses in genome
therapy, immune modulation, and cancer therapy, among others” [1].
Fig. 4 Describing the frequency of existing immunity to viral variants and their distribution across
different geographic regions, using the population of patients with hemophilia A and B as an
illustrative instance [34]
Table 2 Neutralizing
AAV-LK03 AAV3B AAV8
antibodies’ cross-reactivity
with AAV-LK03, 3B, and 8 in AAV-LK03 – 97% 61%
populations [35] AAV3B 67% – 47%
AAV8 79% 87% –
AAV strains, likely due to the high degree of sequence similarity in AAV
capsid proteins. Reporter vectors with genes like GFP or luciferase are used
to measure neutralizing activity against recombinant AAV (rAAV), but the
type of reporter gene and the cell type used can affect the results. TAb tests
detect all capsid-bound antibodies but not non-antibody neutralizing factors.
Choosing recombinant adeno-associated virus capsid-based tools is crucial for
developing antibody tests” [5].
“Factors like donor health and immunosuppressive drugs affect AAV-
specific antibody prevalence. Antibodies can interact with multiple AAV strains
due to a high degree of sequence homology among capsid proteins. Reporter
vectors with genes like GFP or luciferase measure neutralizing activity, but the
choice of gene and cell type impacts results. TAb tests detect capsid-bound
antibodies but not non-antibody neutralizing factors. Selecting recombinant
vector capsid-based tools is crucial for accurate antibody test development”
[38].
5.2. Immunological reactions to the vector: “The viral vectors used in genome
therapy can still induce immune responses in patients without existing immu-
nity, leading to clearance of AAV and decreased gene expression” [39]. “The
immune response involves both natural and acquired immunity, with the detec-
tion of foreign nucleic acids triggering the secretion of inflammatory signaling
Examining the Problems and Possibility of Immunological Control … 603
Fig. 5 The Perocheau et al. study examined the prevalence and titers of neutralizing antibodies and
IgG to AAV serotypes AAV-LK03, AAV-3B, and AAV-8 in populations. The results were presented
in eight sections, including the prevalence and titers of antibodies based on age, sex, and cross-
reactivity between serotypes. The results were shown using vertical bars and box-and-whisker plots
[37]
machinery, including the DNA damage response (DDR) proteins, and induce
host-mediated silencing of the transgene. This silencing can occur through
various mechanisms such as DNA methylation and histone modifications. The
selection of AAV shell may also impact host-mediated silencing. The human
silencing hub (HUSH) complex, made up of MPP8, TASOR, and PPHLN1, is
involved in silencing retroelements and can also silence rAAV genomes. The
NP220 protein can recruit the human silencing hub complex and the enzyme
SETDB1 that adds suppressive histone modifications on retroviral genomes,
but without requiring the HUSH complex, it can repress the gene performance
from the extrachromosomal DNA of retroviruses that are closely relevant, such
as HIV-1 and Mason-Pfizer monkey virus. The selection of the AAV capsid
(serotype) has a significant impact on these mechanisms and can ascertain the
gene repression of integrated viral DNA mediated by the host” [12].
5.4. Adjuvants and immune modulators: “The use of adjuvants or immune modu-
lators is a potential approach to boost the immunological reaction to AAV
vectors in gene therapy” [43]. “Adjuvants can stabilize pathogens and raise
the immunological function, especially in older adults and immunocompro-
mised individuals. The most widely used adjuvant is alum, but research is
ongoing to develop combined adjuvant formulations that elicit strong humoral
and cellular immune responses” [44]. “Recombinant adenoviruses are gaining
popularity as COVID-19 vaccines due to their ability to stimulate B and T cell-
initiated immunological function. However, endeavors to enhance their ability
to stimulate an immune response by administering them with conventional
immunostimulants have been mostly ineffective. The use of STING ligands,
like 2' 3' -cGAMP, before transgene expression can reduce immunity, while the
parallel activity of 2' 3' -cGAMP with the introduced gene can enhance induc-
tion of the STING signaling pathway in advanced stages of viral contamination.
The timing of adjuvant effect is crucial and may have broader implications for
the application of other non-biological immunostimulants” [45].
5.5. Alternative vector systems: “Alternative vector systems have been explored to
address the immune challenges associated with AAV vectors. Lentiviral vectors
and non-viral gene delivery methods are potential alternatives to AAV vectors”
[46]. “There are various drawbacks associated with using AAV vectors as a
means of delivering genes, including immunogenicity, cytotoxicity, and the
risk of gene disruption through viral integration. When viral genetic material
integrates into the wrong location on a chromosome, it can lead to abnormal
changes in cells leading to cancer development. Research in populations has
also shown that this process is often associated with an Immunological reaction.
Techniques that utilize physical means to facilitate the delivery of nucleic acids
are restricted to DNA applications. Nonviral vectors are more attractive than
viral vectors since they may address issues related to potential adverse effects
on the immune system and cellular health. Nonviral vectors are also a less
expensive option and are easy to generate at a massive level” [47].
5.6. Immune evasion strategies: “There are ongoing efforts to develop immune
evasion strategies for AAV vectors. For example, engineering AAV vectors
Examining the Problems and Possibility of Immunological Control … 605
Fig. 6 Illustration of manipulating capsids to enhance their capability to target the brain and
spinal cord. a Techniques for generating diverse AAV capsids. b Procedure for modifying capsids,
which involves selecting from multiple species and customizing their capability to selectively target
particular cells evade the immune system, and package the virus [48]
with modified capsid proteins or using novel AAV serotypes can help to evade
pre-existing immunity and enhance gene expression” [48] (Fig. 6).
“The structure of AAV capsids can be altered to improve vector delivery
efficiency and avoid triggering immunological defenses. The development of
engineered AAV capsids that target special tissue involves three main tech-
niques: natural discovery, deliberate engineering, and evolutionary optimiza-
tion. Natural discovery involves obtaining AAV from various hosts and identi-
fying a distinct genetic code for the capsids. The code is cloned and analyzed to
confirm the modified capsids’ ability to target the special tissue. Rational design
involves utilizing current understanding of capsid biology to insert binding
proteins into specific regions of the AAV capsid protein, thereby enhancing
the therapeutic properties of the resulting capsids. Directed evolution uses
mutagenesis techniques to create a large number of capsid libraries, which are
then exposed to specific conditions that favor the development of the desired
characteristics.
While these methods offer great potential, they also present several chal-
lenges. Natural discovery is limited by low delivery efficiency and the time-
consuming process of isolating and characterizing novel capsids. Intelligent
606 A. Hamidi
help to regulate the immunological responses to the viral vectors and improve
their effectiveness” [45].
6.4. Use of immune-privileged sites: “Administering Adeno-associated-virus
vectors to immunologically protected areas, like CNS or eye, can help to avoid
immune responses. However, this approach is not always feasible, and the
effectiveness of the therapy may be limited by the site of administration” [49].
6.5. Selection of AAV serotypes: “Selecting AAV serotypes that are less prone
to inducing immune responses can also help to mitigate immunological
challenges” [19].
6.6. Addressing pre-existing immunity: “Strategies to address pre-existing immu-
nity, such as selecting AAV serotypes that are less commonly encountered or
employing methods to deplete or improve neutralizing antibodies, can also
enhance the impact of gene therapy using AAV vectors” [20].
“rAAV has surfaced as a potential gene transfer mechanism for addressing mono-
genic diseases in various tissues. When compared to adenoviral vectors, rAAV
exhibits lower immunogenicity due to their weaker activation of natural defense
mechanisms and specialized immune cells. However, recombinant adeno-associated
virus vectors carrying diverse transgenes can provoke strong immune reactions
in different conditions, resulting in the removal of genetically modified cells and
decreased activity of the engineered gene. Delivery of rAAV in humans and primates
may activate considerable cytotoxic CD8+T cell reactions to both vector and
transgene-encoded protein, ultimately resulting in the removal of transduced cells
and complete loss of transgene expression. Moreover, the development of antibodies
that bind to and inactivate the capsid proteins can hinder subsequent vector adminis-
tration and speed up the degradation of the therapeutic agent. Consequently, crafting
approaches to overcome immunological reactions and ensuring the sustained produc-
tion of therapeutic proteins is the primary challenge for translating rAAV vectors to
clinical settings” [50].
“Although AAV vectors have size limitations for delivering transgenes, these limi-
tations can be addressed by modifying the genes to remove unnecessary protein struc-
tures and shorten the cDNA. For instance, AAV vectors can be used for Duchenne
muscular dystrophy by eliminating protein segments from the dystrophin gene
sequence, which is over 11 kilobases in size. A similar approach has been used
in gene therapies for hemophilia A, where the normal FVIII protein-gene sequence
is approximately 7 kb. By using B-domain-deleted FVIII protein isoforms, the gene
sequence for the FVIII protein has been decreased to just over the maximum size
for encapsidation. This allows successful packaging of vector genomes in the viral
capsids, modifying the size of vector genomes can lead to changes in gene expression
when relative to the conventional-sized ones. Research conducted on mice utilizing
AAV-8 found that a larger vector, containing FVIII and exceeding the standard size at
5.1 kb, resulted in 2–3 times less expression of FVIII protein in plasma and decreased
vector genome levels in hepatocytes after 3 months, as compared to a typical-sized
vector at 4.6 kb” [51].
608 A. Hamidi
“Scientists have developed a more efficient and effective delivery system for prime
editing, a gene editing technique that corrects mutations without inducing ds-DNA
cleavage. The team made improvements to the reverse transcriptase (RT) moiety and
truncated the prime editing components to overcome the viral vector size restrictions,
which at present, there is limited support for effective transfer of the bulky prime
editing tools. Consequently, a smaller prime editing system with a gene sequence opti-
mized for expression and size advantages was developed. The team also optimized
the split intein prime editing system and identified Cas9 breaking points that coupled
with the shortened prime editing components transferred by double-stranded AAVs
triggered improved AAV levels and prime editing productivity. Furthermore, they
demonstrated the split prime editing components transferred by double-stranded-
AAV1, particularly altered prime editing system 1024, where able to induce changes
in nucleotides and insert genetic material at four natural locations within human
tissue cultures and they also demonstrated successful editing of the Dnmt1 gene in
the retina of live adult mice” [52]. “Another group of researchers constructed four
split prime editing systems using Rma intein and identified two efficient split prime
editing systems that were able to facilitate point substitution and addition of endoge-
nous areas in human cells when delivered by dual-AAV1. They then successfully
edited Dnmt1 in adult mouse retina in vivo using one of the identified efficient split
prime editing systems” [53].
“Epigenome editing techniques have proven effective in manipulating specific
gene targets, but their delivery through adeno-associated virus (AAV) vectors has
been constrained by the small assembly restrictions of these vectors. Researchers
have addressed the issue by developing a compact deactivated Cas9 repression system
that can be packaged into a single optimized adeno-associated virus. The system
uses a smaller variant of Cas9, derived from Staphylococcus aureus (Sa), and a
novel repressor composed of the condensed transcription repression domain (TRD)
from MeCP2 fused with the KRAB inhibitory domain. This construct, along with
its associated gRNA, may efficiently fit into adeno-associated viruses. The platform
was successfully used to silence APOE, a gene associated with Alzheimer’s disease,
in cell culture as well as in living organisms. The method offers a more efficient and
effective delivery system for epigenome editing” [54].
“The modification of histones by the Human Silencing Hub (HUSH) complex
and NP220 is essential for regulating AAV gene expression by silencing them.
Deletion of NP220 had the most significant effect on the expression of AAV trans-
genes, suggesting a crucial role in controlling the epigenetic suppression of AAV
genomes. However, NP220 may recruit other epigenetic regulators besides the HUSH
complex. NP220 is present and highly conserved in diverse vertebrate species, but
the molecules responsible for inhibiting viral gene activity may vary based on the
virus and host organism. For example, Rous Sarcoma virus is unresponsive to NP220-
induced suppression, perhaps due to the relatively limited amount of cytidine present
in the long terminal repeat regions. An alternative approach to enhance AAV trans-
gene expression is to use low biomolecular weight inhibitors of HDACs, which can
promote histone deacetylation and inhibit host-silencing mechanisms. The impact of
Examining the Problems and Possibility of Immunological Control … 609
“The future of AAV vectors in gene editing and transfer is promising with continued
development likely resulting in safer and more efficient delivery of therapeutic genes
or proteins. Researchers are exploring ways to optimize AAV vectors for specific
applications, enhance their specificity, reduce immunogenicity, and increase their
cargo capacity” [1].
“Researchers are focusing on designing innovative AAV serotypes to more effec-
tively target particular cell populations, which may lead to new treatments for diseases
that are currently difficult to target” [48].
“Scientists are studying methods to combine AAV vectors with other genome
editing tools like CRISPR/Cas9 for better precision and efficacy in gene editing
with fewer off-target effects” [3]. Moreover, “they are constantly improving the
technology of AAV or its substitutes such as Base editing, Prime editing, Grand
editing, and epigenome editing, to achieve more effective gene editing in “in vivo
projects”. The refinement and enhancement of these viral vectors play a crucial role
in advancing these associated tools” [4, 9–12, 14, 15].
“Improving AAV vector design to penetrate the blood–brain barrier and deliver
therapeutic genes or proteins for the treatment of neurodegenerative diseases like
Parkinson’s and Huntington’s is a current research focus. This involves engineering
AAV vectors that can cross the blood–brain barrier and transport the curative agents
directly to the brain” [6].
“Researchers are working to improve the safety and efficiency of AAV vectors
by exploring strategies to reduce the chances of immune responses. One approach
is using small molecules to inhibit the immunological response” [43–45]. “They are
investigating methods to optimize the packaging AAV productivity to increase the
delivery of therapeutic genes or proteins to target cells” [6, 51, 54].
“New techniques are being developed to enhance the manufacturing and scala-
bility of AAV vectors. This is crucial for making AAV-based gene therapies more
widely available and affordable” [18].
“Scientists are developing viral vectors that can target stem cells, which exhibit the
capacity to differentiate into multiple cell types within the organism and are desirable
610 A. Hamidi
for gene therapy applications. These vectors can selectively deliver therapeutic genes
to stem cells, creating new opportunities for regenerative healthcare” [55].
“Current research is centered on the development of adeno-associated viruses that
can treat genetic disorders resulting from mutations in non-coding areas. These areas
make up most of the human genome, and their role in gene expression and regulation
is not fully understood. Scientists are investigating the use of AAV vectors to deliver
therapeutic genes or RNA molecules to non-coding areas, aiming to correct gene
expression and treat conditions such as diabetes, obesity, and heart disease” [8].
Furthermore, “AAV vectors are being investigated as a potential therapy for viral
infections such as hepatitis B and HIV. These vectors can be customized to deliver
therapeutic RNA molecules or genes that disrupt viral replication, potentially leading
to a functional cure for the illness” [23].
The ongoing advancement of AAV vector technology is expected to produce
even more robust tools for gene editing and transfer, which hold the promise of
transforming the management of diverse illnesses.
References
1. Kang, L., Jin, S., Wang, J., Lv, Z., Xin, C., Tan, C., Zhao, M., Wang, L., Liu, J.: AAV vectors
applied to the treatment of CNS disorders: clinical status and challenges. J. Control. Release
355, 458–473 (2023). https://doi.org/10.1016/j.jconrel.2023.01.067. PMID: 36736907
2. Huang, J., Zhou, Y., Li, J., Lu, A., Liang, C.: CRISPR/Cas systems: delivery and application
in gene therapy. Front. Bioeng. Biotechnol. 22(10), 942325 (2022). https://doi.org/10.3389/
fbioe.2022.942325. PMID:N/A;PMCID:N/A. [Review]
3. Wang, S.W., Gao, C., Zheng, Y.M., et al.: Current applications and future perspective of
CRISPR/Cas9 gene editing in cancer. Mol. Cancer 21(1), 57 (2022). https://doi.org/10.1186/
s12943-022-01518-8
4. Raguram, A., Banskota, S., Liu, D.R.: Therapeutic in vivo delivery of gene editing agents. Cell
185(15), 2806–2827 (2022). https://doi.org/10.1016/j.cell.2022.03.045
5. Schulz, M., Levy, D.I., Petropoulos, C.J., Bashirians, G., Winburn, I., Mahn, M., et al.: Binding
and neutralizing anti-AAV antibodies: detection and implications for rAAV-mediated gene
therapy. Mol. Ther. 31(3), 616–630 (2023). Table 1. https://doi.org/10.1016/j.ymthe.2023.
01.010
6. Marrone, L., Marchi, P.M., Azzouz, M.: Circumventing the packaging limit of AAV-mediated
gene replacement therapy for neurological disorders. Expert Opin. Drug Deliv. 19(10):1163–
1176 (2022). Figure 1. https://doi.org/10.1080/14712598.2022.2012148. PMID: 35144839
7. Arjomandnejad, M., Dasgupta, I., Flotte, T.R., et al.: Immunogenicity of Recombinant Adeno-
Associated Virus (AAV) vectors for gene transfer. BioDrugs (2023). https://doi.org/10.1007/
s40259-023-00585-7. Published March 2 2023. Accessed April 30 2023
8. Chavez, M., Chen, X., Finn, P.B., et al.: Advances in CRISPR therapeutics. Nat. Rev. Nephrol.
19, 9–22 (2023). https://doi.org/10.1038/s41581-022-00636-2. Epub 2022 Oct 24 PMID:
34812125
9. Porto, E.M., Komor, A.C., Slaymaker, I.M., et al.: Base editing: advances and therapeutic
opportunities. Nat. Rev. Drug Discov. 19, 839–859 (2020). https://doi.org/10.1038/s41573-
020-0084-6
10. Kweon, J., Hwang, H.Y., Ryu, H., Jang, A.H., Kim, D., Kim, Y.: Targeted genomic transloca-
tions and inversions generated using a paired prime editing strategy. Mol. Ther. 31(1), 249–259
(2023). https://doi.org/10.1016/j.ymthe.2022.09.008. PMID: 34598185
Examining the Problems and Possibility of Immunological Control … 611
11. Wang, J., He, Z., Wang, G., Zhang, R., Duan, J., Gao, P., Lei, X., Qiu, H., Zhang, C., Zhang, Y.,
Yin, H.: Efficient targeted insertion of large DNA fragments without DNA donors. Nat. Methods
19(10), 1093–1098 (2022). https://doi.org/10.1038/s41592-022-01399-1. PMID: 34616111
12. Das, A., Vijayan, M., Walton, E.M., Stafford, V.G., Fiflis, D.N., Asokan, A.: Epigenetic
silencing of recombinant adeno-associated virus genomes by NP220 and the HUSH complex.
J. Virol. 96(4), e02039–e2121 (2022). https://doi.org/10.1128/JVI.02039-21. PMID: 34918051
13. Hatada, I., Morita, S., Horii, T.: Epigenome editing in mice: the dawn of the reverse epigenetics
era. Gene Genome Editing 3–4, 100012 (2022). ISSN 2666-3880. https://doi.org/10.1016/j.gge
dit.2022.100012
14. Lau, C.H., Suh, Y.: In vivo epigenome editing and transcriptional modulation using CRISPR
technology. Transgenic Res. 27(6), 489–509 (2018). https://doi.org/10.1007/s11248-018-
0096-8. Epub 2018 Oct 4 PMID: 30288626
15. Philippidis, A.: This is epic: epigenomic CRISPR startup goes big by thinking small. GEN
Edge 4(1), 539–546 (2022). https://doi.org/10.1089/genedge.4.1.89
16. Ren, D., Fisson, S., Dalkara, D., Ail, D.: Immune responses to gene editing by viral and non-
viral delivery vectors used in retinal gene therapy. Pharmaceutics 14(9), 1973 (2022). https://
doi.org/10.3390/pharmaceutics14091973
17. Nasimuzzaman, M., Villaveces, S., van der Loo, J.C.M., Alla, S.: Process development for the
production and purification of adeno-associated virus (AAV)2 vector using baculovirus-insect
cell culture system. J. Vis. Exp. 179, e62829 (2022). https://doi.org/10.3791/62829
18. Kenjo, E., Hozumi, H., Makita, Y., et al.: Low immunogenicity of LNP allows repeated adminis-
trations of CRISPR-Cas9 mRNA into skeletal muscle in mice. Nat. Commun. 12, 7101 (2021).
https://doi.org/10.1038/s41467-021-26714-w
19. Florea, M., Nicolaou, F., Pacouret, S., Zinn, E.M., Sanmiguel, J., Andres-Mateos,
E., Unzu, C., Wagers, A.J., Vandenberghe, L.H.: High-efficiency purification of diver-
gent AAV serotypes using AAVX affinity chromatography. Mol. Ther. Methods
Clin. Dev. 28, 146–159 (2023). https://doi.org/10.1016/j.omtm.2022.12.009. Figure 2.
PMID:35694980;PMCID:PMC8742189.fsdfsd
20. Sierra-Delgado, J.A., Likhite, S., Bautista, P.K., Gómez-Ochoa, S.A., Echeverría, L.E., Guío,
E., Vargas, C., Serrano, N.C., Meyer, K.C., Rincon, M.Y.: Prevalence of neutralizing antibodies
against adeno-associated virus serotypes 1, 2, and 9 in non-injected latin american patients with
heart failure—ANVIAS study. Int. J. Mol. Sci. 24(6), 5579 (2023). https://doi.org/10.3390/ijm
s24065579
21. Ronzitti, G., Gross, D.A., Mingozzi, F.: Human immune responses to Adeno-Associated
Virus (AAV) vectors. Front. Immunol. 17(11), 670 (2020). https://doi.org/10.3389/fimmu.2020.
00670. PMID:32362844;PMCID:PMC7190319
22. Schulz, M., Levy, D.I., Petropoulos, C.J., Somanathan, S., Cheng, S.H., Byrne, B.J.: Mech-
anisms for the inhibition of vector transduction and transgene expression by neutralizing
anti-AAV antibodies. Mol. Ther. 31(3), 616–630 (2023). https://doi.org/10.1016/j.ymthe.
2023.01.010. PMID:34610483. Figure 3. Adapted from Schulz et al. Mol Ther. 2023
Mar;31(3):616–630
23. Bucher, K., Rodríguez-Bocanegra, E., Wissinger, B., et al.: Extra-viral DNA in adeno-
associated viral vector preparations induces TLR9-dependent innate immune responses in
human plasmacytoid dendritic cells. Sci. Rep. 13(1), 1890 (2023). https://doi.org/10.1038/s41
598-023-28830-7
24. McNally, J.: Pre-existing immunogenicity: how will cell and gene therapies change our testing
approach? Int. Pharm. Ind. 14(4), 34 (2022)
25. Earley, J., Piletska, E., Ronzitti, G., Piletsky, S.: Evading and overcoming AAV neutralization
in gene therapy. Trends Biotechnol. S0167–7799(22), 00303–00311 (2022). https://doi.org/10.
1016/j.tibtech.2022.11.006
26. Nyberg, W.A., Ark, J., To, A., Clouden, S., Reeder, G., Muldoon, J.J., Chung, J.Y., Xie, W.H.,
Allain, V., Steinhart, Z., Chang, C., Talbot, A., Kim, S., Rosales, A., Havlik, L.P., Pimentel,
H., Asokan, A., Eyquem, J.: An evolved AAV variant enables efficient genetic engineering of
murine T cells. Cell 186(2), 446–460.e19 (2023). https://doi.org/10.1016/j.cell.2022.12.022.
PMID: 34987467
612 A. Hamidi
27. Seo, Y.E., Baine, S.H., Kempton, A.N., Rogers, O.C., Lewis, S., Adegboye, K., Haile, A.,
Griffin, D.A., Peterson, E.L., Pozsgai, E.R., Potter, R.A., Rodino-Klapac, L.R.: Systemic γ-
sarcoglycan AAV gene transfer results in dose-dependent correction of muscle deficits in the
LGMD 2C/R5 mouse model. Mol. Ther. Methods Clin. Dev. 28, 284–299 (2023). https://doi.
org/10.1016/j.omtm.2023.01.004. PMID:33738450;PMCID:PMC7975753
28. Sichani, A., Saber, R., Ranjbar, M., Baneshi, M., Sanati-Nezhad, A.: A Review on advanced
CRISPR-based genome-editing tools: base editing and prime editing. Mol. Biotechnol. (2022).
https://doi.org/10.1007/s12033-022-00639-1
29. Gao, X., Yang, J., Tsang, S.H., Garg, S.: Adeno-associated virus-mediated gene delivery for
the treatment of retinal diseases. Expert Opin. Drug Deliv. 16(8), 833–845 (2019). https://doi.
org/10.1080/17425247.2019.1628199. PMID: 31189496
30. Wang, D., Tai, P.W., Gao, G.: Adeno-associated virus vector as a platform for gene therapy
delivery. Nat. Rev. Drug Discov. 18(5), 358–378 (2019)
31. Gottwick, C., Carambia, A., Herkel, J.: Harnessing the liver to induce antigen-specific immune
tolerance. Semin Immunopathol. 44(4), 475–484 (2022). https://doi.org/10.1007/s00281-022-
00942-8
32. Perocheau, D.P., Cunningham, S.C., Lee, J., Antinao Diaz, J., Waddington, S.N., Gilmour, K.,
Eaglestone, S., Lisowski, L., Thrasher, A.J., Alexander, I.E., Gissen, P., Baruteau, J.: Age-
related seroprevalence of antibodies against AAV-LK03 in a UK population cohort. Human
Gene Ther. 30(1), 79–87 (2019). https://doi.org/10.1089/hum.2018.098. [Epub ahead of print:
October 19, 2018; Epub ahead of editing: July 20, 2018]
33. Ahmadi, S.E., Soleymani, M., Shahriyary, F. et al.: Viral vectors and extracellular vesicles:
innate delivery systems utilized in CRISPR/Cas-mediated cancer therapy. Cancer Gene Ther.
(2023). https://doi.org/10.1038/s41417-023-00597-z
34. Schulz et al.: Binding and neutralizing anti-AAV antibodies: detection and implications for
rAAV-mediated gene therapy. Mol Ther. 31(3), 616–630 (2023). Figure 4
35. Perocheau DP, Cunningham SC, Lee J, Diaz JA, Waddington SN, Gilmour K, Eaglestone S,
Lisowski L, Thrasher AJ, Alexander IE, Gissen P, Baruteau J. Age-related seroprevalence of
antibodies against AAV-LK03 in a UK population cohort. Human Gene Ther. 30(1), 79–87
(2019). Table 2. Seroprevalence of antibodies against AAV-LK03 in a UK population cohort
by age group. https://doi.org/10.1089/hum.2018.098
36. Perocheau, D.P., Cunningham, S.C., Lee, J., Diaz JA, Waddington SN, Gilmour K, et al.
Age-related seroprevalence of antibodies against AAV-LK03 in a UK population cohort
[Figure 4]. Human Gene Ther. 30(1), 79-87 (2019). https://doi.org/10.1089/hum.2018.098.
PMID: 30346857
37. Perocheau, D.P., Cunningham, S.C., Lee, J., Diaz, J.A., Waddington, S.N., Gilmour, K.,
et al.: Age-related seroprevalence of antibodies against AAV-LK03 in a UK population cohort
[Figure 5]. Human Gene Ther. 30(1), 79-87 (2019). https://doi.org/10.1089/hum.2018.098.
PMID: 30346857
38. Rapti, K., Grimm, D.: Adeno-Associated Viruses (AAV) and host immunity—A race between
the hare and the hedgehog. Front. Immunol. 29(12), 753467 (2021). https://doi.org/10.3389/
fimmu.2021.753467
39. Mingozzi, F., High, K.A.: Immune responses to AAV vectors: overcoming barriers to successful
gene therapy. Blood 122(1), 23–36 (2013). https://doi.org/10.1182/blood-2013-01-306647
40. Kang, Q., Sun, W., Zhang, J., et al.: Adeno-associated virus vectors: current status and future
perspectives in gene therapy. Int. J. Mol. Sci. 22(10), 5171 (2021). https://doi.org/10.3390/ijm
s22105171
41. Gorovits, B., Azadeh, M., Buchlis, G., et al.: Evaluation of cellular immune response to adeno-
associated virus-based gene therapy. AAPS J. 25(3), 47 (2023). https://doi.org/10.1208/s12
248-023-00814-5. PMID: 32346741
42. Whitehead, M., Osborne, A., Yu-Wai-Man, P., Martin, K.: Humoral immune responses to AAV
gene therapy in the ocular compartment. Biol. Rev. (2021). https://doi.org/10.1111/brv.12718
43. Nidetz, N.F., McGee, M.C., Tse, L.V., Li, C., Cong, L., Li, Y., Huang, W.: Adeno-associated
viral vector-mediated immune responses: understanding barriers to gene delivery. Pharmacol.
Ther. 207, 107453 (2020). https://doi.org/10.1016/j.pharmthera.2019.107453
Examining the Problems and Possibility of Immunological Control … 613
44. Retnakumar, S.V., Bonam, S.R., Hu, H., Bayry, J.: Vaccines and vaccine adjuvants/
immunomodulators for infectious diseases. Vaccines 11(2), 383 (2023). https://doi.org/10.
3390/vaccines11020383
45. Padron-Regalado, E., Ulaszewska, M., Douglas, A.D., et al.: STING-pathway modulation to
enhance the immunogenicity of adenoviral-vectored vaccines. Sci. Rep. 12(1), 14464 (2022).
https://doi.org/10.1038/s41598-022-18750-3
46. Farzanehpour, M., Miri, A., Alvanegh, A.G., Esmaeili, G.H.: Viral vectors, exosomes, and
vexosomes: potential armamentarium for delivering CRISPR/Cas to cancer cells. Biochem.
Pharmacol. 212, 115555 (2023). https://doi.org/10.1016/j.bcp.2023.115555. PMID: 34119860
47. Df Kanvinde, S., Kulkarni, T., Deodhar, S., Bhattacharya, D., Dasgupta, A.: Non-viral vectors
for delivery of nucleic acid therapies for cancer. Biotech 11(1), 6 (2022). https://doi.org/10.
3390/biotech11010006
48. Ghauri, M.S., Ou, L.: AAV engineering for improving tropism to the central nervous system.
Biology 12(2), 186 (2023). Figure 6. https://doi.org/10.3390/biology12020186
49. Bucher, K., Rodríguez-Bocanegra, E., Dauletbekov, D., Fischer, M.D.: Immune responses to
retinal gene therapy using adeno-associated viral vectors—Implications for treatment success
and safety. Prog Retin Eye Res. 83, 100915 (2021). ISSN 1350-9462. https://doi.org/10.1016/
j.preteyeres.2020.100915
50. Muhuri, M., Levy, D.I., Schulz, M., McCarty, D., Gao, G.: Durability of transgene expression
after rAAV gene therapy. Mol. Ther. 30(4), 1364–1380 (2022). https://doi.org/10.1016/j.ymthe.
2022.03.004. PMID: 35261636
51. Handyside, B., Ismail, A.M., Zhang, L., Yates, B., Xie, L., Sihn, C.R., Murphy, R., Bouwman,
T., Kim, C.K., De Angelis, R., Karim, O.A., McIntosh, N.L., Doss, M.X., Shroff, S., Pungor,
E., Bhat, V.S., Bullens, S., Bunting, S., Fong, S.: Vector genome loss and epigenetic modi-
fications mediate decline in transgene expression of AAV5 vectors produced in mammalian
and insect cells. Mol. Ther. 30(12), 3570–3586 (2022). https://doi.org/10.1016/j.ymthe.2022.
11.001. PMID: 34857541
52. Gao, Z., Ravendran, S., Mikkelsen, N.S., Haldrup, J., Cai, H., Ding, X., Paludan, S.R., Thomsen,
M.K., Mikkelsen, J.G., Bak, R.O.: A truncated reverse transcriptase enhances prime editing by
split AAV vectors. Mol. Ther. 30(9), 2942–2951 (2022). https://doi.org/10.1016/j.ymthe.2022.
07.001. PMID: 34304054
53. Zhi, S., Chen, Y., Wu, G., Wen, J., Wu, J., Liu, Q., Li, Y., Kang, R., Hu, S., Wang, J., Liang,
P., Huang, J.: Dual-AAV delivering split prime editor system for in vivo genome editing. Mol.
Ther. 30(1), 283–294 (2022). https://doi.org/10.1016/j.ymthe.2021.07.011
54. Kantor, B., Odonovan, B., Rittiner, J., Lindner, N., Dong, W., Zhang, A., Nicholls, P., Chiba-
Falek, O.: All-in-one AAV-delivered epigenome-editing platform: proof-of-concept and ther-
apeutic implications for neurodegenerative disorders. bioRxiv (2023). https://doi.org/10.1101/
2023.04.14.536951
55. Ferrari, S., Jacob, A., Cesana, D., Laugel, M., Beretta, S., Varesi, A., Unali, G., Conti, A.,
Canarutto, D., Albano, L., Calabria, A., Vavassori, V., Cipriani, C., Castiello, M.C., Esposito,
S., Brombin, C., Cugnata, F., Adjali, O., Ayuso, E., Merelli, I., Villa, A., Di Micco, R., Kajaste-
Rudnitski, A., Montini, E., Penaud-Budloo, M., Naldini, L.: Choice of template delivery miti-
gates the genotoxic risk and adverse impact of editing in human hematopoietic stem cells. Cell
Stem Cell 29(10), 1428-1444.e9 (2022). https://doi.org/10.1016/j.stem.2022.09.001
614 A. Hamidi
O
Ophthalmology, 345, 397, 483 R
Recent trends and future perspectives, 575
Recombinant DNA technology, 48, 50, 51,
P 61
Pco and Cop system, 580 Reconstructive surgery, 119
Peptide/DNA-gold nanoparticle conjugates, Regenerative medicine, 74, 153, 181, 361,
573 362, 368, 372, 377, 385, 395
Subject Index 619