1meckling K A Ed Nutrient Drug Interactions PDF
1meckling K A Ed Nutrient Drug Interactions PDF
1meckling K A Ed Nutrient Drug Interactions PDF
Nutrient–Drug
Interactions
Edited by
Kelly Anne Meckling
This book contains information obtained from authentic and highly regarded sources. Reprinted mate-
rial is quoted with permission, and sources are indicated. A wide variety of references are listed. Reason-
able efforts have been made to publish reliable data and information, but the author and the publisher
cannot assume responsibility for the validity of all materials or for the consequences of their use.
No part of this book may be reprinted, reproduced, transmitted, or utilized in any form by any elec-
tronic, mechanical, or other means, now known or hereafter invented, including photocopying, micro-
filming, and recording, or in any information storage or retrieval system, without written permission
from the publishers.
For permission to photocopy or use material electronically from this work, please access www.copy-
right.com (http://www.copyright.com/) or contact the Copyright Clearance Center, Inc. (CCC) 222
Rosewood Drive, Danvers, MA 01923, 978-750-8400. CCC is a not-for-profit organization that provides
licenses and registration for a variety of users. For organizations that have been granted a photocopy
license by the CCC, a separate system of payment has been arranged.
Trademark Notice: Product or corporate names may be trademarks or registered trademarks, and are
used only for identification and explanation without intent to infringe.
RM302.4.N85 2006
615’.70452--dc22 2006042561
Introduction
The idea of food as medicine has been around for millennia. However, the
acceptance of specific foods, food components, extracts, herbals and supplements
as part of conventional medical practice in industrialized countries is still a highly
controversial subject. Western medicine has largely depended on the availability
of pharmacologic agents or “drugs” for the prevention and treatment of human
disease with lesser emphasis on lifestyle and dietary habits as major contributory
factors, particularly with respect to chronic disease prevention. Furthermore, there
are many additional tools in the repertoire of complementary medical practitioners
that go far beyond what is ingested, inhaled or spread on the skin to ward off
disease or treat illness.
The goal of this volume is not to consider the entirety of complementary
medicine, but to focus on food, herbals and their chemical constituents as con-
tributors to human health through control of metabolism, primarily as they relate
to chronic disease development and treatment. More specifically, we will consider
how what is consumed affects response, whether on a population or individual
level, to pharmacologic agents that are the mainstay of chronic disease treat-
ment/prevention around the world. Many of these drugs have their history as
natural compounds isolated from flora and fauna in our environment; however
the public perception has often been that “drugs” come with significant risk and
are less safe than “foods” or “natural health products.” Promulgating this view
are vocal members of the public who perceive a conspiracy by big business and
the pharmacologic giants to protect their billion-dollar stocks by presenting alter-
native medical practice as “flawed.” Furthermore, many complementary practi-
tioners themselves will say that they usually do not consider that adverse events
may occur in response to their therapies, nor do they ask their patients to report
any side effects,1 which exacerbates the problem.
Regardless of the particular approach that is taken, patients must demand of
their practitioners that applied therapies be safe and effective. Thus, evidence-based
medical practice must continue to be the cornerstone of health care delivery for the
foreseeable future. While testing through phase I, II and III clinical trials following
extensive testing in model systems is the standard for determining the safety and
efficacy of pharmacologic agents and synthetic food additives, this has not been
the case for many “natural products.” Assumptions of safety and efficacy for natural
health products have often come from demonstration of historical use over many
years, or possibly centuries, in specific populations with specific lifestyle and
cultural practices. This limited scope of use can hardly be expected to predict the
complicated behaviors that could occur in individuals or populations following very
different lifestyles or using them in combination with other therapies.
DK5836_C000.fm Page vi Monday, May 22, 2006 2:39 PM
There have been incredible advances in our understanding of how the chem-
icals in foods and herbs we consume interact with natural and synthetic drugs
used to prevent or treat human disease. It is standard practice to examine the
effects of food consumption on the absorption and pharmacokinetics of new
drugs, but the relevant issues have become much greater than “should this med-
icine be taken with or without food.” Long-term use of many medications has
the potential to modify gastrointestinal function and alter the uptake and handling
of both macronutrients, micronutrients and many phytochemicals that we now
know have impacts on nutritional status and overall health. Manipulating the diet
or the administration of supplements, orally or systemically, can help prevent
nutritional deficiencies that could develop during chronic drug administration. A
much more difficult task is to understand the potential for the thousands of
chemical compounds found in food and natural health products to modify drug
bioavailability, biodistribution, toxicity, efficacy and metabolism. In some cases
a single food or supplement could profoundly increase or decrease the toxicity
and/or efficacy of a single drug.
A major emphasis, from a clinical point of view, has been to identify these
substances and remove them from the diets or supplement list of patients under-
going drug therapy. The focus, then, has been almost exclusively on simplifying
the drug treatment plan. An alternative approach would be to use our knowledge
of the mechanisms by which food chemicals modify drug action and to develop
adjuvant therapy protocols that use food (functional foods and nutraceuticals)
and drugs together to optimize treatment outcome. This could result in a decreased
requirement, by the patient, for expensive medications, possibly increased efficacy
with decreased toxicity toward normal or uninfected tissues, and the maintenance
of a food substance with multiple healthful components.
In this collection of reviews, these major issues will be addressed by experts
in their relevant disciplines. The organization of the book will place the focus on
the ailment being treated or prevented and, thus, on the targets of therapy. Within
each section a comprehensive examination of macronutrient, micronutrient and
phytochemical impacts on drug action will be undertaken. Where a given drug
affects nutritional status, the appropriate diet modification or supplement will be
suggested. The major focus of each section will be on the molecular mechanism(s)
by which the food or chemical is thought to modify disease process and drug
behavior. Where specific active chemicals have been identified, their precise
molecular targets, including regulation of gene expression, will be described. The
book will finish with a description of the roles of genetic variation and polymor-
phism in determining nutrient/drug responses, and how individuals might be
“profiled” in future to identify those likely to demonstrate specific interactions
and who would benefit most from adjuvant or complementary therapies.
REFERENCE
1. Abbott, A., Survey questions safety of alternative medicine. Nature, 436 (7053):
898, 2005.
DK5836_C000.fm Page viii Monday, May 22, 2006 2:39 PM
DK5836_C000.fm Page ix Monday, May 22, 2006 2:39 PM
The Editor
Kelly Anne Meckling, Ph.D., completed an Honors B.Sc. with Distinction in
Biochemistry with a minor in psychology from the University of Calgary in 1984,
and her M.Sc. with Dr. Tony Pawson from the University of British Columbia.
She then transferred to the University of Toronto to complete her Ph.D. in medical
biophysics in 1989, examining the signaling pathways unique to human leukemia
and normal blood cell development.
Dr. Meckling’s travels took her to the University of Alberta in Edmonton in
1989, where she examined the role of growth factors and cancer development on
transport of chemotherapeutic drugs. In 1991, she became a faculty member in
the Department of Nutritional Sciences at the University of Guelph, with no
formal nutrition training. She rapidly acquired this knowledge through reading,
attending undergraduate and graduate lectures and through her teaching activities.
Since that time, she has continued as an active researcher in the area of nutrient
signaling and the prevention/treatment of chronic diseases, including cancer,
cardiovascular disease and obesity.
Dr. Meckling currently is associated with the Natural Sciences Engineering
Research Council of Canada and the Breast Cancer Society, where she carries
out both in vitro studies in model systems and human clinical intervention trials.
She teaches undergraduate biology to 2000 students per year as well as advanced
courses in nutrition and the metabolic control of disease. Also, Dr. Meckling is
academic advisor to students majoring in nutritional and nutraceutical sciences.
DK5836_C000.fm Page x Monday, May 22, 2006 2:39 PM
DK5836_C000.fm Page xi Monday, May 22, 2006 2:39 PM
Contributors
Marica Bakovic James B. Kirkland
Department of Human Health and Human Health and Nutritional
Nutritional Sciences Sciences
University of Guelph University of Guelph
Guelph, Ontario, Canada Guelph, Ontario, Canada
Amir Baluch
Department of Anesthesiology Alexandre Loktionov
Louisiana State University Colonix Ltd.
New Orleans, Louisiana Babraham Research Campus
Cambridge, United Kingdom
Matthew Chronowic
Department of Human Health and
Kelly Anne Meckling
Nutritional Sciences
Human Health and Nutritional
University of Guelph
Sciences
Guelph, Ontario, Canada
University of Guelph
Guelph, Ontario, Canada
Ennio Esposito
Istituto di Ricerche Farmacologiche
Mario Negri André J. Scheen
Santa Maria Imbaro, Italy Division of Diabetes, Nutrition and
Metabolic Disorders
Jason M. Hoover Department of Medicine
Department of Anesthesiology Sart Tilman Hospital
Louisiana State University Liège, Belgium
New Orleans, Louisiana
Contents
Chapter 1
Diabetes, Obesity, and Metabolic Syndrome .......................................................1
André J. Scheen
Chapter 2
Hypolipidemic Therapy: Drugs, Diet, and Interactive Effects ..........................31
Alexandre Loktionov
Chapter 3
Phytochemicals, Xenobiotic Metabolism, and Carcinogenesis..........................63
James B. Kirkland
Chapter 4
Nutrient and Phytochemical Modulation of Cancer Treatment .........................95
Kelly Anne Meckling
Chapter 5
Role of Nutritional Antioxidants in the Prevention and Treatment of
Neurodegenerative Disorders............................................................................129
Ennio Esposito
Chapter 6
Nutrients and Herbals in the Pharmacotherapy of
Unipolar/Major Depression...............................................................................179
Matthew Chronowic
Chapter 7
Supplements and Anesthesiology .....................................................................209
Alan D. Kaye, Amir Baluch, and Jason M. Hoover
Chapter 8
Nutrient- and Drug-Responsive Polymorphic Genes as Nutrigenomic
Tools for Prevention of Cardiovascular Disease and Cancer...........................237
Kevin Wood and Marica Bakovic
DK5836_C000.fm Page xiv Monday, May 22, 2006 2:39 PM
Chapter 9
Nutrigenomics and Pharmacogenomics of Human Cancer:
On the Road from Nutrigenetics and Pharmacogenetics .................................261
Alexandre Loktionov
Index..................................................................................................................319
DK5836_C001.fm Page 1 Monday, May 22, 2006 11:50 AM
CONTENTS
1.1 Introduction..................................................................................................1
1.2 Diet Intervention..........................................................................................3
1.2.1 Healthy Diet.....................................................................................3
1.2.2 Special Foods and Nutrients ...........................................................4
1.2.2.1 Fibers .................................................................................4
1.2.2.2 Dietary Supplements and Herbal Products.......................5
1.2.2.3 Grapefruit Juice.................................................................5
1.3 Pharmacological Intervention......................................................................6
1.3.1 Glucose-Lowering Agents ...............................................................6
1.3.1.1 Sulfonylureas.....................................................................6
1.3.1.2 Metformin..........................................................................8
1.3.1.3 Thiazolidinediones (TZDs) ...............................................9
1.3.1.4 α-Glucosidase Inhibitors.................................................10
1.3.1.5 Meglitinide Derivatives ...................................................12
1.3.2 Antiobesity Agents ........................................................................13
1.3.2.1 Orlistat .............................................................................13
1.3.2.2 Sibutramine .....................................................................15
1.3.3 Lipid-Lowering Agents .................................................................16
1.3.3.1 Statins ..............................................................................16
1.3.3.2 Fibrates ............................................................................17
1.3.3.3 Resins ..............................................................................18
1.3.3.4 Ezetimibe.........................................................................18
1.3.3.5 Sterols and Stanols..........................................................19
1.4 Conclusions................................................................................................20
References ...........................................................................................................20
1.1 INTRODUCTION
Metabolic disorders, such as diabetes mellitus, overweight/obesity, and the so-
called metabolic syndrome (including atherogenic dyslipidemia) are rapidly
1
DK5836_C001.fm Page 2 Monday, May 22, 2006 11:50 AM
2 Nutrient–Drug Interactions
increasing in the westernized world because of poor lifestyle habits favoring fat-
and sucrose-enriched meals and low physical activity or sedentariness.1 Medical
nutrition therapy is an integral component of diabetes mellitus,2 obesity3 and
metabolic syndrome management.4 Therapy should be individualized, with con-
sideration given to the individual’s usual food and eating habits, metabolic profile,
treatment goals, and desired outcomes. When diet, associated with regular phys-
ical exercise and healthy lifestyle habits, is insufficient to control blood glucose,
reduce body weight and/or improve the metabolic profile, pharmacological inter-
ventions may be considered to reach these objectives.5 While insulin therapy is
essential in all patients with type 1 diabetes, drug therapy is now a key element
in the management of type 2 diabetes mellitus and is becoming more and more
sophisticated with the recent launch of new compounds and the increased use of
combined therapy.6,7 In contrast, pharmacological intervention for obesity still
remains a controversial issue because of only modest long-term efficacy and/or
concern about safety.8,9 Finally, the recent recognition of the important health
problem associated with the metabolic syndrome10 has stimulated researchers to
develop drugs aimed at specifically improving insulin sensitivity and correcting
associated metabolic disorders.4,11
Because nutrition therapy and pharmacological intervention are major com-
ponents of the management of metabolic disorders, it is interesting to consider
potential food–drug or nutrient–drug interactions in this particular medical field
(Figure 1.1). An interaction is said to take place when the effects of one drug are
changed by the presence of another drug, food, drink or by some environmental
chemical agent.12 Interactions between food and drugs may inadvertently reduce
or increase the effect of the drug, resulting in therapeutic failure (i.e., hypergly-
cemia in the case of diabetes mellitus) or increased toxicity (i.e., hypoglycemia
in the case of diabetes). The majority of clinically relevant food–drug interactions
are caused by food-induced changes in the bioavailability of the drug.13,14 Indeed,
since the bioavailability and clinical effect of most drugs are correlated, the
bioavailability is an important pharmacokinetic effect parameter. However, in
order to evaluate the clinical relevance of a food–drug interaction, the impact of
food intake on the clinical effect of the drug has to be quantified as well.15 This
may be particularly relevant as far as food-related metabolic diseases are con-
cerned. Thus, in the field of metabolic disorders, where nutrition plays a major
role in the overall treatment, the potential influence of food and nutrient intake
on drug therapeutic effect may be crucial.
Pharmacokinetic interferences often occur as a result of a change in drug
metabolism. Cytochrome P450 system oxidizes a broad spectrum of drugs by a
number of metabolic processes that can be enhanced or reduced by various drugs
(known as inducers or inhibitors, respectively).16 The clinical importance of any
drug interaction depends on factors that are drug-, patient- and administration-
related. As reviewed extensively,17 dietary changes can alter the expression and
activity of hepatic drug metabolizing enzymes, and dietary components may
influence the gastrointestinal metabolism and transports of drugs.18 Athough this
can lead to alterations in the systemic elimination kinetics of drugs metabolized
DK5836_C001.fm Page 3 Monday, May 22, 2006 11:50 AM
TYPE 2 DIABETES
DIGESTIVE ORAL
TRACT BIOAVAILABILITY
F D
O CYP HEPATIC
INTERFERENCES METABOLISM
R
O U
D G
NUTRIENT PHARMACO-
EFFECTS DYNAMICS
OBESITY
METABOLIC SYNDROME
by these enzymes, the magnitude of the change is generally small,13,14 with the
exception of the potentially major influence of grapefruit juice.18 The coadmin-
istration of certain drugs with grapefruit juice can markedly elevate drug bio-
availability and can alter both pharmacokinetic and pharmacodynamic parameters
of the drug.19–21
4 Nutrient–Drug Interactions
principles also concern diet recommendations for the treatment and prevention
of diabetes mellitus and related disorders,2,22 and of overweight and obesity.3
A number of factors influence glycemic responses to foods, including the
amount of carbohydrates, type of sugar, nature of the starch, cooking and food
processing and food form, as well as other food components. Although it is clear
that carbohydrates do have differing glycemic responses, the data reveal no clear
trend in outcome benefits. If there are long-term effects on glycemia and lipids,
these effects appear to be modest.22
In persons with type 2 diabetes (on weight-maintenance diets), replacing
carbohydrates with monosaturated fats reduces postprandial glycemia and tri-
glyceridemia. However, there is concern that increased fat intake in ad libitum
diets may promote weight gain. Therefore, the contributions of carbohydrates
and monosaturated fat to energy intake should be individualized based on nutrition
assessment, metabolic profiles and treatment goals.2,22
Total energy intake remains the most important factor in dietary interventions
in obese individuals, but strategies based on emerging factors, such as glycemic
index, fiber intake, dietary variety and energy density, may improve efficacy and
tolerability of energy restriction.3
diseases, such as obesity, diabetes mellitus and metabolic syndrome, the effects
of fiber content on availability of drugs commonly used for treating such patients
have not been specifically investigated. One trial showed that guar gum decreases
the absorption of metformin in healthy subjects.26
Pharmaceutical research conducted over the past 3 decades shows that natural
products are a potential source of molecules for drug development, including for
diabetes treatment.27 Moreover, the use of botanical products has increased in
recent years. Herbal products are widely used as dietary supplements and are part
of complementary or alternative medicine. Importantly, herbal products are often
used concurrently with prescribed or over-the-counter medications. The use of
herbal products often escapes standard mechanisms for protecting persons from
harmful effects of drugs or drug interactions.28,29 Concurrent use of botanicals
with approved prescription drug products can result in therapeutic failures or
adverse events.30 Interactions of herbs with cytochrome P450 have been exten-
sively described.31 For instance, recent publications have shown that coadminis-
tration of St. John’s wort (Hypericum perforatum) decreases plasma levels of
several concomitant drugs (cyclosporin, indinavir, etc.), and the underlying mech-
anism appears to be induction of the intestinal CYP3A and P-glycoprotein.29,32
The potential effects of herbal products on drugs classically used in patients
with diabetes, obesity or metabolic syndrome are largely unknown. One Japanese
study reported that St. John’s wort treatment (300 mg tid [3 times per day] for
14 days) significantly reduced simvastatin area under the curve (AUC) by 50%,
but did not interfere with pravastatin pharmacokinetics.33 Thus, the potential for
clinically significant drug interactions between St. John’s wort with many
CYP3A4 substrates is a real possibility, one that may not be evident from routine
“screening” for traditional drug–drug interactions.29 The potent effects of herbal
products coupled with their risk of interactions with other commonly used drugs
and the lack of governmental regulation have created a public health dilemma
and encouraged the publication of a conference report.
6 Nutrient–Drug Interactions
1.3.1.1 Sulfonylureas
Sulfonylureas were the first oral drugs for treatment of type 2 diabetes in clinical
practice.40,41 They act at the pancreatic β-cell membrane by causing closure of
adenosine triphosphate (ATP)-sensitive potassium channels, which leads to an
enhanced insulin secretion. They are commonly prescribed alone or in combina-
tion with metformin, a thiazolidinedione, or an α-glucosidase inhibitor to reach
the glucose control targets.42 Various sulfonylureas have been, or are currently,
used in clinical practice. Those with the largest experience are tolbutamide and
chlorpropamide (first generation), and glibenclamide, glipizide, gliclazide and
glimepiride (second generation).43
The kinetic-dynamic relations of sulfonylureas are complex. The timing of
food intake, rather than the composition of diet, has been particularly analyzed
with the different sulfonylurea compounds. The objective is to accelerate sulfo-
nylurea gut absorption (leading to short tmax and high Cmax), to maximize meal-
related, early (first phase) insulin secretion and to reduce postprandial hypergly-
cemia.44 The rate of absorption of chlorpropamide or tolbutamide, but not its
extent, is decreased by food.45 Even if concomitant food intake may not delay
DK5836_C001.fm Page 7 Monday, May 22, 2006 11:50 AM
the absorption of tolbutamide, its efficacy may be improved if the drug is given
half an hour before meals rather than with meals.46 Chlorpropamide is more
slowly absorbed than tolbutamide and has the longest elimination half-life of all
sulfonylureas. Hence, the timing of the daily dose is less important, and it is
irrelevant whether chlorpropamide is ingested before or with breakfast.43 Prior
food intake has no influence on rate and extent of absorption of the classical form
of glibenclamide.47 Although concomitant food intake does not seem to delay the
absorption of glibenclamide, its efficacy may be increased if given before meals,
at least in the short term. Indeed, 2.5 mg given half an hour before breakfast was
more effective than 7.5 mg given together with breakfast.48
Food significantly delays absorption of gliclazide and peak concentrations
also may be reduced;49 however, it is not known whether intake before meals
makes gliclazide more effective than does intake together with meals.43 The
absorption of glipizide is also retarded by food,50,51 and is more rapidly absorbed
when taken before breakfast than when ingested together with breakfast. Intake
before breakfast is also associated with a more appropriate timing of insulin
release relative to meal, and with enhanced efficacy of the drug.51 Food delays
the absorption of a tablet of gliquidone only insignificantly.52 Glimepiride has an
absolute bioavailability of 100%,53 and there were no significant differences in
pharmacokinetic and pharmacodynamic responses between glimepiride given 30
min before or immediately after a meal in healthy subjects54 or between glime-
piride given 30 min or directly before breakfast in patients with type 2 diabetes.55
In clinical practice, glimepiride is usually prescribed once daily in the morn-
ing.56,57
Gliclazide is a weak acid with a good lipophilicity and a pH-dependent
solubility.58 Gliclazide is practically insoluble in acidic media and its solubility
increases as the pH becomes more alkaline. The absorption rate depends upon
the gastric emptying time and upon the dissolution rate in the small intestine
where the compound is soluble. The variability in absorption of gliclazide could
also be related to an early dissolution in the stomach leading to more variability
in the absorption in the intestine. From this, it can be anticipated that both delay
in gastric emptying and change in pH accompanying food intake might affect the
dissolution of the compound in the gastrointestinal tract. In two studies, food
altered the pharmacokinetics of gliclazide in type 2 diabetic patients.49,59 Two
mechanisms were thought to be involved: (1) delay in the absorption of gliclazide
when given 30 min after breakfast49 and (2) decrease in the extent of absorption
of gliclazide when given 30 min before a meal.59 Some decrease in gastric
emptying is noted; a possible interaction of gliclazide with dietary components,
as well as a change in pH, could explain these observations.
A new modified release (MR) formulation containing 30 mg of gliclazide
was developed to obtain a better predictable release of the active principle and
to allow once-daily dosing regimen at breakfast.60 In a food-effect study in healthy
volunteers, one tablet was given either fasted or 10 min after the start of a
standardized breakfast.61 No significant difference was observed in tmax, t1/2, Cmax
and AUC of gliclazide after administration of the 30 mg MR tablet under fasted
DK5836_C001.fm Page 8 Monday, May 22, 2006 11:50 AM
8 Nutrient–Drug Interactions
and fed conditions. Thus, the new MR formulation of gliclazide can be given
without regard to meals, i.e., before, during or after breakfast.
Chlorpropamide is less commonly used since the launch of second-generation
sulfonylureas. One reason for this change results from a specific adverse effect
— the chlorpropamide–alcohol flush.62 Indeed, facial flushing after alcohol occurs
much more frequently in type 2 diabetic patients taking chlorpropamide than in
those receiving other sulfonylureas. Both alcohol and chlorpropamide concentra-
tions are critical in determining the chlorpropamide–alcohol flush.63 The mech-
anism that produces the flush is poorly understood. It has been suggested that
chlorpropamide acts as a noncompetitive inhibitor of aldehyde dehydrogenase
resulting in increased acetaldehyde concentrations. There is, however, uncertainty
whether the flush can be explained solely by a rise in acetaldehyde concentrations.
Prostaglandin inhibitors, such as aspirin, have been shown to attenuate the flush
in some patients. Thus, alcohol intake should be avoided in diabetic patients
treated with chlorpropamide.
1.3.1.2 Metformin
although this effect remains controversial.72 Vitamin B12 malabsorption was found
in 30% of diabetic patients during long-term metformin treatment.72,73 Although
it seems to be reversible, it may be persistent.74 Megalobastic anemia due to
vitamin B12 malabsorption associated with long-term metformin treatment has
been reported, but vitamin B12 treatment was successful in correcting such an
abnormality.75 The mechanism of biguanide-induced B12 malabsorption remains
uncertain, but it may be attributed to direct intestinal effects or bacterial over-
growth with binding of the intrinsic factor B12 complex.74
Alcohol potentiates the glucose-lowering and hyperlactatemic effect of bigu-
anides and should be limited in patients on chronic treatment with metformin.67
Alcohol itself decreases lactate oxidation and gluconeogenesis from lactate. The
inhibition of gluconeogenesis by alcohol also promotes hypoglycemia, especially
in the fasting state.
Diet–metformin interactions deserve further evaluation. In the Diabetes Pre-
vention Program,76 individuals with impaired glucose tolerance were randomized
to one of three intervention groups, which included the intensive nutrition and
exercise counseling (“lifestyle”) group or either of two masked medication treat-
ment groups — the biguanide metformin group or the placebo group. The latter
interventions were combined with standard diet and exercise recommendations.
After an average follow-up of 2.8 years, a 58% relative reduction in the progres-
sion to diabetes was observed in the lifestyle group, and a 31% relative reduction
in the progression to diabetes was observed in the metformin group, compared
with controlled subjects (placebo group). Considering these two remarkable pro-
tective effects provided by intensive lifestyle intervention on the one hand and
by metformin on the other hand, it would be interesting to investigate whether a
combination of the two interventions in the same at-risk population is able to
provide a further reduction in the incidence of type 2 diabetes.
TZDs are now widely used as part of antidiabetic treatments.77–79 These agents
act by targeting insulin resistance instead of stimulating insulin secretion. They
interact with the gamma type of the peroxisome proliferator-activated receptor
(PPAR-gamma).79,80 PPAR-gamma, a member of the nuclear receptor subfamily,
stimulates gene expression of proteins involved in glucose metabolism. This
results in an increase in insulin sensitivity in skeletal muscle, and adipose and
liver tissues. Two TZDs are currently commercialized and widely prescribed —
pioglitazone81,82 and rosiglitazone.83,84 Unlike sulfonylureas and metformin, TZDs
are not used commonly as first-line therapy. However, there is considerable
evidence for the incorporation of TZDs into combination regimens, with either
sulfonylureas or metformin, particularly in patients who do not achieve glycemic
goals with conventional regimens.85 Pharmacokinetic characteristics of both
pioglitazone86 and rosiglitazone87 have been described extensively. TZDS are
metabolized through the cytochrome P450 system and, thus, are subject to
pharmacokinetic interactions.88 While some drug–drug interactions have been
DK5836_C001.fm Page 10 Monday, May 22, 2006 11:50 AM
10 Nutrient–Drug Interactions
12 Nutrient–Drug Interactions
Significantly greater insulin secretion was observed when nateglinide was taken
before a meal compared to when given in the fasted state or in response to merely
a meal. When given before meals, nateglinide produced rapid and short-lived
insulin secretion, effectively reducing mealtime glucose excursions, yet lowering
the risk of hypoglycemic episodes.
When nateglinide was given to healthy subjects 10 min before ingestion of
a high-fat meal, the rate of absorption was increased relative to when it was given
during a continued fast, as evidenced by a 12% increase in Cmax and a 52%
decrease in tmax.117 However, there was no significant effect on the extent of
absorption as assessed by the AUC. Alternatively, when nateglinide was given
after the meal, a food effect was observed that was characterized by a decrease
in the rate of absorption: a 34% decrease in Cmax and a 22% increase in tmax, but
no significant effect on AUC. Regardless of timing, the combination of a meal
and nateglinide produced a larger increase in plasma insulin levels than did
nateglinide alone.
The influence of timing of administration of nateglinide on the glucose profile
and β-cell secretory response to a standardized test meal was studied, as well as
the effect of meal composition on the pharmacokinetic and pharmacodynamic
profile.118 Nateglinide administration 10 min before a meal resulted in a more
rapid rise and a higher peak nateglinide plasma concentration than when it was
given 10 min after the start of the meal, irrespective of the meal composition
(i.e., high in carbohydrates, fat or protein). Preprandial administration of nateg-
linide was more effective in reducing prandial glucose excursions, compared with
post-meal dosing, a consequence of the earlier insulin response. Meal composition
had no effect on the plasma nateglinide profile, which is of particular importance
because nateglinide is a D-phenylalanine derivative and, thus, there was the
possibility that its absorption could be influenced by the presence of high amino
acid concentrations. The findings of this study clearly demonstrated that, even in
the presence of a high-protein meal (32% of calories derived from protein), no
effect on absorption of nateglinide was observed.
1.3.2.1 Orlistat
14 Nutrient–Drug Interactions
1.3.2.2 Sibutramine
16 Nutrient–Drug Interactions
1.3.3.1 Statins
1.3.3.2 Fibrates
The two most commonly used fibric derivatives are gemfibrozil in the U.S.161,162
and fenofibrate163–165 in Europe. Fibrates are metabolized by the hepatic cyto-
chrome P450 (CYP) 3A4 and, thus, are subject to drug interactions.166
DK5836_C001.fm Page 18 Monday, May 22, 2006 11:50 AM
18 Nutrient–Drug Interactions
1.3.3.3 Resins
Resins (cholestyramine and colestipol) act by binding bile salts in the gastrointes-
tinal lumen.139 Both compounds are large polymers that are not absorbed system-
ically. They bind bile acids by exchanging them for chloride ions; binding is
irreversible. The bound bile salts are not reabsorbed, which leads to a depletion
of the bile acid pool. The liver compensates by increasing synthesis of bile acids,
which results in intracellular cholesterol depletion leading to increased expression
of LDL receptors on the hepatocyte cell membrane and, thereby, increased LDL
clearance. These drugs are given in the form of a powder, which is usually taken
with meals as a suspension in juice or water. Compliance is the major problem
on the grounds of palatability and adverse gastrointestinal effects. The absorption
of several drugs may be impaired by resins, including statins, digoxin, amio-
darone, thyroxine, warfarin, thiazide diuretics, and beta blockers. Reduced
absorption of folic acid and fat-soluble vitamins may occur, but clinical deficiency
is rare. Resins are less commonly used since the expansion of statin prescription
and the recent launch of ezetimibe, a selective inhibitor of cholesterol intestinal
absorption.
1.3.3.4 Ezetimibe
20 Nutrient–Drug Interactions
1.4 CONCLUSIONS
Metabolic disorders generally require combined dietary and pharmacological
interventions. While food–drug or nutrient–drug interactions might be of partic-
ular importance in patients with metabolic diseases, only scarce data are available
in the scientific literature, as compared to the overwhelming information regarding
drug–drug interactions. Obviously, food composition may influence the efficacy
of drugs affecting glucose, lipid and energy metabolism. In addition, since meal-
related kinetics is crucial in controlling postprandial blood glucose in diabetic
patents, the influence of food on drug bioavailability may also be important,
especially for drugs that stimulate insulin secretion. Furthermore, meal content
and/or timing may also influence drug gastrointestinal tolerability, as described
with metformin, α-glucosidase inhibitors and orlistat. Finally, as food may inter-
fere with CYP P450, potential food–drug interactions may be suspected in using
pharmacological agents metabolized via the cytochrome system. A clear interac-
tion with grapefruit juice was described with statins metabolized via CYP3A4,
especially lovastatin and simvastatin. From a public health point of view, an
interaction between drug and food or drink might reveal more difficulties than a
drug–drug interaction. It is important that the awareness for this potential
food–drug interaction will increase, and actions must be taken in order to increase
the efficacy of diet–drug combination in metabolic diseases and to prevent
undesired and harmful clinical consequences.
REFERENCES
1. Zimmet P. Globalization, coca-colonization and the chronic disease epidemic: can
the doomsday scenario be averted? J Intern Med 2002; 247: 301–310.
2. Franz MJ, Bantle JP, Beebe CA, Brunzell JD, Chiasson J-L, Garg A, Holzmeister
LA, Hoogwerf B, Mayer-Davis E, Mooradian AD, Purnell JQ, Wheeler M. Evi-
dence-based nutrition principles and recommendations for the treatment and pre-
vention of diabetes and related complications (Technical review). Diabetes Care
2002; 25: 148–198.
3. Hajduk CL, Roberts SB, Saltzman E. Dietary treatment of obesity. Curr Opin
Endocrinol Diabetes 2001; 8: 240–246.
4. Scheen AJ. Management of the metabolic syndrome. Minerva Endocrinol 2004;
29: 31–45.
5. Scheen AJ. Current management of coexisting obesity and type 2 diabetes. Drugs
2003; 63: 1165–1184.
6. Scheen AJ, Lefèbvre PJ. Oral antidiabetic agents: a guide to selection. Drugs
1998; 55: 225–236.
7. Inzucchi SE. Oral antihyperglycemic therapy for type 2 diabetes: scientific review.
JAMA 2002; 287: 360–372.
DK5836_C001.fm Page 21 Monday, May 22, 2006 11:50 AM
8. Scheen AJ, Lefèbvre PJ. Pharmacological treatment of obesity: present status. Int
J Obes 1999; 23 (Suppl 1): 47–53.
9. Fernandez-Lopez J-A, Remesar X, Foz M, Alemany M. Pharmacological
approaches for the treatment of obesity. Drugs 2002; 62: 915–944.
10. Grundy SM, Brewer HB Jr, Cleeman JI, Smith SC, Lenfant C, for Conference
Participants. Definition of metabolic syndrome: report of the National Heart, Lung,
and Blood Institute/American Heart Association conference on scientific issues
related to definition. Circulation 2004; 109: 433–438.
11. Grundy SM, Hansen B, Smith SC, Cleeman JI, Kahn RA, for Conference Partic-
ipants. Clinical management of metabolic syndrome. Report of the American Heart
Association/National Heart, Lung, and Blood Institute/American Diabetes Asso-
ciation Conference on scientific issues related to management. Circulation 2004;
109: 551–556.
12. Sorensen JM. Herb–drug, food–drug, nutrient–drug, and drug–drug interactions:
mechanisms involved and their medical implications. J Altern Complement Med
2002; 8: 293–308.
13. Anderson KE. Influences of diet and nutrition on clinical pharmacokinetics. Clin
Pharmacokinet 1988; 14: 325–346.
14. Walter-Sack I, Klotz U. Influence of diet and nutritional status on drug metabolism.
Clin Pharmacokinet 1996; 31: 47–64.
15. Schmidt LE, Dalhoff K. Food–drug interactions. Drugs 2002; 62: 1481–1502.
16. Dresser GK, Spence JD, Bailey DG. Pharmacokinetic–pharmacodynamic conse-
quences and clinical relevance of cytochrome P450 3A4 inhibition. Clin Pharma-
cokinet 2000; 38: 41–57.
17. Ioannides C. Effect of diet and nutrition on the expression of cytochromes P450.
Xenobiotica 1999; 29: 109–154.
18. Evans AM. Influence of dietary components on the gastrointestinal metabolism
and transport of drugs. Ther Drug Monit 2000; 22: 131–136.
19. Bailey DG, Malcolm J, Arnold O, Spence JD. Grapefruit juice–drug interactions.
Br J Clin Pharmacol 1998; 46: 101–110.
20. Fuhr U. Drugs interactions with grapefruit juice. Extent, probable mechanism and
clinical relevance. Drug Saf 1998; 18: 251–272.
21. Dahan A, Altman H. Food-drug interaction: grapefruit juice augments drug bio-
availability — mechanism, extent and relevance. Eur J Clin Nutr 2004; 58: 1–9.
22. American Diabetes Association. Nutrition principles and recommendations in
diabetes. Diabetes Care 2004; 27 (Suppl 1): S36–46.
23. Krauss RM, Eckel RH, Howard B, Appel LJ, Daniels SR, Deckelbaum RJ, Erdman
JW Jr, Kris-Etherton P, Goldberg IJ, Kotchen TA, Lichtenstein AH, Mitch WE,
Mullis R, Robinson K, Wylie-Rosett J, St Jeor S, Suttie J, Tribble DL, Bazzarre
TL. AHA Dietary Guidelines: Revision 2000: a statement for healthcare profes-
sionals from the Nutrition Committee of the American Heart Association. Circu-
lation 2000; 102: 2284–2299
24. Anderson JA, Midgley WR, Wedman B. Fiber and diabetes. Diabetes Care 1979;
2: 369–379.
25. Howard NC, Saltzman E, Roberts SB. Dietary fiber and weight regulation. Nutr
Rev 2001; 59: 129–139.
26. Gin H, Orgerie MB, Aubertin J. The influence of guar gum on absorption of
metformin from the gut in healthy volunteers. Horm Metab Res 1989; 21: 81–83.
DK5836_C001.fm Page 22 Monday, May 22, 2006 11:50 AM
22 Nutrient–Drug Interactions
27. Oubre AY, Carlson TJ, King SR, Reaven GM. From plant to patient: an ethno-
medical approach to the identification of new drugs for the treatment of NIDDM.
Diabetologia 1997; 40: 614–617.
28. Miller L. Herbal medicinals: selected clinical considerations focusing on known
or potential drug–herb interactions. Arch Intern Med 1998; 158: 2200–2211.
29. Huang S-M, Hall SD, Watkins P, Love LA, Serabjit-Singh C, Betz JM, Hoffman
FA, Honig P, Coates PM, Bull J, Chen ST, Kearns GL, Murray MD. Drug
interactions with herbal products and grapefruit juice: A conference report. Clin
Pharmacol Ther 2004; 75: 1–12.
30. Zhou S, Koh HL, Gao ZY, Lee EJ. Herbal bioactivation: the good, the bad and
the ugly. Life Sci 2004; 74: 935–968.
31. Zhou S, Gao Y, Jiang W, Huang M, Xu A, Paxton JW. Interactions of herbs with
cytochrome P450. Drug Metab Rev 2003; 35: 35–98.
32. Wang Z, Gorski JC, Hamman MA, Huang SM, Lesko LJ, Hall SD. The effect of
St John’s wort (Hypericum perforatum) on human cytochrome P450 activity. Clin
Pharmacol Ther 2001; 70: 317–326.
33. Sugimoto K, Ohmori M, Tsuruoka S, Nishiki K, Kawaguchi A, Harada K,
Arakawa M, Sakamoto K, Masada M, Miyamori I, Fujimura A. Different effects
of St John’s wort on the pharmacokinetics of simvastatin and pravastatin. Clin
Pharmacol Ther 2001; 70: 518–524.
34. Spence JD. Drug interactions with grapefruit: whose responsibility is it to warn
the public? Clin Pharmacol Ther 1997; 61: 395–400.
35. Kuhlmann J, Puls W. Oral antidiabetics. Handbook of Experimental Pharmacol-
ogy. Vol. 119. Berlin: Springer, 1996.
36. Scheen AJ. Drug treatment of non-insulin-dependent diabetes mellitus in the
1990s: achievements and future developments. Drugs 1997; 54: 355–368.
37. Brian WR. Hypoglycemic agents. In Levy RH, Thummel KE, Trager WF, Hansten
PD, Eichelbaum M, Eds. Metabolic Drug Interactions. Philadelphia: Lippincott
Williams & Wilkins, 2000: 529–543.
38. Scheen AJ, Lefèbvre PJ. Antihyperglycaemic agents. Drug interactions of clinical
importance. Drug Safety 1995; 12: 32–45.
39. Scheen AJ. Drug interactions of clinical importance with antihyperglycaemic
agents: an update. Drug Safety 2005; 28: 601–631.
40. Groop LC. Sulfonylureas and NIDDM. Diabetes Care 1992; 15: 737–754.
41. Lebovitz HE, Melander A. Sulfonylureas: basic aspects and clinical uses. In Alberti
KGMM, Zimmet P, DeFronzo RA, Keen H, Eds. International Textbook of Dia-
betes Mellitus. 2nd ed. Chichester, John Wiley & Sons, 1997: 817–840.
42. Rendell M. The role of sulphonylureas in the management of type 2 diabetes
mellitus. Drugs 2004; 64: 1339–1358.
43. Groop L, Neugebauer G. Clinical pharmacology of sulfonylureas. In Kuhlmann
J, Puls W, Eds. Oral antidiabetics. Handbook of Experimental Pharmacology. Vol.
119. Berlin, Springer, 1996: 199–259.
44. Del Prato S, Tiengo A. The importance of first-phase insulin secretion: implica-
tions for the therapy of type 2 diabetes mellitus. Diabetes Metab Res Rev 2001;
17: 164–174.
45. Sartor G, Melander A, Scherstén B, Wahlin-Boll E. Influence of food and age on
the single-dose kinetics and effects of tolbutamide and chlorpropamide. Eur J
Clin Pharmacol 1980; 17: 285–293.
DK5836_C001.fm Page 23 Monday, May 22, 2006 11:50 AM
46. Silins RA, Butcher MA, Marlin GE. Improved effect of tolbutamide when given
before food in patients on long-term therapy (letter). Br J Clin Pharmacol 1984;
18: 647–648.
47. Sartor G, Melander A, Scherstén B, Wahlin-Boll E. Serum glibenclamide in
diabetic patients and influence of food on the kinetics and effects of glibenclamide.
Diabetologia 1980; 18: 17–22.
48. Sartor G, Lundquist I, Melander A, Schersten B, Wahlin-Boll E. Improved effect
of glibenclamide on administration before breakfast. Eur J Clin Pharmacol 1982;
21: 403–408.
49. Batch J, Ma A, Bird D, Noble R, Charles B, Ravenscroft P, Cameron D. The
effects of ingestion time of gliclazide in relationship to meals on plasma glucose,
insulin and C-peptide levels. Eur J Clin Pharmacol 1990; 38: 465–467.
50. Sartor G, Melander A, Scherstén B, Wahlin-Boll E. Comparative single-dose
kinetics and effects of four sulfonylureas in healthy volunteers. Acta Med Scand
1980; 208: 301–307.
51. Wahlin-Boll E, Melander A, Sartor G, Scherstén B. Influence of food intake on
the absorption and effect of glipizide in diabetics and healthy subjects. Eur J Clin
Pharmacol 1980; 18: 279–283.
52. Talaulicar M, Willms B. Investigations on Glurenorm blood levels. Diab Croat
1976; 5: 613–622.
53. Rosskamp R, Wernicke-Panten K, Draeger E. Clinical profile of the novel sulpho-
nylurea glimepiride. Diab Res Clin Pract 1996; 31 Suppl: S33–S42.
54. Malerczyk V, Badian M, Korn A, Lehr K-H, Waldhäusl W. Dose linearity assess-
ment of glimepriride (Amaryl) tablets in healthy volunteers. Drug Metab Drug
Interactions 1994; 11: 341–357.
55. Rosskamp R, Herz M. Effect of the time of ingestion of the sulfonylurea glime-
piride on the daily blood glucose profile in NIDDM-patients (abstract). Proceed-
ings of the 15th International Federation Congress, Kobe, Japan, 1994, No.
10A5PP1050.
56. Langtry HD, Balfour JA. Glimepiride: a review of its use in the management of
type 2 diabetes mellitus. Drugs 1998; 55: 563-584.
57. McCall AL. Clinical review of glimepiride. Expert Opin Pharmacother 2001; 2:
699-713.
58. Palmer KJ, Brodgen RN. Gliclazide, an update on its pharmacological properties
and therapeutic efficacy in non-insulin-dependent diabetes mellitus. Drugs 1993;
46: 92–125.
59. Ishibashi F, Takashina S. The effect of timing on gliclazide absorption and action.
Hiroshima J Med Sci 1990; 39: 7–9.
60. Mc Gavin JK, Perry CM, Goa KL. Gliclazide modified release. Drugs 2002; 62:
1357–1364.
61. Delrat P, Paraire M, Jochemsen R. Complete bioavailability and lack of food effect
on pharmacokinetics of gliclazide 30 mg modified release in healthy volunteers.
Biopharm Drug Dispos 2002; 23: 151–157.
62. Fitzgerald MG, Gaddie R, Malins JM, O’Sullivan DJ. Alcohol sensitivity in
diabetics receiving chlorpropamide. Diabetes 1962; 11: 40–43.
63. Groop L, Eriksson CJP, Huupponen R, Ylikahri R, Pelkonen R. Roles of chlor-
propamide, alcohol and acetaldehyde in determining the chlorpropamide-alcohol
flush. Diabetologia 1984; 26: 34–38.
DK5836_C001.fm Page 24 Monday, May 22, 2006 11:50 AM
24 Nutrient–Drug Interactions
64. Hermann LS. Metformin: a review of its pharmacological properties and thera-
peutic use. Diab Metab 1979; 5: 233–245.
65. Caspary WF. Biguanides and intestinal absorption function. Act Hep Gastr 1977;
24: 473–480.
66. Dunn CJ, Peters DH. Metformin A review of its pharmacological properties and
therapeutic use in non-insulin-dependent diabetes mellitus. Drugs 1995; 49:
721–749.
67. Cusi K, DeFronzo RA. Metformin: a review of its metabolic effects. Diabetes Rev
1998; 6: 89–131.
68. Setter SM, Iltz JL, Thams J, Campbell RK. Metformin hydrochloride in the
treatment of type 2 diabetes mellitus: a clinical review with a focus on dual therapy.
Clin Ther 2003; 25: 2991–3027.
69. Scheen AJ. Clinical pharmacokinetics of metformin. Clin Pharmacokin 1996; 30:
359–371.
70. Brookes LG, Sambol NC, Lin ET, Gee W, Benet LZ. Effect of dosage form, dose
and food on the pharmacokinetics of metformin (abstract). Pharm Res 1991; 8
(Suppl): 32.
71. Berchtold P, Dahlqvist A, Gustafson A, Asp NG. Effects of a biguanide (met-
formin) on vitamin B12 and folic acid absorption and intestinal enzyme activities.
Scand J Gastroenterol 1971; 6: 751–754.
72. Carpentier J-L, Bury J, Luyckx A, Lefèbvre P. Vitamin B12 and folic acid serum
levels in diabetics under various therapeutic regimens. Diab Metab 1976; 2:
187–190.
73. Tomkin GH, Hadden DR, Weaver JA, Montgomery DAD. Vitamin B12 status in
patients on long-term metformin therapy. Br Med J 1971; 2: 685–687.
74. Adams JF, Clark JS, Ireland JT, Kesson CM, Watson WS. Malabsorption of
vitamin B12 and intrinsic factor secretion during biguanide therapy. Diabetologia
1983; 24: 16–18.
75. Callaghan TS, Hadden DR, Tomkin GH. Megaloblastic anaemia due to vitamin
B12 malabsorption associated with long-term metformin treatment. Br Med J 1980;
280: 1214–1215.
76. Diabetes Prevention Program Research Group. Reduction in the incidence of type
2 diabetes with lifestyle intervention or metformin. N Engl J Med 2002; 346:
393–403.
77. Diamant M, Heine RJ. Thiazolidinediones in type 2 diabetes mellitus: current
clinical evidence. Drugs 2003; 63: 1373–1405.
78. Meriden T. Progress with thiazolidinediones in the management of type 2 diabetes
mellitus. Clin Ther 2004; 26: 177–190.
79. Yki-Järvinen H. Thiazolidinediones. N Engl J Med 2004; 351: 1106–1118.
80. Gurnell M, Savage DB, Chatterjee VKK, O’Rahilly S. The metabolic syndrome:
peroxisome proliferator-activated receptor γ and its therapeutic modulation. J Clin
Endocrinol Metab 2003; 88: 2412–2421
81. Gillies PS, Dunn CJ. Pioglitazone. Drugs 2000; 60: 333–343.
82. Chilcott J, Tappenden P, Jones ML, Wight JP. A systematic review of the clinical
effectiveness of pioglitazone in the treatment of type 2 diabetes mellitus. Clin
Ther 2001; 23: 1792–1823.
83. Balfour JA, Plosker GL. Rosiglitazone. Drugs 1999; 57: 921–930.
84. Wagstaff AJ, Goa KL. Rosiglitazone. A review of its use in the management of
type 2 diabetes mellitus. Drugs 2002; 62: 1805–1837.
DK5836_C001.fm Page 25 Monday, May 22, 2006 11:50 AM
26 Nutrient–Drug Interactions
103. Wolever TM, Chiasson JL, Josse RG, Hunt JA, Palmason C, Rodger NW, Ross
SA, Ryan EA, Tan MH. Small weight loss on long-term acarbose therapy with
no change in dietary pattern or nutrient intake of individuals with non-insulin-
dependent diabetes. Int J Obes 1997; 21: 756–763.
104. Van Gaal L, Nobels F, De Leeuw I. Effects of acarbose on carbohydrate metab-
olism, electrolytes, minerals and vitamins in fairly well-controlled non-insulin-
dependent diabetes mellitus. Z Gastroenterol 1991; 29: 642–644.
105. Dornhorst A. Insulinotropic meglitinide analogues. Lancet 2001; 358: 1709–1716.
106. Owens DR. Repaglinide — prandial glucose regulator: a new class of oral antidi-
abetic drug. Diabetic Med 1998; 15 Suppl. 4: S28–S36.
107. Cully CR, Jarvis B. Repaglinide: a review of its therapeutic use in type 2 diabetes
mellitus. Drugs 2001; 61: 1625–1660.
108. Dunn CJ, Faulds D. Nateglinide. Drugs 2000; 60: 607–615.
109. Norman P, Rabasseda X. Nateglinide: a structurally novel insulin secretion agent.
Drugs of Today 2001; 37 Suppl. F: 1–16.
110. Hatorp V. Clinical pharmacokinetics and pharmacodynamics of repaglinide. Clin
Pharmacokinet 2002; 41: 471–483.
111. Bidstrup TB, Bjornsdottir I, Sidelman UG, Thomsen MS, Hansen KT. CYP2C8
and CYP3A4 are the principal enzymes involved in the human in vitro biotrans-
formation of the insulin secretagogue repaglinide. Br J Clin Pharmacol 2003; 56:
305–314.
112. Weaver ML, Orwig BA, Rodriguez LC, Graham ED, Chin JA, Shapiro MJ,
McLeod JF, Mangold JB. Pharmacokinetics and metabolism of nateglinide in
humans. Drug Metab Dispos 2001; 29: 415–421.
113. McLeod JF. Clinical pharmacokinetics of nateglinide. A rapidly-absorbed, short-
acting insulinotropic agent. Clin Pharmacokinet 2004; 43: 97–120.
114. Hatorp V, Bayer T. Repaglinide bioavailability in the fed or fasting state (abstract).
J Clin Pharmacol 1997; 37: 875.
115. Damsbo P, Marbury TC, Hatorp V, Clauson P, Müller PG. Flexible prandial glucose
regulation with repaglinide in patients with Type 2 diabetes. Diabetes Res Clin
Pract 1999; 45: 31–39.
116. Keilson L, Mather S, Walter YH, Subramanian S, McLeod JF. Synergistic effects
of nateglinide and meal administration on insulin secretion in patients with type
2 diabetes mellitus. J Clin Endocrinol Metab 2000; 85: 1081–1086.
117. Karara AH, Dunning BE, McLeod JF. The effect of food on the oral bioavailability
and the pharmacodynamic actions on the insulinotropic agent nateglinide in
healthy subjects. J Clin Pharmacol 1999; 39: 172–179.
118. Luzio SD, Anderson DM, Owens DR. Effects of timing of administration and
meal composition on the pharmacokinetic and pharmacodynamic characteristic
of the short-acting oral hypoglycemic agent nateglinide in healthy subjects. J Clin
Endocrinol Metab 2001; 86: 4874–4880.
119. Alemani M, Remesar X, Fernandez-Lopez JA. Drug strategies for the treatment
of obesity. Drugs 2003; 6: 566–572.
120. McNeely W, Benfield P. Orlistat. Drugs 1998; 56: 241–249.
121. O’Meara S, Riemsma R, Shirran L, Mather L, ter Riet G. A systematic review of
the clinical effectiveness of orlistat used for the management of obesity. Obesity
Rev 2004; 5: 51–68.
122. Scheen AJ, Ernest PH. Antiobesity treatment in type 2 diabetes: results of clinical
trials with orlistat and sibutramine. Diabetes Metab 2002; 28: 437–445.
DK5836_C001.fm Page 27 Monday, May 22, 2006 11:50 AM
123. Torgerson JS, Hauptman J, Boldrin MN, Sjöström L. Xenical in the Prevention
of Diabetes in Obese Subjects (XENDOS) study. A randomized study of orlistat
as an adjunct to lifestyle for the prevention of type 2 diabetes in obese patients.
Diabetes Care 2004; 27: 155–161.
124. Muls E, Kolanowski J, Scheen A, Van Gaal L, for the Obelhyx Study Group. The
effects of orlistat on weight and on serum lipids in obese patients with hypercho-
lesterolemia: a randomized, double-blind, placebo-controlled, multicenter study.
Int J Obesity 2001; 25: 1713–1721.
125. Hauptman JB, Jeunet FS, Hartmann D. Initial studies in humans with the novel
gastrointestinal lipase inhibitor ro-18-0647 (tetrahydrolipstatin). Am J Clin Nutr
1992; 55 Suppl: 309–313.
126. Guzelhan C, Odink J, Niestijl Jansen-Zuidema JJ, Hartmann D. Influence of
dietary composition on the inhibition of fat absorption by orlistat. J Int Med Res
1994; 22: 255–265.
127. Hartmann D, Hussain Y, Guzelhan C, Odink J. Effect on dietary fat absorption of
orlistat, administered at different times relative to meal intake. Br J Clin Phar-
macol 1993; 36: 266–270.
128. Melia AT, Koss-Twardy SG, Zhi J. The effect of orlistat, an inhibitor of dietary
fat absorption, on the absorption of vitamins A and E in healthy volunteers. J Clin
Pharmacol 1996; 36: 647–653.
129. Drent ML, Larsson I, William-Olsson T, Quaade F, Czubayko F, von Bergmann
K, Strobel W, Sjostrom L, van der Veen EA. Orlistat (RO 18-0647), a lipase
inhibitor, in the treatment of human obesity: a multiple dose study. Int J Obesity
1995; 19: 221–226.
130. Zhi Zhi J, Melia AT, Guerciolini R, Koss-Twardy SG, Passe SM, Rakhit A,
Sadowski JA. The effect of orlistat, an inhibitor of dietary fat absorption, on the
pharmacokinetics of beta-carotene in healthy volunteers. J Clin Pharmacol 1996;
36: 152–159.
131. Sjostrom L, Rissanen A, Andersen T, Boldrin M, Golay A, Koppeschaar HP,
Krempf M, for the European Multicentre Orlistat Study Group. Randomized
placebo-controlled trial of orlistat for weight loss and prevention of weight regain
in obese patients. Lancet 1998; 352: 167–173.
132. McNeely W, Goa KL. Sibutramine. A review of its contribution to the management
of obesity. Drugs 1998; 56: 1093–1124.
133. James WP, Astrup A, Finer N, Hilsted J, Kopelman P, Rossner S, Saris WH, Van
Gaal LF, for the Storm Study Group. Effect of sibutramine on weight maintenance
after weight loss: a randomized trial. Lancet 2000; 356: 2119–2125.
134. O’Meara S, Riemsma R, Shirran L, Mather L, ter Riet G. The clinical effectiveness
and cost-effectiveness of sibutramine in the management of obesity: a technology
assessment. Health Technol Assess 2002; 6: 1–97.
135. Rolls BJ, Shide DJ, Thorwart ML, Ulbrecht JS. Sibutramine reduces food intake
in non-dieting women with obesity. Obes Res 1998; 6: 1–11.
136. Chapelot D, Marmonier C, Himaya A, Hanotin C, Thomas F. Modalities of the
food intake-reducing effect of sibutramine (abstract). Int J Obes 1998; 22 (Suppl
3): S270.
137. Saris WHM, STORM Study Group. Sibutramine trial of obesity reduction and
maintenance. Lifestyle and diet during the 6-month run-in phase (abstract). Int J
Obes 1998; 22 (Suppl 3): S271.
DK5836_C001.fm Page 28 Monday, May 22, 2006 11:50 AM
28 Nutrient–Drug Interactions
138. Garratt CJ, Hind ID, Haddock RE. Sibutramine metabolite bioavailability: effect
of dose level and food (abstract). J Clin Pharmacol 1995; 35: 927.
139. Knopp R. Drug treatment of lipid disorders. N Engl J Med 1999; 341: 498–511.
140. Evans M, Roberts A, Davies S, Rees A. Medical lipid-lowering therapy: current
evidence, ongoing trials and future developments. Drugs 2004; 64: 1181–1196.
141. Law MR, Wald NJ, Rudnicka AR. Quantifying effect of statins on low density
lipoprotein cholesterol, ischaemic heart disease, and stroke: systematic review and
meta-analysis. BMJ 2003; 326: 1423–1427.
142. Bonetti PO, Lerman LO, Napoli C, Lerman A. Statin effects beyond lipid lowering
—are they clinically relevant? Eur Heart J 2003; 24: 225–248.
143. Snow V, Aronson MD, Hornbake ER, Mottur-Pilson C, Weiss KB, for the Clinical
Efficacy Assessment Subcommittee of the American College of Physicians. Lipid
control in the management of type 2 diabetes mellitus: a clinical practice guideline
from the American College of Physicians. Ann Intern Med 2004; 140: 644–649.
144. Steiner G. Fibrates in the metabolic syndrome and in diabetes. Endocrinol Metab
Clin North Am 2004; 33: 545–555.
145. Bays HE, Dujovne CA. Drug interactions of lipid-altering drugs. Drug Saf 1998;
9: 355–371.
146. Williams D, Feely J. Pharmacokinetic–pharmacodynamic drug interactions with
HMG-CoA reductase inhibitors. Clin Pharmacokinet 2002; 41: 343–370.
147. Martin J, Krum H. Cytochrome P450 drug interactions within the HMG-CoA
reductase inhibitor class: are they clinically relevant? Drug Safety 2003; 26: 13–21.
148. Bellosta S, Paoletti R, Corsini A. Safety of statins. Focus on clinical pharmaco-
kinetics and drug interactions. Circulation 2004; 109 (Suppl. III): III50–57.
149. Dobrinska MR, Stubbs RJ, Gregg MH, et al. Effects of dose and food on HMG-
CoA reductase inhibitor profiles after lovastatin (mevacor) (abstract). Pharm Res
1988; 5: S182.
150. Richter WO, Jacob BG, Schwandt P. Interaction between fibre and lovastatin
(letter). Lancet 1991; 338: 706.
151. Pan HY, DeVault AR, Brescia D, Willard DA, McGovern ME, Whigan DB,
Ivashkiv E. Effect of food on pravastatin pharmacokinetics and pharmacodynam-
ics. Int J Clin Pharmacol Ther Toxicol 1993; 31: 291–294.
152. Radulovic LL, Cilla DD, Posvar EL, Sedman AJ, Whitfield LR. Effect of food on
the bioavailability of atorvastatin, an HMG-CoA reductase inhibitor. J Clin Phar-
macol 1995; 35: 990–994.
153. Whitfield LR, Stern RH, Sedman AJ, Abel R, Gibson DM. Effect of food on the
pharmacodynamics and pharmacokinetics of atorvastatin, an inhibitor of HMG-
CoA reductase. Eur J Drug Metab Pharmacokinet 2000; 25: 97–101.
154. Dujovne CA, Davidson MH. Fluvastatin administration at bedtime versus the
evening meal: a multicenter comparison of bioavailability, safety, and efficacy.
Am J Med 1994; 96 (Suppl 6A): S37–40.
155. Kantola T, Kivisto KT, Neuvonen PJ. Grapefruit juice greatly increases serum
concentrations of lovastatin and lovastatin acid. Clin Pharmacol Ther 1998; 63:
397–402.
156. Lilja JJ, Kivisto KT, Neuvonen PJ. Grapefruit juice increases serum concentrations
of atorvastatin and has no effect on pravastatin. Clin Pharmacol Ther 1999; 66:
118–127.
DK5836_C001.fm Page 29 Monday, May 22, 2006 11:50 AM
157. Lilja JJ, Kivisto KT, Neuvonen PJ. Grapefruit juice–simvastatin interaction: effect
on serum concentrations of simvastatin, simvastatin acid, and HMG-CoA reduc-
tase inhibitors. Clin Pharmacol Ther 1998; 64: 477–483.
158. Rogers JD, Zhao J, Liu L, Amin RD, Gagliano KD, Porras AG, Blum RA, Wilson
MF, Stepanavage M, Vega JM. Grapefruit juice has minimal effects on plasma
concentrations of lovastatin-derived 3-hydroxy-3-methylglutaryl coenzyme A
reductase inhibitors. Clin Pharmacol Ther 1999; 66: 358–366.
159. Kane GC, Lipsky JJ. Drug-grapefruit juice interactions. Mayo Clin Proc 2000;
75: 933–942.
160. Rosenson RS. Rosuvastatin: a new inhibitor of HMG-coA reductase for the treat-
ment of dyslipidemia. Expert Rev Cardiovasc Ther 2003; 1: 495–505.
161. Todd PA, Ward A. Gemfibrozil. A review of its pharmacodynamic and pharmaco-
kinetic properties, and therapeutic use in dyslipidaemia. Drugs 1988; 36: 314–339.
162. Spencer CM, Barradell LB. Gemfibrozil. A reappraisal of its pharmacological
properties and place in the management of dyslipidaemia. Drugs 1996; 51:
982–1018.
163. Balfour JA, McTavish D, Heel RC. Fenofibrate. A review of its pharmacodynamic
and pharmacokinetic properties and therapeutic use in dyslipidemia. Drugs 1990;
40: 260–290.
164. Adkins JC, Faulds D. Micronized fenofibrate: A review of its pharmacodynamic
properties and clinical efficacy in the management of dyslipidaemia. Drugs 1997;
54: 615–633.
165. Najib J. Fenofibrate in the treatment of dyslipidemia: a review of the data as they
relate to the new suprabioavailable tablet formulation. Clin Ther 2002; 24:
2022–2050.
166. Miller DB, Spence JD. Clinical pharmacokinetics of fibric acid derivatives
(fibrates). Clin Pharmacokinet 1998; 34: 155–162.
167. Luner PE, Babu SR, Radebaugh GW. The effects of bile salts and lipids on the
physicochemical behavior of gemfibrozil. Pharm Res 1994; 11: 1755–1760.
168. Keating GM, Ormrod D. Micronized fenofibrate. An updated review of its clinical
efficacy in the management of dyslipidaemia. Drugs 2002; 62: 1909–1944.
169. Abbott Laboratories. TricorR (fenofibrate capsules) prescribing information [on
line]. Available from URL: http://www.tricorx.com [Accessed 2002 March 5].
170. Bays H. Ezetimibe. Expert Opin Invest Drugs 2002; 11: 1587–1604.
171. Jeu LA, Cheng JWM. Pharmacology and therapeutics of ezetimibe (SCH 58235),
a cholesterol-absorption inhibitor. Clin Ther 2003; 25: 2352–2387.
172. Courtney RD, Kosoglou T, Statkevich P, et al. Effect of food on the oral bioavail-
ability of ezetimibe (abstract). Clin Pharmacol Ther 2002; 71: 80.
173. Punwani N, Pai S, Bach C, et al. Effect of food on oral bioavailability of SCH
58235 in healthy male volunteers (abstract). AAPS Pharm Sci 1998; 1: 2147.
174. Dujovne C, Held J, Lipka L, et al. Does cholesterol and/or fat intake affect plasma
lipid efficacy of ezetimibe? (abstract). J Am Coll Cardiol 2002; 39 (Suppl A):
227A.
175. Cater NB. Historical and scientific basis for the development of plant stanol ester
foods as cholesterol-lowering agents. Eur Heart J 1999; 1: S36–44.
176. Blair SN, Capuzzi DM, Gottlieb SO, Nguyen T, Morgan JM, Cater NB. Incre-
mental reduction of serum total cholestrol and low-density lipoprotein cholesterol
with the addition of plant stanol ester-containing spread to statin therapy. Am J
Cardiol 2000; 86: 46–52.
DK5836_C001.fm Page 30 Monday, May 22, 2006 11:50 AM
30 Nutrient–Drug Interactions
177. Vorlat A, Conraads VM, Vrints CJ. Regular use of margarine-containing stanol/ste-
rol esters reduces total and low-density lipoprotein (LDL) cholesterol and allows
reduction of statin therapy after cardiac transplantation: preliminary observations.
J Heart Lung Transplant 2003; 22:1059–1062.
DK5836_C002.fm Page 31 Monday, May 22, 2006 12:37 PM
2 Hypolipidemic Therapy:
Drugs, Diet, and
Interactive Effects
Alexandre Loktionov
CONTENTS
2.1 Introduction................................................................................................31
2.2 Therapy of CVD with Hypocholesterolemic and
Hypolipidemic Drugs ...............................................................................32
2.2.1 HMG-CoA Reductase Inhibitors (Statins)....................................33
2.2.2 Fibric Acid Derivatives (Fibrates).................................................34
2.2.3 Bile Acid Transport Inhibitors ......................................................35
2.2.4 Niacin (Nicotinic Acid) .................................................................36
2.2.5 Cholesterol Absorption Inhibitors .................................................37
2.2.6 Emerging and Future Directions in Hypolipidemic Therapy .......37
2.3 Dietary Factors in Lipid Homeostasis and Their Interactions with
Hypolipidemic Drugs ................................................................................38
2.3.1 Dietary Ingredients Possessing Natural Hypolipidemic and
Cardioprotective Properties ...........................................................39
2.3.2 Interactions between Dietary Components and
Hypolipidemic Drugs ....................................................................44
2.4 Conclusion .................................................................................................48
References ...........................................................................................................49
2.1 INTRODUCTION
In the past, the relationship between dietary factors and cardiovascular disease
(CVD) was often regarded as primarily and almost exclusively dependent on lipid
(especially saturated fat and cholesterol) consumption and metabolism leading
to increased serum cholesterol; in particular, low density lipoprotein (LDL) levels
resulting in atherosclerotic vascular changes. This idea constituted the core of
the traditional “diet–heart” hypothesis.1 With the development of scientific knowl-
edge on both CVD pathogenesis and bioactive properties of nutrients, the field
is rapidly becoming more and more complicated. The range of known pathways
31
DK5836_C002.fm Page 32 Monday, May 22, 2006 12:37 PM
32 Nutrient–Drug Interactions
34 Nutrient–Drug Interactions
CHOLESTEROL
BYOSYNTHESIS
INHIBITION
(SMALL LDL
ANTIOXIDANT REDUCTION)
PLAQUE
EFFECTS STABILIZATION
(REDUCTION OF
oxLDL)
ANTITHROMBOTIC VASOMOTION
EFFECT
STATINS NORMALIZATION
INCREASE
OF ENDOTHELIAL
NITRIC OXIDE FUNCTION
BIOAVAILABILITY IMPROVEMENT
INHIBITION
OF
INFLAMMATION
FIGURE 2.1 Pleiotropic beneficial effects of statin therapy. (Dotted arrows indicate inter-
active links between different mechanisms.)
effects exists.17 Dietary influences on statin effects are possible and will be
considered later in this chapter.
The role of activated PPARα in HDL level modulation is less clear; however,
it is believed that induction of the transcription of ApoA-I and Apo A-II lipopro-
teins in hepatocytes may contribute to the increase of plasma HDL concentrations
and a more efficient reverse cholesterol transport.39,40 Given that PPARs are known
to exert multiple biological effects, it is not surprising that a growing body of
information regarding pleiotropic effects of fibrates now emerges. Antiinflamma-
tory action and favorable effects on both blood coagulation and fibrinolysis
system40 look especially interesting from a clinical point of view. It is also
important to stress that other members of the PPAR family, especially PPARγ,
are deeply involved in lipid metabolism regulation. PPARγ agonists of the thia-
zolidinedione family are already in use for the treatment of diabetes mellitus;
however, their hypolipidemic potential has recently emerged as a promising sign
for the development of new approaches to CVD treatment.41 Perspectives of
developing new drugs combining PPARα and PPARγ agonist activity39 may
provide a new step in this direction.
Four members of the fibrate family are now in wide clinical use: bezafibrate,
ciprofibrate, fenofibrate, and gemfibrosil.37 Drugs of this group are especially
efficient in combination with statins or as monotherapy in patients with normal
LDL cholesterol levels, in particular for the treatment of cardiovascular manifes-
tations of the metabolic syndrome.37,42–44 Although fibrates are demonstrated to
be well tolerated in most cases, a few side effects, including changes of hepatic
function (hepatic transaminase level elevation), gastrointestinal side effects, and
myopathy have been reported.45,46 Some effects have been associated with par-
ticular members of the fibrate family, like rhabdomyolysis with gemfibrozil35,47
or hyperhomocysteinemia with fenofibrate.48 Like statins, fibrates have been
shown to be carcinogenic in rodents;36 however, there is no clinical information
on this potential problem. It has also been repeatedly stressed that combined
hypolipidemic therapy with statins and fibrates may be associated with drug
interactions and higher risk of adverse effects.35,49 Dietary influences should be
taken into account in this context and will be discussed later in this chapter.
36 Nutrient–Drug Interactions
forms nonabsorbable complexes with bile acids in the gastrointestinal tract, has
been reported four to six times more potent than other bile acid sequestrants, and
especially recommended for combination therapy with statins.53
Another family of new drugs affecting bile acid transport is represented by
agents inhibiting ileal sodium-dependent bile acid transporter (IBAT), which
reduce serum cholesterol level by suppressing the reuptake of bile acids in the
ileum. Results from animal experiments with a few synthetic compounds have
provided encouraging results;54–56 however, drugs of this family have not hitherto
been introduced into clinical practice.
38 Nutrient–Drug Interactions
40 Nutrient–Drug Interactions
TABLE 2.1
Dietary Ingredients Exerting Beneficial Influences on Hyperlipidemic and
Dyslipidemic Conditions
Active Ingredients Dietary Sources Physiological Effects Refs.
Fatty Acids
Monounsaturated fatty Canola, olive, Mild hypocholesterolemic effect, 83–85
acids (MUFA): oleic safflower, possible reduction of oxLDL
acid (OA) sunflower oils, and (additional nonlipid beneficial effects
nuts, olives, possible)
avocado
Omega-6 Corn, safflower, Mild hypocholesterolemic effect 86–88
polyunsaturated fatty sunflower oils (downregulation of LDL production
acids (ω-6 PUFA): and enhancement of its clearance);
linoleic acid (LA) adverse effects on cytokines and
platelet aggregation are not excluded
Omega-3 Canola, flaxseed, Mild hypocholesterolemic effect, serum 81, 86, 88,
polyunsaturated fatty soybean oils, triglyceride lowering effect; additional 89–92
acids (ω-3 PUFA): walnuts (nonlipid) beneficial effects include
linoleic acid (ALA) antiarrhythmic (especially EPA and
DHA) and probably antithrombotic
action.
Eicosapentaenoic acid Fish, shellfish
(EPA) and
docosahexaenoic acid
(DHA)
Flavonoids
Flavonols: quercetin, Apple, onion, kale, Mild hypocholesterolemic effect; 93–98
kaemprefol, broccoli, cabbage, atioxidant action is believed to protect
myrocetin, fisetin cherries, berries, from oxLDL accumulation; additional
tea, red wine (nonlipid) effects include
antiinflammatory action, reduction of
platelet aggregation, improved
vascular reactivity, weak estrogenic
activity (only isoflavones)
Flavanols (catechins): Green and black
catechin, epicatechin, tea, red wine,
epicatechin 3-gallate, apples, black
epigallocatechin, chocolate, cocoa
epigallocatechin 3-
gallate, gallocatechin
Flavones: apigenin, Parsley, celery,
luteolin thyme
DK5836_C002.fm Page 41 Monday, May 22, 2006 12:37 PM
Flavanones: Oranges,
naringenin, grapefruit, and
hesperetin, their juices
eriodictyol
Anthocyanins: Berries, cherries,
(anthocyanidins) red wine
Cyanidin,
pelargonidin,
delphinidin, peonidin,
petunidin, malvinidin
Isoflavones: genistein, Soy and soy-
daidzein, dihydro- containing foods,
daidzein, O-desmethy- chick peas
langolensin, equol
Antioxidant Vitamins
α-Tocopherol Vegetable oils Experimental evidence of lipid 99–105
(vitamin E) (sunflower, olive), peroxidation inhibition: controversial
nuts, seeds results regarding protection against
CVD in humans
Ascorbic acid Fruit (especially Experimental evidence of lipid 99, 100,
(vitamin C) citrus), vegetables peroxidation inhibition: controversial 102,
results regarding protection against 104–106
CVD in humans
Carotenoids: Green leafy, root, Antioxidant action was suggested as a 99,
β-carotene and fruiting protective factor against oxLDL 102–105,
vegetables accumulation; antiinflammatory effect 107
possible; controversial results
regarding protection against CVD in
humans
Lycopene Tomatoes, tomato Antioxidant action may provide lipid 108–112
products, peroxidation inhibition; protective
watermelon effect against CVD reported
Lutein Leafy vegetables, Antioxidant effect may influence lipid 113
fruit peroxidation; protective effect against
CVD requires confirmation
DK5836_C002.fm Page 42 Monday, May 22, 2006 12:37 PM
42 Nutrient–Drug Interactions
Sulfur-Containing
Compounds
Allicin, ajoene, Allium vegetables, Mild hypocholesterolemic effect 114–117
S-allylcysteine, especially garlic repeatedly described in experimental
diallyl disulfide, and its and small human studies, but needs
S-methylcysteine preparations further confirmation; possible
sulfoxide, mechanisms may involve inhibition of
S-allylcysteine hepatic cholesterol synthesis and
sulfoxide antioxidant effect; possible additional
(nonlipid) effects include
enhancement of fibrinolytic activity,
inhibition of platelet aggregation,
blood pressure lowering
Fiber
Soluble fiber Cereals, legumes Well-documented mild (118–123)
(psyllium, pectins, guar hypocholesterolemic effect; LDL
gum, some reduction might be related to intestinal
hemicelluloses) binding of bile acids and/or cholesterol
followed by LDL receptor
upregulation and increased LDL
clearance; additional (nonlipid) effects
include C-reactive protein reduction
and normalization of blood pressure
Protein
Legume protein Soy, legumes Legume proteins, especially those 124–127
derived from soy, appear to have
hypocholesterolemic effects;
mechanisms are not clear, difficult to
separate from isoflavone effects
risk.99–101 The idea has been tested in a number of controlled trials investigating
the effects of antioxidant vitamin supplements on CVD risk. It is remarkable that
the results of these studies have been largely disappointing,101,103–105,141 leading to
the conclusion that there is no basis for recommending that patients take vitamin
C or E supplements or other antioxidants for the express purpose of preventing
or treating coronary artery disease.141,142
Likewise, there is some controversy regarding the use of garlic as a potential
lipid-lowering and cardioprotective food component. Commonly used garlic prep-
arations and supplements have varying amounts of allicin, which is believed to
be at the core of garlic bioactivity (see Table 2.1), so making a definite conclusion
on their lipid-reducing efficiency is difficult.114–117 Nevertheless, despite all prob-
lems discussed above, beneficial effects of antioxidant-rich diets based on fruit,
vegetables, and products derived from these sources are evident,97,143–145 and such
diets are widely recommended to reduce CVD risk.141 It appears to be very likely
that consumption of natural products containing different antioxidant components
acting synergistically may be much more efficient in providing cardiovascular
protection than isolated action of food supplements.97
Mild hypocholesterolemic effect of dietary soluble fiber is well documented;
however, precise mechanisms of this phenomenon are not clear and may be
partially related to other components of fiber-rich cereals and legumes.118 Pro-
motion of increased fiber consumption, especially with whole grains, should be
considered as one of the strategic ways of changing overall Western dietary
patterns towards CVD prevention.
The amount and type of dietary protein is closely related to the problem of
fat intake. It was repeatedly shown that substitution of soy for animal protein
results in LDL and triglyceride reduction.124–127 At the same time, the mechanism
of this reduction is unknown and, especially in the case of soy, it is difficult to
exclude an impact of isoflavones.124,126 Overall food composition rather than
protein type appears to be more important for serum lipid balance. It has been
suggested that consumption of animal protein per se may not be harmful, but is
commonly associated with an increased intake of saturated fat.146 To avoid this
unhealthy association, it can be recommended to shift the main sources of dietary
protein by replacing red meat with nuts, soybeans, legumes, poultry, and fish.80
The use of natural cholesterol-lowering properties of plant sterols and stanols
inhibiting intestinal cholesterol absorption has emerged as an attractive strategy
for serum lipid level correction, especially in mildly hypercholesterolemic sub-
jects.130 New polyunsaturated margarines with added sterols and stanols have
recently been made available for wide consumption.129 It should be noticed,
however, that the use of these products can also reduce the absorption of fat
soluble vitamins.129 Possible consequences of long-term sterol/stanol intake are
not entirely known, so caution is needed in the use of this approach.
Food components discussed above are widely used as ingredients of different
diets directed on both CVD prevention and attenuation of hyperlipidemia in CVD
patients. The search for new natural substances with hypolipidemic properties
continues, and such agents as policosanol (a mixture of alcohols isolated from
DK5836_C002.fm Page 44 Monday, May 22, 2006 12:37 PM
44 Nutrient–Drug Interactions
substances from entering the cell and eliminating products of xenobiotic detoxi-
fication reactions.155 Intestinal P-glycoprotein (a member of the multidrug resis-
tance transporter family) expressed in the apical membrane of enterocytes appears
to exert its transporter functions in concert with the metabolic role of CYP3A4,
thus constituting a dynamic subsystem controlling xenobiotic metabolism/elim-
ination rate.159 Even from the brief description given above, it becomes apparent
that disruption of proper functioning of this complex system can have hazardous
consequences.
The problem of diet–drug interactions was suddenly highlighted by an unfore-
seen discovery in 1989 of the effect of grapefruit juice (GJ) on bioavailability of
felodipine, a calcium channel blocker used for hypertension treatment.160 Since
then, multiple studies have shown GJ interactions with various drugs of different
families often lead to enhanced adverse effects (see References 153, 161). GJ
interactions with hypolipidemic drugs (statins) and some other medicines often
prescribed to CVD patients are listed in Table 2.2. Investigation of the mecha-
nisms of GJ–drug interaction has revealed that oral intake of GJ selectively
inhibits CYP3A4 in the small intestine,176–178 resulting in a considerable reduction
of oxidative metabolism of the substrates of CYP3A4, including numerous drugs.
Only intestinal enzyme inhibition was observed, whereas liver CYP3A4 activity
appeared to be unaffected177 as well as pharmacodynamics of drugs metabolized
by this enzyme after their intravenous administration.170,179,180
However, it has recently been reported that consumption of large amounts of
GJ can inhibit hepatic CYP3A4 as well181; thus the effect may be dose-dependent
with a much higher threshold for the enzyme in the liver. The search for the active
GJ ingredients was focused on furanocoumarins (psoralens) and isoflavones as
contributors to CYP3A4 inhibition. GJ furanocoumarins, especially bergamottin
and 6′,7′-dihydroxybergamottin are regarded as major CYP3A4 inhibi-
tors,178,182–184 but binding properties and enzyme inhibition kinetics of the two
furanocoumarins are different,185 suggesting that different inhibition mechanisms
can be involved. In addition, it has been shown that grapefruit flavonoids, in
particular, naringin and its derivative naringenin, can act as competitive inhibitors
of CYP3A4.186 The influence of GJ on the intestinal drug metabolism appears to
be even more complex since it is not limited by the effects on CYP3A4. GJ
components have also been shown to suppress the P-glycoprotein transporter
activity,153,161,187,188 thus further increasing ingested drug bioavailability.
From the point of view of hypolipidemic therapy, interactions between GJ
and HMG-CoA reductase inhibitors are especially important since consumption
of the juice by individuals taking statins predominantly metabolized by CYP3A4
(atorvastatin, lovastatin or simvastatin) can significantly increase the risk of
adverse effects.161,164,166 Interactions are much less likely with fluvastatin, which
is mainly metabolized by CYP2C9,162 or pravastatin, rosuvastatin, and pitavasta-
tin, which are largely metabolized through P450-independent pathways.163–165,189
It is also apparent that considerable interindividual variation in responses to statin
therapy strongly depends on genetic heterogeneity of human populations. Indeed,
numerous polymorphic variants of the CYP3A4 gene have been identified,190–193
DK5836_C002.fm Page 46 Monday, May 22, 2006 12:37 PM
46 Nutrient–Drug Interactions
TABLE 2.2
Grapefruit Juice Interactions with Drugs Used for the Treatment of
Hyperlipidemia and Other Manifestations of Cardiovascular Disease
Drug Classes, Conditions
Individual Drugs Treated Reported Adverse Effects Refs.
HMG-CoA Reductase
Inhibitors (Statins)
Simvastatin, lovastatin, Hyperlipidemia Myopathy, rhabdomyolysis, 161–166
atorvastatin acute renal failure
Pravastatin, fluvastatin, Little or no influence
rosuvastatin, pitavastatin
Calcium Channel
Blockers
(Dihydropyridines)
Felodipine, nicardipine, Hypertension, Excessive vasodilatation, 153, 161,
nifedipine,nimoldipine, angina pectoris hypotension, tachycardia 167–171
nisoldipine, pranidipine, treatment with
nitrendipine, verapamil verapamil
Amlodipine, barnidipine, Little or no influence
diltiazem
Angiotensin II Receptor 1
Antagonistsa
Losartan Hypertension Reduced therapeutic effect 161, 172
Beta-Blockersa
Carvedilol Hypertension, Increased drug toxicity 161, 172
heart failure
treatment
Celiprolol Decreased drug bioavailability
Antiarrhythmic Agentsa
Amiodarone, quinidine, Arrhythmias Increased drug toxicity 161, 174,
disopyramide, 175
propafenone
a No information on interactions with other drugs of this group.
and some of them have already been shown to affect lipid profiles in patients
receiving statin therapy.194,195 Moreover, potential influence of multiple polymor-
phisms of MDR1 gene encoding P-glycoprotein196,197 or other transporter-encod-
ing genes is impossible to exclude. Interestingly, it has recently been reported
that polymorphisms of organic anion transporting polypeptide C have been
associated with plasma pravastatin concentrations.198 At the same time, organic
anion-transporting polypeptides may be inhibited by GJ,161,199 constituting another
interaction area.
DK5836_C002.fm Page 47 Monday, May 22, 2006 12:37 PM
48 Nutrient–Drug Interactions
2.4 CONCLUSION
Serum lipid lowering is a pivotal component among measures applied in order
to prevent CVD or delay its progression. Dietary approaches can be successfully
used for primary prevention of cardiovascular conditions, especially coronary
heart disease. General dietary strategy should include recommendations to change
fat intake habits (substitute polyunsaturated and monounsaturated fats for satu-
rated fats, increase intake of ω-3 PUFA) and to consume food rich in fruits,
vegetables, nuts, and whole grains (i.e., products rich in natural antioxidants,
soluble fiber, vegetable protein). It appears that natural products containing com-
plex mixtures of bioactive ingredients acting synergistically provide more bene-
ficial effects compared to most presently available artificial dietary supplements
and additives.
Secondary CVD prevention directed to delaying disease progress in individ-
uals with existing conditions is now based upon application of combined thera-
peutic strategies including hypolipidemic drug therapy and additional dietary
measures. The development of new lipid-lowering agents is progressing very fast
through introduction of new potent drugs often providing beneficial pleiotropic
effects affecting different mechanisms involved in CVD pathogenesis. HMG-CoA
reductase inhibitors (statins) have emerged as the central element of hypolipi-
demic therapy, especially in cases with seriously increased LDL levels. Avail-
ability of other types of hypolipidemic medicines that can be combined with
statins considerably widens therapeutic options, allowing development of indi-
vidualized treatment schemes. Successful combination of statin therapy with the
use of fibrates, bile acid transport inhibitors, niacin, and cholesterol absorption
inhibitors provides an expanding arsenal of therapeutic approaches. Dietary rec-
ommendations, which are generally similar to those used for primary CVD
DK5836_C002.fm Page 49 Monday, May 22, 2006 12:37 PM
prevention, should be specifically adapted for each individual case. This require-
ment is especially important in view of accumulating evidence of possible inter-
actions between food components and medicines, especially orally ingested drugs.
It appears that in many cases food–drug interactions, if properly assessed and
understood, can potentially be employed to enhance beneficial therapeutic effects.
This understanding, however, requires much better knowledge of individual char-
acteristics of each patient determined by genetic background. “Genetic profiling”
of individuals subjected to hypolipidemic therapy can greatly reduce the risk of
adverse effects and facilitate determination of optimal therapeutic and dietary
schemes applied for CVD treatment.
REFERENCES
1. Gordon T. The diet-heart idea. Outline of a history. Am J Epidemiol 1988; 127:
220–225.
2. Lusis AJ. Atherosclerosis. Nature 2000; 407: 233–241.
3. Glass CK, Witztum JL. Atherosclerosis: the road ahead. Cell 2001; 104: 503–516.
4. Libby P. Inflammation in atherosclerosis. Nature 2002; 420: 868–874.
5. Maas R, Böger RH. Old and new cardiovascular risk factors: from unresolved
issues to new opportunities. Atherosclerosis 2003; Suppl. 4: 5–17.
6. Haffner S, Taegtmeyer H. Epidemic obesity and the metabolic syndrome. Circu-
lation 2003, 108: 1541–1545.
7. Deedwania PC. Metabolic syndrome and vascular disease: is nature or nurture
leading the new epidemic of cardiovascular disease? Circulation 2004; 109: 2–4.
8. Moller DE, Kaufman KD. Metabolic syndrome: a clinical and molecular perspec-
tive. Ann Rev Med 2005; 56: 45–62.
9. Ford ES, Giles WH, Dietz WH. Prevalence of the metabolic syndrome among
U.S. adults: findings from the third National Health and Nutrition Examination
Survey. JAMA 2002; 287: 356–359.
10. Fruchart JC, Nierman MC, Stroes ES, Kastelein JJ, Duriez P. New risk factors
for atherosclerosis and patient risk assessment. Circulation 2004; 109; III15–III19.
11. Splaver A, Lamas GA, Hennekens CH. Homocysteine and cardiovascular disease:
biological mechanisms, observational epidemiology, and the need for randomized
trials. Am Heart J 2004; 148: 34-40.
12. Ross R. Atherosclerosis — an inflammatory disease. N Engl J Med 1999; 340:
115–126.
13. Shishehbor MH, Hansen SL. Inflammatory and oxidative markers in atheroscle-
rosis: relationship to outcome. Curr Atheroscler Rep 2004; 6: 243–250.
14. Diaz MN, Frei B, Vita JA, Keaney JF Jr. Antioxidants and atherosclerotic heart
disease. N Engl J Med 1997; 337: 408–416.
15. Rosenson RS. Statins in atherosclerosis: lipid-lowering agents with antioxidant
capabilities. Atherosclerosis 2004; 173: 1–12.
16. Stocker R, Keaney JF Jr. Role of oxidative modifications in atherosclerosis. Phys-
iol Rev 2004; 84: 1381–1478.
17. Evans M, Roberts A, Davies S, Rees A. Medical lipid-regulating therapy: current
evidence, ongoing trials and future developments. Drugs 2004; 64: 1181–1196.
DK5836_C002.fm Page 50 Monday, May 22, 2006 12:37 PM
50 Nutrient–Drug Interactions
33. Amarenco P, Lavallee P, Touboul PJ. Stroke prevention, blood cholesterol, and
statins. Lancet Neurol 2004; 3: 271–278.
34. Collins R, Armitage J, Parish S, Sleight P, Peto R; Heart Protection Study Col-
laborative Group. MRC/BHF Heart Protection Study of cholesterol-lowering with
simvastatin in 5963 people with diabetes: a randomised placebo-controlled trial.
Lancet 2003; 361:2005–2016.
35. Keech A, Colquhoun D, Best J, Kirby A, Simes RJ, Hunt D, Hague W, Beller E,
Arulchelwam M, Baker J, Tonkin A; Lipid Study Group. Secondary prevention
of cardiovascular events with long-term pravastatin in patients with diabetes or
impaired fasting glucose: results from the LIPID trial. Diabetes Care 2003; 26:
2713–2721.
36. Newman TB, Hulley SB. Carcinogenicity of lipid-lowering drugs. JAMA 1996;
275: 55–60.
37. Chapman MJ. Fibrates in 2003: therapeutic action in atherogenic dyslipidemia
and future perspectives. Atherosclerosis 2003; 171: 1–13.
38. Rader DJ. Effects of nonstatin lipid drug therapy on high-density lipoprotein
metabolism. Am J Cardiol 2003; 91: 18E–23E.
39. Staels B, Dallongeville J, Auwerx J, Schoonjans K, Leitersdorf E, Fruchart JC.
Mechanism of action of fibrates on lipid and lipoprotein metabolism. Circulation
1998; 98: 2088–2093.
40. Barbier O, Pineda Torra I, Duguay Y, Blanquart C, Fruchart JC, Glineur C, Staels
B. Pleiotropic actions of peroxisome proliferator-activated receptors in lipid
metabolism and atherosclerosis. Arterioscler Thromb Vasc Biol 2002; 22:
717–726.
41. Blaschke F, Bruemmer D, Law RE. Will the potential of peroxisome proliferators-
activated receptor agonists be realized in the clinical setting? Clin Cardiol 2004;
27 (Suppl 4): IV3–IV10.
42. Robins SJ. Cardiovascular disease with diabetes or the metabolic syndrome:
should statins or fibrates be first line lipid therapy? Curr Opin Lipidol 2003; 14:
575–583.
43. Fazio S, Linton MF. The role of fibrates in managing hyperlipidemia: mechanisms
of action and clinical efficacy. Curr Atheroscler Rep 2004; 6: 148–157.
44. Maki KC. Fibrates for treatment of the metabolic syndrome. Curr Atheroscler
Rep 2004; 6: 45–51.
45. Tomlinson B, Chan P, Lan W. How well tolerated are lipid-lowering drugs? Drugs
Aging 2001; 18: 665-683.
46. Muscari A, Puddu GM, Puddu P. Lipid-lowering drugs: are adverse effects pre-
dictable and reversible? Cardiology 2002; 97: 115–121.
47. Layne RD, Sehbai AS, Stark LJ. Rhabdomyolysis and renal failure associated
with gemfibrozil monotherapy. Ann Pharmacother 2004; 38: 232–234.
48. Dierkes J, Westphal S, Luley C. Fenofibrate-induced hyperhomocysteinaemia:
clinical implications and management. Drug Safety 2003; 26: 81–91.
49. Graham DJ, Staffa JA, Shatin D, Andrade SE, Schech SD, La Grenade L, Gurwitz
JH, Chan KA, Goodman MJ, Platt R. Incidence of hospitalized rhabdomyolysis
in patients treated with lipid-lowering drugs. JAMA 2004; 292: 2585–2590.
50. Ast M, Frishman WH. Bile acid sequestrants. J Clin Pharmacol 1990; 30: 99–106.
DK5836_C002.fm Page 52 Monday, May 22, 2006 12:37 PM
52 Nutrient–Drug Interactions
51. Davidson MH, Dillon MA, Gordon B, Jones P, Samuels J, Weiss S, Issacsohn J,
Toth P, Burke SK. Colesevelam hydrochloride (cholestagel): a new, potent bile
acid sequestrant associated with a low incidence of gastrointestinal side effects.
Arch Intern Med 1999; 159: 1893–1900.
52. Melian EB, Plosker GL. Colesevelam. Am J Cardiovasc Drugs 2001; 1: 141–146.
53. Steinmetz KL. Colesevelam hydrochloride. Am J Health Syst Pharmacol 2002;
59: 932–939.
54. Higaki J, Hara S, Takasu N, Tonda K, Miyata K, Shike T, Nagata K, Mizui T.
Inhibition of ileal Na+/bile acid cotransporter by S-8921 reduces serum cholesterol
and prevents atherosclerosis in rabbits. Arterioscler Thromb Vasc Biol 1998; 18:
1304–1311.
55. Root C, Smith CD, Sundseth SS, Pink HM, Wilson JG, Lewis MC. Ileal bile acid
transporter inhibition, CYP7A1 induction, and antilipemic action of 264 W94. J
Lipid Res 2002; 43: 1320–1330.
56. Li H, Xu G, Shang Q, Pan L, Shefer S, Batta AK, Bollineni J, Tint GS, Keller
BT, Salen G. Inhibition of ileal bile acid transport lowers plasma cholesterol levels
by inactivating hepatic farnesoid X receptor and stimulating cholesterol 7 alpha-
hydroxylase. Metabolism 2004; 53: 927–932.
57. Rosenson RS. Antiatherothrombotic effects of nicotinic acid. Atherosclerosis
2003; 171: 87–96.
58. McKenney J. New perspectives on the use of niacin in the treatment of lipid
disorders. Arch Intern Med 2004; 164: 697–705.
59. Meyers CD, Kamanna VS, Kashyap ML. Niacin therapy in atherosclerosis. Curr
Opin Lipidol 2004; 15: 659–665.
60. Ganji SH, Tavintharan S, Zhu D, Xing Y, Kamanna VS, Kashyap ML. Niacin
noncompetitively inhibits DGAT2 but not DGAT1 activity in HepG2 cells. J Lipid
Res 2004; 45: 1835–1845.
61. Sakai T, Kamanna VS, Kashyap ML. Niacin, but not gemfibrozil, selectively
increases LP-AI, a cardioprotective subfraction of HDL, in patients with low HDL
cholesterol. Arterioscler Thromb Vasc Biol 2001; 21: 1783–1789.
62. Wise A, Foord SM, Fraser NJ, Barnes AA, Elshourbagy N, Eilert M, Ignar DM,
Murdock PR, Steplewski K, Green A, Brown AJ, Dowell SJ, Szekeres PG, Hassall
DG, Marshall FH, Wilson S, Pike NB. Molecular identification of high and low
affinity receptors for nicotinic acid. J Biol Chem 2003; 278: 9869–9874.
63. Soga T, Kamohara M, Takasaki J, Matsumoto S, Saito T, Ohishi T, Hiyama H,
Matsuo A, Matsushime H, Furuichi K. Molecular identification of nicotinic acid
receptor. Biochem Biophys Res Commun 2003; 303: 364–369.
64. Tunaru S, Kero J, Schaub A, Wufka C, Blaukat A, Pfeffer K, Offermans S. PUMA-
G and HM 74 are receptors for nicotinic acid and mediate its anti-lipolytic effect.
Nature Med 2003; 9: 352–355.
65. Karpe F, Frayn KN. The nicotinic acid receptor — a new mechanism for an old
drug. Lancet 2004; 363: 1892–1894.
66. Rubic T, Trottmann M, Lorenz RL. Simulation of CD36 and the key effector of
reverse cholesterol transport ATP-binding cassette A1 in monocytoid cells by
niacin. Biochem Pharmacol 2004; 67: 411–419.
67. Zellner C, Pullinger CR, Aouizerat BE, Frost PH, Kwok PY, Malloy MJ, Kane
JP. Variations in human HM74 (GPR109B) and HM74A (GPR109A) niacin recep-
tors. Hum Mutat 2005; 25: 18–21.
DK5836_C002.fm Page 53 Monday, May 22, 2006 12:37 PM
68. Darkes MJ, Poole RM, Goa KL. Ezetimibe. Am J Cardiovasc Drugs 2003; 3:
67–76.
69. Sudhop T, Lutjohann D, Kodal A, Igel M, Tribble DL, Shah S, Perevozskaya I,
von Bergmann K. Inhibition of intestinal cholesterol absorption by ezetimibe in
humans. Circulation 2002: 106: 1943–1948.
70. Gagne C, Gaudet D, Bruckert E, Ezetimibe Study Group. Efficacy and safety of
ezetimibe coadministered with atorvastatin or simvastatin in patients with
homozygous familial hypercholesterolemia. Circulation 2002; 105: 2469–2475.
71. Murdoch D, Scott LJ. Ezetimibe/Simvastatin: a review of its use in the manage-
ment of hypercholesterolemia. Am J Cardiovasc Drugs 2004; 4: 405–422.
72. Reyderman L, Kosoglou T, Statkevich P, Pember L, Boutros T, Maxwell SE,
Affrime M, Batra V. Assessment of a multiple-dose drug interaction between
ezetimibe, a novel selective cholesterol absorption inhibitor and gemfibrozil. Int
J Clin Pharmacol Ther 2004; 42: 512–518.
73. Bays H, Stein EA. Pharmacotherapy for dyslipidaemia — current therapies and
future agents. Expert Opin Pharmacother 2003; 4: 1901–1938.
74. Tardif JC, Gregoire J, L’Allier PL, Anderson TJ, Bertrand O, Reeves F, Title LM,
Alfonso F, Schampaert E, Hassan A, McLain R, Pressler ML, Ibrahim R, Lesper-
ance J, Blue J, Heinonen T, Rodes-Cabau J, Avisimibe and Progression of Lesions
on UltraSound (A-PLUS) Investigators. Effects of the acyl coenzyme A:choles-
terol acyltransferase inhibitor avasimibe on human atherosclerotic lesions. Circu-
lation 2004; 110: 3372–3377.
75. Robl JA, Sulsky R, Sun CQ, Simpkins LM, Wang T, Dickson JK Jr, Chen Y,
Magnin DR, Taunk , Slusarchyk WA, Biller SA, Lan SJ, Connolly F, Kunselman
LK, Sabrah T, Jamil H, Gordon D, Harrity TW, Wetterau JR. A novel series of
highly potent benzimidazole-based microsomal triglyceride transfer protein inhib-
itors. J Med Chem 2001; 44: 851–856.
76. Magnin DR, Biller SA, Wetterau J, Rohl JA, Dickson JK Jr, Taunk P, Harrity TW,
Lawrence RM, Sun CQ, Wang T, Logan J, Fryszman O, Connolly F, Jolibois K,
Kunselman L. Microsomal triglyceride transfer protein inhibitors: discovery and
synthesis of alkyl phosphonates as potent MTP inhibitors and cholesterol lowering
agents. Bioorg Med Chem Lett 2003; 13: 1337–1340.
77. Brousseau ME, Schaefer EJ, Wolfe ML, Bloedon LT, Digenio AG, Clark RW,
Mancuso JP, Rader DJ. Effects of an inhibitor of cholesteryl ester transfer protein
on HDL cholesterol. N Engl J Med 2004; 350: 1491–1494.
78. Doggrell SA. Raising high-density lipoprotein cholesterol with inhibitors of cho-
lesteryl ester transfer protein — a new approach to coronary artery disease. Expert
Opin Investig Drugs 2004; 13: 1365–1368.
79. Ory DS. Nuclear receptor signaling in the control of cholesterol homeostasis: have
the orphans found a home? Circ Res 2004; 95: 660–670.
80. Hu WB, Willett WC. Optimal diets for prevention of coronary heart disease. JAMA
2002; 2569–2578.
81. Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol
in Adults. Executive summary of the third report of the National Cholesterol
Education Program (NCEP) expert panel on detection, evaluation, and treatment
of high blood cholesterol in adults (Adult Treatment Panel III). JAMA 2001;
2486–2497.
82. Zeisel S. Regulation of “nutraceuticals.” Science 1999; 285: 1853–1855.
DK5836_C002.fm Page 54 Monday, May 22, 2006 12:37 PM
54 Nutrient–Drug Interactions
83. Gardner CD, Kraemer HC. Monounsaturated versus polyunsaturated dietary fat
and serum lipids. A meta-analysis. Arterioscler Thromb Vasc Biol 1995; 15:
1917–1927.
84. Kris-Etherton PM. Monounsaturated fatty acids and risk of cardiovascular disease.
J Nitr 1999; 129: 2280–2284.
85. Perez-Jimenez F, Lopez-Miranda J, Mata P. Protective effect of dietary monoun-
saturated fat on arteriosclerosis: beyond cholesterol. Atherosclerosis 2002; 163:
385–398.
86. Simopoulos AP. Essential fatty acids in health and chronic disease. Am J Clin
Nutr 1999; 70 (Suppl): 560S–569S.
87. Wijendran V, Hayes KC. Dietary n-6 and n-3 fatty acid balance and cardiovascular
health. Annu Rev Nutr 2004; 24: 597–615.
88. Kris-Etherton PM, Hecker KD, Binkoski AE. Polyunsaturated fatty acids and
cardiovascular health. Nutr Rev 2004; 62: 414–426.
89. Albert CM, Hennekens CH, O’Donnell CJ, Ajani UA, Carey VJ, Willett WC,
Ruskin JN, Manson JE. Fish consumption and risk of sudden cardiac death. Jama
1998; 279: 23–28.
90. Simopoulos AP. The importance of the ratio of omega-6/omega-3 essential fatty
acids. Biomed Pharmacother 2002; 56: 365–379.
91. Din JN, Newby DE, Flapan AD. Omega 3 fatty acids and cardiovascular disease
— fishing for a natural treatment. BMJ 2004; 328: 30–35.
92. de Lorgeril M, Salen P. Alpha-linolenic acid and coronary heart disease. Nutr
Metab Cardiovasc Dis 2004; 14: 162–169.
93. Rimm EB, Katan MB, Ascherio A, Stampfer MJ, Willett WC. Relation between
intake of flavonoids and risk for coronary heart disease in male health profession-
als. Ann Intern Med 1996; 125: 384–389.
94. Scalbert A, Williamson G. Dietary intake and bioavailability of polyphenols. J
Nutr 2000; 130 (Suppl): 2073S–2085S.
95. Arts ICW, Hollman PCH, Feskens EJM, Bueno de Mesquita HB, Kromhout D.
Catechin intake might explain the inverse relation between tea consumption and
ischemic heart disease: the Zutphen Elderly Study. Am J Clin Nutr 2001; 74:
227–232.
96. Knekt P Kumpulainen J, Jarvinen R, Rissanen H, Heliovaara M, Reunanen A,
Hakulinen T, Aromaa A. Flavonoid intake and risk of chronic diseases. Am J Clin
Nutr 2002; 76: 560–568.
97. Liu RH. Health benefits of fruit and vegetables are from additive and synergistic
combinations of phytochemicals. Am J Clin Nutr 2003; 78 (Suppl): 517S–520S.
98. Park D, Huang T, Frishman WH. Phytoestrogens as cardioprotective agents. Car-
diol Rev 2005; 13: 13–17.
99. Diaz MN, Frei B, Vita JA, Keaney JF Jr. Antioxidants and atherosclerotic heart
disease. N Engl J Med 1997; 337: 408–416.
100. Carr AC, Zhu BZ, Frei B. Potential antiatherogenic mechanisms of ascorbate
(vitamin C) and alpha-tocopherol (vitamin E). Circ Res 2000; 87: 349–354.
101. Heinecke JW. Is the emperor wearing clothes? Clinical trials of vitamin E and the
LDL oxidation hypothesis. Arterioscler Thromb Vasc Biol 2001; 21: 1261–1264.
102. Gale CR, Ashurst HE, Powers HJ, Martyn CN. Antioxidant vitamin status and
carotid atherosclerosis in the elderly. Am J Clin Nutr 2001 74: 402–408.
DK5836_C002.fm Page 55 Monday, May 22, 2006 12:37 PM
56 Nutrient–Drug Interactions
140. Sesso HD, Gaziano JM, Liu S, Buring JE. Flavonoid intake and the risk of
cardiovascular disease in women. Am J Clin Nutr 2003; 77: 1400–1408.
141. Kris-Etherton PM, Lichtenstein AH, Howard BV, Steinberg D, Witztum JL; for
the Nutrition Committee of the American Heart Association Council on Nutrition,
Physical Activity, and Metabolism. Antioxidant vitamin supplements and cardio-
vascular disease. Circulation 2004; 110: 637–641.
142. Gibbons RJ, Abrams J, Chatterjee K, Daley J, Deedwania PC, Douglas JS, Fer-
guson TB Jr, Fihn SD, Fraker TD Jr, Gardin JM, O’Rourke RA, Pasternak RC,
Williams SV, Gibbons RJ, Alpert JS, Antman EM, Hiratzka LF, Fuster V, Faxon
DP, Gregoratos G, Jacobs AK, Smith SC Jr; American College of Cardiology;
American Heart Association Task Force on Practice Guidelines. Committee on
the Management of Patients with Chronic Stable Angina. ACC/AHA 2002 guide-
line update for the management of patients with chronic stable angina – summary
article: a report of the American College of Cardiology/American Heart Associ-
ation Task Force on Practice Guidelines (Committee on the Management of
Patients with Chronic Stable Angina). Circulation 2003; 107: 149–158.
143. Joshipura KJ, Hu FB, Manson JE, Stampfer MJ, Rimm EB, Speizer FE, Golditz
G, Ascherio A, Rosner B, Spiegelman D, Willett WC. The effect of fruit and
vegetable intake on risk for coronary heart disease. Ann Intern Med 2001; 134:
1106–1114.
144. Hu FB. Plant-based foods and prevention of cardiovascular disease: an overview.
Am J Clin Nutr 2003; 78 (Suppl) 544S–551S.
145. Boyer J, Liu RH. Apple phytochemicals and their health benefits. Nutr J 2004; 3: 5.
146. Hu FB, Stampfer MJ, Manson JE, Rimm E, Colditz GA, Speier FE, Hennekens
CH, Willett WC. Dietary protein and risk of ischemic heart disease in women.
Am J Clin Nutr 1999; 70: 221–227.
147. Janikula M. Policosanol: a new treatment for cardiovascular disease? Altern Med
Rev 2002; 7: 203–217.
148. Urizar NL, Moore DD. Gugulipid: a natural cholesterol-lowering agent. Annu Rev
Nutr 2003; 23: 303–313.
149. Szapary PO, Wolfe ML, Bloedon LT, Cucchiara AJ, DerMarderosian AH, Cir-
igliano MD, Rader DJ. Guggulipid for the treatment of hypercholesterolemia: a
randomized controlled trial. JAMA 2003; 290: 765–772.
150. Journoud M, Jones PJ. Red yeast rice: a new hypolipidemic drug. Life Sci 2004;
74: 2675–2683.
151. Bailey DG, Malcolm J, Arnold O, Spence JD. Grapefruit juice–drug interactions.
Br J Clin Pharmacol 1998; 46: 101–110.
152. Harris RZ, Jang GR, Tsunoda S. Dietary effects on drug metabolism and transport.
Clin Pharmacokinet 2003; 42: 1071–1088.
153. Dahan A, Altman H. Food-drug interaction: grapefruit juice augments drug bio-
availability — mechanism, extent and relevance. Eur J Clin Nutr 2004; 58: 1–9.
154. Kaminsky LS, Zhang QY. The small intestine as a xenobiotic-metabolizing organ.
Drug Metab Dispos 2003; 31: 1520–1525.
155. Dietrich CG, Geier A, Oude Elferink RP. ABC of oral bioavailability: transporters
as gatekeepers in the gut. Gut 2003; 52: 1788–1795.
156. Zhang QY, Dunbar D, Ostrowska A, Zeisloft S, Yang J, Kaminsky LS. Charac-
terization of human small intestinal cytochromes P-450. Drug Metab Dispos 1999;
27: 804–809.
DK5836_C002.fm Page 58 Monday, May 22, 2006 12:37 PM
58 Nutrient–Drug Interactions
60 Nutrient–Drug Interactions
3 Phytochemicals,
Xenobiotic Metabolism,
and Carcinogenesis
James B. Kirkland
CONTENTS
3.1 Introduction................................................................................................63
3.2 Dietary Carcinogens and Xenobiotic Metabolism....................................64
3.3 Animal-Based Research on Cooked Meat Carcinogens...........................68
3.4 Metabolism of Dietary Secondary Carcinogens .......................................68
3.4.1 Metabolism of NAs .......................................................................68
3.4.2 Metabolism of PAH and HCA Families .......................................69
3.4.3 Metabolism of PAHs .....................................................................69
3.4.4 Metabolism of HCAs ....................................................................70
3.5 Aflatoxin and Food Spoilage.....................................................................70
3.6 Effects of Carcinogen Exposure on Xenobiotic Metabolism...................70
3.7 Diet as a Source of Chemopreventative Agents .......................................73
3.8 Phytochemical Activation of the AhR.......................................................76
3.9 Phytochemical Antagonism of the AhR....................................................77
3.10 Phytochemical Inhibition of the Catalytic Activity of P450 ....................79
3.11 Phytochemical Induction of Phase II Enzymes ........................................80
3.12 Experimental Models for Future Work and Opportunities for
Improving Public Health ...........................................................................81
3.13 Public Perception of Risk and Resulting Behavior ..................................83
3.14 Conclusions................................................................................................85
References ...........................................................................................................86
3.1 INTRODUCTION
Nitrite preservatives in processed meats lead to the formation of carcinogenic
nitrosamines (NAs), while smoking and cooking of meats leads to the formation
of polycyclic aromatic hydrocarbons (PAHs) and heterocyclic amines (HCAs).
Cigarette smoke is also a potent source of NA, PAH, and HCA carcinogens.
Conversely, consumption of vegetables, spices, and green tea are associated with
63
DK5836_C003.fm Page 64 Thursday, May 25, 2006 12:52 PM
64 Nutrient–Drug Interactions
decreased cancer risk, and many phytochemicals from these sources are proven
chemoprotective agents in animal models. NAs, PAHs, and HCAs are stable
secondary carcinogens, which are “bioactivated” by P450-based Phase I metab-
olism to form reactive, ultimate carcinogens. This is also required for effective
metabolism, as it allows them to be conjugated (Phase II metabolism) and
excreted, preventing the accumulation of parent compounds in areas like the
adipose and brain tissue. Thus, the goal in cancer prevention is to find the optimal
balance between Phase I and II activities, which allow clearance, while minimiz-
ing the accumulation of reactive intermediates. Smoking and regular consumption
of cooked meats are negative influences, as P450 enzymes are induced and
bioactivation becomes too rapid for Phase II reactions to keep pace. Chemopro-
tective phytochemicals improve the metabolic balance by antagonizing P450
induction, by competitively inhibiting P450 enzymes, and by inducing the expres-
sion of Phase II enzymes. Thus, phytochemical intake can dramatically alter the
carcinogenicity of a given exposure to dietary carcinogens.
FIGURE 3.1 Examples of the three major classes of dietary secondary carcinogens: (A)
Benzo(a)pyrene (BaP), example of a polycyclic aromatic hydrocarbon (PAH), formed
through condensation of small radicals during the incomplete combustion of any burnable
substrate, requiring temperatures above 250°C. (B) 2-amino-3,4-methylimidazo[4,5-
f]quinoline (MeIQ), example of a heterocyclic amine (HCA), formed through condensation
of creatinine, amino acids, and sugars, across a broad range of meat cooking conditions,
enhanced by drying, as seen in the formation of gravy and jerky products. (C) Dimeth-
ylnitrosamine (DMN), example of a nitrosamine (NA), formed through reaction of nitrite
with secondary amines found in preserved meats and seafoods, also central to cigarette
smoke carcinogenesis in the formation nicotine/nitrite reaction products (e.g., NNK).
levels of fat. Animal models making use of practical foods, rather than single
purified carcinogens, will eventually provide unbiased data on these factors. Liver
cancer is associated with alcohol abuse in developed countries and hepatitis B
and aflatoxin exposure in developing countries.
Many of these negative risk factors can be explained by the exposure to
secondary carcinogens. Our major dietary carcinogens are all secondary carcin-
ogens, which are very stable, lipophilic compounds, generally formed during food
processing and cooking.16 The main categories of carcinogens in cooked and
preserved meats are polycyclic aromatic hydrocarbons (PAHs), heterocyclic
amines (HCAs) and nitrosamines (NAs)16 (Figure 3.1). PAHs, like benzo(a)pyrene
(BaP), are formed when material is partially burned, during smoking, grilling,
and barbequing. PAHs will form from any substrate, but flame grilling of high
fat foods is an excellent example.17 PAHs are also formed in the smoke of burning
wood chips and they dissolve into meats during the smoking process.17,18 BaP is
the main focus of regulatory efforts, limited to 5 µg/kg in smoked meats in the
European Community. This is despite the fact that BaP is a relatively small and
variable proportion of total PAH load in cooked or smoked food.18
HCAs are formed when meats are cooked at below carbonization tempera-
tures, as creatinine reacts with various amino acids to form products like 2-amino-
1-methyl-6-phenylimidazo-[4,5-b]-pyridine (PhIP), 2-amino-3,8-dimethylimi-
dazo[4,5-f]quinoxaline (MeIQx), and 2-amino-3-methylimidazo[4,5-f]quinoline
(IQ).3 Sugars may enhance this process when they are added to marinades or
during the processing of beef jerky.19 During the grilling of meat, higher temper-
ature cooking to the same final internal temperature produces significantly higher
levels of HCAs.3 Pan residues used in the production of gravy and food flavor
products tend to have higher levels of HCAs than the meat itself, especially if
the residue dried under heat. This concentrates the reactants and causes large
increases in cooking temperature.20 The recovery of free HCAs from cooked meat
DK5836_C003.fm Page 66 Thursday, May 25, 2006 12:52 PM
66 Nutrient–Drug Interactions
FIGURE 3.2 Secondary carcinogen bioactivation and conjugation. Stable secondary car-
cinogens require metabolism to prevent accumulation, leading to the formation of reactive
intermediates that cause DNA damage and cancer. These can be safely metabolized to
water-soluble conjugates, if the balance of Phase I and II enzymes and conjugating agents
is favorable.
68 Nutrient–Drug Interactions
tissues, and ethanol also causes cytotoxicity and cell division, which may also
increase cancer risk. There is a dramatic synergism between ethanol consumption
and smoking with respect to head and neck cancer risk.28 The smoke-specific
nitrosamine NNK is bioactivated by various P450 enzymes, including CYP2E1
and CYP2A family enzymes.35
NAs are metabolized by P450 to unstable intermediates that break down
spontaneously to form alkyldiazonium ions, which are the ultimate carcinogen
species.36 These are short-lived and Phase II metabolism of these intermediates
is not a popular research area, but they are electrophilic species and the majority
of conjugation occurs with glutathione. Electrophiles are species with a net
positive charge or a local area of positive charge that can react with other
molecules. Glutathione contains a sulfhydryl group with a high electron density
that allows it to react with electrophilic intermediates, preventing them from
alkylating cellular proteins and DNA. This Phase II conjugation is enhanced by
several forms of glutathione-S-transferase, an enzyme with high activity in tissues
that have to metabolize xenobiotics, especially the liver. Oral support of glu-
tathione via N-acetylcysteine supplementation decreased the induction of esoph-
ageal cancer in rats following diethylnitrosamine treatment.37
70 Nutrient–Drug Interactions
FIGURE 3.3 Cytochrome P450 activity and induction. Cytochrome P450 activity is lim-
ited to the catalytic cycle in the upper left of the diagram. Induction patterns, of both Phase
I and II enzymes, play a critical role in the potency of dietary carcinogens. *Xenobiotics
bind to specific receptors, which then act as transcription factors to induce the synthesis
of new P450 enzymes, usually with a high capacity to metabolize that specific xenobiotic.
**Phytochemicals tend to compete directly at the P450 active site to slow carcinogen
bioactivation. Competition also takes place at the AhR, where phytochemicals can decrease
the induction of P450 expression caused by secondary carcinogens. Large exposure to
phytochemicals can activate the AhR, however, leading to procarcinogenic effects.
***Phytochemicals induce multiple forms of Phase II conjugation enzymes and agents.
DK5836_C003.fm Page 72 Thursday, May 25, 2006 12:52 PM
72 Nutrient–Drug Interactions
74 Nutrient–Drug Interactions
One of the basic assumptions driving these clinical trials was that lung cancer
is caused by oxidant stress. It has subsequently been shown that the mutations
present in smoke-induced cancers are not similar to those caused by oxidant
stress. Instead, they are similar to those caused by the two major classes of
secondary carcinogens present in smoke, namely PAHs and smoke-specific nit-
rosamines, like NNK.67 As described earlier in this chapter, PAHs and NAs are
secondary carcinogens that require bioactivation by endogenous P450 enzymes,
and the resulting reactive intermediates are not detoxified by enzymes involved
in reactive oxygen metabolism. Animal models of lung cancer induced by these
classes of chemicals have shown that various phytochemicals, and other agents
that influence Phase I and II metabolism, have potent effects on lung cancer
incidence.68 Conversely, beta-carotene use in animal models has demonstrated
pro-oxidant effects69 and induction of P450 enzymes, which could enhance car-
cinogen bioactivation.70
Similar to the lung cancer situation, the incidence of GI cancers is associated
with the exposure to secondary carcinogens (consumption of well-done and
preserved meats) along with low vegetable and fruit consumption. Experiments
on phytochemical fractions from sources such as vegetables (glucosinolates68),
spices (curcumin71) and green tea (catechins like ECGC72) have shown that these
are very active in the prevention of chemically-induced cancers.73 Several types
of cancers (e.g., GI, lung) are about 50% lower in the upper quartile of vegetable
consumers.74 This relationship is well explained by metabolic interactions
between vegetable phytochemicals and dietary secondary carcinogens.73 Changes
in Phase I and II metabolism largely determine cancer risk from a given exposure
to secondary carcinogens. Chronic exposure to carcinogens alone causes binding
and activation of the aryl hydrocarbon receptor (AhR),45 and other related recep-
tors, which cause a dramatic upregualtion of specific P450 isozymes, like
CYP1A1/246 (Figure 3.2). This increases the rate of formation of reactive inter-
mediates and dramatically increases cancer risk from a given exposure to sec-
ondary carcinogens.25,75 In contrast, the impact of phytochemical exposure on
Phase I and II metabolism is complex, including the following effects:
76 Nutrient–Drug Interactions
78 Nutrient–Drug Interactions
kaempferol, and green tea extract weakly enhanced the AhR function when used
alone (two- to sixfold at 10 µM), and green tea extract, naringenin, and apigenin
significantly antagonized the strong TCDD-induced activation of AhR (10-fold
at 1 nM).91 Curcumin and TCDD produced an additive effect on AhR activation.
Amakura et al. found that green tea catechins and extracts of sage, spinach, and
citrus fruits produced antagonism of TCDD-induced AhR activation.92 In other
studies, I3C and DIM, kaemferol, quercitin, myricetin, and luteolin antagonized
TCDD-induce AhR activation.50,89
Green tea components are the best characterized phytochemicals with respect
to AhR interactions. Individual catechins have shown lower antagonism than
green tea extracts.91 Williams et al. found that a mixture of the four major
catechins displayed a strong antagonism similar to whole tea extract,93 while
Fukuda et al. showed that tea-derived lutein and chlorophyll a and b are also
antagonists of AhR activation.94 Inconsistent responses to epigallocatechin-3-
gallate (EGCG), a major tea catechin, may be explained by the finding that this
phytochemical does not compete at the binding site of the AhR, but appears to
inhibit the conformation changes subsequent to binding, through interactions with
accessory proteins.95 Black tea theaflavins also suppress the transformation of
TCDD-bound AhR to a transcriptionally active form.96
The importance of these observations is that, while many phytochemicals
alone are weak agonists of the AhR, they are also antagonists of the strong
activation of the AhR by compounds like TCDD and planar carcinogens. In a
typical human diet, these compounds will be presented together and the effect of
phytochemicals may be to modulate the AhR activation to a safer level, limiting
the degree of P450 induction and encouraging a safe balance between Phase I
and II metabolism. This has been verified by an elegant series of experiments in
a physiologically relevant rat model by Thapliyal et al.79,97,98 This model is based
on the prevention of BaP and DMN-induced carcinogenesis by the root spice
turmeric, in which the active ingredient is curcumin (diferuloylmethane). In this
model, dietary turmeric provided dramatic protection against the formation of
BaP adducts in liver, lung, and stomach tissues when consumed for 2 weeks
preceding the dose of BaP, but not when the turmeric was introduced after the
BaP dose.98 DMN-induced cancer of the liver was also decreased by ongoing
turmeric consumption, and not by turmeric following DMN treatment.97 Thus,
the effects in this model are mainly in the bioactivation and initiation period as
has been discussed in this chapter. Further experiments showed that dietary
turmeric alone had variable effects on P450 enzymes in the liver, lung, and GI
tract, from small depressions to modest inductions,79 as described in Section 3.8.
When present in the diet during exposure to BaP, turmeric decreased the large
induction of CYP1A1, 1A2, and 2B1 by 10 to 80%.79 These studies also dem-
onstrated direct inhibition of the catalytic activity of the P450 enzymes and the
induction of Phase II enzymes, as will be discussed below.
DK5836_C003.fm Page 79 Thursday, May 25, 2006 12:52 PM
80 Nutrient–Drug Interactions
82 Nutrient–Drug Interactions
mutagenic in vitro and carcinogenic to rodents in large single dose models, but
these lack physiological relevance to human exposures. The literature also lacks
data on the effects of chronic exposure to low levels of complex mixtures of
PAHs, HCAs, and NAs. Once a model for this is established, it may be used to
examine cancer incidence, short-term biomarkers of exposure and injury, and the
protective impact of plant products and phytochemical fractions.
In 2003, American consumers spent $1.8 billion on hot dogs in supermarkets
alone128 and much more at locations outside the home ($24.2 million in major
league baseball parks128). Hot dogs are representative of a larger group of cured
meats, including luncheon meats, bacon, and sausages. Norat et al. found that
the 70th percentile of processed meat intake was around 40 g/d, while the 90th
percentile was over 80 g/d,5 representing 1 to 2 meals per day. While processed
meats are often viewed as sources of NA exposure, these products are also smoked
(causing PAH formation) and many are slow cooked to a low-moisture content
(causing HCA formation). Children represent a high-risk group that consumes
even greater levels of these products on a body weight basis, and at a young age
are more susceptible to carcinogen exposure.
Consumers remain poorly informed about meat intake and cancer. Red and
processed meat intake is not diminishing and barbequing appears to be increasing
in popularity.129 Interactions between phytochemicals and secondary carcinogens
represent a potent approach to decrease the cancer risks associated with cooked
meats. These phytochemicals can be introduced into processed meats, creating
functional foods that play a role in disease prevention. Green tea is a promising
source of active phytochemicals for this project. Green tea consumption is asso-
ciated with a decreased risk of several human cancers,130 and numerous animal-
based experiments have shown that green tea phytochemical fractions and purified
catechins inhibit high-dose PAH, HCA, and NA-induced cancers.72 As mentioned
earlier, PAHs, HCAs, and NAs are all secondary carcinogens, requiring bioactiva-
tion by P450 enzymes to cause DNA damage. There are about 60 different P450
gene products, with complex regulation in response to exposure to dietary car-
cinogens and phytochemicals.16 This regulation occurs through receptors, like the
AhR, which bind foreign compounds and become transcription factors that lead
to increased expression of P450 proteins. The AhR is very active in the induction
of CYP1A1/2 genes, which form P450 enzymes that bioactivate PAHs25 and
HCAs.75 In human experiments, regular consumption of well-done meat increases
intestinal CYP1A activity,46 an effect that will increase cancer risk from a given
carcinogen exposure. Basic research has demonstrated that green tea phytochem-
icals compete with carcinogens for binding to the AhR, and lessen the degree of
P450 induction caused by carcinogens alone.78 In addition, these phytochemicals
compete directly at the active site of the P450 enzyme, decreasing the rate of
production of reactive intermediates from carcinogens.82 Lastly, green tea phy-
tochemicals induce the expression of Phase II enzymes and conjugating agents.48
The balance of these phytochemical effects is slower production and more rapid
conjugation of reactive intermediates, decreasing the accumulation of harmful
intermediates, DNA adducts, mutations, and cancer.
DK5836_C003.fm Page 83 Thursday, May 25, 2006 12:52 PM
84 Nutrient–Drug Interactions
FIGURE 3.4 Risk models for extrapolation of cancer data. The top panel shows risk
models used by toxicologists and public health policy makers. The bottom panel shows
risk models perceived by the public, in which much higher risk is attached to artificial
components in the food supply, while natural products are perceived as increasingly
beneficial at high exposures, in the form of natural health products.
Baldwin have examined published data for evidence of hormesis in various areas
of toxicology and found that close to 10% of toxicological studies demonstrate
hormesis, although few are properly designed to do so. Among the different
categories of compounds, metals are very likely to produce hormetic dose
response curves, with herbicides, pesticides, insecticides, and hydrocarbons aver-
aging around 5% of studies. Hormesis is poorly dealt with by health policy and
basically incomprehensible in the forum of public risk perception.
The last curve to consider is the public perception curve, where a food additive
or residue shown to cause cancer in rodents at high doses is considered to be as
dangerous, or more so, in trace amounts in human populations. This is the top
curve in the lower panel, and represents the illogical public perception of pesticide
residues and additives like aspartame. To gain an accurate working knowledge
of diet and cancer, the public has to stop considering natural products as healthy
and artificial products as dangerous. They share common pathways of
DK5836_C003.fm Page 85 Thursday, May 25, 2006 12:52 PM
metabolism, and many of the benefits of phytochemicals are due to the fact that
they are xenobiotics and they interact directly with more potent carcinogens in
Phase I and II metabolism. Some of the problems resulting from public perception
of risk are summarized below.
One of the strongest public fears is of pesticide residues in food, which is
often focused on fruits and vegetables. This may lead to decreased intake through
avoidance or due to the higher cost of organic products. Insufficient intake of
fruits and vegetables is the major nutritional problem in developed countries. A
low intake of fruits and vegetables may lead to increased meat intake, with
associated exposure to carcinogens produced by cooking and preservation.
Producers of natural health products are taking advantage of the public desire
to attain the benefits of fruit and vegetable consumption without incurring the
risk of pesticide exposure or the basic lack of desire to eat these foods. This leads
us to the final curve shown in Figure 3.4. Indole-3-carbinol is a breakdown product
of brassica glucosinolates, known to be associated with decreased cancer risk in
human populations (through consumption of brassica vegetables) and shown to
prevent cancer, at low levels, in rodent models. An unbiased observer might place
this data point in the lowest region of a hormetic response curve and expect that
at high levels the health benefits would diminish and the compound would
eventually become toxic. This follows the basic assumptions of toxicology that
all substances become toxic if the dose is increased sufficiently (Paracelsus, 1493-
1541). This has been established for indole-3-carbinol, which is shown to promote
cancer development at high doses in rodent models. The public views this natural
health product in a different light, however, and assumes that the benefits will
increase with increasing intake, as shown in the bottom curve in the lower panel
of Figure 3.4. Indole-3-carbinol is currently marketed in 200 mg capsules, with
recommended intakes of 2 per day, equivalent to 30 pounds of cabbage per day.
The justification for this extremely high intake by marketers of these compounds
uses data from the regression of cervical dysplasia, which is actually an example
of chemotherapy-like treatment of a dividing cell population, and in themselves
could be interpreted as a toxic end point. This chemotherapy-like dose of I3C is
being recommended and marketed to an unsuspecting public.
3.14 CONCLUSIONS
Current dietary patterns in developed countries favor exposure to carcinogens
derived from meat processing and cooking techniques, and dietary habits have
decreased the intake of chemopreventative phytochemicals. The growth of
research, business, and public interest in functional foods and nutraceuticals
provides opportunities to bring some of the beneficial metabolic effects back to
the modern, highly processed diet. This movement also increases the risk of
toxicity due to unnaturally high exposures to natural xenobiotics. Further research
will help to maximize the benefits and minimize the risks associated with the
novel use of natural products in cancer prevention.
DK5836_C003.fm Page 86 Thursday, May 25, 2006 12:52 PM
86 Nutrient–Drug Interactions
REFERENCES
1. Ames BN, Gold LS. Environmental pollution, pesticides, and the prevention of
cancer: misconceptions. FASEB J. 1997; 11: 1041–52.
2. Parkin DM. Global cancer statistics in the year 2000. Lancet Oncol. 2001; 2:
533–43.
3. Knize MG, Felton JS. Formation and human risk of carcinogenic heterocyclic
amines formed from natural precursors in meat. Nutr. Rev. 2005; 63: 158–65.
4. Chao A, Thun MJ, Connell CJ, et al. Meat consumption and risk of colorectal
cancer. JAMA 2005; 293: 172–82.
5. Norat T, Bingham S, Ferrari P, et al. Meat, fish, and colorectal cancer risk: the
European Prospective Investigation into cancer and nutrition. J. Natl. Cancer Inst.
2005; 97: 906–16.
6. Sinha R, Chow WH, Kulldorff M, et al. Well-done, grilled red meat increases the
risk of colorectal adenomas. Cancer Res. 1999; 59: 4320–4.
7. Peters JM, Preston-Martin S, London SJ, Bowman JD, Buckley JD, Thomas DC.
Processed meats and risk of childhood leukemia (California). Cancer Causes
Control 1994; 5: 195–202.
8. Levi F, Pasche C, Lucchini F, Bosetti C, La Vecchia C. Processed meat and the
risk of selected digestive tract and laryngeal neoplasms in Switzerland. Ann.
Oncol. 2004; 15: 346–9.
9. Armstrong RW, Imrey PB, Lye MS, Armstrong MJ, Yu MC, Sani S. Nasopharyn-
geal carcinoma in Malaysian Chinese: salted fish and other dietary exposures. Int.
J. Cancer 1998; 77: 228–235.
10. Ke L, Yu P, Zhang ZX. Novel epidemiologic evidence for the association between
fermented fish sauce and esophageal cancer in South China. Int. J. Cancer 2002;
99: 424–426.
11. Ngoan LT, Mizoue T, Fujino Y, Tokui N, Yoshimura T. Dietary factors and stomach
cancer mortality. Br. J. Cancer 2002; 87: 37–42.
12. Jian L, Zhang DH, Lee AH, Binns CW. Do preserved foods increase prostate
cancer risk? Br. J. Cancer 2004; 90: 1792–1795.
13. Dietrich M, Block G, Pogoda JM, Buffler P, Hecht S, Martin SP. A review: dietary
and endogenously formed N-nitroso compounds and risk of childhood brain
tumors. Cancer Causes Control 2005; 16: 619–635.
14. Sinha R. An epidemiologic approach to studying heterocyclic amines. Mutat. Res.
2002; 506-507: 197–204.
15. Sinha R, Cross A, Curtin J, et al. Development of a food frequency questionnaire
module and databases for compounds in cooked and processed meats. Mol. Nutr.
Food Res. 2005; 49: 648–655.
16. Jagerstad M, Skog K. Genotoxicity of heat-processed foods. Mutat. Res. 2005;
574: 156–172.
17. Lijinsky W. The formation and occurrence of polynuclear aromatic hydrocarbons
associated with food. Mutat. Res. 1991; 259: 251–261.
18. Simko P. Factors affecting elimination of polycyclic aromatic hydrocarbons from
smoked meat foods and liquid smoke flavorings. Mol. Nutr. Food Res. 2005; 49:
637–647.
19. Nerurkar PV, Le Marchand L, Cooney RV. Effects of marinating with Asian
marinades or western barbecue sauce on PhIP and MeIQx formation in barbecued
beef. Nutr. Cancer 1999; 34: 147–152.
DK5836_C003.fm Page 87 Thursday, May 25, 2006 12:52 PM
20. Laser RA, Skog K, Jagerstad M. Effects of creatine and creatinine content on the
mutagenic activity of meat extracts, bouillons and gravies from different sources.
Food Chem. Toxicol. 1987; 25: 747–754.
21. Martin FL, Cole KJ, Phillips DH, Grover PL. The proteolytic release of genotoxins
from cooked beef. Biochem. Biophys. Res. Commun. 2002; 293: 1497–1501.
22. Choi SY, Chung MJ, Sung NJ. Volatile N-nitrosamine inhibition after intake
Korean green tea and Maesil (Prunus mume SIEB. et ZACC.) extracts with an
amine-rich diet in subjects ingesting nitrate. Food Chem. Toxicol. 2002; 40:
949–957.
23. Dietrich M, Block G, Pogoda JM, Buffler P, Hecht S, Preston-Martin S. A review:
dietary and endogenously formed N-nitroso compounds and risk of childhood
brain tumors. Cancer Causes Control 2005; 16: 619–635.
24. Wogan GN, Hecht SS, Felton JS, Conney AH, Loeb LA. Environmental and
chemical carcinogenesis. Semin. Cancer Biol. 2004; 14: 473–486.
25. Xue W, Warshawsky D. Metabolic activation of polycyclic and heterocyclic aro-
matic hydrocarbons and DNA damage: a review. Toxicol. Appl. Pharmacol. 2005;
206: 73–93.
26. Blanck A, Lindhe B, Porsch H, I, Lindeskog P, Gustafsson JA. Influence of
different levels of dietary casein on initiation of male rat liver carcinogenesis with
a single dose of aflatoxin B1. Carcinogenesis 1992; 13: 171–176.
27. Mori Y, Koide A, Kobayashi Y, Morimura K, Kaneko M, Fukushima S. Effect of
ethanol treatment on metabolic activation and detoxification of esophagus carci-
nogenic N-nitrosamines in rat liver. Mutagenesis 2002; 17: 251–256.
28. Castellsague X, Quintana MJ, Martinez MC, et al. The role of type of tobacco
and type of alcoholic beverage in oral carcinogenesis. Int. J. Cancer 2004; 108:
741–749.
29. Marzullo L. An update of N-acetylcysteine treatment for acute acetaminophen
toxicity in children. Curr. Opin. Pediatr. 2005; 17: 239–245.
30. Cross AJ, Sinha R. Meat-related mutagens/carcinogens in the etiology of colorec-
tal cancer. Environ. Mol. Mutagen. 2004; 44: 44–55.
31. Mirvish SS, Haorah J, Zhou L, Clapper ML, Harrison KL, Povey AC. Total N-
nitroso compounds and their precursors in hot dogs and in the gastrointestinal
tract and feces of rats and mice: possible etiologic agents for colon cancer. J. Nutr.
2002; 132: 3526S–3529S.
32. Pence BC, Landers M, Dunn DM, Shen CL, Miller MF. Feeding of a well-cooked
beef diet containing a high heterocyclic amine content enhances colon and stomach
carcinogenesis in 1,2-dimethylhydrazine-treated rats. Nutr. Cancer 1998; 30:
220–226.
33. Heddle JA, Knize MG, Dawod D, Zhang XB. A test of the mutagenicity of cooked
meats in vivo. Mutagenesis 2001; 16: 103–107.
34. Shen CL, Purewal M, San Francisco S, Pence BC. Absence of PhIP adducts, p53
and Apc mutations, in rats fed a cooked beef diet containing a high level of
heterocyclic amines. Nutr. Cancer 1998; 30: 227–231.
35. Smith GB, Bend JR, Bedard LL, Reid KR, Petsikas D, Massey TE. Biotransfor-
mation of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK) in peripheral
human lung microsomes. Drug Metab. Dispos. 2003; 31: 1134–1141.
36. Teiber JF, Mace K, Hollenberg PF. Metabolism of the beta-oxidized intermediates
of N-nitrosodi-n-propylamine: N-nitroso-beta-hydroxypropylpropylamine and N-
nitroso-beta-oxopropylpropylamine. Carcinogenesis 2001; 22: 499–506.
DK5836_C003.fm Page 88 Thursday, May 25, 2006 12:52 PM
88 Nutrient–Drug Interactions
53. Sinha R, Rothman N, Brown ED, et al. Pan-fried meat containing high levels of
heterocyclic aromatic amines but low levels of polycyclic aromatic hydrocarbons
induces cytochrome P4501A2 activity in humans. Cancer Res. 1994; 54:
6154–6159.
54. McCullough ML, Robertson AS, Chao A, et al. A prospective study of whole
grains, fruits, vegetables and colon cancer risk. Cancer Causes Control 2003; 14:
959–970.
55. Omenn GS, Goodman GE, Thornquist MD, et al. Risk factors for lung cancer and
for intervention effects in CARET, the beta-carotene and retinol efficacy trial. J.
Natl. Cancer Inst. 1996; 88: 1550–1559.
56. Decker EA, Warner K, Richards MP, Shahidi F. Measuring antioxidant effective-
ness in food. J. Agric. Food Chem. 2005; 53: 4303–4310.
57. Aviram M, Dornfeld L, Kaplan M, et al. Pomegranate juice flavonoids inhibit
low-density lipoprotein oxidation and cardiovascular diseases: studies in athero-
sclerotic mice and in humans. Drugs Exp. Clin. Res. 2002; 28: 49–62.
58. Natella F, Belelli F, Gentili V, Ursini F, Scaccini C. Grape seed proanthocyanidins
prevent plasma postprandial oxidative stress in humans. J. Agric. Food Chem.
2002; 50: 7720–7725.
59. Marin E, Kretzschmar M, Arokoski J, Hanninen O, Klinger W. Enzymes of
glutathione synthesis in dog skeletal muscles and their response to training. Acta
Physiol. Scand. 1993; 147: 369–373.
60. Carlsen H, Myhrstad MC, Thoresen M, Moskaug JO, Blomhoff R. Berry intake
increases the activity of the gamma-glutamylcysteine synthetase promoter in trans-
genic reporter mice. J. Nutr. 2003; 133: 2137–2140.
61. Myhrstad MC, Carlsen H, Nordstrom O, Blomhoff R, Moskaug JO. Flavonoids
increase the intracellular glutathione level by transactivation of the gamma-
glutamylcysteine synthetase catalytical subunit promoter. Free Radic. Biol. Med.
2002; 32: 386–393.
62. Gunduz F, Senturk UK, Kuru O, Aktekin B, Aktekin MR. The effect of one year’s
swimming exercise on oxidant stress and antioxidant capacity in aged rats. Physiol.
Res. 2004; 53: 171–176.
63. Metin G, Gumustas MK, Uslu E, Belce A, Kayserilioglu A. Effect of regular
training on plasma thiols, malondialdehyde and carnitine concentrations in young
soccer players. Chin. J. Physiol. 2003; 46: 35–39.
64. Virtamo J, Pietinen P, Huttunen JK, et al. Incidence of cancer and mortality
following alpha-tocopherol and beta-carotene supplementation: a postintervention
follow-up. JAMA 2003; 290: 476–485.
65. Goodman GE, Thornquist MD, Balmes J, et al. The beta-carotene and retinol
efficacy trial: incidence of lung cancer and cardiovascular disease mortality during
6-year follow-up after stopping beta-carotene and retinol supplements. J. Natl.
Cancer Inst. 2004; 96: 1743–1750.
66. Albanes D, Heinonen OP, Huttunen JK, et al. Effects of alpha-tocopherol and
beta-carotene supplements on cancer incidence in the Alpha-Tocopherol Beta-
Carotene Cancer Prevention Study. Am. J. Clin. Nutr. 1995; 62: 1427S–1430S.
67. Pfeifer GP, Denissenko MF, Olivier M, Tretyakova N, Hecht SS, Hainaut P.
Tobacco smoke carcinogens, DNA damage and p53 mutations in smoking-asso-
ciated cancers. Oncogene 2002; 21: 7435–7451.
68. Stoner GD, Morse MA. Isothiocyanates and plant polyphenols as inhibitors of
lung and esophageal cancer. Cancer Lett. 1997; 114: 113–119.
DK5836_C003.fm Page 90 Thursday, May 25, 2006 12:52 PM
90 Nutrient–Drug Interactions
69. El Agamey A, Lowe GM, McGarvey DJ, et al. Carotenoid radical chemistry
and antioxidant/pro-oxidant properties. Arch. Biochem. Biophys. 2004; 430:
37–48.
70. Paolini M, Antelli A, Pozzetti L, et al. Induction of cytochrome P450 enzymes
and over-generation of oxygen radicals in beta-carotene supplemented rats. Car-
cinogenesis 2001; 22: 1483–1495.
71. Duvoix A, Blasius R, Delhalle S, et al. Chemopreventive and therapeutic effects
of curcumin. Cancer Lett. 2005; 223: 181–190.
72. Crespy V, Williamson G. A review of the health effects of green tea catechins in
in vivo animal models. J. Nutr. 2004; 134: 3431S–3440S.
73. van Iersel ML, Verhagen H, van Bladeren PJ. The role of biotransformation in
dietary (anti)carcinogenesis. Mutat. Res. 1999; 443: 259–270.
74. Food, Nutrition and the Prevention of Cancer: a Global Perspective. Washington,
DC: American Institute for Cancer Research (AICR), 1997.
75. Kim D, Guengerich FP. Cytochrome P450 activation of arylamines and heterocy-
clic amines. Annu. Rev. Pharmacol. Toxicol. 2005; 45: 27–49.
76. Kassie F, Uhl M, Rabot S, et al. Chemoprevention of 2-amino-3-methylimi-
dazo[4,5-f]quinoline (IQ)-induced colonic and hepatic preneoplastic lesions in the
F344 rat by cruciferous vegetables administered simultaneously with the carcin-
ogen. Carcinogenesis 2003; 24: 255–261.
77. Steinkellner H, Rabot S, Freywald C, et al. Effects of cruciferous vegetables and
their constituents on drug metabolizing enzymes involved in the bioactivation of
DNA-reactive dietary carcinogens. Mutat. Res. 2001; 480–481: 285–97.
78. Williams SN, Shih H, Guenette DK, et al. Comparative studies on the effects of
green tea extracts and individual tea catechins on human CYP1A gene expression.
Chem. Biol. Interact. 2000; 128: 211–229.
79. Thapliyal R, Maru GB. Inhibition of cytochrome P450 isozymes by curcumins in
vitro and in vivo. Food Chem. Toxicol. 2001; 39: 541–547.
80. Barcelo S, Gardiner JM, Gescher A, Chipman JK. CYP2E1-mediated mechanism
of anti-genotoxicity of the broccoli constituent sulforaphane. Carcinogenesis
1996; 17: 277–282.
81. Smith TJ, Guo Z, Guengerich FP, Yang CS. Metabolism of 4-(methylnitrosamino)-
1-(3-pyridyl)-1-butanone (NNK) by human cytochrome P450 1A2 and its inhibi-
tion by phenethyl isothiocyanate. Carcinogenesis 1996; 17: 809–813.
82. Muto S, Fujita K, Yamazaki Y, Kamataki T. Inhibition by green tea catechins of
metabolic activation of procarcinogens by human cytochrome P450. Mutat. Res.
2001; 479: 197–206.
83. Thapliyal R, Deshpande SS, Maru GB. Effects of turmeric on the activities of
benzo(a)pyrene-induced cytochrome P-450 isozymes. J. Environ. Pathol. Toxicol.
Oncol. 2001; 20: 59–63.
84. Nakajima M, Yoshida R, Shimada N, Yamazaki H, Yokoi T. Inhibition and inac-
tivation of human cytochrome P450 isoforms by phenethyl isothiocyanate. Drug
Metab. Dispos. 2001; 29: 1110–1113.
85. Moskaug JO, Carlsen H, Myhrstad MC, Blomhoff R. Polyphenols and glutathione
synthesis regulation. Am. J. Clin. Nutr. 2005;81:277S-83S.
86. Bishayee A, Chatterjee M. Anticarcinogenic biological response of Mikania
cordata: reflections in hepatic biotransformation systems. Cancer Lett. 1994; 81:
193–200.
DK5836_C003.fm Page 91 Thursday, May 25, 2006 12:52 PM
87. Jellinck PH, Forkert PG, Riddick DS, Okey AB, Michnovicz JJ, Bradlow HL. Ah
receptor binding properties of indole carbinols and induction of hepatic estradiol
hydroxylation. Biochem. Pharmacol. 1993; 45: 1129–1136.
88. Sanderson JT, Slobbe L, Lansbergen GW, Safe S, van den BM. 2,3,7,8-tetrachlo-
rodibenzo-p-dioxin and diindolylmethanes differentially induce cytochrome P450
1A1, 1B1, and 19 in H295R human adrenocortical carcinoma cells. Toxicol. Sci.
2001; 61: 40–48.
89. Zhang S, Qin C, Safe SH. Flavonoids as aryl hydrocarbon receptor agonists/antag-
onists: effects of structure and cell context. Environ. Health Perspect. 2003; 111:
1877–1882.
90. Gross-Steinmeyer K, Stapleton PL, Liu F, et al. Phytochemical-induced changes
in gene expression of carcinogen-metabolizing enzymes in cultured human pri-
mary hepatocytes. Xenobiotica 2004; 34: 619–632.
91. Allen SW, Mueller L, Williams SN, Quattrochi LC, Raucy J. The use of a high-
volume screening procedure to assess the effects of dietary flavonoids on human
cyp1a1 expression. Drug Metab. Dispos. 2001; 29: 1074–1079.
92. Amakura Y, Tsutsumi T, Nakamura M et al. Preliminary screening of the inhibitory
effect of food extracts on activation of the aryl hydrocarbon receptor induced by
2,3,7,8-tetrachlorodibenzo-p-dioxin. Biol. Pharm. Bull. 2002; 25: 272–274.
93. Williams SN, Pickwell GV, Quattrochi LC. A combination of tea (Camellia
senensis) catechins is required for optimal inhibition of induced CYP1A expres-
sion by green tea extract. J. Agric. Food Chem. 2003; 51: 6627–6634.
94. Fukuda I, Sakane I, Yabushita Y, et al. Pigments in green tea leaves (Camellia
sinensis) suppress transformation of the aryl hydrocarbon receptor induced by
dioxin. J. Agric. Food Chem. 2004; 52: 2499–2506.
95. Palermo CM, Westlake CA, Gasiewicz TA. Epigallocatechin gallate inhibits aryl
hydrocarbon receptor gene transcription through an indirect mechanism involving
binding to a 90 kDa heat shock protein. Biochemistry 2005; 44: 5041–5052.
96. Fukuda I, Sakane I, Yabushita Y, Sawamura S, Kanazawa K, Ashida H. Black tea
theaflavins suppress dioxin-induced transformation of the aryl hydrocarbon recep-
tor. Biosci. Biotechnol. Biochem. 2005; 69: 883–890.
97. Thapliyal R, Naresh KN, Rao KV, Maru GB. Inhibition of nitrosodiethylamine-
induced hepatocarcinogenesis by dietary turmeric in rats. Toxicol. Lett. 2003; 139:
45–54.
98. Thapliyal R, Deshpande SS, Maru GB. Mechanism(s) of turmeric-mediated pro-
tective effects against benzo(a)pyrene-derived DNA adducts. Cancer Lett. 2002;
175: 79–88.
99. Bear WL, Teel RW. Effects of citrus flavonoids on the mutagenicity of heterocyclic
amines and on cytochrome P450 1A2 activity. Anticancer Res. 2000; 20:
3609–3614.
100. Bear WL, Teel RW. Effects of citrus phytochemicals on liver and lung cytochrome
P450 activity and on the in vitro metabolism of the tobacco-specific nitrosamine
NNK. Anticancer Res. 2000; 20: 3323–3329.
101. Henderson MC, Miranda CL, Stevens JF, Deinzer ML, Buhler DR. In vitro
inhibition of human P450 enzymes by prenylated flavonoids from hops, Humulus
lupulus. Xenobiotica 2000; 30: 235–251.
102. Hammons GJ, Fletcher JV, Stepps KR, et al. Effects of chemoprotective agents
on the metabolic activation of the carcinogenic arylamines PhIP and 4-aminobi-
phenyl in human and rat liver microsomes. Nutr. Cancer 1999; 33: 46–52.
DK5836_C003.fm Page 92 Thursday, May 25, 2006 12:52 PM
92 Nutrient–Drug Interactions
4 Nutrient and
Phytochemical
Modulation of Cancer
Treatment
Kelly Anne Meckling
CONTENTS
4.1 Introduction................................................................................................95
4.2 Magnitude of the Problem ........................................................................96
4.3 Effects of Overall Nutritional Status on Treatment Outcomes ................97
4.3.1 Changes in Nutritional Status of the Cancer Patient....................97
4.3.2 The Antioxidant Conundrum.......................................................101
4.4 Specific Molecular Targets ......................................................................102
4.4.1 P-Glycoprotein and Other Drug Efflux Pumps...........................102
4.4.2 NFκB, AP-1, and COX-2 ............................................................104
4.4.3 The Mevalonate Pathway ............................................................106
4.4.4 Immunomodulators......................................................................107
4.5 Polyunsaturated Fatty Acids as Therapeutic and Adjuvant
Chemotherapy Agents .............................................................................108
4.6 Other Foods, Phytochemicals, and Supplements Useful in
Adjuvant Chemotherapy..........................................................................113
4.7 Conclusions..............................................................................................115
References .........................................................................................................115
4.1 INTRODUCTION
Epidemiological data have consistently demonstrated that fruit and vegetable
consumption are associated with decreased risk of cancer at a variety of tissue
sites. A number of nutrients and phytochemical components of these foods have
been identified as potential cancer chemopreventive agents acting through a
variety of mechanisms (as described in Chapter 3). Many of these same com-
pounds or additional ones may also have activity in the later stages of cancer
95
DK5836_C004.fm Page 96 Thursday, May 25, 2006 12:56 PM
96 Nutrient–Drug Interactions
medicine (CAM) and half of these report this use to their treating physician. One-
third of Australian and Finnish women with breast cancer reported substantial
changes to their dietary and lifestyle habits in the months following diagnosis.6
These included smoking cessation, changes in macronutrient composition,
increasing physical activity, and the use of vitamin and herbal supplements as
well as visits to CAM practitioners, including naturopaths. The highest users of
CAM appear to be pediatric oncology patients (84%)7 followed by breast cancer
survivors with more than 66% of Canadian women, living in Ontario, reporting
use of CAM.5 Sixty-one percent of U.S. armed forces veterans diagnosed with
cancer admitted using dietary supplements8 and about 30% of Canadian men with
prostate cancer report using some form of alternative medicine, although only
one-third of these were seeing a CAM practitioner.9
While the move toward informing clinicians about the use of CAM therapies
may be increasing, there are still 30% or more of certain patient groups that fail
to report. In order to provide useful resources for patients and clinicians on
specific therapies, prevalence data and outcome measures need to be available.
Both positive and negative interactions of CAMs with other therapies, including
chemotherapy, need to be identified and reported by both traditional and alterna-
tive healthcare practitioners. If properly applied, diet, nutritional, and herbal
supplements could increase drug efficacy in cancer treatment and potentially
lessen drug toxicity, leading to increased cure and better quality of life for those
suffering from cancer.
98 Nutrient–Drug Interactions
TABLE 4.1
Nutritional Components with Potential Activity in Adjuvant Cancer
Therapy
Nutrient/
Phytochemical Molecular Target(s) or Activity Confounding Factors
itself or from therapies that affect tissue metabolism or the patient’s ability to
assimilate nutrients.10 As well as there being different incidence between cancer
types, there are also differences in the presentation and pattern of body com-
position changes between men and women.11 Regardless of treatment modality,
the incidence of deficiency signs and symptoms frequently increases as treat-
ment progresses.12 These can include the appearance of stomatitis, edema, and
changes in body composition. The anorexia-cachexia syndrome is typical of
patients suffering from many solid tumors and occurs in greater than 50% of
all cancer patients and more than 85% of pancreatic and lung cancer patients.13
Presence of cachexia is associated with increased morbidity and mortality, and
research efforts are focused on improving quality of life and functional capacity
for those suffering.14 Both fat stores (predominantly triglyceride) and skeletal
muscle protein are targets of the hypermetabolic state resulting from a number
of inflammatory mediator and cytokine changes, including release of lipid
mobilizing factor (LIF) and proteolysis inducing factor (PIF) from tumor cells
and other tissues.
Recent studies using oral nutritional supplements containing eicosapen-
taenoic acid (EPA, 20:5n-3) in cachectic patients have demonstrated improve-
ments in weight or decreases in the rate of weight loss, improvements in body
composition, functional status, and quality of life.15–18 Many of the early studies
focused on untreated pancreatic patients where effective therapy and survival are
limited. However, a recent pilot study demonstrated improved outcomes in lung
DK5836_C004.fm Page 100 Thursday, May 25, 2006 12:56 PM
effective therapies for mucositis. Most efforts have been on palliation and pain
control (for review, see Reference 25). One clinical trial demonstrated that sup-
plementation of parenteral nutrition formula with glutamine reduced the severity
of mucositis in patients undergoing high dose chemotherapy followed by autol-
ogous bone marrow transplantation.28 Another found that treatment with human
recombinant keratinocyte growth factor substantially improved oral mucositis
symptoms in patients about to undergo myeloablation and autologous stem cell
transplantation for hematologic malignancies.29 Though not specifically tested in
cancer patients, a traditional herbal medicine, Throat Coat®, containing elm bark,
licorice root, and marshmallow root, substantially improved symptoms of phar-
yngitis in clinical patients.30
burden of chemotherapy combined with the disease process itself would lead to
an increased free radical load and, therefore, tax the antioxidant defenses even
in the presence of apparent dietary adequacy. Given the inconsistencies between
reports and the large variation between specific patient populations, generaliza-
tions about the benefits and/or risks of antioxidant supplementation during che-
motherapy cannot be made. Rather, recommendations for their use must be based
on the specific drug regimen and tumor type being treated and a molecular
understanding of the relevant targets (see Table 4.1).
4.4.4 IMMUNOMODULATORS
Patients undergoing surgery, radiation, or chemotherapy have a high risk of
complications, including wound infections and septicemia. Alterations in host
defense associated with disease or treatment may make the cancer patient par-
ticularly vulnerable. Although there is no single contributor to immune dysfunc-
tion, there are indicators that specific nutritional substrates may be effective in
rebalancing the immune system in cancer patients. As already mentioned, the n-
3 fatty acid, EPA, has anticachectic properties that lead to decreased weight loss
in treated patients. Recent studies have shown that EPA and the amino acids,
arginine or glutamine, when supplemented orally, improved postoperative infec-
tious and wound complications in head and neck cancer97,98 pancreatic cancer,99
colon cancer100 patients, and during bone marrow transplantation.101
Recently, green tea polyphenols were also shown to promote cytotoxic CD8+
T cell invasion of UV-induced tumors and promoted apoptosis via activation of
caspase-3 in animals.102,103 As well, mistletoe lectins (ML-1) have considerable
immunomodulatory activity in human beings. Healthy human subjects adminis-
tered mistletoe lectins responded with an initial proliferation of peripheral blood
mononuclear cells, accompanied by increased production of TNFα and IL-6 and
less pronounced elevation of INF-γ and IL-4 (reviewed by Pryme et al.104).
Expansion in the CD8+ cytotoxic T cell population and NK activity have also
been reported in response to ML-1.105
Additional compounds that have interesting immunomodulatory activity
include a variety of polysaccharides from herbs used in traditional Chinese
medicine (TCM). There are important structure–function relationships that have
not been completely delineated but that seem critical to their activity (reviewed
by Chang106). The most important bioactives appear to have a β1,3 1,4 or 1,6
branching pattern, and the more highly branched and higher molecular weight
species seem to be the most potent.107 More than 200 species have been identified,
mostly fungal or botanicals. One representative example is the polysaccharide,
Lentinan, from Shiitake mushrooms, Lentinus edodes; it inhibits the growth of
transplanted tumors in rodents108 and, in a controlled clinical trial, it increased
survival in recurrent and metastatic gastric and colorectal cancer patients when
given in combination with chemotherapy.109 This latter study was carried out in
Japan, published in Japanese, and, as far as one can tell, has not been reproduced
in larger clinical trials. Thus, the utility of this agent remains to be examined.
DK5836_C004.fm Page 108 Thursday, May 25, 2006 12:56 PM
6-series 3-series
18:2n-6 18:3n-6
(linoleic acid, LA) alpha linolenic acid, LNA)
Δ6-desaturase
18:3n-6 18:4n-3
(gamma linolenic acid, GLA) (steariodonic acid)
elongase
20:3n-6 20:4n-3
(dihomo-gamma-linolenic acid, DGLA) eicosatetraenoic acid)
Δ5-desaturase
20:4n-6 20:5n-3
(arachidonic acid, AA) eicosapentaenoic acid, EPA)
elongase
22:5n-3
(docosapentaenoic acid, DPA)
elongase
24:5n-3
(tetracosapentaenoic acid, TPA)
Δ6-desaturase
24:6n-3
(tetracosahexaenoic acid, THA)
peroxisomal
β-oxidation 22:6n-3
(docosahexaenoic acid, DHA)
20:3n-6 20:5n-3
(DGLA) (EPA)
COX LOX COX LOX
20:4n-6
COX (AA) Cytochrome
P450
PGG2 and
series-2 12-, 16-HETEs
prostaglandins LOX
and thromboxanes
5-, 12-, 15- HPETEs
LOX
5-, 12-, 15- HETEs
and 4-series
leukotrienes
which can substantially stabilize them and extend their half-lives further.126 5-
HETE and 12-HETE tend to be associated with increased cancer cell growth and
may act through specific receptors or receptor-independent pathways.127,128 Some
tumor types, including gliomas, breast cancer, and leukemia cells, both produce
and respond to HETEs in an autocrine fashion; LOX inhibitors have been dem-
onstrated to have antitumor activity in these cell lines in vitro.129 5-HETE is a
particularly important survival factor for prostate cancer cells. When its synthesis
is blocked, both hormone responsive and nonresponsive tumor cells undergo rapid
apoptosis.130,131 12-HETE promotes proliferation and the metastatic phenotype of
colon, pancreatic, breast, melanoma, and prostate tumor cell lines.83,131 Elevated
12-LOX activity is typical of high-grade tumors, which release 12-HETE pro-
moting angiogenesis in neighboring endothelial cells, changes in extracellular
matrix proteins, and invasive properties in the primary tumor.132,133 Molecular
targets of 12-HETE include protein kinase C, activation of Erk1/2, src, and
activation of NFκB.134 Often times, both COX and LOX products have cancer
promoting and antiapoptotic activity in the same tumor. MCF-7, ER+ breast
cancer cells are stimulated by both LOX and COX products of AA; inhibition of
only one or the other produces much less growth inhibition than inhibiting both
routes simultaneously.135
From a therapeutic view, it seems reasonable to use COX and LOX inhibitors
in chemopreventive or in adjuvant cancer therapies. Despite the evidence from
epidemiological data demonstrating clear chemopreventive effects of NSAIDs,
the long-term use of such pharmacologic agents is associated with substantial
side-effects including gastric ulceration, perforation, and obstruction and have
been linked to thousands of deaths per year.136 This is where dietary modification
and nutritional supplementation come into play. The goal here is to reset the
DK5836_C004.fm Page 111 Thursday, May 25, 2006 12:56 PM
insulin, 150 RANKL, 151 HER-2/neu, 152 nuclear PPARs, and mitochondrial
activity153 have all been shown to be influenced by EPA and/or DHA. As well,
DHA appears to down-regulate the expression of inducible nitric oxide synthase
in colon cancer cells, which indirectly leads to decreased COX-2 production and
associated decreases in NFκB, interferons, cyclic GMP, and up-regulation of a
number of cyclin-dependent kinase inhibitors, including p21 and p27.154 All of
these activities have been associated with reduced tumorigenesis, tumor cell arrest
or apoptosis, and decreased tumor progression. As well, recent studies have
suggested that liver and pancreatic function in postoperative cancer patients can
be improved with n-3 supplementation.155 The benefits of n-3 supplementation
alone seem to be relatively limited, though. No “cures” have been demonstrated
with this approach by itself and the levels of fatty acid needed to achieve many
of the effects in vitro were in the high µM range, which is difficult to achieve
through oral supplementation of patients. Where the n-3s are showing the greatest
promise is in combination or adjuvant therapy settings, with surgery, radiation,
and chemotherapy.
More than a decade ago, it was shown that supplementation of fibrosarcoma
and murine leukemia cells with EPA and DHA increased the toxicity of nucleoside
drugs and doxorubicin selectively in the tumor cells while leaving the normal
cells unaffected or even protected.141,142 This resulted in a substantial theoretical
improvement in therapeutic index for arabinosylcytosine (araC) and doxorubicin.
Later, we demonstrated that in vivo supplementation of rat or mouse diets with
purified DHA decreased tumor burden (fibrosarcoma and leukemia) and enhanced
the therapeutic activity of araC in these animals while simultaneously protecting
bone marrow progenitors and gastrointestinal mucosa, two key sites of potential
dose-limiting injury.27,156,157 While we have been able to demonstrate altered
nucleoside transport activity in a variety of cell types incubated with DHA and
EPA,158,159 we showed that at least part of the chemopotentiation by DHA was
due to altered activity of enzymes involved in araC activation and degradation
that was tumor-specific.160 In our models, both in vitro and in vivo, adding vitamin
E did not attenuate the beneficial effects of DHA on drug toxicity, suggesting
that oxidant-stress per se was not critical; however, a study by Colas and
coworkers38 showed that α-tocopherol did suppress mammary tumor sensitivity
to anthracyclines, so this possibility remains open.
In experimental breast cancer, long-chain n-3 PUFAs themselves have anti-
angiogenic and antmetastatic activity, but tumor kill can synergistically be
improved with several drugs, including mitomycin C, doxorubicin, and cyclo-
phosphamide.161,162 Importantly Colas showed that the effectiveness of the n-3
supplementation with doxorubicin in a breast cancer model was attenuated with
α-tocopherol, suggesting that oxidant stress was part of the killing synergism.38
Their studies had previously indicated that women with higher n-3 levels in breast
adipose tissue fared better following chemotherapy to a variety of regimens than
those with low n-3 status in breast adipose.163 It is not clear from the human
studies to date whether the antioxidant status of the patient is important during
the adjuvant treatment with omega-3 supplements and any of the other drugs.
DK5836_C004.fm Page 113 Thursday, May 25, 2006 12:56 PM
Colon cancer, despite its prevalence and the availability of tools that have
generally aided in early detection and removal of preneoplastic polyps resulting
in increased cure, remains one of the most difficult cancers to treat. Less than
25% of colon cancer patients treated with chemotherapy are responsive. The most
efficacious therapy utilizes a combination of 5-fluorouracil with leucovorin res-
cue. Some recent successes have also been demonstrated with new therapeutic
regimens utilizing two other classes of drugs, irinotecan/camptothecin, and oxali-
platin.164,165 While not typically used in colon cancer therapy (Grem et al., 1995),
araC and gemcitabine have recently been explored as antitumor agents in colon
cancer cells.166 We showed that the therapeutic index of araC could be improved
40-fold with supplementation of rat colon tumor cells while protecting normal
colonic cells from toxicity.167 Jordan and Stein demonstrated that a fish oil-based
lipid emulsion, combined with 5-fluorouracil, promoted apoptosis and cell cycle
arrest in Caco-2 human colon cancer cells as well.168 Wynter et al. showed
increased sensitivity of the MAC16 colon tumor in mice when EPA/DHA were
given in combination with epothiline, gemcitabine, 5-fluorouracil, and cyclophos-
phamide.169 As well as sarcomas, breast cancers, colon cancers, and some leuke-
mias, DHA and EPA have also been tested and found effective as adjuvant agents
in prostate cancer with celecoxib170,171 and in cervical carcinoma.172 A recent study
using more than 30 different colon tumor cell lines identified a series of genes
that identified tumor cells that were, or were not, responsive to 5-fluorouracil
and/or camptothecin.165 Signatures such as these may be useful in the future to
predict which adjuvant therapy setting may provide the best outcome in specific
clinical patients (see Chapters 2, 8, and 9).
of treatment with the flaxseed muffin (between the confirmation biopsy and
surgical resection of the tumor) significantly reduced Ki-67 labeling index, HER-
2/neu expression, progesterone, and estrogen receptor levels, and increased apo-
ptosis in tumor tissue.80 It will be interesting to see if this approach results in
long-term successful treatment outcomes and/or benefits those who go on for
radiation or chemotherapy treatments.
Indole-3-carbinol has been shown to inhibit the hepatotoxicity of the tetrahy-
droisoquinoline alkyloid, trabectidin, without compromising the drug’s mammary
carcinoma killing ability in rats.176 Maitake mushroom β-glucan was recently
shown to enhance the in vitro activity of carmustine in drug-resistant prostate
cancer cells;177 human clinical trials using this preparation in hormone refractory
prostate cancer are needed. Several studies have suggested utility of other natural
medicines, including Rasayanas, which is a complex group of herbals used in
Ayurveda traditional Indian medicine.178 This complex formulation showed
reduced leukopenia and enhanced bone marrow cellularity in cyclophosphamide-
treated mice and enhanced NK activity following radiation therapy179 and inhib-
ited tumor growth and metastasis in melanoma and prostate cancer models.180,181
There is a single report of reduced myelosuppression in a pilot study using human
cancer patients receiving radiation or chemotherapy.182
In addition to certain supplementation conditions, there is evidence that
dietary deficiency of some specific nutrients may actually improve therapy out-
come. While this is generally not the case, a specific example where limiting
nutrient availability may be helpful for cancer treatment is methionine restriction.
A number of studies suggest that tumor cells are dependent on methionine, while
normal cells can use homocysteine instead if provided (reviewed by Cellarier et
al.183). Many tumor types including gliomas and melanomas appear to have low
methionine synthase activity compared to normal surrounding tissue. This could
result from enzymatic mutations or defects in the generation of cofactors B12 and
folate for activity. Alternatively, methionine salvage for polyamine biosynthesis
may be limiting because of deficiencies in methylthioadenosine phosphorylase,
which seems typical of nonsmall-cell lung cancer, leukemia, glioma, rectal ade-
nocarcinoma, and melanoma. Additionally, there is evidence that the requirement
for methionine in tumor cells may simply be much higher than in normal cells
because of increased rates of transmethylation. Capitalizing on these metabolic
differences, some success has occurred in animal models combining methionine-
deficient diets with methionine analogs, such as ethionine in prostate, glioma,
and sarcoma.183 Furthermore, because of the link between folate metabolism and
methionine, deficiency combined with 5-fluorouracil treatment has synergistic
antitumor activity in these same models. Benefits have also been observed with
methionine deficiency in combination with doxorubicin, antimitotics (such as
vincristine), with the alkylating agent carmustine and with cisplatin in human
colon and breast cancer models.184–188 Large-scale clinical trials are still lacking
for these approaches, but are certainly worthy of additional study.
DK5836_C004.fm Page 115 Thursday, May 25, 2006 12:56 PM
4.7 CONCLUSIONS
As the population continues to age and the numbers of individuals afflicted with
cancer continues to climb over the next few decades, substantial new treatments
will need to be available. While there have been considerable strides in the
detection and treatment of some cancers, there continues to be a substantial
problem of drug resistance and failure of therapies for a large portion of those
with disease. Furthermore, incidence of secondary or recurrent malignancies
continues to be a problem for those who survive their first diagnosis and go on
to surgery, radiation, and/or chemotherapy. A large number of children and adults
are currently using nutritional strategies to either decrease their risk of developing
cancer or to improve their likelihood of cure. While there are still numerous
unanswered questions regarding mechanisms, interactions with conventional
drugs, and very few human clinical intervention trials, this “adjuvant” strategy is
likely to be adopted and expanded over the next several years. Understanding the
molecular targets of drugs and nutrients/phytochemicals should lead to improve-
ments in cancer therapies, increased cure rates, and much more immediately,
improved quality of life for cancer patients. The findings presented here are
encouraging and give hope that together conventional practitioners and those that
practice complementary and alternative medicine can cooperate to provide the
best possible treatment options for their patients.
REFERENCES
1. Goodman GE, Thornquist MD, Balmes J, Cullen MR, Meyskens FL, Jr., Omenn
GS, Valanis B, Williams JH Jr. The Beta-Carotene and Retinol Efficacy Trial:
incidence of lung cancer and cardiovascular disease mortality during 6-year fol-
low-up after stopping beta-carotene and retinol supplements. J Natl Cancer Inst
2004; 96(23): 1743–1750.
2. Albanes D, Heinonen OP, Huttunen JK, Taylor PR, Virtamo J, Edwards BK,
Haapakoski J, Rautalahti M, Hartman AM, Palmgren J. Effects of alpha-tocopherol
and beta-carotene supplements on cancer incidence in the Alpha-Tocopherol Beta-
Carotene Cancer Prevention Study. Am J Clin Nutr 1995; 62(6 Suppl): 1427S-
1430S.
3. Pisani P, Bray F, Parkin DM. Estimates of the world-wide prevalence of cancer
for 25 sites in the adult population. Int J Cancer 2002; 97(1): 72–81.
4. Bray F, Sankila R, Ferlay J, Parkin DM. Estimates of cancer incidence and
mortality in Europe in 1995. Eur J Cancer 2002; 38(1): 99–166.
5. Boon H, Stewart M, Kennard MA, Gray R, Sawka C, Brown JB, McWilliam C,
Gavin A, Baron RA, Aaron D, Haines-Kamka T. Use of complementary/alternative
medicine by breast cancer survivors in Ontario: prevalence and perceptions. J Clin
Oncol 2000; 18(13): 2515–2521.
6. Salminen E, Bishop M, Poussa T, Drummond R, Salminen S. Dietary attitudes
and changes as well as use of supplements and complementary therapies by
Australian and Finnish women following the diagnosis of breast cancer. Eur J
Clin Nutr 2004; 58(1): 137–144.
DK5836_C004.fm Page 116 Thursday, May 25, 2006 12:56 PM
7. Kelly KM, Jacobson JS, Kennedy DD, Braudt SM, Mallick M, Weiner MA. Use
of unconventional therapies by children with cancer at an urban medical center.
J Pediatr Hematol Oncol 2000; 22(5): 412–416.
8. Jazieh AR, Kopp M, Foraida M, Ghouse M, Khalil M, Savidge M, Sethuraman
G. The use of dietary supplements by veterans with cancer. J Altern Complement
Med 2004; 10(3): 560–564.
9. Boon H, Westlake K, Stewart M, Gray R, Fleshner N, Gavin A, Brown JB, Goel
V. Use of complementary/alternative medicine by men diagnosed with prostate
cancer: prevalence and characteristics. Urology 2003; 62(5): 849–853.
10. Ockenga J, Valentini L. Review article: anorexia and cachexia in gastrointestinal
cancer. Aliment Pharmacol Ther 2005; 22(7): 583–594.
11. Harvie MN, Campbell IT, Thatcher N, Baildam A. Changes in body composition
in men and women with advanced nonsmall cell lung cancer (NSCLC) undergoing
chemotherapy. J Hum Nutr Diet 2003; 16(5): 323–326.
12. Usharani K, Roy RK, Vijayalakshmi, Prakash J. Nutritional status of cancer
patients given different treatment modalities. Int J Food Sci Nutr 2004; 55(5):
363–369.
13. Tisdale MJ. Molecular pathways leading to cancer cachexia. Physiology
(Bethesda) 2005; 20: 340–348.
14. Yeh SS, Hafner A, Chang CK, Levine DM, Parker TS, Schuster MW. Risk factors
relating blood markers of inflammation and nutritional status to survival in cachec-
tic geriatric patients in a randomized clinical trial. J Am Geriatr Soc 2004; 52(10):
1708–1712.
15. Barber MD, Ross JA, Fearon KC. Cancer cachexia. Surg Oncol 1999; 8(3):
133–141.
16. Fearon KC, Von Meyenfeldt MF, Moses AG, Van Geenen R, Roy A, Gouma DJ,
Giacosa A, Van Gossum A, Bauer J, Barber MD, Aaronson NK, Voss AC, Tisdale
MJ. Effect of a protein and energy dense N-3 fatty acid enriched oral supplement
on loss of weight and lean tissue in cancer cachexia: a randomised double blind
trial. Gut 2003; 52(10): 1479-–1486.
17. Isenring E, Capra S, Bauer J, Davies PS. The impact of nutrition support on body
composition in cancer outpatients receiving radiotherapy. Acta Diabetol 2003; 40
Suppl 1: S162–S164.
18. Moses AW, Slater C, Preston T, Barber MD, Fearon KC. Reduced total energy
expenditure and physical activity in cachectic patients with pancreatic cancer can
be modulated by an energy and protein dense oral supplement enriched with n-3
fatty acids. Br J Cancer 2004; 90(5): 996–1002.
19. Bauer JD, Capra S. Nutrition intervention improves outcomes in patients with
cancer cachexia receiving chemotherapy — a pilot study. Support Care Cancer
2005; 13(4): 270–274.
20. Lorite MJ, Cariuk P, Tisdale MJ. Induction of muscle protein degradation by a
tumour factor. Br J Cancer 1997; 76(8): 1035–1040.
21. Smith HJ, Lorite MJ, Tisdale MJ. Effect of a cancer cachectic factor on protein
synthesis/degradation in murine C2C12 myoblasts: modulation by eicosapen-
taenoic acid. Cancer Res 1999; 59(21): 5507–5513.
22. Smith HJ, Wyke SM, Tisdale MJ. Mechanism of the attenuation of proteolysis-
inducing factor stimulated protein degradation in muscle by beta-hydroxy-beta-
methylbutyrate. Cancer Res 2004; 64(23): 8731–8735.
DK5836_C004.fm Page 117 Thursday, May 25, 2006 12:56 PM
23. Alexandre J, Gross-Goupil M, Falissard B, Nguyen ML, Gornet JM, Misset JL,
Goldwasser F. Evaluation of the nutritional and inflammatory status in cancer
patients for the risk assessment of severe haematological toxicity following che-
motherapy. Ann Oncol 2003; 14(1): 36–41.
24. Ingenbleek Y, Carpentier YA. A prognostic inflammatory and nutritional index
scoring critically ill patients Int J Vitam Nutr Res 1985; 55(1): 91–101.
25. Scully C, Epstein J, Sonis S. Oral mucositis: a challenging complication of
radiotherapy, chemotherapy, and radiochemotherapy. Part 2: diagnosis and man-
agement of mucositis. Head Neck 2004; 26(1): 77–84.
26. Ruthig DJ, Meckling-Gill KA. Both (n-3) and (n-6) fatty acids stimulate wound
healing in the rat intestinal epithelial cell line, IEC-6. J Nutr 1999; 129(10):
1791–1798.
27. Atkinson TG, Murray L, Berry DM, Ruthig DJ, Meckling-Gill KA. DHA feeding
provides host protection and prevents fibrosarcoma-induced hyperlipidemia while
maintaining the tumor response to araC in Fischer 344 rats. Nutr Cancer 1997;
28(3): 225–235.
28. Piccirillo N, De Matteis S, Laurenti L, Chiusolo P, Sora F, Pittiruti M, Rutella S,
Cicconi S, Fiorini A, D’Onofrio G, Leone G, Sica S. Glutamine-enriched
parenteral nutrition after autologous peripheral blood stem cell transplantation:
effects on immune reconstitution and mucositis. Haematologica 2003; 88(2):
192–200.
29. Spielberger R, Stiff P, Bensinger W, Gentile T, Weisdorf D, Kewalramani T, Shea
T, Yanovich S, Hansen K, Noga S, McCarty J, LeMaistre CF, Sung EC, Blazar
BR, Elhardt D, Chen MG, Emmanouilides C. Palifermin for oral mucositis after
intensive therapy for hematologic cancers. N Engl J Med 2004; 351(25):
2590–2598.
30. Brinckmann J, Sigwart H, van Houten TL. Safety and efficacy of a traditional
herbal medicine (Throat Coat) in symptomatic temporary relief of pain in patients
with acute pharyngitis: a multicenter, prospective, randomized, double-blinded,
placebo-controlled study. J Altern Complement Med 2003; 9(2): 285–298.
31. Prasad KN, Kumar A, Kochupillai V, Cole WC. High doses of multiple antioxidant
vitamins: essential ingredients in improving the efficacy of standard cancer ther-
apy. J Am Coll Nutr 1999; 18(1): 13–25.
32. Lamson DW, Brignall MS. Antioxidants in cancer therapy; their actions and
interactions with oncologic therapies. Altern Med Rev 1999; 4(5): 304–329.
33. Conklin KA. Dietary antioxidants during cancer chemotherapy: impact on che-
motherapeutic effectiveness and development of side effects. Nutr Cancer 2000;
37(1): 1–18.
34. Kumar A, Qiblawi S, Khan AK, Banerjee S, Rao AR. Chemomodulatory action
of Brassica compestris (var sarason) on hepatic carcinogen metabolizing enzymes,
antioxidant profiles and lipid peroxidation. Asian Pac J Cancer Prev 2004; 5(2):
190–195.
35. Labriola D, Livingston R. Possible interactions between dietary antioxidants and
chemotherapy. Oncology (Williston Park) 1999; 13(7): 1003–1008.
36. Kong Q, Lillehei KO. Antioxidant inhibitors for cancer therapy. Med Hypotheses
1998; 51(5): 405–409.
DK5836_C004.fm Page 118 Thursday, May 25, 2006 12:56 PM
37. Salganik RI, Albright CD, Rodgers J, Kim J, Zeisel SH, Sivashinskiy MS, Van
Dyke TA. Dietary antioxidant depletion: enhancement of tumor apoptosis and
inhibition of brain tumor growth in transgenic mice. Carcinogenesis 2000; 21(5):
909–914.
38. Colas S, Germain E, Arab K, Maheo K, Goupille C, Bougnoux P. Alpha-tocopherol
suppresses mammary tumor sensitivity to anthracyclines in fish oil-fed rats. Nutr
Cancer 2005; 51(2): 178–183.
39. Saintot M, Mathieu-Daude H, Astre C, Grenier J, Simony-Lafontaine J, Gerber
M. Oxidant-antioxidant status in relation to survival among breast cancer patients.
Int J Cancer 2002; 97(5): 574–579.
40. Wagdi P, Fluri M, Aeschbacher B, Fikrle A, Meier B. Cardioprotection in patients
undergoing chemo- and/or radiotherapy for neoplastic disease. A pilot study. Jpn
Heart J 1996; 37(3): 353–359.
41. Wadleigh RG, Redman RS, Graham ML, Krasnow SH, Anderson A, Cohen MH.
Vitamin E in the treatment of chemotherapy-induced mucositis. Am J Med 1992;
92(5): 481–484.
42. Chinery R, Brockman JA, Peeler MO, Shyr Y, Beauchamp RD, Coffey RJ. Anti-
oxidants enhance the cytotoxicity of chemotherapeutic agents in colorectal cancer:
a p53-independent induction of p21WAF1/CIP1 via C/EBPbeta. Nat Med 1997;
3(11): 1233–1241.
43. Norman HA, Butrum RR, Feldman E, Heber D, Nixon D, Picciano MF, Rivlin
R, Simopoulos A, Wargovich MJ, Weisburger EK, Zeisel SH. The role of dietary
supplements during cancer therapy. J Nutr 2003; 133(11 Suppl 1): 3794S–3799S.
44. Mayland CR, Bennett MI, Allan K. Vitamin C deficiency in cancer patients. Palliat
Med 2005; 19(1): 17–20.
45. Kennedy DD, Tucker KL, Ladas ED, Rheingold SR, Blumberg J, Kelly KM. Low
antioxidant vitamin intakes are associated with increases in adverse effects of
chemotherapy in children with acute lymphoblastic leukemia. Am J Clin Nutr
2004; 79(6): 1029–1036.
46. Choi CH. ABC transporters as multidrug resistance mechanisms and the devel-
opment of chemosensitizers for their reversal. Cancer Cell Int 2005; 5: 30.
47. Lautier D, Canitrot Y, Deeley RG, Cole SP. Multidrug resistance mediated by the
multidrug resistance protein (MRP) gene. Biochem Pharmacol 1996; 52(7):
967–977.
48. Ballatori N, Hammond CL, Cunningham JB, Krance SM, Marchan R. Molecular
mechanisms of reduced glutathione transport: role of the MRP/CFTR/ABCC and
OATP/SLC21A families of membrane proteins. Toxicol Appl Pharmacol 2005;
204(3): 238–255.
49. Stride BD, Valdimarsson G, Gerlach JH, Wilson GM, Cole SP, Deeley RG.
Structure and expression of the messenger RNA encoding the murine multidrug
resistance protein, an ATP-binding cassette transporter. Mol Pharmacol 1996;
49(6): 962–971.
50. Mizutani T, Hattori A. New horizon of MDR1 (P-glycoprotein) study. Drug Metab
Rev 2005; 37(3): 489–510.
51. Raaijmakers HG, Van Den BG, Boezeman J, De Witte T, Raymakers RA. Single-
cell image analysis to assess ABC-transporter-mediated efflux in highly purified
hematopoietic progenitors. Cytometry 2002; 49(4): 135–142.
DK5836_C004.fm Page 119 Thursday, May 25, 2006 12:56 PM
69. Yuan H, Pan Y, Young CY. Overexpression of c-Jun induced by quercetin and
resverol inhibits the expression and function of the androgen receptor in human
prostate cancer cells. Cancer Lett 2004; 213(2): 155–163.
70. Kundu JK, Surh YJ. Molecular basis of chemoprevention by resveratrol: NF-κB
and AP-1 as potential targets. Mutat Res 2004; 555(1-2): 65–80.
71. Min JK, Han KY, Kim EC, Kim YM, Lee SW, Kim OH, Kim KW, Gho YS, Kwon
YG. Capsaicin inhibits in vitro and in vivo angiogenesis. Cancer Res 2004; 64(2):
644–651.
72. Oak MH, El Bedoui J, Schini-Kerth VB. Antiangiogenic properties of natural
polyphenols from red wine and green tea. J Nutr Biochem 2005; 16(1): 18.
73. Korutla L, Cheung JY, Mendelsohn J, Kumar R. Inhibition of ligand-induced
activation of epidermal growth factor receptor tyrosine phosphorylation by cur-
cumin. Carcinogenesis 1995; 16(8): 1741–1745.
74. Aggarwal S, Ichikawa H, Takada Y, Sandur SK, Shishodia S, Aggarwal BB.
Curcumin (diferuloylmethane) downregulates expression of cell proliferation,
antiapoptotic and metastatic gene products through suppression of
I{kappa}B{alpha} kinase and AKT activation. Mol Pharmacol Epub, October
11, 2005.
75. Vyas S, Asmerom Y, De Leon DD. Resveratrol regulates insulin-like growth factor-
II in breast cancer cells. Endocrinology 2005; 146(10): 4224–4233.
76. Mousa AS, Mousa SA. Anti-angiogenesis efficacy of the garlic ingredient alliin
and antioxidants: role of nitric oxide and p53. Nutr Cancer 2005; 53(1): 104–110.
77. Mousa SS, Mousa SS, Mousa SA. Effect of resveratrol on angiogenesis and
platelet/fibrin-accelerated tumor growth in the chick chorioallantoic membrane
model. Nutr Cancer 2005; 52(1): 59–65.
78. Singh RP, Tyagi AK, Dhanalakshmi S, Agarwal R, Agarwal C. Grape seed extract
inhibits advanced human prostate tumor growth and angiogenesis and upregulates
insulin-like growth factor binding protein-3. Int J Cancer 2004; 108(5): 733–740.
79. Hong RL, Spohn WH, Hung MC. Curcumin inhibits tyrosine kinase activity of
p185neu and also depletes p185neu. Clin Cancer Res 1999; 5(7): 1884–1891.
80. Thompson LU, Chen JM, Li T, Strasser-Weippl K, Goss PE. Dietary flaxseed
alters tumor biological markers in postmenopausal breast cancer. Clin Cancer Res
2005; 11(10): 3828–3835.
81. Kim HY, Park EJ, Joe EH, Jou I. Curcumin suppresses Janus kinase-STAT inflam-
matory signaling through activation of Src homology 2 domain-containing
tyrosine phosphatase 2 in brain microglia. J Immunol 2003; 171(11): 6072–6079.
82. Labrecque L, Lamy S, Chapus A, Mihoubi S, Durocher Y, Cass B, Bojanowski
MW, Gingras D, Beliveau R. Combined inhibition of PDGF and VEGF receptors
by ellagic acid, a dietary-derived phenolic compound. Carcinogenesis 2005; 26(4):
821–826.
83. Wallace JM. Nutritional and botanical modulation of the inflammatory cascade
— eicosanoids, cyclooxygenases, and lipoxygenases — as an adjunct in cancer
therapy. Integr Cancer Ther 2002; 1(1): 737.
84. Dimberg J, Samuelsson A, Hugander A, Soderkvist P. Differential expression of
cyclooxygenase 2 in human colorectal cancer. Gut 1999; 45(5): 730–732.
85. Sheng H, Shao J, Kirkland SC, Isakson P, Coffey RJ, Morrow J, Beauchamp RD,
DuBois RN. Inhibition of human colon cancer cell growth by selective inhibition
of cyclooxygenase-2. J Clin Invest 1997; 99(9): 2254–2259.
DK5836_C004.fm Page 121 Thursday, May 25, 2006 12:56 PM
102. Mantena SK, Meeran SM, Elmets CA, Katiyar SK. Orally administered green tea
polyphenols prevent ultraviolet radiation-induced skin cancer in mice through
activation of cytotoxic T cells and inhibition of angiogenesis in tumors. J Nutr
2005; 135(12): 2871–2877.
103. Mantena SK, Roy AM, Katiyar SK. Epigallocatechin-3-gallate inhibits photocar-
cinogenesis through inhibition of angiogenic factors and activation of CD8(+) T
cells in tumors. Photochem Photobiol 2005; 81(5): 1174–1179.
104. Pryme IF, Bardocz S, Pusztai A, Ewen SW, Pfuller U. A mistletoe lectin (ML-1)-
containing diet reduces the viability of a murine non-Hodgkin lymphoma tumor.
Cancer Detect Prev 2004; 28(1): 52–56.
105. Baxevanis CN, Voutsas IF, Soler MH, Gritzapis AD, Tsitsilonis OE, Stoeva S,
Voelter W, Arsenis P, Papamichail M. Mistletoe lectin I-induced effects on human
cytotoxic lymphocytes. I. Synergism with IL-2 in the induction of enhanced LAK
cytotoxicity. Immunopharmacol Immunotoxicol 1998; 20(3): 355–372.
106. Chang R. Bioactive polysaccharides from traditional Chinese medicine herbs as
anticancer adjuvants. J Altern Complement Med 2002; 8(5): 559–565.
107. Cleary JA, Kelly GE, Husband AJ. The effect of molecular weight and beta-1,6-
linkages on priming of macrophage function in mice by (1,3)-beta-D-glucan.
Immunol Cell Biol 1999; 77(5): 395–403.
108. Ng ML, Yap AT. Inhibition of human colon carcinoma development by lentinan
from shiitake mushrooms (Lentinus edodes). J Altern Complement Med 2002;
8(5): 581–589.
109. Wakui A, Kasai M, Konno K, Abe R, Kanamaru R, Takahashi K, Nakai Y, Yoshida
Y, Koie H, Masuda H. Randomized study of lentinan on patients with advanced
gastric and colorectal cancer. Tohoku Lentinan Study Group. Gan To Kagaku
Ryoho 1986; 13(4 Pt 1): 1050–1059.
110. Nakazato H, Koike A, Saji S, Ogawa N, Sakamoto J. Efficacy of immunochemo-
therapy as adjuvant treatment after curative resection of gastric cancer. Study
Group of Immunochemotherapy with PSK for Gastric Cancer. Lancet 1994;
343(8906): 1122–1126.
111. Ito K, Nakazato H, Koike A, Takagi H, Saji S, Baba S, Mai M, Sakamoto J, Ohashi
Y. Long-term effect of 5-fluorouracil enhanced by intermittent administration of
polysaccharide K after curative resection of colon cancer. A randomized controlled
trial for 7-year follow-up. Int J Colorectal Dis 2004; 19(2): 157–164.
112. Mitomi T, Tsuchiya S, Iijima N, Aso K, Suzuki K, Nishiyama K, Amano T,
Takahashi T, Murayama N, Oka H. Randomized, controlled study on adjuvant
immunochemotherapy with PSK in curatively resected colorectal cancer. The
Cooperative Study Group of Surgical Adjuvant Immunochemotherapy for Cancer
of Colon and Rectum (Kanagawa). Dis Colon Rectum 1992; 35(2): 123–130.
113. Ogoshi K, Satou H, Isono K, Mitomi T, Endoh M, Sugita M. Immunotherapy for
esophageal cancer. A randomized trial in combination with radiotherapy and
radiochemotherapy. Cooperative Study Group for Esophageal Cancer in Japan.
Am J Clin Oncol 1995; 18(3): 216–222.
114. Go P, Chung CH. Adjuvant PSK immunotherapy in patients with carcinoma of
the nasopharynx. J Int Med Res 1989; 17(2): 141–149.
115. Hayakawa K, Mitsuhashi N, Saito Y, Takahashi M, Katano S, Shiojima K, Furuta
M, Niibe H. Effect of krestin (PSK) as adjuvant treatment on the prognosis after
radical radiotherapy in patients with non-small cell lung cancer. Anticancer Res
1993; 13(5C): 1815–1820.
DK5836_C004.fm Page 123 Thursday, May 25, 2006 12:56 PM
128. O’Flaherty JT, Rogers LC, Paumi CM, Hantgan RR, Thomas LR, Clay CE, High
K, Chen YQ, Willingham MC, Smitherman PK, Kute TE, Rao A, Cramer SD,
Morrow CS. 5-Oxo-ETE analogs and the proliferation of cancer cells. Biochim
Biophys Acta 2005; 1736(3): 228–236.
129. Blomgren H, Kling-Andersson G. Growth inhibition of human malignant glioma
cells in vitro by agents which interfere with biosynthesis of eicosanoids. Antican-
cer Res 1992; 12(3): 981–986.
130. Ghosh J, Myers CE. Central role of arachidonate 5-lipoxygenase in the regulation
of cell growth and apoptosis in human prostate cancer cells. Adv Exp Med Biol
1999; 469: 577–582.
131. Gao X, Honn KV. Biological properties of 12(S)-HETE in cancer metastasis. Adv
Prostaglandin Thromboxane. Leukot Res 1995; 23: 439–444.
132. Pidgeon GP, Tang K, Rice RL, Zacharek A, Li L, Taylor JD, Honn KV. Overex-
pression of leukocyte-type 12-lipoxygenase promotes W256 tumor cell survival
by enhancing alphavbeta5 expression. Int J Cancer 2003; 105(4): 459–471.
133. Kandouz M, Nie D, Pidgeon GP, Krishnamoorthy S, Maddipati KR, Honn KV.
Platelet-type 12-lipoxygenase activates NF-kappaB in prostate cancer cells. Pros-
taglandins Other Lipid Mediat 2003; 71(3-4): 189–204.
134. Szekeres CK, Trikha M, Honn KV. 12(S)-HETE, pleiotropic functions, multiple
signaling pathways. Adv Exp Med Biol 2002; 507: 509–515.
135. Steele VE, Holmes CA, Hawk ET, Kopelovich L, Lubet RA, Crowell JA, Sigman
CC, Kelloff GJ. Lipoxygenase inhibitors as potential cancer chemopreventives.
Cancer Epidemiol Biomarkers Prev 1999; 8(5): 467–483.
136. Miller JL. Decisions loom on selective COX-2 inhibitors. Am J Health Syst Pharm
1999; 56(2): 106–107.
137. Young G, Conquer J. Omega-3 fatty acids and neuropsychiatric disorders. Reprod
Nutr Dev 2005; 45(1): 1–28.
138. Marzo I, Pineiro A, Naval J. Loss of delta6-desaturase activity leads to impaired
docosahexaenoic acid synthesis in Y-79 retinoblastoma cells. Prostaglandins Leu-
kot Essent Fatty Acids 1998; 59(5): 293–297.
139. Marzo I, Martinez-Lorenzo MJ, Anel A, Desportes P, Alava MA, Naval J, Pineiro
A. Biosynthesis of unsaturated fatty acids in the main cell lineages of human
leukemia and lymphoma. Biochim Biophys Acta 1995; 1257(2): 140–148.
140. Lane J, Mansel RE, Jiang WG. Expression of human delta-6-desaturase is asso-
ciated with aggressiveness of human breast cancer. Int J Mol Med 2003; 12(2):
253–257.
141. Atkinson TG, Meckling-Gill K.A. Regulation of nucleoside drug toxicity by
transport inhibitors and omega-3 polyunsaurated fatty acids in mormal and trans-
formed rat-2 fibroblasts. Cell Pharmacol 1995; 2: 259–264.
142. de Salis H, Meckling-Gill KA. EPA and DHA alter nucleoside drug and adria-
mycin toxicity in L1210 leukemia cells but not in normal bone marrow derived
S1 macrophages. Cell Pharmacol 1995; 2: 69–74.
143. Rose DP, Connolly JM, Rayburn J, Coleman M. Influence of diets containing
eicosapentaenoic or docosahexaenoic acid on growth and metastasis of breast
cancer cells in nude mice. J Natl Cancer Inst 1995; 87(8): 587–592.
144. Brand A, Yavin E. Translocation of ethanolamine phosphoglyceride is required
for initiation of apoptotic death in OLN-93 oligodendroglial cells. Neurochem Res
2005; 30(10): 1257–1267.
DK5836_C004.fm Page 125 Thursday, May 25, 2006 12:56 PM
145. Kimura Y. New anticancer agents: in vitro and in vivo evaluation of the antitumor
and antimetastatic actions of various compounds isolated from medicinal plants.
In Vivo 2005; 19(1): 37–60.
146. Kimura Y, Sumiyoshi M. Antitumor and antimetastatic actions of eicosapentaenoic
acid ethylester and its by-products formed during accelerated stability testing.
Cancer Sci 2005; 96(7): 441–450.
147. Jia Y, Turek JJ. Inducible nitric oxide synthase links NF-kappaB to PGE2 in
polyunsaturated fatty acid altered fibroblast in vitro wound healing. Lipids Health
Dis 2005; 4: 14.
148. Singh J, Hamid R, Reddy BS. Dietary fat and colon cancer: modulation of
cyclooxygenase-2 by types and amount of dietary fat during the post-initiation
stage of colon carcinogenesis. Cancer Res 1997; 57(16): 3465–3470.
149. Endres S, von Schacky C. n-3 polyunsaturated fatty acids and human cytokine
synthesis. Curr Opin Lipidol 1996; 7(1): 48–52.
150. Lardinois CK. The role of omega 3 fatty acids on insulin secretion and insulin
sensitivity. Med Hypotheses 1987; 24(3): 243–248.
151. Bhattacharya A, Rahman M, Banu J, Lawrence RA, McGuff HS, Garrett IR,
Fischbach M, Fernandes G. Inhibition of osteoporosis in autoimmune disease
prone MRL/Mpj-Fas(lpr) mice by N-3 fatty acids. J Am Coll Nutr 2005; 24(3)
200–209.
152. Menendez JA, Lupu R, Colomer R. Exogenous supplementation with omega-3
polyunsaturated fatty acid docosahexaenoic acid (DHA; 22:6n-3) synergistically
enhances taxane cytotoxicity and downregulates Her-2/neu (c-erbB-2) oncogene
expression in human breast cancer cells. Eur J Cancer Prev 2005; 14(3): 263–270.
153. Guo W, Xie W, Lei T, Hamilton JA. Eicosapentaenoic acid, but not oleic acid,
stimulates beta-oxidation in adipocytes. Lipids 2005; 40(8): 815–821.
154. Narayanan BA, Narayanan NK, Reddy BS. Docosahexaenoic acid regulated genes
and transcription factors inducing apoptosis in human colon cancer cells. Int J
Oncol 2001; 19(6): 1255–1262.
155. Heller AR, Rossel T, Gottschlich B, Tiebel O, Menschikowski M, Litz RJ, Zim-
mermann T, Koch T. Omega-3 fatty acids improve liver and pancreas function in
postoperative cancer patients. Int J Cancer 2004; 111(4): 611–616.
156. Atkinson TG, Barker HJ, Meckling-Gill KA. Incorporation of long-chain n-3 fatty
acids in tissues and enhanced bone marrow cellularity with docosahexaenoic acid
feeding in post-weanling Fischer 344 rats. Lipids 1997; 32(3): 293–302.
157. Cha MC, Meckling KA, Stewart C. Dietary docosahexaenoic acid levels influence
the outcome of arabinosylcytosine chemotherapy in l1210 leukemic mice. Nutr
Cancer 2002; 44(2): 176–181.
158. Blackmore VL, Meckling-Gill KA. Fish oil and oleic acid-rich oil feeding alter
nucleoside uptake in human erythrocytes. Nutritional Biochemistry 1995; 6:
438–444.
159. Martin D, Meckling-Gill KA. Omega-3 polyunsaturated fatty acids increase purine
but not pyrimidine transport in L1210 leukaemia cells. Biochem J 1996; 315 (Pt
1): 329–333.
160. Cha MC, Meckling-Gill KA. Modifications of deoxycytidine kinase and deami-
nase activities by docosahexaenoic acid in normal and transformed rat fibroblasts.
Biochem Pharmacol 2002; 63(4): 717–723.
161. Rose DP, Connolly JM. Regulation of tumor angiogenesis by dietary fatty acids
and eicosanoids. Nutr Cancer 2000; 37(2): 119–127.
DK5836_C004.fm Page 126 Thursday, May 25, 2006 12:56 PM
162. Hardman WE. (n-3) fatty acids and cancer therapy. J Nutr 2004; 134(12 Suppl):
3427S–3430S.
163. Bougnoux P, Germain E, Chajes V, Hubert B, Lhuillery C, Le Floch O, Body G,
Calais G. Cytotoxic drugs efficacy correlates with adipose tissue docosahexaenoic
acid level in locally advanced breast carcinoma. Br J Cancer 1999; 79(11-12):
1765–1769.
164. Martin MJ. Current stage-specific chemotherapeutic options in colon cancer.
Expert Rev Anticancer Ther 2005; 5(4): 695–704.
165. Mariadason JM, Arango D, Shi Q, Wilson AJ, Corner GA, Nicholas C, Aranes
MJ, Lesser M, Schwartz EL, Augenlicht LH. Gene expression profiling-based
prediction of response of colon carcinoma cells to 5-fluorouracil and camptothecin.
Cancer Res 2003; 63(24): 8791–8812.
166. Grem JL, Geoffroy F, Politi PM, Cuddy DP, Ross DD, Nguyen D, Steinberg SM,
Allegra CJ. Determinants of sensitivity to 1-beta-D-arabinofuranosylcytosine in
HCT 116 and NCI-H630 human colon carcinoma cells. Mol Pharmacol 1995;
48(2): 305–315.
167. Cha MC, Lin A, Meckling KA. Low dose docosahexaenoic acid protects normal
colonic epithelial cells from araC toxicity. BMC Pharmacol 2005; 5(1): 7.
168. Jordan A, Stein J. Effect of an omega-3 fatty acid containing lipid emulsion alone
and in combination with 5-fluorouracil (5-FU) on growth of the colon cancer cell
line Caco-2. Eur J Nutr 2003; 42(6): 324–331.
169. Wynter MP, Russell ST, Tisdale MJ. Effect of n-3 fatty acids on the antitumour
effects of cytotoxic drugs. In Vivo 2004; 18(5): 543–547.
170. Narayanan BA, Narayanan NK, Pttman B, Reddy BS. Adenocarcina of the mouse
prostate growth inhibition by celecoxib: downregulation of transcription factors
involved in COX-2 inhibition. Prostate 2006; 66: 257–265.
171. Narayanan NK, Narayanan BA, Reddy BS. A combination of docosahexaenoic
acid and celecoxib prevents prostate cancer cell growth in vitro and is associated
with modulation of nuclear factor-kappaB, and steroid hormone receptors. Int J
Oncol 2005; 26(3): 785–792.
172. Madhavi N, Das UN, Prabha PS, Kumar GS, Koratkar R, Sagar PS. Suppression
of human T-cell growth in vitro by cis-unsaturated fatty acids: relationship to free
radicals and lipid peroxidation. Prostaglandins Leukot Essent Fatty Acids 1994;
51(1): 33–40.
173. Dabrosin C, Chen J, Wang L, Thompson LU. Flaxseed inhibits metastasis and
decreases extracellular vascular endothelial growth factor in human breast cancer
xenografts. Cancer Lett 2002; 185(1): 31–37.
174. Bylund A, Saarinen N, Zhang JX, Bergh A, Widmark A, Johansson A, Lundin E,
Adlercreutz H, Hallmans G, Stattin P, Makela S. Anticancer effects of a plant
lignan 7-hydroxymatairesinol on a prostate cancer model in vivo. Exp Biol Med
(Maywood) 2005; 230(3): 217–223.
175. Chen J, Hui E, Ip T, Thompson LU. Dietary flaxseed enhances the inhibitory
effect of tamoxifen on the growth of estrogen-dependent human breast cancer
(mcf-7) in nude mice. Clin Cancer Res 2004; 10(22): 7703–7711.
176. Donald S, Verschoyle RD, Greaves P, Colombo T, Zucchetti M, Falcioni C,
Zaffaroni M, D’Incalci M, Manson MM, Jimeno J, Steward WP, Gescher AJ.
Dietary agent indole-3-carbinol protects female rats against the hepatotoxicity of
the antitumor drug ET-743 (trabectidin) without compromising efficacy in a rat
mammary carcinoma. Int J Cancer 2004; 111(6): 961–967.
DK5836_C004.fm Page 127 Thursday, May 25, 2006 12:56 PM
177. Finkelstein MP, Aynehchi S, Samadi AA, Drinis S, Choudhury MS, Tazaki H,
Konno S. Chemosensitization of carmustine with maitake beta-glucan on andro-
gen-independent prostatic cancer cells: involvement of glyoxalase I. J Altern
Complement Med 2002; 8(5): 573–580.
178. Vayalil PK, Kuttan G, Kuttan R. Protective effects of Rasayanas on cyclophos-
phamide- and radiation-induced damage. J Altern Complement Med 2002; 8(6):
787–796.
179. Kumar P, Kuttan R, Kuttan G. Radioprotective effects of Rasayanas. Indian J Exp
Biol 1996; 34(9): 848–850.
180. Menon LG, Kuttan R, Kuttan G. Effect of rasayanas in the inhibition of lung
metastasis induced by B16F-10 melanoma cells. J Exp Clin Cancer Res 1997;
16(4): 365–368.
181. Gaddipati JP, Rajeshkumar NV, Thangapazham RL, Sharma A, Warren J, Mog
SR, Singh AK, Maheshwari RK. Protective effect of a polyherbal preparation,
Brahma rasayana against tumor growth and lung metastasis in rat prostate model
system. J Exp Ther Oncol 2004; 4(3): 203–212.
182. Joseph CD, Praveenkumar V, Kuttan G, Kuttan R. Myeloprotective effect of a
non-toxic indigenous preparation Rasayana in cancer patients receiving chemo-
therapy and radiation therapy. A pilot study. J Exp Clin Cancer Res 1999; 18(3):
325–329.
183. Cellarier E, Durando X, Vasson MP, Farges MC, Demiden A, Maurizis JC, Madel-
mont JC, Chollet P. Methionine dependency and cancer treatment. Cancer Treat
Rev 2003; 29(6): 489–499.
184. Stern PH, Hoffman RM. Enhanced in vitro selective toxicity of chemotherapeutic
agents for human cancer cells based on a metabolic defect. J Natl Cancer Inst
1986; 76(4): 629–639.
185. Poirson-Bichat F, Goncalves RA, Miccoli L, Dutrillaux B, Poupon MF. Methion-
ine depletion enhances the antitumoral efficacy of cytotoxic agents in drug-resis-
tant human tumor xenografts. Clin Cancer Res 2000; 6(2): 643–653.
186. Kokkinakis DM, Hoffman RM, Frenkel EP, Wick JB, Han Q, Xu M, Tan Y, Schold
SC. Synergy between methionine stress and chemotherapy in the treatment of
brain tumor xenografts in athymic mice. Cancer Res 2001; 61(10): 4017–4023.
187. Tan Y, Sun X, Xu M, Tan X, Sasson A, Rashidi B, Han Q, Tan X, Wang X, An
Z, Sun FX, Hoffman RM. Efficacy of recombinant methioninase in combination
with cisplatin on human colon tumors in nude mice. Clin Cancer Res 1999; 5(8):
2157–2163.
188. Hoshiya Y, Kubota T, Matsuzaki SW, Kitajima M, Hoffman RM. Methionine
starvation modulates the efficacy of cisplatin on human breast cancer in nude
mice. Anticancer Res 1996; 16(6B): 3515–3517.
DK5836_C004.fm Page 128 Thursday, May 25, 2006 12:56 PM
DK5836_C005.fm Page 129 Monday, May 22, 2006 1:14 PM
5 Role of Nutritional
Antioxidants in the
Prevention and
Treatment of
Neurodegenerative
Disorders
Ennio Esposito
CONTENTS
5.1 Introduction..............................................................................................130
5.1.1 Major Neurodegenerative Disorders ...........................................130
5.1.2 Oxidative Stress and Antioxidants ..............................................131
5.1.3 Epidemiological Studies..............................................................133
5.2 Natural Dietary Antioxidants ..................................................................134
5.2.1 Presence of Flavonoids in Foods ................................................138
5.2.2 Absorption and Metabolism of Polyphenols ..............................140
5.2.3 Biochemical Actions of Polyphenols ..........................................141
5.2.4 Putative Health Benefits of Flavonoids.......................................142
5.3 ROS, NFκB, and Neurodegenerative Disorders .....................................143
5.4 Alzheimer’s Disease, Oxidative Stress, NFκB, and Antioxidants .........146
5.5 Parkinson’s Disease, Oxidative Stress, NFκB, and Antioxidants ..........149
5.6 Amyotrophic Lateral Sclerosis, Oxidative Stress,
NFκB, and Antioxidants..........................................................................154
5.7 Conclusions..............................................................................................156
5.8 Acknowledgments ...................................................................................157
References .........................................................................................................157
129
DK5836_C005.fm Page 130 Monday, May 22, 2006 1:14 PM
5.1 INTRODUCTION
There is an increasing attention toward the role played by certain nutritional
components found in foods, including dietary flavonoids, in fruit, vegetables and
beverages in the prevention of age-related decreases in cognitive, memory and
learning tasks. Thus, aging is a major risk factor for neurodegenerative diseases
including Alzheimer’s disease (AD), Parkinson’s disease (PD), and amyotrophic
lateral sclerosis (ALS). An unbalanced overproduction of reactive oxygen species
(ROS) may give rise to oxidative stress, which can induce neuronal damage,
ultimately leading to neuronal death by apoptosis or necrosis. A large body of
evidence indicates that oxidative stress is involved in the pathogenesis of AD,
PD and ALS. An increasing number of studies show that nutritional antioxidants
(especially vitamin E and polyphenols) can block neuronal death in vitro, and
may have therapeutic properties in animal models of neurodegenerative diseases
including AD, PD and ALS. Moreover, clinical data suggest that nutritional
antioxidants might exert some protective effect against AD, PD and ALS. In this
chapter, the biochemical mechanisms by which nutritional antioxidants can
reduce or block neuronal death occurring in neurodegenerative disorders are
reviewed. Particular emphasis will be given to the role played by the nuclear
transcription factor-κB (NFκB) in apoptosis, and in the pathogenesis of neuro-
degenerative disorders, such as AD, PD and ALS. The effects of ROS and
antioxidants on NFκB function and their relevance in the pathophysiology of
neurodegenerative diseases will also be examined.
radical scavenger that is effective against peroxyl and hydroxyl radicals, super-
oxide, singlet oxygen and peroxynitrite.34 Also, the lipid-soluble, chain-breaking
antioxidant vitamin E exerts a very important protective function against oxidative
stress in the brain10 and interacts with ascorbate enhancing its antioxidant activ-
ity.35 Little information is available on the levels of carotenoids and flavonoids
in the human brain. The antioxidant enzymes in the brain include Cu/Zn super-
oxide dismutase (SOD-1) and Mn superoxide dismutase (SOD-2), which catalyze
the conversion of O2–. to H2O2.36 H2O2 is then converted to H2O by either catalase
or glutathione peroxidase (GSH-Px). Antioxidant defense mechanisms can be
upregulated in response to increased ROS or peroxide production.37 Although
upregulating antioxidant defense systems may confer protection against ROS,
they are not completely effective in preventing oxidative damage. Moreover, the
efficiency of gene expression may decline with age or become defective as
oxidative damage to the genome increases.
As already mentioned, the brain is especially vulnerable to ROS damage
because of its high oxygen consumption rate, abundant lipid content and relative
paucity of antioxidant enzymes compared with other organs.38 If the increased
demand on the cell’s capacity to detoxify ROS is not met, alterations, such as
aldehydes or isoprostanes from lipid peroxidation, protein carbonyls from protein
oxidation, and oxidized base adducts from DNA oxidation may accumulate.10
Oxidation of polyunsaturated fatty acids (PUFA) results in the production of
multiple aldehydes with different carbon chain lengths including propanal, buta-
nal, pentanal, hexanal and 4-hydroxy-2-trans-nonenal (4-HNE). There is evidence
that 4-HNE is capable of inducing apoptosis in PC12 cells and cultured rat
hippocampal neurons, suggesting that it is a mediator of oxidative stress-induced
apoptosis.39 These findings suggest that in addition to direct ROS damage to
phospholipid membranes, there is an indirect mechanism involving 4-HNE, which
may also be involved in neuronal death. In this regard, it noteworthy that 4-HNE
has been suggested to be involved in the pathogenesis of PD.40,41 Oxidative
damage to proteins can be revealed by measuring protein carbonyl content,42
which was found to be elevated in AD and ALS patients.43 Another indication of
protein oxidation is the formation of nitrotyrosine by peroxynitrite. This might
represent a useful clinical parameter of the occurrence of oxidative stress in
neurodegenerative diseases, inasmuch as increased levels of nitrotyrosine have
been found in AD, PD and ALS.21,44–49 The most useful marker of DNA oxidation
is 8-hydroxy-2′-deoxyguanosine (8-OHdG), which is elevated in patients with
AD, PD and ALS.50–54
Another index of oxidative is the activation of the transcription factor NFκB
(nuclear factor kappa B). Thus, a large body of evidence indicates that ROS can
act as second messengers mediating intracellular responses, including NFκB
activation.55–59 In turn, activated NFκB can influence the expression of a large
number of genes, including SOD-2.55,58,60 Hence, NFκB activation can be con-
sidered as the executive branch of a feed-back mechanism that operates to regulate
the intracellular concentration of ROS, trying to dampen an excessive accumu-
lation of ROS, which can be dangerous for the cell. Moreover, NFκB induces
DK5836_C005.fm Page 133 Monday, May 22, 2006 1:14 PM
the expression of the so-called IAPs (inhibitor of apoptosis proteins), Bcl-2, and
calbindins.60,61 All these biochemical actions of NFκB indicate that this transcrip-
tion factor can exert an antiapoptotic effect, thereby protecting neurons against
degeneration.58,60 As we will discuss below, these data are consistent with clinical
findings showing increased levels of NFκB in vulnerable regions of the central
nervous system of AD, PD and ALS.62–64
have much higher silicon intakes than do Western populations because of higher
intakes of plant-based foods86 and it may be significant that Indians have among
the lowest rates of AD.87 More recently, it was found that subjects with low plasma
vitamin E concentrations are at higher risk of developing a dementia in subsequent
years.88 However, this is a very controversial issue in that data obtained from the
Honolulu–Asia Aging Study, a prospective community-based study of Japanese-
American men who were age 45 to 68 in 1965 to1968, show that midlife dietary
intake of antioxidants does not modify the risk of late-life dementia or its most
prevalent subtypes.89 Thus, intake of β-carotene, vitamin C and flavonoids was
not associated with the risk of dementia and its subtypes either at 6 years or at
26 years of follow-up.89,90
Epidemiological studies have also found an inverse association between high
intake of dietary vitamin E (but not flavonoids or vitamin C) and the occurrence
of PD.69,91 However, these data were not confirmed by other studies,66,92 although
Hellenbrand et al.66 reported a significant statistical trend toward a protective
effect of vitamin C in PD. Individuals over the age of 65 that had higher levels
of β-carotene performed better on learning and memory tests compared with
individuals with low β-carotene levels.93 Lycopene is a carotenoid that has been
suggested to protect against heart disease, stroke and certain cancers.94,95 Lyco-
pene can protect cultured hippocampal neurons against amyloid-beta (Aβ) and
glutamate toxicity.3 Uric acid is markedly effective in protecting cultured neurons
against insults relevant to AD and PD, including exposure to Aβ and iron.96,97
The clinical findings indicating a protective effect of dietary flavonoids against
neurodegenerative disease are supported by data obtained in laboratory animals
showing that a diet supplemented with fruits and vegetables rich in antioxidants
(blueberries, strawberries and spinach) can have beneficial effects on age-related
decline of neuronal and cognitive function in old rats.98
This chapter will focus on the actions of in vitro application of natural
nutritional antioxidants in experimental models of neurodegenerative disorders.
The capability of these compounds to counteract the damaging effects of ROS,
and the relevance of this biochemical effect in their putative neuroprotective action
will be examined. Among the numerous biochemical effects of ROS and antiox-
idants, particular emphasis will be given to their interference with NFκB function,
whose role in the pathophysiology of neurodegenerative disorders is gaining
increasing attention. Moreover, the effects of the administration of “pharmaco-
logical” doses of nutritional antioxidants in animal models and in patients with
AD, PD and ALS will be reviewed. Finally, a detailed analysis on the role of
dietary intake of polyphenols and other antioxidant vitamins in the prevention of
AD and PD will be carried out.
1. Flavones
2. Flavanones
3. Isoflavones
4. Flavonols
5. Catechins
6. Anthocyanins
R1
R1
O
O R2
R2
OH
OH R3
R3
R1 = R2 = OH, R3 = H : Protocatechuic acid R1 = OH : Coumaric acid
R1 = R2 = R3 = OH : Gallic acid R1 = R2 = OH : Caffeic acid
R1 = OCH3 = OH : Ferulic acid
O
HO OH
O
OH
H O
HO H
H OH OH
Chlorogenic acid
Flavonoids
Stilbenes Lignans
CH3O CH2OH
HO
HO CH2OH
OH
OCH3
HO OH
Resveratrol Secoisolariciresinol
Flavonols Flavones
R1
R1
R2
R2
HO O
R3 HO O
R3
OH
OH O
OH O
R2 = OH; R1 = R2 = H : Kaempferol R1 = H ; R2 = OH : Apigenin
R1 = R2 = OH ; R3 = H : Quercetin R1 = R2 = OH : Luteolin
R1 = R2 = R3 = OH : Myricetin
Flavanones
Isoflavones R1
R2
HO O
HO O
R3
R1 O OH O
OH
R1 = H; R2 = OH : Narigenin
R1 = H : Daidzein R1 = R2 = OH : Eriodictyol
R1 = OH : Genistein R1 = OH; R2 = OCH3 : Hesperetin
Flavanols
Anthocyanidins R1
R1
R2
OH
HO O
+
HO O R3
R2
OH
OH
OH
OH R1 = R2 = OH; R3 = H : Catechins
R1 = R2 = R3 = OH : Gallocatechin
R1 = R2 = H : Pelargonidin
R1 = OH; R2 = H : Cyanidin OH
R1 = R2 = OH : Delphinidin
R1 = OCH3; R2 = OH : Petunidin HO O
OH
R1 = R2 = OCH3 : Malvidin
OH
OH
OH
HO O
OH
OH OH
OH
HO O
OH
OH
OH
Trimeric procyanidin
not glycosylated in foods. The tea epicatechin is remarkably stable when exposed
to heat as long as the pH is acidic; only 15% of this substance is degraded after
7 h in boiling water at pH 5.99
Proanthocyanidins, which are also known as condensed tannins, are dimers,
oligomers and polymers of catechins that are bound together by links between
C4 and C8 (or C6). Through the formation of complexes with salivary proteins,
condensed tannins are responsible for the astringent character of fruit (grapes,
peaches, kakis, apples, pears, berries, etc.) and beverages (wine, cider, tea, beer,
etc.) and for the bitterness of chocolate.99 This astringency changes over the course
of maturation and often disappears when the fruit reaches ripeness; this change
has been well explained in the kaki fruit by polymerization reactions with ace-
taldehyde. Such polymerization of tannins probably accounts for the apparent
reduction in tannin content that is commonly seen during the ripening of many
types of fruit. It is difficult to estimate the proanthocyanidin content of foods
because they have a wide range of structures and weights. Anthocyanins are
pigments dissolved in vacuolar sap of the epidermal tissues of flowers and fruit,
to which they impart a pink, red, blue or purple color.99 They exist in different
chemical forms, both colored and uncolored, according to pH. Although they are
highly unstable in the aglycone form (anthocyanidins) while they are in plants,
they are resistant to light, pH and oxidation conditions, which are likely to degrade
them. In the human diet, anthocyanins are found in red wine, certain varieties of
cereals and certain leafy and root vegetables (aubergines [eggplant], cabbage,
beans, onions, radishes), but they are most abundant in fruit.
Cyanidin is the most common anthocyanidin in foods. Food contents are
generally proportional to color intensity and reach values up to 2 to 4 g/kg fresh
weight in blackcurrants or blackberries. These values increase as the fruit ripens.
Anthocyanins are found in the skin of certain types of red fruit and may occur
in the flesh (cherries and strawberries) as well. Wine contains 200 to 350 mg
anthocyanins/l, and these anthocyanins are transformed into various complex
structures as the wine ages.99 Stilbenes are found in only low quantities in the
human diet. One of these, resveratrol, for which anticarcinogenic effects have
appeared during screening of medical plants and which has been extensively
studied, is found in low quantities in wine (0.3 to 7 mg aglycone/l and 15 mg
glycosides/l in red wine). However, because resveratrol is found in such small
quantities in the diet, any protective effect of this molecule is unlikely at normal
nutritional intake.99
In most cases, foods contain complex mixtures of polyphenols, which are
often poorly characterized. Apples, for example, contain flavanol monomers
(mainly epicatechin) or oligomers (procyanidin B2 mainly), chlorogenic acid
and small quantities of other hydroxycinnamic acids, two glycosides of phlo-
retin, several quercetin glycosides and anthocyanins, such as cyanidin 3-galac-
toside in the skin of certain red varieties. Apples are one of the rare types of
food for which fairly precise data on polyphenol composition between varieties
of apples have notably been studied. The polyphenol profiles of all varieties of
apples are practically identical, but concentrations may range from 0.1 to 5 g
DK5836_C005.fm Page 140 Monday, May 22, 2006 1:14 PM
studies have shown that moderate wine consumption can be protective against
neurological disorders, such as age-related macular degeneration127,128 and AD.68
Moreover, in vitro and in vivo preclinical studies have shown the neuroprotective
effect of lyophilized red wine,129 grape polyphenols,130 quercetin,131 trans-
resveratrol132–134 and (+)-catechin.135 Taken together, these findings raise the pos-
sibility that red wine constituents may be beneficial in the prevention of age-
related neurodegenerative disorders. There is also increasing interest for the role
of tea (Camellia sinensis) in maintaining health and in treating disease. Although
tea consists of several components, research has focused on polyphenols, espe-
cially those found in green tea. The green tea polyphenols include (—)-epicatechin
(EC), (—)-epigallocatechin (EGC), (—)-epicatechin-3-gallate (ECG), (—)-epi-
gallocatechin-3-gallate (EGCG). Of these, EGCG generally accounts for greater
than 40% of the total.136 Green tea polyphenols are potent antioxidants.106 EGCG
usually has the greatest antioxidant activity and is the most widely studied
polyphenol for disease prevention.136–138 Many of the putative health benefits of
tea are presumed to stem from its antioxidant effects.
The epidemiological evidence indicating the putative role of nutritional anti-
oxidants in the prevention and attenuation of neurodegenerative disorders is
receiving experimental confirmation in a number of laboratory studies. Thus, the
polyphenol epicatechin was shown to attenuate neurotoxicity induced by oxidized
low-density lipoprotein in mouse-derived striated neurons.138 Tea extracts and
EgCG attenuated the neurotoxic action of 6-OHDA in rat PC12 cells, human
neuroblastoma SH-SY5Y cells,137 and was shown to be neuroprotective in a
mouse model of PD.138 Moreover, recent reports have revealed that flavonoids
may be neuroprotective in neuronal primary cell cultures. For example, the ginkgo
biloba extract, enriched with flavonoids, has been shown to protect hippocampal
neurons from nitric oxide or β-amyloid derived, peptide-induced neurotoxic-
ity.139–141 In addition, the extract of ginkgo biloba, referred to as Egb 761, is one
of the most popular plant extracts used in Europe to alleviate symptoms associated
with a range of cognitive disorders.142,143 The mechanism of action of Egb 761
in the central nervous system is only partially understood, but the main effects
seem to be related to its antioxidant properties, which require the synergistic
action of the flavonoids, the terpenoids (ginkgolides, bilobalide), and the organic
acids, principal constituents of Egb 761.144 These compounds, to varying degrees,
act as scavengers of ROS, which have been considered the mediators of the
excessive lipid peroxidation and cell damage observed in AD.145–147
p65, p50 and an inhibitory subunit called IκB.148,149,154 When IκB is bound to
p50/p65, it is inactive; signals that activate NFκB cause dissociation of IκB
releasing p50/p65, which then translocates to the nucleus and binds to specific
κB DNA consensus sequences in the enhancer region of a variety of κB-respon-
sive genes.58,60,148,149,154,155
In neurons, NFκB is activated by various intracellular signals, including
cytokines, neurotrophic factors, and neurotransmitters.58,154,156 Activation of
glutamate receptors and membrane depolarization lead to activation of NFκB in
hippocampal pyramidal neurons and cerebellar granule neurons in culture.152,157
The mechanism whereby diverse stimulants lead to the activation of NFκB has
been a subject of intense research. Most work has focused on the p50/p65 dimer,
the predominant form of NFκB activated in many cells including neurons,58,60,148
and its association with IB. It is now known that upon stimulation with many
NFκB inducers, IκBα is rapidly phosphorylated on two serine residues (S32 and
S36), which target the inhibitor protein for ubiquitination and subsequent degra-
dation by the 26S proteasome.155 Released NFκB dimer can then translocate to
the nucleus and activate target genes by binding with high affinity to κB elements
in their promoters. The phosphorylation and degradation of IκBα are tightly
coupled events, so it is likely that agents that activate NFκB do so by stimulating
a specific IκB kinases, or alternatively by inactivating a particular phosphatase.
Two IκB kinases (IKKs) termed IKKα and IKKβ have been described in
research.155 IKKα and β have been shown to be activated by important inducers
of NFκB, such as IL-1 and TNF, to specifically phosphorylate S32 and S36 of
IκBα, and to be crucial for NFκB activation by these cytokines.155 The IKKs are
part of a larger multiprotein complex called the IKK signalsome. It appears that
multiple pathways can regulate NFκB, most of which lead to IκB phosphorylation
via the IKK-containing signalsome155 One model has been proposed whereby
diverse agents all activate NFκB by causing oxidative stress.57,149,158 This hypoth-
esis is based on four main lines of evidence:
Thus, in 1992 the protective effect of vitamin E was first described on neurons
in culture against Aβ–induced cell death.201 Following these initial findings, a
number of subsequent studies confirmed the role of oxidative stress in the neu-
rotoxic effect of Aβ peptide. For example, Behl et al.214 found that Aβ can induce
the formation of H2O2 in hippocampal neurons, which causes peroxidation of cell
membranes and ultimately leads to neuronal death. Consistent with these findings,
exposure of cultured hippocampal neurons to Aβ induced a significant increase
in 4-HNE.215 Moreover, it has recently been found that the phenolic antioxidant
curcumin, which is largely used as a food preservative and herbal medicine in
India, reduces oxidative damage and amyloid pathology in a transgenic mouse
model of AD.216 However, in another study, Aβ-induced neurotoxicity in rat
hippocampal neurons in culture was not affected by several antioxidants;217 nev-
ertheless, pretreatment of cultures with Aβ significantly increased the sensitivity
of neurons to H2O2, suggesting that Aβ can render neurons more susceptible to
ROS damage.217 Some of the proteins oxidatively modified by Aβ−induced oxi-
dative stress include membrane transporters, receptors, GTP-binding proteins (G
proteins) and ion channels. Oxidative modifications of tau by 4HNE and other
ROS can promote its aggregation and, thereby, may induce the formation of
neurofibrillary tangles. Aβ also causes mitochondrial oxidative stress and dysreg-
ulation of Ca2+ homeostasis, resulting in impairment of the electron transport
chain, increased production of superoxide anion radicals and decreased produc-
tion of ATP.
In agreement with data obtained in experimental models, clinical findings
indicate that oxidative stress occurs in AD, as indicated by the finding that higher
than normal levels of lipid, protein and DNA oxidation are found in the brains
of AD patients.46,48,145,146,218 Thus, lipid peroxidation, measured as thiobarbituric
acid reactive substances (TBARS), was found to be increased in various brain
regions of AD patients.219–221 Moreover, Mecocci et al.53 found a significant three-
fold increase in mitochondrial DNA oxidation in the parietal cortex of AD
patients. In addition, immunohistochemical analysis of brain sections from AD
patients using an antibody with selectivity for the activated nuclear form of p65
revealed that NFκB was activated in neurons and astrocytes.63 Cells with activated
NFκB were restricted to the close proximity of early plaque stages.63 Thus, it is
possible that Aβ-induced NFκB activation contributes to the pathological changes
observed in AD via the induction of proinflammatory and cytotoxic genes or,
DK5836_C005.fm Page 148 Monday, May 22, 2006 1:14 PM
more likely, that Aβ-induced NFκB activation is part of a cellular defense pro-
gram.
Based on the preclinical and clinical data indicating the presence of oxidative
stress in AD patients, clinical trials were carried out to test the effect of antiox-
idants in this pathological condition. However, as already indicated above, incon-
sistent findings were reported in the trials investigating the effects of antioxidant
vitamins on cognitive function and dementia. Thus, a controlled clinical trial with
dl-α-tocopherol (synthetic form: 2000 IU/d) in patients with moderately severe
impairment from AD showed some beneficial effects with respect to rate of
deterioration of cognitive function.222 In the same dl-α-tocopherol clinical trial,
selegiline (10 mg/d), a monoamine oxidase inhibitor, produced beneficial effects
similar to that produced by dl-α-tocopherol.222 It is interesting to note that there
was no significant difference in effect between the groups receiving a combination
of dl-α-tocopherol and selegiline and those receiving treatment with the individual
agents.146,222 Several possibilities were proposed to explain the lack of additive
effect. One was that selegiline and vitamin E can act by the same mechanism.
Indeed, both reduce the levels of free radicals, although by different molecular
pathways. Vitamin E protects neurons by destroying formed ROS (“quenching”),
whereas selegiline protects neurons by preventing the formation of ROS and by
inhibiting oxidative metabolism of catecholamines. Therefore, clinical studies
involving vitamin E and selegiline support the concept that ROS are one of the
intermediary risk factors for the progression of neurodegeneration in AD.198
However, in the MRC/BHF Hearth Protection Study, which included 20,536
persons allocated to receive either antioxidant vitamin supplementation (vitamins
E and C and β-carotene) or a placebo, no treatment differences were found in
the percentage of persons defined as cognitively impaired or in mean cognitive
scores after 5 years of treatment.223 In addition, no difference was found in the
number of persons who developed dementia during follow-up.
Another clinical study was conducted among 1059 rural, noninstitutionalized
elderly residents of southwestern Pennsylvania, who were participants in the
Monongahela Valley Independent Elders Survey.224 Current use of nutritional
supplements containing vitamins A, C, or E, β-carotene, zinc or selenium was
measured through self-report. After adjustment for age, education and sex, no
significant differences were found in cognitive test performance between antiox-
idant users and nonusers.224 However, in the Rotterdam Study, Jama et al.225
studied 5182 elderly persons and found that dietary and nutritional supplement
intake of β-carotene was inversely associated with cognitive impairment, even
after adjustment for age, sex, education, smoking, total caloric intake and con-
sumption of other antioxidants. The discrepancy between the results of the study
of Mendelsohn et al.224 and that of Jama et al.225 could be due to differences in
the study populations, such as difference in age distribution or socioeconomic
status or the exclusion of demented persons in the Rotterdam study. Although
Jama et al.225 found an association between β-carotene and cognition, they did
not find similar results with vitamins E or C. Nevertheless, the data of a protective
effect of β-carotene against age-related cognitive impairment were not confirmed
DK5836_C005.fm Page 149 Monday, May 22, 2006 1:14 PM
in a more recent study.226 Thus, from the analysis of the data from the Washington
Heights-Inwood Columbia Aging Project (WHICAP), results indicated that the
risk of AD was not associated with supplement, dietary or total intake of car-
otenes, vitamin C or vitamin E.226 In contrast, a recent prospective study has
shown a reduced prevalence and incidence of AD in individuals taking vitamins
E and C in combination.227 However, there was no significant reduction in risk
of incident AD with vitamin E or vitamin C alone or with a multivitamin. There
was also no association between AD risk and use of B-complex vitamins.227 In
two other investigations, an association of high folic acid levels and decreased
homocysteine levels with reduced AD risk was found.228,229 Given the conflicting
data reported in many of the trials to date, it is clear that more effort is necessary
in the future to try to confirm whether or not there is a relationship between
dietary habits (in particular, the amount of polyphenols and other antioxidants
intake) and the risk of dementia.
Increasing evidence suggests that diets high in saturated fats may increase
the risk of AD, whereas diets rich in mono- and polyunsaturated fatty acids may
decrease the risk. Several studies indicate that diets rich in specific long-chain,
polyunsaturated fatty acids of the omega-3 series, such as those found in fish,
can reduce the risk of AD.230 Recent studies have extended and confirmed data
showing the protective effect of omega-3 fatty acids against AD.231 Moreover,
there is epidemiological data suggesting an association between an inadequate
intake of fish oils and a greater than expected incidence of late onset dementia.232
Interestingly, experimental animal studies support these epidemiological data in
that there is evidence that a diet enriched with docosahexaenoic acid (DHA,
22:6n-3) reduces the burden of β-amyloid peptide in a mouse model of AD.233
Although the emerging data linking fatty acids to AD are encouraging, the
potential of dietary modifications of fat intake to affect disease risk remains to
be established. There is also emerging evidence suggesting that cognitively stim-
ulating environments, physical exercise and diets low in energy and fats (choles-
terol and saturated fatty acids) may reduce the risk of AD.193,228,234 Exercise,
cognitive stimulation and dietary restriction may each exert a beneficial effect
through a similar mechanism involving increased production of brain-derived
neurotrophic factor (BDNF).228,235,236 The possibility that the risk of AD can be
reduced by modifications of diet and lifestyle is of considerable interest and
suggests the potential for reducing the incidence of AD by preventive strategies
similar to those that reduce the risk of cardiovascular disease.
DA, and the loss of dopaminergic influence on other structures in the basal ganglia
leads to the classic Parkinsonian symptoms. Moreover, PD is characterized by
degeneration of monoamine-containing neurons in the brain stem nuclei (pre-
dominantly the locus coeruleus) and is variably associated with pathology in
nonnigral systems causing multiple neurotransmitter dysfunctions.239
Parkinson’s disease develops much less frequently than Alzheimer’s disease,
ranging from 0.1 to 0.5% annually. Depending on the study, the annual incidence
for PD ranges from 0.1 to 0.3% for those over 50 years, to 0.4% for those over
80 years and as high as 2.6% for those 85 to 98 years of age.240–242 Incidence
rates for PD increase with age both in men and women, but the rate in men
exceeds that for women by two-fold.241 The average duration of PD is 9 years.
From birth, the lifetime risk of developing PD is about 2% for men and 1.3% for
women.243 Although idiopathic PD is usually sporadic, it is now well established
that there is a genetic component to the disease.244,245 Approximately 5 to 10%
of PD patients have a familial form of Parkinsonism with an autosomal-dominant
pattern of inheritance.245 Case control studies have typically indicated a 2- to 14-
fold increase in incidence in close relatives of PD patients,246 and although
concordance rates between identical twins are low for overt expression of the
disease, they are much higher when subclinical decline in striatal dopaminergic
dysfunction is measured by positron emission tomography (PET) imaging (53%
in monozygotic twins of PD patients, compared with 13% in dizygotic cases).247
A specific mutation in exon 4 of the gene encoding α-synuclein has been identified
as a causative factor of Parkinsonism in a family from southern Italy.248 Subse-
quently, this same mutation was found in other Greek and Brazilian families.246,249
Kruger et al.250 reported a second mutation in a German family. The parkin
gene mutation was first described in a Japanese family with autosomal recessive,
levodopa-responsive disease characterized by degeneration of the substantia nigra
and the absence of Lewy bodies.251 Over 20 different mutations have since been
identified252 and these mutations are now considered to be the most common
cause of familial PD. Nevertheless, in sporadic PD, environmental factors have
been emphasized.253 Epidemiological studies have correlated a number of poten-
tial factors as those that may increase the risk of developing PD.254 These include
exposure to well water, herbicides, industrial chemicals, wood pulp mills, farming
and living in a rural environment. A number of exogenous toxins have been
associated with the development of Parkinsonism, including trace metals, cyanide,
lacquer thinner, organic solvents, carbon monoxide, and carbon disulfide.245 There
has also been interest in the possible role of endogenous toxins, such as tetrahy-
droisoquinolines and β-carbolines. However, no specific toxin has been found in
the brain of PD patients. The most compelling evidence for an environmental
factor in PD relates to the toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine
(MPTP). MPTP is a biproduct of the illicit manufacture of a synthetic meperidine
derivative. Some drug addicts who took MPTP developed a syndrome that strik-
ingly resembled PD, both clinically and pathologically.255,256 MPTP induces tox-
icity through its conversion in astrocytes to the pyridinium ion (MPP+) in a
reaction catalyzed by monoamine oxidase B (MAO-B).11 MPP+ is then taken up
DK5836_C005.fm Page 151 Monday, May 22, 2006 1:14 PM
Various drugs, which can act by reducing oxidative stress, have been used as
potential therapeutic agents in transgenic mice expressing the mutated human
SOD-1 enzyme. Thus, polyamine- or putrescine-modified catalase, an antioxidant
enzyme that removes hydrogen peroxide and has good permeability at the
blood–brain barrier, increases the survival of transgenic mice bearing the human
mSOD-1G93A.337,338 Moreover, the copper chelator and thiol compound penicil-
lamine, the copper chelator trientine, carboxyfullerenes, vitamin E and N-acetyl-
cysteine have been reported to increase the survival time in this mouse model
and/or delay the onset of the disease to a small extent.339–342 The drug riluzole,
which inhibits glutamate release at presynaptic terminals, also extends lifespan
slightly in human mSOD-1G93A transgenic mice.342 Interestingly, riluzole, which
is used clinically in patients with ALS,343 has been shown to have direct antiox-
idative effect on cultured cortical neurons.344 However, no clear evidence for a
beneficial effect of α-tocopherol, selegiline, N-acetylcysteine or an antioxidant
cocktail has been obtained in humans.345–347
Li et al.348 have recently reported that blockade of caspase-1 and caspase-3
activity by N-benzyloxycarbonyl-Val-Asp-fluoromethylketone (zVAD-fmk) pro-
longs the survival of transgenic mice expressing the human mSOD-1G93A that
begin to develop ALS symptoms at the mean age of 3 months. These findings
open new perspectives for the use of caspase inhibitors as potential therapeutic
agents in the treatment of ALS and other neurodegenerative diseases. However,
because of the low oral bioavailability and limited brain penetrance, zVAD-fmk
was delivered by intracerebral administration. Thus, the physicochemical char-
acteristics of zVAD-fmk might limit its clinical usefulness. Based on these find-
ings and on the hypothesis that in transgenic mice expressing the human mSOD-
1G93A an increased formation of ROS occurs, we decided to treat them with
lyophilized red wine (which is rich in antioxidant compounds) dissolved in the
drinking water that was freely available to the animals. This treatment regimen
caused a significant reduction in the overall mortality of the treated mice as
compared with control animals. Thus, lyophilized wine prolonged by 6% the
survival of mSOD1G93A mice.349
In the first series of experiments, the onset of treatment was variable and
ranged from 43 to 66 days of age for the mice.349 We have recently repeated the
experiments on mSOD1G93A mice that were treated with the same concentration
of lyophilized red wine, but the treatment was started earlier, i.e., 30 to 40 days
from birth. By using this protocol, we have found that administration of lyo-
philized red wine significantly increased the mean survival time by 15%, as
compared with control transgenic mice given drinking water only. The calculated
concentration of polyphenolic compounds, expressed as gallic acid equivalent
(GAE), was 4824 mg/l. Considering that each mouse drank about 4 ml of liquid
daily, it is possible to calculate the daily intake of GAE, which was about 20 mg
per mouse. It is tempting to speculate that the mechanism of neuroprotection
exerted by lyophilized red wine on mSOD1G93A mice might be due to its ability
to inhibit caspase-3 activity. This hypothesis is based on in vitro experiments
showing that lyophilized red wine (5 µg/ml) caused a significant inhibition of
DK5836_C005.fm Page 156 Monday, May 22, 2006 1:14 PM
5.7 CONCLUSIONS
There is growing evidence that oxidative stress may play an important role in the
pathogenesis of AD, PD and ALS. However, in spite of the large body of exper-
imental data showing the protective effect of antioxidants in in vitro models of
neurodegeneration and in some in vivo animal models, there is still limited
evidence for a neuroprotective effect of antioxidants in the treatment of neuro-
degenerative disorders in humans. There may be several reasons for this discrep-
ancy between preclinical and clinical data. Many laboratory studies use models
of oxidative stress and investigate rescue by antioxidant agents. These models
normally use acute high doses of antioxidants that far exceed those usually
ingested via dietary sources.350 These types of studies are, therefore, unlikely to
be comparable to dietary exposure to antioxidants. Moreover, it is conceivable
that the therapeutic regimen used so far (e.g., one or two antioxidants) might not
be sufficient to halt the neuropathologic process. As pointed out by others, a more
efficient strategy would be the use of multiple antioxidants in the treatment of
AD, PD and ALS.146 In this regard, it is important to point out that one possible
advantage of the use of extracts of fruits, vegetables or beverages (such as red
wine, green tea or ginkgo biloba) in the treatment of neurodegenerative disorders,
is that they often contain multiple antioxidant compounds that can potentiate each
other. Consistent with this line of reasoning, it has recently been shown that a
complex antiaging dietary supplement composed of 31 ingredients, most of them
with antioxidant activity, is capable of blocking age-related cognitive decline in
transgenic mice expressing high levels of ROS-mediated processes.351 Particularly
important would be the use of lyophilized red wine,349 which is provided with
strong antioxidant capacity.73,74
One possible limitation of the neuroprotective strategy (including antioxidant
administration) might be consequent to the fact that when overt symptomatology
of AD, PD and ALS occurs, a certain amount of neuronal death has already
happened. Thus, the neuroprotective agents (including antioxidants) can, at best,
only rescue the surviving neurons, an effect which might not be sufficient to
attenuate the neurological symptomatology. Nevertheless, recent advances
DK5836_C005.fm Page 157 Monday, May 22, 2006 1:14 PM
suggest that the goal of curing patients with age-related neurodegenerative dis-
orders is worth pursuing. One reason for optimism is that the extent of neuronal
loss in AD and PD patients during the early period of the disease may not be as
great as initially thought because many dysfunctional neurons may be able to
recover.350 It is, therefore, important to start the therapeutic intervention at an
early stage of the disease process. In this regard, it is interesting to note that some
epidemiological studies have shown that dietary habits can influence the incidence
of neurodegenerative disorders. In particular, it was found that a diet rich in
vitamin E can reduce the risk for PD69,91 and that moderate wine consumption
may decrease the risk for AD.67,68 However, there are still few and controversial66,92
epidemiological data on this important point, which might be partly due to the
intrinsic difficulties in performing epidemiological surveys regarding the dietary
habits of large populations. Nevertheless, it is desirable that future studies aimed
at investigating the relationship between dietary antioxidant intake and the relative
risk for neurodegenerative disorders such as AD, PD and ALS will shed more
light on this very important aspect of public health.
5.8 ACKNOWLEDGMENTS
This work was supported by the Italian MIUR (Ministero Istruzione Università
Ricerca) L488/92 project n. s209-p/f. The author is very grateful to Dr. Andreina
Poggi for her critical reading of this manuscript.
REFERENCES
1. Evans DA, Funkenstein HH, Albert MS, Scherr PA, Cook NR, Chown MJ, Hebert
LE, Hennekens CH, Taylor JO. Prevalence of Alzheimer’s disease in a community
population of older persons. JAMA 1989; 262:2551–2556.
2. Mattson MP. Pathways towards and away from Alzheimer’s disease. Nature 2004:
430:631–639.
3. Mattson MP, Chan SL, Duan W. Modification of brain aging and neurodegener-
ative disorders by genes, diet, and behavior. Physiol Rev 2002; 82: 637–672.
4. Zhu H, Guo Q, Mattson MP. Dietary restriction protects neurons against the death-
promoting action of a presenilin-1 mutation. Brain Res 1999; 842:224–229.
5. Bowling AC, Beal MF. Bioenergetic and oxidative stress in neurodegenerative
diseases. Life Sci 1995; 56:1151–1171.
6. Jenner P. Oxidative damage in neurodegenerative disease. Lancet 1994;
344:796–798.
7. Jenner P. Oxidative mechanisms in nigral cell death in Parkinson’s disease. Mov
Disord 1998; 13:24–34.
8. Jenner P, Olanow CW. Oxidative stress and the pathogenesis of Parkinson’s dis-
ease. Neurology 1996; 47(Suppl 3):S161–S170.
9. Olanow CW. A radical hypothesis for neurodegeneration. Trends Neurosci 1993;
16:439–444.
DK5836_C005.fm Page 158 Monday, May 22, 2006 1:14 PM
10. Halliwell B, Gutteridge JMC. Oxygen radicals and the nervous system. Trends
Neurosci 1985; 6:22–26.
11. Simonian NA, Coyle JT. Oxidative stress in neurodegenerative diseases. Annu Rev
Pharmacol Toxicol 1996; 36:83–106.
12. Andersen JK. Oxidative stress in neurodegeneration: cause or consequence? Nat
Med 2004; 10 Suppl:S18–S25.
13. Jain A, Martensson J, Stole E, Auld PA, Meister A. Glutathione deficiency leads
to mitochondrial damage in brain. Proc Natl Acad Sci USA 1991; 88:1913–17.
14. Koppal T, Drake J, Yatin S, Jordan B, Varadarajan S, Bettenhausen L, Butterfield
DA. Peroxynitrite-induced alterations in synaptosomal membrane proteins: insight
into oxidative stress in Alzheimer’s disease. J Neurochem 1999; 72:310–317.
15. Markesbery WR, Carney JM. Oxidative alterations in Alzheimer’s disease. Brain
Pathol 1999; 9:133–48.
16. Schulz JB, Weller M, Klockgether T. Potassium deprivation-induced apoptosis of
cerebellar granule neurons: a sequential requirement for new mRNA and protein
synthesis, ICE-like protease activity, and reactive oxygen species. J Neurosci 1996;
16:4696–4706.
17. Sée V, Loeffler J-P. Oxidative stress induces neuronal death by recruiting a protease
and phosphatase-gated mechanism. J Biol Chem 2001; 276:35049–35059.
18. Sastry PS, Rao KS. Apoptosis and the nervous system. J Neurochem 2000;
74:1–20.
19. Beal MF. Mitochondria, free radicals, and neurodegeneration. Curr Opin Neuro-
biol 1996; 6:661–666.
20. Beal MF. Oxidative damage in neurodegenerative diseases. Neuroscientist 1997,
3:21–27.
21. Beal MF, Ferrante RJ, Browne SE, Matthews RT, Kowall NW, Brown RH Jr.
Increased 3-nitrotyrosine in both sporadic and familial amyotrophic lateral scle-
rosis. Ann Neurol 1997, 42:644–654.
22. Castilho RF, Ward MW, Nicholls DG. Oxidative stress, mitochondrial function,
and acute glutamate excitotoxity in cultured cerebellar granule cells. J Neurochem
1999; 72:1394–1401.
23. Gassem M, Youdim MBH. Free radical scavengers: chemical concepts and clinical
relevance. J Neural Transm 1999; 56:193–210.
24. Ishikawa Y, Satoh T, Enokido Y, Nishio C, Ikeuchi T, Hatanaka H. Generation of
reactive oxygen species, release of L-glutamate and activation of caspases are
required for oxygen-induced apoptosis of embryonic hippocampal neurons in
culture. Brain Res 1999; 824:71–80.
25. Tong L, Toliver-Kinski T, Taglialatela G, Werrbach-Perez K, Wood T, Perez-Polo
R. Signal transduction in neuronal death. J Neurochem 1998; 71:447–459.
26. Yuan J, Yankner BA. Apoptosis in the nervous system. Nature 2000; 407:802–809.
27. Junn E, Mouradian MM. Apoptotic signaling in dopamine-induced cell death: the
role of oxidative stress, p38 mitogen-activated protein kinase, cytochrome c and
caspases. J Neurochem 2001, 78:374–383.
28. Martin LJ. Neuronal death in amyotrophic lateral sclerosis is apoptosis: possible
contribution of a programmed cell death mechanism. J Neuropathol Exp Neurol
1999; 58:459–471.
29. Mochizuki H, Goto K, Mori H, Mizuno Y. Histochemical detection of apoptosis
in Parkinson’s disease. J Neurol Sci 1996; 137:120–123.
DK5836_C005.fm Page 159 Monday, May 22, 2006 1:14 PM
50. Alam ZI, Jenner A, Daniel SE, Lees AJ, Cairns N, Marsden CD, Jenner P, Halliwell
B. Oxidative DNA damage in the parkinsonian brain: an apparent selective
increase in 8-hydroxyguanine levels in the substantia nigra. J Neurochem 1997;
69:1196–1203.
51. Ferrante RJ, Browne SE, Shinobu LA, Bowling AC, Baik MJ, MacGarvey U,
Kowall NW, Brown RH Jr, Beal MF. Evidence of increased oxidative damage in
both sporadic and familial amyotrophic lateral sclerosis. J Neurochem 1997;
69:2064–2074.
52. Gabbita SP, Lovell MA, Markesbery WR. Increased nuclear oxidation in the brain
in Alzheimer’s disease. J Neurochem 1998; 71:2034–2040.
53. Mecocci P, MacGarvey U, Beal MF. Oxidative damage to mitochondrial DNA is
increased in Alzheimer’s disease. Ann Neurol 1994; 36:747–751.
54. Zhang J, Perry G, Smith MA, Robertson D, Olson SJ, Graham DG, Montine TJ.
Parkinson’s disease is associated with oxidative damage to cytoplasmic DNA and
RNA in substantia nigra neurons. Am J Pathol 1999; 154:1423–1429.
55. Dalton TP, Shertzer HG, Puga A. Regulation of gene expression by reactive
oxygen. Annu Rev Pharmacol Toxicol 1999; 39:67–101.
56. Flohé L, Brigelius-Flohé, Saliou C, Traber MG, Packer L. Redox regulation of
NF-ΚB activation. Free Radic Biol Med 1997; 22:1115–1126.
57. Li N, Karin M. Is NF-κB the sensor of oxidative stress? FASEB J 1999;
13:1137–1143.
58. Mattson MP, Culmsee C, Yu ZF, Camaldola S. Roles of nuclear factor-κB in
neuronal survival and plasticity. J Neurochem 2000; 74:443–456.
59. Pinkus R, Weiner LM, Daniel V. Role of oxidants and antioxidants in the induction
of AP-1, NF-κB, and glutathione s-transferase gene expression. J Biol Chem 1996;
271:13422–13429.
60. Mattson MP, Camaldola S. NF-κB in neuronal plasticity and neurodegenerative
disorders. J Clin Invest 2001; 107:247–254.
61. Tamatani M, Che YH, Matsuzaki H, Ogawa S, Okado H, Miyake S, Mizuno T,
Tohyama M. Tumor necrosis factor induces Bcl-2 and bcl-x expression through
NF-κB activation in primary hippocampal neurons. J Biol Chem 1999;
274:8531–8538.
62. Hunot S, Brugg B, Ricard D, Michel PP, Muriel MP, Ruberg M, Facheux BA.
Agid Y, Hirsch EC. Nuclear translocation of NF-κB is increased in dopaminergic
neurons of patients with Parkinson’s disease. Proc Natl Acad Sci USA 1997;
94:7531–7536.
63. Kaltschmidt B, Uherek M, Volk B, Baeuerle PA, Kaltschmidt C. Transcription
factor NF-κB is activated in primary neurons by amyloid β peptides and in neurons
surrounding early plaques from patients with Alzheimer’s disease. Proc Natl Acad
Sci USA 1997; 94:2642–2647.
64. Migheli A, Piva R, Atzori C, Troost D, Schiffer D. c-Jun, JNK/SAPK kinase and
transcription factor NF-κB are selectively activated in astrocytes, but not motor
neurons, in amyotrophic lateral sclerosis. J Neuropathol Exp Neurol 1997;
56:1314–1322.
65. Deschamps V, Barberger-Gateau P, Peuchant E, Orgogozo JM. Nutritional factors
in cerebral aging and dementia: epidemiological arguments for a role of oxidative
stress. Neuroepidemiology 2001; 20:7–15.
DK5836_C005.fm Page 161 Monday, May 22, 2006 1:14 PM
83. in t′ Veld BA, Ruitenberg A, Hofman A, Launer LJ, van Duijn CM, Stijnen T,
Breteler MM, Stricker BH. Nonsteroidal antiinflammatory drugs and the risk of
Alzheimer’s disease. N Engl J Med 2001; 345(21):1515–1521.
84. Brenner S. Antioxidants and the risk of Alzheimer disease. JAMA 2002; 288:2265.
85. Jugdaohsingh R, Anderson SH, Tucker KL, Elliott H, Kiel DP, Thompson RP,
Powell JJ. Dietary silicon intake and absorption. Am J Clin Nutr 2002; 75:887–93.
86. Anasuya A, Bapurao S, Paranjape PK. Fluoride and silicon intake in normal and
endemic fluorotic areas. J Trace Elem Med Biol 1996; 10:149–155.
87. Chandra V, Pandav R, Dodge HH, Johnston JM, Belle SH, DeKosky ST, Ganguli
M. Incidence of Alzheimer’s disease in a rural community in India: the Indo-U.S.
study. Neurology 2001; 57:985–989.
88. Helmer C, Peuchant E, Letenneur L, Bourdel-Marchasson I, Larrieu S, Dartigues
JF, Duborg L, Thomas M-J, Gateau-Barberger P. Association between antioxidant
nutritional indicators and the incidence of dementia: results from PAQUID pro-
spective cohort study. Eur J Clin Pharmacol 2003; 57:1555–1561.
89. Laurin D, Masaki KH, Foley DJ, White LR, Launer LJ. Midlife dietary intake of
antioxidants and risk of late-life incident dementia. The Honolulu-Asia aging
study. Am J Epidemiol 2004; 159:959–967.
90. Laurin D, Foley DJ, Masaki KH, White LR, Launer LJ. Vitamin E and C supple-
ments and risk of dementia. JAMA 2002; 288:2266–2268.
91. Golbe LI, Farrell TM, Davis PH. Case-control study of early life dietary factors
in Parkinson’s disease. Arch Neurol 1998; 45:1350–1353.
92. Logroscino G, Marder K, Cote L, Tang M-X, Shea S, Mayeux R. Dietary lipids
and antioxidants in Parkinson’s: a population-based, case-control study. Ann Neu-
rol 1996; 39:89–94.
93. Perrig WJ, Perrig P, Stahelin HB. The relation between antioxidants and memory
performance in the old and very old. J Am Geriatr Soc 1997; 45:718–724.
94. Rao AV, Agarwal S. Role of antioxidant lycopene in cancer and hearth disease. J
Am Coll Nutr 2000; 19:563–569.
95. Rissanen TH, Voutilainen S, Nyyssonen K, Lakka TA, Sivenius J, Salonen R,
Kaplan GA, Salonen JT. Low serum lycopene concentration is associated with an
excess incidence of acute coronary events and stroke: the Kuopio Ischaemic Hearth
Disease Risk Factor Study. Br J Nutr 2001; 85:749–754.
96. Keller JN, Guo Q, Holtsberg FW, Bruce-Keller AJ, Mattson MP. Increased sen-
sitivity to mitochondrial toxin-induced apoptosis in neural cells expressing mutant
presenilin-1 is linked to perturbed calciun homeostasis and enhanced oxyradical
production. J Neurosci 1998; 18:4439–4450.
97. Yu ZF, Bruce-Keller AJ, Goodman Y, Mattson MP. Uric acid protects neurons
against excitotoxic and metabolic insults in cell culture, and against focal ischemic
brain injury in vivo. J Neurosci Res 1998; 53:613-625.
98. Joseph JA, Shukitt-Hale B, Denisova NA, Bielinski D, Martin A, McEwen JJ,
Bickford PC. Reversal of age-related declines in neuronal signal transduction,
cognitive, and motor behavioral deficits with blueberry, spinach, or strawberry
dietary supplementation. J Neurosci 1999; 19:8114–8121.
99. Manach C, Scalbert A, Morand C, Rémésy C, Jiménez L. Polyphenols: food
sources and bioavailability. Am J Clin Nutr 2004; 79:727–747.
100. Middleton EJ. Effect of plant flavonoids on immune and inflammatory cell func-
tion. Adv Exp Med Biol 1998; 439:175–182.
101. Rice-Evans C. Flavonoid antioxidants. Curr Med Chem 2001; 8:797–807.
DK5836_C005.fm Page 163 Monday, May 22, 2006 1:14 PM
121. Cushman M, Nagaratham D, Burg DL, Geahlen RL. Synthesis and protein-
tyrosine kinase inhibitory activities of flavonoid analogues. J Med Chem 1991;
34:798–806.
122. Mielke K, Herdegen T. JNK a p38 stress kinases: degenerative effectors of signal-
transduction-cascades in the nervous system. Progr Neurobiol 2000; 61:45–60.
123. Davis RJ. Signal transduction by JNK group of MAP kinases. Cell 2000;
103:239–252.
124. Levites Y, Amit T, Youdim MB, Mandel S. Involvement of protein kinase C
activation and cell/survival/cell cycle genes in green tea polyphenol (–)-epigallo-
catechin 3 gallate neuroprotective action. J Biol Chem 2002; 277:30574–30580.
125. Renaud S, De Lorgeril M. Wine, alcohol, platelets, and the French paradox for
coronary heart disease. Lancet 1992; 339:1523–1526.
126. Pace-Asciak CR, Hahn S, Diamandis EP, Soleas G, Goldberg DM. The red wine
phenolics trans-resveratrol and quercetin block human platelet aggregation and
eicosanoid synthesis: implications for protection against coronary heart disease.
Clin Chim Acta 1995; 235:207–209.
127. Tsai SH, Lin-Shiau SY, Lin JK. Suppression of nitric oxide synthase and the
down-regulation of the activation of NF-κB in macrophages by resveratrol. Br J
Pharmacol 1999; 126:673–680.
128. Obisesan TO, Hirsh, R, Kosoko O, Carlson L, Parrott M. Moderate wine con-
sumption is associated with decreased odds of developing age-related macular
degeneration in NHANES-1. J Am Ger Soc 1998; 46:1–7.
129. De Ruvo C, Amodio R, Algeri S, Martelli N, Intilangelo A, D’Ancona GM,
Esposito E. Nutritional antioxidants as antidegenerative agents. Int J Dev Neurosci
2000; 18:359–366.
130. Sun GY, Xia J, Draczynska-Lusiak B, Simonyi A, Sun AY. Grape polyphenols
protect neurodegenerative changes induced by chronic ethanol administration.
NeuroReport 1999; 10:93–96.
131. Shutenko Z, Henry Y, Pinard E, Seylaz J, Potier P, Berthet F, Girard P, Sercombe
R. Influence of the antioxidant quercetin in vivo on the level of nitric oxide
determined by electron paramagnetic resonance in rat brain during global ischemia
and reperfusion. Biochem Pharmacol 1999; 57:199–208.
132. Chanvitayapongs S, Draczynska-Lusiak B, Sun AY. Amelioration of oxidative
stress by antioxidants and resveratrol in PC12 cells. Neuroreport 1997;
8:1499–1502.
133. Karlsson J, Emgård M, Brundin P, Burkitt MJ. trans-Resveratrol protects embry-
onic mesencephalic cells from tert-butyl hydroperoxide: electron paramagnetic
resonance spin trapping evidence for a radical scavenging mechanism. J Neuro-
chem 2000; 75:141–150.
134. Virgili M, Contestabile A. Partial neuroprotection of in vivo excitotoxic brain
damage by chronic administration of the red wine antioxidant agent, trans-resver-
atrol in rats. Neurosci Lett 2001; 281:123–126.
135. Inanami O, Watanabe Y, Syuto B, Nakano M, Tsuji M, Kuwabara M. Oral admin-
istration of (–)catechin protects against ischemia-reperfusion-induced neuronal
death in the gerbil. Free Radic Res 1998; 29:359–365.
136. Hara Y. Influence of tea catechins on digestive tract. J Cell Biochem 1997;
27(Suppl):52-58.
DK5836_C005.fm Page 165 Monday, May 22, 2006 1:14 PM
137. Levites Y, Weinreb O, Maor G, Youdim MBH, Mandel S. Green tea polyphenol
(–)-epigallocatechin-3-gallate prevents N-methyl-4-phenyl-1,2,3,6-tetrahydropy-
ridine-induced dopaminergic neurodegeneration. J Neurochem 2001;
78:1073–1082.
138. Levites Y, Youdim MBH, Maor G, Mandel S. Attenuation of 6-hydroxydopamine
(6-OHDA)-induced nuclear factor-kappaB (NF-κB) activation and cell death by
tea extracts in neuronal cultures. Biochem Pharmacol 2002; 63:21–29.
139. Schroeter H, Williams RJ, Matin R, Iversen L, Rice-Evans CA. Phenolic antiox-
idants attenuate neuronal cell death following uptake of oxidized low-density
lipoprotein. Free Radic Biol Med 2000; 29:1222–1233.
140. Bastianetto, S.; Ramassamy, C.; Dore, S.; Christen, Y.; Poirier, J.; Quirion, R. The
Ginkgo biloba extract (Egb 761) protects hippocampal neurons against cell death
induced by beta-amyloid. Eur J Neurosci 2000, 12:1882–1890.
141. Bastianetto S, Zheng W-H, Quirion R. The Ginkgo biloba extract (Egb 761)
protects and rescues hippocampal cells against nitric oxide-induced toxicity:
involvement of its flavonoid constituents and protein kinase C. J Neurochem 2000;
74:2268–2277.
142. Cott J. Natural product formulations in Europe for psychotropic indication. Psy-
chopharmacol Bull 1995; 31:745–751.
143. Le Bars P, Katz MM, Berman N, Itil TM, Freedman AM, Schatzberg AF. A
placebo-controlled, double blind, randomized trial of an extract of ginkgo biloba
for dementia. JAMA 1997; 278:1327–1332.
144. Kleijnen J, Knipschild P. Ginkgo biloba. Lancet 1992; 340:1136–1139.
145. Pappolla MA, Omar RA, Kim KS, Robakis NK. Immunohistochemical evidence
of antioxidative stress in Alzheimer’s disease. Am J Pathol 1992; 140:621–628.
146. Prasad KD, Cole WC, Hovland AR, Prasad KC, Nahreini P, Kumar B, Edward-
Prasad J, Andreatta CP. Multiple antioxidants in the prevention and treatment of
neurodegenerative disease: analysis of biologic rationale. Curr Opin Neurol 1999;
12:761–770.
147. Smith MA, Hirai K, Hsiao K, Pappolla MA, Harris PLR, Siedlak SL, Tabaton M,
Perry G. Amyloid-β deposition in Alzheimer’s transgenic mice is associated with
oxidative stress. J Neurochem 1998; 70:2212–2215.
148. Baeuerle PA, Baltimore D. NF-κB: ten years after. Cell 1996; 87: 13–20.
149. Baeuerle PA, Henkel T. Function and activation of NF-κB in the immune system.
Annu Rev Immunol 1994; 12:141–79.
150. Siebenlist U, Franzoso G, Brown K. Structure, regulation and function of NF-κB.
Annu Rev Cell Biol 1994; 10:405–455.
151. Kaltschmidt C, Kaltschmidt B, Baeuerle PA. Brain synapses contain inducible
forms of the transcription factor NF-κB. Mech Dev 1993; 43:135–147.
152. Kaltschmidt C, Kaltschmidt B, Baeuerle PA. Stimulation of ionotropic glutamate
receptors activates transcription farctor NF-κB in primary neurons. Proc Natl Acad
Sci USA 1995; 92:9618–9622.
153. Kaltschmidt C, Kaltschmidt B, Neumann H, Wekerle H, Baeuerle PA. Constitutive
NF-κB activity in neurons. Mol Cell Biol 1994; 14:3981–3992.
154. O’Neill LAJ, Kaltschmidt C. NF-κB: a crucial transcription factor for glial and
neuronal cell function. Trends Neurosci 1997; 20:252–258.
155. Bowie A, O’Neill LAJ. Oxidative stress and nuclear factor-κB: a reassessement
of the evidence in the light of recent discoveries. Biochem Pharmacol 2000,
59:13–23.
DK5836_C005.fm Page 166 Monday, May 22, 2006 1:14 PM
187. Glasgow JN, Wood T, Perez-Polo JR. Identification and characterization of nuclear
factor-κB binding sites in the murine bcl-x promoter. J Neurochem 2000;
75:1377–1389.
188. Zong WX, Edelstein LC, Chen C, Bash J, Gelinas C. The prosurvival Bcl-2
homolog Bfl-1/A is a direct transcriptional target of NF-κB that blocks TNFα-
induced apoptosis. Genes Dev 1999; 13:382–387.
189. Grilli M, Memo M. Possible role of NF-κB and p53 in the glutamate-induced
pro-apoptotic neuronal pathway. Cell Death Differ 1999; 6:22–27.
190. Grilli M, Pizzi M, Memo M, Spano P. Neuroprotection by aspirin and sodium
salicylate through blockade of NF-κB activation. Science 1996; 274:1383–1385.
191. Lipton SA. Janus faces of NF-κB: neurodestruction versus neuroprotection. Nat
Med 1997; 3:20–21.
192. Panet H, Barzilai A, Daily D, Melamed E, Offen D. Activation of nuclear tran-
scription factor kappa B (NF-κB) is essential for dopamine-induced apoptosis in
PC12 cells. J Neurochem 2001; 77:391–398.
193. Mayeux P. Epidemiology of neurodegeneration. Annu Rev Neurosci 2003;
26:81–104.
194. Helmer C, Joly P, Letenneur L, Commenges D. Mortality with dementia: results
from a French perspective. Am J Epidemiol 2001; 154: 642–648.
195. Wolfson C, Wolfson DB, Asgharian M, M’Lan CE, Ostbye T, Rockwood K, Hogan
DB; Clinical Progression of Dementia Study Group. A reevaluation of the duration
of survival after the onset of dementia. N Engl J Med 2001; 344:1111–1116.
196. Hofman A, Rocca WA, Brayne C, Breteler MMB, Clarke M, Cooper B, Copeland
JRM, Dartigues JF, Da Silva Droux A, Hagnell O, Heeren TJ, Engeland K, Jonker
C, Lindesay J, Lobo A, Mann AH, Molsa PK, Morgan K, O’Connor DW, Sulkava
R, Kay DWK, Amaducci L. The prevalence of dementia in Europe: a collaborative
study of 1980–1990 findings. Int J Epidemiol 1991; 20:736–748.
197. Rocca WA, Hofman A, Brayne C, Breteler M, Clarke M, Copeland JRM, Dartigues
JF, Engedal K, Hagnell O, Heeren TJ, Jonker C, Lindesay J, Lobo A, Mann AH,
Molsa PK, Morgan K, O’Connor DW, Da Silva Droux A, Sulkawa R, Kay DWK,
Amaducci L. Frequency and distribution of Alzheimer’s disease in Europe: a
collaborative study of 1980–1990 prevalence findings. Ann Neurol 1991;
30:381–390.
198. Behl C. Alzheimer’s disease and oxidative stress: implications for novel therapeu-
tic approaches. Prog Neurobiol 1999; 57:301–323.
199. Roses AD. A model for susceptibility polymorphism for complex diseases: apo-
lipoprotein E and Alzheimer’s disease. Neurogenetics 1997; 1:3–11.
200. Behl C. Vitamin E and other antioxidants in neuroprotection. Int J Vitam Nutr Res
1999; 69:213–219.
201. Behl C, Davis JB, Cole GM, Schubert D. Vitamin E protects nerve cells from
amyloid β protein toxicity. Biochem Biophys Res Commun 1992; 186:944–950.
202. Loo D, Copani A, Pike C, Whittemore E, Walencewicz A, Cotman CW. Apoptosis
is induced by β-amyloid in cultured central nervous system neurons. Proc Natl
Acad Sci USA 1993; 90:7951–7955.
203. Ekinci FJ, Linsley M-D, Shea TB. β-Amyloid-induced calcium influx induces
apoptosis by oxidative stress rather than τ phopshorilation. Brain Res Mol Brain
Res 2000; 76:389–395.
DK5836_C005.fm Page 169 Monday, May 22, 2006 1:14 PM
204. Guo Q, Christakos S, Robinson N, Mattson MP. Calbindin blocks the pro-apoptotic
actions of mutant presenilin-1: reduced oxidative stress and preserved mitochon-
drial function. Proc Natl Acad Sci USA 1998; 95:3227–3232.
205. Guo Q, Fu W, Holtsberg FW, Steiner SM, Mattson MP. Superoxide mediates the
cell-death-enhancing action of presenilin-1 mutations. J Neurosci Res 1999;
56:457–470.
206. Guo Q, Sebastian LS, Sopher BL, Miller MW, Ware CB, Martin GM, Mattson
MP. Increased vulnerability of hippocampal neurons from presenilin-1 mutant
knock-in mice to amyloid β-peptide toxicity: central roles of superoxide produc-
tion and caspase activation. J Neurochem 1999; 72:1019–1029.
207. Keller JN, Pang Z, Geddes JW, Begley JG, Germeyer A, Wang G, Mattson MP.
Impairment of glucose and glutamate transport and induction of mitochondrial
oxidative stress and dysfunction in synaptosomes by amyloid β-peptide: role of
the lipid peroxidation product 4-hydroxynonenal. J Neurochem 1997; 69:273–284.
208. Markesbery WR. Oxidative stress hypothesis in Alzheimer’s disease. Free Radic
Biol Med 1997; 23:134–147.
209. Pappolla MA, Chyan Y-J, Omar RA, Hsiao K, Perry G, Smith MA, Bozner P.
Evidence of oxidative stress and in vivo neurotoxicity of β-amyloid in a transgenic
mouse model of Alzheimer’s disease: a chronic oxidative paradigm for testing
antioxidant therapies in vivo. Am J Pathol 1998; 152:871–877.
210. Goodman Y, Mattson MP. Secreted forms of β-amyloid precursor protein protect
hippocampal neurons against amyloid β-peptide-induced oxidative injury. Exp
Neurol 1994; 128:1–12.
211. Harris ME, Hensley K, Butterfield A, Leedle R, Carney JM. Direct evidence of
oxidative injury produced by the Alzheimer’s β-amyloid peptide (1-40) in cultured
hippocampal neurons. Exp Neurol 1995; 131:193–202.
212. Lucca E, Angeretti N, Forloni G. Influence of cell culture conditions on the
protective effect of antioxidants against β-amyloid toxicity: studies with lazaroids.
Brain Res 1997; 764:293–298.
213. Hensley K, Carney JM, Mattson MP, Aksenova M, Harris M, Wu JF, Floyd RA,
Butterfield DA. A model for β-amyloid aggregation and neurotoxicity based on
free radical generation by the peptide: relevance to Alzheimer’s disese. Proc Natl
Acad Sci USA 1994; 91:3270–3274.
214. Behl C, Davis JB, Lesley R, Schubert D. Hydrogen peroxide mediates amyloid
β protein toxicity. Cell 1994; 77:817–822.
215. Mark RJ, Lovell MA, Markesbery WR, Uchida K, Mattson MP. A role of 4-
hydroxynonenal, an aldehydic product of lipid peroxidation, in disruption of ion
homeostasis and neuronal death induced by β-peptide. J Neurochem 1997;
255–264.
216. Lim GP, Chu T, Yang F, Beech W, Frautschy SA, Cole GM. The curry spice
curcumin reduces oxidative damage and amyloid pathology in an Alzheimer
transgenic mouse. J Neurosci 2001; 21:8370–8377.
217. Lockhart BP, Benicourt C, Junien JL, Privat A. Inhibitors of free radical formation
fail to attenuate direct beta-amyloid 25-35 peptide-mediated neurotoxicity in rat
hippocampal cultures. J Neurosci Res 1994; 4:494–505.
218. Aksenov MY, Aksenova MV, Butterfield DA, Geddes JW, Markesbery WR. Pro-
tein oxidation in the brain in Alzheimer’s disease. Neuroscience 2001;
103:273–283.
DK5836_C005.fm Page 170 Monday, May 22, 2006 1:14 PM
236. Lee J., Duan W, Mattson MP. Evidence that brain-derived neuritrophic factor is
required for basal neurogenesis and mediates, in part, the enhancement of neuro-
genesis by dietary restriction in the hippocampus of adult mice. J Neurochem
2002, 82:1367–1375.
237. Jellinger K. Pathology of Parkinson’s disease. In Calne DB, Ed. Handbook of
Experimental Pharmacology, vol 8. Berlin: Springer, 1989. p. 47–112.
238. Scherman D, Desnos C, Darchen F, Pollak P, Javoy-Agid F, Agid Y. Striatal
dopamine deficiency in Parkinson’s disease: role of aging. Ann Neurol 1989;
26:551–557.
239. Ebadi M, Srinivasan SK, Baxi MD. Oxidative stress and antioxidant therapy in
Parkinson’s disease. Progr Neurobiol 1996; 48:1–19.
240. Bower JH, Maragonore DM, McDonnell SK, Rocca WA. Incidence and distribu-
tion of Parkinsonism in Olmsted County, Minnesota, 1976-1990. Neurology 1999;
52:1214–1220.
241. Baldereschi M, Di Carlo A, Rocca WA, Vanni P, Maggi S, Perissinotto E, Grigo-
letto F, Amaducci L, Inzitari D. Parkinson’s disease and Parkinsonism in a longi-
tudinal study: two-fold higher incidence in men. ILSA Working Group. Italian
Longitudinal Study on Aging. Neurology 2000; 55:1358–1563.
242. de Rijk MC, Launer LJ, Berger K, Breteler MM, Dartigues JF, Baldereschi M,
Fratiglioni L, Lobo A, Martinez-Lage J, Trenkwalder C, Hofman A. Prevalence
of Parkinson’s disease in Europe: a collaborative study of population-based
cohorts. Neurologic Diseases in the Elderly Research Group. Neurology 2000; 54
(11 Suppl 5): S21–S23.
243. Elbaz A, Bower JH, Maraganore DM, McDonnell SK, Peterson BJ, Ahlskog JE,
Schaid DJ, Rocca WA. Risk tables for Parkinsonism and Parkinson’s disease. J
Clin Epidemiol 2002; 55:25–31.
244. Golbe L. The genetics of Parkinson’s disease: a reconsideration. Neurology 1990;
40:7–14.
245. Olanow CW, Tatton WG. Etiology and pathogenesis of Parkinson’s disease. Annu
Rev Neurosci 1999; 22:123–144.
246. Gasser T. Genetics of Parkinson’s disease. Ann Neurol 1998; 44:S53–S57.
247. Brooks DJ. The early diagnosis of Parkinson’s disease. Ann Neurol 1998;
44(Suppl), S19–S31.
248. Polymeropoulos MH, Lavedan C, Leroy E, Ide SE, Dehejia A, Dutra A, Pike B,
Root H, Rubenstein J, Boyer R, Stenroos ES, Chandrasekharappa S, Athanassia-
dou A, Papapetropoulos T, Johnson WG, Lazzarini AM, Duvoisin RC, Di Iorio
G, Golbe LI, Nussbaum RL. Mutation in the alpha-synuclein gene identified in
families with Parkinson’s disease. Science 1997; 276:2045–2007.
249. Teive HA, Raskin S, Iwamoto FM, Germiniani FM, Baran MH, Werneck LC,
Allan N, Quagliato E, Leroy E, Ide SE, Polymeropoulos MH. The G209A mutation
in the alpha-synuclein gene in Brazilian families with Parkinson’s disease. Arq
Neuropsiquiatr 2001; 59:722–724.
250. Kruger R, Kuhn W, Muller T, Woitalla D, Graeber M, Kosel S, Przuntek H, Epplen
JT, Schols L, Riess O. Ala30Pro mutation in the gene encoding alpha-synuclein
in Parkinson’s disease. Nat Genet 1998; 18:106–108.
251. Kitada T, Asakawa S, Hattori N, Matsumine H, Yamamura Y, Minoshima S,
Yokochi M, Mizuno Y, Shimizu N. Mutations in the parkin gene cause autosomal
recessive juvenile Parkinsonism. Nature 1998; 392:605–608.
DK5836_C005.fm Page 172 Monday, May 22, 2006 1:14 PM
252. Lim KL, Dawson VL, Dawson TM. The genetics of Parkinson’s disease. Curr
Neurol Neurosci Rep 2002; 2:439–446.
253. Langston JW Epidemiology versus genetics in Parkinson’s disease: progress in
resolving an age-old debate. Ann Neurol 1998; 44:(Suppl) S45–S52.
254. Tanner CM, Langston J.W. Do environmental toxins cause Parkinsons’s disease?
A critical review. Neurology 1990; 40:17–30.
255. Hornykiewicz O. Aging and neurotoxins as causative factors in idiopathic Parkin-
son’s disease — a critical analysis of the neurochemical evidence. Prog Neuro-
psychopharmacol Biol Psychiatry 1989; 13:319–328.
256. Langston JW, Ballard PA, Tetrud JW, Irwin I. Chronic Parkinsonism in humans
due to a product of meperidine analog synthesis. Science 1983; 219:979–980.
257. Nicklas WJ, Vyas I, Heikkila RE. Inhibition of NADH-linked oxidation in brain
mitochondria by 1-methyl-4-phenyl-pyridine, a metabolite of the neurotoxin 1-
methyl-4-phenyl-1,2,5,6-tetrahydropyridine. Life Sci 1985; 36:2503–2508.
258. Forno L. The Lewy body in Parkinson’s disease. Adv Neurol 1986; 45:35–43.
259. Hornykiewicz O. Neurochemical pathology and the etiology of Parkinson’s dis-
ease: basic facts and hypothetical possibilities. Mt Sinai J Med 1988; 55:11–20.
260. Jellinger K. An overview of morphological changes in Parkinson’s disease. Adv
Neurol 1986; 45:1–16.
261. Louis ED, Goldman JE, Powers JM, Fahn S. Parkinsonism features of eight
pathologically diagnosed cases of diffuse Lewy body disease. Mov Disord 1995;
10:188–194.
262. Jenner P, Dexter DT, Sian J, Schapira AHV, Marsden CD. Oxidative stress as a
cause of nigral cell death in Parkinson’s disease and incident Lewy body disease.
Ann Neurol 1992; 32:582–587.
263. Dexter DT, Carayon A, Javoy-Agid F, Agid Y, Wells SE, Jenner P, Marsden C.
Alterations in the levels of iron, ferritin and other trace metals in Parkinson’s
disease and other neurodegenerative diseases affecting the basal ganglia. Brain
1991; 114:1953–1975.
264. Tipton KF, Singer TP. Advances in our understanding of the mechanisms of the
neurotoxicity of MPTP and related compounds. J Neurochem 1993;
61:1191–1206.
265. Bindoff LA, Birch-Machin M, Cartlidge NEF, Parker WD Jr, Turnbull DM. Mito-
chondrial function in Parkinson’s disease. Lancet 1989; 2:49.
266. Hattori N, Tanaka M, Ozawa T, Mizuno Y. Immunohistochemical studies on
complexes I, II, III, and IV of mitochondria in Parkinson’s disease. Ann Neurol
1991; 30:563–571.
267. Parker WD Jr, Boyson SJ, Park JK. Abnormalities of the electron transport chain
in idiopathic Parkinson’s disease. Ann. Neurol 1989; 26:719–723.
268. Schapira AHV, Cooper JM, Dexter D, Clark JB, Jenner P, Marsden CD. Mito-
chondrial complex I deficiency in Parkinson’s disease. J Neurochem 1990;
54:823–827.
269. Krige D, Carroll MT, Cooper JM, Marsden CD, Schapira AHV. Platelet mito-
chondrial function in Parkinson’s disease. Ann Neurol 1992; 32:782–788.
270. DiMonte DA, Sandy MS, Jewell SA, Adornato B, Tanner CM, Langston JW.
Oxidative phosphorylation by intact muscle mitochondria in Parkinson’s disease.
Neurodegeneration 1993; 2:275–281.
DK5836_C005.fm Page 173 Monday, May 22, 2006 1:14 PM
271. Schapira AHV, Mann VM, Cooper JM, Dexter D, Daniel SE, Jenner P, Clark JB,
Marsden CD. Anatomic and disease specificity of NADH CoQ1 reductase (com-
plex I) deficiency in Parkinson’s disease. J Neurochem 1990; 55:2142–2145.
272. Betarbet R, Sherer TB, Mackenzie G, Garcia-Osuna M, Panov AV, Greenamyre
T. Chronic systemic pesticide exposure reproduces features of Parkinson’s disease.
Nat Neurosci 2000; 3:1301–1306.
273. Dexter DT, Wells FR, Lees AJ, Agid F, Agid P, Jenner P, Marsden CD. Increased
nigral iron content and alterations in other metal ions occurring in the brain in
Parkinson’s disease. J Neurochem 1989; 52:1830–1836.
274. Hirsch EC, Brandel J-P, Galle P, Javoy-Agid F, Agid Y. Iron and aluminum increase
in the substantia nigra of patients with Parkinson’s disease: an x-ray microanalysis.
J Neurochem 1991; 56:446–451.
275. Riederer P, Sofic E, Rausch W-D, Schmidt B, Reynolds GP, Jellinger K, Youdim
BH. Transition metals, ferritin, glutathione, and ascorbic acid in Parkinsonian
brains. J Neurochem 1989; 52:515–520.
276. Pearce RKB, Owen A, Daniel S, Jenner P, Marsden CD. Alterations in the distri-
bution of glutathione in the substantia nigra in Parkinson’s disease. J Neural
Transm 1997; 104:661–677.
277. Perry TL, Godin D, Hansen S. Parkinson’s disease: a disorder due to nigral
glutathione deficiency? Neurosci Lett 1982; 33:305–310.
278. Sian J, Dexter DT, Lees AJ, Daniel S, Agid Y, Javoy-Agid F, Jenner P, Marsden
CD. Alterations in glutathione levels in Parkinson’s disease and other neurode-
generative disorders affecting basal ganglia. Ann Neurol 1994; 36:348–355.
279. Sian J, Dexter DT, Lees AJ, Daniel S, Jenner P, Marsden CD. Glutathione-related
enzymes in brain in Parkinson’s disease. Ann Neurol 1994; 36:356–361.
280. Sofic E, Lange KW, Jellinger K, Riederer P. Reduced and oxidized glutathione in
the substantia nigra of patients with Parkinson’s disease. Neurosci Lett 1992;
142:128–130.
281. Dexter DT, Sian J, Rose S, Hindmarsch JG, Mann VM, Cooper JM, Wells FR,
Daniel SE, Lees AJ, Schapira AHV, Jenner P, Marsden CD. Indices of oxidative
stress and mitochondrial function in individuals with incidental Lewy body dis-
ease. Ann Neurol 1994; 35:38–44.
282. Dexter DT, Carter CJ, Wells FR, Javoy-Agid F, Agid Y, Lees A, Jenner P, Marsden
CD. Basal lipid peroxidation in substantia nigra is increased in Parkinson’s disease.
J Neurochem 1989; 52:381–389.
283. Dexter DT, Holley AE, Flitter WD, Slater TF, Wells FR, Daniel S, Lees AJ,
Marsden CD. Increased levels of lipid hydroperoxides in the Parkinsonian sub-
stantia nigra: an HPLC and ESR study. Mov Disord 1994; 9:92–97.
284. Sanchez-Ramos J, Overik E, Ames BN. A marker of oxyradical-mediated DNA
damage (8-hydroxy-2′deoxyguanosine) is increased in nigro-striatum of Parkin-
son’s disease brain. Neurodegeneration 1994; 3:197–204.
285. Yurek DM, Sladek JR. Dopamine cell replacement: Parkinson’s disease. Annu Rev
Neurosci 1990; 13:415–440.
286. Hagglund J, Aquilonius SM, Erkernas SM, Harving P, Lundquist H, Gullberg P,
Langstrom B. Dopamine receptor properties in Parkinson’s disease and Hunting-
ton’s chorea evaluated by positron emission tomography using C-N-methyl-spip-
erone. Acta Neurol Scand 1987; 75:87–94.
DK5836_C005.fm Page 174 Monday, May 22, 2006 1:14 PM
287. Rinne UK, Rinne JO, Laasko K, Laihinen A, Lonnberg P. Brain receptor changes
in Parkinson’s disease in relation to the disease process and treatment. J Neural
Transm 1983; 18:279–286.
288. Agid Y, Ruberg M, Javoy-Agid F, Hirsch E, Raisman-Vozari R, Vyas S, Faucheux
B, Michel P, Kastner A, Blanchard V, Damier P, Villares J, Zhang P. Are dopa-
minergic neurons selectively vulnerable to Parkinson’s disease? Adv Neurol 1993;
60:148–164.
289. Spina MB, Cohen G. Dopamine turnover and glutathione oxidation: implications
for Parkinson’s disease. Proc Natl Acad Sci USA 1989; 86:1398–1400.
290. Mena MA, Pardo B, Casarejos MJ, Fahn S, de Yebenes JG. Neurotoxicity of
levodopa on catecholamine-rich neurons. Mov Disord 1992; 7:23–31.
291. Spencer Smith T, Parker WD Jr, Bennett JP Jr. L-DOPA increases nigral production
of hydroxyl radicals in vivo: potential L-DOPA toxicity? NeuroReport 1994;
5:1006–1011.
292. Graham DG, Tiffany SM, Bell WR, Gutknecht WF. Autoxidation versus covalent
binding of quinones as the mechanism of toxicity of dopamine, 6-hydroxydopam-
ine, and related compounds toward C1300 neuroblastoma cells in vitro. Mol
Pharmacol 1978; 14:644–653.
293. Wick M, Byers L, Frey E. L-DOPA: selective toxicity for melanoma cells in vitro.
Science 1977; 197:468–469.
294. Hiramatsu M, Kohno M, Mori A, Shiraga H, Pfeiffer RF, Ebadi M. An ESR study
of 6-hydroxydopamine: generated hydroxy radicals and superoxide anions in
brain. Neurosciences 1994; 20:129–138.
295. Golbe LL, Langston JW, Shoulson I. Selegiline and Parkinson’s disease: protective
and symptomatic consideration. Drugs 1990; 39:646–651.
296. Heikkila RE, Manzino L, Duvoisin RC, Cabbat FS. Protection against the dopa-
minergic neurotoxicity of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine by
monoamine oxidase inhibitors. Nature 1984; 311:467–469.
297. Blum D, Torch S, Nisoou M-F, Verna J-M. 6-hydroxydopamine-induced nuclear
factor-kappaB activation in PC12 cells. Biochem Pharmacol 2001; 62:473–481.
298. Burke R.E, Kholodilov NG. Programmed cell death: does it play a role in Par-
kinson’s disease? Ann Neurol 1998; 44:S126–S133.
299. Tatton NA, Maclean-Fraser A, Tatton WG, Perl DP, Olanow CW. A fluorescent
double-labeling method to detect and confirm apoptotic nuclei in Parkinson’s
disease. Ann. Neurol 1998; 44(Suppl):S142–S148.
300. Marsden CD, Olanow CW. The causes of Parkinson’s disease are being unraveled
and rational neuroprotective therapy is close to reality. Ann Neurol 1998;
44(Suppl):S189–S196.
301. Fearnley JM, Lees AJ. Aging and Parkinson’s disease: substantia nigra regional
selectivity. Brain 1991; 114:2283–2301.
302. Parkinson Study Group. Mortality in DATATOP: a multicenter trial in early Par-
kinson’s disease. Ann Neurol 1998; 43:318–325.
303. Shoulson I. DATATOP: a decade of neuroprotective inquiry. Ann Neurol 1998;
44:S160–S166.
304. The Parkinson’s Study Group. Effects of tocopherol and deprenyl on the progres-
sion of disability in early Parkinson’s disease. N Engl J Med 1993; 328:176–183.
305. Wu R-M, Chiueh CC, Pert A, Murphy DL. Apparent antioxidant effect of l-
deprenyl on hydroxyl radical formation and nigral injury elicited by MPP+ in vivo.
Eur J Pharmacol 1993; 243:241–247.
DK5836_C005.fm Page 175 Monday, May 22, 2006 1:14 PM
306. Keller WC. Neuroprotection for Parkinson’s disease. Ann Neurol 1998;
44(Suppl):S155–S159.
307. Fahn S. An open trial of high-dosage antioxidants in early Parkinson’s disease.
Am J Clin Nutr 1991; 53:380S–382S.
308. Ogawa N, Tanaka K, Asanuma M, Kawai M, Masumizu T, Kohno M, Mori A.
Bromocriptine protects mice against 6-hydroxydopamine and scavenges hydroxyl
free radicals in vitro. Brain Res 1994; 657:207–213.
309. Olanow CW, Jenner P, Brooks D. Dopamine agonists and neuroprotection in
Parkinson’s disease. Ann Neurol 1998; 44(Suppl):S167–S174.
310. Schapira AHV. Neuroprotection and dopamine agonists. Neurology 2002;
58(Suppl 1):S9–S18.
311. Sawada H, Ibi M, Kihara T, Urushitani M, Akaike A, Kimura J, Shimohama S.
Dopamine D2-type agonism protect mesencephalic neurons from glutamate neu-
rotoxicity: mechanisms of neuroprotective treatment against oxidative stress. Ann
Neurol 1998; 44:110–119.
312. Rowland LT. Amyotrophic lateral sclerosis. Curr Opin Neurol 1994; 7:310–315.
313. Tandan R, Bradley WG. Amyotrophic lateral sclerosis: part 1. Clinical features,
pathology, and ethical issues in management. Ann Neurol 1985; 18:271–280.
314. Cudkowicz ME, McKenna-Yasek D, Sapp PE, Chin W, Geller B, Hayden DL,
Schoenfeld DA, Hosler BA, Horvitz HR, Brown RH. Epidemiology and mutations
in superoxide dismutase in amyotrophic lateral sclerosis. Ann Neurol 1997;
41:210–221.
315. Deng HX, Hentati A, Tainer JA, Iqbal Z, Cayabyab A, Hung W-Y, Getzoff ED,
Hu P, Herzfeldt B, Roos RP, Warner C, Deng G, Soriano E, Smyth C, Parge HE,
Ahmed A, Roses AD, Hallewell RA, Pericak-Vance M, Siddique T. Amyotrophic
lateral sclerosis and structural defects in Cu,Zn superoxide dismutase. Science
1993; 261:1047–1051.
316. Siddique T, Nijhawan D, Hentati A. Molecular genetic basis of familial ALS.
Neurology 1996; 47(Suppl):27–35.
317. Dal Canto M, Gurney ME. Neuropathological changes in two lines of mice
carrying a transgene for mutant human Cu,Zn SOD, and in mice overexpressing
wild type human SOD: a model of familial amyotrophic lateral sclerosis. Brain
Res 1995; 676:25–40.
318. Ghadge GD, Lee JP, Bindokas VP, Jordan J, Ma L, Miller RJ, Roos RP. Mutant
superoxide dismutase-1-linked familial amyotrophic lateral sclerosis: molecular
mechanisms of neuronal death and protection. J Neurosci 1997; 17:8756–8766.
319. Gurney ME, Pu H, Chiu AY, Dal Canto MC, Polchow CY, Alexander DD, Caliendo
J, Hentati A, Kwon YW, Deng H-X, Chen W, Zhai P, Sufit RL, Siddique T. Motor
neuron degeneration in mice that express a human Cu, Zn superoxide dismutase
mutation. Science 1994; 264:1772–1775.
320. Wong PC, Pardo CA, Borchelt DR, Lee MK, Copeland NG, Jenkins NA, Sisodia
SS, Cleveland DW, Price DL. An adverse property of familial ALS-linked SOD-
1 mutations causes motor neuron disease characterized by vacuolar degeneration
of mitochondria. Neuron 1995; 14:1105–1116.
321. Bowling AC, Schulz JB, Brown RH Jr, Beal MF. Superoxide dismutase activity,
oxidative damage, and mitochondrial energy metabolism in familial and sporadic
amyotrophic lateral sclerosis. J Neurochem 1993; 61:2322–2325.
DK5836_C005.fm Page 176 Monday, May 22, 2006 1:14 PM
322. Rabizadeh S, Gralla EB, Borchelt DR, Gwinn R, Valentine JS, Sisodia S, Wong
P, Lee M, Hahn H, Bredesen DE. Mutations associated with amyotrophic lateral
sclerosis convert superoxide dismutase from an antiapoptotic gene to a proapop-
totic gene: studies in yeast and neural cells. Proc Natl Acad Sci USA 1995;
92:3024–3028.
323. Lee MH, Hyum D-H, Halliwell B, Jenner P. Effect of overexpression of wild-type
mutant Cu/Zn-superoxide dismutases on oxidative stress and cell death induced
by hydrogen peroxide, 4-hydroxynonenal or serum deprivation: potentiation of
injury by ALS-related mutant superoxide dismutases and prevention by Bcl-2. J
Neurochem 2001; 78: 209–220.
324. Przedborski S, Jackson-Lewis V, Kostic V, Carlson E, Epstein CJ, Cadet JL.
Superoxide dismutase, catalase, and glutathione peroxidase activities in cop-
per/zinc-superoxide dismutase transgenic mice. J Neurochem 1992;
58:1760–1767.
325. Borchelt D.R, Lee MK, Slunt HS, Guarnieri M, Xu Z-S, Wong PC, Brown RH,
Price DL, Sisodia SS, Cleveland DW. Superoxide dismutase 1 with mutations
linked to familial amyotrophic lateral sclerosis possess significant activity. Proc
Natl Acad Sci USA 1994; 91:8292–8296.
326. McCord JM. Mutant mice, CuZn superoxide dismutase, and motor neuron degen-
eration. Science 1995; 266:1586–1587.
327. Wiedau-Pazos M, Goto JJ, Rabizadeh S, Gralla EB, Roe JA, Lee MK, Valentine
JS, Bredesen DE. Altered reactivity of superoxide dismutase in familial amyo-
trophic lateral sclerosis. Science 1996; 271:515–517.
328. Bogdanov M, Ramos LE, Xu Z, Beal FM. Elevated “hydroxyl radical” generation
in vivo in an animal model of amyotrophic lateral sclerosis. J Neurochem 1998;
71:1321–24.
329. Hall ED, Andrus PK, Oostveen JA, Fleck TJ, Gurney ME. Relationship of oxygen
radical-induced lipid peroxidative damage to disease onset and progression in a
transgenic model of familial ALS. J Neurosci Res 1998; 53:66–77.
330. Liu D, Wen J, Liu J, Li L. The roles of free radicals in amyotrophic lateral sclerosis:
reactive oxygen species and elevated oxidation of protein, DNA, and membrane
phospholipids. FASEB J 1999; 13:2318–2328.
331. Bogdanov M, Brown RH, Matson W, Smart R, Hayden D, O’Donnell H, Beal
MF, Cudkowicz M. Increased oxidative damage to DNA in ALS patients. Free
Radic Biol Med 2000; 29:652–658.
332. Crow JP, Sampson JB, Zhuang Y, Thompson JA, Beckman JS. Decreased zinc
affinity of amyotrophic superoxide dismutase mutants leads to enhanced catalysis
of tyrosine nitration by peroxynitrite. J Neurochem 1997; 69:1936–1944.
333. Pedersen WA, Fu W, Keller JN, Markesbery WR, Appel S, Smith RG, Kasarkis
E, Mattson MP. Protein modification by the lipid peroxidation product 4-hydrox-
ynonenal in the spinal cords of amyotrophic lateral sclerosis patients. Ann Neurol
1998; 44:819–824.
334. Aguirre T, Van Den Bosch L, Goetshalckx K, Tilkin P, Mathijs G, Cassiman JJ,
Robberecht W. Increased sensitivity of fibroblasts from amyotrophic lateral scle-
rosis patients to oxidative stress. Ann. Neurol. 1998; 43:452–457.
335. Hudson AJ. Amyotrophic lateral sclerosis and its association with dementia, Par-
kinsonism and other neurological disorders: a review. Brain 1981; 104:217–247.
DK5836_C005.fm Page 177 Monday, May 22, 2006 1:14 PM
336. Kostic V, Gurney ME, Deng H-X, Siddique T, Epstein CJ, Przedborski S. Midbrain
dopaminergic neuronal degeneration in a transgenic mouse model of familial
amyotrophic lateral sclerosis. Ann Neurol 1997; 41:497–504.
337. Poduslo JF, Whelan SL, Curran GL, Wengenack TM. Therapeutic benefit of
polyamine-modified catalase as a scavenger of hydrogen peroxide and nitric oxide
in familial amyotrophic lateral sclerosis transgenics. Ann Neurol 2000;
48:943–947.
338. Reinholz MM, Merkle CM, Poduslo JF. Therapeutic benefits of putrescine-mod-
ified catalase in a transgenic mouse model of familial amyotrophic lateral sclerosis.
Exp Neurol 1999; 159:204–216.
339. Andreassen OA, Dedeoglu A, Friedlich A, Ferrante KL, Hughes D, Szabo C, Beal
MF. Effects of an inhibitor of poly(ADP-ribose) polymerase, desmethylselegiline,
trientine, and lipoic acid in transgenic ALS mice. Exp Neurol 2001; 168:419–424.
340. Andreassen OA, Dedeoglu A, Klivenyi P, Beal MF, Bush AI. N-acetyl-L-cysteine
improves survival and preserves motor performance in an animal model of familial
amyotrophic lateral sclerosis. Neuroreport 2000; 11:2491–243.
341. Dugan LL, Lovett EG, Quick KL, Lotharius J, Lin TT, O’Malley KL. Fullerene-
based antioxidants and neurodegenerative disorders. Parkinsonism Relat Disord
2001; 7:243–246.
342. Gurney ME, Cutting FB, Zhai P, Doble A, Taylor C, Andrus PK, Hall ED. Benefit
of vitamin E, riluzole, and gabapentin in a transgenic model of familial amyo-
trophic lateral sclerosis. Ann Neurol 1996; 39:147–157.
343. Bensimon G, Lacomblez L, Meininger V and the ALS/riluzole study group. A
controlled trial of riluzole in amyotrophic lateral sclerosis. N Engl J Med 1994;
330:585-591.
344. Koh J-Y, Kim D-K, Hwang JY, Kim YI, Seo JH. Antioxidative and proapoptotic
effects of riluzole on cultured cortical neurons. J Neurochem 1999; 72:716–723.
345. Desnuelle C, Dib M, Garrel C, Favier A. A double-blind, placebo-controlled
randomized clinical trial of alpha-tocopherol (vitamin E) in the treatment of
amyotrophic lateral sclerosis. ALS riluzole-tocopherol Study Group. Amyotroph
Lateral Scler Other Motor Neuron Disord. 2001; 2:9–18.
346. Lange DJ, Murphy PL, Diamond B, Appel V, Lai EC, Younger DS, Appel SH.
Selegiline is ineffective in a collaborative double-blind, placebo-controlled trial
for treatment of amyotrophic lateral sclerosis. Arch Neurol 1998; 55:93–96.
347. Vyth A, Timmer JG, Bossuyt PM, Louwerse ES, de Jong JM. Survival in patients
with amyotrophic lateral sclerosis, treated with an array of antioxidants. J Neurol
Sci 1996; 139 Suppl:99–103.
348. Li M, Ona VO, Guégan C, Chen M, Jackson-Lewis V, Andrews LJ, Olszewski
AJ, Stieg PE, Lee J-P, Przedborski S, Friedlander RM. Functional role of caspase-
1 and caspase-3 in an ALS transgenic mouse model. Science 2000; 288:335–339.
349. Esposito E, Rossi C, Amodio R, Di Castelnuovo A, Bendotti C, Rotondo T, Algeri
S, Rotilio D. Lyophilized red wine administration prolongs survival in an animal
model of amyotrophic lateral sclerosis. Ann Neurol 2000; 48:686–687.
350. Sano M. Do dietary antioxidants prevent Alzheimer’s disease? Lancet Neurol
2002; 1:342.
351. Lemon JA, Boreham DR, Rollo CD. A dietary supplement abolishes age-related
cognitive decline in transgenic mice expressing elevated free radical processes.
Exp Biol Med 2003; 228:800–810.
DK5836_C005.fm Page 178 Monday, May 22, 2006 1:14 PM
CONTENTS
6.1 Introduction..............................................................................................179
6.2 Unipolar Depression................................................................................180
6.2.1 Amino Acids ...............................................................................182
6.2.2 Folate, Vitamin B12, and Homocysteine......................................186
6.2.3 S-adenosylmethionine..................................................................190
6.2.4 Zinc ..............................................................................................192
6.2.5 St. John’s Wort (Hypericum perforatum)....................................193
6.2.6 Ginkgo Biloba and Panax Ginseng.............................................194
6.2.7 Vitamin B6 ...................................................................................195
6.2.8 Tyramine ......................................................................................196
6.2.9 Omega-3 Fatty Acids...................................................................197
6.3 Conclusion ...............................................................................................198
References .........................................................................................................203
6.1 INTRODUCTION
Approximately 7% of the North American population is currently affected by
mood disorders.1 The bouts of depression and mania that characterize these
conditions cause a great deal of morbidity and increase the risk of mortality,
ranking these disorders among the top 10 causes of disability.1 Therefore, effective
treatment options are of the utmost importance to increase quality of life for these
individuals, as well as to increase the labor capacity of the work force. Originally
the domain of the psychiatrist, the pharmacotherapy of these disorders is now
often in the hands of the general physician. This requires considerable knowledge
179
DK5836_C006.fm Page 180 Monday, May 22, 2006 1:51 PM
associated with these pharmaceuticals, they are often a last line of defense used
in treatment-resistant depression. The current MAOIs in use are phenelzine and
tranylcypromine.2
Also brought onto the market in the 1950s were the tricyclic antidepressants,
which are a major class of drugs used to treat depression. This class of drugs
works by nonselectively blocking the reuptake of monoamines. Reuptake is a
mechanism that prevents over-stimulation of the postsynaptic neuron by bringing
neurotransmitters back into the presynaptic neuron. By nonselectively blocking
monoamine reuptake, the concentrations of serotonin, norepinephrine, epineph-
rine, and dopamine are all increased in the synaptic cleft. Consequently, a number
of different monoamine receptors are activated both in the brain and many other
tissues where receptors are expressed. This lack of selectivity explains why the
tricyclic antidepressants are associated with a plethora of side effects: drowsiness,
dry mouth, urinary retention, constipation, blurred vision, low blood pressure,
weight gain, and cardiac effects.2 The current tricyclic antidepressants in use
include amitriptyline, clomipramine, desipramine, nortriptyline, and imipramine.2
A major development in unipolar depression pharmacotherapy was the intro-
duction of selective serotonin reuptake inhibitors (SSRIs) in the 1980s. This class
of drugs also works by inhibiting the reuptake of neurotransmitters; however, it
differs from tricyclic antidepressants in that it is selective only for serotonin. By
binding only to the serotonin reuptake transporter protein, there is an increase in
serotonin in the synaptic cleft, but the other monoamine concentrations remain
unchanged. This property of the SSRIs results in fewer side effects than the
tricyclic antidepressants, although several are still observed, including nausea,
nervousness, insomnia, sexual dysfunction, and headache.2 Given the increased
selectivity and decreased severity of side effects, SSRIs (fluoxetine, sertraline,
paroxetine, citalopram, and escitalopram) are usually the first-line therapy in
unipolar depression.2
Similar to the SSRIs, another class of drugs named the serotonin and nore-
pinephrine reuptake inhibitors (SNRIs), which block the reuptake of both sero-
tonin and norepinephrine, were developed for the treatment of unipolar depres-
sion. The SNRIs still have fewer side effects than tricyclic antidepressants and
are more efficacious in some individuals than the SSRIs. However, there is some
risk of hypertension as well as other side effects with the SNRIs (venlafaxine
and duloxetine), which requires the doctor’s consideration when prescribing.2
Finally, the last class of drugs used for the treatment of unipolar depression
is the atypical antidepressants. They fall outside the four aforementioned catego-
ries due to differences in both structure and function. Some operate by weakly
inhibiting reuptake of specific neurotransmitters, some by inhibiting neurotrans-
mitter receptors directly, whereas the mechanisms of others still remain to be
clarified. These mechanisms will be elaborated upon as needed in the following
sections. In line with the other classes of drugs, the atypical antidepressants,
mirtazapine, bupropion and trazodone, are also associated with a range of side
effects.2
DK5836_C006.fm Page 182 Monday, May 22, 2006 1:51 PM
There are a few problems with the monoamine theory of unipolar depression.
First of all, it cannot account for the fact that antidepressant drugs produce their
biochemical effects within minutes or hours, yet the onset of therapeutic benefit
usually takes weeks. Furthermore, the severity of the depressive state and the
amount of monoamine depletion do not necessarily correlate.4 Our knowledge of
intracellular signaling has drastically improved over the past decade, and accu-
mulating evidence seems to suggest that protein phosphorylation is part of the
long-term mechanism of antidepressants.4 Evidence has shown that various anti-
depressants can alter both the activity and translocation of second-messenger
regulated protein kinases; in particular, protein kinase C (PKC), cyclic AMP-
dependent protein kinase (PKA), and Ca2+/calmodulin-dependent protein kinase
II (CaMKII).4 Increasing activation of protein kinases allows for increased phos-
phorylation of certain subcellular components (of particular interest are micro-
tubule-associated protein 2 [MAP2] and synaptotagmin) that may consequently
affect cytoskeleton remodeling, a process involved in neurotransmitter release.4
This new theory of the mechanistic basis of antidepressant action continues to
develop at a rapid pace and shows great promise at aiding in the development of
more effective antidepressants as well as furthering our understanding of the
etiology of the disease.
Another theory that accounts for the temporal discrepancy between the bio-
chemical and clinical effects of antidepressants is the idea that these compounds
decrease N-methyl-D-aspartate (NMDA) receptor function. This is supported by
the observation that chronic (14 day) administration, but not acute (1 day), of 17
different antidepressants produces adaptive changes in the binding of radioligand
to NMDA receptors in a mouse model.5 This is an interesting finding as it
corresponds quite well with the time course required for the clinical benefit of
antidepressants.
It is in the context of these mechanisms that the interaction of nutritional and
dietary components with unipolar depression drugs will be examined.
There have been many studies conducted to determine if baseline amino acid
status is correlated to pharmacotherapy success. In these studies, it is common
to use a ratio of tryptophan over the sum of all other large, neutral amino acids
(Trp/LNAA). The sum of plasma LNAA is typically obtained by adding valine,
isoleucine, leucine, tyrosine, and phenylalanine together. This Trp/LNAA ratio
is used because tryptophan competes with the other LNAA to get across the
blood–brain barrier (BBB). Hence, an excess of other LNAAs will hinder tryp-
tophan’s uptake into the brain, causing a subsequent decrease in serotonin syn-
thesis. A similar ratio is used for tyrosine (Tyr/LNAA) when the study is con-
cerned with noradrenergic neurotransmission, except that tryptophan replaces
tyrosine in the denominator.
The baseline Trp/LNAA ratio has been shown to be inversely proportional
to improvement on the tricyclic antidepressants, amitriptyline and clomipramine,
with a Trp/LNAA ratio below the group mean predicting a better treatment
response.7 In addition, the same study also showed that the plasma concentration
of tryptophan was inversely proportional to the clinical response to the SSRIs,
citalopram, and paroxetine.7 Another study using a mixture of SSRIs (fluvoxam-
ine, fluoxetine, and citalopram), along with SNRIs (amitriptyline and clomi-
pramine), and a tricyclic antidepressants (TCA) (nortriptyline), showed that the
Trp/LNAA ratio during the first week of therapy could be used to predict clinical
efficacy of the group as a whole.8 However, no such relationship was observed
when examining treatment of major depression with the MAOI, moclobemide.9
The Tyr/LNAA has also shown some utility in predicting the response to treatment
with the TCA, nortriptyline, with the baseline ratio similarly being inversely
correlated to subsequent clinical improvement on the medication.10 Unfortunately,
not all results are in agreement with the above trends. In one of the larger studies
conducted in this area (n = 147), baseline Trp/LNAA and Tyr/LNAA ratios were
not correlated with the 6-month treatment outcome of depressed subjects placed
on fluoxetine or nortriptyline.11 It was initially hoped that a clear pattern of plasma
amino acid status could suggest the best choice for pharmacotherapy. However,
the mixed nature of the results has made this exceedingly difficult.
It has been suggested that the ability of the 5-HT postsynaptic receptors to
adapt to serotonin availability is more important than the availability of precursor
amino acids.11 This could also mean that a reduction in receptor plasticity is a
marker of treatment resistance in depression.12 If this were the case, then the
baseline amino acid profile could be a more meaningful predictor of treatment
response in those individuals who have a greater ability to regulate their 5-HT
receptors. On the other hand, those individuals who lack such plasticity would
likely be those that do not respond well to increasing monoamine precursors via
diet, or by increasing monoamines directly using antidepressants.
An interesting observation is that subjects given the TCA antidepressant,
clomipramine, for 6 months had their plasma tryptophan concentration reduced
to 28% of its initial value over the treatment period, but this value was still only
68% of the initial value 3 months after successful pharmacotherapy was com-
pleted.13 This was interpreted as a rebound phenomenon due to the fact that
DK5836_C006.fm Page 184 Monday, May 22, 2006 1:51 PM
on monoamine precursors because serotonin itself lacks the ability to cross the
blood–brain barrier, hence making supplementation ineffective.
One study has shown that co-administration of tryptophan with the SSRI,
fluoxetine, caused a significant decrease in clinical symptoms of unipolar depres-
sion in the first week of pharmacotherapy, but at no later points in time.17 This
is an important observation considering that side effects of antidepressants begin
immediately with clinical response beginning after approximately 2 weeks, which
makes the initial phase of treatment notoriously difficult.17 It should be noted that
the over-the-counter sale of L-tryptophan was banned in the U. S. by the U.S.
Food and Drug Administration (USFDA) in 1990 as a result of a number of deaths
related to a contaminated batch.6 This batch caused at least 38 deaths due to
eosinophilia myalgia syndrome.6 However, α-lactalbumin supplementation is still
available over-the-counter and may be a worthwhile alternative.
There is much interest in using the -lactalbumin protein to increase seroton-
ergic activity in the brain. One study showed that a 2-day experimental diet
supplemented with α-lactalbumin could decrease depressive feelings under stress
in subjects that were stress-vulnerable.18 This is likely due to the fact that, when
combined with a regular diet, α-lactalbumin has the capacity to increase the
Trp/LNAA ratio.19 It still remains to be determined if α-lactalbumin co-admin-
istered with antidepressants has a beneficial effect, but there is some evidence
for the utility of 5-HTP in this regard.
5-HTP may be more efficacious than L-tryptophan, or α-lactalbumin, as it
bypasses the tryptophan hydroxylase enzyme in serotonin synthesis, which is the
rate-limiting step, and, in addition, it cannot be shunted into niacin or protein
production.20 A review of the literature suggesting that 5-HTP is effective in
treating unipolar depression showed that only a few studies were of sufficient
quality to show “statistical superiority to placebo” and they were mostly aug-
mentation studies.6 In light of this observation, it is clear that more studies looking
at the augmentation of classical antidepressant pharmacotherapy with 5-HTP are
warranted. It has been suggested that the advent of the SSRIs in the 1980s caused
a loss of interest in 5-HTP as a treatment, but in light of the augmentation studies
perhaps it should be reconsidered as a treatment option.6
Both clinicians, as well as patients, need to be aware of the remote possibility
of serotonin syndrome in regards to co-administration of more than one com-
pound. The most commonly reported substances involved in causing this syn-
drome are the MAOIs in combination with L-tryptophan or fluoxetine, but there
has been a reported case of an over-the-counter cough medicine causing serotonin
syndrome in a man with vascular disease.21 However, there have been no reports
of serotonin syndrome with the use of 5-HTP as a monotherapy, or in augmen-
tation studies with antidepressants.6 Despite these promising results, patients
should probably only use 5-HTP under the supervision of their clinician.
Ultimately, the efficacy of L-tryptophan, 5-HTP, and α-lactalbumin to aug-
ment traditional pharmacotherapy in unipolar depression likely depends on a
number of factors. First, the type of drug being used may determine whether or
not modulating monoaminergic precursors has any beneficial effect. Second,
DK5836_C006.fm Page 186 Monday, May 22, 2006 1:51 PM
individual variation, such as 5-HT receptor plasticity and the relative abundance
of precursor amino acids already available, could also determine whether more
monoamine precursor would be of any clinical benefit. Furthermore, the degree
of inhibition of the liver enzyme tryptophan pyrrolase may very well determine
how much tryptophan is being catabolized and, hence, how much is needed in
the diet. Finally, the involvement of serotonergic deficits, noradrenergic deficits,
or both in unipolar depression etiology could ultimately determine the utility of
the amino acid precursors to monoamines. As our understanding of the biological
and molecular etiology of depression becomes clearer, it is likely that examination
of the above factors in individual patients could suggest which pharmacotherapy
may be the most effective, and whether augmentation with monoamine precursors
would have any therapeutic benefit.
relationship between low baseline folate status and further treatment resistance.28
However, elevated homocysteine levels and low vitamin B12 levels were not
associated.28 Furthermore, this group also showed that low baseline serum folate
levels, but not elevated homocysteine or low vitamin B12 levels, were associated
with increased risk of relapse during the continuation phase of fluoxetine phar-
macotherapy.29 This study examined 71 patients, who had previously remitted
from unipolar depression for at least 3 weeks while undergoing fluoxetine treat-
ment and were followed for a subsequent 28 weeks to be monitored for depressive
relapse.29 These studies suggest that low folate status might have the ability to
predict treatment resistance as well as possibility of relapse, in addition to its
possible ability to predict treatment outcome, as noted above. If this is substan-
tiated by further research, folate status could prove to be a valuable clinical
indicator.
There are a few points of detail that should be considered while contemplating
the above relationships. The first of which is the possibility that macrocytic
anemia could be a predictor of poor treatment response in unipolar depression,
as it is often caused by either a folate or vitamin B12 deficiency. However, one
study found that neither macrocytosis nor anemia could predict antidepressant
refractoriness, in addition to not being able to predict low serum folate or vitamin
B12.30 Based on these results, it does not appear that macrocytosis or anemia is
useful in predicting treatment response. Another important finding is that the
relationship between low folate levels and an increased incidence of unipolar
depression may not exist in all populations. For instance, it was shown that 117
newly admitted Chinese inpatients with unipolar depression had normal levels of
folate, and that folate levels could not predict outcome on standard assessments
of depression.31 However, both the subjects and controls were found to have a
high intake of green vegetables.31 Therefore, the utility of serum folate levels to
predict incidence of depression might be limited in populations with a high intake
of green vegetables. Whether or not this relationship extends the likelihood of a
favorable response to pharmacotherapy remains to be determined.
A few studies have been undertaken to investigate the use of supplemental
folate in augmenting the traditional pharmacotherapy of unipolar depression. It
was shown that the addition of 15 mg daily folate to the standard treatments of
123 subjects, having either major depression or schizophrenia, with borderline
or deficient folate status, resulted in significant clinical and social improvements
in both groups over the course of a 6-month period.32 Later, a study was conducted
to see if subjects with unipolar depression, randomly assigned to treatment with
20 mg of fluoxetine plus 500 µg of folic acid or a similar looking placebo,
responded differently.33 Interestingly, only the female subjects receiving folate
showed a significantly greater improvement compared to placebo, as well as a
significant decrease in plasma homocysteine levels.33 It was suggested that the
dose of folic acid needed to enhance pharmacotherapy in males could be higher
than what was given in the study and should be sufficient to decrease plasma
levels of homocysteine.33 It is clear that more research needs be done in order to
determine the optimum doses for different subjects.
DK5836_C006.fm Page 189 Monday, May 22, 2006 1:51 PM
6.2.3 S-ADENOSYLMETHIONINE
The use of S-adenosylmethionine (SAM) in the treatment of unipolar depression
makes perfect sense considering the previous section discussing one-carbon metab-
olism and its possible relationship to this disease. It functions as a cofactor in the
rate-limiting steps of the tryptophan hydroxylase and tyrosine hydroxylase reactions,
which are responsible for the production of serotonin and dopamine/norepinephrine,
respectively.36 Theoretically, by providing exogenous SAM, one should be able to
improve the capacity for methylation reactions that are necessary for neurotransmitter
synthesis, along with a myriad other reactions. However, the function of SAM might
extend beyond its role in methylation reactions.
As a monotherapy, SAM has shown some clinical utility. One open, multicenter
study showed that in 145 patients with unipolar depression, the parenteral adminis-
tration of 400 mg/day SAM for 15 days caused scores of depression to decrease
after both 7 days and 15 days.37 This is an important finding as most traditional
antidepressants have a 2-week lag period, during which there is no clinical benefit.
This rapid onset of action of SAM treatment is a common finding, with improvement
seen anywhere from 2 days to 2 weeks after treatment begins.36 A number of studies
have compared the efficacy of SAM monotherapy to traditional antidepressants. One
small study comparing intravenous administration of SAM at 400 mg/day with oral
administration of the tricyclic antidepressant, imipramine, found that SAM produced
superior results at the end of the first week and, by the end of the study, 66% of the
patients in the SAM group and only 22% of the patients in the imipramine group
had a clinically significant improvement.38 Later, two multicenter studies, each with
approximately 300 subjects with unipolar depression, were conducted simulta-
neously to compare 1600 mg/day SAM given orally and 400 mg/day given intra-
muscularly, both to 150 mg imipramine/day given orally.39 The clinical responses
for the SAM and imipramine arms did not differ, but significantly fewer side effects
were observed in subjects treated with SAM.39 Overall, in six of the eight studies
that have compared SAM with tricyclic antidepressants, SAM was of equivalent
efficacy, and in one study it was superior to imipramine.36 These are promising results
for SAM as a monotherapy, considering that it seems to be well tolerated with few
side effects and has an onset of action that is faster than traditional antidepressants.
In addition to these clinical trials, electroencephalogram/event-related potential map-
ping identified SAM as an antidepressant when given intravenously at 800 mg/day.40
Unfortunately, there are far fewer studies that have looked at augmenting
traditional pharmacotherapy of unipolar depression with SAM. It has been shown
DK5836_C006.fm Page 191 Monday, May 22, 2006 1:51 PM
6.2.4 ZINC
Some studies have shown that plasma zinc levels in depressed subjects are
significantly lower than levels in healthy controls,45 yet this relationship was not
statistically significant in other studies.46 However, regardless of baseline zinc
levels, both these studies showed that plasma zinc levels significantly increased
after successful treatment with antidepressants.45,46 Furthermore, in a rat model,
treatment with citalopram and imipramine as well as electroconvulsive shock
therapy, all elevated hippocampal zinc concentrations.47 These studies suggest
that zinc may have a role in the pharmacotherapy of unipolar depression.
A number of studies have investigated the use of zinc supplementation to
augment traditional antidepressant efficacy. One study showed that zinc (ZnSO4)
at a dose of 30 mg/kg and/or imipramine at 30 mg/kg reduced the immobility
time in the forced swim test, also known as Porsolt’s test, in both mice and rats.48
Furthermore, a combination of zinc and imipramine at ineffective doses (1 and
5 mg/kg, respectively) was clinically effective in rats.48 Research has suggested
that the state of immobility in the forced swim test reflects lowered mood in rats
and is improved with antidepressant treatment as well as electroconvulsive shock
therapy.49 Therefore, the aforementioned study may suggest that augmentation of
traditional pharmacotherapy with zinc may have the capacity to improve treatment
of unipolar depression. This notion was further supported by the finding that low,
otherwise ineffective doses of imipramine and citalopram, administered together
with low, ineffective doses of zinc were effective in decreasing immobility times
in mice subjected to the forced swim test.50 In addition to these findings, it has
been shown that the use of zinc hydroaspartate alone has antidepressant properties
in both the forced swim test as well as the olfactory bulbectomy animal models
of depression.51 Furthermore, there was a concomitant rise in serum levels of zinc
in association with the antidepressant-like effects, and no tolerance was developed
following chronic treatment.51 These results all support a role for zinc status in
the treatment of unipolar depression.
In addition to the research conducted in animal models of depression, there
is some preliminary evidence showing zinc augmentation of traditional pharma-
cotherapy in humans. One small (n = 14), placebo-controlled, double-blind study
showed that zinc supplementation was able to augment the treatment of unipolar
depression with tricyclic antidepressants as well as SSRIs.52 Subjects in the zinc
supplementation group showed a significant decrease in both Hamilton Depres-
sion Rating Scale and Beck Depression Inventory measures of clinical symptoms
after 6 and 12 weeks of supplementation, as compared with placebo supplemen-
tation.52 These preliminary findings are encouraging and emphasize the need for
additional clinical intervention trials of zinc and conventional antidepressants.
The mechanistic basis of zinc’s action in the treatment of unipolar depression
is an area of great interest. One line of evidence suggests that the chronic
administration of antidepressants ultimately results in a region-specific dampen-
ing of N-methyl-D-aspartate (NMDA) receptor function, and conversely, the
administration of compounds that reduce transmission at NMDA receptors act as
DK5836_C006.fm Page 193 Monday, May 22, 2006 1:51 PM
antidepressants.53 In accordance with this theory, it has been shown that zinc has
the capacity to modulate binding of NMDA receptor agonists and antagonists to
NMDA receptors in the forced swim test, resulting in antidepressant-like effects.54
Furthermore, it has been shown that there is a significant decrease in the potency
of zinc to inhibit binding at NMDA receptors in the hippocampal tissue of suicide
victims; a group with a high likelihood of suffering from depression.55 This
interaction of zinc with NMDA receptors requires further research, as animal
models of depression are somewhat controversial, and not all suicide subjects
had a diagnosis of depression.55
It has been suggested that it is possible that zinc may reduce NMDA receptor
function via single or multiple mechanisms, but experiments to reveal the exact
nature of zinc’s influence on the NMDA receptor are currently being conducted.56
The observed actions of zinc include a direct antagonism of the NMDA receptor,
the ability to act on the AMPA receptor, and also to inhibit group I metabotropic
glutamate receptor function, all of which may ultimately affect NMDA receptor
function (reviewed in Nowak56).
In addition to the work on NMDA receptors, it has been shown that zinc has
the ability to potentiate the action of acetylcholine on the nicotinic acetylcholine
receptors (nAChRs), and fluoxetine has the ability to inhibit acetylcholine’s
action.57 Furthermore, when the two substances are combined, fluoxetine is able
to greatly reduce or even abolish the potentiating action of zinc.57 How this action
of zinc is involved in the etiology and treatment of depression, and whether it is
related to the NMDA mechanism in some way, still remains to be elucidated.
Another interesting finding is that zinc was able to enhance 5-HT uptake in
certain areas of adult rat brain, and that zinc reversed the inhibition of 5-HT
uptake exerted by fluoxetine, imipramine, and 6-nitroquipazine.58 This recent
finding suggests that zinc may very well play a role in modulating serotonergic
neurotransmission. Whether this is related to its ability to inhibit NMDA receptor
function, or interact with nAChRs, requires additional study.
Overall, it appears as though zinc has the capacity to augment pharmacother-
apy in animal models of depression as well as in a preliminary trial in human
subjects. Although the mechanistic basis of this still requires much investigation,
zinc has shown some ability to modulate NMDA receptor activity, nAChR activ-
ity, and influences 5-HT uptake. It is possible one or more of these mechanisms
may explain zinc’s capacity to augment antidepressant pharmacotherapy, but
much more work is needed to clarify these relationships.
While it does not promote growth and, as such, cannot be classified as a nutrient,
St. John’s wort is included in this chapter as it is commonly taken by individuals
suffering from unipolar depression, either as a monotherapy or in combination
with other antidepressant compounds. However, individuals taking this extract
should be aware of the spectrum of drugs with which it can interact. This chapter
will discuss only the interactions related to antidepressants, but for a more
DK5836_C006.fm Page 194 Monday, May 22, 2006 1:51 PM
complete list of drug interactions associated with St. John’s wort, see Zhou.59
Evidence of the efficacy of St. John’s wort in the treatment of major depression
is inconsistent, as reported by a recent meta-analysis.60 Nevertheless, it is still
extremely popular for the treatment of depression, possibly because of the mis-
informed and commonly held notion that all “natural” compounds are safe and
superior to pharmaceuticals.
A review of the literature suggests that co-administration of St. John’s wort
with the tricyclic antidepressant, amitriptyline, results in decreased plasma levels
of the drug as well as two of its metabolites.59 Therefore, co-supplementation of
St. John’s wort with amitriptyline can have drastic effects on the plasma concen-
tration of the drug, reducing its capacity for effective treatment. Cytochrome P450
3A4 (CYP 3A4), which is found in the liver and intestine, plays a role in two
different steps of amitriptyline metabolism.61,62 A literature review showed that
St. John’s wort had the capacity to induce both hepatic and intestinal CYP 3A4
expression,59 which likely results in increased clearance of amitriptyline. Obvi-
ously, healthcare practitioners need to be aware of this important interaction when
treating subjects with unipolar depression.
In addition to its capacity to increase clearance of the tricyclic antidepressant
amitriptyline, St. John’s wort may also contribute to induction of the serotonin
syndrome when taken along with an SSRI. A number of case reports have linked
co-consumption of St. John’s wort and an SSRI with symptoms typical of the
serotonin syndrome.63-65 A number of biologically active components of St. John’s
wort, including hyperforin, adhyperforin, and procyanidins, have the capacity to
inhibit synaptosomal uptake of neurotransmitters.66 St. John’s wort can be con-
sidered a nonselective reuptake inhibitor as it has the capacity to inhibit reuptake
of serotonin, dopamine, and norepinphrine, as well as GABA and glutamate. In
the case of the latter two substances, these are not typically affected by other
antidepressants.67 Therefore, the combination of SSRI treatment, which also
inhibits the reuptake of serotonin, with St. John’s wort could have the capacity
to cause an accumulation of excess serotonin in the synaptic cleft, resulting in
serotonin syndrome. Due to our limited knowledge of the safety of combining
SSRIs with St. John’s wort, it has been suggested that combining these two
substances should be avoided.59
It is crucial that healthcare practitioners, as well as patients being treated with
antidepressants, be aware of the interactions between amitriptyline, SSRIs, and
St. John’s wort. Combining these substances can not only have negative effects
on the outcome of pharmacotherapy, but could also be life threatening. Further-
more, St. John’s wort interacts with a multitude of other drugs, some of which
unipolar depressed patients may be taking for other health problems. Clearly its
use must be carefully monitored by this population.59
6.2.7 VITAMIN B6
There is a fairly well-established relationship between treatment with the MAOI,
phenelzine and vitamin B6 deficiency. Over the years, there have been a number
of reports describing vitamin B6 deficiency associated with phenelzine use.74–76
One open treatment study detected no effects of phenelzine on vitamin B6 levels,
but these results may have been confounded by concomitant use of vitamin
supplements by the subjects.77
Despite finding an average 54% reduction of vitamin B6 levels in subjects
treated with phenelzine as compared to a control group, one study did not find
a correlation between the pyridoxal phosphate (active form of vitamin B6 in the
DK5836_C006.fm Page 196 Monday, May 22, 2006 1:51 PM
body) level and daily dose.76 This indicates that there could be other factors
involved, yet still suggests that phenelzine use can induce a vitamin B6 deficiency.
However, administration of a pyridoxine supplement along with phenelzine has
alleviated the symptoms of vitamin B6 deficiency in patients.74,75
The suspected reason for the vitamin B6 deficiency associated with phenelzine
use, as discussed in Malcolm et al.,76 is the nonenzymatic formation of a pyri-
doxalhydrazone complex involving the two compounds. This could result in a
rapid decrease in plasma levels of pyridoxal phosphate after taking phenelzine.
As is the case for the other supplements listed above, healthcare practitioners as
well as patients need to be aware of this relationship in order to avoid deficiencies
of vitamin B6 while taking phenelzine.
6.2.8 TYRAMINE
Tyramine is a monoamine derived from the amino acid tyrosine and can be found
in many foods. The enzyme, monoamine oxidase, is responsible for the oxidative
deamination of tyramine and, hence, its metabolism. It has been well established
that individuals being treated with a MAOI can have a hypertensive crisis if they
consume large amounts of tyramine. When an individual is taking a MAOI drug,
MAO-A type in the gut wall and liver is inhibited, allowing tyramine from the diet
to enter the circulation.78 Tyramine has vasopressor activity of its own, but also
promotes the release norepinephrine from nerve terminals, both of which contribute
to an increase in blood pressure.78 The hyperadrenergic state induced by consumption
of large quantities of tyramine while taking a MAOI is commonly referred to as “the
cheese reaction” and can result in a number of symptoms. This condition can take
three different forms as reviewed by Brown et al.78 The most common is characterized
by a sudden, severe headache, along with pallor, chills, and neck stiffness; the second
form relates almost exclusively to the cardiovascular system and is characterized by
sudden palpitations, hypertension, chest pain, apprehension, headache, pallor, and
collapse; the third and most devastating manifestation of the condition is character-
ized by intracerebral hemorrhage and death.78
A diet has been devised in order to assure that individuals taking MAOI drugs
do not consume large amounts of tyramine. This diet has gone through extensive
refining in order to be as unrestrictive as possible, helping to ensure compliance
with medications. Foods that are to be avoided on this diet, due to their high
levels of tyramine, include aged cheese, aged or cured meats, any potentially
spoiled meat/poultry/fish, broad (fava) bean pods, marmite (concentrated yeast
extract), sauerkraut, soy sauce and soy bean condiments, and tap beer.79 However,
pizza bought from large-chain commercial outlets appears to be safe, even with
double-cheese and double-pepperoni.80 Nevertheless, it has been suggested that
caution be exercised if ordering from smaller pizza outlets or if purchasing a
gourmet pizza that could contain aged cheeses.80 By receiving accurate dietary
advice about the MAOI diet, patients taking this class of drug should be able to
prevent potentially lethal hypertensive crises.
DK5836_C006.fm Page 197 Monday, May 22, 2006 1:51 PM
Another suggests that omega-3 fatty acids have some capacity to regulate sero-
tonergic neurotransmission.87 Thirdly, it has been noted that unipolar depression
is associated with an acute phase response, with increased secretions of proin-
flammatory eicosanoids, and excessive secretion of proinflammatory cytokines.88
Therefore, by limiting the available omega-3 fatty acids, the relative abundance
of omega-6 PUFAs is increased (including arachidonic acid), which leads to
excess synthesis of proinflammatory eicosanoids. (For further discussion and
metabolic pathway descriptions, see Chapter 4.) Each of these mechanisms rep-
resents a large area of study currently undergoing intensive research. How, and
to what extent, each of these mechanisms is involved in the therapeutic benefit
of omega-3 fatty acids in unipolar depression requires much clarification. How-
ever, it is possible, and even likely, that these mechanisms are integrated or that
more than one mechanism contributes to the beneficial properties of these fatty
acids. Overall, the omega-3 fatty acids show promise in the augmentation of
traditional antidepressant treatments for unipolar depression. Nevertheless, more
research is required
6.3 CONCLUSION
It appears that there are a number of nutrients that may have the capacity to
interact with current antidepressants in a positive manner. A better understanding
of these relationships could theoretically allow for nutritional modifications, or
the use of dietary supplements, which could improve the efficacy of unipolar
depression pharmacotherapy. Furthermore, such dietary alterations could decrease
the required dose of traditional antidepressants resulting in a consequent lessening
of negative side effects. In addition, there are a number of dietary components
that can cause various detrimental effects when combined with antidepressants.
A clear understanding of these relationships is important for healthcare practi-
tioners when prescribing unipolar depression medications in order to avoid any
unnecessary suffering for the patient.
Understanding the mechanistic basis of the beneficial nutrient–drug interac-
tions is of the utmost importance for their utilization in the pharmacotherapy of
individuals suffering from unipolar depression. However, many of these interac-
tions are currently explained by seemingly disparate mechanisms, which is a good
indication that much more research is needed in this area. Conversely, under-
standing the mechanistic basis of the detrimental nutrient–drug interactions could
help prevent adverse side effects from occurring as well as help improve our
understanding of the etiology of the condition.
While the “monoamine theory of unipolar depression” has proved at least
partly useful in developing treatments for unipolar depression, it has also left
much room for improvement. It is likely that pharmacotherapy will advance
greatly as we attain a better understanding of the molecular and cellular basis of
this condition. Furthermore, such an understanding will also allow for enhanced
utilization of dietary modifications that assist in pharmacotherapy, and avoidance
of dietary components that could result in unwanted side effects.
TABLE 6.1
Summary of Nutrients and Herbals in the Pharmacotherapy of Unipolar/Major Depression
Nutrient or
Herbal Molecular Target or Possible Confounders or
Supplement Suspected Activity Possible Negative Interactions
Other large neutral amino acids compete to get across blood–brain barrier
Tryptophan can be shunted into niacin or protein production (5-HTP cannot)
The type of drug being taken can determine efficacy of tryptophan
Subject variation: 5-HT receptor plasticity, the relative abundance of precursor
amino acids already available, stage of pharmacotherapy
DK5836_C006.fm Page 199 Monday, May 22, 2006 1:51 PM
Folate, vitamin B12, 1. Decreased recycling of homocysteine decreases the body’s Dose of folate needed to augment pharmacotherapy may be higher in males
(and methylation capacity, which could impair production of than females (it may have to be high enough to decrease homocysteine levels)
homocysteine) neurotransmitters and membrane phospholipids Interactions between folate, B12, and homocysteine may make interpretations
(continued) 2. Decreased production of tetrahydrobiopterin (BH4), which is of results difficult
a cofactor for tryptophan hydroxylase and tyrosine hydroxylase
(rate-limiting steps in monoamine synthesis)
DK5836_C006.fm Page 200 Monday, May 22, 2006 1:51 PM
Ginkgo biloba Bilobalide may increase GABA levels in the brain by increasing Negative Interaction:
glutamic acid decarboxylase levels Suspected to have caused a coma in an elderly woman with Alzheimer’s
May increase CYP 3A4 activity resulting in increased conversion disease, when taken along with trazodone
of trazodone to its active metabolite
Panax ginseng Ginseng sapoins observed to block nAChRs Negative Interaction:
Ability to inhibit NMDA receptors When taken with phenelzine, it may result in mania
Vitamin B6 B6 and phenelzine nonenzymatically form a complex, resulting Negative Interaction:
in a loss of available B6 Phenelzine treatment observed to cause B6 deficiency
Confounders:
Mixed results of the studies might be the result of concomitant use of vitamin
supplements in some studies
201
202
Tyramine Monoamine oxidase (MAO) is needed for the oxidative Negative Interaction:
deamination of tyramine A hypertensive crisis may result if foods high in tyramine are consumed by
When MAO is blocked, tyramine enters the circulation an individual taking an MAOI drug
Tyramine has vasopressor activity and also releases
norepinephrine from nerve terminals
The end result is an increase in blood pressure
DK5836_C006.fm Page 202 Monday, May 22, 2006 1:51 PM
Omega-3 PUFAs Three theories suggest how fatty acids may relate to mood Confounders:
(EPA and DHA) disorders: EPA, DHA, and the combination of EPA + DHA may have differing efficacies
1. Mood disorders may be caused by impaired phospholipids Patient variables, such as baseline fatty acid profiles, may also determine
metabolism and impaired fatty-acid related signal transduction efficacy
2. Omega-3 fatty acids may have the ability to regulate Loss of blinding may occur in supplementation studies due to a fishy aftertaste
serotonergic neurotransmission after consuming the fish oil
3. Limitation of omega-3 PUFAs leads to excess omega-6 PUFAs,
which are the precursors to proinflammatory eicosanoids
Nutrient–Drug Interactions
DK5836_C006.fm Page 203 Monday, May 22, 2006 1:51 PM
REFERENCES
1. Kalia M. Neurobiological basis of depression: an update. Metab Clin Exp 2005;
54, pt. Suppl.(5): 24–27.
2. To SE, Zepf RA, and Woods AG. The symptoms, neurobiology, and current
pharmacological treatment of depression. J Amer Assoc Neurosci Nurse 2005;
37(2): 102–107.
3. Birmes P, Coppin D, Schmitt L, and Lauque D. Serotonin syndrome: a brief review.
Can Med Assoc J 2003; 168(11): 1439–1442.
4. Popoli M, Brunello N, Perez J and Racagni G. Second messenger-regulated protein
kinases in the brain: their functional role and the action of antidepressant drugs.
J Neurochem 2000; 74(1): 21–33.
5. Skolnick P, Layer RT, Popik P, Nowak G, Paul IA and Trullas R. Adaptation of
N-methyl-D-aspartate (NMDA) receptors following antidepressant treatment:
implications for the pharmacotherapy of depression. PharmacoPsych 1996; 29(1):
23–26.
6. Turner EH, Loftis JM and Blackwell AD. Serotonin a la carte: supplementation
with the serotonin precursor 5-hydroxytryptophan. Pharmacol Ther 2006; 109(3):
325–338.
7. Møller SE. 5-HT uptake inhibitors and tricyclic antidepressants: relation between
trytophan availability and clinical response in depressed patients. Eur J Neurop-
sychopharmacology 1990; 1(1): 41–44.
8. Lucini V, Lucca A, Catalano M and Smeraldi E. Predictive value of tryp-
tophan/large neutral amino acids ratio to antidepressant response. J Affect Disord
1996; 36(3-4): 129–133.
9. Møller SE. Plasma amino acid profiles in relation to clinical response to moclobe-
mide in patients with major depression. J Affect Disord 1993; 27(4): 225–231.
10. Møller SE, Ødum K, Kirk L, Bjerre M, Fog-Møller F and Knudsen A. Plasma
tyrosine/neutral amino acid ratio correlated with clinical response to nortriptyline
in endogenously depressed patients. J Affect Disord 1985 9(3): 223–229.
11. Porter RJ, Mulder RT, Joyce PR and Luty SE. Tryptophan and tyrosine availability
and response to antidepressant treatment in major depression. J Affect Disord
2005; 86(2-3): 129–134.
12. Porter RJ, Mulder RT and Joyce PR. Baseline prolactin and L-tryptophan avail-
ability predict response to antidepressant treatment in major depression. Psycho-
pharmacology (Berl) 2003; 165(3): 216.
13. Fekkes D, Timmerman L and Pepplinkhuizen L. Effects of clomipramine on
plasma amino acids and serotonergic parameters in panic disorder and depression.
Europ Neuropsychopharma 1997; 7(3): 235–239.
14. Mctavish SFB, Mannie ZN and Cowen PJ. Tyrosine depletion does not cause
depressive relapse in antidepressant-treated patients. Psychopharmacology (Berl)
2004; 175(1): 124.
15. Delgado PL, Miller HL, Salomon RM, Licinio J, Krystal JH, Moreno FA, Heninger
GR, Charney DS. Tryptophan-depletion challenge in depressed patients treated
with desipramine or fluoxetine: implications for the role of serotonin in the
mechanism of antidepressant action. Biol Psych 1999; 46(2): 212–220.
DK5836_C006.fm Page 204 Monday, May 22, 2006 1:51 PM
16. Booij L, Van der Does AJW, Haffmans PMJ and Riedel WJ. Acute tryptophan
depletion in depressed patients treated with a selective serotonin-noradrenalin
reuptake inhibitor: augmentation of antidepressant response? J Affect Disord 2005;
86(2-3): 305–311.
17. Levitan RD, Shen J, Jindal R, Driver HS, Kennedy SH and Shapiro CM. Prelim-
inary randomized double-blind placebo-controlled trial of tryptophan combined
with fluoxetine to treat major depressive disorder: antidepressant and hypnotic
effects. J Psychiatr Neurosci 2000; 25(5): 439.
18. Markus CR, Olivier B, Panhuysen GE, Van Der Gugten J, Alles MS, Tuiten A,
Westenberg HG, Fekkes D, Koppeschaar HF, de Haan EE. The bovine protein
alpha-lactalbumin increases the plasma ratio of tryptophan to the other large
neutral amino acids, and in vulnerable subjects raises brain serotonin activity,
reduces cortisol concentration, and improves mood under stress. Am J Clin Nutr
2000; 71(6): 1536–1544.
19. Beulens JWJ, Bindels JG, de Graaf C, Alles MS and Wouters-Wesseling W. Alpha-
lactalbumin combined with a regular diet increases plasma Trp–LNAA ratio.
Physiol Behav 2004; 81(4): 585–593.
20. Birdsall TC. 5-Hydroxytryptophan: a clinically-effective serotonin precursor.
Alternat Med Rev 1998; 3(4): 271–280.
21. Skop BP, Finkelstein JA, Mareth TR, Magoon MR and Brown TM. The serotonin
syndrome associated with paroxetine, an over-the-counter cold remedy, and vas-
cular disease. Am J Emerg Med 1994; 12(6): 642–644.
22. Coppen A, Bolander-Gouaille C. Treatment of depression: time to consider folic
acid and vitamin B12. J Psychopharma 2005; 19(1): 59.
23. Morris MS, Fava M, Jacques PF, Selhub J and Rosenberg IH. Depression and
folate status in the U.S. population. Psychother Psychosom 2003; 72(2): 80–89.
24. Fava M, Borus JS and Alpert JE. Folate, vitamin B12, and homocysteine in major
depressive disorder. Am J Psych 1997; 154: 426–428.
25. Reutens S, Sachdev P. Homocysteine in neuropsychiatric disorders of the elderly.
Int J Geriatr Psych 2002; 17(9): 859.
26. Hintikka J, Tolmunen T, Tanskanen A and Viinamaki H. High vitamin B12 level
and good treatment outcome may be associated in major depressive disorder. BMC
Psych [electronic resource] 2003; 3: 17. E-pub: 2003 Dec 02.
27. Scott TM, Tucker KL, Bhadelia A, Benjamin B, Patz S, Bhadelia R, Liebson E,
Price LL, Griffith J, Rosenberg I, Folstein MF. Homocysteine and B vitamins
relate to brain volume and white-matter changes in geriatric patients with psychi-
atric disorders. Amer J Geriatr Psych 2004; 12(6): 631–638.
28. Papakostas GI, Petersen T, Mischoulon D, Green CH, Nierenberg AA, Bottiglieri
T, Rosenbaum JF, Alpert JE, Fava M. Serum folate, vitamin B12, and homocys-
teine in major depressive disorder, Part 1: predictors of clinical response in flu-
oxetine-resistant depression. J Clin Psych 2004; 65(8): 1090–1095.
29. Papakostas GI, Petersen T, Mischoulon D, Green CH, Nierenberg AA, Bottiglieri
T, Rosenbaum JF, Alpert JE, Fava M. Serum folate, vitamin B12, and homocys-
teine in major depressive disorder, Part 2: predictors of relapse during the contin-
uation phase of pharmacotherapy. J Clin Psych 2004; 65(8): 1096–1098.
30. Mischoulon D, Burger JK, Spillmann MK, Worthington JJ, Fava M and Alpert
JE. Anemia and macrocytosis in the prediction of serum folate and vitamin B12
status, and treatment outcome in major depression. J Psychosom Res 2000; 49(3):
183–187.
DK5836_C006.fm Page 205 Monday, May 22, 2006 1:51 PM
31. Lee S, Wing YK and Fong S. A controlled study of folate levels in Chinese
inpatients with major depression in Hong Kong. J Affect Disord 1998; 49(1):
73–77.
32. Godfrey PS, Toone BK, Carney MW, Flynn TG, Bottiglieri T, Laundy M, Chanarin
I, Reynolds EH. Enhancement of recovery from psychiatric illness by methyl-
folate. Lancet 1990; 336(8712): 392–395.
33. Coppen A, Bailey J. Enhancement of the antidepressant action of fluoxetine by
folic acid: a randomised, placebo controlled trial. J Affect Disord 2000; 60(2):
121–130.
34. Alpert JE, Mischoulon D, Rubenstein GEF, Bottonari K, Nierenberg AA and Fava
M. Folinic acid (leucovorin) as an adjunctive treatment for SSRI-refractory depres-
sion. Annal Clin Psych 2002; 14(1): 33–38.
35. Taylor MJ, Carney S, Geddes J and Goodwin G. Folate for depressive disorders.
Cochrane database of systematic reviews (Online: Update Software) 2003; (2):
CD003390.
36. Mischoulon D, Fava M. Role of S-adenosyl-L-methionine in the treatment of
depression: a review of the evidence. Amer J Clin Nutr 2002; 76(5): 1158S–61S.
37. Fava M, Giannelli A, Rapisarda V, Patralia A and Guaraldi GP. Rapidity of onset
of the antidepressant effect of parenteral S-adenosyl-l-methionine. Psych Res
1995; 56(3): 295–297.
38. Bell KM, Plon L and Bunney WE. S-adenosylmethionine treatment of depression:
a controlled clinical trial. Am J Psych 1988; 145: 1110–1114.
39. Delle Chiaie R, Pancheri P and Scapicchio P. Efficacy and tolerability of oral and
intramuscular S-adenosyl-L-methionine 1,4-butanedisulfonate (SAMe) in the
treatment of major depression: comparison with imipramine in 2 multicenter
studies. Am J Clin Nutr 2002; 76(5): 1172S–1176S.
40. Saletu B, Anderer P, Di Padova C, Assandri A and Saletu-Zyhlarz GM. Electro-
physiological neuroimaging of the central effects of S-adenosyl-L-methionine by
mapping of electroencephalograms and event-related potentials and low-resolution
brain electromagnetic tomography. Am J Clin Nutr 2002; 76(5): 1162S–71S.
41. Berlanga C, Ortega-Soto Hé A, Ontiveros M and Senties Hé. Efficacy of S-
adenosyl-L-methionine in speeding the onset of action of imipramine. Psych Res
1992; 44(3): 257–262.
42. Consogno E, Tiraboschi E, Iuliano E, Gennarelli M, Racagni G and Popoli M.
Long-term treatment with s-adenosylmethionine induces changes in presynaptic
cam kinase II and synapsin I. Biol Psych 2001; 50(5): 337–344.
43. Bellido I, Gomez-Luque A, Plaza A, Rius F, Ortiz P and Sanchez de la Cuesta, F.
S-adenosyl-l-methionine prevents 5-HT1A receptors up-regulation induced by
acute imipramine in the frontal cortex of the rat. Neurosci Lett 2002; 321(1-2):
110–114.
44. Young SN. Are SAMs and 5-HTP safe and effective treatments for depression? J
Psychiatr Neurosci 2003; 28(6): 471.
45. McLoughlin IJ, Hodge JS. Zinc in depressive disorder. Acta Psychiatr Scand 1990;
82(6): 451–453.
46. Narang RL, Gupta KR, Narang AP and Singh R. Levels of copper and zinc in
depression. Indian J Physiol Pharmacol 1991; 35(4): 272–274.
47. Nowak G, Schlegel-Zawadzka M. Alterations in serum and brain trace element
levels after antidepressant treatment: part I. Zinc. Biol Trace Elem Res 1999; 67(1):
85–92.
DK5836_C006.fm Page 206 Monday, May 22, 2006 1:51 PM
65. Spinella M, Eaton LA. Hypomania induced by herbal and pharmaceutical psy-
chotropic medicines following mild traumatic brain injury. Brain Injury 2002;
16(4): 359.
66. Wonnemann M, Singer A, Siebert B and Muller WE. Evaluation of synaptosomal
uptake inhibition of most relevant constituents of St. John's wort. PharmacoPsych
2001; 34 Suppl 1: S148–S151.
67. Nathan PJ. Hypericum perforatum (St. John's Wort): a non-selective reuptake
inhibitor? A review of the recent advances in its pharmacology. J Psychopharma-
col 2001; 15(1): 47.
68. Galluzzi S, Zanetti O, Binetti G, Trabucchi M and Frisoni GB. Coma in a patient
with Alzheimer's disease taking low dose trazodone and gingko biloba. J Neurol
Neurosurg Psych 2000; 68(5): 679–680.
69. Hu Z, Yang X, Ho PC, Chan SY, Heng PW, Chan E, Duan W, Koh HL, Zhou S.
Herb-drug interactions: a literature review. Drugs 2005; 65(9): 1239–1282.
70. Sasaki K, Hatta S, Haga M and Ohshika H. Effects of bilobalide on gamma-
aminobutyric acid levels and glutamic acid decarboxylase in mouse brain. Eur J
Pharmacol 1999; 367(2-3): 165–173.
71. Shinozuka K, Umegaki K, Kubota Y, Tanaka N, Mizuno H, Yamauchi J, Nakamura
K, Kunitomo M. Feeding of Ginkgo biloba extract (GBE) enhances gene expres-
sion of hepatic cytochrome P-450 and attenuates the hypotensive effect of nicar-
dipine in rats. Life Sci 2002; 70(23): 2783–2792.
72. Sala F, Mulet J, Choi S, Jung SY, Nah SY, Rhim H, Valor LM, Criado M, Sala
S. Effects of ginsenoside Rg2 on human neuronal nicotinic acetylcholine recep-
tors. J Pharmacol Exp Ther 2002; 301(3): 1052–1059.
73. Kim S, Ahn K, Oh TH, Nah SY and Rhim H. Inhibitory effect of ginsenosides
on NMDA receptor-mediated signals in rat hippocampal neurons. Biochem Bio-
phys Res Commun 2002; 296(2): 247–254.
74. Demers RG, McDonagh PH and Moore RJ. Pyridoxine deficiency with phenelzine.
South Med J 1984; 77(5): 641–642.
75. Stewart JW, Harrison W, Quitkin F and Liebowitz MR. Phenelzine-induced pyri-
doxine deficiency. J Clin Psychopharmacol 1984; 4(4): 225–226.
76. Malcolm DE, Yu PH, Bowen RC, O'Donovan C, Hawkes J and Hussein M.
Phenelzine reduces plasma vitamin B6. J Psychiatr Neurosci 1994; 19(5):
332–334.
77. Lydiard RB, Laraia MT, Howell EF and Fossey MD. Phenelzine treatment of
panic disorder: lack of effect on pyridoxal phosphate levels. J Clin Psychophar-
macol 1989; 9(6): 428–431.
78. Brown C, Taniguchi G and Yip K. The monoamine oxidase inhibitor-tyramine
interaction. J Clin Pharmacol 1989; 29(6): 529–532.
79. Gardner DM, Shulman KI, Walker SE and Tailor SA. The making of a user-
friendly MAOI diet. J Clin Psych 1996; 57(3): 99–104.
80. Shulman KI, Walker SE. Refining the MAOI diet: tyramine content of pizzas and
soy products. J Clin Psych 1999; 60(3): 191–193.
81. Tanskanen A, Hibbeln JR, Tuomilehto J, Uutela A, Haukkala A, Viinamaki H,
Lehtonen J, Vartiainen E. Fish consumption and depressive symptoms in the
general population in Finland. Psychiatr Serv 2001; 52(4): 529–531.
82. Maes M, Christophe A, Delanghe J, Altamura C, Neels H and Meltzer HY.
Lowered omega-3 polyunsaturated fatty acids in serum phospholipids and cho-
lesteryl esters of depressed patients. Psych Res 1999; 85(3): 275–291.
DK5836_C006.fm Page 208 Monday, May 22, 2006 1:51 PM
7 Supplements and
Anesthesiology
Alan D. Kaye, Amir Baluch, and
Jason M. Hoover
CONTENTS
7.1 Introduction..............................................................................................210
7.2 Minerals ...................................................................................................210
7.2.1 Calcium........................................................................................210
7.2.2 Chromium ....................................................................................210
7.2.3 Magnesium ..................................................................................211
7.2.4 Iron...............................................................................................212
7.2.5 Selenium ......................................................................................212
7.2.6 Zinc ..............................................................................................212
7.3 Vitamins...................................................................................................213
7.3.1 Vitamin A.....................................................................................213
7.3.2 Vitamin B12 ..................................................................................213
7.3.3 Vitamin C.....................................................................................214
7.3.4 Vitamin D ....................................................................................215
7.3.5 Vitamin E.....................................................................................215
7.3.6 Folate ...........................................................................................215
7.4 Herbals.....................................................................................................216
7.4.1 Saw Palmetto ...............................................................................216
7.4.2 St. John’s Wort ............................................................................216
7.4.3 Echinacea.....................................................................................217
7.4.4 Feverfew ......................................................................................220
7.4.5 Ephedra ........................................................................................220
7.4.6 Ginger ..........................................................................................221
7.4.7 Garlic ...........................................................................................222
7.4.8 Gingko Biloba .............................................................................223
7.4.9 Kava Kava....................................................................................224
7.4.10 Ginseng ........................................................................................225
7.5 Conclusion ...............................................................................................226
References .........................................................................................................226
209
DK5836_C007.fm Page 210 Monday, May 22, 2006 1:57 PM
7.1 INTRODUCTION
The use of supplements such as minerals, vitamins, and herbal products has
increased dramatically in recent years. Reasons for such an increase in prevalence
include anecdotal reports on efficacy, impressive advertisement, lower cost of
products compared to prescription medications, and ease of attainment of the
supplements. Regardless of the reasons, it is important that physicians, particu-
larly the anesthesiologist, be cognizant of the effects of these agents, whether
beneficial or harmful.
7.2 MINERALS
7.2.1 CALCIUM
It may be reasonable for patients to supplement their diet with calcium, as calcium
deficiency is a common problem.1 Many women supplement with calcium to
improve symptoms associated with premenstrual syndrome.2
Calcium may interfere with a host of commonly used drugs. The anesthesi-
ologist must be aware of patients with cardiac problems that may be taking
calcium channel blockers or beta-blockers. The effects of calcium channel block-
ers may be affected by calcium supplementation, as calcium has been shown to
antagonize the effects of verapamil.3 In fact, calcium has recently been used in
the successful management of calcium channel blocker overdose.4 Calcium sup-
plementation may also decrease levels of beta-blockers, leading to a greater
chronotropic and inotropic presentation than one would expect.5
Thiazide diuretics have been shown to increase serum calcium concentrations,
possibly leading to hypercalcemia due to increased reabsorption of calcium in
the kidneys. Dysrhythmias may occur in patients taking digitalis and calcium
together. The antibiotic effect of tetracyclines and quinolone, and pharmacological
blood levels of bisphosphonates and levothyroxine may be decreased with calcium
supplementation; these medications should not be taken within 2 hours of calcium
intake.6,7
Calcium supplementation may also affect the choice of anesthesia used in
operative procedures. Elevated levels of calcium may complicate cardiopulmo-
nary bypass procedures by worsening mechanical injury to erythrocytes. Recent
data suggest that the use of propofol may have a protective effect on erythrocytes
in patients with elevated levels of calcium.8 Documenting the use of calcium by
patients preoperatively may prevent many of these drug interactions.
7.2.2 CHROMIUM
Chromium is an essential nutrient involved in metabolism of carbohydrates and
lipids. Recently, chromium has received attention from consumers in the belief
that it may improve glucose tolerance in diabetics, reduce body fat, and reduce
atherosclerotic formation. These purported effects stem from chromium’s effect
DK5836_C007.fm Page 211 Monday, May 22, 2006 1:57 PM
on insulin resistance. However, the evidence regarding its use for insulin resis-
tance and mildly impaired glucose tolerance is inconclusive.9–12
A double blind trial with 180 patients concluded that high doses of chromium
supplementation (1000 mg) could have beneficial effects on hemoglobin A1c,
insulin, cholesterol, and overall glucose control in type 2 diabetics.13 The anes-
thesiologist should consider asking any diabetics if they supplement with chro-
mium in an attempt to attain these effects. Because of chromium’s effects on
insulin resistance and impaired glucose control, some patients will supplement
with this mineral to prevent risk of cardiovascular disease. Human studies have
shown decreased levels of cholesterol and triglycerides in elderly patients taking
200 µg twice a day.14
Chromium is generally well tolerated; however, some patients may experience
nervous system symptoms, such as perceptual, cognitive, and motor dysfunction
with doses as low as 200 to 400 µg.15 In addition, toxicity has been reported with
chromium consumption. In one case, a woman developed anemia, thrombocy-
topenia, hemolysis, weight loss, and liver and renal toxicity when attempting
weight loss with 1200 to 2400 µg of chromium picolinate. These problems
resolved after discontinuation of chromium ingestion.16 A lower dose of only 600
µg was demonstrated to have resulted in interstitial nephritis in another female
patient.17
7.2.3 MAGNESIUM
Magnesium plays many important roles in structure, function, and metabolism
and is involved in numerous essential physiologic reactions in the human body.
The supplementing of magnesium has been used extensively by patients with
cardiovascular disease, diabetes, osteoporosis, asthma, and migraines, although
most individuals consume adequate levels in their diet.18 Patients with a history
of these illnesses may be supplementing with magnesium and, therefore, should
be questioned.
The most obvious anesthetic consideration in treating a patient taking mag-
nesium supplementation has to do with its effect on muscle relaxants. The mineral
can potentiate the effects of nondepolarizing skeletal muscle relaxants, such as
tubocurarine. Therefore, it may be advisable to ask patients about their magne-
sium usage preoperatively to avoid complications.6
Anesthesiologists caring for obstetrical patients must be aware of the effects
of magnesium sulfate in the patient undergoing caesarean section. Literature
suggests that the duration of action of relaxant anesthetics, such as mivacuronium,
may be affected by subtherapeutic serum magnesium levels.19 Magnesium may
also interfere with the absorption of antibiotics, such as tetracyclines, flurorqui-
nolones, nitrofurantoins, penicillamine, ACE inhibitors, phenytoin, and H2 block-
ers. Absorption problems can be ameliorated by not taking doses of magnesium
within 2 hours from these other medications.20–23 The mineral may also make
oral hypoglycemics, specifically sulfonylureas, more effective when used, thus
increasing the risk of hypoglycemic episodes.24
DK5836_C007.fm Page 212 Monday, May 22, 2006 1:57 PM
7.2.4 IRON
In both developed and underdeveloped countries, iron deficiency is the most
common nutrient deficiency. Worldwide, at least 700 million individuals have
iron-deficiency anemia.25 More than just a constituent of hemoglobin and myo-
globin, iron is a key component in nearly every living organism and, in humans,
is associated with hundreds of enzymes and other proteins structures. People have
supplemented with iron in order to treat iron deficiency anemia, alleviate poor
cognitive function in children, increase athletic performance, and suppress restless
legs syndrome (RLS).
High concentrations of iron in the blood may worsen neuronal injury sec-
ondary to cerebral ischemia.26 Increased iron levels during pregnancy may lead
to preterm delivery and neonatal asphyxia.27 These complications may occur even
with normal iron intake if the patient also takes vitamin C, as high doses of the
vitamin can increase iron absorption.28
Iron may inhibit absorption of many drugs including levodopa, methyldopa,
carbidopa, penicillamine, thyroid hormone, captopril, and antibiotics in the qui-
nolone and tetracycline family.29–33 Some medications may decrease iron absorp-
tion and lead to decreased therapeutic levels of the mineral. This includes antacids,
histamine (H2) receptor antagonists, proton pump inhibitors, and cholestyramine
resin.6,7 Iron should not be given orally within 2 hours of other pharmaceuticals
to avoid alterations in drug or mineral absorption.
7.2.5 SELENIUM
Selenium, an essential trace element, functions in a variety of enzyme-dependent
pathways, especially those utilizing selenoproteins. Much of its supplemental
efficacy is due to its antioxidant properties. Glutathione peroxidase incorporates
this mineral at its active site and, as dietary selenium intake decreases, glutathione
levels drop.34
Patients supplement with selenium for a variety of reasons, most notably a
supposed improvement in immune status. Elderly patients may be inclined to
supplement with selenium for this reason.Toxicity with selenium supplementation
begins at intake greater than 750 µg/day and may manifest as garlic-like breath,
loss of hair and fingernails, gastrointestinal distress, or central nervous system
changes.35,36 Few interactions with other pharmacological agents have been
found.6
7.2.6 ZINC
Zinc deficiency was first described in 1961, when it was found to be associated
with “adolescent nutritional dwarfism” in the Middle East.37 Deficiency is this
mineral is thought to be quite common in infants, adolescents, women, and the
elderly.38–41 The most well-known use for zinc supplementation is in treatment
of the common cold, caused principally by the rhinovirus.
DK5836_C007.fm Page 213 Monday, May 22, 2006 1:57 PM
7.3 VITAMINS
7.3.1 VITAMIN A
The term “vitamin A” refers to a large number of related compounds: preformed
retinol (an alcohol) and retinal (an aldehyde). Vitamin A deficiency is common
in teenagers, lower socioeconomic groups, and in developing countries.44 Fur-
thermore, some studies indicate that diabetic patients are at an increased risk for
vitamin A deficiency.45 This deficiency may manifest as night blindness, immune
deterioration, birth defects, or decreased red blood cell production.46 Purported
therapeutic uses for vitamin A include diseases of the skin, acute promyelotic
leukemia, and viral infections.
Retinoids (a class of chemical compounds that are related chemically to
vitamin A) have been used as pharmacologic agents to treat disorders of the skin.
Psoriasis, acne, and rosacea have been treated with natural or synthetic retinoids.
Moreover, retinoids are effective in treating symptoms associated with congenital
keratinization disorder syndromes. Therapeutic effects stem from its antineoplas-
tic activity.47 Patients suffering from these illnesses may be supplementing with
vitamin A and their dosages should be explored.
Vitamin A may increase anticoagulant effects of warfarin.48 This interaction
could increase the risk of bleeding complications in surgical patients, which can
be avoided by informing the patient of this effect preoperatively
Excess vitamin A intake during pregnancy, as well as deficiency, may lead
to birth defects. For this reason, pregnant woman who are not vitamin A deficient
should not consume more than 2600 IU/day of supplemental retinol.49 Patients
using isotretinoin and pregnant women taking valproic acid, likewise, are at
increased risk for vitamin A toxicity.46,50 Finally, alcohol consumption decreases
the liver toxicity threshold for vitamin A, thereby narrowing its therapeutic win-
dow in alcoholics.51
7.3.3 VITAMIN C
Ascorbic acid, also known as vitamin C, is an essential water-soluble vitamin.
The symptoms of scurvy, which include bleeding and easy bruising, can be
prevented with as little as 10 mg of vitamin C due to its association with collagen,
but it can also be used to prevent a host of other disease processes.62
Numerous people moderately supplement their diet with vitamin C in order
to prevent infection from viruses responsible for the common cold, yet research
reviews over the past 20 years conclude that there is no significant impact on the
incidence of infection.63 However, there are a few studies that show that certain
groups of people who are susceptible to low dietary intake of vitamin C, such as
marathon runners, may be less susceptible when supplementation is used. Fur-
thermore, vitamin C may decrease the duration or severity of colds via an anti-
histamine effect when taken in large doses.64
There is some evidence that patients taking vitamin C supplements may have
a reduced anticoagulant effect from warfarin or heparin. Increased doses of these
anticoagulants might be advised to achieve therapeutic levels.65,66 It is recom-
mended that patients on anticoagulation therapy should limit vitamin C intake to
1 g/day. As always, the precise dosage regimen must be monitored by the appro-
priate lab studies. Since high doses may also interfere with certain laboratory
tests, such as serum bilirubin, creatinine, and stool guaiac assay, it is crucial to
inquire about any over-the-counter supplementation with the vitamin.6 There is
evidence that vitamin C may increase the inotropic effect of dobutamine in
patients with abnormal left ventricular function. Infusion of vitamin C into indi-
viduals with normal heart function was shown to increase contractility of the left
ventricle.67 High doses of vitamin C may increase acetaminophen levels, while
aspirin and oral contraceptives may lower serum levels of vitamin C.68–70
DK5836_C007.fm Page 215 Monday, May 22, 2006 1:57 PM
7.3.4 VITAMIN D
Vitamin D deficiency does occur in the elderly and shows increased incidence in
people that live in Northern latitudes.71,72 The main function of this vitamin is in
calcium homeostasis. Individuals with osteoporosis frequently have a deficiency
in vitamin D.73 With increasing age, vitamin D and calcium metabolism increase
the risk of deficiency. Studies show a clear benefit of vitamin D and calcium
supplementation in older postmenopausal women. Supplementation results in
increased bone density, decreased bone turnover, and decreased nonvertebral
fractures, as well as decreases in fall risk and body sway.74
Hypervitaminosis D can occur with high doses of the vitamin. Symptoms
include nausea, vomiting, loss of appetite, polydipsia, polyuria, itching, muscular
weakness, joint pain, and, in some cases, can lead to coma and death.46 In order
to prevent the syndrome, the Food and Nutrition Board has set an upper limit of
supplementation at 2000 IU/day for adults.75
The cardiac patient taking calcium channel blockers may present for surgery
while concurrently taking supplemental vitamin D and calcium. The combination
of vitamin D and calcium may interfere with calcium channel blockers by antag-
onizing its effect. Hypercalcemia exacerbates arrhythmias in patients taking dig-
italis. A state of hypercalcemia may be induced by the concomitant use of thiazide
diuretics with vitamin D, which may lead to these complications. Conversely,
anticonvulsants, cholesterol-lowering medications, and the fat substitute olestra
may decrease the absorption of vitamin D.76
7.3.5 VITAMIN E
Antioxidant properties define the primary function of vitamin E. Dietary defi-
ciency is quite prevalent even in the developed world; therefore, supplementation
is reasonable.77 The anesthesiologist must be keenly aware of vitamin E supple-
mentation, as it may increase the effects of anticoagulant and antiplatelet drugs.
Concomitant use of vitamin E with these drugs may increase the risk of hemor-
rhage.78 Further, preliminary evidence suggests that type 2 diabetics may have
an increased risk of hypoglycemia since vitamin E may enhance insulin sensitivity
and, therefore, adjustment of oral hypoglycemics would be advisable.79,80
Cholestyramine, colestipol, isoniazid, mineral oil, orlistat, sucralfate, and the fat
substitute olestra may possibly decrease the absorption of vitamin E, leading to
decreased levels in the serum.6
7.3.6 FOLATE
Folic acid and folate have been used interchangeably, although the most stable
form that is used by the human body is folic acid. This water-soluble, B-complex
vitamin occurs naturally in foods and in metabolically active forms.81 Since 1998,
the fortification of cereal with folate has significantly decreased the prevalence
of folate deficiency.82
DK5836_C007.fm Page 216 Monday, May 22, 2006 1:57 PM
Excess folate intake has not been associated with any significant adverse
effects. Patients taking large amounts of nonsteroidal antiinflammatory drugs
(NSAIDs), such as aspirin or ibuprofen, experience interference in folate metab-
olism, although regular use shows no significant changes. Patients suffering from
seizures who use phenytoin for therapy may report decrease in seizure threshold
when taking folate supplements.83 The body’s ability to absorb or utilize folate
may be decreased if taking nitrous oxide, antacids, bile acid sequestrants, H2
blockers, certain anticonvulsants, and high-dose triamterene. Supplementation of
folic acid may also correct for megaloblastic anemia due to B12 deficiency, but
the neurological damage will not be prevented. In these cases, one must be careful
to pinpoint the true cause of the anemia to prevent neurological complications.43
7.4 HERBALS
7.4.1 SAW PALMETTO
Saw palmetto is used mainly for treatment of benign prostatic hyperplasia with
free fatty acids and sterols being the main components.84 Despite an uncertain
mechanism, the literature does demonstrate antagonism at the androgen receptor
for dihydrotestosterone and 5α-reductase enzyme.84 Though prostate size and
prostate-specific antigen level are not decreased by saw palmetto, biopsies have
demonstrated decreases in transitional zone epithelia in prostates of men treated
with this agent compared to a placebo.84 When compared with finasteride, a 5α-
reductase inhibitor, saw palmetto use resulted in fewer side effects and increased
urine flow.84 However, a study of patients with prostatitis/chronic pelvic pain
syndrome that evaluated the safety and efficacy of saw palmetto compared to
finasteride reported that at the end of the investigation, more patients opted to
continue finasteride treatment rather than the saw palmetto treatment. The
researchers found that in patients with the studied condition, saw palmetto had
no appreciable long-term improvement and, with the exception of voiding,
patients on finasteride experienced significant improvement in all other analyzed
parameters.85
Adverse reactions to saw palmetto are rare, but there are reports of mild
gastrointestinal symptoms and headaches.84 Results of a recent investigation
indicated that recommended doses of saw palmetto are not likely to alter the
pharmacokinetics of co-administered medications dependent on the cytochrome
P-450 isoenzymes CYP2D6 or CYP3A4, such as dextromethorphan and alpra-
zolam.86 Further, there are few herbal–drug interactions in the literature regarding
saw palmetto, but, as always, care and responsibility should be exercised when
taking this agent.84
7.4.3 ECHINACEA
Echinacea is part of the daisy family found throughout North America. There are
nine species of echinacea in total and the medicinal preparations are derived from
three of these: Echinacea purpurea (purple coneflower), Echinacea pallida (pale
purple coneflower), and Echinacea angustifolia (narrow-leaved coneflower).96,98,99
Echinacea is recommended as a prophylactic and treatment substance for upper
respiratory infections. However, data are insufficient at present to support the
former.84 It has alkylamide and polysaccharide substances that possess significant
DK5836_C007.fm Page 218 Monday, May 22, 2006 1:57 PM
TABLE 7.1
Herbal Agents, Potential Side Effects, and Anesthesia Considerations
Herbal
Agents Potential Side Effects Anesthesia Considerations
Note: MAOIs = monoamine oxidase inhibitors, SSRIs = selective serotonin reuptake inhibitors
Source: Modified from Kaye AD, Clarke RC, Sabar R, et al. Herbal medicines: current trends in
anesthesiology practice — a hospital survey. J Clin Anest 12: 468–471, 2000.
7.4.4 FEVERFEW
Feverfew is used to treat headache, fever, menstrual abnormalities, and prevent
migraines.111 The name is derived from the Latin word febrifugia, which means
“fever reducer.”112 Although feverfew is commonly used for migraine headaches,
the literature is inconclusive regarding its efficacy.113,114 In a study reviewing
evidence from double-blind, randomized controlled trials of the clinical efficacy
of feverfew vs. placebo for migraine prophylaxis, investigators found insufficient
evidence to suggest a benefit of feverfew over placebo for the prevention of
migraines.115 As with most herbal compounds, analyses of feverfew-based prod-
ucts have yielded significant variations in the parthenolide contents, which are
believed to be the active ingredients.116
Regarding the effects of the antiinflammatory lactone parthenolide, a German
study indicated that parthenolide may support T-cell survival by downregulating
the CD95 system. The CD95 system is a critical component of the apoptotic, or
programmed cell death pathway of activated T-cells. Further, the authors reported
that pathenolide can have therapeutic potential as an antiapoptotic substance
blocking the activation-induced cell death of T-cells.117
Feverfew also has demonstrated inhibition of serotonin release from aggre-
gating platelets. This mechanism may be related to the inhibition of arachidonic
acid release via a phosholipase pathway.118–120 It has also been found that feverfew
has decreased approximately 86 to 88% of prostaglandin production without
exhibiting inhibition of the cyclooxygenase enzyme.121
Adverse reactions to feverfew include apthous ulcers, abdominal pain, nausea,
and vomiting. A rebound headache may occur with abrupt cessation of this
herbal.111–112 Better tolerance to feverfew has been suggested when compared to
conventional migraine medications because in studies feverfew use resulted in
no alteration in heart rate, blood pressure, body weight, or blood chemistry like
conventional migraine medications.111 A condition known as “post-feverfew syn-
drome” can occur in long-term users, which manifests as fatigue, anxiety, head-
aches, insomnia, arthralgias, and muscle and joint stiffness.111,122
Feverfew may inhibit platelet action; therefore, it is reasonable to avoid the
concomittant use of this herb in patients taking medications, such as heparin,
warfarin, NSAIDs, aspirin, and vitamin E.123,124 Furthermore, herbs, like feverfew,
can interact with iron preparations, thereby reducing the bioavailability of that
substance.106
7.4.5 EPHEDRA
Since the U.S. government’s ban on ephedra-based products, there has been an
obvious decline in its prevalent use in this country. However, patients may still
present for perioperative assessment with a history of use of ephedra. Ma huang,
an ephedra-based alkaloid, is similar in structure to amphetamines and is tradi-
tionally indicated for the treatment of various respiratory disorders, such as the
flu, the common cold, allergies, and bronchitis. Additionally, it is commonly used
DK5836_C007.fm Page 221 Monday, May 22, 2006 1:57 PM
7.4.6 GINGER
Ginger has been used for the treatment of nausea, vomiting, motion sickness, and
vertigo.87 A study of the effects of ginger on subjects with vertigo found that no
subjects experienced nausea after caloric stimulation of the vestibular system, in
contrast to those treated with a placebo.131 It is postulated that ginger may
be superior to dimenhydrinate in decreasing motion sickness.132 For vomiting
DK5836_C007.fm Page 222 Monday, May 22, 2006 1:57 PM
episodes, this herbal has also been effective in decreasing symptoms associated
with hyperemesis gravidarum.133
The effect of ginger on the clotting pathway has also been investigated. Ginger
has exhibited potent inhibition of thromboxane synthetase and this results in an
increased bleeding time.134 The ability of ginger constituents and related sub-
stances to inhibit arachidonic acid-induced platelet activation in human whole
blood has also been investigated. The data from that study revealed ginger com-
pounds and derivatives are more potent antiplatelet agents than aspirin under
conditions employed in the study. [8]-Paradol, a constituent of ginger, was iden-
tified the most potent antiplatelet aggregation agent and cyclooxygenase 1 inhib-
itor.135 In another study, administration of ginger has also resulted in decreases
in blood pressure, serum cholesterol, and serum triglycerides in diabetic rats.136
Thus, further investigation into these effects in this disease is warranted.
Adverse effects of ginger include bleeding dysfunction and its use is con-
traindicated in patients with coagulation abnormalities or those on anticoagulant
medications such as NSAIDs, aspirin, warfarin, and heparin.87 Ginger may
increase bleeding risk, enhance barbiturate effects, and, as a result of an inotropic
effect, interfere with cardiac medications. Large quantities of ginger may also
cause cardiac arrhythmias and central nervous system depression.137
7.4.7 GARLIC
Garlic’s use is prevalent and is available in powdered, dried, and fresh forms.84
Allicin, the main active ingredient in garlic, contains sulfur, and crushing the
clove activates the enzyme allinase, thus facilitating the conversion of alliin to
allicin.96
Recommended uses for garlic have focused on treating hypercholesterolemia,
hypertension, and cardiovascular disease and studies have targeted its hypo-
cholesterolemic and vasodilatory activity.84,138–142 Investigations have found that
garlic may lead to inhibition of the HMG-CoA reductase and 14α-demethylase
enzyme systems, thereby exerting a lipid-reducing effect.84 Garlic may also be
used for its antiplatelet, antioxidant, and fibrinolytic actions.140,143,144 There is
minimal data present to support the use of garlic for hypertension, as its depressor
effects on systolic and diastolic blood pressure appear to range from minimal to
modest.84,96
Chronic oral use of garlic has been reported to augment the endogenous
antioxidants of the heart.146 A recent study hypothesized that garlic-induced
cardiac antioxidants may provide protection against acute adriamycin-induced
cardiotoxicity. Using the rat model, researchers discovered an increase in oxida-
tive stress as evidenced by a significant increase in myocardial thiobarbituric acid
reactive substances (TBARS) and a decrease in myocardial superoxide dismutase
(SOD), catalase, and glutathione peroxidase activity in the adriamycin group.
However, in the garlic treated rats, the increase in myocardial TBARS and a
decrease in endogenous antioxidants by adriamycin was significantly attenuated.
Therefore, one may conclude garlic administration may help prevent this form
DK5836_C007.fm Page 223 Monday, May 22, 2006 1:57 PM
of drug-induced cardiotoxicity.145 The effects of allicin in the feline and rat lung
vasculature have also been studied. Allicin has shown significant vasodepressor
activity in the pulmonary vascular bed of the rat and cat.146 Although allicin has
been found to lower blood pressure, insulin, and triglycerides levels in fructose-
fed rats, it has also been considered important to investigate its effect on the
weight of animals.
Recent data indicate garlic may be an effective treatment against methicillin-
resistant Staphylococcus aureus (MRSA) infection. In a study using mice, inves-
tigators demonstrated that the garlic extracts, diallyl sulphide and diallyl disul-
phide, showed protective qualities against MRSA infection. Such conclusions,
coupled with further investigation, may result in the use of such extracts in MRSA
infection treatment.147
Side effects of garlic are minimal, with odor and gastrointestinal discomfort
being the most commonly reported.84 Induction of the cytochrome P-450 system
may occur as evidenced by reduction of serum levels of one medication.84 Anes-
thesiologists must be aware that garlic may augment the effects of warfarin,
heparin, aspirin, and may result in an abnormal bleeding time. This effect can
result in increased risk of peri-operative hemorrhage.148 Thus, monitoring of
cardiovascular function in this period is crucial.
side effects of kava kava use include visual changes, a pellagra-like syndrome with
characteristic ichthyosiform dermopathy, and hallucinations.165,169,170
Regarding drug interactions, kava kava may react adversely with the benzo-
diazepine alprazolam, other central nervous system depressants, statins, alcohol,
and levodopa, consequently resulting in excessive sedation, among other side
effects; therefore, the supplement should be avoided in those patients with endog-
enous depression.165,171–173 Finally, kava kava may also affect platelets in an
antithrombotic fashion by inhibiting cyclooxygenase and, thus, attenuating throm-
boxane production.165 Pain relief mechanisms utilized by the herbal may be
similar to local anesthetic responses and could be dependent on a nonopiate
sensitive pathway.174,175
7.4.10 GINSENG
There are three main groups of ginseng that are classified based on their geo-
graphic origin.84 These are Asian ginseng, American ginseng, and Siberian gin-
seng, with the pharmacologically active ingredient in ginseng being ginsenos-
ides.84,89,112 Asian and American ginsengs have been used to increase resistance
to environmental stress, promote diuresis, stimulate the immune system, and aid
digestion.176,177 Further, while Asian ginseng has shown promise in improving
cognition when combined with the herbal agent gingko, American ginseng has
been studied for its potential to stimulate human tumor necrosis factor alpha
(TNF-alpha) production in cultured human white blood cells.177,178 American
ginseng may also possess hypoglycemic activity.179,180 Such effects have been
observed in both normal and diabetic subjects and may be attributed to ginseng’s
components, specifically ginsenoside Rb2 and panaxans I, J, K, and L.181–185
Typically ginseng is well tolerated, but side effects, such as bleeding abnor-
malities secondary to antiplatelet effects, headache, vomiting, Stevens-Johnson
syndrome, epistaxis, and hypertension have been reported.186–192 Drug interactions
between Asian ginseng and calcium channel blockers, warfarin, phenelzine, and
digoxin have also been noted.84 It may be advisable for patients on anticoagulant
medications, such as warfarin, heparin, aspirin, and NSAIDs, to avoid ginseng.
Further, because of ginseng’s association with hypertension and the deleterious
outcomes linked to chronic hypertension, the anesthesiologist should be aware
of whom and for how long patients may have been taking this herbal product.
Since many anesthetic agents can cause generalized vasodilation, hemodynamic
lability may be seen.
Regarding ginseng’s interaction with antidepressants, such as monoamine
oxidase inhibitors, concurrent use of ginseng with phenelzine sulphate should be
avoided as manic episodes have been reported with routine use of both.193,194
Finally, as a result of ginseng’s potential to cause decreased blood glucose levels,
it should be used cautiously in diabetic patients on insulin or other oral hypogly-
cemic agents, and blood glucose levels should be monitored.
DK5836_C007.fm Page 226 Monday, May 22, 2006 1:57 PM
7.5 CONCLUSION
The growing use of supplements, such as minerals, vitamins, and herbals, in the
world warrants a more comprehensive understanding of these agents by the
medical community. It is important for the anesthesiologist to recognize certain
facts regarding these supplements. For example, there are about 1300 g of calcium
in a 70 kg adult and the mineral magnesium activates approximately 300 enzyme
systems in the human body; most of these systems are involved in energy metab-
olism.195 Aside from these, the anesthesiologist must regulate the patient’s phys-
iological functions and appreciate the effect of these supplements on such func-
tions during various operative procedures. As demonstrated in this chapter, the
use of these compounds may prove beneficial for some patients, but result in
alterations in normal physiologic functions in others, thus potentially resulting
in deleterious consequences. These agents, in addition to all other medications
taken by the patient, should be screened by medical practitioners, in particular
anesthesiologists, as some of these compounds may interact with chosen anes-
thetics during the stages of anesthesia. Furthermore, education of patients regard-
ing the serious potential drug-supplement interactions should be an integral com-
ponent of the preoperative assessment. Specifically, the American Society of
Anesthesiologists (ASA) recommends that all herbal medications be discontinued
2 to 3 weeks prior to elective surgery.
Due to current lax regulations in some countries, some of these agents are
poorly categorized and standardized, thus resulting in a high risk of adverse effects
when used by an uninformed or misinformed public. Within the past few decades,
hundreds of deaths have been linked to the use of these agents, specifically the
herbals. Data also suggest that less than 1% of adverse effects associated with
herbals are reported. In general, whether the patient is taking minerals, vitamins,
and/or herbals, one thing is for certain, an open line of communication between
medical practitioner and patient should exist regarding all of these agents. This
communication is essential to ensure quality patient treatment, a stable and secure
rapport, and a properly informed and educated general public.
REFERENCES
1. McCarron DA, Hatton D. Dietary calcium and lower blood pressure: we can all
benefit. JAMA 1996; 275: 1128–1129.
2. Thys-Jacobs S, Starkey P, Bernstein D, et al. Calcium carbonate and the premen-
strual syndrome: effects on premenstrual and menstrual symptoms. Premenstrual
Syndrome Study Group. Am J Obstet Gynecol 1998; 179: 444–452.
3. Bar-Or D, Gasiel Y. Calcium and calciferol antagonize effect of verapamil in atrial
fibrillatin. Br Med J (Clin Res Ed) 1981; 282: 1585–1586.
4. Durward A. Guerguerian AM. Lefebvre M. Shemie SD. Massive diltiazem over-
dose treated with extracorporeal membrane oxygenation. [Case Reports. Journal
Article] Ped Crit Care Med 2003; 4(3): 372–376.
DK5836_C007.fm Page 227 Monday, May 22, 2006 1:57 PM
26. Davolos A, Castillo J, Marrugat J, et al. Body iron stores and early neurologic
deterioration in acute cerebral infarction. Neurology 2000; 54: 1568–1574.
27. Lao TT, Tam K, Chan LY. Third trimester iron status and pregnancy outcome in
non-anemic women; pregnancy unfavourably affected by maternal iron excess.
Hum Reprod 2000; 15: 1843–1848.
28. Siegenberg D, Baynes RD, Bothwell TH, et al. Ascorbic acid prevents the dose-
dependent inhibitory effects of polyphenols and phytates on nonheme-iron absorp-
tion. Am J Clin Nutr 1991; 53: 537–541.
29. Lehto P, Kivisto KT, Neuvonen PJ. The effect of ferrous sulphate on the absorption
of norfloxacin, ciprofloxacin and ofloxacin. Br J Clin Pharmacol 1994; 37: 82–85.
30. Campbell NR, Hasinoff BB. Iron supplements: a common cause of drug interac-
tions. Br J Clin Pharmacol 1991; 31: 251–255.
31. Heinrich HC, Oppitz KH, Gabbe EE. Inhibition of iron absorption in man by
tetracycline [German]. Klin Wochenschr. 1974; 52: 493–498.
32. Osman MA, Patel RB, Schuna A, et al. Reduction in oral penicillamine absorption
by food, antacid, and ferrous sulfate. Clin Pharmacol Ther 1983; 33: 465–470.
33. Campbell NR, Hasinoff BB, Stalts H, et al. Ferrous sulfate reduces thyroxine
efficacy in patients with hypothyroidism. Ann Intern Med 1992; 117: 1010–1013.
34. Ursini F, Heim S, Kiess M, et al. Dual function of the selenoprotein PHGPx during
sperm maturation. Science 1999; 285(5432): 1393–1396.
35. Patterson BH, Levander OA. Naturally occurring selenium compounds in cancer
chemoprevention trials: a workship summary. Cancer Epidemiol Biomarkers Prev
1997; 6: 63–69.
36. Fan AM, Kizer KW. Selenium: nutritional, toxicologic, and clinical aspects, West
J Med 1990; 153: 160–167.
37. Prasad AS, Halsted JA, Nadimi M. Syndrome of iron deficiency anemia,
hepatosplenomegaly, hypogonadism, dwarfism, and geophagia. Am J Med 1961;
31: 532–546.
38. Sandstead HH. Is zinc deficiency a public health problem? Nutrition 1995; 11:
87–92.
39. Goldenberg RL, Tamura T, Neggers Y, et al. The effect of zinc supplementation
on pregnancy outcome. JAMA 1995; 274: 463–468.
40. Ma J, Betts NM. Zinc and copper intakes and their major food sources for older
adults in the 1994-96 continuing survey of food intakes by individuals (CSFII).
J Nutr 2000; 130: 2838–2843.
41. Prasad AS. Zinc deficiency in women, infants, and children. J Am Coll Nutr 1996;
15: 113–120.
42. Bratman S, Girman AM. Handbook of Herbs and Supplements and their Thera-
peutic Uses. St. Louis: Mosby, Inc., 2003.
43. Mikszewski JS, Saunders HM, Hess RS. Zinc-associated acute pancreatitis in a
dog. J Small Anim Pract 2003; 44(4): 177–180.
44. Combs GF. The Vitamins: Fundamental Aspects in Nutrition and Health. 2nd ed.
San Diego: Academic Press, 1998: 5–6.
45. Queiroz E, Ramalho A, Saunders C, et al. Vitamin A status in diabetic children.
Diabetes Nutr Metab. 2000; 13(5): 298–299.
46. Higdon, J. An Evidence-Based Approach to Vitamins and Minerals. New York:
Thieme Medical Publishers, Inc., 2003: 148–-156.
47. Brzezinska-Wcislo L, Pierzchala E, Kaminska-Budzinska G, et al. [The use of
retinoids in dermatology] [Polish]. Wiad Lek 2004; 57(1-2): 63–69.
DK5836_C007.fm Page 229 Monday, May 22, 2006 1:57 PM
48. Harris JE. Interaction of dietary factors with oral anticoagulants: review and
applications. J Am Diet Assoc 1995; 95: 580–584.
49. Binkley N, Krueger D. Hypervitaminosis A and bone. Nutr Rev 2000; 58(5):
138–144.
50. Nau H, Tzimas G, Mondry M, et al. Antiepileptic drugs alter endogenous retinoid
concentrations: a possible mechanism of teratogenesis of anticonvulsant therapy.
Life Sci 1995; 57: 53–60.
51. Leo MA, Lieber CS. Alcohol, vitamin A, and beta-carotene: adverse interactins,
including hepatotoxicity and carcinogenicity. Am J Clin Nutr 1999; 69(6):
1071–1085.
52. Baik HW, Russel RM. Vitamin B12 deficiency in the elderly. Annu Rev Nutr 1999;
19: 357–377.
53. Loikili NG, Noel E. Blaison G, et al. Update of pernicious anemia. A retrospective
study of 49 cases. [French] Rev Med Interne 2004; 25(8): 556–561.
54. Weimann J. Toxicity of nitrous oxide. Best Pract Res Clin Anaesthesiol 2003;
17(1): 47–61.
55. Webb DI, Chodos RB, Mahar CQ, et al. Mechanism of vitamin B12 malabsorption
in patients receiving colchicines. N Engl J Med 1968; 279: 845–850.
56. Adams JF, Clark JS, Ireland JT, et al. Malabsorption of vitamin B12 and intrinsic
factor secretion during biguanide therapy. Diabetologia 1983; 24: 16–18.
57. Flippo TS, Holder WD Jr. Neurologic degeneration associated with nitrous oxide
anesthesia in patients with vitamin B12 deficiency. Arch Surg 1993; 128:
1391–1395.
58. Baum MK, Javier JJ, Mantero-Atienza E, et al. Zidovudine-associated adverse
reactions in a longitudinal study of asymptomatic HIV-1-infected homosexual
males. J Acquir Immune Defic Syndr 1991; 4: 1218–1226.
59. Marcuard SP, Albernaz L, Khazanie PG. Omeprazole therapy causes malabsorp-
tion of cyanocobalamin (vitamin B12). Ann Intern Med 1994; 120: 211–215.
60. Streeter AM, Goulston KJ, Bathur FA, et al. Cimetidine and malabsorption of
cobalamin. Dig Dis Sci 1982; 27: 13–16.
61. Aymard JP, Aymard B, Netter P, et al. Haematological adverse effects of histamine
H2-receptor antagonists. Med Toxicol Adverse Drug Exp 1988; 3: 430–448.
62. Sauberlich HE. A history of scurvy and vitamin C. In Packer L, Fuchs J, Ed.
Vitamin C in Health and Disease. New York: Marcel Decker, 1997: 1–24.
63. Hemila H. Vitamin C intake and susceptibility to the common cold. Br J Nutr
1997; 77(1): 59–72.
64. Johnston CS, Martin LJ, Cai X. Antihistamine effect of supplemental ascorbic
acid and neutrophil chemotaxis. J Am Coll Nutr 1992; 11(2): 172–176.
65. Rosenthal G. Interaction of ascorbic acid and warfarin [letter]. JAMA 1971; 215:
1671.
66. Harris JE. Interaction of dietary factors with oral anticoagulants: review and
applications. J Am Diet Assoc 1995; 95: 580–584.
67. Mak S, Newton GE. Vitamin C augments the inotropic response to dobutamine
in humans with normal left ventricular function. Circulation 2001; 103: 826–830.
68. Houston JB, Levy G. Drug biotransformation interactions in man. VI. Acetami-
nophen and ascorbic acid. J Pharm Sci 1976; 65 :1218–1221.
69. Molloy TP, Wilson CW. Protein-binding of ascorbic acid. 2. Interaction with
acetyl-salicylic acid. Int J Vitam Nutr Res 1980; 50: 387–392.
DK5836_C007.fm Page 230 Monday, May 22, 2006 1:57 PM
70. Rivers JM, Devine MM. Plasma ascorbic acid concentrations and oral contracep-
tives. Am J Clin Nutr 1972; 25: 684–689.
71. Utiger R. The need for more vitamin D. N Engl J Med 1998; 338: 828–829.
72. Semba RD, Garrett E, Johnson BA, et al. Vitamin D deficiency among older
women with and without disability. Am J Clin Nutr 2000; 72: 1529–1534.
73. Mezquita-Raya P, Munoz-Torres M, De Dios Luna J, et al. Relation between
vitamin D insufficiency, bone density, and bone metabolism in healthy postmeno-
pausal women. J Bone Miner Res 2001; 16: 1408–1415.
74. Malabanan AO, Holick MF. Vitamin D and bone health in postmenopausal women.
J Womens Health (Larchmt) 2003; 12(2): 151–156.
75. Food and Nutrition Board, Institute of Medicine. Vitamin D. Dietary Reference
Intakes: Calcium, Phosphorus, Magnesium, Vitamin D, and Fluoride. Washington,
D.C.: National Academy Press, 1997: 250–287.
76. Vitamins. Drug Facts and Comparisons. St. Louis: Facts and Comparisons, 6–33.
77. Ford ES, Sowell A. Serum alpha-tocopherol status in the United States population:
findings from the Third National Health and Nutrition Examination Survey. Am
J Epidemiol 1999; 150: 290–300.
78. Liede KE, Haukka JK, Saxen LM, et al. Increased tendency towards gingival
bleeding caused by joint effect of alpha-tocopherol supplementation and acetyl-
salicylic acid. Ann Med 1998; 30: 542–546.
79. Paolisso G, D’Amore A, Galzerano D, et al. Daily vitamin E supplements improve
metabolic control but not insulin secretion in elderly type II diabetic patients.
Diabetes Care 1993; 16: 1433–1437.
80. Paolisso G, D’Amore A, Giugliano D, et al. Pharmacologic doses of vitamin E
improve insulin action in healthy subjects and non-insulin-dependent diabetic
patients. Am J Clin Nutr 1993; 57: 650–656.
81. Food and Nutrition Board, Institute of Medicine. Folic acid. Dietary Reference
Intakes: Thiamin, Riboflavin, Niacin, Vitamin B-6, Vitamin B-12, Pantothenic Acid,
Biotin, and Choline. Washington, D.C.: National Academy Press, 1998: 193–305.
82. Cembrowski GS, Zhang MM, Prosser CI, et al. Folate is not what it is cracked
up to be. Arch Intern Med 1999; 159: 2747–2748.
83. Lewis DP, Van Dyke DC, Willhite LA, et al. Phenytoin-folic acid interaction. Ann
Pharmacother 1995; 29: 726–735.
84. Hughes EF, Jacobs BP, Berman BM. Complementary and alternative medicine.
In Tierney Jr. LM, McPhee SJ, Papadakis MA, Eds. Current Medical Diagnosis
and Treatment. New York: Lange Medical Books/McGraw-Hill, 2004: 1681–1703.
85. Kaplan SA, Volpe MA, Te AE. Prospective, 1-year trial using saw palmetto versus
finasteride in the treatment of category III prostatitis/chronic pelvic pain syndrome.
J Urol 2004; 171: 284–288.
86. Markowitz JS, Donovan JL, Devane CL, et al. Multiple doses of saw palmetto
(serenoa repens) did not alter cytochrome P450 2D6 and 3A4 activity in normal
volunteers. Clin Pharmacol Ther 2003; 74: 536–542.
87. Kaye AD, Sabar R, Vig S, et al. Neutraceuticals-current concepts and the role of
the anesthesiologist. Am J Anesthesiol 2000; 27: 405–407.
88. Jellin JM, Gregory PJ, Batz F, et al, Eds. St. John’s Wort. Natural Medicines:
Comprehensive Database. 4th ed. Stockton, CA: Therapeutic Research Faculty,
2002; 1180–1184.
DK5836_C007.fm Page 231 Monday, May 22, 2006 1:57 PM
107. Eaton EA, Walle UK, Lewis AJ, et al. Flavinoids, potent inhibitors of the human
form of phenolsulfotransferase: potential role in drug metabolism and chemopre-
vention. Drug Met Disp 1996; 24: 232–237.
108. Schubert W, Eriksson U, Edgar B, et al. Flavinoids in grapefruit juice inhibit the
in vitro hepatic metabolism of 17 beta-estradiol. Eur J Drug Metab Pharmacokinet
1995; 20: 219–224.
109. Gorski JC, Huang SM, Pinto A, et al. The effect of echinacea (Echinacea purpurea
root) on cytochrome P450 activity in vivo. Clin Pharmacol Ther 2004; 75: 89–100.
110. Chavez ML, Chavez PI. Echinacea. Hosp Pharm 1998; 33: 180–188.
111. Jellin JM, Gregory PJ, Batz F, et al, Eds. Feverfew. Natural Medicines: Compre-
hensive Database, 5th ed. Stockton: Therapeutic Research Faculty, 2003:
541–543.
112. Kaye AD, Sabar R, Vig S, et al. Neutraceuticals-current concepts and the role of
the anesthesiologist. Am J Anesthesiol 2000; 27: 467–471.
113. Murphy J, Heptinstall S, Mitchell JR, et al. Randomized double-blind, placebo-
controlled trial of feverfew in migrane prevention. Lancet 1988; 2: 189–192.
114. De Weerdt C, Bootsma H, Hendricks H. Herbal medicines in migrane prevention:
randomized double-blind, placebo-controlled crossover trial of a feverfew prepa-
ration. Physomed 1996; 3: 225–230.
115. Pittler M, Ernst E. Feverfew for preventing migraine. Cochrane Database Syst
Rev 1: CD002286, 2004.
116. Nelson MH, Cobb SE, Shelton J. Variations in parthenolide content and daily dose
of feverfew products. Am J Health Syst Pharm 2002; 59: 1527–1531.
117. Li-Weber M, Giaisi M, Baumann S, et al. The anti-inflammatory sesquiterpene
lactone parthenolide suppresses CD95-mediated activation-induced-cell-death in
T-cells. Cell Death Differ 2002; 9: 1256–1265.
118. Marles RJ, Kaminski J, Arnason JT, et al. A bioassay of inhibition of serotonin
release from bovine platelets. J Nat Prod 1992; 55: 1044–1056.
119. Fozard JR. 5-Hydroxytryptamine in the pathophysiology of migrane. In Bevan
JA, Ed. Vascular Neuroeffector Mechanisms. Amsterdam: Elsevier, 1985:
321–328.
120. Makheja AN, Bailey JM. A platelet phospholipase inhibitor from the medicinal
herb feverfew (Tanacatum parthenium). Proostaglandins Leukot Med 1982; 8:
653–660.
121. Collier HO, Butt NM, McDonald-Gibson WJ, et al. Extract of feverfew inhibits
prostaglandins biosynthesis. Lancet 1980; 2: 922–923.
122. Baldwin CA, Anderson LA, Phillipson JD, et al. What pharmacists should know
about feverfew. J Pharm Pharmacol 1987; 239: 237–238.
123. Heptinstall S, Groenwegen WA, Spangenberg P, et al. Extracts of feverfew may
inhibit platelet behavior neutralization of sulphydryl groups. J Pharm Pharmacol
1987; 39: 459–465.
124. Makheja AN, Bailey J. The active principle in feverfew [letter]. Lancet 1981; 2:
1054.
125. Tinkleman DG, Avner SE. Ephedrine therapy in asthmatic children. Clinical
tolerance and absence of side effects. JAMA 1977; 237: 553–557.
126. Fields AM, Kaye AD, Richards TA, et al. Pulmonary vascular responses to ma
huang extract. J Altern Complement Med 2003; 9: 727–733.
DK5836_C007.fm Page 233 Monday, May 22, 2006 1:57 PM
127. Gurley BJ, Gardner SF, White LM, et al. Ephedrine pharmacokinetics after inges-
tion of nutritional supplements containing ephedra sinica (ma huang). Ther Drug
Monit 1998; 20: 439–445.
128. Centers for Disease Control and Prevention (CDC). Adverse events associated
with ephedrine-containing products — Texas, December 1993–September 1995.
MMWR Morb Mortal Wkly Rep 1996, August 16; 45: 689–693.
129. Jurgensen K, Stevens C, Eds. Finally, a Ban on Ephedra. USA Today Newspaper,
p. 22A, April 13, 2004.
130. Gruenwald J, Brendler T, Jaenicke C, et al. PDR for Herbal Medicines, Ed. 1.
Montvale: Medical Economics Company, 1998: 826-827.
131. Grontved A, Hentzer E. Vertigo-reducing effect of ginger root. J Otolaryngol
1986; 48: 282–286.
132. Holtmann S, Clarke AH, Scherer H, et al. The anti-motion sickness mechanism
of ginger. Acta Otolaryngol (Stockh) 1989; 108: 168–174.
133. Fischer-Rasmussen W, Kjaer SK, Dahl C, et al. Ginger treatment of hyperemesis
gravidarum. Eur J Obstet Gyn Rep Biol 1990; 38 :19–24.
134. Backon J. Ginger: inhibition of thromboxane synthetase and stimulation of pros-
tacyclin: relevance for medicine and psychiatry. Med Hypoth 1986; 20: 271–278.
135. Nurtjahja-Tjendraputra E, Ammit AJ, Roufogalis BD, et al. Effective anti-platelet
and COX-1 enzyme inhibitors from pungent constituents of ginger. Thromb Res
2003; 111: 259–265.
136. Akhani SP, Vishwakarma SL, Goyal RK. Anti-diabetic activity of Zingiber offici-
nale in streptozotocin-induced type I diabetic rats. J Pharm Pharmacol 2004; 56:
101–105.
137. Jellin JM, Batz F, Hitchens K, et al, Eds. Ginger. Natural Medicines: Compre-
hensive Database, 2nd ed. Stockton: Therapeutic Research Faculty, 1999:
416–418.
138. Jain AK, Vargas R, Gotzowsky S, et al. Can garlic reduce levels of serum lipids?
A controlled clinical study. Am J Med 1993; 94: 632–635.
139. Silagy CA, Neil HAW. A meta-analysis of the effect of garlic on blood pressure.
J Hyperten 1994; 12: 463–468.
140. Neil HAW, Silagy CA, Lancaster T, et al. Garlic powder in the treatment of
moderate hyperlipidemia: a controlled trial and meta-analysis. J R Coll Physician
1996; 30: 329–334.
141. Berthold HK, Sudhop T, von Bergmann K. Effect of a garlic oil preparation on
serum lipoproteins and cholesterol metabolism: a randomized controlled trial.
JAMA 1998; 279: 1900–1902.
142. Cooperative group for essential oil of garlic: the effect of essential oil of garlic
on hyperlipidemia and platelet aggregation: an analysis of 308 cases. J Trad Chin
Med 1986; 6: 117–120.
143. Reuter HD. Allium sativum and allium ursinum, part 2: pharmacology and medic-
inal applications. Phytomedicine 1995; 2: 73–91.
144. Beaglehole R. Garlic for flavor, not cardioprotection. Lancet 1996; 348:
1186–1187.
145. Mukherjee S, Banerjee SK, Maulik M, et al. Protection against acute adriamycin-
induced cardiotoxicity by garlic: role of endogenous antioxidants and inhibition
of TNF-alpha expression. BMC Pharmacol. 2003; 3: 16.
DK5836_C007.fm Page 234 Monday, May 22, 2006 1:57 PM
146. Kaye AD, Nossaman BD, Ibrahim IN, et al. Analysis of responses of allicin, a
compound from garlic, in the pulmonary vascular bed of the cat and in the rat.
Eur J Pharmacol 1995; 276: 21–26.
147. Tsao SM, Hsu CC, Yin MC. Garlic extract and two diallyl sulphides inhibit
methicillin-resistant Staphylococcus aureus infection in BALB/cA mice. J Anti-
microb Chemother 2003; 52: 974–980.
148. Bordia A. Effect of garlic on human platelet aggregation in vitro. Atherosclerosis
1978; 30: 355–360.
149. Bauer U. Six-month double-blind randomized clinical trial of gingko biloba extract
versus placebo in two parallel groups in patients suffering from peripheral arterial
insufficiency. Arz-neimittelforschung 1984; 34: 716–720.
150. Peters H, Kieser M, Holscher U. Demonstration of the efficacy of Gingko biloba
special extract EGB 761 on intermittent claudication — a placebo controlled,
double-blind multicenter trial. Vasa 1998; 27: 106–110.
151. Braquet P. BN 52021 and related compounds: a new series of highly specific PAF-
acether receptor antagonists isolated from gingko biloba. Blood Vessels 1985; 16:
559–572.
152. Braquet P, Bourgain RH. Anti-anaphylactic properties of BN 52021: a potent
platelet activating factor antagonist. Adv Exp Med Biol 1987; 215: 215–233.
153. Marcocci L. The nitric oxide scavenging properties of gingko biloba extract
Egb761: inhibitory effect on nitric oxide production in the macrophage cell line
RAW 264.7. Biochem Pharmacol 1997; 53: 897–903.
154. Kobuchi H, Ldroy-Lefaix MT, Christen Y, et al. Gingko biloba extract (Egb 761):
inhibitory effect on nitric oxide production in macrophage cell line RAW 264.7.
Biochem Pharmacol 1997; 53: 897–903.
155. LeBars PL, Katz MM, Berman N, et al. A placebo controlled, double-blind,
randomized trial of an extract of gingko biloba for dementia. JAMA 1997; 278:
1327–1332.
156. Zeybek N, Gorgulu S, Yagci G, et al. The effects of gingko biloba extract (EGb
761) on experimental acute pancreatitis. J Surg Res 2003; 115: 286–293.
157. Rosenblatt M, Mindel J. Spontaneous hyphema associated with ingestion of
gingko biloba extract [letter]. NEJM 1997; 336: 1108.
158. Rowin J, Lewis SL. Spontaneous bilateral subdural hematomas associated with
chronic gingko biloba ingestion have also occurred. Neurology 1996; 46:
1775–1776.
159. Gilbert GJ. Gingko biloba [commentary]. Neurology 1997; 48: 1137.
160. Vale S. Subarachanoid hemorrhage associated with Gingko biloba [letter]. Lancet
1998; 352: 36.
161. Mauro VF, Mauro LS, Kleshinski JF, et al. Impact of ginkgo biloba on the
pharmacokinetics of digoxin. Am J Ther 2003; 10: 247–251.
162. Nowakowska E, Ostrowicz A, Chodera A. Kava-kava preparations-alternative
anxiolytics. Pol Merkuriusz Lek 1998; 4: 179–180a.
163. Skidmore-Roth L. Kava. Mosby’s Handbook of Herbs and Natural Supplements.
St. Louis: Mosby-Harcourt Health Sciences, 2001: 486–490.
164. Uebelhack R, Franke L, Schewe HJ. Inhibition of platelet MAO-B by kava pyrone-
enriched extract from Piper methysticum Forster (kava-kava). Pharmacopsychia-
try 1998; 31: 187–192.
165. Jellin JM, Gregory PJ, Batz F, et al, Eds. Kava. Natural Medicines: Comprehensive
Database, 4th ed. Stockton: Therapeutic Research Faculty, 2002: 759–761.
DK5836_C007.fm Page 235 Monday, May 22, 2006 1:57 PM
166. Volz HP, Kieser M. Kava-kava extract WS 1490 versus placebo in anxiety disor-
ders a randomized placebo-controlled 25-week outpatient trial. Pharmacopsychi-
atry 1997; 30: 1–5.
167. Forget L, Goldrosen J, Hart JA, et al, Eds. Herbal Companion to AHFS DI.
Bethesda, MD: American Society of Health-System Pharmacists, Inc., 2000.
168. Seitz U, Schule A, Gleitz J. [3H]-monoamine uptake inhibition properties of kava
pyrones. Planta Med 1997; 63: 548–549.
169. Winslow LC, Kroll DJ. Herbs as medicines. Arch Intern Med 1998; 158:
2192–2199.
170. Garner LF, Klinger JD. Some visual effects caused by the beverage kava. J
Ethnopharm 1985; 13: 307–311.
171. Jellin JM, Gregory PJ, Batz F, et al, Eds. Kava. Natural Medicines: Comprehensive
Database, 5th ed. Stockton: Therapeutic Research Faculty, 2003: 788–791.
172. Jamieson DD, Duffield PH. Positive interaction of ethanol and kava resin in mice.
Clin Exp Pharm Physiol 1990; 17: 509–514.
173. Gruenwald J, Brendler T, Jaenicke C, et al. PDR for Herbal Medicines, 1st ed.
Montvale: Medical Economics Company, 1998: 1043–1045.
174. Jamieson DD, Duffield PH. The antinociceptive actions of kava components in
mice. Clin Exp Pharm Physio 1990; 17: 495–507.
175. Singh YN. Effects of kava on neuromuscular transmission and muscular contrac-
tility. J Ethnopharm 1983; 7: 267–276.
176. Ng TB, Li WW, Yeung HW. Effects of ginsenosides, lectins and Momordica
charantia insulin-like peptide on corticosterone production by isolated rat adrenal
cells. J Ethnopharm 1987; 21: 21–29.
177. Jellin JM, Gregory PJ, Batz F, et al, Eds. Ginseng, American, Ginseng, Panax.
Natural Medicines: Comprehensive Database, 5th ed. Stockton: Therapeutic
Research Faculty, 2003: 614–619.
178. Zhou DL, Kitts DD. Peripheral blood mononuclear cell production of TNF-alpha
in response to North American ginseng stimulation. Can J Physiol Pharmacol
2002; 80: 1030–1033.
179. Jie YH, Cammisuli S, Baggiolini M, et al. Immunomodulatory effects of panax
ginseng: CA Meyer in the mouse. Agents Actions Suppl 1984; 15: 386–391.
180. Sotaniemi EA, Haapakkoski E, Rautio A, et al. Ginseng therapy in non-insulin
dependent diabetic patients. Diabetes Care 1995; 18: 1373–1375.
181. Yokozawa T, Kobayashi T, Oura H, et al. Studies on the mechanism of hypogly-
cemic activity of ginsenoside-Rb2 in streptozotocin-diabetic rats. Chem Pharm
Bull 1985; 33: 869–872.
182. Oshima Y, Kkonno C, Hikono H. Isolation and hypoglycemic activity of panaxans
I, J, K and L, glycans of panax ginseng roots. J Ethnopharm 1985; 14: 255–259.
183. Konno C, Murakami M, Oshima Y, et al. Isolation and hypoglycemic activity of
panaxans Q, R, S, T and U, glycans of panax ginseng roots. J Ethnopharm 1985;
14: 69–74.
184. Konno C, Sugiyama K, Oshima Y, et al. Isolation and hypoglycemic activity of
panaxans A, B, C, D and E glycans of panax ginseng roots. Planta Med 1984;
50: 436–438.
185. Tokmoda M, Shimada K, Konno M, et al. Partial structure of panax A: a hypogly-
cemic glycan of panax ginseng roots. Planta Med 1984; 50: 436–438.
186. Baldwin CA. What pharmacists should know about ginseng. Pharm J 1986; 237:
583–586.
DK5836_C007.fm Page 236 Monday, May 22, 2006 1:57 PM
187. Hammond TG, Whitworth JA. Adverse reactions to ginseng [letter]. Med J Aust
1981; 1: 492.
188. Dega H, Laporte J, Frances C, et al. Ginseng a cause of Steven-Johnson syndrome?
[letter] Lancet 1996; 347: 1344.
189. Greenspan EM. Ginseng and vaginal bleeding [letter]. JAMA 1983; 249: 2018.
190. Hopkins MP, Androff L, Benninghoff AS, et al. Ginseng face cream and unex-
pected vaginal bleeding. Am J Obs Gyn 1988; 159: 1121–1122.
191. Palmer BV, Montgomery AC, Monterio JC, et al. Ginseng and mastalgia. BMJ
1978; 1: 1284.
192. Kuo SC, Teng CM, Lee JG, et al. Antiplatelet components in panax ginseng.
Planta Med 1990; 56: 164–167.
193. Shader RI, Greenblatt DJ. Phenelzine and the dream machine-ramblings and
reflections [edit.] J Clin Psychopharmacol 1985; 5: 65.
194. Jones BD, Runikis AM. Interactions of ginseng with phenelzine. J Clin Psychop-
harm 1987; 7: 201–202.
195. Kaye AD, Grogono AW. Fluid and electrolyte physiology. In Miller RD, Cucchiara
RF, Miller ED Jr, et al, Eds. Anesthesia vol. 1, 5th ed. Philadelphia: Churchill
Livingstone, 2000: 1586–1612.
DK5836_C008.fm Page 237 Monday, May 22, 2006 2:04 PM
CONTENTS
8.1 Introduction..............................................................................................238
8.2 Genetic Polymorphisms and CVD..........................................................239
8.2.1 Apolipoprotein E and CVD ........................................................240
8.2.2 APOE Genotype and Saturated Fat Intake .................................240
8.2.3 APOE Genotype and Sucrose Intake ..........................................241
8.2.4 Methylenetetrahydrofolate Reductase and CVD ........................241
8.2.5 MTHFR 677 C→T Polymorphism, Folate Intake, and CVD ....242
8.2.6 Hepatic Lipase and CVD ............................................................243
8.2.7 HL-514 C→T Polymorphism and CVD.....................................244
8.2.8 Statins, Niacin, and Polymorphisms Related to the
Lowering of Cholesterol..............................................................244
8.3 Genetic Polymorphisms and Cancer .......................................................246
8.3.1 Vitamin D and the Vitamin D Receptor .....................................247
8.3.2 VDR Polymorphisms, Vitamin D Intake, and
Colorectal Cancer ........................................................................248
8.3.3 Methylenetetrahydrofolate Reductase, Folate Status,
and Cancer ...................................................................................249
8.3.4 MTHFR 677 C→T Polymorphism, Folate Intake, and
Colorectal Cancer ........................................................................250
8.3.5 MTHFR 677 C→T and 1298 A→C Polymorphisms and
Acute Leukemia...........................................................................250
237
DK5836_C008.fm Page 238 Monday, May 22, 2006 2:04 PM
8.1 INTRODUCTION
Nutrigenomics is an integrative science combining biotechnology, molecular
medicine, and pharmacogenomics, which is revolutionizing how nutrition and
diet are viewed.1 A working definition of this new branch of genomics, by Kaput
and Rodriguez, is that “it seeks to provide a genetic understanding for how dietary
chemicals can affect the balance between health and disease by altering the
expression of an individual’s genetic makeup.”2 In an attempt to clarify the term,
they have proposed the following five tenets:
structure and function when the nucleotide base substitution occurs in a gene’s
coding region.6 Alternatively, a polymorphism may have no effect on the protein
product because the base substitution occurs in a DNA region that is not involved
in gene transcription or translation; however, it could be a part of regulatory
promoters.
Molecular techniques for detecting SNPs (polymerase chain reaction, restric-
tion enzyme detection) have been used extensively in pharmaceutical, toxicolog-
ical, and clinical research disciplines, but not so much in nutritional sciences.4
The recent development of extensive genomic databases and high throughput
genetic screening made it possible for the study of the interactions of interindi-
vidual variation and nutrition. The greatest potential for benefit from those studies
will be for health maintenance by preventing or slowing the early stages of diet-
related diseases. Nutrigenomics could also provide the means to develop better
molecular biomarkers of early changes between health maintenance and disease
progression.
The most common diseases, such as cardiovascular disease (CVD) and cancer,
are known to be polygenic, meaning that the incidence of the disease is attribut-
able to polymorphisms of multiple interacting genes.6,7 These multifactorial dis-
eases involve the interaction of many genes as well as gene–environment and
gene–diet interactions. Complex interactions of multiple internal and external
factors determine the probability of developing these multifactorial diseases.
Since the interactions can make it increasingly difficult to identify and charac-
terize what combinations of genetic variants are relevant to CVD and cancer, the
study of gene–diet interactions can help to establish the strongest relationships
between gene variations and phenotypes in healthy individuals in order to relate
their specific genotype to biomarkers of disease risk.5 Defining these pathophys-
iologic mechanisms in each individual can aid in identifying individuals at risk
for disease and to apply diet and lifestyle changes to modify disease risk factors.6
This chapter focuses on certain polymorphisms where the intake of a specific
nutrient related to that polymorphism affects the pathogenesis of CVD and/or
cancer.
particles in E2 carriers, and the opposite or no effect was observed in the E4 and
E3 carriers. This suggests that carriers of the E2 variant allele may actually be
particularly susceptible to coronary disease when exposed to high saturated fat
diets. It was found that the E2 allele has a more detrimental influence on the
lipoprotein profile in individuals eating a high saturated fat diet compared to low
saturated fat, and the mechanism for this response may be due to the effect of
the E2 allele on receptor-mediated VLDL clearance. Compared to E3, E2 binds
poorly to lipoprotein receptors leading to delayed clearance and increased accu-
mulation of chylomicron and VLDL remnants.11
Deoxyuridylate
Diet Intake Folate Dihydrofolate
dUMP
Serine
Observational studies have shown that when folate status is less than adequate,
individuals with the MTHFR 677 TT genotype have lower blood folate and higher
plasma homocysteine concentrations than those with the MTHFR 677 CC gen-
otype.22–26 One of the most recent studies in this area, by Shelnutt et al.,27 recruited
only 677 CC and TT, nonpregnant healthy women, aged 20 to 30 years old and
placed them on folate deficient and folate rich diets. The folate depletion diet
provided approximately 115 µg dietary folate equivalents (DFE) per day for 7
weeks, and the folate repletion diet consisted of a combination of the depletion
diet, plus 400 µg DFE/d for 7 weeks. Serum folate concentrations did not differ
between the two genotypes at baseline. Results showed that TT individuals were
less capable of maintaining normal blood folate concentration during the deple-
tion phase than CC individuals.27 Mean plasma homocysteine concentration was
DK5836_C008.fm Page 243 Monday, May 22, 2006 2:04 PM
response to LDL-C lowering and those carrying the E4 allele have a significantly
lower response.51 Plasma total cholesterol, LCL-C, and triglyceride responses
were significantly greater for E2 subjects as compared to E3 and E4 subjects
when taking atorvastatin, one of the most effective statins available.
Other polymorphisms involved in the individual response to statins are the
PvuII and AvaII polymorphisms of the LDL-receptor gene. These polymorphisms
have been shown to influence the levels of LDL-C in both normo- and hyperc-
holesterolemic subjects and may influence the response to treatment with statins.52
There has been minimal work done on these polymorphisms with conflicting
results. Studies have reported that subjects with the P2 allele of the PvuII poly-
morphism have a better response than those homozygous for the P1 allele, but
this has yet to be reproduced.45,53 Lahoz has shown a significant trend in the AvaII
polymorphism showing that those homozygous for the (−) allele responded less
than heterozygotes, who in turn responded less than homozygotes for the (+)
allele. Clinical importance for these polymorphisms is currently unknown and
requires further studies to determine an effect.
The −514C/T hepatic lipase and apolipoprotein A-1 promoter polymor-
phisms can also influence the effectiveness of statin therapy. Individuals who
are homozygous for the C allele had scarce modification of HDL-C with
pravastatin treatment while in carriers of the T allele, the increase in HDL-C
was almost 7%.54 However, it has been reported that those with the CC genotype
may benefit more from statins since they appear to have greater improvement
in coronary lesions than CT and TT genotypes. Carriers of the A allele for the
apoA-1 polymorphism showed HDL-C concentrations significantly higher than
G allele carriers with pravastatin treatment. The mechanisms by which the G/A
polymorphism influences HDL-C levels and its response to pravastatin are not
known. This is the case for many of the above-mentioned polymorphisms
interacting with statins. The value of determining genetic polymorphisms in
order to predict the effects of statin treatment is still unclear, but will improve
with further research.
Niacin (nicotinic acid) has been used as a lipid lowering agent to treat
dyslipidemia since 1954.55–57 Niacin is one of the most effective agents available
for raising HDL-C and lowering triglycerides and, hence, adding niacin to statin
therapy may be desirable. There has been reluctance to take immediate-release
niacin due to possible side effects of flushing and skin rash, but extended-release
niacin is both better tolerated and safer.58 Many studies suggest that combination
niacin–statin therapy leads to improved lipoprotein profiles and clinical outcomes
over monotherapy using either niacin or a statin.59,60 The addition of 2000 mg/d
of extended-release niacin to patients on statin therapy can decrease total choles-
terol an additional 21%, LDL-C by 31%, triglycerides by 27%, and increase
HDL-C by an additional 27%. Therefore, drug–nutrient interactions need to be
considered for each individual and these drug therapies could be more effective
if they are tailored better to individual needs.
DK5836_C008.fm Page 246 Monday, May 22, 2006 2:04 PM
for colon cancer was observed for individuals with a high intake of calcium,
vitamin D, and low-fat dairy products, and with the SS poly-A or BB BsMI
genotypes. Also, calcium, vitamin D, and low-fat dairy products were inversely
associated with rectal cancer in women, but not in men. These results are sup-
ported by Kim et al.93 who showed that those with the BB BsMI genotype and
the lowest calcium and vitamin D intake were at a reduced risk of colorectal
adenomas relative to those with the bb BsMI genotype and the highest levels of
intake, although this interaction wasn’t statistically significant.92 It has been
proposed that the antineoplastic activity of the BB and SS alleles is due to the
increased 1,25(OH)2D levels associated with those alleles.92 It may also be pos-
sible that different VDR genotypes interact differently with VDREs, which could
lead to altered ability to transcriptionally regulate target genes.
Because colorectal carcinomas are derived from rapidly proliferating tissues, they
have a great requirement for DNA synthesis and are affected by the metabolic
fate of folic acid. Epidemiological studies have shown that colorectal cancer was
reduced (~50%) in individuals with the MTHFR 677 TT genotype when folate
intake was adequate, compared to those with the MTHFR 677 CC genotype.71
Slattery and Potter96 analyzed a mixed-ethnicity population of individuals aged
30 to 79 who were diagnosed with first primary colon cancer matched with control
subjects. Dietary food frequency questionnaires were distributed. After genotyp-
ing for the MTHFR 677 C→T polymorphism, frequencies of the CC, CT, and
TT genotypes among all cases were 45.9%, 44.7%, and 9.5%, respectively, and
were very similar for the controls. High intakes of folate were associated with a
30 to 40% risk reduction in colon cancer among those with the TT genotype
relative to those with low folate intake who were CC genotypes.97 Shannon et
al.98 provided further support by showing that the MTHFR TT genotype conferred
an increased risk of colorectal cancer in subjects older but not younger than 70
years of age. Hence, the variant TT form of the MTHFR gene appears to be
slightly inversely associated with colon cancer, while the heterozygote CT and
wild-type CC genotypes appear to have similar associations with colon cancer.97
When examining joint effects between the two polymorphisms, it was found that
the double heterozygotes (677 CT/1298 AC) were approximately five times less
likely than 677 CC/1298 AA individuals to develop acute lymphocytic leukemia.95
8.4 CONCLUSIONS
In this new millennium, research in nutrition and disease prevention must now
prioritize in understanding the molecular basis by which nutrients affect the
disease process.68,100,101 A well-coordinated, multidisciplinary effort must be made
amongst all types of scientists — nutritional scientists, molecular biologists,
geneticists, statisticians, and clinical cancer researchers — in order to move this
research forward. There are many more genes related to the pathogenesis of
cancer and CVD that have not been included in this paper because there hasn’t
yet been information for those genes showing relevance for dietary interventions.
For each gene there may be one or more polymorphisms associated with it, hence,
creating even more possible interactions between genetic polymorphisms and
diet. This creates an ongoing challenge in trying to solve multifactorial, polygenic
diseases. The individualization of treatments has been one of the strongest ther-
apeutic tools, yet also one of the biggest challenges of the pregenomic era.
However, now, with the availability of comprehensive genetic and molecular
profiles and with improved molecular techniques, it will be possible to respond
to diseases with individual strategies based on genetic profiles.16 Nutrigenomics
DK5836_C008.fm Page 252 Monday, May 22, 2006 2:04 PM
has a powerful message — food and nutrients have the potential to affect the
balance between health and disease by affecting gene expression and function.102
Exploring the role that functional gene polymorphisms play in determining
risk and in determining the levels of intermediate phenotypes is important to our
understanding of the key metabolic pathways and physiology in both the disease
and disease-free states.7 More research in this area is still required, as there are
many more genetic polymorphisms and nutrient interactions that are related to
cancer and CVD. As was seen for the added benefit of including niacin with
statins for treating hypercholesterolemia, there is great potential to improve treat-
ment by adding a simple nutrient to drug therapy. These benefits will vary
according to each individual’s genotype. Yet, also, there is the potential for adverse
effects with adding a nutrient to a drug therapy, which can also vary depending
on one’s genotype; hence, caution is required as well. It seems that only the
surface has been scratched in the pursuit of solving the genetic–nutrient–drug
mechanisms of CVD and cancer, but a great deal of research will undoubtedly
follow. The key now is for researchers to always think of nutrient, drug, and gene
interactions when designing experiments because the three are no longer solely
independent of each other, but inevitably linked.
REFERENCES
1. Fogg-Johnson, N. and Merolli, A. (2000) Nutrigenomics: The next wave in nutri-
tion research. Nutraceuticals World March 2000: 86–95.
2. Kaput, J. and Rodriguez, R. L. (2004) Nutritional genomics: the next frontier in
the postgenomic era. Physiol Genomics 16(2): 166–177.
3. Bakovic, M. (2005) An Introduction to Nutrigenomics: What Consumers Need to
Know. FAQ’s and Facts Sheet; Dietitians of Canada (Available online: http://www.
dietitians.ca)
4. Elliott, R. and Ong, T. J. (2002) Science, medicine, and the future. Nutritional
genomics. BM J 324: 1438–1442.
5. Kirk, B. W., Feinsod, M., Favis, R., Kilman, R. M., and Barany, F. (2002) Single
nucleotide polymorphism seeking long-term association with complex disease.
Nucleic Acids Res 30: 3295–3311.
6. Iannuzzi, M. C., Maliarik, M., and Rybicki, B. (2002) Genetic polymorphism in
lung disease: bandwagon or breakthrough? Respir Res 3: 15.
7. Loktionov, A. (2003) Common gene polymorphisms and nutrition: emerging links
with pathogenesis of multifactorial chronic diseases (Review). J Nutr Biochem
14: 426–451.
8. Stephens, J. W. and Humphries, S. E. (2003) The molecular genetics of cardio-
vascular disease: clinical implications. J Intern Med 253: 120–127.
9. Tai, E. S., Corella, D., Deurenberg-Yap, M., Cutter, J., Chew, S. K., Tan, C. E.,
and Ordovas, J. M. (2003) Dietary fat interacts with the −514 CT polymorphism
in the hepatic lipase gene promoter on plasma lipid profiles in a multiethnic Asian
population: the 1998 Singapore national health survey. J Nutr 133: 3399–3408.
DK5836_C008.fm Page 253 Monday, May 22, 2006 2:04 PM
10. Hagberg, J. M., Wilund, K. R., and Ferrell, R. E. (2000) APOE gene and gene-
environment effects on plasma lipoprotein-lipid levels. Physiol Genomics 4:
101–108.
11. Erkkila, A. T., Sarkkinen, E. S., Lindi, V., Lehto, S., Laakso, M., and Uusitupa,
M. I. (2001) APOE polymorphism and the hypertryglyceridimic effect of dietary
sucrose. Am J Clin Nutr 73: 746–752.
12. Campos, H., D’Agostino, M., and Ordovas, J. M. (2001) Gene–diet interactions
and plasma lipoproteins: role of apolipoprotein E and habitual saturated fat intake.
Genet Epidemiol 20: 117–128.
13. Corella, D., Tucker, K., Lahoz, C., Coltell, O., Cupples, L. A., Wilson, P., Schaefer,
E. J., and Ordovas, J. M. (2001) Alcohol drinking determines the effect of the
APOE locus on LDL-cholesterol concentrations in men: the Framingham Off-
spring Study. Am J Clin Nutr 73: 736–745.
14. Eichner, J. E., Dunn, S. T., Perveen, G., Thompson, D. M., Stewart, K. E., and
Stroehla, B. C. (2002) Apolipoprotein E polymorphism and cardiovascular disease:
a HuGE review. Am J Epidemiol 155(6): 487–495.
15. Jimenez-Sanchez, G., Childs, B., and Valle, D. (2001) Human disease genes.
Nature 409: 853–855.
16. Gerling, I. C., Solomen, S. S., and Bryer-Ash, M. (2003) Genomes, transcriptomes,
and proteomes. Arch Intern Med 163: 190–198.
17. Fodinger, M., Horl, W. H., and Sunder-Plassmann, G. (2000). Molecular biology
of 5,10-methylenetetrahydrofolate reductase. J Nephrol 13: 20–33.
18. Frosst, P., Blom, H. J., Milos, R., Gryette, P., Sheppard, C. A., Matthews, R. G.,
Boers, G. J. H., Heijer, M., Kluijtmans, L. A. J., Heuval, L. P., and Rozen, R.
(1995) A candidate genetic risk factor for vascular disease: a common mutation
in methylenetetrahydrofolate reductase. Nat Genet 10: 111–113.
19. Ueland, P. M., Hustad, S., Schneede, J., Refsum, H., and Vollset, S. E. (2001)
Biological and clinical implications of the MTHFR C677T polymorphism. Trend
Pharmacol Sci 22: 195–201.
20. Kang, S. S., Passen, E. L., Ruggie, N., Wond, P. W., and Sora, H. (1993) Ther-
molabile defect of methylenetetrahydrofolate reductase in coronary artery disease.
Circulation 88: 1463–1469.
21. Refsum, H., Ueland, P. M., Nygard, O., and Vollset, S. E. (1998) Homocysteine
and cardiovascular disease. Annu Rev Med 49: 31–62.
22. Molloy, A. M., Daly, S., Mills, J. L., Kirke, P. N., Whitehead, A. S., Ramsbottom,
D., Conley, M. R., Weir, D. G., and Scott, J. M. (1997) Thermolabile variant of
5,10-methylenetetrahydrofolate reductase associated with low red-cell folates:
implications for folate intake recommendations. Lancet 349: 1591–1593.
23. de Bree, A., Verschuren, W. M., Bjorke-Monsen, A. L., van der Put, N. M., Heil,
S. G, Trijbels, F. J., and Blom, H. J. (2003) Effect of the methylenetetrahydrofolate
reductase 677 CT mutation on the relations among folate intake and plasma folate
and homocysteine concentrations in a general population sample. Am J Clin Nutr
77: 687–693.
24. Brattstrom, L., Wilcken, D. E., Ohrvik, J. and Brudin, L. (1998) Common meth-
ylenetetrahydrofolate reductase gene mutation leads to hyperhomocysteinemia but
not to vascular disease: the result of a meta-analysis. Circulation 98: 2520–2526.
DK5836_C008.fm Page 254 Monday, May 22, 2006 2:04 PM
25. Zittoun, J., Tonetti, C., Bories, D., Pignon, J. M., and Tulliez, M. (1998) Plasma
homocysteine levels related to interactions between folate status and methylene-
tetrahydrofolate reductase: a study in 52 healthy subjects. Metabolism 47:
1413–1418.
26. Lievers, K. J., Boers, G. H., Verhoef, P., den Heijer, M., Kluijtmans, L. A., van
der Put, N. M., Trijbels, F. J. and Blom, H. J. (2001) A second common variant
in the methylenetetrahydrofolate reductase (MTHFR) gene and its relationship to
MTHFR enzyme activity, homocysteine, and cardiovascular disease risk. J Mol
Med 79: 522–528.
27. Shelnutt, K. P., Kauwell, G. P., Chapman, C. M., Gregory III, J. F, Maneval, D.
R., Browdy, A. A., Theriaque, D. W., and Bailey, L. B. (2003) Folate status
response to controlled folate intake is affected by the methylenetetrahydrofolate
reductase 677 CT polymorphism in young women. J Nutr 133: 4107–4111.
28. Ashfield-Watt, P. A., Pullin, C. H., Whiting, J. M., Clark, Z. E., Moat, S. J.,
Newcombe, R. G., Burr, M. L., Lewis, M. J., Powers, H. J., and McDowell, I. F.
Methylenetetrahydrofolate reductase 677 CT genotype modulates homocysteine
responses to a folate-rich diet or a low-dose folic acid supplement: a randomized
controlled trial. Am J Clin Nutr 76: 180–186.
29. van’t Hooft, F. M., Lundahl, B., Ragogna, F., Karpe, F., Olivecrona, G., and
Hamsten, A. (2000) Functional characterization of 4 polymorphisms in promoter
region of hepatic lipase gene. Arterioscler Thromb Vasc Biol 20: 1335–1339.
30. Connelly P. W. (1999) The role of hepatic lipase in lipoprotein metabolism. Clin
Chim Acta 286: 243–245.
31. Galan, X., Robert, M. Q., Llobera, M., and Ramirez, I. (2000) Secretion of hepatic
lipase by perfused liver and isolated hepatocytes. Lipids 35: 1017–1026.
32. Campos, H., Dreon, D. M., and Krauss, R. M. (1995) Associations of hepatic and
lipoprotein lipase activities with changes in dietary composition and low-density
lipoproteins subclasses. J Lipid Res 36: 462–472.
33. Deeb, S. S. and Peng, R. (2000) The C-514T polymorphism in the human hepatic
lipase gene promoter diminishes its activity. J Lipid Res 41: 155–158.
34. Kuusi, T., Saarinen, P., and Nikkila, E. A. (1980) Evidence for the role of hepatic
endothelial lipase in the metabolism of plasma high-density lipoprotein2 in man.
Atherosclerosis 36B: 589–593.
35. Grundy, S. M., Vega, G. L., Otvos, J. D., Rainwater, D. L., and Cohen, J. C. (1999)
Hepatic lipase activity influences high density lipoprotein subclass distribution in
normotriglyceridaemic men: genetic and pharmacological evidence. J Lipid Res
40: 229–234.
36. Hegele, R.A, Tu, L., and Connelly, P. W. (1992) Human hepatic lipase mutations
and polymorphisms. Hum Mutat 1: 320–324.
37. Hegele, R. A., Little, J. A., Vezina, C., Maguire, G. F., Tu, L., Wolever, T. S.,
Jenkins, D. A., and Connelly, P. W. (1993) Hepatic lipase deficiency: clinical,
biochemical, and molecular genetic characteristics. Arterioscler Throm 13:
720–728.
38. Pihlajamaki, J., Karjalainen, L., Karhapaa, P. Vauhkonen, I., Taskinen, M. R.,
Deeb, S. S., and Laakso, M. (2000) G-250A substitution in promoter of hepatic
lipase gene is associated with dyslipidemia and insulin resistance in healthy control
subjects and in members of families with familial combined hyperlipidemia.
Arteriosclerosis 20: 1789–1795.
DK5836_C008.fm Page 255 Monday, May 22, 2006 2:04 PM
39. Couture, P., Otvos, J. D., Cupples, L. A., Lahoz, C., Wilson, P. W., Schaefer, E.
J., and Ordovas, J. M. (2000) Association of the C-514T polymorphism in the
hepatic lipase gene with variations in lipoprotein subclass profiles: The Framing-
ham Offspring Study. Arterioscler Thromb Vasc Biol 20: 815–822.
40. Guerra, R., Wang, J., Grundy, S. M., and Cohen, J. C. (1997) A hepatic lipase
(LIPC) allele associated with high plasma concentration of high density lipopro-
tein cholesterol. Proc Natl, Acad Sci USA 94: 4532–4537.
41. Zambon, A., Deeb, S. S., Hokanson, J. E., Brown, B. G., and Brunzell, J. D.
(1998) Common variants in the promoter of the hepatic lipase gene are associated
with lower levels of hepatic lipase activity, buoyant LDL, and higher HDL2
cholesterol. Arterioscler Thromb Vasc Biol 18: 1723–1729.
42. Shohet, R. V., Vega, G. L., Anwar, A. Cigarroa, J. E., Grundy, S. M., and Cohen,
J. C. (1999) Hepatic lipase (LIPC) promoter polymorphism in men with coronary
artery disease. Allele frequency and effects on hepatic lipase activity and plasma
HDL-C concentrations. Arterioslcer Thromb Vasc Biol 19: 1975–1978.
43. Ordovas, J. M., Corella, D., Demissie, S., Cupples L. A., Couture, P., Coltell, O.,
Wilson, P. W., Schaefer, E. J., and Tucker, K. L. (2002) Dietary fat intake deter-
mines the effect of a common polymorphism in the hepatic lipase gene promoter
on high-density lipoprotein metabolism. Circulation 106: 2315–2321.
44. Pedro-Botet, J., Schaefer, E. J., Bakker-Arkema, R. G., Black, D. M., Stein, E.
M., Corella, D., and Ordovas, J. M. (2001) Apolipoprotein E genotype affects
plasma lipid response to atorvastatin in a gender specific manner. Atherosclerosis
158: 183–193.
45. Lahoz, C., Pena, R., Mostaza, J. M., Laguna, F., Garcia-Iglesias, M. F., Taboada,
M., and Pinto, X. (2005) Baseline levels of low-density lipoprotein cholesterol
and lipoprotein (a) and the AvaII polymorphism of the low-density lipoprotein
receptor gene influence the response of low-density lipoprotein cholesterol to
pravastatin treatment. Metab Clin Exper 54: 741–747.
46. Shepherd, J., Cobbe, S. M., Ford, I., Isles, C. G., Lorimer, A. R., MacFarlane, P.
W., McKillop, J. H., and Packard, C. J. (2004) Prevention of coronary heart disease
with pravastatin in men with hypercholesterolemia. Atherscler Supples 5: 91–97.
47. Downs, J. R., Clearfeld, M., Weis, S., Whitney, E., Shapiro, D. R., Beere, P. A.,
Langendorfer, A., Stein, E. A., Kruyer, W., and Gotto Jr, A. M. (1998) Primary
prevention of acute coronary events with lovastatin in men and women with
average cholesterol levels: results of AFCAPS/TexCAPS. Air Force/Texas Coro-
nary Athersclerosis Prevention Study JAMA 279: 1615–1622.
48. Sacks, R. M., Pfeffer, M. A., Moye, L. A., Rouleau, J. L., Rutherford, J. D., Cole,
T. G., Brown, L., Warnica, J. W., Arnold, J. M., Wun, C. C., Davis, B. R., and
Braunwald, E. (1996) The effect of pravastatin on coronary events after myocardial
infarction in patients with average cholesterol levels. Cholesterol and Recurrent
Events Trial investigators. N Engl J Med 335: 1001–1009.
49. Nestel, P., Simons, L., Barter, P., Clifton, P., Colquhoun, D., Hamilton-Craig, I.,
Sikaris, K., and Sullivan, D. (1997) A comparative study of the efficacy of sim-
vastatin and gemfibrozil in combined hyperlipoproteinemia: prediction of response
by baseline lipids, apoE genotype, lipoprotein(a) and insulin. Athersclerosis 129:
231–239.
DK5836_C008.fm Page 256 Monday, May 22, 2006 2:04 PM
50. Sanllehy, C., Casals, E., Rodriguez-Villar, C., Zambon, D., Ojuel, J., Ballesta, A.
M., and Ros, E. (1998) Lack of interaction of apolipoprotein E phenotype with
the lipoprotein response to lovastatin or gemfibrozil in patients with primary
hypercholesterolemia. Metabolism 47: 560–565.
51. Ordovas, J. M., Lopez-Miranda, J., Perez-Jimenez, F., Rodriguez, C., Park, J. S.,
Cole, T., and Schaefer, E. J. (1995) Effect of apolipoprotein E and A-IV phenotypes
on the low-density lipoprotein response to HMG CoA reductase inhibitor therapy.
Atherosclerosis 113: 157–166.
52. Salazar, L. A., Hirata, M. H., Giannini, S. D., Forti, N., Diament, J., Issa, J. S.,
and Hirata, R. D. (1999) Effects of AvaII and HincII polymorphisms at the LDL
receptor gene on serum lipid levels of Brazilian individuals with high risk for
coronary heart disease. J Clin Lab Anal 13: 251–258.
53. Salazar, L. A., Hirata, M. H., Quintao, E. C., and Hirata, R. D. (2000) Lipid-
lowering response of the HMG-CoA reductase inhibitor fluvastatin is influenced
by polymorphisms in the low-density lipoprotein receptor gene in Brazilian
patients with primary hypercholesterolemia. J Clin Lab Anal 14: 125–131.
54. Lahoz, C., Pena, R., Mostaza, J. M., Laguna, F., Garcia-Iglesias, M. F., Taboada,
M., and Pinto, X. (2005) The -514C/T polymorphism of the hepatic lipase gene
significantly modulates the HDL-cholesterol response to statin treatment. Athero-
sclerosis 182: 129–134.
55. Kashyap, M. L., McGovern, M. E., Berra, K., Guyton, J. R., Kwiterovich, P. O.,
Harper, W. L., Toth, P. D., Favrot, L. K., Kerzner, B., Nash, S. D., Bays, H. E.,
and Simmons, P. D. (2002) Long-term safety and efficacy of a once-daily nia-
cin/lovastatin formulation for patients with dyslipidemia. Am J Cardiol 89:
672–678.
56. Birjmohun, R. S., Hutten, B. A., Kastelein, J. J. P., and Stroes, E. S. G. (2004)
Increasing HDL cholesterol with extended-release nicotinic acid: from promise
to practice. Neth J Med 62: 229–234.
57. Ganji, S. H., Kamanna, V. S., and Kashyap, M. L. (2003) Niacin and cholesterol:
role in cardiovascular disease (review). J Nutr Biochem 14: 298–305.
58. Wolfe, M. L., Vartanian, S. F., Ross, J. L., Bansavich, L., Mohler, E. R., Meagher,
E, Friedrich, C. A., and Rader, D. J. (2001) Safety and effectiveness of niaspan
when added sequentially to a statin for treatment of dyslipidemia. Am J Cardiol
87: 476–479.
59. Ito, M. K. (2003) Advances in the understanding and management of dyslipidemia:
using niacin-based therapies. Am J Health-Syst Pharm 60: S15–S21.
60. Rosenson, R. S. (2003) Antiatherothrombotic effects of nicotinic acid. Atheroscle-
rosis 171: 87–96.
61. Rock, C. L., Lampe, J. W., and Patterson, R. E. (2000) Nutrition, genetics, and
risks of cancer. Annu Rev Public Health 21: 47–64.
62. Greenwald, P. (1996) The potential of dietary modification to prevent cancer. Prev
Med 25: 41–43.
63. Lai, C. and Shields, P. G. (1999) The role of interindividual variation in human
carcinogenesis. J Nutr 129: 552S–555S.
64. Perera, F. P. (1997) Environment and cancer: who are susceptible? Science 278:
1067–1073.
65. Steinmetz, K. A. and Potter, J. D. (1996) Vegetables, fruit, and cancer prevention:
a review. J Am Diet Assoc 96 (10): 1027–1039.
DK5836_C008.fm Page 257 Monday, May 22, 2006 2:04 PM
66. Mathers, J. C. (2003) Nutrition and cancer prevention: diet-gene interactions. Proc
Nutr Soc 62: 605–610.
67. Patterson, R. E, Eaton, D. L. and Potter, J. D. (1999) The genetic revolution:
change and challenge for the dietetics profession. J Am Diet Assoc 99 (11):
1412–1420.
68. Milner, J. A. (2002) Strategies for cancer prevention: the role of diet. Br J Nutr
87: S265–S272.
69. Greenwald, P., Milner, J. A. and Clifford, C. K. (2000) Creating a new paradigm
in nutrition research within the national cancer institute. J Nutr 130: 3103–3105.
70. Cotton, S. C., Sharp, L., Little, J., and Brockton, N. (2000) Glutathione S-trans-
ferase polymorphisms and colorectal cancer: a HuGE review. Am J Epidemiol
151(1): 7–32.
71. Milner, J. A., McDonald, S. S., Anderson, D. E., and Greenwald, P. (2001)
Molecular targets for nutrients involved with cancer prevention. Nutr Cancer
41(1&2): 1–16.
72. Platz, E. A., Leitzmann, M. F., Hollis, B. W., Willett, W. C., and Giovannucci, E.
(2004) Plasma 1,25-dihydroxy- and 25-hydroxyvitamin D and subsequent risk of
prostate cancer. Canc Causes Cont 15: 255–265.
73. Giovannucci, E. (1998) Dietary influences of 1,25(OH)2 vitamin D in relation to
prostate cancer: a hypothesis. Canc Causes Cont 9: 657–582.
74. Platz, E. A., Hankinson, S. E., Hollis, B. W., Colditz, G. A., Hunter, D. J., Speizer,
F. E., and Giovannucci, E. (2000) Plasma 1,25-dihydroxy- and 25-hydroxyvitamin
D and adenomatous polyps of the distal colorectum. Canc Epidemiol Biomark
Prev 9: 1059–1065.
75. Kallay, E., Pietschmann, P., Toyokuni, S., Bajna, E., Hahn, P., Mazzucco, K.,
Bieglmayer, C., Kato, S., and Cross, H. S. (2001) Characterization of a vitamin
D receptor knockout mouse as a model of colorectal hyperproliferation and DNA
damage. Carcinogenesis 22: 1429–1435.
76. Liu, M., Lee, M. H., Cohen, M., Bommakanti, M., and Freedman, L. P. (1996)
Transcriptional activation of the Cdk inhibitor p21 by vitamin D3 leads to the
induced differentiation of the myelomonocytic cell line U937. Genes Dev 10:
142–153.
77. Palmer, H. G., Gonzalez-Sancho, J. M., and Espada, J. (2001) Vitamin D(3)
promotes the differentiation of colon carcinoma cells by the induction of E-
cadherin and the inhibition of beta-catenin signaling. J Cell Biol 154: 369–387.
78. Mathiasen, I. S., Lademann, U. and Jaattela, M. (1999) Apoptosis induced by
vitamin D compounds in breast cancer cells is inhibited by Bcl-2 but does not
involve known caspases or p53. Canc Res 59: 4848–4856.
79. Donohue, M. M. and Demay, M. B. (2002) Rickets in VDR null mice is secondary
to decreased apoptosis of hypertrophic chondrocytes. Endocrinology 143:
3691–3694.
80. Morrison, N. A., Qi, J. C., Tokita, A., Kelly, P. J., Crofts, L., Nguyen, T. V.,
Sambrook, P. N., and Eisman, J. A. (1994) Prediction of bone density from vitamin
D receptor alleles. Nature 367: 284–287.
81. Ingles, S. A., Wang, J., Coetzee, G. A., Lee, E. R., Frank, H. D., and Haile, R.
W. (2001) Vitamin D receptor polymorphisms and risk of colorectal adenomas.
Canc Causes Cont 12: 607–614.
DK5836_C008.fm Page 258 Monday, May 22, 2006 2:04 PM
82. Habuchi, T., Suzuki, T., Sasaki, R., Wang, L., Sato, K., Satoh, S., Akao, T.,
Tsuchiya, N., Shimoda, N., Wada, Y., Koizumi, A., Chihara, J., Ogawa, O., and
Kato, T. (2000) Association of vitamin D receptor gene polymorphism with
prostate cancer and benign prostatic hyperplasia in a Japanese population. Canc
Res 60: 305–308.
83. Sinha, R. and Caporaso, N. (1999) Diet, genetic susceptibility and human cancer
etiology. J Nutr 129: 556S–559S.
84. Wong, H-L., Seow, A., Arakawa, K., Lee, H-P., Yu, M. C., and Ingles, S. A. (2003)
Vitamin D receptor start codon polymorphism and colorectal risk: effect modifi-
cation by dietary calcium and fat in Singapore Chinese. Carcinogenesis 24:
1091–1095.
85. Peters, U., McGlynn, K. A., Chatterjee, N., Gunter, E., Garcia-Closas, M, Roth-
man, N., and Sinha, R. (2001) Vitamin D, calcium, and vitamin D receptor
polymorphism in colorectal adenomas. Canc Epidemiol Biomark Prev 10:
1267–1274.
86. Niv, Y., Sperber, A. D., Figer, A., Igael, D., Shany, S., Fraser, G., and Schwartz,
B. (1999) In colorectal carcinoma patients, serum vitamin D levels vary according
to the stage of carcinoma. Cancer 86: 391–397.
87. Xue, L., Lipkin, M, Newmark, H., and Wang, J. (1999) Influence of dietary
calcium and vitamin D on diet-induced epithelial cell hyperproliferation in mice.
J Natl Canc Inst 91: 176–181.
88. Tong, W. M., Kallay, E., Hofer, H., Hulla, W., Manhardt, T., Peterlik, M., and
Cross, H. S. (1999) Growth regulation of human colon cancer cells by epidermal
growth factor and 1,25-dihydroxyvitamin D3 is mediated by mutual modulation
of receptor expression. Eur J Canc 34: 2119–2125.
89. Diaz, G. D., Paraskeva, C., Thomas, M. G., Binderup, L., and Hague, A. (2000)
Apoptosis is induced by the active metabolite of vitamin D3 and its analogue,
EB1089 in colorectal adenoma and carcinoma cells: possible implications for
prevention and therapy. Canc Res 60: 2304–2312.
90. Evans, S. R., Nolla, J., Hanfelt, J., Shabahang, M., Nauta, R. J., and Shchepotin,
I. B. (1998) Vitamin D receptor expression as a predictive marker of biological
behavior in human colorectal cancer. Clin Canc Res 4: 1591–1595.
91. Slattery, M. L., Yakumo, K., Hoffman, M., and Newhausen, S. (2001) Variants of
the VDR gene and risk of colon cancer. Canc Causes Cont 12: 359–364.
92. Slattery, M. L., Newhausen, S. L., Hoffman, M., Caan, B., Curtin, K., Ma, K.,
and Samowitz, W. (2004) Dietary calcium, vitamin D, VDR genotypes and col-
orectal cancer. Int J Canc 111: 750–756.
93. Kim, H. S., Newcomb, P. A., Ulrich, C. M., Keener, C. L., Bigler, J., and Farin,
F. M. (2001) Vitamin D receptor polymorphism and the risk of colorectal ade-
nomas: evidence of interaction with dietary vitamin D and calcium. Canc Epide-
miol Biomark Prev 10: 869–874.
94. Bailey, L. B. and Gregory III, J. F. (1999) Polymorphisms of methylenetetrahy-
drofolate reductase and other enzymes: metabolic significance, risks and impact
on folate requirement. J Nutr 129: 919–922.
95. Skibola, C. F., Smith, M. T., Kane, E., Roman, E., Rollinson, S., Cartwright, R.
A., and Morgan, G. (1999) Polymorphisms in the methylenetetrahydrofolate
reductase gene are associated with susceptibility to acute leukemia in adults. PNAS
96: 12810–12815.
DK5836_C008.fm Page 259 Monday, May 22, 2006 2:04 PM
96. Ma, J., Stampfer, M. J., Christensen, B., Giovannucci, E., Hunter, D. J., Chen, J.,
Willett, W. C., Selhub, J., Hennekens, C. H., Gravel, R., and Rozen, R. (1999) A
polymorphism of the methionine synthase gene: association with plasma folate,
vitamin B12, homocysteine, and colorectal cancer risk. Canc Epidemiol Biomark
Prev 8: 825–829.
97. Slattery, M. L., Potter, J. D., Samowitz, W., Schaffer, D., and Leppert, M. (1999)
Methylenetetrahydrofolate reductase, diet, and risk of colon cancer. Canc Epide-
miol Biomark Prev 8: 513–518.
98. Shannon, B., Gnanasampanthan, S., Beilby, J. and Lacopetta, B. (2002) A poly-
morphism in the methylenetetrahydrofolate reductase gene predisposes to colorec-
tal cancers with microsatellite instability. Gut 50: 520–524.
99. Ulvik, A., Vollset, S. E., Hansen, S., Gislefoss, R., Jellum, E., and Ueland, P. M.
Colorectal cancer and the methylenetetrahydrofolate reductase 677C→T and
methionine synthase 2756A→G polymorphisms: a study of 2168 case-control
pairs from the JANUS cohort. Canc Epidemiol Biomark Prev 13: 2175–2180.
100. Rankinen, T. and Tiwari, H. (2004) Genome scans for human nutritional traits:
what have we learned? Nutr 20: 9–13.
101. Kaput, J. (2004) Diet-disease gene interactions. Nutrition 20(1): 26–31.
102. Bakovic, M. (2005) Nutrigenomics: balancing diet to gene function proposes a
new-age recipe for better health Resource Information; Dietitians of Canada
Available online: http://www.dietitians.ca
DK5836_C008.fm Page 260 Monday, May 22, 2006 2:04 PM
DK5836_C009.fm Page 261 Monday, May 22, 2006 2:15 PM
9 Nutrigenomics and
Pharmacogenomics of
Human Cancer: On the
Road from Nutrigenetics
and Pharmacogenetics
Alexandre Loktionov
CONTENTS
9.1 Introduction..............................................................................................262
9.2 Diet, Genetic Background, and Cancer Risk..........................................263
9.2.1 Xenobiotic Metabolism-Controlling Pathways ...........................264
9.2.2 Folate Metabolism and DNA Methylation .................................271
9.2.3 Alcohol Metabolism ....................................................................272
9.2.4 DNA Repair.................................................................................273
9.2.5 Energy Balance Regulation .........................................................274
9.2.6 Other Physiological Pathways, Genes, and
Research Perspectives..................................................................274
9.3 Advances in Cancer Pharmacogenomics and
Links to Nutrigenomics...........................................................................276
9.3.1 Drug-Metabolizing Phase I and Phase II Enzymes....................276
9.3.2 Enzymes of Purine/Pyrimidine Metabolism ...............................290
9.3.3 Enzymes and Regulatory Factors of Folate Metabolism............291
9.3.4 DNA Repair Effects on Cancer Treatment .................................291
9.3.5 ATP-Binding Cassette Transporters ............................................292
9.3.6 Other Drug Targets and Recent Advances..................................293
9.4 Conclusion ...............................................................................................294
References .........................................................................................................295
261
DK5836_C009.fm Page 262 Monday, May 22, 2006 2:15 PM
9.1 INTRODUCTION
Recent rapid progress in the determination of the human genome sequence has
strongly stimulated the development of genomic approaches to the investigation
of a broad spectrum of medical problems.1,2 Functional genomics is now expand-
ing into several bioscientific domains, which previously had little relation to
genetic research. Emerging amalgamation of functional genomics with nutritional
sciences has recently been called nutrigenomics; however this new discipline is
just entering its embryonal state, being mainly discussed in numerous reviews
devoted to future opportunities rather than existing reality.3–6 This dream-like
state of the subject has brought about a very vague concept of suggested bound-
aries of nutrigenomics, which often tend to incorporate transcriptomics, proteom-
ics, and, eventually, metabolomics. However, the integrative ideas, more appro-
priately attributable to the systems biology, still have little impact on our
knowledge regarding the relationship between human genome, nutritional factors,
and genesis of major human chronic diseases, such as cardiovascular disease and
cancer.
Cancer remained one of the most enigmatic conditions still challenging sci-
entists and physicians at the beginning of the new millennium. Although it is
generally accepted that genetic component plays a leading role in the pathogenesis
of neoplastic disorders, only a minor fraction of all human cancers (i.e., hereditary
cancer syndromes accounting for 5 to 10% of all cancer cases) inevitably results
from specific deteriorating mutations transmitted through germ line.7,8 In contrast,
the development of sporadic tumors is regarded as a complex pathogenetic process
involving interactions of multiple contributing factors, both internal (intrinsic host
factors) and external.9 In this context, diet obviously constitutes an omnipresent
combination of external influences potentially important for all malignancies at
different stages of neoplastic growth. Furthermore, dietary factors can interact
with anticancer drugs used at later stages of the disease, thus affecting chemo-
therapy outcome.
Although it would be extremely tempting to have a chance to look at cancer
pathogenesis and medicamentous treatment through the prism of genome-gov-
erned network of interactions between nutrition, chemotherapeutic agents, and
biological systems in all their complexity, the existing knowledge is obviously
insufficient to allow any fact-based analysis of this type. In the context of this
chapter, it appears to be justified to use a limited meaning of the term “nutrige-
nomics,” analogous to “pharmacogenomics,” which is often defined as a field
aiming to investigate the genetic basis for interindividual differences in drug
response using genome-wide approaches.10 It is, however, clear that the effects
of drugs, which are administered as pure compounds in precise doses, are much
easier to assess than combined and interfering effects of numerous food constit-
uents.4 Nevertheless, this chapter is an attempt to analyze the present state of
nutrigenomic and pharmacogenomic research in relation to human cancer.
DK5836_C009.fm Page 263 Monday, May 22, 2006 2:15 PM
Dietary influences
Carcinogenic Protective
Effects on the malignant growth are largely unknown Life support
Protective Carcinogenic (interactions with chemotherapeutic drugs possible) (anti-cahexia
measures)
Adjuvant
Chemotherapy
therapy
and
Radiotherapy
studied in relation to cancer risk; however, no firm evidence in favor of this link
has been produced.28 It should be noted that the initial concept of the P450 family,
regarding it mainly as a hepatic xenobiotic detoxification system, is now being
expanded upon the discovery of multiple additional functions of these
enzymes.26,27 It also has become clear that some CYPs are closely involved in
the metabolism of anticancer chemotherapeutic agents.28 These interactions are
going to be addressed later in this chapter.
Available information on interactions of dietary factors with polymorphic
variants of the CYP genes is presented in the beginning of Table 9.1. Although
the number of studies approaching gene–nutrient interactions as a factor in human
cancer development is limited, it is obvious that polymorphic genes of the P450
family have been investigated mostly in the context of association between cooked
red meat consumption and colorectal neoplasia. Several reports indicate possible
risk-modulating effects of CYP gene variants (see Table 9.1), which might be in
line with the idea of enhanced activation of meat processing-generated carcino-
gens, such as polycyclic aromatic hydrocarbons (PAH), heterocyclic amines
(HCA), and N-nitroso compounds by some variant CYP enzymes.70–72
However, reported effects and interactions are usually weak and often difficult
to reproduce. Moreover, the complexity of human diets and abundant presence
of multiple bioactive dietary components makes investigation of isolated
gene–nutrient interactions notoriously difficult. Furthermore, effects of different
gene polymorphisms often overlap as several examples given in Table 9.1 show.
For instance it is very difficult to separate influence of CYP family (especially
CYP1A1 and CYP1A2) and N-acetyltransferase (NAT1 and NAT2) genes on
colorectal cancer risk associated with cooked meat consumption. CYP family
genes encode numerous xenobiotic-metabolizing enzymes, but CYP3A4, being
the most abundant P450 enzyme in the human liver and gut73–75 and a major factor
in drug metabolism, deserves particular attention. Although over 70 SNPs are
recently described in the corresponding gene,75 knowledge of their physiological
significance is still very scarce. Correlations between CYP3A4 gene polymor-
phisms and cancer risk only start to emerge,75 and there is little doubt that further
investigation of this gene can produce abundant information on carcinogenesis,
responses to cancer treatment, and modulation of these processes by active com-
ponents of diet (see also Chapter 2, Hypolipidemic Therapy: Drugs, Diet, and
Interactive Effects).
Whereas polymorphic xenobiotic-metabolizing enzymes of Phase I are
mostly responsible for a generation of active carcinogenic substances, Phase II
metabolism is directed on detoxification of carcinogenic and mutagenic sub-
stances through conjugation reactions producing soluble and easily excretable
products. Inherited deficiency of some Phase II enzymes, such as “Null” variants
of glutathione S-transferases M1 (GSTM1) and T1 (GSTT1) caused by long
deletions in the corresponding genes, has repeatedly been reported to be associ-
ated with increased risk of cancers of different sites.76 GST family enzymes are
strongly involved in the metabolism of PAH,77 thus changes in their activity
are likely to affect the probability of cancer initiation. Table 9.1 shows that
266
TABLE 9.1
Interactions between Common “Metabolic” Gene Variants and Dietary Factors in Cancer Risk Modulation
Gene Tumor Site/Type Dietary Factor(s) Interactive Effects Refs.
CYP1A2 rapid phenotype is associated with certain genetic variants, e.g., (32)
CYP1A2*F.
Ovarian cancer Caffeine The associations of caffeine and coffee intake and ovarian cancer risk were (33)
stronger in women with CYP1A2 (high-rapid) genotype.
CYP2A6 Colorectal cancer Cooked red meat CYP2A6 high activity was associated with an increased risk of colorectal (34, 35)
cancer, particularly among consumers of well-done meat (see also effect
of GSTA1 genotype).
Information on the role CYP2A6 gene variants in the enzyme activity (36)
modulation is scarce; however, the effect appears to be likely.
CYP2E1 Colorectal cancer Red meat and Individuals with a 5’ 96-bp promoter region insertion variant of the CYP2E1 (37)
processed meat gene had an increased rectal cancer risk associated with high red meat or
processed meat consumption.
Nutrient–Drug Interactions
Incomplete intestinal Salted food (salted Association between incomplete intestinal metaplasia and salted food intake (38)
metaplasia (stomach meat, dehydrated was observed among subjects bearing homozygous c1/c1 CYP2E1 variant
cancer precursor) salted vegetables, (defined by RsaI polymorphism in 5′ region). (See also effects of GSTM1
salted raw fish) and GSTT1 genotypes.)
Upper aerodigestive Alcohol The highest risk of oral cavity/pharyngeal cancer was observed among the (39)
tract cancer heaviest drinkers with either the CYP2E1 C (defined by DraI
polymorphism in intron 6) or the CYP2E1 c2 (defined by RsaI
polymorphism in 5′ region) allele.
Myeloperoxidase Breast cancer Antioxidant-rich fruits The A-allele (G463A promoter region SNP) was associated with reduced (40)
(MPO) and vegetables breast cancer risk in women (especially premenopausal) who consumed
higher amounts of fruits and vegetables.
Genes Encoding Enzymes with Dual (Predominantly Phase II, but Some Phase I Metabolism) Function
DK5836_C009.fm Page 267 Monday, May 22, 2006 2:15 PM
N-acetyltransferase 1 Breast cancer Cooked red meat Elevated breast cancer risk related to increased well-done red meat (41, 42)
(NAT1) consumption was observed in women with putative rapid acetylation-
associated NAT1*11 or NAT1*10 alleles.
Colorectal adenomas Cooked red meat Stronger association of well-done red meat consumption with colorectal (43)
adenoma development was observed in individuals with rapid acetylation-
related NAT1 genotypes.
N-acetyltransferase 2 Colorectal cancer Cooked red meat Well-done red meat consumption was associated with an increased risk of (31)
(NAT2) colorectal cancer among ever-smokers with NAT2 rapid phenotype (and
Nutrigenomics and Pharmacogenomics of Human Cancer
Hepatocellular cancer Red meat A trend of increased hepatocellular carcinoma risk with increasing red meat (47)
intake was observed in rapid acetylators (defined here as subjects
homozygous for the wild type NAT2*4 allele).
Sulfotransferase 1A1 Breast cancer Red meat The risk of breast cancer was elevated with increasing doneness of red meat (48)
(SULT1A1) intake among women with the Arg/Arg or Arg/His genotype, but not with
the His/His homozygocity (lower enzyme activity-associated) at codon 213.
Prostate cancer An increased risk of prostate cancer was associated with the presence of the (49)
*1/*1 (His/His at 213) SULT1A1 genotype; however, there was no clear
DK5836_C009.fm Page 268 Monday, May 22, 2006 2:15 PM
genotype.
Prostate cancer Men with GSTM1-positive genotype had the greatest reduction of cancer (60)
risk.
Head and neck cancer Consumption of raw cruciferous vegetables was associated with cancer (61)
incidence reduction only in GSTM1-Null individuals.
Breast cancer Consumption of cruciferous vegetables, particularly broccoli, was (62)
marginally protective, but risk was unaffected by GSTM1 genotype.
Glutathione Lung cancer Red meat Among smokers with high animal fat and protein intake and low (52)
Nutrigenomics and Pharmacogenomics of Human Cancer
S-transferase P1 carbohydrate and fiber intake (“unhealthy” dietary pattern) individuals with
(GSTP1) Ile/Ile 105 homozygocity were at significantly higher lung cancer risk.
Glutathione Lung cancer Intake of Low intake of isothiocyanates was associated with marginally increased (53)
S-transferase T1 isothiocyanate-rich cancer risk in GSTT1-Null individuals.
(GSTT1) cruciferous
vegetables (esp.
broccoli)
269
270
dietary supplements antioxidant intake, were at higher risk of developing breast cancer.
No correlation of breast cancer risk with either MnSOD genotype or (67)
antioxidant intake.
Prostate cancer Beta-carotene Homozygocity for the Ala(-9) in combination with high antioxidant levels (68)
treatment, was associated with lower relative risk for both total and fatal prostate
antioxidants cancer.
Catalase (CAT) Breast cancer Fruit and vegetable The high-activity (wild type, CC) catalase homozygocity in combination (69)
consumption with higher vegetable and, especially, fruit consumption was associated
with breast cancer risk reduction.
Nutrient–Drug Interactions
DK5836_C009.fm Page 271 Monday, May 22, 2006 2:15 PM
China, Japan, and other populations of East Asia. Studies of other genes of this
group have shown that the presence of the low activity allele of the ADH3
(ADH3*2), in combination with alcohol drinking, appeared to result in elevated
risk for the development of upper digestive tract tumors.114–116 However, other
investigators reported no effect.117 Also, a higher risk of developing breast
cancer118 and colorectal adenomas119 was observed in individuals homozygous
for the ADH3*1, which is associated with increased activity of the enzyme. It
was also found that the presence of ADH1C “rapid” allele ADH1C*1 in alcoholics
was associated with increased risk of upper aerodigestive tract cancer.120
There is no doubt that obesity and related disorders, especially metabolic syn-
drome,9,136 may increase cancer risk. While caloric restriction is believed to be
cancer-protective,127 overeating is regarded as a major avoidable cause of cancer,
and there is sufficient evidence linking obesity with cancer of the colon, breast,
endometrium, kidney, and esophagus.23,137 Although regulation of energy balance
now attracts considerable attention of the scientific community,9,138 the knowledge
of its involvement in cancer pathogenesis remains at best fragmentary.
Recent studies of genes encoding factors regulating food/energy intake have
revealed the existence of multiple polymorphic variants.9 Publications suggesting
impact of some of these variants in cancer risk determination are just starting to
emerge. Among polymorphic genes encoding factors regulating food/energy
intake leptin gene variants have been demonstrated to be associated with suscep-
tibility to prostate cancer139 and non-Hodgkin lymphomas;139,140 in the latter case,
a leptin receptor gene SNP has been shown to be interactive.140,141 Polymorphisms
in the ghrelin and neuropeptide Y genes also affected non-Hodgkin lymphoma
risk.142 Variation of the gene encoding melanocortin-1 receptor (an important
factor in central regulation of energy intake) is proven to be a factor in skin cancer
and melanoma risk,143 but these links may reflect the key role of the gene in skin
pigmentation regulation. Moreover, there is a growing body of evidence linking
polymorphisms in the insulin and, especially, insulin-like growth factor pathway
genes with colorectal,144,145 breast,146 lung,147 prostate,148,149 and oral cavity150
cancers.
Modulation of cancer risk has also been reported for genes encoding proteins
involved in energy expenditure control; however this effect may not be directly
related to nutritional factors. Briefly, there were reports linking polymorphisms
in the 2- and 3-adrenergic receptor genes with susceptibility to breast,151 colorec-
tal,152 and endometrial153 cancer. Multifunctional factors of the arachidonic acid
pathway, in particular, peroxisome proliferator-activated receptor gamma (PPAR)
can be mentioned here as well, since PPAR polymorphisms have been shown to
affect risk of colorectal tumours,154–156 bladder,157 renal,158 and endometrial159
cancer. Interestingly, there appeared to be colorectal cancer risk-affecting inter-
actions between polymorphic PPAR gene variants and diet155,156; however, these
findings require confirmation.
Limits of this chapter do not allow extensive analysis of this complex
field, ramifications of which expand far beyond the area of nutrigenetics and
nutrigenomics.
Other regulatory and metabolic systems are certainly involved in cancer patho-
genesis, but it is very difficult to analyze them in relation to gene–nutrient
DK5836_C009.fm Page 275 Monday, May 22, 2006 2:15 PM
TABLE 9.2
Polymorphic Genes Encoding Factors Affecting Outcome of Cancer Chemotherapy and Radiotherapy
Therapeutic Agents Reported Clinical Effects or Functional Importance
Gene Malignancy Site and Type (Chemo- or Radio-) (References given in parentheses)
Ovarian cancer Cisplatinum, alkylating agents Combination of GSTM1-Null and GSTT1-Null was
associated with better responses to chemotherapy (188).
Paclitaxel, cisplatinum The GSTM1-Null genotype was associated with
significantly longer survival time (189).
Several chemotherapy schemes Patients carrying the GSTM1-Null were less likely to
have disease progression (190).
Malignant glioma Nitrosoureas The GSTM1-Null genotype was associated with better
DK5836_C009.fm Page 280 Monday, May 22, 2006 2:15 PM
Stomach cancer 5-FU/cisliplatinum chemotherapy The presence of the Val105 allele was associated with
significantly improved survival (203).
Malignant glioma Nitrosoureas Homozygous simultaneous presence of Ile105 and
Ala114 (Ala114Val) in combination with GSTM1-Null
was associated with better survival, but higher
probability of adverse effects (191).
Childhood acute lymphoblastic Multiagent chemotherapy The presence of the Val105 homozygocity was associated
DK5836_C009.fm Page 282 Monday, May 22, 2006 2:15 PM
Childhood acute lymphoblastic Methotrexate The presence of the T677T + A1298 (A1298G SNP)
leukemia (ALL) haplotype (especially in combination with TS high-
activity genotype) was associated with reduced event-
free survival (224).
The presence of the T677 allele was associated with
higher risk of relapse (225).
Thymidylate synthase (TS) Colorectal cancer 5-FU-based chemotherapy schemes, The presence of TS genotypes associated with high activity
DK5836_C009.fm Page 284 Monday, May 22, 2006 2:15 PM
Oxaliplatin/5-FU The presence of the AAT codon 118 allele was associated
with higher chemotherapy response rate (242).
Xeroderma pigmentosum Non-small-cell lung cancer Platinum-based chemotherapy The presence of the Asn312 allele (Asp312Asn XPD
group D (XPD or ERCC2) polymorphism) was associated with reduced survival
(243).
Colorectal cancer Oxaliplatin/5FU The presence of the Lys751 homozygocity (Lys751Gln
XPD polymorphism) was associated with better
DK5836_C009.fm Page 286 Monday, May 22, 2006 2:15 PM
survival (244).
Acute myeloid leukemia Multiagent chemotherapy The presence of the Lys751 homozygocity was
associated with better survival (245).
Excision repair cross Superficial bladder cancer Bacillus Calmette–Guerin treatment The presence of the Val1097 allele (Met1097Val ERCC6
complementation group 6 polymorphism) was associated with reduced
(ERCC6) recurrence-free survival (246).
Xeroderma pigmentosum Superficial bladder cancer Bacillus Calmette–Guerin treatment The presence of the A-4 allele (G-4A SNP in the 5′
group A (XPA) untranslated region) was associated with reduced
recurrence-free survival (246).
X-ray repair cross Colorectal cancer Oxaliplatin/5-FU The presence of the Gln399 allele (Arg399Gln XRCC1
complementation group 1 polymorphism) was associated with resistance to
(XRCC1) chemotherapy and reduced survival (247).
Breast cancer Radiotherapy The presence of the Trp(194) allele (Arg194Trp XRCC1
polymorphism) was associated with adverse responses
to radiotherapy (248).
Nutrient–Drug Interactions
The presence of the Gln399 allele was associated with
protection against adverse effects of radiotherapy (249).
Non-small-cell lung cancer Radiotherapy Combined analysis of polymorphisms at codons 194,
280, and 399 has shown correlations between XRCC1
haplotypes and survival (250).
Therapy-related acute myeloid Radiotherapy The presence of the Gln399 allele was associated with a
leukemia (t-AML) decreased risk of developing t-AML (251).
Apurinic/apyrimidinic Breast cancer Radiotherapy The presence of the Glu148 allele (Asp148Glu APE1
endonuclease 1 polymorphism) was associated with protection against
(APE1/APEX1) the development of acute adverse effects of
radiotherapy (249).
DNA mismatch repair gene Therapy-related acute myeloid Cyclophosphamide procarbazine The presence of the (-6)C allele (T-6C SNP at the 3′
hMSH2 leukemia (t-AML) splice acceptor site of exon 13) was associated with an
DK5836_C009.fm Page 287 Monday, May 22, 2006 2:15 PM
(257).
Nasopharyngeal carcinoma Irinotecan Higher irinotecan to SN-38 conversion of was related to
the absence of CTCA deletion in the 5′ flanking region
(256).
Polymorphic Genes Encoding Other Drug Targets (Receptors, Regulatory Factors, etc.)
Epidermal growth factor Colorectal cancer (rectum) Adjuvant and neoadjuvant Patients with Arg497 homozygocity (Arg497Lys SNP)
receptor (EGFR or HER-1) chemoradiation or lower number (<20) of CA repeats (Inthron 1 CAn
repeat polymorphism) had a higher recurrence risk
(258).
Colorectal cancer 5-FU/oxaliplatin Patients with less than 20 CA repeats were more likely
to show disease progression (259).
HER-2 receptor (HER-2, c-erb- Ovarian cancer Cisplatin, paclitaxel The presence of Val655 homozygocity (Ile655Val
B2 or neu) polymorphism) was associated with reduced overall
survival (260).
Nutrient–Drug Interactions
Immunoglobulin G Fc receptor Follicular lymphoma Rituximab The presence of His131 homozygocity (Arg131His
II (FcRIIa, CD32) polymorphism) was associated with better response rate
and freedom from progression (261).
Immunoglobulin G Fc receptor Follicular lymphoma Rituximab The presence of Val158 homozygocity (Phe158Val
III (FcRIIIa, CD16) polymorphism) was associated with better response rate
and freedom from progression (261).
Id vaccination The presence of Val158 homozygocity was associated
with longer progression-free survival (262).
Interleukin-1β (IL-1B) Stomach cancer Palliative chemotherapy The presence of T-511 (C-511T SNP) and C-31 (T-31C
SNP) was associated with reduced progression-free and
overall survival, but only in patients with wild-type
genotype of interleukin-1 receptor antagonist (IL-RN)
(263).
DK5836_C009.fm Page 289 Monday, May 22, 2006 2:15 PM
Interleukin-6 (IL-6) Breast cancer Anthracycline-based adjuvant The presence of the C-174 allele (G-174C SNP) was
chemotherapy and multiagent high- associated with better disease-free and overall survival
dose chemotherapy (264).
Interleukin 8 receptor CXCR1 Colorectal cancer 5-FU/oxaliplatin Patients with CXCR1 2607 heterozygocity (G2607C
SNP) were more likely to show disease progression
(254).
Interferon-γ (IFN-γ) Melanoma Cisplatin, vinblastine, and dacarbazine The presence of T834 allele (A874T SNP) was
combined with interleukin-2 and associated with better response, progression-free, and
Nutrigenomics and Pharmacogenomics of Human Cancer
9.4 CONCLUSION
The presented overview of pharmacogenomic studies highlights a few impressive
examples of successful utilization of information regarding genetic profiles of
oncological patients in the development of individual-oriented (“tailored”) che-
motherapeutic strategies. These examples include interactions between UGT1A1
polymorphisms and treatment with irinotecan as well as TPMT variants and
thiopurine chemotherapy. Furthermore, promising results have already emerged
from studies of the effects of thymydilate synthase alleles on fluoropyrimidine-
based chemotherapy, influence of ERCC1 genotype on platinum resistance, and
effects of GST-family gene genotypes in different schemes of cytostatic treatment.
This chapter was focused predominantly on the role of inherited structural DNA
variations (polymorphisms); however, it should be understood that other mecha-
nisms, especially changes in gene expression in the malignant tissue, are not less
important in terms of the development of individually tailored schemes of cancer
therapy. There is no doubt that clinical significance of pharmacogenomic infor-
mation is going to increase rapidly with introduction of modern genome-wide
analytical approaches.
In contrast, nutrigenomics of cancer is only starting to come into existence.
Although serious attempts to reveal links between carcinogenesis and nutrition
have been undertaken for decades, making firm conclusions remains difficult.
Cooked red meat consumption appears to be the only dietary influence, which
can be regarded as a proven risk factor for several types of cancer. A number of
nutrients have been shown to have cancer-preventive potential, but further studies
are needed to develop reliable strategies for their wide application. Introduction
of nutrigenomic approaches into this area has brought little clarification so far;
thus, full realization of potential strengths of this approach belongs to the future.
Parallel analysis of genetic factors involved in the metabolism of nutritional
agents and anticancer drugs reveals a striking similarity, which is not surprising,
given the plant-derived nature of many anticancer agents.266 This similarity results
in multiple opportunities for drug–nutrient interactions that is further modulated
by variants of gene structure and expression (see schematic representation in
Figure 9.2). Some situations of nutrient–drug interactions are recognized and
partially investigated, which is exemplified by studies on modulation of drug
transport and metabolism (MDR-1 and P-glycoprotein system and CYP3A
enzymes, respectively) by bioactive substances present in grapefruit juice or St.
John’s wort. However, significance of these known interactions for anticancer
therapy remains obscure. Only further studies in this direction will help to elu-
cidate their importance.
Although it is obvious that successful development of nutrigenomics is com-
plicated by many unsolved problems, the breathtaking speed of the technical
progress introduces numerous new powerful research tools in the field. This
innovation, along with the revision of some inefficient old investigative
approaches in nutritional science, brings expectations that serious breakthroughs
await us in the near future.
DK5836_C009.fm Page 295 Monday, May 22, 2006 2:15 PM
Biosystem (organism)
Genetic background
Targets
E
DIET
E
E
T1 T2
Phase I Phase II
interactions interactions interactions
T1 T2
DRUG E
E
E
Targets
Genetic background
REFERENCES
1. Peltonen L, McKusick VA. Genomics and medicine. Dissecting human disease in
the postgenomic era. Science 2001; 291: 1224–1229.
2. Guttmacher AE, Collins FS. Genomic medicine — a primer. N Engl J Med 2002;
347: 1512–1520.
3. Elliott R, Ong TJ. Nutritional genomics. BMJ. 2002; 324: 1438–1442.
4. Müller M, Kersten S. Nutrigenomics: goals and strategies. Nat Rev Genet 2003;
3: 912–920.
5. German JB, Roberts MA, Watkins SM. Genomics and metabolomics as markers
for the interaction of diet and health: lessons from lipids. J Nutr 2003; 133 (Suppl
1): 2078S–2083S.
6. Kaput J, Rodriguez RL. Nutritional genomics: the next frontier in the postgenomic
era. Physiol Genomics 2004; 16: 166–177.
7. Nagy R, Sweet K, Eng C. Highly penetrant hereditary cancer syndromes. Onco-
gene 2004; 23: 6445–6470.
DK5836_C009.fm Page 296 Monday, May 22, 2006 2:15 PM
24. Prentice RL, Willett WC, Greenwald P, Alberts D, Bernstein L, Boyd NF, Byers
T, Clinton SK, Fraser G, Freedman L, Hunter D, Kipnis V, Kolonel LN, Kristal
BS, Kristal A, Lampe JW, McTiernan A, Milner J, Patterson RE, Potter JD, Riboli
E, Schatzkin A, Yates A, Yetley E. Nutrition and physical activity and chronic
disease prevention: research strategies and recommendations. J Natl Cancer Inst
2004; 96: 1276–1287.
25. Vineis P, Malats N, Lang M, d’Errico A, Caporaso N, Cuzick J, Boffetta P. (Eds.)
Metabolic Polymorpisms and Susceptibility to Cancer. International Agency for
Research on Cancer (IARC) Scientific Publication 148. Lyon: IARC, 2000.
26. Nebert DW, Russell DW. Clinical importance of the cytochromes P450. Lancet
2002; 360: 1155–1162.
27. Guengerich FP. Cytochrome P450: what have we learned and what are the future
issues? Drug Metab Rev 2004; 36: 159–197.
28. Ingelman-Sundberg M. Polymorphism of cytochrome P450 and xenobiotic toxic-
ity. Toxicology 2002; 181–182: 447–452.
29. Terry KL, Titus-Ernstoff L, Garner EO, Vitonis AF, Cramer DW. Interaction
between CYP1A1 polymorphic variants and dietary exposures influencing ovarian
cancer risk. Cancer Epidemiol Biomarkers Prev 2003; 12: 187–190.
30. Murtaugh MA, Sweeney C, Ma K, Caan BJ, Slattery ML. The CYP1A1 genotype
may alter the association of meat consumption patterns and preparation with the
risk of colorectal cancer in men and women. J Nutr 2005; 135: 179–186.
31. Le Marchand L, Hankin JH, Wilkens LR, Pierce LM, Franke A, Kolonel LN,
Seifried A, Custer LJ, Chang W, Lum-Jones A, Donlon T. Combined effects of
well-done red meat, smoking, and rapid N-acetyltransferase 2 and CYP1A2 phe-
notypes in increasing colorectal cancer risk. Cancer Epidemiol Biomarkers Prev
2001; 10: 1259–1266.
32. Sachse C, Brockmöller J, Bauer S, Roots I. Functional significance of CA poly-
morphism in intron 1 of the cytochrome P450 1A2 (CYP1A2) gene tested with
caffeine. Br J Clin Pharmacol 1999; 47: 445–449.
33. Goodman MT, Tung KH, McDuffie K, Wilkens LR, Donlon TA. Association of
caffeine intake and CYP1A2 genotype with ovarian cancer. Nutr Cancer 2003;
46: 23–29.
34. Sweeney C, Coles BF, Nowell S, Lang NP, Kadlubar FF. Novel markers of
susceptibility to carcinogens in diet: associations with colorectal cancer. Toxicol-
ogy 2002; 181–182: 83–87.
35. Nowell S, Coles B, Sinha R, MacLeod S, Luke Ratnasinghe D, Stotts C, Kadlubar
FF, Ambrosone CB, Lang NP. Analysis of total meat intake and exposure to
individual heterocyclic amines in a case-control study of colorectal cancer: con-
tribution of metabolic variation to risk. Mutat Res 2002; 30: 175–185.
36. Sachse C, Smith G, Wilkie MJ, Barrett JH, Waxman R, Sullivan F, Forman D,
Bishop DT, Wolf CR; Colorectal Cancer Study Group. A pharmacogenetic study
to investigate the role of dietary carcinogens in the etiology of colorectal cancer.
Carcinogenesis 2002; 23: 1839–1849.
37. Le Marchand L, Donlon T, Seifried A, Wilkens LR. Red meat intake, CYP2E1
genetic polymorphisms, and colorectal cancer risk. Cancer Epidemiol Biomarkers
Prev 2002; 11: 1019–1024.
DK5836_C009.fm Page 298 Monday, May 22, 2006 2:15 PM
38. Chen SY, Liu TY, Shun CT, Wu MS, Lu TH, Lin JT, Sheu JC, Santella RM, Chen
CJ. Modification effects of GSTM1, GSTT1 and CYP2E1 polymorphisms on
associations between raw salted food and incomplete intestinal metaplasia in a
high-risk area of stomach cancer. Int J Cancer 2004; 108: 606–612.
39. Bouchardy C, Hirvonen A, Coutelle C, Ward PJ, Dayer P, Benhamou S. Role of
alcohol dehydrogenase 3 and cytochrome P-4502E1 genotypes in susceptibility
to cancers of the upper aerodigestive tract. Int J Cancer 2000; 87: 734–740.
40. Ahn J, Gammon MD, Santella RM, Gaudet MM, Britton JA, Teitelbaum SL, Terry
MB, Neugut AI, Josephy PD, Ambrosone CB. Myeloperoxidase genotype, fruit
and vegetable consumption, and breast cancer risk. Cancer Res 2004; 64:
7634–7639.
41. Zheng W, Deitz AC, Campbell DR, Wen WQ, Cerhan JR, Sellers TA, Folsom AR,
Hein DW. N-acetyltransferase 1 genetic polymorphism, cigarette smoking, well-
done meat intake, and breast cancer risk. Cancer Epidemiol Biomarkers Prev
1999; 8: 233–239.
42. Krajinovic M, Ghadirian P, Richer C, Sinnett H, Gandini S, Perret C, Lacroix A,
Labuda D, Sinnett D. Genetic susceptibility to breast cancer in French-Canadians:
role of carcinogen-metabolizing enzymes and gene-environment interactions. Int
J Cancer 2001; 92: 220–225.
43. Ishibe N, Sinha R, Hein DW, Kulldorff M, Strickland P, Fretland AJ, Chow WH,
Kadlubar FF, Lang NP, Rothman N. Genetic polymorphisms in heterocyclic amine
metabolism and risk of colorectal adenomas. Pharmacogenetics 2002; 12:
145–150.
44. Tiemersma EW, Voskuil DW, Bunschoten A, Hogendoorn EA, Witteman BJ,
Nagengast FM, Glatt H, Kok FJ, Kampman E. Risk of colorectal adenomas in
relation to meat consumption, meat preparation, and genetic susceptibility in a
Dutch population. Cancer Causes Control 2004; 15: 225–236.
45. Deitz AC, Zheng W, Leff MA, Gross M, Wen WQ, Doll MA, Xiao GH, Folsom
AR, Hein DW. N-Acetyltransferase-2 genetic polymorphism, well-done meat
intake, and breast cancer risk among postmenopausal women. Cancer Epidemiol
Biomarkers Prev 2000; 9: 905–910.
46. Delfino RJ, Sinha R, Smith C, West J, White E, Lin HJ, Liao SY, Gim JS, Ma
HL, Butler J, Anton-Culver H. Breast cancer, heterocyclic aromatic amines from
meat and N-acetyltransferase 2 genotype. Carcinogenesis 2000; 21: 607–615.
47. Huang YS, Chern HD, Wu JC, Chao Y, Huang YH, Chang FY, Lee SD. Polymor-
phism of the N-acetyltransferase 2 gene, red meat intake, and the susceptibility
of hepatocellular carcinoma. Am J Gastroenterol 2003; 98: 1417–1422.
48. Zheng W, Xie D, Cerhan JR, Sellers TA, Wen W, Folsom AR. Sulfotransferase
1A1 polymorphism, endogenous estrogen exposure, well-done meat intake, and
breast cancer risk. Cancer Epidemiol Biomarkers Prev 2001; 10: 89–94.
49. Nowell S, Ratnasinghe DL, Ambrosone CB, Williams S, Teague-Ross T, Trimble
L, Runnels G, Carrol A, Green B, Stone A, Johnson D, Greene G, Kadlubar FF,
Lang NP. Association of SULT1A1 phenotype and genotype with prostate cancer
risk in African-Americans and Caucasians. Cancer Epidemiol Biomarkers Prev
2004; 13: 270–276.
50. Cortessis V, Siegmund K, Chen Q, Zhou N, Diep A, Frankl H, Lee E, Zhu QS,
Haile R, Levy D. A case-control study of microsomal epoxide hydrolase, smoking,
meat consumption, glutathione S-transferase M3, and risk of colorectal adenomas.
Cancer Res 2001; 61: 2381–2385.
DK5836_C009.fm Page 299 Monday, May 22, 2006 2:15 PM
51. Ulrich CM, Bigler J, Whitton JA, Bostick R, Fosdick L, Potter JD. Epoxide
hydrolase Tyr113His polymorphism is associated with elevated risk of colorectal
polyps in the presence of smoking and high meat intake. Cancer Epidemiol
Biomarkers Prev 2001; 10: 875–882.
52. Tsai YY, McGlynn KA, Hu Y, Cassidy AB, Arnold J, Engstrom PF, Buetow KH.
Genetic susceptibility and dietary patterns in lung cancer. Lung Cancer 2003; 41:
269–281.
53. Spitz MR, Duphorne CM, Detry MA, Pillow PC, Amos CI, Lei L, de Andrade
M, Gu X, Hong WK, Wu X. Dietary intake of isothiocyanates: evidence of a joint
effect with glutathione S-transferase polymorphisms in lung cancer risk. Cancer
Epidemiol Biomarkers Prev 2000; 9: 1017–1020.
54. London SJ, Yuan JM, Chung FL, Gao YT, Coetzee GA, Ross RK, Yu MC.
Isothiocyanates, glutathione S-transferase M1 and T1 polymorphisms, and lung-
cancer risk: a prospective study of men in Shanghai, China. Lancet 2000; 356:
724–729.
55. Zhao B, Seow A, Lee EJ, Poh WT, Teh M, Eng P, Wang YT, Tan WC, Yu MC,
Lee HP. Dietary isothiocyanates, glutathione S-transferase -M1, -T1 polymor-
phisms and lung cancer risk among Chinese women in Singapore. Cancer Epi-
demiol Biomarkers Prev 2001; 10: 1063–1067.
56. Wang LI, Giovannucci EL, Hunter D, Neuberg D, Su L, Christiani DC. Dietary
intake of cruciferous vegetables, Glutathione S-transferase (GST) polymorphisms
and lung cancer risk in a Caucasian population. Cancer Causes Control 2004; 15:
977–985.
57. Slattery ML, Kampman E, Samowitz W, Caan BJ, Potter JD. Interplay between
dietary inducers of GST and the GSTM-1 genotype in colon cancer. Int J Cancer
2000; 87: 728–733.
58. Seow A, Yuan JM, Sun CL, Van Den Berg D, Lee HP, Yu MC. Dietary isothio-
cyanates, glutathione S-transferase polymorphisms and colorectal cancer risk in
the Singapore Chinese Health Study. Carcinogenesis 2002; 23: 2055–2061.
59. Lin HJ, Probst-Hensch NM, Louie AD, Kau IH, Witte JS, Ingles SA, Frankl HD,
Lee ER, Haile RW. Glutathione transferase null genotype, broccoli, and lower
prevalence of colorectal adenomas. Cancer Epidemiol Biomarkers Prev 1998; 7:
647–652.
60. Joseph MA, Moysich KB, Freudenheim JL, Shields PG, Bowman ED, Zhang Y,
Marshall JR, Ambrosone CB. Cruciferous vegetables, genetic polymorphisms in
glutathione S-transferases M1 and T1, and prostate cancer risk. Nutr Cancer 2004;
50: 206–213.
61. Gaudet MM, Olshan AF, Poole C, Weissler MC, Watson M, Bell DA. Diet, GSTM1
and GSTT1 and head and neck cancer. Carcinogenesis 2004; 25: 735–740.
62. Ambrosone CB, McCann SE, Freudenheim JL, Marshall JR, Zhang Y, Shields
PG. Breast cancer risk in premenopausal women is inversely associated with
consumption of broccoli, a source of isothiocyanates, but is not modified by GST
genotype. J Nutr 2004; 134: 1134–1138.
63. Yeh CC, Hsieh LL, Tang R, Chang-Chieh CR, Sung FC. Vegetable/fruit, smoking,
glutathione S-transferase polymorphisms and risk for colorectal cancer in Taiwan.
World J Gastroenterol 2005; 11: 1473–1480.
64. Turner F, Smith G, Sachse C, Lightfoot T, Garner RC, Wolf CR, Forman D, Bishop
DT, Barrett JH. Vegetable, fruit and meat consumption and potential risk modi-
fying genes in relation to colorectal cancer. Int J Cancer 2004; 112: 259–264.
DK5836_C009.fm Page 300 Monday, May 22, 2006 2:15 PM
65. Ambrosone CB, Freudenheim JL, Thompson PA, Bowman E, Vena JE, Marshall
JR, Graham S, Laughlin R, Nemoto T, Shields PG. Manganese superoxide dis-
mutase (MnSOD) genetic polymorphisms, dietary antioxidants, and risk of breast
cancer. Cancer Res 1999; 59: 602–606.
66. Cai Q, Shu XO, Wen W, Cheng JR, Dai Q, Gao YT, Zheng W. Genetic polymor-
phism in the manganese superoxide dismutase gene, antioxidant intake, and breast
cancer risk: results from the Shanghai Breast Cancer Study. Breast Cancer Res
2004; 6: R647–R655.
67. Tamimi RM, Hankinson SE, Spiegelman D, Colditz GA, Hunter DJ. Manganese
superoxide dismutase polymorphism, plasma antioxidants, cigarette smoking, and
risk of breast cancer. Cancer Epidemiol Biomarkers Prev 2004; 13: 989–996.
68. Li H, Kantoff PW, Giovannucci E, Leitzmann MF, Gaziano JM, Stampfer MJ,
Ma J. Manganese superoxide dismutase polymorphism, prediagnostic antioxidant
status, and risk of clinical significant prostate cancer. Cancer Res 2005; 65:
2498–2504.
69. Ahn J, Gammon MD, Santella RM, Gaudet MM, Britton JA, Teitelbaum SL, Terry
MB, Nowell S, Davis W, Garza C, Neugut AI, Ambrosone CB. Associations
between breast cancer risk and the catalase genotype, fruit and vegetable con-
sumption, and supplement use. Am J Epidemiol 2005; 162: 943–952.
70. Roberts-Thomson IC, Butler WJ, Ryan P. Meat, metabolic genotypes and risk for
colorectal cancer. Eur J Cancer Prev 1999; 8: 207–211.
71. Le Marchand L. Meat intake, metabolic genes and colorectal cancer. IARC Sci
Publ. 2002; 156: 481–485.
72. Cross AJ, Sinha R. Meat-related mutagens/carcinogens in the etiology of colorec-
tal cancer. Environ Mol Mutagen 2004; 44: 44–55.
73. Lamba JK, Lin YS, Schuetz EG, Thummel KE. Genetic contribution to variable
human CYP3A-mediated metabolism. Adv Drug Deliv Rev 2002; 54: 1271–1294.
74. Kaminsky LS, Zhang QY. The small intestine as a xenobiotic-metabolizing organ.
Drug Metab Dispos 2003; 31: 1520–1525.
75. Keshava C, McCanlies EC, Weston A. CYP3A4 polymorphisms—potential risk
factors for breast and prostate cancer: a HuGE review. Am J Epidemiol 2004; 160:
825–841.
76. Strange RC, Spiteri MA, Ramachandran S, Fryer AA. Glutathione-S-transferase
family of enzymes. Mutat Res 2001; 482: 21–26.
77. Cotton SC, Sharp L, Little J, Brockton N. Glutathione S-transferase polymor-
phisms and colorectal cancer: a HuGE review. Am J Epidemiol 2000; 151: 7–32.
78. Lampe JW, Peterson S. Brassica, biotransformation and cancer risk: genetic poly-
morphisms alter the preventive effects of cruciferous vegetables. J Nutr 2002;
132: 2991–2994.
79. Seow A, Vainio H, Yu MC. Effect of glutathione-S-transferase polymorphisms on
the cancer preventive potential of isothiocyanates: an epidemiological perspective.
Mutat Res 2005; 592: 58–67.
80. Lampe JW, Chen C, Li S, Prunty J, Grate MT, Meehan DE, Barale KV, Dightman
DA, Feng Z, Potter JD. Modulation of human glutathione S-transferases by botan-
ically defined vegetable diets. Cancer Epidemiol Biomarkers Prev 2000; 9:
787–793.
DK5836_C009.fm Page 301 Monday, May 22, 2006 2:15 PM
81. Wells PG, Mackenzie PI, Chowdhury JR, Guillemette C, Gregory PA, Ishii Y,
Hansen AJ, Kessler FK, Kim PM, Chowdhury NR, Ritter JK. Glucuronidation
and the UDP-glucuronosyltransferases in health and disease. Drug Metab Dispos
2004; 32: 281–290.
82. Basu NK, Ciotti M, Hwang MS, Kole L, Mitra PS, Cho JW, Owens IS. Differential
and special properties of the major human UGT1-encoded gastrointestinal UDP-
glucuronosyltransferases enhance potential to control chemical uptake. J Biol
Chem 2004; 279: 1429–1441.
83. Fang JL, Lazarus P. Correlation between the UDP-glucuronosyltransferase
(UGT1A1) TATAA box polymorphism and carcinogen detoxification phenotype:
significantly decreased glucuronidating activity against benzo(a)pyrene-7,8-dihy-
drodiol(-) in liver microsomes from subjects with the UGT1A1*28 variant. Cancer
Epidemiol Biomarkers Prev 2004; 13: 102–109.
84. Peterson S, Bigler J, Horner NK, Potter JD, Lampe JW. Cruciferae interact with
the UGT1A1*28 polymorphism to determine serum bilirubin levels in humans. J
Nutr 2005; 135: 1051–1055.
85. Girard H, Thibaudeau J, Court MH, Fortier LC, Villeneuve L, Caron P, Hao Q,
von Moltke LL, Greenblatt DJ, Guillemette C. UGT1A1 polymorphisms are
important determinants of dietary carcinogen detoxification in the liver. Hepatol-
ogy 2005; 42: 448–457.
86. Butler LM, Duguay Y, Millikan RC, Sinha R, Gagne JF, Sandler RS, Guillemette
C. Joint effects between UDP-glucuronosyltransferase 1A7 genotype and dietary
carcinogen exposure on risk of colon cancer. Cancer Epidemiol Biomarkers Prev
2005; 14: 1626–1632.
87. Klaunig JE, Kamendulis LM. The role of oxidative stress in carcinogenesis. Annu
Rev Pharmacol Toxicol 2004; 44: 239–267.
88. Friso S, Choi SW. Gene-nutrient interactions and DNA methylation. J Nutr 2002;
132(Suppl): 2382S–2387S.
89. Friso S, Choi SW, Girelli D, Mason JB, Dolnikowski GG, Bagley PJ, Olivieri O,
Jacques PF, Rosenberg IH, Corrocher R, Selhub J. A common mutation in the
5,10-methylenetetrahydrofolate reductase gene affects genomic DNA methylation
through an interaction with folate status. Proc Natl Acad Sci USA 2002; 99:
5606–5611.
90. Friso S, Girelli D, Trabetti E, Olivieri O, Guarini P, Pignatti PF, Corrocher R,
Choi SW. The MTHFR 1298A>C polymorphism and genomic DNA methylation
in human lymphocytes. Cancer Epidemiol Biomarkers Prev 2005; 14: 938–943.
91. Chen J, Gammon MD, Chan W, Palomeque C, Wetmur JG, Kabat GC, Teitelbaum
SL, Britton JA, Terry MB, Neugut AI, Santella RM. One-carbon metabolism,
MTHFR polymorphisms, and risk of breast cancer. Cancer Res 2005; 65:
1606–1614.
92. Yamada K, Chen Z, Rozen R, Matthews RG. Effects of common polymorphisms
on the properties of recombinant human methylenetetrahydrofolate reductase.
Proc Natl Acad Sci USA 2001; 98: 14853–14858.
93. Potter JD. Methyl supply, methyl metabolizing enzymes and colorectal neoplasia.
J Nutr 2002; 132(Suppl): 2410S–2412S.
94. Bailey LB. Folate, methyl-related nutrients, alcohol, and the MTHFR 677C–>T
polymorphism affect cancer risk: intake recommendations. J Nutr 2003; 133
(Suppl): 3748S–3753S.
DK5836_C009.fm Page 302 Monday, May 22, 2006 2:15 PM
124. van Gils CH, Bostick RM, Stern MC, Taylor JA. Differences in base excision
repair capacity may modulate the effect of dietary antioxidant intake on prostate
cancer risk: an example of polymorphisms in the XRCC1 gene. Cancer Epidemiol
Biomarkers Prev 2002; 11: 1279–1284.
125. Shen J, Terry MB, Gammon MD, Gaudet MM, Teitelbaum SL, Eng SM, Sagiv
SK, Neugut AI, Santella RM. MGMT genotype modulates the associations
between cigarette smoking, dietary antioxidants and breast cancer risk. Carcino-
genesis 2005; 26: 2131–2137.
126. Bonner MR, Rothman N, Mumford JL, He X, Shen M, Welch R, Yeager M,
Chanock S, Caporaso N, Lan Q. Green tea consumption, genetic susceptibility,
PAH-rich smoky coal, and the risk of lung cancer. Mutat Res 2005; 582: 53–60.
127. Hursting SD, Lavigne JA, Berrigan D, Perkins SN, Barrett JC. Calorie restriction,
aging, and cancer prevention: mechanisms of action and applicability to humans.
Annu Rev Med 2003; 54: 131–152.
128. Patel AC, Nunez NP, Perkins SN, Barrett JC, Hursting SD. Effects of energy
balance on cancer in genetically altered mice. J Nutr 2004; 134 (Suppl):
3394S–3398S.
129. Rao KS. Dietary calorie restriction, DNA-repair and brain aging. Mol Cell Bio-
chem 2003; 253: 313–318.
130. Stuart JA, Karahalil B, Hogue BA, Souza-Pinto NC, Bohr VA. Mitochondrial and
nuclear DNA base excision repair are affected differently by caloric restriction.
FASEB J 2004; 18: 595–597.
131. Seo YR, Sweeney C, Smith ML. Selenomethionine induction of DNA repair
response in human fibroblasts. Oncogene 2002; 21: 3663–3669.
132. Seo YR, Kelley MR, Smith ML. Selenomethionine regulation of p53 by a ref1-
dependent redox mechanism. Proc Natl Acad Sci USA 2002; 99: 14548–14553.
133. Collins AR, Harrington V, Drew J, Melvin R. Nutritional modulation of DNA
repair in a human intervention study. Carcinogenesis 2003; 24: 511–515.
134. Moller P, Loft S. Interventions with antioxidants and nutrients in relation to
oxidative DNA damage and repair. Mutat Res 2004; 551: 79–89.
135. Kotsopoulos J, Narod SA. Towards a dietary prevention of hereditary breast cancer.
Cancer Causes Control 2005; 16: 125–138.
136. Ukkola O, Bouchard C. Clustering of metabolic abnormalities in obese individu-
als: the role of genetic factors. Ann Med 2001; 33: 79–90.
137. Calle EE, Thun MJ. Obesity and cancer. Oncogene 2004; 23: 6365–6378.
138. Broberger C. Brain regulation of food intake and appetite: molecules and networks.
J Intern Med 2005; 258: 301–327.
139. Ribeiro R, Vasconcelos A, Costa S, Pinto D, Morais A, Oliveira J, Lobo F, Lopes
C, Medeiros R. Overexpressing leptin genetic polymorphism (-2548 G/A) is
associated with susceptibility to prostate cancer and risk of advanced disease.
Prostate 2004; 59: 268–274.
140. Skibola CF, Holly EA, Forrest MS, Hubbard A, Bracci PM, Skibola DR, Hegedus
C, Smith MT. Body mass index, leptin and leptin receptor polymorphisms, and
non-hodgkin lymphoma. Cancer Epidemiol Biomarkers Prev 2004; 13: 779–786.
141. Willett EV, Skibola CF, Adamson P, Skibola DR, Morgan GJ, Smith MT, Roman
E. Non-Hodgkin’s lymphoma, obesity and energy homeostasis polymorphisms.
Br J Cancer 2005; 93: 811–816.
DK5836_C009.fm Page 305 Monday, May 22, 2006 2:15 PM
142. Skibola DR, Smith MT, Bracci PM, Hubbard AE, Agana L, Chi S, Holly EA.
Polymorphisms in ghrelin and neuropeptide Y genes are associated with non-
Hodgkin lymphoma. Cancer Epidemiol Biomarkers Prev 2005; 14: 1251–1256.
143. Sturm RA. Skin colour and skin cancer — MC1R, the genetic link. Melanoma
Res 2002; 12: 405–416.
144. Morimoto LM, Newcomb PA, White E, Bigler J, Potter JD. Insulin-like growth
factor polymorphisms and colorectal cancer risk. Cancer Epidemiol Biomarkers
Prev 2005; 14: 1204–1211.
145. Slattery ML, Murtaugh M, Caan B, Ma KN, Neuhausen S, Samowitz W. Energy
balance, insulin-related genes and risk of colon and rectal cancer. Int J Cancer
2005; 115: 148–154.
146. Fletcher O, Gibson L, Johnson N, Altmann DR, Holly JM, Ashworth A, Peto J,
Silva Idos S. Polymorphisms and circulating levels in the insulin-like growth factor
system and risk of breast cancer: a systematic review. Cancer Epidemiol Biom-
arkers Prev 2005; 14: 2–19.
147. Moon JW, Chang YS, Ahn CW, Yoo KN, Shin JH, Kong JH, Kim YS, Chang J,
Kim SK, Kim HJ, Kim SK. Promoter -202 A/C polymorphism of insulin-like
growth factor binding protein-3 gene and non-small cell lung cancer risk. Int J
Cancer 2006; 118: 353–356.
148. Ho GY, Melman A, Liu SM, Li M, Yu H, Negassa A, Burk RD, Hsing AW,
Ghavamian R, Chua SC Jr. Polymorphism of the insulin gene is associated with
increased prostate cancer risk. Br J Cancer 2003; 88: 263–269.
149. Tsuchiya N, Wang L, Horikawa Y, Inoue T, Kakinuma H, Matsuura S, Sato K,
Ogawa O, Kato T, Habuchi T. CA repeat polymorphism in the insulin-like growth
factor-I gene is associated with increased risk of prostate cancer and benign
prostatic hyperplasia. Int J Oncol 2005; 26: 225–231.
150. Zavras AI, Pitiphat W, Wu T, Cartsos V, Lam A, Douglass CW, Diehl SR. Insulin-
like growth factor II receptor gene-167 genotype increases the risk of oral squa-
mous cell carcinoma in humans. Cancer Res 2003; 63: 296–297.
151. Huang XE, Hamajima N, Saito T, Matsuo K, Mizutani M, Iwata H, Iwase T, Miura
S, Mizuno T, Tokudome S, Tajima K. Possible association of beta-2 and beta-3
adrenergic receptor gene polymorphisms with susceptibility to breast cancer.
Breast Cancer Res 2001; 3: 264–269.
152. Takezaki T, Hamajima N, Matsuo K, Tanaka R, Hirai T, Kato T, Ohashi K, Tajima
K. Association of polymorphisms in the beta-2 and beta-3 adrenoceptor genes
with risk of colorectal cancer in Japanese. Int J Clin Oncol 2001; 6: 117–122.
153. Babol K, Przybylowska K, Lukaszek M, Pertynski T, Blasiak J. An association
between the Trp64Arg polymorphism in the beta3-adrenergic receptor gene and
endometrial cancer and obesity. J Exp Clin Cancer Res 2004; 23: 669–674.
154. Gong Z, Xie D, Deng Z, Bostick RM, Muga SJ, Hurley TG, Hebert JR. The
PPAR{gamma} Pro12Ala polymorphism and risk for incident sporadic colorectal
adenomas. Carcinogenesis 2005; 26: 579–585.
155. Siezen CL, van Leeuwen AI, Kram NR, Luken ME, van Kranen HJ, Kampman
E. Colorectal adenoma risk is modified by the interplay between polymorphisms
in arachidonic acid pathway genes and fish consumption. Carcinogenesis 2005;
26: 449–457.
DK5836_C009.fm Page 306 Monday, May 22, 2006 2:15 PM
156. Murtaugh MA, Ma KN, Caan BJ, Sweeney C, Wolff R, Samowitz WS, Potter JD,
Slattery ML. Interactions of peroxisome proliferator-activated receptor {gamma}
and diet in etiology of colorectal cancer. Cancer Epidemiol Biomarkers Prev 2005;
14: 1224–1229.
157. Leibovici D, Grossman HB, Dinney CP, Millikan RE, Lerner S, Wang Y, Gu J,
Dong Q, Wu X. Polymorphisms in inflammation genes and bladder cancer: from
initiation to recurrence, progression, and survival. J Clin Oncol 2005; 23:
5746–5756.
158. Smith WM, Zhou XP, Kurose K, Gao X, Latif F, Kroll T, Sugano K, Cannistra
SA, Clinton SK, Maher ER, Prior TW, Eng C. Opposite association of two PPARG
variants with cancer: overrepresentation of H449H in endometrial carcinoma cases
and underrepresentation of P12A in renal cell carcinoma cases. Hum Genet 2001;
109: 146–151.
159. Cao Y, Cao R, Brakenhielm E. Antiangiogenic mechanisms of diet-derived
polyphenols. J Nutr Biochem 2002; 13: 380–390.
160. Kanadaswami C, Lee LT, Lee PP, Hwang JJ, Ke FC, Huang YT, Lee MT. The
antitumor activities of flavonoids. In Vivo 2005; 19: 895–909.
161. Hoensch HP, Kirch W. Potential role of flavonoids in the prevention of intestinal
neoplasia: a review of their mode of action and their clinical perspectives. Int J
Gastrointest Cancer 2005; 35: 187–195.
162. Thompson HJ, McGinley JN, Spoelstra NS, Jiang W, Zhu Z, Wolfe P. Effect of
dietary energy restriction on vascular density during mammary carcinogenesis.
Cancer Res 2004; 64: 5643–5650.
163. Lambert JD, Hong J, Yang GY, Liao J, Yang CS. Inhibition of carcinogenesis by
polyphenols: evidence from laboratory investigations. Am J Clin Nutr 2005;
81(Suppl): 284S–291S.
164. Guy M, Lowe LC, Bretherton-Watt D, Mansi JL, Peckitt C, Bliss J, Wilson RG,
Thomas V, Colston KW. Vitamin D receptor gene polymorphisms and breast
cancer risk. Clin Cancer Res 2004; 10: 5472–5481.
165. Slattery ML, Neuhausen SL, Hoffman M, Caan B, Curtin K, Ma KN, Samowitz
W. Dietary calcium, vitamin D, VDR genotypes and colorectal cancer. Int J Cancer
2004; 111: 750–756.
166. Uitterlinden AG, Fang Y, Van Meurs JB, Pols HA, Van Leeuwen JP. Genetics and
biology of vitamin D receptor polymorphisms. Gene 2004; 338: 143–156.
167. Chen WY, Bertone-Johnson ER, Hunter DJ, Willett WC, Hankinson SE. Associ-
ations between polymorphisms in the vitamin D receptor and breast cancer risk.
Cancer Epidemiol Biomarkers Prev 2005; 14: 2335–2339.
168. Chew BP, Park JS. Carotenoid action on the immune response. J Nutr 2004; 134:
257S–261S.
169. Roynette CE, Calder PC, Dupertuis YM, Pichard C. n-3 polyunsaturated fatty
acids and colon cancer prevention. Clin Nutr 2004; 23: 139–151.
170. Nagasubramanian R, Innocenti F, Ratain MJ. Pharmacogenetics in cancer treat-
ment. Annu Rev Med 2003; 54: 437–452.
171. Ulrich CM, Robien K, McLeod HL. Cancer pharmacogenetics: polymorphisms,
pathways and beyond. Nat Rev Cancer 2003; 3: 912–920.
172. Loktionov A. Common gene polymorphisms, cancer progression and prognosis.
Cancer Lett 2004; 208: 1–33.
DK5836_C009.fm Page 307 Monday, May 22, 2006 2:15 PM
186. Nowell SA, Ahn J, Rae JM, Scheys JO, Trovato A, Sweeney C, MacLeod SL,
Kadlubar FF, Ambrosone CB. Association of genetic variation in tamoxifen-
metabolizing enzymes with overall survival and recurrence of disease in breast
cancer patients. Breast Cancer Res Treat 2005; 91: 249–258.
187. Ambrosone CB, Sweeney C, Coles BF, Thompson PA, McClure GY, Korourian
S, Fares MY, Stone A, Kadlubar FF, Hutchins LF. Polymorphisms in glutathione
S-transferases (GSTM1 and GSTT1) and survival after treatment for breast cancer.
Cancer Res 2001; 61: 7130–7135.
188. Howells RE, Holland T, Dhar KK, Redman CW, Hand P, Hoban PR, Jones PW,
Fryer AA, Strange RC. Glutathione S-transferase GSTM1 and GSTT1 genotypes
in ovarian cancer: association with p53 expression and survival. Int J Gynecol
Cancer 200; 11: 107–112.
189. Medeiros R, Pereira D, Afonso N, Palmeira C, Faleiro C, Afonso-Lopes C, Freitas-
Silva M, Vasconcelos A, Costa S, Osorio T, Lopes C. Platinum/paclitaxel-based
chemotherapy in advanced ovarian carcinoma: glutathione S-transferase genetic
polymorphisms as predictive biomarkers of disease outcome. Int J Clin Oncol
2003; 8: 156–161.
190. Beeghly A, Katsaros D, Chen H, Fracchioli S, Zhang Y, Massobrio M, Risch H,
Jones B, Yu H. Glutathione S-transferase polymorphisms and ovarian cancer
treatment and survival. Gynecol Oncol 2006; 100: 330–337.
191. Okcu MF, Selvan M, Wang LE, Stout L, Erana R, Airewele G, Adatto P, Hess K,
Ali-Osman F, Groves M, Yung AW, Levin VA, Wei Q, Bondy M. Glutathione S-
transferase polymorphisms and survival in primary malignant glioma. Clin Cancer
Res 2004; 10: 2618–2625.
192. Stanulla M, Schrappe M, Brechlin AM, Zimmermann M, Welte K. Polymorphisms
within glutathione S-transferase genes (GSTM1, GSTT1, GSTP1) and risk of
relapse in childhood B-cell precursor acute lymphoblastic leukemia: a case-control
study. Blood 2000; 95: 1222–1228.
193. Rocha JC, Cheng C, Liu W, Kishi S, Das S, Cook EH, Sandlund JT, Rubnitz J,
Ribeiro R, Campana D, Pui CH, Evans WE, Relling MV. Pharmacogenetics of
outcome in children with acute lymphoblastic leukemia. Blood 2005; 105:
4752–4758.
194. Davies SM, Bhatia S, Ross JA, Kiffmeyer WR, Gaynon PS, Radloff GA, Robison
LL, Perentesis JP. Glutathione S-transferase genotypes, genetic susceptibility, and
outcome of therapy in childhood acute lymphoblastic leukemia. Blood 2002; 100:
67–71.
195. Takanashi M, Morimoto A, Yagi T, Kuriyama K, Kano G, Imamura T, Hibi S,
Todo S, Imashuku S. Impact of glutathione S-transferase gene deletion on early
relapse in childhood B-precursor acute lymphoblastic leukemia. Haematologica
2003; 88: 1238–1244.
196. Voso MT, D’Alo’ F, Putzulu R, Mele L, Scardocci A, Chiusolo P, Latagliata R,
Lo-Coco F, Rutella S, Pagano L, Hohaus S, Leone G. Negative prognostic value
of glutathione S-transferase (GSTM1 and GSTT1) deletions in adult acute myeloid
leukemia. Blood 2002; 100: 2703–2707.
197. Davies SM, Robison LL, Buckley JD, Tjoa T, Woods WG, Radloff GA, Ross JA,
Perentesis JP. Glutathione S-transferase polymorphisms and outcome of chemo-
therapy in childhood acute myeloid leukaemia. J Clin Oncol 2001; 19: 1279–1287.
DK5836_C009.fm Page 309 Monday, May 22, 2006 2:15 PM
210. Relling MV, Hancock ML, Rivera GK, Sandlund JT, Ribeiro RC, Krynetski EY,
Pui CH, Evans WE. Mercaptopurine therapy intolerance and heterozygosity at the
thiopurine S-methyltransferase gene locus. J Natl Cancer Inst 1999; 91:
2001–2008.
211. Ando M, Ando Y, Hasegawa Y, Sekido Y, Shimokata K, Horibe K. Genetic
polymorphisms of thiopurine S-methyltransferase and 6-mercaptopurine toxicity
in Japanese children with acute lymphoblastic leukaemia. Pharmacogenetics
2001; 11: 269–273.
212. Evans WE, Hon YY, Bomgaars L, Coutre S, Holdsworth M, Janco R, Kalwinsky
D, Keller F, Khatib Z, Margolin J, Murray J, Quinn J, Ravindranath Y, Ritchey
K, Roberts W, Rogers ZR, Schiff D, Steuber C, Tucci F, Kornegay N, Krynetski
EY, Relling MV. Preponderance of thiopurine S-methyltransferase deficiency and
heterozygosity among patients intolerant to mercaptopurine or azathioprine. J Clin
Oncol 2001; 19: 2293–2301.
213. Stanulla M, Schaeffeler E, Flohr T, Cario G, Schrauder A, Zimmermann M, Welte
K, Ludwig WD, Bartram CR, Zanger UM, Eichelbaum M, Schrappe M, Schwab
M. Thiopurine methyltransferase (TPMT) genotype and early treatment response
to mercaptopurine in childhood acute lymphoblastic leukemia. JAMA 2005; 293:
1485–1489.
214. van Kuilenburg AB, Haasjes J, Richel DJ, Zoetekouw L, Van Lenthe H, De Abreu
RA, Maring JG, Vreken P, van Gennip AH. Clinical implications of dihydropyri-
midine dehydrogenase (DPD) deficiency in patients with severe 5-fluorouracil-
associated toxicity: identification of new mutations in the DPD gene. Clin Cancer
Res 2000; 6: 4705–4712.
215. Raida M, Schwabe W, Hausler P, Van Kuilenburg AB, Van Gennip AH, Behnke
D, Hoffken K. Prevalence of a common point mutation in the dihydropyrimidine
dehydrogenase (DPD) gene within the 5'-splice donor site of intron 14 in patients
with severe 5-fluorouracil (5-FU)- related toxicity compared with controls. Clin
Cancer Res 2001; 7: 2832–2839.
216. Van Kuilenburg AB, Meinsma R, Zoetekouw L, Van Gennip AH. High prevalence
of the IVS14 + 1G>A mutation in the dihydropyrimidine dehydrogenase gene of
patients with severe 5-fluorouracil-associated toxicity. Pharmacogenetics 2002
Oct; 12(7): 555–558.
217. Yonemori K, Ueno H, Okusaka T, Yamamoto N, Ikeda M, Saijo N, Yoshida T,
Ishii H, Furuse J, Sugiyama E, Kim SR, Kikura-Hanajiri R, Hasegawa R, Saito
Y, Ozawa S, Kaniwa N, Sawada J. Severe drug toxicity associated with a single-
nucleotide polymorphism of the cytidine deaminase gene in a Japanese cancer
patient treated with gemcitabine plus cisplatin. Clin Cancer Res 2005; 11:
2620–2624.
218. Shi JY, Shi ZZ, Zhang SJ, Zhu YM, Gu BW, Li G, Bai XT, Gao XD, Hu J, Jin
W, Huang W, Chen Z, Chen SJ. Association between single nucleotide polymor-
phisms in deoxycytidine kinase and treatment response among acute myeloid
leukaemia patients. Pharmacogenetics 2004; 14: 759–768.
219. Cohen V, Panet-Raymond V, Sabbaghian N, Morin I, Batist G, Rozen R. Meth-
ylenetetrahydrofolate reductase polymorphism in advanced colorectal cancer: a
novel genomic predictor of clinical response to fluoropyrimidine-based chemo-
therapy. Clin Cancer Res 2003; 9: 1611–1615.
DK5836_C009.fm Page 311 Monday, May 22, 2006 2:15 PM
243. Gurubhagavatula S, Liu G, Park S, Zhou W, Su L, Wain JC, Lynch TJ, Neuberg
DS, Christiani DC. XPD and XRCC1 genetic polymorphisms are prognostic
factors in advanced non-small-cell lung cancer patients treated with platinum
chemotherapy. J Clin Oncol 2004; 22: 2594–2601.
244. Park DJ, Stoehlmacher J, Zhang W, Tsao-Wei DD, Groshen S, Lenz HJ. A
xeroderma pigmentosum group D gene polymorphism predicts clinical outcome
to platinum-based chemotherapy in patients with advanced colorectal cancer.
Cancer Res 2001; 61: 8654–8658.
245. Allan JM, Smith AG, Wheatley K, Hills RK, Travis LB, Hill DA, Swirsky DM,
Morgan GJ, Wild CP. Genetic variation in XPD predicts treatment outcome and
risk of acute myeloid leukemia following chemotherapy. Blood 2004; 104:
3872–3877.
246. Gu J, Zhao H, Dinney CP, Zhu Y, Leibovici D, Bermejo CE, Grossman HB, Wu
X. Nucleotide excision repair gene polymorphisms and recurrence after treatment
for superficial bladder cancer. Clin Cancer Res 2005; 11: 1408–1415.
247. Stoehlmacher J, Ghaderi V, Iobal S, Groshen S, Tsao-Wei D, Park D, Lenz HJ.
A polymorphism of the XRCC1 gene predicts for response to platinum-based
treatment in advanced colorectal cancer. Anticancer Res 2001; 21: 3075–3079.
248. Moullan N, Cox DG, Angele S, Romestaing P, Gerard JP, Hall J. Polymorphisms
in the DNA repair gene XRCC1, breast cancer risk, and response to radiotherapy.
Cancer Epidemiol Biomarkers Prev 2003; 12: 1168–1174.
249. Chang-Claude J, Popanda O, Tan XL, Kropp S, Helmbold I, von Fournier D,
Haase W, Sautter-Bihl ML, Wenz F, Schmezer P, Ambrosone CB. Association
between polymorphisms in the DNA repair genes, XRCC1, APE1, and XPD and
acute side effects of radiotherapy in breast cancer patients. Clin Cancer Res 2005;
11: 4802–4809.
250. Yoon SM, Hong YC, Park HJ, Lee JE, Kim SY, Kim JH, Lee SW, Park SY, Lee
JS, Choi EK. The polymorphism and haplotypes of XRCC1 and survival of non-
small-cell lung cancer after radiotherapy. Int J Radiat Oncol Biol Phys. 2005; 63:
885–891.
251. Seedhouse C, Bainton R, Lewis M, Harding A, Russell N, Das-Gupta E. The
genotype distribution of the XRCC1 gene indicates a role for base excision repair
in the development of therapy-related acute myeloblastic leukemia. Blood 2002;
100: 3761–3766.
252. Worrillow LJ, Travis LB, Smith AG, Rollinson S, Smith AJ, Wild CP, Holowaty
EJ, Kohler BA, Wiklund T, Pukkala E, Roman E, Morgan GJ, Allan JM. An intron
splice acceptor polymorphism in hMSH2 and risk of leukemia after treatment
with chemotherapeutic alkylating agents. Clin Cancer Res 2003; 9: 3012–3020.
253. Illmer T, Schuler US, Thiede C, Schwarz UI, Kim RB, Gotthard S, Freund D,
Schakel U, Ehninger G, Schaich M. MDR1 gene polymorphisms affect therapy
outcome in acute myeloid leukemia patients. Cancer Res 2002; 62: 4955–4962.
254. Kafka A, Sauer G, Jaeger C, Grundmann R, Kreienberg R, Zeillinger R, Deissler
H. Polymorphism C3435T of the MDR-1 gene predicts response to preoperative
chemotherapy in locally advanced breast cancer. Int J Oncol 2003; 22: 1117–1121.
255. Jamroziak K, Mlynarski W, Balcerczak E, Mistygacz M, Trelinska J, Mirowski
M, Bodalski J, Robak T. Functional C3435T polymorphism of MDR1 gene: an
impact on genetic susceptibility and clinical outcome of childhood acute lympho-
blastic leukemia. Eur J Haematol 2004; 72: 314–321.
DK5836_C009.fm Page 314 Monday, May 22, 2006 2:15 PM
256. Zhou Q, Sparreboom A, Tan EH, Cheung YB, Lee A, Poon D, Lee EJ, Chowbay
B. Pharmacogenetic profiling across the irinotecan pathway in Asian patients with
cancer. Br J Clin Pharmacol 2005; 59: 415–424.
257. de Jong FA, Marsh S, Mathijssen RH, King C, Verweij J, Sparreboom A, McLeod
HL. ABCG2 pharmacogenetics: ethnic differences in allele frequency and assess-
ment of influence on irinotecan disposition. Clin Cancer Res 2004; 10: 5889–5894.
258. Zhang W, Park DJ, Lu B, Yang DY, Gordon M, Groshen S, Yun J, Press OA,
Vallbohmer D, Rhodes K, Lenz HJ. Epidermal growth factor receptor gene poly-
morphisms predict pelvic recurrence in patients with rectal cancer treated with
chemoradiation. Clin Cancer Res 2005; 11: 600–605.
259. Zhang W, Stoehlmacher J, Park DJ, Yang D, Borchard E, Gil J, Tsao-Wei DD,
Yun J, Gordon M, Press OA, Rhodes K, Groshen S, Lenz HJ. Gene polymorphisms
of epidermal growth factor receptor and its downstream effector, interleukin-8,
predict oxaliplatin efficacy in patients with advanced colorectal cancer. Clin Col-
orectal Cancer 2005; 5: 124–131.
260. Pinto D, Pereira D, Portela C, da Silva JL, Lopes C, Medeiros R. The influence
of HER2 genotypes as molecular markers in ovarian cancer outcome. Biochem
Biophys Res Commun 2005; 335: 1173–1178.
261. Weng WK, Levy R. Two immunoglobulin G fragment C receptor polymorphisms
independently predict response to rituximab in patients with follicular lymphoma.
J Clin Oncol 2003; 21: 3940–3947.
262. Weng WK, Czerwinski D, Timmerman J, Hsu FJ, Levy R. Clinical outcome of
lymphoma patients after idiotype vaccination is correlated with humoral immune
response and immunoglobulin G Fc receptor genotype. J Clin Oncol 2004; 22:
4717–4724.
263. Graziano F, Ruzzo A, Santini D, Humar B, Tonini G, Catalano V, Berardi R,
Pizzagalli F, Arduini F, Bearzi I, Scartozzi M, Cascinu S, Testa E, Ficarelli R,
Magnani M. Prognostic role of interleukin-1beta gene and interleukin-1 receptor
antagonist gene polymorphisms in patients with advanced gastric cancer. J Clin
Oncol 2005; 23: 2339–2345.
264. DeMichele A, Martin AM, Mick R, Gor P, Wray L, Klein-Cabral M, Athanasiadis
G, Colligan T, Stadtmauer E, Weber B. Interleukin-6 -174G–>C polymorphism
is associated with improved outcome in high-risk breast cancer. Cancer Res 2003;
63: 8051–8056.
265. Liu D, O'Day SJ, Yang D, Boasberg P, Milford R, Kristedja T, Groshen S, Weber
J. Impact of gene polymorphisms on clinical outcome for stage IV melanoma
patients treated with biochemotherapy: an exploratory study. Clin Cancer Res
2005; 11: 1237–1246.
266. McFadyen MC, Melvin WT, Murray GI. Cytochrome P450 enzymes: novel
options for cancer therapeutics. Mol Cancer Ther 2004; 3: 363–371.
267. Shimada T, Yamazaki H, Mimura M, Inui Y, Guengerich FP. Interindividual vari-
ations in human liver cytochrome P-450 enzymes involved in the oxidation of
drugs, carcinogens and toxic chemicals: studies with liver microsomes of 30
Japanese and 30 Caucasians. J Pharmacol Exp Ther 1994; 270: 414–423.
268. Kivisto KT, Niemi M, Fromm MF. Functional interaction of intestinal CYP3A4
and P-glycoprotein. Fundam Clin Pharmacol 2004; 18: 621–626.
269. Williams JA, Cook J, Hurst SI. A significant drug-metabolizing role for CYP3A5?
Drug Metab Dispos 2003; 31: 1526–1530.
DK5836_C009.fm Page 315 Monday, May 22, 2006 2:15 PM
270. Lamba JK, Lin YS, Thummel K, Daly A, Watkins PB, Strom S, Zhang J, Schuetz
EG. Common allelic variants of cytochrome P4503A4 and their prevalence in
different populations. Pharmacogenetics 2002; 12: 121–132.
271. Schuetz EG. Lessons from the CYP3A4 promoter. Mol Pharmacol 2004; 65:
279–281.
272. Matsumura K, Saito T, Takahashi Y, Ozeki T, Kiyotani K, Fujieda M, Yamazaki
H, Kunitoh H, Kamataki T. Identification of a novel polymorphic enhancer of the
human CYP3A4 gene. Mol Pharmacol 2004; 65: 326–334.
273. Xie HG, Wood AJ, Kim RB, Stein CM, Wilkinson GR. Genetic variability in
CYP3A5 and its possible consequences. Pharmacogenomics 2004; 5: 243–272.
274. Roy JN, Lajoie J, Zijenah LS, Barama A, Poirier C, Ward BJ, Roger M. CYP3A5
genetic polymorphisms in different ethnic populations. Drug Metab Dispos 2005;
33: 884–887.
275. Fujita K. Food-drug interactions via human cytochrome P450 3A (CYP3A). Drug
Metabol Drug Interact 2004; 20: 195–217.
276. Paine MF, Criss AB, Watkins PB. Two major grapefruit juice components differ
in time to onset of intestinal CYP3A4 inhibition. J Pharmacol Exp Ther 2005;
312: 1151–1160.
277. Veronese ML, Gillen LP, Burke JP, Dorval EP, Hauck WW, Pequignot E, Waldman
SA, Greenberg HE. Exposure-dependent inhibition of intestinal and hepatic
CYP3A4 in vivo by grapefruit juice. J Clin Pharmacol 2003; 43: 831–839.
278. Markowitz JS, Donovan JL, DeVane CL, Taylor RM, Ruan Y, Wang JS, Chavin
KD. Effect of St. John's wort on drug metabolism by induction of cytochrome
P450 3A4 enzyme. JAMA 2003; 290: 1500–1504.
279. Reif S, Nicolson MC, Bisset D, Reid M, Kloft C, Jaehde U, McLeod HL. Effect
of grapefruit juice intake on etoposide bioavailability. Eur J Clin Pharmacol 2002;
58: 491–494.
280. van Schaik RH. Implications of cytochrome P450 genetic polymorphisms on the
toxicity of antitumor agents. Ther Drug Monit 2004; 26: 236–240.
281. Marsh S, Xiao M, Yu J, Ahluwalia R, Minton M, Freimuth RR, Kwok PY, McLeod
HL. Pharmacogenomic assessment of carboxylesterases 1 and 2. Genomics 2004;
84: 661–668.
282. Rooseboom M, Commandeur JN, Vermeulen NP. Enzyme-catalyzed activation of
anticancer prodrugs. Pharmacol Rev 2004; 56: 53–102.
283. Salavaggione OE, Wang L, Wiepert M, Yee VC, Weinshilboum RM. Thiopurine
S-methyltransferase pharmacogenetics: variant allele functional and comparative
genomics. Pharmacogenet Genomics 2005; 15: 801–815.
284. Noguchi T, Tanimoto K, Shimokuni T, Ukon K, Tsujimoto H, Fukushima M,
Noguchi T, Kawahara K, Hiyama K, Nishiyama M. Aberrant methylation of
DPYD promoter, DPYD expression, and cellular sensitivity to 5-fluorouracil in
cancer cells. Clin Cancer Res 2004; 10: 7100–7107.
285. Ukon K, Tanimoto K, Shimokuni T, Noguchi T, Hiyama K, Tsujimoto H, Fuku-
shima M, Toge T, Nishiyama M. Activator protein accelerates dihydropyrimidine
dehydrogenase gene transcription in cancer cells. Cancer Res 2005; 65:
1055–1062.
286. Longley DB, Harkin DP, Johnston PG. 5-fluorouracil: mechanisms of action and
clinical strategies. Nat Rev Cancer 2003; 3: 330–338.
DK5836_C009.fm Page 316 Monday, May 22, 2006 2:15 PM
287. Sellers TA, Alberts SR, Vierkant RA, Grabrick DM, Cerhan JR, Vachon CM,
Olson JE, Kushi LH, Potter JD. High-folate diets and breast cancer survival in a
prospective cohort study. Nutr Cancer 2002; 44: 139–144.
288. Hooijberg JH, de Vries NA, Kaspers GJ, Pieters R, Jansen G, Peters GJ. Multidrug
resistance proteins and folate supplementation: therapeutic implications for anti-
folates and other classes of drugs in cancer treatment. Cancer Chemother Phar-
macol 2005; 1 (Online): 1–12.
289. Guminski AD, Harnett PR, deFazio A. Scientists and clinicians test their metal-
back to the future with platinum compounds. Lancet Oncol 2002; 3: 312–318.
290. Altaha R, Liang X, Yu JJ, Reed E. Excision repair cross complementing-group 1:
gene expression and platinum resistance. Int J Mol Med 2004; 14: 959–970.
291. Yu JJ, Mu C, Lee KB, Okamoto A, Reed EL, Bostick-Bruton F, Mitchell KC,
Reed E. A nucleotide polymorphism in ERCC1 in human ovarian cancer cell lines
and tumor tissues. Mutat Res 1997; 382: 13–20.
292. Yu JJ, Lee KB, Mu C, Li Q, Abernathy TV, Bostick-Bruton F, Reed E. Comparison
of two human ovarian carcinoma cell lines (A2780/CP70 and MCAS) that are
equally resistant to platinum, but differ at codon 118 of the ERCC1 gene. Int J
Oncol 2000; 16: 555–560.
293. Vonderheide AP, Wrobel K, Kannamkumarath SS, B'Hymer C, Montes-Bayon M,
Ponce De Leon C, Caruso JA. Characterization of selenium species in Brazil nuts
by HPLC-ICP-MS and ES-MS. J Agric Food Chem 2002; 50: 5722–5728.
294. Lockhart AC, Tirona RG, Kim RB. Pharmacogenetics of ATP-binding cassette
transporters in cancer and chemotherapy. Mol Cancer Ther 2003; 2: 685–698.
295. Benet LZ, Cummins CL, Wu CY. Unmasking the dynamic interplay between
efflux transporters and metabolic enzymes. Int J Pharm 2004; 277: 3–9.
296. Schwab M, Eichelbaum M, Fromm MF. Genetic polymorphisms of the human
MDR1 drug transporter. Annu Rev Pharmacol Toxicol 2003; 43: 285–307.
297. Sakaeda T, Nakamura T, Okumura K. Pharmacogenetics of MDR1 and its impact
on the pharmacokinetics and pharmacodynamics of drugs. Pharmacogenomics
2003; 4: 397–410.
298. Dahan A, Altman H. Food-drug interaction: grapefruit juice augments drug bio-
availability—mechanism, extent and relevance. Eur J Clin Nutr 2004; 58: 1–9.
299. Chearwae W, Wu CP, Chu HY, Lee TR, Ambudkar SV, Limtrakul P. Curcuminoids
purified from turmeric powder modulate the function of human multidrug resis-
tance protein 1 (ABCC1). Cancer Chemother Pharmacol 2006; 57: 376–388.
300. Dürr D, Stieger B, Kullak-Ublick GA, Rentsch KM, Steinert HC, Meier PJ,
Fattinger K. St. John's Wort induces intestinal P-glycoprotein/MDR1 and intestinal
and hepatic CYP3A4. Clin Pharmacol Ther 2000; 68: 598–604.
301. Adams GP, Weiner LM. Monoclonal antibody therapy of cancer. Nat Biotechnol
2005; 23: 1147–1157.
302. Zhang Z, Li M, Rayburn ER, Hill DL, Zhang R, Wang H. Oncogenes as novel
targets for cancer therapy (part I): growth factors and protein tyrosine kinases.
Am J Pharmacogenomics 2005; 5: 173–190.
303. Wakeling AE. Inhibitors of growth factor signalling. Endocr Relat Cancer 2005;
12 (Suppl): S183–S187.
304. Yan L, Beckman RA. Pharmacogenetics and pharmacogenomics in oncology
therapeutic antibody development. Biotechniques 2005; 39: 565–568.
305. Tisdale MJ. Cachexia in cancer patients. Nat Rev Cancer 2002; 2: 862–871.
DK5836_C009.fm Page 317 Monday, May 22, 2006 2:15 PM
306. Zigman JM, Elmquist JK. From anorexia to obesity—the yin and yang of body
weight control. Endocrinology 2003; 144: 3749–3756.
307. Inui A. Cancer anorexia-cachexia syndrome: current issues in research and man-
agement. CA Cancer J Clin 2002; 52: 72–91.
308. Rubin H. Cancer cachexia: its correlations and causes. Proc Natl Acad Sci USA.
2003; 100: 5384–5389.
309. Barber MD, Powell JJ, Lynch SF, Fearon KC, Ross JA. A polymorphism of the
interleukin-1 beta gene influences survival in pancreatic cancer. Br J Cancer 2000;
83: 1443–1447.
310. Halma MA, Wheelhouse NM, Barber MD, Powell JJ, Fearon KC, Ross JA.
Interferon-gamma polymorphisms correlate with duration of survival in pancreatic
cancer. Hum Immunol 2004; 65: 1405–1408.
DK5836_C009.fm Page 318 Monday, May 22, 2006 2:15 PM
DK5836_Index.fm Page 319 Monday, May 22, 2006 2:32 PM
Index
319
DK5836_Index.fm Page 320 Monday, May 22, 2006 2:32 PM
Index 321
Brain-derived neurotrophic factor (BDNF), 149 gene polymorphisms and dietary factors,
Brazil nuts, 292 265–271, See also Cancer
Breast cancer, 64, 96 nutrigenomics
antioxidants and, 101 green tea phytochemicals, 82
COX-2 overexpression and, 105 epidemiology, 96–97
diet and lifestyle risk factors, 97 oxidant stress and, 75
fatty acid supplementation effects on patient weight loss, 97
chemotherapy, 112 phytochemical induction of Phase II
flaxseed phytoestrogens and, 113 enzymes, 80–81
genetic polymorphisms and dietary factors, procarcinogenic phytochemicals, 77–78
267–270 public perception of risk, 83–85
mushroom polysaccharides and, 108 Cancer chemotherapy, 95–96, 106–107,
polymorphic genes and chemotherapy 263–264
outcomes, 278–282, 286, 287, 289 antioxidants and, 101–102
vitamin D and, 247 chemosensitizers, 102–103
Breast cancer resistance protein (BCRP), 288 drug resistance development, 102
Broccoli, 76, 80, 135, 138 genetic polymorphisms, drug interactions
Brussels sprouts, 76 and outcomes, 276–290, See also
Bupropion, 181 Cancer pharmacogenomics and
Buthionine sulphoximine, 131 pharmacogenetics
Butyrate, 105 interindividual response variation, 100
in vitro and in vivo bioavailability, 103
nutrigenomics, See Cancer nutrigenomics
C nutritional status and treatment outcomes,
97, 99–102
Cabbage, 76 beneficial dietary deficiencies, 114
Cachexia, 99–100, 107, 293 pharmacogenomics, 276–294, See also
Caffeic acid, 141 Cancer pharmacogenomics and
Caffeine, 79, 266 pharmacogenetics
Calbindins, 133 Cancer chemotherapy, adjuvant agents, 98–99
Calcium/calmodulin-dependent protein kinase antifolate chemotherapeutic agents, 291
II (CaMKII), 182, 191 flaxseed phytoestrogens, 113–114
Calcium channel blockers, 210, 215, See also gastrointestinal toxicity treatment, 100–101
Beta blockers hematopoietic toxicity treatment, 100
cancer chemotherapy chemosensitizer, 102 immunomodulators, 107–108
ginseng and, 225 monoclonal antibodies, 293
grapefruit juice and, 45, 46 NSAIDs, 105, 110
Calcium supplements, 210, 215 nutritional cocktails, 106
Caloric restriction, cancer preventive/protective pharmacogenomics, See Cancer
effects, 273, 274 pharmacogenomics and
Camptothecin, 113 pharmacogenetics
Cancer, 262, See also specific types polyunsaturated fatty acids, 108–113
angiogenesis modulation, 275 targets
body composition changes, 99 COX inhibition, 105–106, 110
cachexia, 99–100, 107, 293 drug transporting glycoprotein
calorie restriction and, 273, 274 interactions, 102–104
changes in nutritional status, 97, 99–101 transcription factors, 104–105
COX-2 overexpression and, 105–106 Cancer nutrigenomics, 246–252, 271–272
diet and lifestyle risk factors, 64, 96–97 alcohol/aldehyde dehydrogenase
dietary chemopreventative/chemoprotective polymorphisms, 272–273
factors, 73–75, 95–96, 263, See alcohol metabolism, 272–273
also Cancer chemotherapy, angiogenesis modulation, 275
adjuvant agents; specific nutrients diet, genetics, and risk, 263–264
or phytochemicals DNA repair enzymes, 273
DK5836_Index.fm Page 322 Monday, May 22, 2006 2:32 PM
Index 323
Index 325
Index 327
Index 329
Index 331
P Phenformin, 214
Phenobarbital, 72, 219, 224
Paclitaxel, 276, 277, 280, 288 Phenytoin, 216, 219, 224
Pancreatic cancer, 106 Phloretin, 76
Pancreatitis, 223 Phosphoinositide 3-kinase (PI 3-kinase), 141
Paradol, 222 Phospholipase A2, 141
Parkinson’s disease (PD), 130, 149, See also pH-sensitive drug absorption, 7
Neurodegenerative disorders Phytoestrogens, 113–114, 138, See also
antioxidants and, 134, 153, 157 Isoflavones
apoptotic mechanisms, 131, 152–153 Pioglitazone, 6, 9–10
cytoskeletal pathology, 151 Pitavastatin, 33, 45, 46
environmental factors and, 150–151 Platelet-derived growth factor (PDGF), 106
epidemiology and genetics, 150 Platinum-based chemotherapy, 281, 285, 286,
lifestyle and dietary habits and, 134 292, See also specific agents
mitochondrial dysfunction, 151 Policosanol, 43
neuroimaging and autopsy data, 153 Polycyclic aromatic hydrocarbons (PAHs),
oxidative stress and, 130, 151–153 63–70, 265
Paroxetine, 181 animal-based research, 68
Parsley, 138 cooked/processed meat, 65, 82
Parthenolide, 220 experimental models and future research,
PBN, 147 81–83
PCBs, 72 green tea phytochemicals and, 82
Pecans, 105 metabolism of, 69–70, 265
PEITC, 79 phytochemicals and formation during meat
Penicillamine, 155, 212, 213 processing, 82
Pernicious anemia, 213–214 tobacco smoke, 66, 75
Peroxisome proliferator-activated receptor, 9, xenobiotic metabolism, 64–68
34–35, 38, 72, 274 Polymorphic genes, See Genetic
Peroxynitrite, 131, 141 polymorphisms
P-glycoprotein, 45 Polyphenols, 131, 135, See also Flavonoids
chemopreventive agents and, 96 absorption and metabolism, 140–141
herb interactions, 5 apples, 136–137
phytochemical interactions and cancer biological actions, 141–142
chemotherapy, 102–104, 292–293 “French paradox,” 142–143
Pharmacogenomics, 262, 276, See also Cancer green tea, 143, See also Catechins
pharmacogenomics and red wine protective effects, 142
pharmacogenetics structures, 136
Pharyngitis treatment, 101 Polyunsaturated fatty acids (PUFAs), See also
Phase I xenobiotic metabolic reactions, 44–45 Omega-3 (ω-3) fatty acids;
cancer nutrigenomics and, 264–268 Omega-6 (ω-6) fatty acids
cancer pharmacogenomics and, 276 adjuvant chemotherapy, 108–113
meat carcinogen bioactivation, 64 beneficial effects, 39
phytochemical exposure and, 75–77 mood disorders and, 197
Phase II xenobiotic conjugation neurodegenerative diseases and, 149
cancer nutrigenomics and, 266–271 Potatoes, 137
cancer pharmacogenomics and, 276–290 Pramipexole, 153
hypolipidemic drug interactions, 44–45 Pranidipine, 46
phytochemical exposure and, 75–77 Pravastatin, 5, 17, 33, 45, 46, 47, See also Statins
phytochemical induction, 80–81 Pregnancy
secondary carcinogen metabolism, 66–70 iron levels and, 212
xenobiotic exposure and regulation of, magnesium-anesthetic interactions, 211
72–73 vitamin A and, 213
Phenelzine, 181, 195–196, 221, 225 8-Prenylnaringenin, 76
Phenethyl isothiocyanate, 76 Proanthocyanidins, 136, 138
DK5836_Index.fm Page 333 Monday, May 22, 2006 2:32 PM
Index 333
Procyanidins, 98, 104, 194 Red wine, 135, 138, 156, See also Wine
Proline, 106 consumption
Propafenine, 46 ALS and, 155
Propofol, 210 cardiovascular protective effects, 142
Prostaglandin D2, 36 flavonoids in, 135, 138
Prostaglandin E2, 105, 106, 111 neurodegenerative diseases and, 143
Prostate cancer, 64 stilbenes, 136
COX-2 and, 106 Red yeast rice, 44
DNA repair enzymes and, 273 Reduced folate carrier 1 (RFC1), 285
fatty acid metabolism, 110 Reductase inhibitors, 216
fatty acid supplementation effects on Repaglinide, 6, 12, 18
chemotherapy, 113 Resins (bile salt binders), 18
genetic polymorphisms and dietary factors, Resveratrol
268, 269, 270 ALS and, 156
leptin gene and, 274 chemotherapy adjuvant, 98, 106
mushroom glucan and, 114 protein kinase inhibition, 142
patient use of alternative medicine, 97 red wine protective effects, 142
vitamin D and, 247 stilbenes, 136
Protein, dietary, cancer risk and, 67 transcription factors and, 104
Protein kinase B, 141 xenobiotic metabolism and, 76, 77
Protein kinase C (PKC), 141, 182 Retinoid X receptor (RXR), 72
Protein kinases, 141, 182 Retinoids, 213
Protein oxidation, 132 Rifampin, 219
Proteolysis inducing factor (PIF), 99–100 Riluzole, 155
Pseudoephedrine hydrochloride, 217 Rituximab, 289, 293
Psoralens, 45 Rosiglitazone, 6, 9–10
Public risk perceptions, 83–85 Rosuvastatin, 17, 33, 45, 46
Purine/pyrimidine metabolism, 290–291 Rotenone, 151
Rutin, 79
Q
S
Quercetin, 76, 78, 79, 103, 135, 136, 138, 141,
142, 156 S-adenosylmethionine (SAM), 186, 190–191,
Quinidine, 46 200, 249
Quinolone, 210, 211 Sage, 78
Quinone reductase, 80 Sausages, 66, 82
Saw palmetto, 216
Scutellaria baiacalensis, 106
R Secoisolariciresinol diglycoside (SDG), 113
Selective serotonin reuptake inhibitors (SSRIs),
Radiotherapy 181, 184–185, See also Depression
hormesis (beneficial low-level effects), 83 pharmacotherapy; specific drugs
polymorphic genes and chemotherapy St. John’s wort and, 194, 217
outcomes, 286–287 tryptophan and, 183
Ras mutation activation, 107 Selegiline, 148, 155
Rasayanas, 114 Selenium, 212
Raspberries, 105 Selenomethionine, 292
Reactive oxygen species (ROS), 131, See also Serotonin (5-HT)
Oxidative stress depression pharmacotherapy and, 183, 199
antioxidant defenses, 74, See also major depression etiology, 180
Antioxidants St. John’s wort and, 194
phase II xenobiotic conjugation and, 73 zinc and brain uptake, 193
DK5836_Index.fm Page 334 Monday, May 22, 2006 2:32 PM
Serotonin and norepinephrine reuptake Sucrose intake, cardiovascular health and, 241
inhibitors (SNRIs), 15–16, 181, Suicide victims, 193
183, 184, See also Depression Sulfonylureas, 6–8, 211
pharmacotherapy; specific drugs Sulforaphane, 76, 79–80, 98
Serotonin syndrome, 180, 185, 194, 217 Sulfotransferase, 81, 268, 282–283
Sertraline, 181 Superoxide, 131
Shiitake mushrooms, 107 Superoxide dismutase, 132, 270
Sibutramine, 15–16 mutation and ALS, 154–156
Silicon, dietary, neurodegenerative disorders Synapsin, 191
and, 133–134 Synaptotagmin, 182
Simvastatin, 5, 17, 33, 45, 46, 47, See also α-Synuclein, 151
Statins
Single nucleotide polymorphisms, See Genetic
polymorphisms T
Skin cancer, 274
Skin disorders, 213 Tamoxifen, 108, 276, 277, 279, 282
SN-38G, 290 Tannic acid, 79
Soy, 41, 43, 76, 81, 98, 138, See also Daidzein; Tannins, 136
Genistein TCDD, 75, 78
Spinach, 78 Tea, See Black tea; Green tea
Squamous cell carcinoma, 106 Tegafur, 276, 277
St. John’s wort, 216–217 Tetracyclines, 210, 211, 213
antidepressant interactions, 193–194, 217 Tetrahydrobiopterin (BH4), 189
cancer chemotherapy and, 104 Theaflavins, 79, 135
drug metabolism interactions, 5, 72 Thearubigins, 135
hypolipidemic drug interactions, 47 Thiazide diuretics, 18, 210, 215
P-glycoprotein activity and, 293 Thiazolidinediones (TZDs), 6, 9–10, 35
xenobiotic metabolism and, 80, 104, 277 Thiobarbituric acid reactive substances
Stanols, 19, 42, 43 (TBARS), 147, 222
Statins, 18, 33–34, See also specific drugs Thiocyanates, 76
antitumor activity, 106–107 6-Thioguanine, 283, 290
cytochrome P450-mediated metabolism, 17 Thiopurine S-methyltransferase (TPMT), 283,
ezetimibe and, 37 290
fibrate interaction, 47 Thiotepa, 276
food effects on bioavailability, 17 Throat Coat, 101
genetic polymorphisms and cardiovascular Thromboxane A2 (TXA2), 106
health, 244–245 Thromboxane synthetase, 222
grapefruit juice and, 17, 45, 46 Thymidylate synthase, 283, 291
herb interactions, 5 Thyroid hormone, 212
kava interactions, 225 Thyroxine, 18
niacin and, 245 Tobacco smoke, carcinogens in, 66, 69, 75
Phase II glucuronidation, 44 Tobacco smokers, chemopreventative dietary
St. John’s wort and, 47 components, 74–75
Steroid and xenobiotic receptor (SXR), 72 α-Tocopherol, See Vitamin E
Sterol regulatory element-binding proteins Tolbutamide, 6–7
(SREBs), 38 Tomatoes, 137, 138
Sterols, 19, 42, 43 Topotecan, 276
Stilbenes, 136 Torcetrapib, 37
Stomach cancer, 64, 75, See also Trabectidin, 114
Gastrointestinal tract cancers Trans-fats, 3
polymorphic genes and chemotherapy Transcription regulators, hypolipidemic therapy
outcomes, 284, 289 applications, 38
Strawberries, 105, 135 Tranylcypromine, 181
Sucralfate, 215 Trastuzumab, 293
DK5836_Index.fm Page 335 Monday, May 22, 2006 2:32 PM
Index 335
V
W
Vascular endothelial growth factor (VEGF), 104
Venlafaxine, 181 Walnuts, 105
Verapamil, 46, 210 Warfarin, 18, 213, 214, 223, 224, 225
Very low density lipoproteins (VLDL) Watercress, 80
apolipoprotein E polymorphisms and, 240 Weight-reducing drugs, 15–16
statins and, 33 Wine consumption, See also Red wine
Vinblastine, 276, 289 neurodegenerative diseases and, 134, 157
Vincristine, 114, 276 neurological disorders and, 143
Vitamin A, 15, 135, 213 WS 1490, 224
DK5836_Index.fm Page 336 Monday, May 22, 2006 2:32 PM