Mi 4 2 137 PDF

Download as pdf or txt
Download as pdf or txt
You are on page 1of 19

MEDICINE INTERNATIONAL 4: 13, 2024

Immune checkpoint inhibitors in metastatic


melanoma therapy (Review)
VEDANT SHAH, VIRAJ PANCHAL, ABHI SHAH, BHAVYA VYAS,
SIDDHARTH AGRAWAL and SANKET BHARADWAJ

Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel
Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India

Received October 31, 2023; Accepted January 26, 2024

DOI: 10.3892/mi.2024.137

Abstract. An increase in the incidence of melanoma has significant breakthrough in the treatment of metastatic mela‑
been observed in recent decades, which poses a signifi‑ noma, providing improved outcomes compared to traditional
cant challenge due to its poor prognosis in the advanced therapies.
and metastatic stages. Previously, chemotherapy and
high doses of interleukin‑2 were available treatments for
melanoma; however, they offered limited survival benefits Contents
and were associated with severe toxicities. The treatment
of metastatic melanoma has been transformed by new 1. Introduction
developments in immunotherapy. Immune checkpoint 2. Immune checkpoint inhibitor drugs and their role in meta‑
inhibitors (ICIs), monoclonal antibodies that target cytotoxic static melanoma
T‑lymphocyte‑associated antigen‑4 (CTLA‑4), programmed 3. Mechanisms underlying the effectiveness of ICIs in meta‑
cell death protein 1 (PD‑1) and its ligand, PDL‑1, have static melanoma
emerged as promising therapeutic options. Commonly used 4. Combination therapy with immune checkpoint inhibitors
ICIs, such as ipilimumab, nivolumab and pembrolizumab, in melanoma
have been found to be associated with an improved median 5. Comparison between the immune checkpoint inhibitors
overall survival, recurrence‑free survival and response rates 6. Biomarkers
compared to traditional chemotherapies. Combination thera‑ 7. Factors affecting drug use
pies involving different types of ICIs, such as anti‑PD1 with 8. Adverse effects
anti‑CTLA‑4, have further enhanced the overall survival and 9. Mechanisms of resistance
response rates by targeting various phases of T‑cell activa‑ 10. Emerging newer therapeutic strategies: Targeting tumor
tion. Additionally, the development of novel biomarkers has metabolic dependencies
facilitated the assessment of responses to ICI therapy, with 11. Conclusion and future perspectives
tissue and serum‑based prognostic and predictive biomarkers
now available. The increased response observed with ICIs
also provides potential for immune‑related adverse effects 1. Introduction
on various organ systems. Further research is required to
evaluate the efficacy and safety of various combinations of The incidence of melanoma has increased over the past
ICIs, while ongoing clinical trials explore the potential of two decades (1). Each year, melanoma affects more than
newer ICIs. Concerns regarding the development of resis‑ 325,000 people. Males experience more frequent occur‑
tance to ICIs also warrant attention. The present review rences, with 174,000 yearly cases compared to females, with
summarizes and discusses the advent of ICIs with a marked 151,000 cases (2). Of note, 20% of patients with melanoma
eventually develop unresectable or distant metastatic disease
labeled as stage III/IV (3). Unfortunately, for such an
advanced‑stage disease, the prognosis remains bleak. However,
Correspondence to: Dr Viraj Panchal, Department of Medicine, understanding the advanced stages of melanoma growth and
Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel progression has led to the development of promising new
Institute of Medical Sciences and Research (SVPISMR), 1, Santoor therapeutic alternatives.
Bunglows, Iscon‑Ambali Road, Ahmedabad, Gujarat 380058, India Until 2011, chemotherapy was the initial treatment for meta‑
E‑mail: virajpanchal611@gmail.com static melanoma; however, it only provided a 6‑month median
survival time and a 25% 1‑year overall survival rate. High‑dose
Key words: ipilimumab, nivolumab, pembrolizumab, melanoma, interleukin‑2 (IL‑2) was the only immunotherapy available, but
immune checkpoint inhibitors was associated with severe toxicities and only benefited a limited
number of patients (4). Currently, advances in immunotherapy
2 SHAH et al: IMMUNE CHECKPOINT INHIBITORS IN MELANOMA

and studies on cell cycle regulatory molecules have facilitated 3. Mechanisms underlying the effectiveness of ICIs in
the creation of immune checkpoint inhibitors (ICIs), a group of metastatic melanoma
monoclonal antibodies that block co‑inhibitory molecules, such
as cytotoxic T‑lymphocyte‑associated antigen‑4 (CTLA‑4), CTLA‑4 and anti‑CTLA‑4 drugs. CTLA‑4, a B7/CD28 family
programmed cell death protein 1 (PD‑1) and its ligand, member, is a coinhibitory receptor expressed on the surface
PDL1 (5‑7). Ipilimumab, nivolumab and pembrolizumab were of T‑cells that eventually inhibits T‑cells, and it is expressed
the first class of medications shown to improve the overall by regulatory T‑cells (Tregs) (14). Discovered in 1987, it was
survival of patients with metastatic melanoma (4). considered to function as a negative regulator of T‑cell activa‑
The present review provides comprehensive evidence tion until the mid‑1990s (15‑17).
regarding the role of ICIs and their utilization in advanced When CTLA‑4 is expressed on the surface of CD4+ and
melanoma cases. The outcomes of ICIs, such as ipilimumab, CD8+ T‑cells, the binding affinity increases and is higher
nivolumab and pembrolizumab are highlighted, including the to CD80 and CD86, which are the costimulatory receptors
improved survival rates and response rates associated with present on antigen‑presenting cells (APCs) compared to CD28
their use compared to traditional chemotherapies, while also which is another costimulatory receptor (18). The expression of
focusing on the mechanisms and demonstrating the potentially CTLA‑4 increases when there is an activation of T‑cell recep‑
adverse effects of these therapies. tors and the release of cytokines such as IL‑12 and interferon
Furthermore, the present review focuses on combination (IFN)‑γ. This upregulation creates a feedback inhibition loop
therapies, including anti‑PD1 with anti‑CTLA‑4, showcasing on T‑effector cells which are activated, leading to CTLA‑4
their importance compared to monotherapy. The improved acting as a natural ‘brake’ on CD4+ and CD8+ T‑cell activation
outcomes of combination therapies over traditional therapies induced by APCs, as shown in Fig. 3.
are highlighted, with an emphasis on the need for ongoing Tregs also play a key role in maintaining immune homeo‑
research, optimized treatment approaches and strategies which stasis by inhibiting excessive immune responses. One of the
can be used to overcome resistance. In addition to discussing mechanisms through which Tregs suppress effector T‑cell
the development of novel biomarkers for assessing ICI thera‑ activity is via CTLA‑4 signaling (19). Two anti‑CTLA‑4 drugs
peutic responses in both tissue and serum‑based prognostic have been studied in patients with melanoma: i) Ipilimumab,
and predictive markers, tumor metabolic dependencies and the first ICI evaluated and approved for the treatment of
targeting the metabolic pathways by combining ICIs are also melanoma is a fully human immunoglobulin anti‑CTLA‑4
discussed. This could provide an improved efficacy, which, to monoclonal antibody (20,21); ii) tremelimumab, a fully human
the best of our knowledge, has not been described commonly immunoglobulin anti‑CTLA‑4 monoclonal antibody which is
in the available literature focusing on ICIs used in melanoma. still under investigation (12).
There are two major mechanisms through which these
2. Immune checkpoint inhibitor drugs and their role in drugs act. First, the inhibition of CTLA‑4 signaling in cyto‑
metastatic melanoma toxic T‑cells that specifically target tumors can directly affect
these cells by enabling them to evade a state of anergy and
The surface of immune cells, such as T‑cells, assists in the enter an active proliferative effector phase. Once activated,
regulation of the immune response through various receptors. these effector T‑cells are more likely to penetrate the tumor
When activated by certain ligands, these receptors inhibit and exhibit direct cytotoxic effects on tumor cells, while also
immune cells from attacking the body's own cells. However, releasing cytokines such as IL‑2 and IFN‑γ to stimulate an
in cancer, tumor cells can take advantage by binding to these immunogenic tumor microenvironment. Thus, by blocking
checkpoint‑inhibitory receptors through their own ligands and the CTLA‑4 pathway, T‑cells that were previously inactive
suppressing the immune response, as illustrated in Fig. 1. can become activated and effectively target the tumor cells,
Commonly implicated inhibitory receptors include causing a more powerful immune response against the cancer.
(CTLA‑4, PD‑1, T‑cell immunoglobulin domain and mucin This new approach holds promise as a potential immuno‑
domain‑3 (TIM‑3), killer cell immunoglobulin‑like receptor therapy for the treatment of cancer (22).
(KIR), lymphocyte‑activation gene 3 (LAG3), glucocorti‑ The second major mechanism driving these drugs is the
coid‑induced tumor necrosis factor receptor (GITR), B‑ and blocking of CTLA‑4 signaling in Tregs, which may impair
T‑lymphocyte attenuator (BTLA) and V‑domain immuno‑ their ability to halt the activity of effector T‑cells. This inhibi‑
globulin (Ig)‑containing suppressor of T‑cell activation tion of CTLA‑4 signaling can either cause a decrease in the
(VISTA) (8‑10), as presented in Fig. 2. number of Tregs or reduce their function without affecting
Over time, several drugs have been introduced targeting their population size. Therefore, blocking CTLA‑4 on Tregs
these receptors. Ipilimumab was one of the first ICI drugs to may disrupt this suppression and lead to increased immune
be approved by the Food and Drug Administration (FDA) for activation against tumor cells (19,23,24).
the treatment of metastatic melanoma, which functions by Patients with melanoma are treated with the primary aim
blocking CTLA‑4 (11). There are numerous additional compa‑ of suppressing the molecular interplay between the melanoma
rable drugs in early phase III, phase II, or preclinical research. cells and immune effector cells. Ipilimumab, which has mainly
These include pidilizumab, atezolizumab, durvalumab and been approved for the treatment of more advanced stages, such
tremelimumab (formerly known as ticilimumab) (12,13). To as unresectable or metastatic melanoma, has been shown to be
increase the immune system's defense against cancer cells, associated with a marked overall survival rate confirmed from
these medications also work against various immunological a phase 3 clinical trial (25). The interference of ipilimumab
checkpoints (12,13). on CTLA‑4 expressed on the subset of tumor‑specific T‑cell
MEDICINE INTERNATIONAL 4: 13, 2024 3

Figure 1. Illustration demonstrating the role of immune checkpoint inhibitors in suppressing the activation of T‑cells or preventing the release of cytokines
from tumor cells.

Figure 2. Various types of immune receptors on T‑cells and their interaction with ligands present on tumor cells or immune checkpoint inhibitors in the
treatment of melanoma. APC, antigen‑presenting cell; CTLA‑4, cytotoxic T‑lymphocyte‑associated antigen‑4; PD‑1, programmed cell death protein 1;
LAG‑3, lymphocyte‑activation gene‑3; TIM‑3, T‑cell immunoglobulin domain and mucin domain‑3; BTLA, B‑ and T‑lymphocyte attenuator; KIR, killer cell
immunoglobulin‑like receptor; VISTA, V‑domain immunoglobulin (Ig)‑containing suppressor of T‑cell activation; NK cell, natural killer cell.
4 SHAH et al: IMMUNE CHECKPOINT INHIBITORS IN MELANOMA

Figure 3. Illustration demonstrating the interaction of CTLA‑4 expression on T‑cells and CD80 and CD86 on APCs. CTLA‑4, cytotoxic T‑lymphocyte‑associated
antigen‑4; APC, antigen‑presenting cell; MHC, major histocompatibility complex.

proliferation and B7 molecules on antigen‑presenting cells is fully humanized and dostarlimab), as well as those that inhibit
expected to prevent tumor development (26). PD‑L1 (such as avelumab, atezolizumab, and durvalumab) are
being evaluated for use in melanoma cases and various other
PD‑1 pathway and anti‑PD‑1/PD‑L1 drugs. PD‑1 is a protein malignancies (NCT04020809, NCT04274816, NCT03313206,
present on the surface of T‑cells, B‑cells and natural killer NCT03842943 and NCT05928962). However, it is not yet
(NK) cells. It functions as an inhibitory molecule by binding known which inhibitor, PD‑1 or PD‑L1, is more efficient (13).
to PD‑L1 (or B7‑H1) and PD‑L2 (B7‑H2). PD‑L1 is expressed Of these, nivolumab and pembrolizumab are the two major
in numerous types of tissue, including hematopoietic cells and FDA‑approved anti‑PD‑1 monoclonal antibodies available for
certain tumors such as melanoma, where they are expressed in the treatment of advanced and metastatic melanomas.
40‑50% of cases. PD‑L2 is mainly expressed in hematopoietic Melanoma cells exhibit increased levels of PD‑L1, which
cells. The binding of PD‑1 to PD‑L1/2 inhibits the death of promotes the apoptosis of the likewise increased levels
tumor cells and promotes the conversion of T‑effector cells of T‑cells (33). It has also been found that the circulating
into Tregs, while also inducing exhaustion in peripheral melanoma antigen‑specific T‑cells and tumor‑infiltrating
T‑effector cells, as illustrated in Fig. 4 (27,28). lymphocytes express PD‑1 abnormally. It is considered that
PD‑1 and/or PD‑L1 are also expressed on cells, such as NK melanoma cells are capable of initiating, as well as sustaining
cells, monocytes and dendritic cells (27,28). The PD‑1 pathway PD‑1 signals, T‑cell fatigue and dysfunction (33). Hence, by
operates through various mechanisms, such as reducing blocking PD‑1 in patients with melanoma, one could possibly
the activity of T‑cells during an inflammatory response, restore abnormal activation and signaling and eventually
increasing the proliferation and suppressive activity of Tregs, recover the immune effect. Pembrolizumab or lambrolizumab
and reducing the lytic activity of B‑cells and NK cells (29). were used in unresectable or metastatic melanoma in the study
The affinity between PD‑1 and PD‑L1 is 3‑fold stronger by Hamid et al (34), in an aim to elucidate the effects of PD‑1
than the affinity between PD‑1 and PD‑L2. When PD‑L1 medications in melanoma.
binds with PD‑1 on T‑cells, it results in T‑cell exhaustion, However, due to the heterogeneous nature of tumors, the
dysfunction, neutralization and the production of IL‑10 within expression of PD‑L1 is not uniform throughout. The extent of
the tumor mass. This process allows tumors that overexpress PD‑L1 expression can differ in various locations within the
PD‑L1 to protect themselves from being attacked and killed by tumor, resulting in varying levels of PD‑L1 in immunohisto‑
CD8+ cytotoxic T‑cells (30). chemical staining. Moreover, the effectiveness of PD‑L1/PD‑1
Pro‑effector cytokines, namely IL‑12 and IFN‑ γ, can inhibitors can also be influenced by several other factors,
upregulate the expression of PD‑1 and PD‑L1/L2, which helps such as the type of cancer, the patient's immune system and
to prevent excessive T‑effector cell activity. It is worth noting the genetic profile of the tumor. Thus, a more in‑depth under‑
that PD‑L1 has also been shown to inhibit CD80, indicating standing of these factors is essential for developing effective
the existence of complex interactions between CTLA‑4, PD‑1 treatment strategies that consider the heterogeneity of tumors
and other pathways (31,32). and the variability in PD‑L1 expression (35).
The PD‑1 and PD‑L1 antibody inhibitors were created The combination ICI therapy used in the treatment
with the aim of preventing the PD‑1 or PD‑L1 side from of patients with metastatic melanoma primarily involves
functioning, reactivating T‑cells and promoting an immune CTLA‑1 and PD‑1 inhibitors. This amplified the inhibitions
response against cancer cells (13). that can be simultaneously intervened during different phases
Based on promising results from clinical trials, antibodies of the interaction among melanoma cells and the immune
that inhibit PD‑1 (such as pembrolizumab, nivolumab‑IgG4 system. This, for example, includes anti‑CTLA‑4 inhibiting
MEDICINE INTERNATIONAL 4: 13, 2024 5

Figure 4. Illustration demonstrating the interaction between PD‑1 and PD‑L1 among T‑cells and tumor cells. PD‑1, programmed cell death protein 1; PD‑L1,
programmed cell death protein ligand 1; CTLA‑4, cytotoxic T‑lymphocyte‑associated antigen‑4; MHC, major histocompatibility complex.

the priming stage at the same time anti‑PD‑1 inhibits the 4. Combination therapy with immune checkpoint inhibitors
effector stage (36,37). It has also been noted that the use of in melanoma
anti‑CTLA‑4 inhibitors results in an increased expression
of PD‑1; hence, using combination therapy results in a more The ability of anti‑CTLA‑4 and anti‑PD‑1/PD‑L1 mono‑
robust treatment response in patients with melanoma (38). clonal antibodies to target various T‑cell activation locations
and phases is the rationale for their combined use. PD‑1 is
Newer drugs. An increased understanding of immunological primarily expressed on antigen‑experienced T‑cells in periph‑
mechanisms has led to the identification of additional potential eral tissues, while CTLA‑4 is expressed by naive T‑cells in the
targets for checkpoint inhibition in the treatment of cancer. lymph nodes. According to pre‑clinical research, combining
Some of these potential targets include BTLA, VISTA, TIM‑3, ICIs is more effective than treatment with with monotherapy
CD47 and LAG‑3. i) The blockade of BTLA has been shown for managing melanoma (50‑53).
to enhance New York esophageal squamous cell carcinoma In pre‑clinical investigations, anti‑CTLA‑ 4 and
1) specific CD8+ T‑cell function and enhance the efficacy of anti‑PD‑1/PD‑L1 monoclonal antibodies have been shown to
anti‑PD‑1 (39‑41). ii) VISTA blockade has been shown to induce the infiltration of CD8+ T‑cells and the expansion of
increase T‑cell infiltration and function in tumors, thereby an inducible T‑cell co‑stimulator (ICOS)+ T helper 1‑like CD4
reducing tumor growth (10,42). iii) TIM‑3 blockade causes fraction, which in turn induces the response of CD4+ effector
T‑helper‑1 cell hyperproliferation and cytokine release, leading T‑cells. Based on this, the sequencing or combination of
to tumor shrinkage in a mouse model when combined with nivolumab with ipilimumab in metastatic cutaneous melanoma
anti‑CTLA‑4 or anti‑PD‑1 (43‑47). iv) Targeting CD47 with has been researched (50‑53). Other combination studies are on
a humanized anti‑CD47 monoclonal antibody in combination nivolumab, relatlimab and combination therapy with pembro‑
with rituximab has shown to lead to objective responses in half lizumab with low‑dose ipilimumab (54,55). The data from the
of the heavily pretreated patients with relapsed or refractory CheckMate and RELATIVITIY047 trials on the combination
non‑Hodgkin's lymphoma, including a complete response in of ICIs are presented in Table I, which demonstrate a favorable
more than one‑third of patients (48). v) An immune pathway response for such therapies (54,56,57). Table II presents data
known as LAG‑3 has been identified as a potential complement from a meta‑analysis, comparing monotherapy and combina‑
to the PD‑1/PDL1 pathway in enhancing the immune response tion therapy (47‑64).
against cancer. LAG‑3 is an immune checkpoint receptor that
regulates the function of T‑cells. BMS‑986016 is a therapy 5. Comparison between the immune checkpoint inhibitors
that targets LAG‑3 and is currently under investigation in
combination with nivolumab, which targets PD‑1, to enhance Along with the major breakthrough in melanoma treatment
the immune response against cancer cells. The combination with the use of selective BRAF inhibitors, after ~6 months of
of these two therapies has the potential to create a synergistic the median duration, resistance to therapy began to develop.
effect, leading to improved treatment outcomes for patients The BRAF mutation drives the tumor proliferation exponen‑
with cancer (49). tially by activating mitogen‑activated kinase pathway (MAP),
6 SHAH et al: IMMUNE CHECKPOINT INHIBITORS IN MELANOMA

Table I. Summary of the results of the CheckMate and RELATIVITY047 trials performed on the combination of immune
checkpoint inhibitors for the treatment of melanoma.

Objective Median 5‑year Median 5‑Year overall


Study name Treatment group response rate (%) PFS PFS rate overall survival survival rate (Refs.)

CheckMate 064 Nivolumab followed 41 ‑ ‑ 76% ‑ (56)


by ipilimumab
CheckMate 064 Ipilimumab followed 20 ‑ ‑ 54% ‑ (56)
by nivolumab
Checkmate 067 Nivolumab‑ipilimumab 58 11.5 months 36% 36.9 months 52% (57)
Checkmate 067 Nivolumab 45 6.9 months 29% ‑ 44% (57)
Checkmate 067 Ipilimumab 19 2.9 months 8% 19.9 months 26% (57)
RELATIVITY‑047 Nivolumab‑relatlimab 47.7 10.1 months ‑ ‑ ‑ (54)
RELATIVITY‑047 Nivolumab 36 4.6 months ‑ ‑ ‑ (54)

PFS, progression‑free survival.

Table II. Summary of results obtained from a previous meta‑analysis of clinical trials conducted by Pradeep et al (58), which
compared the efficacy and safety of ICIs between monotherapy and combined ICI therapy in advanced melanomas.

Outcome [(Refs.); as in the Effect size in terms


meta‑analysis by Pradeep et al (58)] Comparison of HR or RR (95% CI) P‑value

Overall survival (52‑56) Nivolumab with ipilimumab vs. monotherapy HR, 0.65 (0.53‑0.79) <0.0001
Nivolumab with ipilimumab vs. nivolumab alone HR, 0.84 (0.71‑0.99) 0.04
Nivolumab with ipilimumab vs. ipilimumab alone HR, 0.54 (0.48‑0.62) <0.00001
Progression‑free survival (53,55‑58) Nivolumab with ipilimumab vs. monotherapy HR, 0.48 (0.38‑0.60) <0.0001
Nivolumab with ipilimumab vs. nivolumab alone HR, 0.68 (0.49‑0.94) 0.02
Nivolumab + ipilimumab vs. ipilimumab alone HR, 0.42 (0.37‑0.47) <0.00001
Overall response rate (52,54‑57,59) Nivolumab with ipilimumab vs. monotherapy RR, 2.15 (1.63‑2.84) <0.00001
Nivolumab with ipilimumab vs. nivolumab alone RR, 1.32 (1.22‑1.43) <0.00001
Nivolumab with ipilimumab vs. ipilimumab alone RR, 3.09 (2.74‑3.50) <0.00001

In the meta‑analysis by Pradeep et al (58), overall survival, progression‑free survival and objective response rates were the outcomes studied.
HR, hazard ratio; RR, relative risk.

and the development of resistance to BRAF inhibitors in patients who received ipilimumab plus gp100 had a longer
both MAP kinase‑dependent and MAP kinase‑independent median overall survival rate of 10 months compared to
pathways (65‑67). Resistance in MAP kinase‑dependent path‑ 6.4 months for those who received gp100 alone, with a hazard
ways includes secondary mutations in NRAS, the increased ratio (HR) for mortality of 0.68 and a statistically significant
expression of COT kinase, CRAF activation and acquired P‑value of <0.001. The median overall survival rate of patients
mutations in MEK1 (65,68‑71). MAP kinase‑independent who received ipilimumab alone was 10.1 months (HR, 0.66;
pathways include the upregulation of platelet‑derived growth P=0.003) compared to those who received gp100 alone (20).
factor receptor, additional receptor tyrosine kinases activation In the study by Robert et al (25), patients with untreated
including AXL, Erb‑B2 receptor tyrosine kinase 4) and insulin metastatic melanoma were randomly assigned to receive
like growth factor 1 receptor, the activation of PI3K/AKT either ipilimumab plus dacarbazine or dacarbazine alone. A
signaling, and the loss of phosphatase and tensin homolog significantly longer median overall survival (11.2 months) was
(PTEN) (65,69,71‑75). found in the ipilimumab plus dacarbazine group compared
to the dacarbazine alone group (9.1 months). In a follow‑up
Ipilimumab. Ipilimumab is a fully human monoclonal anti‑ maintenance study, patients who received ipilimumab
body developed to antagonize CTLA‑4. A clinical study was plus dacarbazine had a higher 5‑year survival rate (18.2%)
conducted on patients who had unresectable stage III or IV compared to those who received dacarbazine alone (8.8%).
melanoma, where they were randomly assigned in a 3:1:1 These findings suggest that ipilimumab in combination with
ratio to receive ipilimumab (3 mg/kg) plus the glycoprotein dacarbazine may be an effective treatment option for meta‑
100 (gp100) vaccine, ipilimumab alone, or gp100 alone. The static melanoma patients (76).
MEDICINE INTERNATIONAL 4: 13, 2024 7

In a trial for ipilimumab as an adjuvant treatment performed a more effective treatment option for some patients (81).
by Eggermont et al (77), patients with stage III cutaneous mela‑ Another study was also carried out to compare the efficacy
noma who had undergone complete resection were randomly of nivolumab compared to ipilimumab as an adjuvant therapy
administered either ipilimumab (10 mg/kg) (n=475) or a in patients who had resected advanced melanoma. In patients
placebo (n=476). Following a median follow‑up of 5.3 years, with stage III or stage IV melanoma, adjuvant therapy was
the ipilimumab group had a 5‑year recurrence‑free survival administered with either nivolumab (n=453) or ipilimumab
rate of 40.8%, while the placebo group had a rate of 30.3% (n=453) and follow‑up was performed after 18 months (82).
(HR, 0.76; P<0.001). In terms of the 5‑year overall survival The 12‑month rate of recurrence‑free survival was signifi‑
rate, the ipilimumab group had a rate of 65.4% compared to cantly higher in the nivolumab group at 70.5%, vs. 60.8% in
54.4% in the placebo group (HR, 0.72; P=0.001) (77). the ipilimumab group with a HR of 0.65 (P<0.001). It was also
In the study by Ascierto et al (78), patients with unresect‑ noted that treatment‑related adverse events were 14.4% for
able stage III or IV melanoma were randomly assigned to patients treated with nivolumab and 45.9% for those treated
receive either 10 or 3 mg/kg ipilimumab. The median overall with ipilimumab. Therefore, patients who received ipilimumab
survival rate was 15.7 months for the 10 mg/kg group and therapy experienced more severe side‑effects than those who
11.5 months for the 3 mg/kg group (HR, 0.84; P=0.04). Overall, received nivolumab therapy. This suggests that nivolumab may
their study suggested that ipilimumab treatment improved the be a more effective and tolerable treatment option for patients
survival outcomes of patients with unresectable stage III or IV with stage IIIB, IIIC, or IV melanoma following surgical
melanomas, with higher doses of the drug (10 mg/kg) leading resection (82).
to an improved overall survvial compared to lower doses
(3 mg/kg) (78). Pembrolizumab. Pembrolizumab is a monoclonal antibody
which functions by blocking the PD‑1 on T‑cells and allowing
Nivolumab. Nivolumab is a monoclonal antibody that these T‑cells to identify and kill cancer cells. Similar to
inhibits the interaction of PD‑1 with PD‑L1. The clinical nivolumab, pembrolizumab was also compared with ICC in
study performed by Robert et al (79) compared the efficacy ipilimumab‑refractory melanoma. A randomized controlled
of nivolumab with the standard therapy of dacarbazine. In study was conducted on patients with advanced melanoma and
their study, patients who had metastatic melanoma without a have progressed even after receiving ipilimumab and/or standard
BRAF mutation were randomly divided into two groups (1:1 BRAF therapy (83). Patients were divided into three groups as
with nivolumab at 3 mg/kg once every 2 weeks (n=210) and follows: One group (n=181) received 10 mg/kg pembrolizumab,
dacarbazine (n=208). The survival rate at 1 year was 72.9% in one group (n=180) received 2 mg/kg pembrolizumab, and
patients treated with nivolumab compared to 42.1% in patients another group (n=179) received ICC. The 6‑month progres‑
who were assigned dacarbazine (HR, 0.42, P<0.001). The sion‑free survival rate was found to be 38% in patients treated
objective response rate was 40% with nivolumab compared with pembrolizumab at 10 mg/kg (HR, 0.5 vs. ICC; P<0.0001),
to 13.9% with dacarbazine (odds ratio, 4.06; P<0.001) (79). 34% in the 2 mg/kg group (HR, 0.57 vs. ICC; P<0.0001) and
Another randomized controlled trial was carried out between 16% in the ICC group (83). Another study was conducted by
2012‑2014 on patients with advanced melanoma who Robert et al (84), this time comparing pembrolizumab with
progressed after ipilimumab therapy or a combination of ipili‑ ipilimumab. Patients with advanced melanoma were divided at
mumab and a BRAF inhibitor if they were found to be positive a 1:1:1 ratio to receive pembrolizumab at 10 mg/kg once every
for a V600E mutation (80). That study assessed the role of 2 weeks or pembrolizumab at 2 mg/kg once every 3 weeks or
nivolumab as a second‑line treatment in the management of four doses of ipilimumab at 3 mg/kg for once every 3 weeks. An
patients with advanced melanoma. Patients were divided into interim analysis was performed which revealed that the 6‑month
three groups in a 2:1 pattern where one group (n=272) received progression‑free survival of the patients treated with pembroli‑
nivolumab at 3 mg/kg once every 2 weeks and another group zumab once every 2 weeks was 47.3% (HR, 0.58 vs. ipilimumab;
(n=133) received the investigator's choice of chemotherapy P<0.001), 46.4% for those treated with pembrolizumab once
(ICC), which was either dacarbazine or paclitaxel plus carbo‑ every 3 weeks (HR, 0.58 vs. ipilimumab; P<0.001) and 26.5%
platin (80). An interim analysis of that study found that, in the for those treated with ipilimumab (84). A final analysis revealed
first 120 patients of the nivolumab group, 38 patients (31.7%) that the median overall survival rate was not reached in both
experienced confirmed objective responses, whereas only 5 out pembrolizumab groups; however, it was noted to be 16 months in
of the 47 patients (10.8%) receiving the ICC treatment exhib‑ the ipilimumab group (HR, 0.68 for pembrolizumab once every
ited similar responses. Subsequently, upon further analysis of 2 weeks vs. ipilimumab, P=0.0009; and HR, 0.68 for pembro‑
that trial, it was revealed that the median overall survival rate lizumab once every 3 weeks vs. ipilimumab, P=0.0008) (85).
of patients who received nivolumab was 16 months, while for Similarly, in the study by Robert et al (84) the 24‑month overall
those who received ICC, it was 14 months (81). The HR was survival rate was 55% in the group treated once every 2 weeks,
0.95, indicating that nivolumab did not improve the survival 55% in the group treated once every 3 weeks and 43% in the
rate of patients who had ipilimumab‑refractory metastatic ipilimumab group. Not only do nivolumab and pembrolizumab
melanoma when compared to ICC. However, nivolumab prolong overall survival, but they also maintain the quality of
had a higher overall response rate of 27% vs. 10% for ICC, life of patients with melanoma (86,87). These findings have led
and the median duration of response was also longer for to the FDA approval of pembrolizumab for ipilimumab and/or
nivolumab at 32 months compared to 13 months for ICC (77). BRAF inhibitory refractory advanced melanoma. A summary
Hence, nivolumab exhibiting a higher overall response rate of the comparison among ICIs and their outcomes in patients is
and a longer duration of response suggests that it may be presented in Table III.
8 SHAH et al: IMMUNE CHECKPOINT INHIBITORS IN MELANOMA

Table III. Individual immune checkpoint inhibitors with the various outcomes affecting clinical decisions regarding their use.

ICI drug Compared drug Outcome (Refs.)

Ipilimumab Ipilimumab (3 mg/kg) (n=137) vs. Median overall survival (months): 10.1 vs. (20)
Gp100 (n=136) vs. ipilimumab (3 mg/kg) 6.4 vs. 10.0
with gp100 (n=403)
Ipilimumab (10 mg/kg) with dacarbazine (n=251) vs. Median overall survival (months): (76)
dacarbazine (n=251) 11.2 vs. 9.1
Ipilimumab (10 mg/kg) adjuvant (n=475) vs. Recurrence‑free survival (%): 40.8 vs. 30.3 (77)
placebo (n=476)
Ipilimumab (10 mg/kg) (n=365) vs. Median overall survival (months): (78)
ipilimumab (3 mg/kg) (n=362) 15.7 vs. 11.5
Nivolumab Nivolumab(3 mg/kg) once every 2 weeks (n=210) vs. 1‑Year survival rate (%): 72.9 vs. 42.1; (79)
dacarbazine (n=208) objective response rate (%): 40 vs. 13.9
Nivolumab (3 mg/kg) (n=272) vs. ICC (dacarbazine Median overall survival rate (months): (80)
or paclitaxel plus carboplatin) (n=133) 16 vs. 14; overall response rate (%):
27 vs. 10
Nivolumab (n=453) vs. ipilimumab (n=453) 12‑Month recurrence‑free survival (82)
rate (%): 70.5 vs. 60.8; treatment‑related
adverse events (%): 14.4 vs. 45.9
Pembrolizumab Pembrolizumab (10 mg/kg) (n=181) vs. 6‑Month progression‑free survival rate (%): (83)
pembrolizumab (2 mg/kg) (n=180) vs. ICC 38 vs. 34 vs. 16
(paclitaxel plus carboplatin, paclitaxel, carboplatin,
dacarbazine, or oral temozolomide) (n=179)
Pembrolizumab (10 mg/kg once every 2 weeks) 6‑Month progression‑free survival rate (%): (85)
(n=279) vs. pembrolizumab (2 mg/kg once every 47.3 vs. 46.4 vs. 26.5; median overall
3 weeks) (n=277) vs. four doses of ipilimumab survival rate (months): NA vs. NA vs. 16;
(3 mg/kg once every 3 weeks) (n=278) 24‑month overall survival rate (%): 55 vs.
55 vs. 43

ICC, investigator's choice of chemotherapy; NA, not available.

6. Biomarkers ii) Histotype: The majority of melanoma histotypes are not


considered prognostic when looked at individually from tumor
The prognostic marker for melanoma traditionally used is thickness, and are therefore not included in the American Joint
the depth of invasion and the associated mitotic count of the Committee on Cancer staging system (90,109,110). However,
affected cells. With advancements being made, newer prog‑ a nodular melanoma is an independent predictor which can
nostic markers have been found and used. Prognostic and be used for the measurement of recurrence and its association
predictive biomarkers have gained importance, particularly in with mortality due to melanoma (111).
the treatment of melanoma. iii) Digital images trained from AI: New advancements
have allowed for the development of deep learning‑based
Serum biomarkers. The role of serum biomarkers in the early biomarkers, which can help to stratify the stages of melanoma
detection of melanoma is described in Table IV (88‑106). into risk groups, and thus associate disease‑specific survival
with two independent validating cohorts to accurately predict
Tissue markers the prognosis of patients with early‑stage melanoma (112).
Prognostic markers. i) Tumor infiltrating lymphocyte (TIL) iv) Melanoma cell adhesion molecule (MCAM): Expressed
patterns are often divided into grades, such as ‘absent’, in 80% of metastatic tumors, MCAM is a cell adhesion
which is no presence of any lymphocytes within the tumor, marker (113). Those who are positive for MCAM have signifi‑
‘non‑brisk’, which suggest few foci of lymphocytes within cantly worse 5‑year survival rates than those who are negative
the tumor, or ‘brisk’, which is a large diffuse infiltration of for MCAM, and there is an inverse association between the
lymphocytes within the tumor (107). As demonstrated by amount of marker expressed and survival (114,115).
Clark et al (107) in 1989, as well as by others, the presence of v) Ki‑67: Ki‑67 is a unique nuclear antigen that can function
brisk TILs in a vertical growth pattern is often associated with as a marker for cellular proliferation during the active phase
a favorable disease‑specific survival and overall survival rate of the cell cycle (116). For melanomas who have a thickness
after non‑brisk and absent patterns of TILs (107,108). <1 mm, the risk of metastasis increases with the expression of
Table IV. Role of serum biomarkers in melanoma.

Biomarker Use Type Information (Refs.)

LDH Prognostic and predictive Protein‑based The AJCC staging scheme includes blood LDH as an independent prognostic biomarker for metastatic
melanoma. A univariate analysis revealed that an increased amount of LDH was associated with a lesser
response to anti‑PD‑1 medications in patients who received anti‑PD‑1 monotherapy (88).
It was previously found that patients with elevated LDH and NLR levels, despite receiving ICIs, had poor
a OS and PFS (89).
Elevated serum LDH levels increased the disease stage of patients with metastasis to stage IV M1c (1)
metastatic disease, which included individuals with metastases to the lungs, as well as to other visceral
sites apart from the central nervous system (90,91).
S100 Prognostic Protein‑based S100B levels in serum are affected by the tumor load, and higher values of S100B may be used as a
prognostic biomarker and a sign of disease progression (92‑94).
It was previously found that the discriminative ability with serum S100B levels for detecting disease
relapse was much higher than that of serum LDH (95).
miRNA Prognostic Genomic The role of miRNAs as post‑transcriptional inhibitors of translation has been substantially
investigated (96‑100). It has been discovered that these small non‑coding RNAs are detectable in exosomes
in serum (101). It has been demonstrated that aberrant levels of expression of certain miRNAs are
associated with the stage and recurrence of disease (102,103).
miRNAs are crucial in the deregulation of a number of oncogenic pathways in uveal melanoma, which can
facilitate the metastatic spread of disease. The study by Wróblewska et al (104) found that it may be
beneficial to use differentially expressed miRNAs as a biomarker for the evaluation of the risk of
metastasis in patients with uveal melanoma.
MEDICINE INTERNATIONAL 4: 13, 2024

ctDNA Prognostic Genetic It has been demonstrated that circulating tumor DNA testing may detect BRAF V600e mutations, which is
helpful for determining how effectively the BRAF/MEK inhibitor therapy works. Imaging scan
measurements of disease progression and treatment effectiveness have been found to be associated with
the amount of circulating tumor DNA (105,106).
Both univariate and multivariate analyses have revealed an association between ctDNA detectability and a
shorter OS. Patients with distant metastases (79%) were more likely to have ctDNA detected than those
with no distant metastases or only intracranial tumors (32%). Elevated protein S100 and CRP levels were
more closely linked to detectable ctDNA than LDH (104).

AJCC, American Joint Committee on Cancer; LDH, lactate dehydrogenase; PD‑1, programmed cell death protein 1; NLR, neutrophil‑lymphocyte ratio; ICI, immune checkpoint inhibitor; OS, overall
survival; PFS, progression‑free survival; miRNA, microRNA; ctDNA, circulating tumor DNA.
9
10 SHAH et al: IMMUNE CHECKPOINT INHIBITORS IN MELANOMA

Ki‑67 and an increased mitotic rate (117). However, with the among women of reproductive age. As opposed to this, post‑
increasing thickness of melanomas, Ki‑67 can serve as a more menopausal women have a relatively low incidence of the
effective prognostic marker than the mitotic rate, and is often disease, thus raising the possibility that sex hormones such as
associated with ulceration within the tumor, necrosis, higher estrogen may be involved in the growth of the disease (158).
level Clark's level of invasion, and even vascular invasion (118). As a result, estrogen levels should be considered an important
In addition, with recurrent melanomas, higher values of Ki‑67 biomarker in advanced melanoma. Elderly patients aged ≥65
exhibit an independent association with a decreased overall treated with combination therapy comprising of ipilimumab
survival (119). and nivolumab have not exhibited a considerable difference
vi) Lymphatic invasion: In research on primary mela‑ in overall mortality. When prior exposure to ipilimumab is
nomas with a thickness >1 mm, D2‑40 staining was assessed considered, women have a 2.82‑fold increased risk of mortality
for lymphatic invasion, which is an antibody against sialo‑ as compared to prior‑exposed males with ipilimumab (158).
glycoprotein that selectively attaches on endothelial cells of Moderate colitis which does not require the use of intrave‑
lymphatic vessels and helps detect sentinel lymph node metas‑ nous steroids is consistent with an improved overall survival
tasis (120‑122). of patients with stage IV melanoma when treated with a single
vii) Osteopontin: Overexpressed in numerous visceral anti‑CTLA‑4 drug, but not with combination drugs. This
malignancies, osteopontin is known as an integrin‑binding holds true even after the completion of therapy (159). Multiple
protein and used as a biomarker to measure tumor progress nonrandomized studies have shown excellent results in patients
and metastasis (123‑125). It functions as an independent who discontinue treatment after being treated for 1‑2 years
predictor for the prognosis of melanoma and was found to be and disease progression is also uncommon in the following
associated with increased sentinel lymph node positivity in a 2‑5 years of treatment termination (160‑162). This is in contrast
cohort of 345 patients who had primary melanoma detected to the progression of disease of patients with non‑small cell
using immunohistochemical analysis (126). lung cancer, for whom treatment continuation led to improved
viii) Driver mutations: It has been found that BRAF results compared to treatment termination (163).
and NRAS are associated with a significantly lower
melanoma‑specific survival in high‑risk tumors, such as 8. Adverse effects
a stage >2 (127). NF1 mutations has also been found to be
associated with a lower disease‑specific survival and overall The use of PD‑1 inhibitors, namely nivolumab and pembro‑
survival (128). However, further research is required to iden‑ lizumab, and the anti‑CTLA‑4 drug, ipilimumab, has been
tify patients with BRAF mutations and uncover the role of shown to be associated with a steady regression in malignan‑
BRAF mutations in directing the treatment strategy. cies, including metastatic melanoma (164).
Predictive markers. The role of predictive markers in PD‑1 inhibitors function in the tumor setting, while
melanoma and its clinical importance is summarized in CTLA‑4 inhibitors act on lymphoid tissue, resulting in a
Table V (61,129‑151). wide and different set of adverse events (165). Combination
therapies with nivolumab and ipilimumab have been proven
7. Factors affecting drug use to be more effective with a response rate of 59% as compared
to when used alone, with response rate of 43% for nivolumab
Therapy with nivolumab affects the frequencies of innate and 15‑20% for ipilimumab. Moreover, an increased response
lymphoid cells (ILCs) in peripheral blood in patients with mela‑ rate is associated with an increase in adverse events, resulting
noma. The frequency, as well as the secretory activity of ILC in an overall increase in adverse events with the combination
subsets, particularly ILC2s, are affected by treatment. Albeit of nivolumab with ipilimumab, as compared to nivolumab or
nivolumab was found to not effectively alter serum cytokine ipilimumab monotherapy (36,62).
profiles, pro‑inflammatory and angiogenic substances such as A CTLA‑4 blockade with or without anti‑PD‑1 antibody
IL‑1, IL‑6, CCL2, CXCL8 and VEGF had levels outside the produces adverse events in a dose‑dependent manner (166,167).
normal range in 7 of the 18 cytokines. In addition, the produc‑ Considering that older patients are more inclined to develop
tion of IL‑5 and IL‑13 was affected, which are released during rheumatologic events and female patients are also at an
parasite infections and allergic reactions (152). In malignant increased risk, the toxicity profile may vary according to age
melanoma, type 3 ILC is suspected in tumor suppression (153). and sex (168,169).
Serum levels of IL‑6, CXCL8 and CCL2 in particular, surge However, as these molecules are targeted, due to the
during melanoma progression, while mature NKp44+ ILC3s resulting immune response, an increase in the incidence of
protect against melanoma (154). autoimmune conditions is observed; these adverse events are
As previously demonstrated, the advancement of melanoma known as immune‑related adverse events (irAE). If severe
was comparable with aging, although the treatment outcome irAEs occur with one of the drugs, then it is a safe practice
did not differ significantly, and there was no significant change to re‑challenge the patient with a different class of drug (165).
in the survival outcomes of elderly patients as compared to These drugs have the following on the following systems.
young ones. Moreover, it was recommended that both age
groups should be treated in similar manner (155). Primary Gastrointestinal tract. Diarrhea is the most frequent irAE
and secondary resistance are also a key factor affecting drug with incidences between 10 and 13% (164).
use (84,156). Combination therapy with ipilimumab and
nivolumab, as approved by the FDA, has been proven to be Endocrine disorders. In decreasing order, the first endocrine
efficient (157). There is an increased incidence of melanoma system that is most affected by ICIs is the thyroid gland
MEDICINE INTERNATIONAL 4: 13, 2024 11

Table V. Role of predictive biomarkers and their clinical relevance.

Type Biomarker Clinical relevance (Refs.)

Tumor intrinsic TMB/neoantigen Tumors with a higher TMB are potentially more responsive to ICIs; a reason for this
profile may be the fact that these tumors express more neoantigen and hence, may be
recognized easily as a target by T‑cells (129). With the increasing evidence of the
ability of TMBs to predict the response to immunotherapy in melanoma, various
studies have been conducted (130,131). In a previous study, 321 patients with
melanoma were treated with ICIs and it was found that with a higher TMB, one could
predict an increased survival following treatment (132).
Driver mutations Immunotherapy was found to be more effective in patients with activating NRAS genes
than in those without such mutations. According to the CheckMate 067 study, those
patients with BRAF‑mutation in melanoma had a 4‑year overall survival rate of 62%
with combination therapy with ipilimumab and nivolumab, when compared to only
50% with nivolumab alone and 33% with ipilimumab alone (61,133).
Tumor MHC Longer overall survival, a greater response rate, and an increased infiltration of CD4+
microenvironment and CD8+ T‑cells in the tumor microenvironment were all associated with elevated
levels of MHC‑II expression. Conversely, ipilimumab treatment was found to be
associated with a higher risk of disease progression when MHC‑I expression was
lower (<30%), with a 100% negative predictive value (134‑136).
HLA supertypes Two HLA supertypes were identified and linked to CTLA‑4 inhibition therapeutic
outcomes: HLA‑B44 was associated with a longer survival, while HLA‑B62 was
associated with a shorter survival (137).
PD‑L1 PD‑L1 is a complex biomarker to study since it is regulated by several pathways, which
is vulnerable to high sampling error, and expressed on numerous immune cells in the
microenvironment (138). Treatment with PD‑1 inhibitors has been demonstrated to be
successful in both patients with and without PD‑L expression; however, the quality
of immunohistochemical staining for PD‑L1 is not dependable for clinical use (139,141).
Immune ICI‑sensitive melanomas exhibited more oxidative phosphorylation and lipid
metabolism metabolism than ICI‑resistant tumors using high resolution mass spectrometry. In
addition, elevated lipid metabolism increases antigen presentation, which may be the
reason why ICI‑sensitive cancers have a better response to therapy (142).
TIL/TCF‑7 It has been demonstrated that TCF‑7, a transcription factor, promotes a central memory
stem‑like state, and that TCF7‑expressing CD8+ cells have the ability to self‑renew
and differentiate into effector cells (143,144). TCF‑7‑positive cells appear to be the
ones that multiply following anti‑PD1 therapy (145). According to this finding, TCF‑7
expression has been linked to successful clinical results in patients with melanoma
receiving ICIs (146). In light of this, further research is required to establish the role of
TCF7 expression as a predictive biomarker in patients with melanoma who are being
treated with ICIs and its role as a therapeutic target.
GEP Research has been performed to identify a gene expression profile (GEP) that can
predict the response to pembrolizumab. It was discovered that the 18‑gene profile
could identify aspects of the tumor microenvironment that are pertinent to predicting
the clinical outcome of pembrolizumab that are independent of tumor type. This
indicates that GEPs, such as the 18‑gene profile, can be utilized to predict how different
types of cancer will respond to PD‑1/PD‑L1 inhibitors (147‑149).
IPRES The IPRES transcriptional signature is a peculiar pattern of gene expression that is
present in tumors which are resistant to anti‑PD‑1 medications. Increased activity in
genes is related to an increase in transition through the mesenchyme, helping in cell
adhesion, extracellular matrix remodeling, increase in angiogenesis, and wound
repair which are the hallmark of this pattern. The response rate to anti‑PD‑1 therapy can
be enhanced with the identification of transcriptomic features that are associated with
anti‑PD‑1 resistance, which may suggest the mitigation of IPRES‑related biological
process and eventually, an enhance response rate to treatment (147)
12 SHAH et al: IMMUNE CHECKPOINT INHIBITORS IN MELANOMA

Table V. Continued.

Type Biomarker Clinical relevance (Refs.)

Host factors Gut microbiome Patients with high‑fiber diets were 5‑fold more likely than those with low‑fiber diets
to respond to anti‑PD‑1 therapy. In addition, regardless of the type of antibiotic, indi
viduals who had received antibiotics for >30 days prior to commencing ICI treatment
suffered worse outcomes. A past but not concurrent antibiotic treatment has an impact
on the gut microbiome's ‘activating’ response to ICIs (150).
Stress A solid tumor line was grafted into mice, and they were then treated with an anti‑PD‑1
monoclonal antibody. Subsequently, they were made to experience social defeat stress,
and compared to the mice who did not experience this type of stress; the mice who were
socially defeated with stress responded to PD‑1 inhibition less effectively (151).
Others AI According to recent research, pre‑treatment computerized H&E slides can be used to
artificially predict the probability of a response to ICIs (112).

TMB, tumor mutational burden; ICI, immune checkpoint inhibitor; MHC, major histocompatibility complex; HLA, human leukocyte antigen;
CTLA‑4, cytotoxic T‑lymphocyte‑associated antigen‑4; PD‑L1, programmed cell death protein ligand 1; TCF‑7, transcription factor 7; IPRES,
innate anti‑PD1 resistance; AI, artificial intelligence; H&E, hematoxylin and eosin.

(typically hypothyroidism observed following a transient proteins (such as tyrosinase and TRP‑2), the interference of
thyroiditis‑induced thyrotoxicosis) followed by the rest of the PD‑1 signaling may result in autoimmune vitiligo (180). This
endocrine organs. The median time frame from the start of offers a reasonable explanation for the onset and durability of
the treatment to the development of thyroid symptoms, most depigmentation in patients receiving immunotherapy.
commonly hypothyroidism, is 6 weeks, followed by pituitary
(hypophysitis), adrenals (primary adrenal insufficiency) and Lungs. Case series studies have demonstrated that patients
β‑cells of the pancreas (insulin deficient diabetes, analogous to develop organizing pneumonia, diffuse alveolar damage,
type 1 diabetes) (170). These are different from the side‑effects acute respiratory distress syndrome (ARDS) and non‑specific
brought on by conventional cytotoxic chemotherapy or even interstitial pneumonia, which is then managed by intravenous
more recent molecular‑targeted medicines, which infrequently and oral steroids (181‑185).
result in endocrine dysfunction (171).
Liver and kidneys. A previous meta‑analysis revealed adverse
Skin disorders. Non‑specific adverse events such as macu‑ effects associated with the use of anti‑PD‑1/PD‑L1 mono‑
lopapular rash, pruritus, psoriasiform, eczematous and clonal antibodies for malignancies with an increased incidence
lichenoid dermatosis are among the most prevalent (172,173). of pancreatitis, and increased levels of liver enzymes, such as
Compared to anti‑PD‑1 monotherapy, the maculopapular aspartate aminotransferase and alanine transaminase, elevated
rash phenotype is more prevalent when CTLA‑4 inhibition creatinine levels, nephritis and renal failure (164).
is implemented (21). Bullous pemphigoid, vitiligo‑like skin
hypopigmentation/depigmentation and alopecia are other 9. Mechanisms of resistance
less‑common irCAEs (174,175). Although severe reactions,
such as Stevens‑Johnson syndrome, toxic epidermal necrolysis A myriad of ongoing clinical trials and practices have discov‑
and drug reaction with eosinophilia and systemic symp‑ ered multiple mechanisms leading to resistance to ICIs. More
toms are uncommon, cutaneous consequences are typically precisely, these include changes in the tumor microenviron‑
self‑limiting (174‑176). Early diagnosis and the administration ment prohibiting T‑cell interaction, tumor invasion and tumor
of corticosteroids or antitumor necrosis factor‑agents are the cell destruction by effector mechanisms. The key to tumor
foundation of treatment algorithms for irCAEs (176,177). cell destruction via effector T‑cells is through the processing
However, the use of corticosteroids before or after ICI initiation of tumor antigens to antigen‑presenting cells. The failure of
may result in a diminished antitumor efficacy. Anti‑CTLA‑4 antigen‑presenting components in this pathway is a major
and anti‑PD1 therapy have both been associated with reports cause of resistance in melanoma (186). β2 microglobulin is
of vitiligo (178). The occurrence of skin hypopigmentation or an key molecule responsible for the folding and transporta‑
depigmentation such as vitiligo has been linked to an extensive tion of major histocompatibility complex‑1 to the surface of
anticancer benefit from drug treatment in patients with mela‑ cells. Mutations in these molecules have been noted in patients
noma. Vitiligo has been proven as a positive predictive factor in with melanoma at the time of anti‑PD1 treatment failure (187).
measuring the tumor response to treatment. In comparison with Other mechanisms responsible for limiting T‑cell trafficking in
the general population, patients with melanoma have a 10‑fold the tumor microenvironment include mutations in BRAF, and
increased incidence of drug‑related cutaneous hypopigmenta‑ the inhibition of PTEN. This leads to the increased expression
tion and depigmentation (179). Since the PD‑L1:PD1 pathway of immunosuppressive molecules, such as VEGF (188). It also
mostly regulates the peripheral tolerance of melanosomal inhibits the migration and trafficking of effector T‑cells (189).
MEDICINE INTERNATIONAL 4: 13, 2024 13

In addition to these tumor‑intrinsic mechanisms, various it was combined with an anti‑PD1 antibody, it markedly
tumor‑extrinsic mechanisms also play a role in the develop‑ improved the survival and increased immune response with
ment of resistance to ICIs. These include the development of reduced tumor cell mass (199). With such promising results, a
new inhibitory checkpoints, immunosuppressive cytokines number of newer anti‑PI3K medicines are being developed and
and molecules in the tumor microenvironment suppressing tested to increase efficacy (NCT01390818). Despite this, more
immune cell function. One such example is the production of novel promising approaches are needed to prove the success
transforming growth factor β (TGF‑β) by tumor cells. TGF‑β is of combining anti‑PI3K drugs with ICIs in the treatment of
an immunosuppressive cytokine that functions by stimulating melanoma (200).
Tregs and inhibiting the cytotoxicity of effector T‑cells (190).
To summarize, understanding and investigating the 11. Conclusion and future perspectives
potential mechanisms that lead to resistance to ICIs is crucial
in developing effective strategies to guide therapy. Further The development of ICIs and targeted therapies has played a
studies are required to identify new mechanisms and develop crucial role in revolutionizing the management of melanomas
targeted therapies to improve the clinical outcome of patients by improving the overall and progression‑free survival.
undertaking immunotherapy. Although both of these therapies have advantages and disad‑
vantages, combination therapy (ICI + ICI, or ICI + targeted
10. Emerging newer therapeutic strategies: Targeting therapies) has been found to be more effective in improving
tumor metabolic dependencies patient outcomes. However, there is limited literature avail‑
able regarding combination therapies and different types of
Tumor cells sustain themselves by utilizing altered metabolic potential combinations. There are also insufficient data on
pathways by using nutrients, such as glucose, tryptophan and patients and their responses to draw sufficient conclusions. The
arginine to produce toxic metabolites such as adenosine, lactate development of drug‑related adverse effects with the use of
and kynurenine (191,192). Such toxic metabolites produce an combination therapies is also a debatable question. However,
unfavorable environment for the antitumor cells to function when developing newer ICIs to achieve a more effective
resulting in increased expression of immune checkpoints and response, a focus should certainly be placed on the integration
expansion of Tregs (193). of nanotechnology or antibody engineering. Through these,
The mechanism that tumor cells use is the mutation in the one can increase drug delivery to a specific target and thus
myelocytomatosis oncogene (MYC) and PI3K/AKT/mamma‑ increase overall response. In addition, focusing on epigenetic
lian target of rapamycin (mTOR) signaling pathways. The modulation and developing ICIs that target those changes can
increased expression of hypoxia‑inducible factor‑1‑ α leads enhance the responsiveness of ICIs.
to the overexpression of the PI3K/AKT/mTOR pathway, as There may be concerns regarding resistance to ICIs in
well as glucose transporters such as glucose transporter 1, patients with melanoma. Some patients may have resistance
leading to increased glucose consumption and acidification of to certain ICIs from the beginning or may develop them
the tumor microenvironment (194,195). As hypoxia is gener‑ as an acquired resistance with subsequent treatment after
ated, glucose depletion occurs and increased toxic waste is progression of a tumor with clinical benefit. Further treat‑
produced within the tumor microenvironment, resulting in the ment decisions shall be made on the basis of evaluation of
inhibition of tumor antigen presentation by APCs (196). Thus, the tumor and factors related to the patient, focusing on
there is an overall decrease in the antitumor immune response targeted therapeutic drugs, other immunotherapy drugs,
by T‑effector, macrophages or NK cells, while pro‑tumor cellular therapies, intralesional therapies, or chemotherapy. It
immune cells such as Tregs proliferate to increase the expres‑ is important to tailor ICI treatment based on an individual's
sion of inhibitory checkpoint ligand PD‑1 on immune cells, genetic makeup and tumor characteristics to decrease the
inhibiting the antitumor immunity (197). With the advance‑ resistance. The early identification of tumor biomarkers can
ment of technologies, newer therapeutic strategies that target predict future responses to particular ICIs and may help to
the immunosuppressive tumor microenvironment generated select a personalized treatment strategy. Furthermore, with
by tumor cells may be developed to reprogram the behavior of the use of tumor metabolic pathway inhibitors in combina‑
immune cells, leading to an improved efficacy in terms of the tion with ICIs, targeting signaling pathways and immune
treatment response. responses can be better used to overcome potential resistance
One of the important T‑cellular processes is the activation to ICIs than when used alone.
of the PI3K pathway, which plays a vital role in proliferation Trials are being conducted on newer inhibitory immune
and differentiation. Monotherapy, which inhibits the PI3K checkpoint targets, as well as certain inhibitory targets beyond
pathway, has not yielded any significant results in the treat‑ immune checkpoints. These include LAF‑3, TIM‑3, B7‑H3
ment of cancer; however, combining PI3K inhibitors and the and B7‑H4, CD73, etc. which are immune checkpoints, and
PD‑1‑PDL1 blockade has shown some notable results (198). CEACAM1, CEACAM5/6, CCL2/CCR2, etc. which are other
The loss of PTEN, which is a PI3K‑inhibiting tumor suppressor inhibitory targets (201). It is essential to maintain enrollment
often mutated in tumor cells, results in the uncontrolled growth in clinical trials so that newer ICIs, additional inhibitory treat‑
of tumor cells and escapes the immune destruction imposed ments, combination therapy, and mechanisms of resistance
on it. As previously demonstrated, when mice with PTEN‑null and methods of overcoming the resistance can all be further
melanoma were treated in vivo with the PI3K β inhibitor, investigated.
GSK2636771, this resulted in a decreased AKT phosphoryla‑ In conclusion, with the increasing incidence of mela‑
tion and the activation of mTOR targets. Additionally, when noma over the past two decades, managing it with different
14 SHAH et al: IMMUNE CHECKPOINT INHIBITORS IN MELANOMA

treatment modalities has become cumbersome. With the 4. Rosenberg SA, Lotze MT, Yang JC, Topalian SL, Chang AE,
Schwartzentruber DJ, Aebersold P, Leitman S, Linehan WM,
limited effectiveness of the traditional approach using Seipp CA, et al: Prospective randomized trial of high‑dose
chemotherapy and immunotherapy, the role of newer interleukin‑2 alone or in conjunction with lymphokine‑activated
treatment modalities should be given equal emphasis. killer cells for the treatment of patients with advanced cancer.
J Natl Cancer Inst 85: 622‑632, 1993.
Novel approaches using ICIs have been a revolution in the 5. Bronte V and Mocellin S: Suppressive influences in the immune
therapeutic approach by unleashing the immune system's response to cancer. J Immunother 32: 1‑11, 2009.
ability to recognize and eliminate cancer cells. Ipilimumab, 6. Mellman I, Coukos G and Dranoff G: Cancer immunotherapy
comes of age. Nature 480: 480‑489, 2001.
nivolumab and pembrolizumab have been shown to lead to 7. Pardoll DM: The blockade of immune checkpoints in cancer
a substantial improvement in the overall survival of patients immunotherapy. Nat Rev Cancer 12: 252‑264, 2012.
with advanced melanoma, particularly in high‑risk meta‑ 8. Okazaki T, Okazaki IM, Wang J, Sugiura D, Nakaki F, Yoshida T,
Kato Y, Fagarasan S, Muramatsu M, Eto T, et al: PD‑1 and
static melanoma compared to traditional therapies. However, LAG‑3 inhibitory co‑receptors act synergistically to prevent
with ICIs, it is paramount to monitor any side‑effects, and autoimmunity in mice. J Exp Med 208: 395‑407, 2011.
to ensure the optimal outcome is achieved using personal‑ 9. Fourcade J, Sun Z, Pagliano O, Chauvin JM, Sander C, Janjic B,
Tarhini AA, Tawbi HA, Kirkwood JM, Moschos S, et al: PD‑1
ized treatment approaches. and Tim‑3 regulate the expansion of tumor antigen‑specific
CD8+ T cells induced by melanoma vaccines. Cancer Res 74:
Acknowledgements 1045‑1055, 2014.
10. Lines JL, Pantazi E, Mak J, Sempere LF, Wang L, O'Connell S,
Ceeraz S, Suriawinata AA, Yan S, Ernstoff MS and Noelle R:
Not applicable. VISTA is an immune checkpoint molecule for human T‑cells.
Cancer Res 74: 1924‑1932, 2014.
11. Hanaizi Z, van Zwieten‑Boot B, Calvo G, Lopez AS, van
Funding Dartel M, Camarero J, Abadie E and Pignatti F: The European
medicines agency review of ipilimumab (Yervoy) for the treat‑
No funding was received. ment of advanced (unresectable or metastatic) melanoma in
adults who have received prior therapy: Summary of the scientific
assessment of the committee for medicinal products for human
Availability of data and materials use. Eur J Cancer 48: 237‑242, 2012.
12. Tarhini AA: Tremelimumab: A review of development to date in
solid tumors. Immunotherapy 5: 215‑229, 2013.
Not applicable. 13. Wang D, Wang T, Liu J, Yu H, Jiao S, Feng B, Zhou F, Fu Y,
Yin Q, Zhang P, et al: Acid‑activatable versatile micelleplexes
Authors' contributions for PD‑L1 blockade‑enhanced cancer photodynamic immuno‑
therapy. Nano Lett 16: 5503‑5513, 2016.
14. Ottaviano M, De Placido S and Ascierto PA: Recent success and
VS, VP and AS were involved in the conceptualization of limitations of immune checkpoint inhibitors for cancer: A lesson
the study. VS, VP, AS, BV, SB and SA were involved in the from melanoma. Virchows Arch 474: 421‑432, 2019.
15. Chambers CA, Sullivan TJ and Allison JP: Lymphoproliferation
curation of data from the literature for inclusion in the present in CTLA‑4‑deficient mice is mediated by costimulation-
review. VS, VP and AS were involved in selecting the relevant dependent activation of CD4 + T‑cells. Immunity 7: 885‑895,
literature. VS, VP and AS were involved in project admin‑ 1997.
16. Tivol EA, Borriello F, Schweitzer AN, Lynch WP, Bluestone JA
istration. VS supervised the study. VS, VP, AS, BV, SB and and Sharpe AH: Loss of CTLA‑4 leads to massive lymphop‑
SA were involved in the writing of the original draft. VS, VP roliferation and fatal multiorgan tissue destruction, revealing
and AS were involved in the writing, review and editing of a critical negative regulatory role of CTLA‑4. Immunity 3:
541‑547, 1995.
the manuscript. All authors have read and approved the final 17. Waterhouse P, Penninger JM, Timms E, Wakeham A,
manuscript. Data authentication is not applicable. Shahinian A, Lee KP, Thompson CB, Griesser H and Mak TW:
Lymphoproliferative disorders with early lethality in mice defi‑
cient in Ctla‑4. Science 270: 985‑988, 1995.
Ethics approval and consent to participate 18. Walker LSK and Sansom DM: The emerging role of CTLA4 as a
cell‑extrinsic regulator of T cell responses. Nat Rev Immunol 11:
Not applicable. 852‑863, 2011.
19. Ménard C, Ghiringhelli F, Roux S, Chaput N, Mateus C,
Grohmann U, Caillat‑Zucman S, Zitvogel L and Robert C: Ctla‑4
Patient consent for publication blockade confers lymphocyte resistance to regulatory T‑cells in
advanced melanoma: Surrogate marker of efficacy of tremelim‑
umab? Clin Cancer Res 14: 5242‑5249, 2008.
Not applicable. 20. Hodi FS, O'Day SJ, McDermott DF, Weber RW, Sosman JA,
Haanen JB, Gonzalez R, Rober t C, Schadendor f D,
Competing interests Hassel JC, et al: Improved survival with ipilimumab in patients
with metastatic melanoma. N Engl J Med 363: 711‑723, 2010.
21. Phan GQ, Yang JC, Sherry RM, Hwu P, Topalian SL,
The authors declare that they have no competing interests. Schwartzentruber DJ, Restifo NP, Haworth LR, Seipp CA,
Freezer LJ, et al: Cancer regression and autoimmunity induced
by cytotoxic T lymphocyte‑associated antigen 4 blockade in
References patients with metastatic melanoma. Proc Natl Acad Sci USA 100:
8372‑8377, 2003.
1. Finn L, Markovic SN and Joseph RW: Therapy for metastatic 22. Malek TR and Castro I: Interleukin‑2 receptor signaling: At
melanoma: The past, present, and future. BMC Med 10: 23, 2012. the interface between tolerance and immunity. Immunity 33:
2. Arnold M, Singh D, Laversanne M, Vignat J, Vaccarella S, 153‑165, 2010.
Meheus F, Cust AE, de Vries E, Whiteman DC and Bray F: 23. Reuben JM, Lee BN, Li C, Gomez‑Navarro J, Bozon VA,
Global burden of cutaneous melanoma in 2020 and projections Parker CA, Hernandez IM, Gutierrez C, Lopez‑Berestein G and
to 2040. JAMA Dermatol 158: 495‑503, 2022. Camacho LH: Biologic and immunomodulatory events after
3. Miller AJ and Mihm MC Jr: Melanoma. N Engl J Med 355: CTLA‑4 blockade with ticilimumab in patients with advanced
51‑65, 2006. malignant melanoma. Cancer 106: 2437‑2444, 2006.
MEDICINE INTERNATIONAL 4: 13, 2024 15

24. Ribas A, Comin‑Anduix B, Economou JS, Donahue TR, 44. Anderson AC, Anderson DE, Bregoli L, Hastings WD,
de la Rocha P, Morris LF, Jalil J, Dissette VB, Shintaku IP, Kassam N, Lei C, Chandwaskar R, Karman J, Su EW,
Glaspy JA, et al: Intratumoral immune cell infiltrates, FoxP3, and Hirashima M, et al: Promotion of tissue inflammation by the
indoleamine 2,3‑dioxygenase in patients with melanoma under‑ immune receptor Tim‑3 expressed on innate immune cells.
going CTLA4 blockade. Clin Cancer Res 15: 390‑399, 2009. Science 318: 1141‑1143, 2007.
25. Robert C, Thomas L, Bondarenko I, O'Day S, Weber J, Garbe C, 45. Zhu C, Anderson AC, Schubart A, Xiong H, Imitola J, Khoury SJ,
Lebbe C, Baurain JF, Testori A, Grob JJ, et al: Ipilimumab plus Zheng XX, Strom TB and Kuchroo VK: The Tim‑3 ligand
dacarbazine for previously untreated metastatic melanoma. galectin‑9 negatively regulates T helper type 1 immunity. Nat
N Engl J Med 364: 2517‑2526, 2011. Immunol 6: 1245‑1252, 2005.
26. Buchbinder EI and Desai A: CTLA‑4 and PD‑1 pathways: 46. Sabatos CA, Chakravarti S, Cha E, Schubart A, Sánchez‑Fueyo A,
Similarities, differences, and implications of their inhibition. Am Zheng XX, Coyle AJ, Strom TB, Freeman GJ and Kuchroo VK:
J Clin Oncol 39: 98‑106, 2016. Interaction of Tim‑3 and Tim‑3 ligand regulates T helper type 1
27. Amarnath S, Mangus CW, Wang JCM, Wei F, He A, Kapoor V, responses and induction of peripheral tolerance. Nat Immunol 4:
Foley JE, Massey PR, Felizardo TC, Riley JL, et al: The 1102‑1110, 2003.
PDL1‑PD1 axis converts human TH1 cells into regulatory 47. Ngiow SF, von Scheidt B, Akiba H, Yagita H, Teng MWL and
T‑cells. Sci Transl Med 3: 111ra120, 2011. Smyth MJ: Anti‑TIM3 antibody promotes T cell IFN‑γ‑mediated
28. Spranger S, Spaapen RM, Zha Y, Williams J, Meng Y, Ha TT and antitumor immunity and suppresses established tumors. Cancer
Gajewski TF: Up‑regulation of PD‑L1, IDO, and T(regs) in the Res 71: 3540‑3551, 2011.
melanoma tumor microenvironment is driven by CD8(+) T cells. 48. Advani R, Flinn I, Popplewell L, Forero A, Bartlett NL,
Sci Transl Med 5: 200ra116, 2013. Ghosh N, Kline J, Roschewski M, LaCasce A, Collins GP, et al:
29. Sun Z, Fourcade J, Pagliano O, Chauvin JM, Sander C, CD47 blockade by Hu5F9‑G4 and rituximab in non‑Hodgkin's
Kirkwood JM and Zarour HM: IL10 and PD‑1 cooperate to lymphoma. N Engl J Med 379: 1711‑1721, 2018.
limit the activity of tumor‑specific CD8+ T cells. Cancer Res 75: 49. Ascierto PA, Melero I, Bhatia S, Bono P, Sanborn RE,
1635‑1644, 2015. Lipson EJ, Callahan MK, Gajewski T, Gomez‑Roca CA,
30. Zou W and Chen L: Inhibitory B7‑family molecules in the Hodi FS, et al: Initial efficacy of anti‑lymphocyte activa‑
tumour microenvironment. Nat Rev Immunol 8: 467‑477, 2008. tion gene‑3 (anti‑LAG‑3; BMS‑986016) in combination with
31. Kinter AL, Godbout EJ, McNally JP, Sereti I, Roby GA, nivolumab (nivo) in pts with melanoma (MEL) previously
O'Shea MA and Fauci AS: The common gamma‑chain cytokines treated with anti‑PD‑1/PD‑L1 therapy. J Clin Orthod 35 (15
IL‑2, IL‑7, IL‑15, and IL‑21 induce the expression of programmed Suppl): S9520, 2017.
death‑1 and its ligands. J Immunol 181: 6738‑6746, 2008. 50. Wei SC, Levine JH, Cogdill AP, Zhao Y, Anang NAS,
32. Yang J, Riella LV, Chock S, Liu T, Zhao X, Yuan X, Paterson AM, Andrews MC, Sharma P, Wang J, Wargo JA, Pe'er D and
Watanabe T, Vanguri V, Yagita H, et al: The novel costimulatory Allison JP: Distinct cellular mechanisms underlie anti‑CTLA‑4
programmed death ligand 1/B7.1 pathway is functional in inhibiting and anti‑PD‑1 checkpoint blockade. Cell 170: 1120‑1133.e17,
alloimmune responses in vivo. J Immunol 187: 1113‑1119, 2011. 2017.
33. Krönig H, Julia Falchner K, Odendahl M, Brackertz B, Conrad H, 51. Rotte A, Jin JY and Lemaire V: Mechanistic overview of immune
Muck D, Hein R, Blank C, Peschel C, Haller B, et al: PD‑1 expres‑ checkpoints to support the rational design of their combinations
sion on Melan‑A‑reactive T cells increases during progression to in cancer immunotherapy. Ann Oncol 29: 71‑83, 2018.
metastatic disease. Int J Cancer 130: 2327‑2336, 2012. 52. Tarhini A: Immune‑mediated adverse events associated
34. Hamid O, Robert C, Daud A, Hodi FS, Hwu WJ, Kefford R, with ipilimumab ctla‑4 blockade therapy: The underlying
Wolchok JD, Hersey P, Joseph RW, Weber JS, et al: Safety and mechanisms and clinical management. Scientifica (Cairo) 2013:
tumor responses with lambrolizumab (anti‑PD‑1) in melanoma. 857519, 2013.
N Engl J Med 369: 134‑144, 2013. 53. Michot J M, Bigenwa ld C, Cha mpiat S, Collins M,
35. Francisco LM, Salinas VH, Brown KE, Vanguri VK, Freeman GJ, Carbonnel F, Postel‑Vinay S, Berdelou A, Varga A, Bahleda R,
Kuchroo VK and Sharpe AH: PD‑L1 regulates the development, Hollebecque A, et al: Immune‑related adverse events with
maintenance, and function of induced regulatory T cells. J Exp immune checkpoint blockade: A comprehensive review. Eur J
Med 206: 3015‑3029, 2009. Cancer 54: 139‑148, 2016.
36. Larkin J, Chiarion‑Sileni V, Gonzalez R, Grob JJ, Cowey CL, 54. Tawbi HA, Schadendorf D, Lipson EJ, Ascierto PA, Matamala L,
Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, et al: Castillo Gutiérrez E, Rutkowski P, Gogas HJ, Lao CD, De
Combined nivolumab and ipilimumab or monotherapy in untreated Menezes JJ, et al: Relatlimab and nivolumab versus nivolumab
melanoma. N Engl J Med 373: 23‑34, 2015. in untreated advanced melanoma. N Engl J Med 386: 24‑34,
37. Wolchok JD, Chiarion‑Sileni V, Gonzalez R, Rutkowski P, 2022.
Grob JJ, Cowey CL, Lao CD, Wagstaff J, Schadendorf D, 55. Olson DJ, Eroglu Z, Brockstein B, Poklepovic AS, Bajaj M,
Ferrucci PF, et al: Overall survival with combined nivolumab Babu S, Hallmeyer S, Velasco M, Lutzky J, Higgs E, et al:
and ipilimumab in advanced melanoma. N Engl J Med 377: Pembrolizumab plus ipilimumab following anti‑PD‑1/L1 failure
1345‑1356, 2017. in melanoma. J Clin Oncol 39: 2647‑2655, 2021.
38. Marconcini R, Spagnolo F, Stucci LS, Ribero S, Marra E, Rosa F, 56. Weber JS, Gibney G, Sullivan RJ, Sosman JA, Slingluff CL Jr,
Picasso V, Di Guardo L, Cimminiello C, Cavalieri S, et al: Lawrence DP, Logan TF, Schuchter LM, Nair S, Fecher L, et al:
Current status and perspectives in immunotherapy for metastatic Sequential administration of nivolumab and ipilimumab with a
melanoma. Oncotarget 9: 12452‑12470, 2018. planned switch in patients with advanced melanoma (CheckMate
39. Watanabe N, Gavrieli M, Sedy JR, Yang J, Fallarino F, Loftin SK, 064): An open‑label, randomised, phase 2 trial. Lancet Oncol 17:
Hurchla MA, Zimmerman N, Sim J, Zang X, et al: BTLA is a 943‑955, 2016.
lymphocyte inhibitory receptor with similarities to CTLA‑4 and 57. Shoushtari AN, Wagstaff J, Ascierto PA, Butler MO, Lao CD,
PD‑1. Nat Immunol 4: 670‑679, 2003. Marquez‑Rodas I, Chiarion‑Sileni V, Dummer R, Ferrucci PF,
40. Murphy KM, Nelson CA and Sedý JR: Balancing co‑stimulation Lorigan P, et al: CheckMate 067: Long‑term outcomes in patients
and inhibition with BTLA and HVEM. Nat Rev Immunol 6: with mucosal melanoma. J Clin Orthod 38 (15 Suppl): S10019,
671‑681, 2006. 2020.
41. Fourcade J, Sun Z, Pagliano O, Guillaume P, Luescher IF, 58. Pradeep J, Win TT, Aye SN and Sreeramareddy CT: Efficacy and
Sander C, Kirkwood JM, Olive D, Kuchroo V and Zarour HM: safety of immune checkpoint inhibitors for advanced malignant
CD8(+) T cells specific for tumor antigens can be rendered melanoma: A meta‑analysis on monotherapy vs combination
dysfunctional by the tumor microenvironment through upregula‑ therapy. J Cancer 13: 3091‑3102, 2022.
tion of the inhibitory receptors BTLA and PD‑1. Cancer Res 72: 59. Amaria RN, Reddy SM, Tawbi HA, Davies MA, Ross MI,
887‑896, 2012. Glitza IC, Cormier JN, Lewis C, Hwu WJ, Hanna E, et al:
42. Le Mercier I, Chen W, Lines JL, Day M, Li J, Sergent P, Noelle RJ Neoadjuvant immune checkpoint blockade in high‑risk resect‑
and Wang L: VISTA regulates the development of protective able melanoma. Nat Med 24: 1649‑1654, 2018.
antitumor immunity. Cancer Res 74: 1933‑1944, 2014. 60. Hodi FS, Chesney J, Pavlick AC, Robert C, Grossmann KF,
43. Monney L, Sabatos CA, Gaglia JL, Ryu A, Waldner H, Chernova T, McDer mott DF, Linette GP, Meyer N, Giguere J K,
Manning S, Greenfield EA, Coyle AJ, Sobel RA, et al: Agarwala SS, et al: Combined nivolumab and ipilimumab
Th1‑specific cell surface protein Tim‑3 regulates macrophage versus ipilimumab alone in patients with advanced melanoma:
activation and severity of an autoimmune disease. Nature 415: 2‑Year overall survival outcomes in a multicentre, randomised,
536‑541, 2002. controlled, phase 2 trial. Lancet Oncol 17: 1558‑1568, 2016.
16 SHAH et al: IMMUNE CHECKPOINT INHIBITORS IN MELANOMA

61. Hodi FS, Chiarion‑Sileni V, Gonzalez R, Grob JJ, Rutkowski P, 78. Ascierto PA, Del Vecchio M, Robert C, Mackiewicz A,
Cowey CL, Lao CD, Schadendorf D, Wagstaff J, Dummer R, et al: Chiarion‑Sileni V, Arance A, Lebbé C, Bastholt L, Hamid O,
Nivolumab plus ipilimumab or nivolumab alone versus ipilim‑ Rutkowski P, et al: Ipilimumab 10 mg/kg versus ipilimumab
umab alone in advanced melanoma (CheckMate 067): 4‑Year 3 mg/kg in patients with unresectable or metastatic melanoma:
outcomes of a multicentre, randomised, phase 3 trial. Lancet A randomised, double‑blind, multicentre, phase 3 trial. Lancet
Oncol 19: 1480‑1492, 2018. Oncol 18: 611‑622, 2017.
62. Larkin J, Chiarion‑Sileni V, Gonzalez R, Grob JJ, Rutkowski P, 79. Robert C, Long GV, Brady B, Dutriaux C, Maio M, Mortier L,
Lao CD, Cowey CL, Schadendorf D, Wagstaff J, Dummer R, et al: Hassel JC, Rutkowski P, McNeil C, Kalinka‑Warzocha E, et al:
Five‑year survival with combined nivolumab and ipilimumab in Nivolumab in previously untreated melanoma without BRAF
advanced melanoma. N Engl J Med 381: 1535‑1546, 2019. mutation. N Engl J Med 372: 320‑330, 2015.
63. Postow MA, Chesney J, Pavlick AC, Robert C, Grossmann K, 80. Weber JS, D'Angelo SP, Minor D, Hodi FS, Gutzmer R, Neyns B,
McDer mot t D, Li net te GP, Meyer N, Gig uere J K, Hoeller C, Khushalani NI, Miller WH Jr, Lao CD, et al:
Agarwala SS, et al: Nivolumab and ipilimumab versus ipilim‑ Nivolumab versus chemotherapy in patients with advanced mela‑
umab in untreated melanoma. N Engl J Med 372: 2006‑2017, noma who progressed after anti‑CTLA‑4 treatment (CheckMate
2015. 037): A randomised, controlled, open‑label, phase 3 trial. Lancet
64. Long GV, Atkinson V, Lo S, Sandhu S, Guminski AD, Brown MP, Oncol 16: 375‑384, 2015.
Wilmott JS, Edwards J, Gonzalez M, Scolyer RA, et al: 81. Larkin J, Minor D, D'Angelo S, Neyns B, Smylie M, Miller WH
Combination nivolumab and ipilimumab or nivolumab alone in Jr, Gutzmer R, Linette G, Chmielowski B, Lao CD, et al:
melanoma brain metastases: A multicentre randomised phase 2 Overall survival in patients with advanced melanoma who
study. Lancet Oncol 19: 672‑681, 2018. received nivolumab versus investigator's choice chemotherapy in
65. Wagle N, Emery C, Berger MF, Davis MJ, Sawyer A, CheckMate 037: A randomized, controlled, open‑label phase III
Pochanard P, Kehoe SM, Johannessen CM, Macconaill LE, trial. J Clin Oncol 36: 383‑390, 2018.
Hahn WC, et al: Dissecting therapeutic resistance to RAF 82. Weber J, Mandala M, Del Vecchio M, Gogas HJ, Arance AM,
inhibition in melanoma by tumor genomic profiling. J Clin Cowey CL, Dalle S, Schenker M, Chiarion‑Sileni V,
Oncol 29: 3085‑3096, 2011. Marquez‑Rodas I, et al: Adjuvant nivolumab versus ipilim‑
66. Gorre ME, Mohammed M, Ellwood K, Hsu N, Paquette R, umab in resected stage III or IV melanoma. N Engl J Med 377:
Rao PN and Sawyers CL: Clinical resistance to STI‑571 cancer 1824‑1835, 2017.
therapy caused by BCR‑ABL gene mutation or amplification. 83. Ribas A, Puzanov I, Dummer R, Schadendorf D, Hamid O,
Science 293: 876‑880, 2001. Robert C, Hodi FS, Schachter J, Pavlick AC, Lewis KD, et al:
67. Ellis LM and Hicklin DJ: Resistance to targeted therapies: Pembrolizumab versus investigator‑choice chemotherapy
Refining anticancer therapy in the era of molecular oncology. for ipilimumab‑refractory melanoma (KEYNOTE‑002): A
Clin Cancer Res 15: 7471‑7478, 2009. randomised, controlled, phase 2 trial. Lancet Oncol 16: 908‑918,
68. Nazarian R, Shi H, Wang Q, Kong X, Koya RC, Lee H, Chen Z, 2015.
Lee MK, Attar N, Sazegar H, et al: Melanomas acquire resistance 84. Robert C, Ribas A, Schachter J, Arance A, Grob JJ,
to B‑RAF(V600E) inhibition by RTK or N‑RAS upregulation. Mortier L, Daud A, Carlino MS, McNeil CM, Lotem M, et al:
Nature 468: 973‑977, 2010. Pembrolizumab versus ipilimumab in advanced melanoma
69. Johannessen CM, Boehm JS, K im SY, T homas SR, (KEYNOTE‑006): Post‑hoc 5‑year results from an open‑label,
Wardwell L, Johnson LA, Emery CM, Stransky N, Cogdill AP, multicentre, randomised, controlled, phase 3 study. Lancet
Barretina J, et al: COT drives resistance to RAF inhibition Oncol 20: 1239‑1251, 2019.
through MAP kinase pathway reactivation. Nature 468: 968‑972, 85. Schachter J, Ribas A, Long GV, A rance A, Grob JJ,
2010. Mortier L, Daud A, Carlino MS, McNeil C, Lotem M, et al:
70. Montagut C, Sharma SV, Shioda T, McDermott U, Ulman M, Pembrolizumab versus ipilimumab for advanced melanoma:
Ulkus LE, Dias‑Santagata D, Stubbs H, Lee DY, Singh A, et al: Final overall survival results of a multicentre, randomised,
Elevated CRAF as a potential mechanism of acquired resistance open‑label phase 3 study (KEYNOTE‑006). Lancet 390:
to BRAF inhibition in melanoma. Cancer Res 68: 4853‑4861, 1853‑1862, 2017.
2008. 86. Long GV, Atkinson V, Ascierto PA, Robert C, Hassel JC,
71. Villanueva J, Vult ur A, L ee J T, Somasunda ra m R, Rutkowski P, Savage KJ, Taylor F, Coon C, Gilloteau I, et al:
Fukunaga‑Kalabis M, Cipolla AK, Wubbenhorst B, Xu X, Effect of nivolumab on health‑related quality of life in patients
Gimotty PA, Kee D, et al: Acquired resistance to BRAF inhibitors with treatment‑naïve advanced melanoma: Results from the
mediated by a RAF kinase switch in melanoma can be overcome phase III CheckMate 066 study. Ann Oncol 27: 1940‑1946,
by cotargeting MEK and IGF‑1R/PI3K. Cancer Cell 18: 683‑695, 2016.
2010. 87. Schadendorf D, Dummer R, Hauschild A, Rober t C,
72. Turke AB, Zejnullahu K, Wu YL, Song Y, Dias‑Santagata D, Hamid O, Daud A, van den Eertwegh A, Cranmer L, O'Day S,
Lifshits E, Toschi L, Rogers A, Mok T, Sequist L, et al: Puzanov I, et al: Health‑related quality of life in the randomised
Preexistence and clonal selection of MET amplification in EGFR KEYNOTE‑002 study of pembrolizumab versus chemotherapy
mutant NSCLC. Cancer Cell 17: 77‑88, 2010. in patients with ipilimumab‑refractory melanoma. Eur J
73. Engelman JA, Zejnullahu K, Mitsudomi T, Song Y, Hyland C, Cancer 67: 46‑54, 2016.
Park JO, Lindeman N, Gale CM, Zhao X, Christensen J, et al: 88. Nosrati A, Tsai KK, Goldinger SM, Tumeh P, Grimes B, Loo K,
MET amplification leads to gefitinib resistance in lung cancer by Algazi AP, Nguyen‑Kim TDL, Levesque M, Dummer R, et al:
activating ERBB3 signaling. Science 316: 1039‑1043, 2007. Evaluation of clinicopathological factors in PD‑1 response: deri‑
74. Guix M, Faber AC, Wang SE, Olivares MG, Song Y, Qu S, vation and validation of a prediction scale for response to PD‑1
Rinehart C, Seidel B, Yee D, Arteaga CL and Engelman JA: monotherapy. Br J Cancer 116: 1141‑1147, 2017.
Acquired resistance to EGFR tyrosine kinase inhibitors in 89. Zhang Y, Liu B, Kotenko S and Li W: Prognostic value of
cancer cells is mediated by loss of IGF‑binding proteins. J Clin neutrophil‑lymphocyte ratio and lactate dehydrogenase in mela‑
Invest 118: 2609‑2619, 2008. noma patients treated with immune checkpoint inhibitors: A
75. Paraiso KHT, Xiang Y, Rebecca VW, Abel EV, Chen YA, systematic review and meta‑analysis. Medicine (Baltimore) 101:
Mu n ko AC, Wo o d E, Fe doren ko I V, Sond a k V K, e29536, 2022.
Anderson AR, et al: PTEN loss confers BRAF inhibitor 90. Gershenwald JE, Scolyer RA, Hess KR, Sondak VK,
resistance to melanoma cells through the suppression of BIM Long GV, Ross MI, Lazar AJ, Faries MB, Kirkwood JM,
expression. Cancer Res 71: 2750‑2760, 2011. McArthur GA, et al: Melanoma staging: Evidence‑based changes
76. Maio M, Grob JJ, Aamdal S, Bondarenko I, Robert C, Thomas L, in the American joint committee on cancer eighth edition cancer
Garbe C, Chiarion‑Sileni V, Testori A, Chen TT, et al: Five‑year staging manual. CA Cancer J Clin 67: 472‑492, 2017.
survival rates for treatment‑naive patients with advanced mela‑ 91. Balch CM, Gershenwald JE, Soong SJ, Soong SJ, Thompson JF,
noma who received ipilimumab plus dacarbazine in a phase III Atkins MB, Byrd DR, Buzaid AC, Cochran AJ, Coit DG, et al:
trial. J Clin Oncol 33: 1191‑1196, 2015. Final version of 2009 AJCC melanoma staging and classifica‑
77. Eggermont AMM, Chiarion‑Sileni V, Grob JJ, Dummer R, tion. J Clin Oncol 27: 6199‑6206, 2009.
Wolchok JD, Schmidt H, Hamid O, Robert C, Ascierto PA, 92. Hauschild A, Engel G, Brenner W, Gläser R, Mönig H, Henze E
Richards JM, et al: Prolonged survival in stage III melanoma and Christophers E: S100B protein detection in serum is a signif‑
with ipilimumab adjuvant therapy. N Engl J Med 375: 1845‑1855, icant prognostic factor in metastatic melanoma. Oncology 56:
2016. 338‑344, 1999.
MEDICINE INTERNATIONAL 4: 13, 2024 17

93. Jury CS, McAllister EJ and MacKie RM: Rising levels of serum 114. Lei X, Guan CW, Song Y and Wang H: The multifaceted role
S100 protein precede other evidence of disease progression in of CD146/MCAM in the promotion of melanoma progression.
patients with malignant melanoma. Br J Dermatol 143: 269‑274, Cancer Cell Int 15: 3, 2015.
2000. 115. Pacifico MD, Grover R, Richman PI, Daley FM, Buffa F and
94. Mårtenson ED, Hansson LO, Nilsson B, von Schoultz E, Wilson GD: Development of a tissue array for primary mela‑
Månsson Brahme E, Ringborg U and Hansson J: Serum S‑100b noma with long‑term follow‑up: Discovering melanoma cell
protein as a prognostic marker in malignant cutaneous mela‑ adhesion molecule as an important prognostic marker. Plast
noma. J Clin Oncol 19: 824‑831, 2001. Reconstr Surg 115: 367‑375, 2005.
95. Janka EA, Várvölgyi T, Sipos Z, Soós A, Hegyi P, Kiss S, 116. Weinstein D, Leininger J, Hamby C and Safai B: Diagnostic and
Dembrovszky F, Csupor D, Kéringer P, Pécsi D, et al: Predictive prognostic biomarkers in melanoma. J Clin Aesthet Dermatol 7:
performance of serum S100B versus LDH in melanoma patients: 13‑24, 2014.
A systematic review and meta‑analysis. Front Oncol 11: 772165, 117. Gimotty PA, Van Belle P, Elder DE, Murry T, Montone KT,
2021. Xu X, Hotz S, Raines S, Ming ME, Wahl P and Guerry D:
96. Friedman RC, Farh KKH, Burge CB and Bartel DP: Most Biologic and prognostic significance of dermal Ki67 expression,
mammalian mRNAs are conserved targets of microRNAs. mitoses, and tumorigenicity in thin invasive cutaneous mela‑
Genome Res 19: 92‑105, 2009. noma. J Clin Oncol 23: 8048‑8056, 2005.
97. Lim LP, Glasner ME, Yekta S, Burge CB and Bartel DP: 118. Ladstein RG, Bachmann IM, Straume O and Akslen LA: Ki‑67
Vertebrate microRNA genes. Science 299: 1540, 2003. expression is superior to mitotic count and novel proliferation
98. Lagos‑Quintana M, Rauhut R, Lendeckel W and Tuschl T: markers PHH3, MCM4 and mitosin as a prognostic factor in
Identification of novel genes coding for small expressed RNAs. thick cutaneous melanoma. BMC Cancer 10: 140, 2010.
Science 294: 853‑858, 2001. 119. Tu TJ, Ma MW, Monni S, Rose AE, Yee H, Darvishian F,
99. Lau NC, Lim LP, Weinstein EG and Bartel DP: An abundant class Polsky D, Berman RS, Shapiro RL, Pavlick AC, et al: A high
of tiny RNAs with probable regulatory roles in Caenorhabditis proliferative index of recurrent melanoma is associated with
elegans. Science 294: 858‑862, 2001. worse survival. Oncology 80: 181‑187, 2011.
100. Lee RC and Ambros V: An extensive class of small RNAs in 120. Kahn HJ, Bailey D and Marks A: Monoclonal antibody D2‑40,
Caenorhabditis elegans. Science 294: 862‑864, 2001. a new marker of lymphatic endothelium, reacts with Kaposi's
101. Pfeffer SR, Grossmann KF, Cassidy PB, Yang CH, Fan M, sarcoma and a subset of angiosarcomas. Mod Pathol 15:
Kopelovich L, Leachman SA and Pfeffer LM: Detection of 434‑440, 2002.
exosomal miRNAs in the plasma of melanoma patients. J Clin 121. Kahn HJ and Marks A: A new monoclonal antibody, D2‑40,
Med Res 4: 2012‑2027, 2015. for detection of lymphatic invasion in primary tumors. Lab
102. Lin N, Zhou Y, Lian X and Tu Y: Expression of microRNA‑106b Invest 82: 1255‑1257, 2002.
and its clinical significance in cutaneous melanoma. Genet Mol 122. Niakosari F, Kahn HJ, McCready D, Ghazarian D, Rotstein LE,
Res 14: 16379‑16385, 2015. Marks A, Kiss A and From L: Lymphatic invasion identified
103. Friedman EB, Shang S, de Miera EVS, Fog JU, Teilum MW, by monoclonal antibody D2‑40, younger age, and ulceration:
Ma MW, Berman RS, Shapiro RL, Pavlick AC, Hernando E, et al: Predictors of sentinel lymph node involvement in primary cuta‑
Serum microRNAs as biomarkers for recurrence in melanoma. neous melanoma. Arch Dermatol 144: 462‑467, 2008.
J Transl Med 10: 155, 2012. 123. Rittling SR and Chambers AF: Role of osteopontin in tumour
104. Wróblewska JP, Lach MS, Ustaszewski A, Kulcenty K, Ibbs M, progression. Br J Cancer 90: 1877‑1881, 2004.
Jagiełło I, Suchorska WM and Marszałek A: The potential role 124. Rudland PS, Platt‑Higgins A, El‑Tanani M, Silva Rudland S,
of selected miRNA in uveal melanoma primary tumors as early Barraclough R, Winstanley JH, Howitt R and West CR:
biomarkers of disease progression. Genes (Basel) 11: 271, 2020. Prognostic significance of the metastasis‑associated protein
105. Tsao SCH, Weiss J, Hudson C, Christophi C, Cebon J, Behren A osteopontin in human breast cancer. Cancer Res 62: 3417‑3427,
and Dobrovic A: Monitoring response to therapy in melanoma 2002.
by quantifying circulating tumour DNA with droplet digital 125. Pan HW, Ou YH, Peng SY, Liu SH, Lai PL, Lee PH, Sheu JC,
PCR for BRAF and NRAS mutations. Sci Rep 5: 11198, 2015. Chen CL and Hsu HC: Overexpression of osteopontin is associ‑
106. Girotti MR, Gremel G, Lee R, Galvani E, Rothwell D, Viros A, ated with intrahepatic metastasis, early recurrence, and poorer
Mandal AK, Lim KH, Saturno G, Furney SJ, et al: Application prognosis of surgically resected hepatocellular carcinoma.
of sequencing, liquid biopsies, and patient‑derived xenografts Cancer 98: 119‑127, 2003.
for personalized medicine in melanoma. Cancer Discov 6: 126. Rangel J, Nosrati M, Torabian S, Shaikh L, Leong SP, Haqq C,
286‑299, 2016. Miller JR II, Sagebiel RW and Kashani‑Sabet M: Osteopontin
107. Clark WH Jr, Elder DE, Guerry D IV, Braitman LE, Trock BJ, as a molecular prognostic marker for melanoma. Cancer 112:
Schultz D, Synnestvedt M and Halpern AC: Model predicting 144‑150, 2008.
survival in stage I melanoma based on tumor progression. J Natl 127. Thomas NE, Edmiston SN, Alexander A, Groben PA, Parrish E,
Cancer Inst 81: 1893‑1904, 1989. Kricker A, Armstrong BK, Anton‑Culver H, Gruber SB,
108. Clemente CG, Mihm MC Jr, Bufalino R, Zurrida S, Collini P and From L, et al: Association between NRAS and BRAF mutational
Cascinelli N: Prognostic value of tumor infiltrating lymphocytes status and melanoma‑specific survival among patients with
in the vertical growth phase of primary cutaneous melanoma. higher‑risk primary melanoma. JAMA Oncol 1: 359‑368, 2015.
Cancer 77: 1303‑1310, 1996. 128. Cirenajwis H, Lauss M, Ekedahl H, Törngren T, Kvist A,
109. Mandalà M and Massi D: Tissue prognostic biomarkers in Saal LH, Olsson H, Staaf J, Carneiro A, Ingvar C, et al:
primary cutaneous melanoma. Virchows Arch 464: 265‑281, NF1‑mutated melanoma tumors harbor distinct clinical and
2014. biological characteristics. Mol Oncol 11: 438‑451, 2017.
110. Balch CM, Murad TM, Soong SJ, Ingalls AL, Halpern NB and 129. Schumacher TN and Schreiber RD: Neoantigens in cancer
Maddox WA: A multifactorial analysis of melanoma: Prognostic immunotherapy. Science 348: 69‑74, 2015.
histopathological features comparing Clark's and Breslow's 130. Va n A llen EM, M iao D, Sch illing B, Shu k la SA,
staging methods. Ann Surg 188: 732‑742, 1978. Blank C, Zimmer L, Sucker A, Hillen U, Foppen MHG,
111. Lattanzi M, Lee Y, Simpson D, Moran U, Darvishian F, Kim RH, Goldinger SM, et al: Genomic correlates of response to CTLA‑4
Hernando E, Polsky D, Hanniford D, Shapiro R, et al: Primary blockade in metastatic melanoma. Science 350: 207‑211, 2015.
melanoma histologic subtype: Impact on survival and response 131. Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM,
to therapy. J Natl Cancer Inst 111: 180‑188, 2019. Desrichard A, Walsh LA, Postow MA, Wong P, Ho TS, et al:
112. Robinson E, Kulkarni PM, Pradhan JS, Gartrell RD, Yang C, Genetic basis for clinical response to CTLA‑4 blockade in mela‑
Acs B, Rohr B, Phelps R, Ferringer T, Horst B, et al: Prediction noma. N Engl J Med 371: 2189‑2199, 2014.
of distant melanoma recurrence from primary tumor digital 132. Samstein RM, Lee CH, Shoushtari AN, Hellmann MD, Shen R,
H&E images using deep learning. J Clin Orthod 37 (15 Suppl): Janjigian YY, Barron DA, Zehir A, Jordan EJ, Omuro A, et al:
S9577, 2019. Tumor mutational load predicts survival after immunotherapy
113. Lehmann JM, Holzmann B, Breitbart EW, Schmiegelow P, across multiple cancer types. Nat Genet 51: 202‑206, 2019.
Riethmüller G and Johnson JP: Discrimination between benign 133. Johnson DB, Lovly CM, Flavin M, Panageas KS, Ayers GD,
and malignant cells of melanocytic lineage by two novel anti‑ Zhao Z, Iams WT, Colgan M, DeNoble S, Terry CR, et al:
gens, a glycoprotein with a molecular weight of 113,000 and Impact of NRAS mutations for patients with advanced mela‑
a protein with a molecular weight of 76,000. Cancer Res 47: noma treated with immune therapies. Cancer Immunol Res 3:
841‑845, 1987. 288‑295, 2015.
18 SHAH et al: IMMUNE CHECKPOINT INHIBITORS IN MELANOMA

134. Johnson DB, Bordeaux J, Kim JY, Vaupel C, Rimm DL, 151. Yang H, Xia L, Chen J, Zhang S, Martin V, Li Q, Lin S, Chen J,
Ho TH, Joseph RW, Daud AI, Conry RM, Gaughan EM, et al: Calmette J, Lu M, et al: Stress‑glucocorticoid‑TSC22D3
Quantitative spatial profiling of PD‑1/PD‑L1 interaction and axis compromises therapy‑induced antitumor immunity. Nat
HLA‑DR/IDO‑1 predicts improved outcomes of anti‑PD‑1 ther‑ Med 25: 1428‑1441, 2019.
apies in metastatic melanoma. Clin Cancer Res 24: 5250‑2560, 152. Altan‑Bonnet G and Mukherjee R: Cytokine‑mediated commu‑
2018. nication: A quantitative appraisal of immune complexity. Nat
135. Joh nson DB, Est rada M V, Sa lgado R, Sa nchez V, Rev Immunol 19: 205‑217, 2019.
Doxie DB, Opalenik SR, Vilgelm AE, Feld E, Johnson AS, 153. Eisenring M, vom Berg J, Kristiansen G, Saller E and Becher B:
Greenplate AR, et al: Melanoma‑specific MHC‑II expres‑ IL‑12 initiates tumor rejection via lymphoid tissue‑inducer
sion represents a tumour‑autonomous phenotype and predicts cells bearing the natural cytotoxicity receptor NKp46. Nat
response to anti‑PD‑1/PD‑L1 therapy. Nat Commun 7: 10582, Immunol 11: 1030‑1038, 2010.
2016. 154. Cristiani CM, Capone M, Garofalo C, Madonna G, Mallardo D,
136. Ro d ig SJ, Gusen leit ner D, Jack son D G, Gji n i E, Tuffanelli M, Vanella V, Greco M, Foti DP, Viglietto G, et al:
Giobbie‑Hurder A, Jin C, Chang H, Lovitch SB, Horak C, Altered frequencies and functions of innate lymphoid cells in
Weber JS, et al: MHC proteins confer differential sensitivity to melanoma patients are modulated by immune checkpoints
CTLA‑4 and PD‑1 blockade in untreated metastatic melanoma. inhibitors. Front Immunol 13: 811131, 2002.
Sci Transl Med 10: eaar3342, 2018. 155. Joshi K, Atwal D, Ravilla R, Pandey Y, Yarlagadda N, Kakadia S,
137. Chowell D, Morris LGT, Grigg CM, Weber JK, Samstein RM, Makhoul I, Hutchins L and Mahmoud F: Immunotherapy outcomes
Makarov V, Kuo F, Kendall SM, Requena D, Riaz N, et al: Patient in advanced melanoma in relation to age. Perm J 24: 19.093, 2020.
HLA class I genotype influences cancer response to checkpoint 156. Seidel JA, Otsuka A and Kabashima K: Anti‑PD‑1 and
blockade immunotherapy. Science 359: 582‑587, 2018. anti‑CTLA‑4 therapies in cancer: Mechanisms of action, effi‑
138. Sanlorenzo M, Vujic I, Floris A, Novelli M, Gammaitoni L, cacy, and limitations. Front Oncol 8: 86, 2018.
Giraudo L, Macagno M, Leuci V, Rotolo R, Donini C, et al: 157. Wei SC, Duffy CR and Allison JP: Fundamental mechanisms
BRAF and MEK inhibitors increase PD‑1‑positive mela‑ of immune checkpoint blockade therapy. Cancer Discov 8:
noma cells leading to a potential lymphocyte‑independent 1069‑1086, 2018.
synergism with anti‑PD‑1 antibody. Clin Cancer Res 24: 158. Jang SR, Nikita N, Banks J, Keith SW, Johnson JM, Wilson M
3377‑3385, 2018. and Lu‑Yao G: Association between sex and immune checkpoint
139. Eggermont AMM, Blank CU, Mandala M, Long GV, Atkinson V, inhibitor outcomes for patients with melanoma. JAMA Netw
Dalle S, Haydon A, Lichinitser M, Khattak A, Carlino MS, et al: Open 4: e2136823, 2021.
Adjuvant pembrolizumab versus placebo in resected stage III 159. Anstadt EJ, Chu B, Yegya‑Raman N, Han X, Doucette A,
melanoma. N Engl J Med 378: 1789‑1801, 2018. Poi r ier K, Moh iuddin JJ, Ma ity A, Facciabene A,
140. Rimm DL, Han G, Taube JM, Yi ES, Bridge JA, Flieder DB, Amaravadi RK, et al: Moderate colitis not requiring intrave‑
Homer R, West WW, Wu H, Roden AC, et al: A prospective, nous steroids is associated with improved survival in stage IV
multi‑institutional, pathologist‑based assessment of 4 immuno‑ melanoma after anti‑CTLA4 monotherapy, but not combination
histochemistry assays for PD‑L1 expression in non‑small cell therapy. Oncologist 27: 799‑808, 2022.
lung cancer. JAMA Oncol 3: 1051‑1058, 2017. 160. Jansen YJL, Rozeman EA, Mason R, Goldinger SM, Geukes
141. Rizk EM, Gartrell RD, Barker LW, Esancy CL, Finkel GG, Foppen MH, Hoejberg L, Schmidt H, van Thienen JV,
Bordbar DD and Saenger YM: Prognostic and predictive Haanen JBAG, Tiainen L, et al: Discontinuation of anti‑PD‑1
immunohistochemistry‑based biomarkers in cancer and immu‑ antibody therapy in the absence of disease progression or
notherapy. Hematol Oncol Clin North Am 33: 291‑299, 2019. treatment limiting toxicity: Clinical outcomes in advanced
142. Harel M, Ortenberg R, Varanasi SK, Mangalhara KC, melanoma. Ann Oncol 30: 1154‑1161, 2019.
Mardamshina M, Markovits E, Baruch EN, Tripple V, 161. Robert C, Ribas A, Hamid O, Daud A, Wolchok JD, Joshua AM,
Arama‑Chayoth M, Greenberg E, et al: Proteomics of melanoma Hwu WJ, Weber JS, Gangadhar TC, Joseph RW, et al: Durable
response to immunotherapy reveals mitochondrial dependence. complete response after discontinuation of pembrolizumab in
Cell 179: 236‑250.e18, 2019. patients with metastatic melanoma. J Clin Orthod 36: 1668‑1674,
143. Zhou X, Yu S, Zhao DM, Harty JT, Badovinac VP and Xue HH: 2018.
Differentiation and persistence of memory CD8(+) T cells 162. Betof Warner A, Palmer JS, Shoushtari AN, Goldman DA,
depend on T cell factor 1. Immunity 33: 229‑240, 2010. Panageas KS, Hayes SA, Bajwa R, Momtaz P, Callahan MK,
144. Kratchmarov R, Magun AM and Reiner SL: TCF1 expres‑ Wolchok JD, et al: Long‑term outcomes and responses to retreat‑
sion marks self‑renewing human CD8+ T cells. Blood Adv 2: ment in patients with melanoma treated with PD‑1 blockade.
1685‑1690, 2018. J Clin Oncol 38: 1655‑1663, 2020.
145. Im SJ, Hashimoto M, Gerner MY, Lee J, Kissick HT, Burger MC, 163. Waterhouse DM, Garon EB, Chandler J, McCleod M, Hussein M,
Shan Q, Hale JS, Lee J, Nasti TH, et al: Defining CD8+ T Jotte R, Horn L, Daniel DB, Keogh G, Creelan B, et al:
cells that provide the proliferative burst after PD‑1 therapy. Continuous versus 1‑year fixed‑duration nivolumab in previ‑
Nature 537: 417‑421, 2016. ously treated advanced non‑small‑cell lung cancer: CheckMate
146. Sade‑Feldman M, Yizhak K, Bjorgaard SL, Ray JP, de 153. J Clin Orthod 38: 3863‑3873, 2020.
Boer CG, Jenkins RW, Lieb DJ, Chen JH, Frederick DT, 164. Wang PF, Chen Y, Song SY, Wang TJ, Ji WJ, Li SW, Liu N
Barzily‑Rokni M, et al: Defining T cell states associated with and Yan CX: Immune‑related adverse events associated with
response to checkpoint immunotherapy in melanoma. Cell 175: anti‑PD‑1/PD‑L1 treatment for malignancies: A meta‑analysis.
998‑1013.e20, 2018. Front Pharmacol 8: 730, 2017.
147. Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, 165. Santini FC, Rizvi H, Plodkowski AJ, Ni A, Lacouture ME,
Hu‑Lieskovan S, Berent‑Maoz B, Pang J, Chmielowski B, Gambarin‑Gelwan M, Wilkins O, Panora E, Halpenny DF,
Cherry G, et al: Genomic and transcriptomic features of Long NM, et al: Safety and efficacy of re‑treating with immu‑
response to anti‑PD‑1 therapy in metastatic melanoma. Cell 165: notherapy after immune‑related adverse events in patients with
35‑44, 2016. NSCLC. Cancer Immunol Res 6: 1093‑1099, 2018.
148. Ott PA, Bang YJ, Piha‑Paul SA, Razak ARA, Bennouna J, 166. Lebbé C, Meyer N, Mortier L, Marquez‑Rodas I, Robert C,
Soria JC, Rugo HS, Cohen RB, O'Neil BH, Mehnert JM, et al: Rutkowski P, Menzies AM, Eigentler T, Ascierto PA,
T‑cell‑inflamed gene‑expression profile, programmed death Smylie M, et al: Evaluation of two dosing regimens for nivolumab
ligand 1 expression, and tumor mutational burden predict effi‑ in combination with ipilimumab in patients with advanced
cacy in patients treated with pembrolizumab across 20 cancers: melanoma: Results from the phase IIIb/IV CheckMate 511 trial.
KEYNOTE‑028. J Clin Oncol 37: 318‑327, 2019. J Clin Oncol 37: 867‑875, 2019.
149. Ayers M, Lunceford J, Nebozhyn M, Murphy E, Loboda A, 167. Tarhini AA, Lee SJ, Hodi FS, Rao UNM, Cohen GI, Hamid O,
Kaufman DR, Albright A, Cheng JD, Kang SP, Shankaran V, et al: Hutchins LF, Sosman JA, Kluger HM, Eroglu Z, et al: Phase III
IFN‑γ‑related mRNA profile predicts clinical response to PD‑1 study of adjuvant ipilimumab (3 or 10 mg/kg) versus high‑dose
blockade. J Clin Invest 127: 2930‑2940, 2017. interferon alfa‑2b for resected high‑risk melanoma: North
150. Pinato DJ, Howlett S, Ottaviani D, Urus H, Patel A, Mineo T, American intergroup E1609. J Clin Oncol 38: 567‑575, 2020.
Brock C, Power D, Hatcher O, Falconer A, et al: Association of 168. Betof AS, Nipp RD, Giobbie-Hurder A, Johnpulle RAN, Rubin K,
prior antibiotic treatment with survival and response to immune Rubinstein SM, Flaherty KT, Lawrence DP, Johnson DB and
checkpoint inhibitor therapy in patients with cancer. JAMA Sullivan RJ: Impact of age on outcomes with immunotherapy
Oncol 5: 1774‑1778, 2019. for patients with melanoma. Oncologist 22: 963‑971, 2017.
MEDICINE INTERNATIONAL 4: 13, 2024 19

169. Zamami Y, Niimura T, Okada N, Koyama T, Fukushima K, 184. Watanabe S, Kimura H, Takato H, Waseda Y, Hara J, Sone T,
Izawa‑Ishizawa Y and Ishizawa K: Factors associated with Abo M, Maeda S, Matsushita T and Kasahara K: Severe pneu‑
immune checkpoint inhibitor‑related myocarditis. JAMA monitis after nivolumab treatment in a patient with melanoma.
Oncol 5: 1635‑1637, 2019. Allergol Int 65: 487‑489, 2016.
170. Ta n M H, Iyenga r R, M izoka m i‑Stout K, Yentz S, 185. Nishino M, Sholl LM, Hatabu H, Ramaiya NH and Hodi FS:
MacEachern MP, Shen LY, Redman B and Gianchandani R: Anti‑PD‑1‑related pneumonitis during cancer immunotherapy.
Spectrum of immune checkpoint inhibitors‑induced endocri‑ N Engl J Med 373: 288‑290, 2015.
nopathies in cancer patients: A scoping review of case reports. 186. Mir MA: T‑cell costimulation and its applications in diseases.
Clin Diabetes Endocrinol 5: 1, 2019. Dev Costimulatory Mol Immunother Dis 29: 255‑292, 2015.
171. Wright JJ, Powers AC and Johnson DB: Endocrine toxicities of 187. Tarazona R, Duran E and Solana R: Natural killer cell
immune checkpoint inhibitors. Nat Rev Endocrinol 17: 389‑399, recognition of melanoma: New clues for a more effective immu‑
2021. notherapy. Front Immunol 6: 649, 2015.
172. Minkis K, Garden BC, Wu S, Pulitzer MP and Lacouture ME: 188. Rieth J and Subramanian S: Mechanisms of intrinsic tumor
The risk of rash associated with ipilimumab in patients with resistance to immunotherapy. Int J Mol Sci 19: 1340, 2018.
cancer: A systematic review of the literature and meta‑analysis. 189. Fisher DT, Appenheimer MM and Evans SS: The two faces
J Am Acad Dermatol 69: e121‑e128, 2013. of IL‑6 in the tumor microenvironment. Semin Immunol 26:
173. Coleman E, Ko C, Dai F, Tomayko MM, Kluger H and 38‑47, 2014.
Leventhal JS: Inflammatory eruptions associated with immune 190. O'Donnell JS, Long GV, Scolyer RA, Teng MWL and Smyth MJ:
checkpoint inhibitor therapy: A single‑institution retrospective Resistance to PD1/PDL1 checkpoint inhibition. Cancer Treat
analysis with stratification of reactions by toxicity and implica‑ Rev 52: 71‑81, 2017.
tions for management. J Am Acad Dermatol 80: 990‑997, 2019. 191. Vander Heiden MG, Cantley LC and Thompson CB:
174. Sibaud V, Meyer N, Lamant L, Vigarios E, Mazieres J and Understanding the Warburg effect: The metabolic requirements
Delord JP: Dermatologic complications of anti‑PD‑1/PD‑L1 of cell proliferation. Science 324: 1029‑1033, 2009.
immune checkpoint antibodies. Curr Opin Oncol 28: 254‑263, 192. Ahn CS and Metallo CM: Mitochondria as biosynthetic facto‑
2016. ries for cancer proliferation. Cancer Metab 3: 1, 2015.
175. Sibaud V: Dermatologic reactions to immune checkpoint 193. O'Donnell JS, Teng MWL and Smyth MJ: Cancer immunoed‑
inhibitors: Skin toxicities and immunotherapy. Am J Clin iting and resistance to T cell‑based immunotherapy. Nat Rev
Dermatol 19: 345‑361, 2018. Clin Oncol 16: 151‑167, 2019.
176. Inno A, Metro G, Bironzo P, Grimaldi AM, Grego E, Di 194. DeBerardinis RJ and Chandel NS: Fundamentals of cancer
Nunno V, Picasso V, Massari F and Gori S: Pathogenesis, metabolism. Sci Adv 2: e1600200, 2016.
clinical manifestations and management of immune checkpoint 195. Robey IF, Lien AD, Welsh SJ, Baggett BK and Gillies RJ:
inhibitors toxicity. Tumori 103: 405‑421, 2017. Hypoxia‑inducible factor‑1alpha and the glycolytic phenotype
177. Kumar V, Chaudhary N, Garg M, Floudas CS, Soni P and in tumors. Neoplasia 7: 324‑330, 2005.
Chandra AB: Current diagnosis and management of immune 196. Franco F, Jaccard A, Romero P, Yu YR and Ho PC: Metabolic
related adverse events (irAEs) Induced by immune checkpoint and epigenetic regulation of T‑cell exhaustion. Nat Metab 2:
inhibitor therapy. Front Pharmacol 8: 49, 2017. 1001‑1012, 2020.
178. Bryce J and Boers‑Doets CB: Non‑rash dermatologic adverse 197. Jiang Y, Li Y and Zhu B: T‑cell exhaustion in the tumor micro‑
events related to targeted therapies. Semin Oncol Nurs 30: environment. Cell Death Dis 6: e1792, 2015.
155‑168, 2014. 198. Sirico M, D'Angelo A, Gianni C, Casadei C, Merloni F and De
179. Geisler AN, Phillips GS, Barrios DM, Wu J, Leung DYM, Giorgi U: Current state and future challenges for PI3K inhibitors
Moy AP, Kern JA and Lacouture ME: Immune checkpoint in cancer therapy. Cancers (Basel) 15: 703, 2023.
inhibitor‑related dermatologic adverse events. J Am Acad 199. Peng W, Chen JQ, Liu C, Malu S, Creasy C, Tetzlaff MT, Xu C,
Dermatol 83: 1255‑1268, 2020. McKenzie JA, Zhang C, Liang X, et al: Loss of PTEN promotes
180. Tewalt EF, Cohen JN, Rouhani SJ, Guidi CJ, Qiao H, Fahl SP, resistance to T cell‑mediated immunotherapy. Cancer Discov 6:
Conaway MR, Bender TP, Tung KS, Vella AT, et al: Lymphatic 202‑216, 2016.
endothelial cells induce tolerance via PD‑L1 and lack of costim‑ 200. Li X, Wenes M, Romero P, Huang SCC, Fendt SM and Ho PC:
ulation leading to high‑level PD‑1 expression on CD8 T cells. Navigating metabolic pathways to enhance antitumour immu‑
Blood 120: 4772‑4782, 2012. nity and immunotherapy. Nat Rev Clin Oncol 16: 425‑441, 2019.
181. Sano T, Uhara H, Mikoshiba Y, Kobayashi A, Uchiyama R, 201. Marin‑Acevedo JA, Kimbrough EO and Lou Y: Next genera‑
Tateishi K, Yamamoto H and Okuyama R: Nivolumab‑induced tion of immune checkpoint inhibitors and beyond. J Hematol
organizing pneumonia in a melanoma patient. Jpn J Clin Oncol 14: 45, 2021.
Oncol 46: 270‑272, 2016.
182. Nakashima K, Naito T, Omori S, Yoshikawa S, Endo M,
Kiyohara Y and Takahashi T: Organizing pneumonia induced Copyright © 2024 Shah et al. This work is licensed
by nivolumab in a patient with metastatic melanoma. J Thorac u nder a Creat ive C om mons At t r ibut ion 4.0
Oncol 11: 432‑433, 2016. International (CC BY 4.0) License.
183. Koelzer VH, Rothschild SI, Zihler D, Wicki A, Willi B, Willi N,
Voegeli M, Cathomas G, Zippelius A and Mertz KD: Systemic
inflammation in a melanoma patient treated with immune
checkpoint inhibitors‑an autopsy study. J Immunother Cancer 4:
13, 2016.

You might also like

pFad - Phonifier reborn

Pfad - The Proxy pFad of © 2024 Garber Painting. All rights reserved.

Note: This service is not intended for secure transactions such as banking, social media, email, or purchasing. Use at your own risk. We assume no liability whatsoever for broken pages.


Alternative Proxies:

Alternative Proxy

pFad Proxy

pFad v3 Proxy

pFad v4 Proxy