Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Feb 1.
Published in final edited form as: Am J Clin Dermatol. 2020 Feb;21(1):1–11. doi: 10.1007/s40257-019-00475-1

Biomarkers predictive of survival and response to immune checkpoint inhibitors (ICI) in melanoma

Emanuelle M Rizk 1, Angelina M Seffens 2, Megan H Trager 2, Michael R Moore 3, Larisa J Geskin 4, Robyn D Gartrell-Corrado 5, Winston Wong 1, Yvonne M Saenger 1
PMCID: PMC6994371  NIHMSID: NIHMS1541132  PMID: 31602560

Abstract

Immunotherapy has revolutionized the treatment of melanoma. Targeting of the immune checkpoints cytotoxic T-lymphocyte-associated protein 4 and programmed cell death protein 1 has led to improved survival in a subset of patients. Unfortunately, the use of immune checkpoint inhibitors is associated with significant side effects and many patients do not response to treatment. Thus, there is an urgent need both for prognostic biomarkers to estimate risk and for predictive biomarkers to determine which patients are likely to respond to therapy. In this review, prognostic and predictive biomarkers that are an active area of research are outlined. Of note, certain transcriptomic signatures are already used in the clinic, albeit not routinely, to prognosticate patients. In the predictive setting, programmed cell death protein ligand 1 expression has been shown to correlate with benefit but is not precise enough to be used as an exclusionary biomarker. Future investigation will need to focus on biomarkers that are easily reproducible, cost-effective, and accurate. The use of readily available clinical material, such as serum or hematoxylin and eosin-stained images, may offer one such path forward.

1. Introduction

In the United States, melanoma is the fifth most common type of cancer; an estimated 96,480 new cases of melanoma will be diagnosed in 2019.1 Incidence rates for melanoma are still increasing,1 and as such an increased focus on both prevention and treatment of the disease is urgent. The development of immunotherapy has revolutionized the treatment of melanoma. In particular, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) immune checkpoint inhibitors (ICI) such as ipilimumab, nivolumab, and pembrolizumab have shown significant survival benefits in patients with advanced melanoma. In patients with unresectable stage III or IV melanoma, treatment with the CTLA-4 checkpoint blockade agent ipilimumab led to improved overall survival (OS) compared to treatment with gp100 peptide vaccine (10.1 months and 6.4 months, respectively).2 For patients with previously untreated BRAF wild type stage III melanoma, treatment with the PD1 checkpoint blockade agent nivolumab resulted in a 40.0% objective response rate (ORR) and progression-free survival (PFS) of 5.1 months, compared to chemotherapy with 13.0% ORR and 2.2 month PFS.3 Pembrolizumab, another PD-1 inhibitor, has shown prolonged PFS relative to ipilimumab in patients with unresectable stage III or IV melanoma, with a 6-month estimated PFS of 47.3% using biweekly pembrolizumab and 26.5% using ipilimumab.4,5 Such findings have led to the FDA approval of both pembrolizumab and nivolumab for the treatment of patients with unresectable or metastatic melanoma and, more recently, for the adjuvant treatment of stage III disease.6 Ipilimumab has also been FDA approved for the treatment of unresectable or metastatic melanoma in patients 12 years and older and in the adjuvant setting.7 In 2015, the combination of nivolumab and ipilimumab was also FDA approved in patients with unresectable or metastatic melanoma based on data indicating that such combination treatment led to a 61% ORR as opposed to an 11% ORR with ipilimumab alone.8

Despite the success of ICI in the treatment of melanoma, these treatments are also associated with serious adverse events such as colitis.9 For example, a phase 3 study evaluating combined ipilimumab with nivolumab against single agent nivolumab and single agent ipilimumab found that 59%, 21%, and 28% of patients experienced a grade 3 or 4 treatment-related adverse event, respectively.10 Thus, the development of biomarkers is critical to helping clinicians determine whether patients’ possible benefits from immunotherapy outweigh potential toxicities. Such biomarkers may play a key role in the adjuvant setting in identifying which patients have a low risk of recurrence and should be spared treatment.

Biomarkers for cancer can be subdivided into two categories: prognostic and predictive. While prognostic biomarkers aim to estimate a patient’s prognosis (e.g., whether that patient will recur) regardless of therapy, predictive biomarkers are meant to determine a patient’s response to a specific therapy, such as ICI.11,12 This review summarizes current prognostic biomarkers and predictive biomarkers for ICI in melanoma and highlights biomarkers that are an active focus of research.

Traditional prognostic biomarkers for melanoma include the histological factors involved in disease staging, such as depth of invasion, ulceration, and presence of microsatellite lesions.13 The Tumor, Node, Metastasis (TNM) classification in particular is used clinically to assess a patient’s risk category based on tumor thickness and ulceration (T category), lymph node involvement and presence of microsatellites (N category), and presence of sites of distant metastases (M category).13 Tumor depth and ulceration are both inversely correlated with survival, with thicker tumors and ulcerated tumors being associated with lower rates of survival. Tumor thickness and ulceration also act as independent predictors of metastases to the sentinel lymph node (SLN).14,15 SLN metastases remain the most important predictors of recurrence in patients with primary T2 and T3 tumors, and thus SLN biopsies are routinely used in these patients to determine nodal involvement.16,17 In thin melanomas, guidelines from the American Society for Clinical Oncology (ASCO) recommend that SLN biopsy be considered in patients with high-risk features, such as ulceration and high mitotic rate.18 However, given the cost of biopsy, its potential morbidity, and the low incidence of SLN metastasis in patients with thin melanoma, a more concrete understanding of predictors of SLN positivity would be useful in clinical decision-making.1922

2. Determining risk: prognostic biomarkers

A number of biomarkers have been examined to prognosticate patients with melanoma aside from SLN positivity. Many have been extensively validated and may thus prove useful in stratifying patients into risk groups. Several of the biomarkers discussed below, such as tumor infiltrating lymphocytes (TILs), osteopontin, and lymphatic invasion, have also been examined for correlation to SLN positivity. Such biomarkers may be of particular use in thin melanomas (under 1 millimeter), where the SLN positivity rate is only around 5%23 and where stratification of patients could help avoid unnecessary costs and potential morbidity of SLN biopsy.

2.1. Tissue-based biomarkers

2.1.1. Pathology-based biomarkers

TILs, which are visible via hematoxylin and eosin (H&E) staining, have been extensively examined both as prognostic biomarkers and as biomarkers predictive of SLN positivity. TIL patterns are classified as grades: absent, with no lymphocytes in the tumor; non-brisk, with a foci or multiple foci of lymphocytes within the tumor; and brisk, with diffuse lymphocyte infiltration within the tumor.24 In early stage melanoma, the presence of brisk TILs in vertical growth phase tumors is associated with the most favorable disease-specific survival (DSS) and OS, followed by non-brisk and absent TILs, as demonstrated by Clark et al. in 1989.24,25 More recently, a study of 1,865 patients with a single melanoma greater than or equal to 0.75 mm in thickness found that TILs were independent predictors of SLN status.26 The study demonstrated a clear inverse relationship between TIL grade and SLN status, with a positive SLN biopsy in 27.8% of patients with absent TILs, as opposed to a positive SLN biopsy in 5.6% of patients with brisk TILs.26

Melanomas have significant morphologic diversity. A melanoma can be classified as superficial spreading, nodular, lentigo maligna, or acral lentiginous.27 For the most part, melanoma histotypes have not been found to be prognostic when considered independently from tumor thickness and are thus not included in the American Join Committee on Cancer (AJCC) staging system.13,28,29 However, a recent paper has challenged this belief by finding that nodular melanoma was an independent predictor for recurrence and death from melanoma.30

Most recently, deep learning-based computational methods have been developed to predict a patient’s likelihood of recurrence based on morphological features in digitized pathology images. Some of these methods are intended to assess tumor subtype,31 while others have used deep learning to accurately grade a tumor.32 In melanoma, we have developed one such deep learning-based biomarker that stratifies patients with stage I, II, and III melanoma into risk groups.33 This biomarker correlates with DSS in two independent validation cohorts, and with further validation may be used to accurately predict prognosis in patients with early stage melanoma.33

2.1.2. Protein biomarkers

Cell adhesion proteins are important in the development of metastatic disease, and as such have been examined as potential prognostic biomarkers. Melanoma cell adhesion molecule (MCAM), the predominant cell adhesion marker in melanoma, is expressed in over 80% of metastatic tumors but is rarely expressed on benign nevi.34 MCAM positive melanoma is associated with a significantly worse 5-year survival than MCAM negative melanoma, and survival decreases with increased expression of this marker.35,36

Another promising protein biomarker is the expression of Ki-67. Ki-67 is a nuclear antigen that is a marker of cellular proliferation during the active stages of the cell cycle.37 In thin melanoma (<1 mm), both Ki-67 expression and increased mitotic rate are associated with increased risk of metastasis.38 Of note, mitotic rate was recently removed from the AJCC staging system for thin melanomas given that tumor thickness and ulceration were stronger predictors of survival, although assessing mitotic rate is still recommended across melanomas of different thicknesses.13 However, in thick melanoma, Ki-67 may be a better prognostic indicator than mitotic rate, and its expression is associated with increased melanoma thickness, presence of tumor ulceration, necrosis, increased Clark’s level of invasion, and vascular invasion.39 Additionally, in recurrent melanomas, high Ki-67 expression was found to be independently associated with decreased OS.40

The presence of lymphatic invasion has been shown to act as a predictor of locoregional cutaneous recurrence.41 D2–40 is an antibody to a sialoglycoprotein and is selective for the endothelium of lymphatic vessels.42,43 In a study of primary melanomas with a thickness equal to or greater than 1mm, lymphatic invasion as assessed by D2–40 staining was associated with SLN metastasis.44 The same association between lymphatic invasion and SLN positivity has been reported in a study focusing on thin to intermediate depth melanomas.45

Osteopontin, an integrin-binding protein that drives IL-17 expression and that is overexpressed in many malignancies including colorectal, lung, breast, liver, and prostate cancer has been identified as another potential biomarker associated with tumor progression and metastasis.4648 In a cohort of 345 primary melanoma patients examined by immunohistochemical (IHC) analysis of a tissue microarray, osteopontin expression was found to correlate with an increased risk of SLN positivity and was an independent predictor of prognosis in melanoma.49 Several other studies have also found osteopontin to be overexpressed in melanoma and that its presence at higher levels is associated with disease progression.5053

Previous studies have identified macrophages at the leading edge of the tumor to be prognostic in early-stage melanoma.54 In addition, in depth analyses of TILs have found that CD8+ cytotoxic T lymphocytes (CTLs) are best associated with survival.55 Closer evaluation using quantitative immunofluorescence (qIF) allows for location-specific concatenation of markers. Using qIF, a study of 104 primary stage II-III melanomas combined CTL and macrophage density in the stroma, finding that the CTL/macrophage ratio demonstrated improved prognostic power for both OS and DSS over independent CTL and macrophage density.56

2.1.3. Genomic biomarkers

Gene expression profiles can provide additional information for risk stratification in patients with melanoma. The DecisionDx®-Melanoma Prognostic test (Castle Biosciences) is one such gene expression profile that was developed to classify patients with early melanoma (stage I and II) into low-risk (class 1) or high-risk (class 2) groups based on the expression of 31 genes in primary melanoma.57 In studies using it in conjunction with the AJCC clinical staging system, DecisionDx®-Melanoma was shown to improve identification of patients with stage I and II disease who have a high risk of developing recurrent or metastatic disease.58 However, this gene expression profile has not been widely adopted in melanoma management. Another gene expression profile has been developed specifically for primary stage II-III melanoma, which is a diagnosis of significant clinical uncertainty and thus for which risk stratification is especially important in guiding therapy. Melanoma immune profile (MIP), a 53-immune gene transcriptomic signature score, predicts the risk of distant metastatic recurrence and DSS in patients with stage II-III melanoma.59 In creating a system for risk stratification, gene expression profiles provide an important clinical tool in guiding systemic therapy in melanoma.

2.1.4. Genetic biomarkers

Somatic cancer genomes have a large number of mutations within individual tumors and, in one study that used whole-genome sequencing, melanoma was found to be the most frequently mutated tumor.60 Mutations that promote cancer progression are known as driver mutations. The most common driver mutations associated with the development of melanoma occur in the BRAF, NRAS, and NF1 genes. These mutations lead to oncogenic activation of the MAPK pathway, which is involved in cell growth, proliferation, and survival.61

BRAF mutations have been identified in 66% of melanomas, the most common being V600E, which constitutes approximately 80% of those mutations.62 NRAS is mutated in approximately 15–25% of melanomas,63 and NF1 is mutated in approximately 14% of these tumors.64 Each of these driver mutations impacts disease prognosis.

In high risk tumors (≥stage T2b), BRAF and NRAS are associated with a significantly lower melanoma specific survival.65 NF1 mutations are also associated with a lower DSS and OS.66 In addition to providing prognostic information, the identification of patients with BRAF mutations is important in directing therapy. Targeted therapy, including dabrafenib (BRAF inhibitor) plus trametinib (MEK inhibitor), improves outcomes in patients with resected stage III melanoma with the BRAF mutation. Patients treated with a combination of dabrafenib plus trametinib have increased OS and metastasis-free survival, highlighting the importance of identifying these mutations in patients with melanoma.67 Finally, activating mutations in the KIT gene, although less common than the mutations described above, arise most often in mucosal or acral lentiginous melanomas and can be treated with imatinib, a kinase inhibitor.68,69

2.2. Serum biomarkers

Serum biomarkers have the potential to provide a minimally invasive means of determining a patient’s prognosis. Lactate dehydrogenase (LDH) catalyzes the conversion of pyruvate to lactate, and is thus important for tumor metabolism.70 Several studies have pointed to high LDH levels as an unfavorable prognostic factor in patients with cancer.71,72 A meta-analysis of 76 solid tumor studies found that higher serum LDH was associated with poor survival, with melanoma being one of the tumor types in which this association was most significant.73 Studies that have investigated the role of LDH in melanoma prognosis have confirmed this association.7476 Serum LDH is included as an independent prognostic biomarker for metastatic melanoma in the AJCC staging system, and until recently high levels of serum LDH upstaged patients with metastatic disease to stage IV M1c, which includes patients with metastases to the lungs and other visceral sites, excluding the central nervous system.13,77

Similarly, S100B is a soluble calcium-binding protein that is secreted into the bloodstream by malignant melanocytes.78 S100B levels in the serum have been shown to be dependent on tumor load and several studies have found that high levels of S100B may act as prognostic biomarkers and evidence of disease progression.7981

miRNAs have also been proposed as potential serum biomarkers in melanoma. Ever since their discovery in 2001, micro-RNAs have been extensively studied as post-transcriptional repressors of translation.8286 These short, noncoding RNAs have been found to be detectable and stable in exosomes in the serum.87 Expression of various miRNAs at abnormal levels has been shown to correlate with disease stage and recurrence.88,89

Finally, circulating tumor DNA (ctDNA) found in the plasma has been investigated as a means to determine disease status in patients with advanced melanoma.90 This ctDNA can both provide insights into total tumor load and into tumor-associated mutations. Studies have shown that evaluation of circulating DNA can assist in detection of BRAFV600 mutations, thus providing a useful tool for monitoring the efficacy of treatment with BRAF/MEK inhibitors.91,92 Disease progression and treatment response as assessed by imaging scans have both been shown to correlate with levels of ctDNA that increase or decrease, respectively.93,94

3. Predicting response to ICI: predictive biomarkers

As discussed above, a number of prognostic biomarkers have been developed in melanoma. Further, mutation of the BRAF gene is a widely used predictive biomarker for treatment with targeted therapy. On the other hand, the development of predictive biomarkers for response to ICI remains in a more experimental phase. No predictive biomarkers are yet in routine use for ICI in melanoma; a selection of promising biomarkers is discussed below.

3.1. Tissue-based biomarkers

3.1.1. Pathology-based biomarkers

As discussed above, the use of artificial intelligence (AI) to gather information from digitized H&E slides is an active area of research.31,32 This technique has been extended to the development of predictive biomarkers as well. Recent data suggested that AI could potentially be used to predict the likelihood of response to ICI using pre-treatment digitized H&E slides.95

3.1.2. Protein biomarkers

Immune infiltration by CD8+ TILs has been proposed as a predictive biomarker for response to immunotherapy in patients with melanoma. In a study of patients with advanced melanoma treated with pembrolizumab, patients who experienced a tumor response were found to have a higher CD8+ T cell density at the invasive tumor margin.96 Another study similarly examined the role of CD8+ TILs in response to PD-1 inhibition, finding that pretreatment CD8+ lymphocytic infiltration as assessed by quantitative immunofluorescence was associated with prolonged survival after treatment.97

Because ICI such as nivolumab and pembrolizumab target PD-1, which is often expressed on TILs, there has been significant interest in the potential correlation between the expression of PD-L1 on tumor cells and response to these immunotherapies.98 Although PD-L1 is also expressed on cells of the myeloid lineage, its presence on tumor cells has garnered the most interest.99101 A tumor is generally defined as PD-L1 positive when 5% of its cells express the marker, and several studies have found that patients whose tumors express PD-L1 have a greater benefit from treatment with anti-PD1 therapy.3,102 One such study found that in melanoma patients treated with nivolumab, those with a PD-L1 positive tumor status had a median PFS of 14.0 months and an ORR of 57.5%, whereas those with PD-L1 negative tumors had a median PFS of 5.3 months and an ORR of 41.3%.98 Of note, PD-L1 levels are already commonly assessed in biopsies both in the clinic and in trials. However, treatment with PD-1 inhibitors has been shown to be effective both in patients with and without overexpression of PD-L1,103 and the quality of IHC staining for PD-L1 is not reliable enough for clinical application.11,104 This may in part be due to the fact that tumor heterogeneity may lead to heterogeneous expression of PD-L1, thus complicating interpretation of PD-L1 immunohistochemistry.105 As a result, PD-L1 expression is not currently used in the clinic as an exclusionary biomarker for immunotherapy treatment.106

Human Leukocyte Antigen (HLA) molecules are responsible for antigen presentation by both immune cells and cancer cells, and are thus key players in the immune system’s response to cancer.107 HLA class II molecules, which mediate presentation of foreign peptides and are generally found on antigen-presenting cells (APCs), are frequently expressed on melanoma cells.108 High expression of HLA-DR, a class II HLA molecule, was found to predict response to anti-PD-1 therapy in patients with metastatic melanoma.108

Because ICI is thought to target and activate TILs, there has been interest in further phenotyping the TILs that create a tumor environment receptive to ICI.96 TCF7, a transcription factor, has garnered interest as a potential predictive biomarker in TILs for immunotherapy.109,110 The presence of TCF7 has been shown to promote a central memory stem-like phenotype, with CD8+ cells that express higher levels of TCF7 having the capacity to both self-renew and to differentiate into effector cells.111,112 Further, TCF7-positive cells seem to be those that proliferate after treatment with anti-PD1.109 In line with this observation, expression of TCF7 was found to correlate with positive clinical outcome in patients with melanoma treated with ICI.110 Thus, with further validation TCF7 expression could be used as a predictive biomarker in melanoma patients treated with ICI, and may potentially be used as a target for treatment.

3.1.3. Genomic biomarkers

Several studies have investigated whether specific gene expression signatures could predict sensitivity or resistance to treatment with ICIs. For example, an IFNɣ-related 18-gene signature has been found to correlate with response to pembrolizumab in patients with melanoma and has been tested in a number of other cancer types including head and neck squamous cell carcinoma (HNSCC) and gastric cancer.113 This T cell-inflamed gene expression profile has been evaluated in solid tumors treated with pembrolizumab in the KEYNOTE 028 trial, validating that this gene expression pattern is associated with improved ORR.114 Another signature, the innate anti-PD-1 resistance (IPRES) signature, involves upregulation of genes involved in such processes as the mesenchymal transition, angiogenesis, and cell adhesion and is enriched in patients resistant to treatment with anti-PD1 checkpoint inhibition.115

3.1.4. Genetic biomarkers

A tumor’s mutational burden (TMB), which is generally assessed by whole exome sequencing, is a well-established predictive biomarker in cancer types such as non-small cell lung cancer.116118 Tumors with a high TMB are thought to be more susceptible to ICI because they express more neoantigens and can thus be more easily recognized and targeted by T cells.119 There has recently been an increasing interest in the potential for the TMB to predict response to immunotherapy in melanoma.120,121 A study that included 321 patients with melanoma who were treated with ICI found that a high TMB could predict improved survival after treatment. Of note, the TMB was not found to be prognostic, as there was no association between TMB and outcome in patients who were not treated with ICI.122

Specific driver mutations have also been found to correlate with response to treatment. Patients with activating NRAS mutations were found to respond more favorably to immunotherapy than patients without NRAS mutations.123 Further, the CheckMate 067 trial reported that patients with BRAF-mutant melanoma had a 62% 4-year OS with the combination of ipilimumab and nivolumab as opposed to 50% with nivolumab alone and 33% with ipilimumab alone.10,124 Thus, determining mutation status may inform treatment for patients with melanoma.

HLA class I molecules are used by cells to present ‘self,’ and therefore are one means by which tumor cells would present tumor-derived neoantigens to the immune system. However, many cancer cells lose their expression of HLA class I molecules due to selective pressure by the immune system, thus allowing them to evade detection.125 Because ICI relies on immune response to the presentation of tumor neoantigens,126 downregulation of HLA-I expression or lack of HLA-I heterozygosity (and thus presentation of a lower diversity of antigens) has been proposed as a predictive biomarker for ICIs.127,128 In an analysis of 1,535 patients with cancer, over 500 of whom had melanoma, HLA class I homozygosity in at least one locus was found to correlate with reduced survival in response to ICI.127

3.2. Serum biomarkers

In addition to its value as a prognostic biomarker, serum LDH has been found to be a predictive blood-based biomarker in melanoma patients receiving ICI. As mentioned above, high serum LDH levels have been widely investigated as a poor prognostic factor in many cancers including melanoma.13,75,129131 Recently there has been increasing interest in the potential of serum LDH to act as a predictive biomarker, as well. In a univariable analysis of patients treated with anti-PD-1 monotherapy, elevated LDH was found to be associated with a decreased response to anti-PD1 therapy.132 Other blood biomarkers such as absolute monocyte counts, myeloid-derived suppressor cells, and regulatory T cell frequency were also found to be associated with outcome in response to ipilimumab.133 Gene signatures predictive of response to immunotherapy have also been developed using peripheral blood, such as a four-gene signature predictive of survival in patients treated with tremelimumab, an anti-CTLA-4 agent.134

4. Discussion

The field of prognostic and predictive biomarkers in melanoma is very promising and it is anticipated that in the near future one or several of the biomarkers under development reviewed above may be integrated into standard clinical care algorithms.

In the prognostic setting, multiple biomarkers have been developed to help guide the clinician in melanoma risk assessment, especially in cases where the tumor thickness is less than 1 millimeter. However, no standardized recommendations exist at this time on how to best utilize such biomarkers together to assess disease risk. We believe that genomic biomarkers in particular hold the potential to change clinical management of early stage melanoma. For example, the DecisionDx®-Melanoma test, which is commercially available for stage I-II melanoma, improves identification of early-stage melanoma patients at high risk for developing metastatic disease. Thus, the use of such tests in addition to conventional staging methods may allow clinicians to adjust screening intervals and interventions based on an individual patient’s disease risk. These biomarkers may especially benefit from additional validation on robust sample sets such as tissues from E1697, the historical cooperative group trial of adjuvant interferon. Doing so may help establish their prognostic ability and allow for their integration into standard AJCC staging. Other biomarkers, such as protein-based biomarkers, will still require further investigation, while genetic biomarkers are firmly established in their ability to separate high-risk from low-risk patients.

In the predictive setting, while PD-L1 staining does generally correlate with benefit, it is not sufficiently accurate for wide scale clinical use due to problems such as tumor heterogeneity, and limited effective options are available beyond PD-1 blockade. However, several other biomarkers such as the tumor mutational burden, the IFNɣ-related gene expression profile, and driver mutations can be implemented using widely available and robust tests. Such biomarkers may be easily incorporated into clinical practice to help predict whether a patient will respond to immune checkpoint inhibition. Other biomarkers mentioned in this review such as LDH, CD8+ TILs, and HLA heterozygosity will need more testing standardization and evaluation, although they hold significant promise as treatment predictors in anti-PD-1 therapy.

Further investigation into biomarkers for melanoma will need to prioritize sensitivity, specificity, accuracy, precision, and cost. Utilizing biomarkers that rely on readily available material such as blood-based biomarkers or H&E-based biomarkers may offer a path forward as a means of systematically estimating patient risk and response in a time-efficient and easily reproducible manner.

Key points.

Biomarkers for melanoma are urgently needed to determine which patients are at highest risk for recurrence and which patients will most benefit from treatment with immune checkpoint inhibitors.

The development of prognostic and predictive biomarkers is an active focus of research in the melanoma field, but these biomarkers have yet to be implemented into routine clinical care.

Biomarkers that use readily available clinical material such as blood-based biomarkers or hematoxylin and eosin-based biomarkers may provide a quick, cost-effective way of developing biomarkers for melanoma.

Acknowledgments

Funding sources: The authors of this publication were supported by the National Institutes of Health through Grant Numbers R01FD006108 (Y.M. Saenger) and KL2TR001874 (R.D. Gartrell-Corrado). Yvonne Saenger is also supported by funding from the Melanoma Research Alliance and by an Irving Assistant Professorship at Columbia University’s NIH/NCATS CTSA Program hub: UL1TR001873. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. Robyn Gartrell-Corrado is also supported by Swim Across America. Megan Trager is supported by the Dean’s Research Fellowship at Columbia. Angelina Seffens is supported by the Howard Hughes Medical Institute Medical Student Fellowship. The funding sources had no role preparation of the manuscript or the decision to submit for publication.

Compliance with Ethical Standards

Conflicts of interest: E.M. Rizk, A.M. Seffens, M.H. Trager, and M.R. Moore declare that they have no conflicts of interest that might be relevant to the contents of this manuscript. E.M. Rizk received travel funding to a conference from nanoString. Y.M. Saenger receives funding from Amgen and Regeneron. R.D. Gartrell-Corrado receives funding from nanoString and travel support and honoraria from Northwest Biotherapeutics and PerkinElmer. L.J. Geskin receives funding, honoraria, travel support, and royalties from Helsinn, Mallincrodt, Kyowa Kirin, Medivir, Medscape, and UpToDate. L.J. Geskin has also provided expert testimony to Medivir. None of these funding sources have impacted the contents of this manuscript.

References

  • 1.Siegel RL, Miller KD, Jemal A: Cancer statistics, 2019. CA Cancer J Clin 69:7–34, 2019 [DOI] [PubMed] [Google Scholar]
  • 2.Hodi FS, O’Day SJ, McDermott DF, et al. : Improved survival with ipilimumab in patients with metastatic melanoma. N Engl J Med 363:711–23, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Robert C, Long GV, Brady B, et al. : Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 372:320–30, 2015 [DOI] [PubMed] [Google Scholar]
  • 4.Robert C, Schachter J, Long GV, et al. : Pembrolizumab versus Ipilimumab in Advanced Melanoma. N Engl J Med 372:2521–32, 2015 [DOI] [PubMed] [Google Scholar]
  • 5.Administration FaD: FDA approves pembrolizumab for adjuvant treatment of melanoma, 2019
  • 6.Administration FaD: FDA grants regular approval to nivolumab for adjuvant treatment of melanoma, (ed 12/21/2017), 2017
  • 7.Administration FaD: Yervoy (ipilimumab) prescribing information, 2017
  • 8.Postow MA, Chesney J, Pavlick AC, et al. : Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med 372:2006–17, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Weber J, Mandala M, Del Vecchio M, et al. : Adjuvant Nivolumab versus Ipilimumab in Resected Stage III or IV Melanoma. N Engl J Med 377:1824–1835, 2017 [DOI] [PubMed] [Google Scholar]
  • 10.Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. : Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N Engl J Med 377:1345–1356, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Rizk EM, Gartrell RD, Barker LW, et al. : Prognostic and Predictive Immunohistochemistry-Based Biomarkers in Cancer and Immunotherapy. Hematol Oncol Clin North Am 33:291–299, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Oldenhuis CN, Oosting SF, Gietema JA, et al. : Prognostic versus predictive value of biomarkers in oncology. Eur J Cancer 44:946–53, 2008 [DOI] [PubMed] [Google Scholar]
  • 13.Gershenwald JE, Scolyer RA, Hess KR, et al. : Melanoma staging: Evidence-based changes in the American Joint Committee on Cancer eighth edition cancer staging manual. CA Cancer J Clin 67:472–492, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Rousseau DL Jr., Ross MI, Johnson MM, et al. : Revised American Joint Committee on Cancer staging criteria accurately predict sentinel lymph node positivity in clinically node-negative melanoma patients. Ann Surg Oncol 10:569–74, 2003 [DOI] [PubMed] [Google Scholar]
  • 15.Munsch C, Lauwers-Cances V, Lamant L, et al. : Breslow thickness, clark index and ulceration are associated with sentinel lymph node metastasis in melanoma patients: a cohort analysis of 612 patients. Dermatology 229:183–9, 2014 [DOI] [PubMed] [Google Scholar]
  • 16.Gershenwald JE, Thompson W, Mansfield PF, et al. : Multi-institutional melanoma lymphatic mapping experience: the prognostic value of sentinel lymph node status in 612 stage I or II melanoma patients. J Clin Oncol 17:976–83, 1999 [DOI] [PubMed] [Google Scholar]
  • 17.Wong SL, Hurley P, Lyman GH: Sentinel Lymph Node Biopsy for Melanoma: American Society of Clinical Oncology and Society of Surgical Oncology Joint Clinical Practice Guideline . J Oncol Pract 8:e65–e66, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Wong SL, Balch CM, Hurley P, et al. : Sentinel lymph node biopsy for melanoma: American Society of Clinical Oncology and Society of Surgical Oncology joint clinical practice guideline. J Clin Oncol 30:2912–8, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Andtbacka RH, Gershenwald JE: Role of sentinel lymph node biopsy in patients with thin melanoma. J Natl Compr Canc Netw 7:308–17, 2009 [DOI] [PubMed] [Google Scholar]
  • 20.Han D, Zager JS, Shyr Y, et al. : Clinicopathologic predictors of sentinel lymph node metastasis in thin melanoma. J Clin Oncol 31:4387–93, 2013 [DOI] [PubMed] [Google Scholar]
  • 21.Wat H, Senthilselvan A, Salopek TG: A retrospective, multicenter analysis of the predictive value of mitotic rate for sentinel lymph node (SLN) positivity in thin melanomas. J Am Acad Dermatol 74:94–101, 2016 [DOI] [PubMed] [Google Scholar]
  • 22.Herbert G, Karakousis GC, Bartlett EK, et al. : Transected thin melanoma: Implications for sentinel lymph node staging. J Surg Oncol 117:567–571, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Warycha MA, Zakrzewski J, Ni Q, et al. : Meta-analysis of sentinel lymph node positivity in thin melanoma (<or=1 mm). Cancer 115:869–79, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Clark WH Jr., Elder DE, Guerry Dt, et al. : Model predicting survival in stage I melanoma based on tumor progression. J Natl Cancer Inst 81:1893–904, 1989 [DOI] [PubMed] [Google Scholar]
  • 25.Clemente CG, Mihm MC Jr., Bufalino R, et al. : Prognostic value of tumor infiltrating lymphocytes in the vertical growth phase of primary cutaneous melanoma. Cancer 77:1303–10, 1996 [DOI] [PubMed] [Google Scholar]
  • 26.Azimi F, Scolyer RA, Rumcheva P, et al. : Tumor-infiltrating lymphocyte grade is an independent predictor of sentinel lymph node status and survival in patients with cutaneous melanoma. J Clin Oncol 30:2678–83, 2012 [DOI] [PubMed] [Google Scholar]
  • 27.Jelfs PL, Giles G, Shugg D, et al. : Cutaneous malignant melanoma in Australia, 1989. Med J Aust 161:182–7, 1994 [DOI] [PubMed] [Google Scholar]
  • 28.Mandala M, Massi D: Tissue prognostic biomarkers in primary cutaneous melanoma. Virchows Arch 464:265–81, 2014 [DOI] [PubMed] [Google Scholar]
  • 29.Balch CM, Murad TM, Soong SJ, et al. : A multifactorial analysis of melanoma: prognostic histopathological features comparing Clark’s and Breslow’s staging methods. Ann Surg 188:732–42, 1978 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Lattanzi M, Lee Y, Simpson D, et al. : Primary Melanoma Histologic Subtype: Impact on Survival and Response to Therapy. J Natl Cancer Inst 111:180–188, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Coudray N, Ocampo PS, Sakellaropoulos T, et al. : Classification and mutation prediction from non-small cell lung cancer histopathology images using deep learning. Nat Med 24:1559–1567, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Arvaniti E, Fricker KS, Moret M, et al. : Automated Gleason grading of prostate cancer tissue microarrays via deep learning. Sci Rep 8:12054, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Robinson E, Kulkarni PM, Pradhan JS, et al. : Prediction of distant melanoma recurrence from primary tumor digital H&E images using deep learning. Journal of Clinical Oncology 37:9577–9577, 2019 [Google Scholar]
  • 34.Lehmann JM, Holzmann B, Breitbart EW, et al. : Discrimination between benign and malignant cells of melanocytic lineage by two novel antigens, a glycoprotein with a molecular weight of 113,000 and a protein with a molecular weight of 76,000. Cancer Res 47:841–5, 1987 [PubMed] [Google Scholar]
  • 35.Lei X, Guan C-W, Song Y, et al. : The multifaceted role of CD146/MCAM in the promotion of melanoma progression. Cancer cell international 15:3–3, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Pacifico MD, Grover R, Richman PI, et al. : Development of a tissue array for primary melanoma with long-term follow-up: discovering melanoma cell adhesion molecule as an important prognostic marker. Plast Reconstr Surg 115:367–75, 2005 [DOI] [PubMed] [Google Scholar]
  • 37.Weinstein D, Leininger J, Hamby C, et al. : Diagnostic and prognostic biomarkers in melanoma. The Journal of clinical and aesthetic dermatology 7:13–24, 2014 [PMC free article] [PubMed] [Google Scholar]
  • 38.Gimotty PA, Van Belle P, Elder DE, et al. : Biologic and Prognostic Significance of Dermal Ki67 Expression, Mitoses, and Tumorigenicity in Thin Invasive Cutaneous Melanoma. Journal of Clinical Oncology 23:8048–8056, 2005 [DOI] [PubMed] [Google Scholar]
  • 39.Ladstein RG, Bachmann IM, Straume O, et al. : Ki-67 expression is superior to mitotic count and novel proliferation markers PHH3, MCM4 and mitosin as a prognostic factor in thick cutaneous melanoma. BMC Cancer 10:140, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Tu TJ, Ma MW, Monni S, et al. : A high proliferative index of recurrent melanoma is associated with worse survival. Oncology 80:181–187, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Borgstein PJ, Meijer S, van Diest PJ: Are locoregional cutaneous metastases in melanoma predictable? Ann Surg Oncol 6:315–21, 1999 [DOI] [PubMed] [Google Scholar]
  • 42.Kahn HJ, Bailey D, Marks A: Monoclonal antibody D2–40, a new marker of lymphatic endothelium, reacts with Kaposi’s sarcoma and a subset of angiosarcomas. Mod Pathol 15:434–40, 2002 [DOI] [PubMed] [Google Scholar]
  • 43.Kahn HJ, Marks A: A new monoclonal antibody, D2–40, for detection of lymphatic invasion in primary tumors. Lab Invest 82:1255–7, 2002 [DOI] [PubMed] [Google Scholar]
  • 44.Niakosari F, Kahn HJ, McCready D, et al. : Lymphatic invasion identified by monoclonal antibody D2–40, younger age, and ulceration: predictors of sentinel lymph node involvement in primary cutaneous melanoma. Arch Dermatol 144:462–7, 2008 [DOI] [PubMed] [Google Scholar]
  • 45.Fohn LE, Rodriguez A, Kelley MC, et al. : D2–40 lymphatic marker for detecting lymphatic invasion in thin to intermediate thickness melanomas: association with sentinel lymph node status and prognostic value-a retrospective case study. J Am Acad Dermatol 64:336–45, 2011 [DOI] [PubMed] [Google Scholar]
  • 46.Rittling SR, Chambers AF: Role of osteopontin in tumour progression. Br J Cancer 90:1877–81, 2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Rudland PS, Platt-Higgins A, El-Tanani M, et al. : Prognostic significance of the metastasis-associated protein osteopontin in human breast cancer. Cancer Res 62:3417–27, 2002 [PubMed] [Google Scholar]
  • 48.Pan HW, Ou YH, Peng SY, et al. : Overexpression of osteopontin is associated with intrahepatic metastasis, early recurrence, and poorer prognosis of surgically resected hepatocellular carcinoma. Cancer 98:119–27, 2003 [DOI] [PubMed] [Google Scholar]
  • 49.Rangel J, Nosrati M, Torabian S, et al. : Osteopontin as a molecular prognostic marker for melanoma. Cancer 112:144–50, 2008 [DOI] [PubMed] [Google Scholar]
  • 50.Zhou Y, Dai DL, Martinka M, et al. : Osteopontin expression correlates with melanoma invasion. J Invest Dermatol 124:1044–52, 2005 [DOI] [PubMed] [Google Scholar]
  • 51.Kiss T, Ecsedi S, Vizkeleti L, et al. : The role of osteopontin expression in melanoma progression. Tumour Biol 36:7841–7, 2015 [DOI] [PubMed] [Google Scholar]
  • 52.Kashani-Sabet M, Nosrati M, Miller JR 3rd, et al. : Prospective Validation of Molecular Prognostic Markers in Cutaneous Melanoma: A Correlative Analysis of E1690. Clin Cancer Res 23:6888–6892, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Kashani-Sabet M, Venna S, Nosrati M, et al. : A multimarker prognostic assay for primary cutaneous melanoma. Clin Cancer Res 15:6987–92, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Jensen TO, Schmidt H, Møller HJ, et al. : Macrophage Markers in Serum and Tumor Have Prognostic Impact in American Joint Committee on Cancer Stage I/II Melanoma. Journal of Clinical Oncology 27:3330–3337, 2009 [DOI] [PubMed] [Google Scholar]
  • 55.Erdag G, Schaefer JT, Smolkin ME, et al. : Immunotype and immunohistologic characteristics of tumor-infiltrating immune cells are associated with clinical outcome in metastatic melanoma. Cancer Res 72:1070–80, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Gartrell RD, Marks DK, Hart TD, et al. : Quantitative Analysis of Immune Infiltrates in Primary Melanoma. Cancer Immunol Res 6:481–493, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Cook RW, Middlebrook B, Wilkinson J, et al. : Analytic validity of DecisionDx-Melanoma, a gene expression profile test for determining metastatic risk in melanoma patients. Diagnostic pathology 13:13–13, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Ferris LK, Farberg AS, Middlebrook B, et al. : Identification of high-risk cutaneous melanoma tumors is improved when combining the online American Joint Committee on Cancer Individualized Melanoma Patient Outcome Prediction Tool with a 31-gene expression profile-based classification. J Am Acad Dermatol 76:818–825.e3, 2017 [DOI] [PubMed] [Google Scholar]
  • 59.Gartrell RD, Marks DK, Rizk EM, et al. : Validation of Melanoma Immune Profile (MIP), a Prognostic Immune Gene Prediction Score for Stage II–III Melanoma. Clinical Cancer Research 25:2494, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Lawrence MS, Stojanov P, Polak P, et al. : Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 499:214–218, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Burotto M, Chiou VL, Lee JM, et al. : The MAPK pathway across different malignancies: a new perspective. Cancer 120:3446–56, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Davies H, Bignell GR, Cox C, et al. : Mutations of the BRAF gene in human cancer. Nature 417:949–54, 2002 [DOI] [PubMed] [Google Scholar]
  • 63.Jakob JA, Bassett RL Jr, Ng CS, et al. : NRAS mutation status is an independent prognostic factor in metastatic melanoma. Cancer 118:4014–23, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Cancer Genome Atlas N: Genomic Classification of Cutaneous Melanoma. Cell 161:1681–96, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Thomas NE, Edmiston SN, Alexander A, et al. : Association Between NRAS and BRAF Mutational Status and Melanoma-Specific Survival Among Patients With Higher-Risk Primary Melanoma. JAMA Oncol 1:359–68, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Cirenajwis H, Lauss M, Ekedahl H, et al. : NF1-mutated melanoma tumors harbor distinct clinical and biological characteristics. Molecular oncology 11:438–451, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Long GV, Hauschild A, Santinami M, et al. : Adjuvant Dabrafenib plus Trametinib in Stage III BRAF-Mutated Melanoma. N Engl J Med 377:1813–1823, 2017 [DOI] [PubMed] [Google Scholar]
  • 68.Goldinger SM, Murer C, Stieger P, et al. : Targeted therapy in melanoma - the role of BRAF, RAS and KIT mutations. EJC Suppl 11:92–6, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Guo J, Si L, Kong Y, et al. : Phase II, open-label, single-arm trial of imatinib mesylate in patients with metastatic melanoma harboring c-Kit mutation or amplification. J Clin Oncol 29:2904–9, 2011 [DOI] [PubMed] [Google Scholar]
  • 70.Warburg O, Wind F, Negelein E: The Metabolism of Tumors in the Body. J Gen Physiol 8:519–30, 1927 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Zhang Y, Xu T, Wang Y, et al. : Prognostic Role of Lactate Dehydrogenase Expression in Urologic Cancers: A Systematic Review and Meta-Analysis. Oncol Res Treat 39:592–604, 2016 [DOI] [PubMed] [Google Scholar]
  • 72.Faloppi L, Del Prete M, Casadei Gardini A, et al. : The correlation between LDH serum levels and clinical outcome in advanced biliary tract cancer patients treated with first line chemotherapy. Sci Rep 6:24136, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Petrelli F, Cabiddu M, Coinu A, et al. : Prognostic role of lactate dehydrogenase in solid tumors: a systematic review and meta-analysis of 76 studies. Acta Oncol 54:961–70, 2015 [DOI] [PubMed] [Google Scholar]
  • 74.Gao D, Ma X: Serum lactate dehydrogenase is a predictor of poor survival in malignant melanoma. Panminerva Med 59:332–337, 2017 [DOI] [PubMed] [Google Scholar]
  • 75.de Lecea MV, Palomares T, Al Kassam D, et al. : Indoleamine 2,3 dioxygenase as a prognostic and follow-up marker in melanoma. A comparative study with LDH and S100B. J Eur Acad Dermatol Venereol 31:636–642, 2017 [DOI] [PubMed] [Google Scholar]
  • 76.Kelderman S, Heemskerk B, van Tinteren H, et al. : Lactate dehydrogenase as a selection criterion for ipilimumab treatment in metastatic melanoma. Cancer Immunol Immunother 63:449–58, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Balch CM, Gershenwald JE, Soong S-j, et al. : Final Version of 2009 AJCC Melanoma Staging and Classification. Journal of Clinical Oncology 27:6199–6206, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Ghanem G, Loir B, Morandini R, et al. : On the release and half-life of S100B protein in the peripheral blood of melanoma patients. Int J Cancer 94:586–90, 2001 [DOI] [PubMed] [Google Scholar]
  • 79.Hauschild A, Engel G, Brenner W, et al. : S100B protein detection in serum is a significant prognostic factor in metastatic melanoma. Oncology 56:338–44, 1999 [DOI] [PubMed] [Google Scholar]
  • 80.Jury CS, McAllister EJ, MacKie RM: Rising levels of serum S100 protein precede other evidence of disease progression in patients with malignant melanoma. Br J Dermatol 143:269–74, 2000 [DOI] [PubMed] [Google Scholar]
  • 81.Martenson ED, Hansson LO, Nilsson B, et al. : Serum S-100b protein as a prognostic marker in malignant cutaneous melanoma. J Clin Oncol 19:824–31, 2001 [DOI] [PubMed] [Google Scholar]
  • 82.Friedman RC, Farh KK, Burge CB, et al. : Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 19:92–105, 2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Lim LP, Glasner ME, Yekta S, et al. : Vertebrate microRNA genes. Science 299:1540, 2003 [DOI] [PubMed] [Google Scholar]
  • 84.Lagos-Quintana M, Rauhut R, Lendeckel W, et al. : Identification of novel genes coding for small expressed RNAs. Science 294:853–8, 2001 [DOI] [PubMed] [Google Scholar]
  • 85.Lau NC, Lim LP, Weinstein EG, et al. : An abundant class of tiny RNAs with probable regulatory roles in Caenorhabditis elegans. Science 294:858–62, 2001 [DOI] [PubMed] [Google Scholar]
  • 86.Lee RC, Ambros V: An extensive class of small RNAs in Caenorhabditis elegans. Science 294:862–4, 2001 [DOI] [PubMed] [Google Scholar]
  • 87.Pfeffer SR, Grossmann KF, Cassidy PB, et al. : Detection of Exosomal miRNAs in the Plasma of Melanoma Patients. J Clin Med 4:2012–27, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Lin N, Zhou Y, Lian X, et al. : Expression of microRNA-106b and its clinical significance in cutaneous melanoma. Genet Mol Res 14:16379–85, 2015 [DOI] [PubMed] [Google Scholar]
  • 89.Friedman EB, Shang S, de Miera EV, et al. : Serum microRNAs as biomarkers for recurrence in melanoma. J Transl Med 10:155, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Calapre L, Warburton L, Millward M, et al. : Circulating tumour DNA (ctDNA) as a liquid biopsy for melanoma. Cancer Lett 404:62–69, 2017 [DOI] [PubMed] [Google Scholar]
  • 91.Ascierto PA, Minor D, Ribas A, et al. : Phase II trial (BREAK-2) of the BRAF inhibitor dabrafenib (GSK2118436) in patients with metastatic melanoma. J Clin Oncol 31:3205–11, 2013 [DOI] [PubMed] [Google Scholar]
  • 92.Schreuer M, Meersseman G, Van Den Herrewegen S, et al. : Quantitative assessment of BRAF V600 mutant circulating cell-free tumor DNA as a tool for therapeutic monitoring in metastatic melanoma patients treated with BRAF/MEK inhibitors. J Transl Med 14:95, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Tsao SC, Weiss J, Hudson C, et al. : Monitoring response to therapy in melanoma by quantifying circulating tumour DNA with droplet digital PCR for BRAF and NRAS mutations. Sci Rep 5:11198, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Girotti MR, Gremel G, Lee R, et al. : Application of Sequencing, Liquid Biopsies, and Patient-Derived Xenografts for Personalized Medicine in Melanoma. Cancer Discov 6:286–99, 2016 [DOI] [PubMed] [Google Scholar]
  • 95.Johannet P, Coudray N, Jour G, et al. : Using machine learning algorithms to predict response and toxicity to immune checkpoint inhibitors (ICIs) in melanoma patients. Journal of Clinical Oncology 37:2581–2581, 2019. 31246523 [Google Scholar]
  • 96.Tumeh PC, Harview CL, Yearley JH, et al. : PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 515:568–71, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Wong PF, Wei W, Smithy JW, et al. : Multiplex Quantitative Analysis of Tumor-Infiltrating Lymphocytes and Immunotherapy Outcome in Metastatic Melanoma. Clin Cancer Res 25:2442–2449, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Larkin J, Chiarion-Sileni V, Gonzalez R, et al. : Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med 373:23–34, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Dong H, Strome SE, Salomao DR, et al. : Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med 8:793–800, 2002 [DOI] [PubMed] [Google Scholar]
  • 100.Herbst RS, Soria JC, Kowanetz M, et al. : Predictive correlates of response to the anti-PDL1 antibody MPDL3280A in cancer patients. Nature 515:563–7, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Taube JM, Klein A, Brahmer JR, et al. : Association of PD-1, PD-1 ligands, and other features of the tumor immune microenvironment with response to anti-PD-1 therapy. Clin Cancer Res 20:5064–74, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Weber JS, D’Angelo SP, Minor D, et al. : Nivolumab versus chemotherapy in patients with advanced melanoma who progressed after anti-CTLA-4 treatment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial. Lancet Oncol 16:375–84, 2015 [DOI] [PubMed] [Google Scholar]
  • 103.Eggermont AMM, Blank CU, Mandala M, et al. : Adjuvant Pembrolizumab versus Placebo in Resected Stage III Melanoma. N Engl J Med 378:1789–1801, 2018 [DOI] [PubMed] [Google Scholar]
  • 104.Rimm DL, Han G, Taube JM, et al. : A Prospective, Multi-institutional, Pathologist-Based Assessment of 4 Immunohistochemistry Assays for PD-L1 Expression in Non-Small Cell Lung Cancer. JAMA Oncol 3:1051–1058, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Madore J, Vilain RE, Menzies AM, et al. : PD-L1 expression in melanoma shows marked heterogeneity within and between patients: implications for anti-PD-1/PD-L1 clinical trials. Pigment Cell Melanoma Res 28:245–53, 2015 [DOI] [PubMed] [Google Scholar]
  • 106.Patel SP, Kurzrock R: PD-L1 Expression as a Predictive Biomarker in Cancer Immunotherapy. Mol Cancer Ther 14:847–56, 2015 [DOI] [PubMed] [Google Scholar]
  • 107.Rock KL, Reits E, Neefjes J: Present Yourself! By MHC Class I and MHC Class II Molecules. Trends Immunol 37:724–737, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Johnson DB, Estrada MV, Salgado R, et al. : Melanoma-specific MHC-II expression represents a tumour-autonomous phenotype and predicts response to anti-PD-1/PD-L1 therapy. Nat Commun 7:10582, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Im SJ, Hashimoto M, Gerner MY, et al. : Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537:417–421, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Sade-Feldman M, Yizhak K, Bjorgaard SL, et al. : Defining T Cell States Associated with Response to Checkpoint Immunotherapy in Melanoma. Cell 175:998–1013 e20, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Zhou X, Yu S, Zhao DM, et al. : Differentiation and persistence of memory CD8(+) T cells depend on T cell factor 1. Immunity 33:229–40, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Kratchmarov R, Magun AM, Reiner SL: TCF1 expression marks self-renewing human CD8(+) T cells. Blood Adv 2:1685–1690, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Ayers M, Lunceford J, Nebozhyn M, et al. : IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J Clin Invest 127:2930–2940, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Ott PA, Bang YJ, Piha-Paul SA, et al. : T-Cell-Inflamed Gene-Expression Profile, Programmed Death Ligand 1 Expression, and Tumor Mutational Burden Predict Efficacy in Patients Treated With Pembrolizumab Across 20 Cancers: KEYNOTE-028. J Clin Oncol 37:318–327, 2019 [DOI] [PubMed] [Google Scholar]
  • 115.Hugo W, Zaretsky JM, Sun L, et al. : Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell 165:35–44, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Rizvi NA, Hellmann MD, Snyder A, et al. : Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 348:124–8, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Hellmann MD, Nathanson T, Rizvi H, et al. : Genomic Features of Response to Combination Immunotherapy in Patients with Advanced Non-Small-Cell Lung Cancer. Cancer Cell 33:843–852 e4, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Hellmann MD, Ciuleanu TE, Pluzanski A, et al. : Nivolumab plus Ipilimumab in Lung Cancer with a High Tumor Mutational Burden. N Engl J Med 378:2093–2104, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Schumacher TN, Schreiber RD: Neoantigens in cancer immunotherapy. Science 348:69–74, 2015 [DOI] [PubMed] [Google Scholar]
  • 120.Van Allen EM, Miao D, Schilling B, et al. : Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 350:207–211, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Snyder A, Makarov V, Merghoub T, et al. : Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 371:2189–2199, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Samstein RM, Lee CH, Shoushtari AN, et al. : Tumor mutational load predicts survival after immunotherapy across multiple cancer types. Nat Genet 51:202–206, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Johnson DB, Lovly CM, Flavin M, et al. : Impact of NRAS mutations for patients with advanced melanoma treated with immune therapies. Cancer Immunol Res 3:288–295, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Hodi FS, Chiarion-Sileni V, Gonzalez R, et al. : Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. Lancet Oncol 19:1480–1492, 2018 [DOI] [PubMed] [Google Scholar]
  • 125.Garcia-Lora A, Algarra I, Garrido F: MHC class I antigens, immune surveillance, and tumor immune escape. J Cell Physiol 195:346–55, 2003 [DOI] [PubMed] [Google Scholar]
  • 126.Gubin MM, Zhang X, Schuster H, et al. : Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 515:577–81, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Chowell D, Morris LGT, Grigg CM, et al. : Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 359:582–587, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Rodig SJ, Gusenleitner D, Jackson DG, et al. : MHC proteins confer differential sensitivity to CTLA-4 and PD-1 blockade in untreated metastatic melanoma. Sci Transl Med 10, 2018. [DOI] [PubMed] [Google Scholar]
  • 129.Abusaif S, Jradi Z, Held L, et al. : S100B and lactate dehydrogenase as response and progression markers during treatment with vemurafenib in patients with advanced melanoma. Melanoma Res 23:396–401, 2013 [DOI] [PubMed] [Google Scholar]
  • 130.Egberts F, Kotthoff EM, Gerdes S, et al. : Comparative study of YKL-40, S-100B and LDH as monitoring tools for Stage IV melanoma. Eur J Cancer 48:695–702, 2012 [DOI] [PubMed] [Google Scholar]
  • 131.Weide B, Martens A, Hassel JC, et al. : Baseline Biomarkers for Outcome of Melanoma Patients Treated with Pembrolizumab. Clin Cancer Res 22:5487–5496, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Nosrati A, Tsai KK, Goldinger SM, et al. : Evaluation of clinicopathological factors in PD-1 response: derivation and validation of a prediction scale for response to PD-1 monotherapy. Br J Cancer 116:1141–1147, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Martens A, Wistuba-Hamprecht K, Geukes Foppen M, et al. : Baseline Peripheral Blood Biomarkers Associated with Clinical Outcome of Advanced Melanoma Patients Treated with Ipilimumab. Clin Cancer Res 22:2908–18, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Saenger Y, Magidson J, Liaw B, et al. : Blood mRNA expression profiling predicts survival in patients treated with tremelimumab. Clin Cancer Res 20:3310–8, 2014 [DOI] [PubMed] [Google Scholar]

RESOURCES