Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 3531 2023-01-30 18:32:40 |
2 format -1 word(s) 3530 2023-01-31 04:08:09 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Seyhan, A.A.;  Carini, C. Insights and Strategies of Melanoma Immunotherapy. Encyclopedia. Available online: https://encyclopedia.pub/entry/40614 (accessed on 02 July 2024).
Seyhan AA,  Carini C. Insights and Strategies of Melanoma Immunotherapy. Encyclopedia. Available at: https://encyclopedia.pub/entry/40614. Accessed July 02, 2024.
Seyhan, Attila A., Claudio Carini. "Insights and Strategies of Melanoma Immunotherapy" Encyclopedia, https://encyclopedia.pub/entry/40614 (accessed July 02, 2024).
Seyhan, A.A., & Carini, C. (2023, January 30). Insights and Strategies of Melanoma Immunotherapy. In Encyclopedia. https://encyclopedia.pub/entry/40614
Seyhan, Attila A. and Claudio Carini. "Insights and Strategies of Melanoma Immunotherapy." Encyclopedia. Web. 30 January, 2023.
Insights and Strategies of Melanoma Immunotherapy
Edit

Despite the successes and durable responses with immune checkpoint inhibitors (ICI), many cancer patients, including those with melanoma, do not derive long-term benefits from ICI therapies. The lack of predictive biomarkers to stratify patients to targeted treatments has been the driver of primary treatment failure and represents an unmet medical need in melanoma and other cancers. Understanding genomic correlations with response and resistance to ICI will enhance cancer patients’ benefits. Building on insights into interplay with the complex tumor microenvironment (TME), the ultimate goal should be assessing how the tumor ’instructs’ the local immune system to create its privileged niche with a focus on genomic reprogramming within the TME. It is hypothesized that this genomic reprogramming determines the response to ICI. Furthermore, emerging genomic signatures of ICI response, including those related to neoantigens, antigen presentation, DNA repair, and oncogenic pathways, are gaining momentum. 

immune checkpoint inhibitors melanoma responders and non-responders biomarkers

1. Introduction

Immune checkpoint inhibitors (ICIs) are revolutionizing the treatment of melanoma and other types of cancers, with 7 agents now approved in the US and a rich pipeline of new agents and mechanisms in development [1][2][3][4]. Notwithstanding the excitement around these developments, there is a significant unmet medical need in the form of patient stratification and therapy resistance. Melanoma affects more than 1 million Americans, and there is an increasing incidence of melanoma worldwide. Approx. 300,000 new cases are diagnosed in the US each year [5], with the average annual cost for treatment estimated at $3.3 billion [6].
Numerous successes have been achieved with anti-PD1, anti-CTLA4, or combination therapies [5][7] in treating melanoma and other types of cancers. The groundbreaking finding by Leach et al. [8] showed that antibodies blocking the T cell co-inhibitory receptor CTLA-4 can augment immune responses against tumor cells in mice. This finding gave rise to ipilimumab, the first ICI to increase the survival of patients with metastatic melanoma [9][10] which was granted FDA approval in 2011 for the treatment of metastatic melanoma.
In 2014, additional T cell immune checkpoint blocking antibodies, anti-PD-1 (pembrolizumab) and anti-PD-L1 (nivolumab) [11], received FDA approval. The combination of anti-PD-1 (pembrolizumab) and anti-PD-L1 (nivolumab) resulted in the augmented survival of patients with metastatic melanoma compared to patients treated with ipilimumab alone or chemotherapy alone [12][13][14].
In 2022, relatlimab, which targets lymphocyte-activation gene 3 (LAG-3), was approved by the FDA for adult and pediatric use with metastatic melanoma [15].
Following those developments, high levels of PD-L1 expression in cancer cells and tumor mutational burden (TMB) have been shown in melanoma to correlate with clinical responses to ICI [13].
Despite these data, a substantial number of melanoma patients fail to respond to ICI, leading to premature death [16][17][18]. Considerable effort has been made to identify biomarkers that predict clinical response/resistance to ICI [19]. Despite the successes of ICI, even with the combination of ipilimumab and nivolumab, the five-year survival for the intention-to-treat (ITT) population was only 53%. This means that 47% of patients do not reach long-lasting benefits and succumb to the disease [20], suggesting the need for predictive biomarkers of response to overcome ICI resistance. Presently, physicians have no idea which patients will or will not respond to ICI in the absence of predictive biomarkers of response.

2. Melanoma Biomarkers of Response to ICI

2.1. Established Clinical Biomarkers

The earliest approved clinical biomarkers that helped to inform the prognosis of metastatic melanoma relied on baseline clinical characteristics, such as serum levels of lactate dehydrogenase (LDH), a marker of tumor burden [21]. Elevated serum LDH levels have been demonstrated to be a negative prognostic marker, irrespective of the given treatment [22][23][24][25]. Generally, high LDH levels are associated with poor overall survival (OS) compared with normal LDH levels. Elevated levels of LDH have been used as a biomarker to assess patient staging [26]. For example, it has been shown that among ca. 30% of patients with 4–5-year OS following treatment with BRAF and MEK inhibitors, only a few of those patients had high LDH levels before the initiation of therapy [27].
Likewise, elevated serum levels of S100B have demonstrated to have a prognostic value in both metastatic [28][29] and high-risk resected melanoma settings [30].
Gene expression profiling has shown to have a prognostic value that complements existing biomarkers in patients with melanoma [27][31][32].
In metastatic melanoma, a well-characterized predictive biomarker of response guiding the therapeutic decision process is the BRAF V600 mutation, which is somehow predictive of response to BRAF ± MEK inhibition with low rates of primary resistance [33][34][35]. The response rate to BRAF and MEK inhibitors in metastatic melanoma patients with the BRAF V600 mutation is ca. 70% in selected patients, with less than 10% of patients having the highest response to progressive disease [35][36][37][38].
Several oncogenic driver mutations have been identified as predictive biomarkers of response from targeted agents, including NRAS, NF-1, and c-KIT, that provide insight into the probability of therapeutic response to a specific treatment [39][40][41][42][43]. Presently, the presence of a BRAF V600 mutation is the only validated predictive biomarker for melanoma patients.

2.2. Emerging Predictive Biomarkers

Predictive biomarkers of response include TMB, neoantigen load [7][44][45][46][47][48], HLA-I genotype [49][50], cytolytic activity [51], aneuploidy [52], and T cell repertoire [26], which exhibit high predictability to ICI response. Other predictive biomarkers of response are PDL-1 expression, LAG-3 expression, CD8+ T cells at tumor invasive margin, IL-17 expression, immune-related gene expression signatures, and T-cell receptor (TCR) signature.
Additional biomarkers that are used to assess ICI response include ctDNA profiles, absolute lymphocyte count, proliferating CD8+ T cells, increase of T-cell subsets and checkpoint molecules (PDL-1, LAG-3), granzyme B expression, and TCR signature [53][54][55][56].
Unfortunately, many of those potential biomarkers of ICI response have not yet been validated [57][58][59][60][61].
A recent study by Carter et al. [57] questioned the validity of the immuno-predictive score (IMPRES), a predictor of ICI response in melanoma consisting of 15 pairwise transcriptomic signatures that analyze the relationship between immune checkpoint genes reported by Auslander et al. [59]. The IMPRES is context-dependent and could not reproducibly predict ICI response in the context of metastatic melanoma [59].
Moreover, Xiao et al. [60] questioned the reproducibility of the Immune Cells.Sig [62] signature in melanoma, demonstrating inconsistencies in the prediction capability of ImmuneCells.Sig across different RNA-seq datasets [60]. The performance of the ImmuneCells.Sig signature in predicting ICI outcomes in four melanoma patient datasets, using the same implementation scheme as Xiong et al. [62], showed that there were inconsistencies across different datasets [60].

2.2.1. Gene Expression Signatures

Gene expression signatures (GES) have also been identified as predictive biomarkers of response to ICI and have been validated in several independent datasets (e.g., immune-predictive score, IMPRES consisting of 15 immune genes). IFN-γ-responsive genes were also used to predict ICI response in metastatic melanoma [26][51][52][59][63][64][65][66][67][68][69][70].
A panel of pan-tumor T cell-inflamed GES consisting of 18 IFN-γ-responsive genes was validated and confirmed to predict the response to ICI in pre-treatment tumor specimens from nine types of cancers, including melanoma [67].
MHC-I/II gene signatures have also been explored as predictive biomarkers of ICI response in melanoma [69][71][72].
Carter et al. [57] reported that immuno-predictive score (IMPRES), a predictor of ICI response in melanoma encompassing 15 pairwise transcriptomics relations between immune checkpoint gene [59], did not reproducibly predict the response to ICI in metastatic melanoma.
It was argued that many factors may contribute to the limited successes of those biomarkers, such as: (1) the predictive biomarkers have been derived from pre-clinical studies; (2) evaluation of the biomarkers in clinical specimens only included baseline biopsies and peripheral blood samples; (3) batch effect, lack of reproducibility might have contributed to the failure of the biomarkers for ICI response [57][58][60][61].
To address these issues, numerous researchers have developed predictive biomarkers to reduce batch noise and other technical issues. Expanded predictive biomarker panels have resulted in higher reproducibility as opposed to predictive signatures based on individual biomarkers [73][74][75][76].
Tian et al. [75] reported that the combined BRAF, KRAS, and PI3KCA mutation signature resulted in a favorable predictive response to cetuximab for patients with colorectal cancer [75].

2.2.2. Gene Expression Signatures at Baseline and On-Treatment Tumor Specimens

Genome-wide analysis of transcriptomic and genomic profiles of baseline and on-therapy tumor specimens from patients treated with ICI provides a comprehensive view into the mechanisms underlying tumor response and resistance to ICI [77].
Grasso et al. [77] reported that the mechanism of action of ICI is based on the interaction between immune effector cells and cancer cell targets. Tumor studies conducting comprehensive analyses of transcriptomic and genomic profiling have focused not only on the genetic alterations and gene expression profiles of cancer cells [63][69][72][78], but also on the composition of immune infiltrates and expression of immune-activating gene programs [26][46][58][64][66][67][69][70][72][79][80][81][82][83][84].
Du et al. [85] reported that pathway-based signatures derived from on-treatment tumor specimens were predictive of the response to anti-PD1 blockade in patients with metastatic melanoma.
Other studies of breast cancer suggested that post-treatment tumor samples were more informative than pre-treatment samples [86][87][88].
Conversely, Wallin et al. developed adaptive immune signatures based on tumor samples obtained during the early course of treatment, showing that the signatures were highly predictive of the response to ICI in patients with metastatic melanoma [89].
Auslander et al. built an immune-predictive score (IMPRES), which encompasses 15 pairwise transcriptomic relations between immune checkpoint genes, to predict the response of metastatic melanoma to ICB therapy [59].
The IMPRES signature produced better predictive scores with post-treatment samples than with pre-treatment samples in two independent datasets [59].
In support of these findings, a recent proteome profiling study of samples from patients with metastatic melanoma undergoing either tumor infiltrating lymphocyte based or anti-PD1 immunotherapy demonstrated that the fatty acid oxidation pathway was significantly enriched in responder patients. These results underlined the critical role of mitochondrial metabolism, including fatty acid metabolism, in conferring response to immunotherapy [90].
It is now widely accepted that post-treatment tumor specimens are generally much more informative than pre-treatment specimens and may provide more valuable insight into dynamic changes at the transcriptional level that correlate with clinical response, resulting in a higher predictive score.
In conclusion, although pathway signatures derived from post-treatment samples are highly predictive of therapeutic response to anti-PD1 in patients with metastatic melanoma, further studies are warranted to confirm the predictive value of those signatures in larger cohorts of patients with metastatic melanoma.

2.2.3. Pathway Signatures

Du et al. [85] developed pathway-specific signatures in pre-treatment (PASS-PRE) and on-treatment (PASS-ON) tumor specimens based on transcriptomic data and clinical information from a large dataset of metastatic melanoma patients treated with anti-PD1. Both PASS-PRE and PASS-ON signatures were validated in three independent datasets of metastatic melanoma. Compared to existing molecular signatures, it was concluded that the on-treatment (PASS-ON) tumor specimen signature exhibited a robust and better predictive value for metastatic melanoma patients who responded to anti-PD1 across all four datasets.
The pre-treatment pathway signatures included six pathways for predicting the response to anti-PD1 treatment, including: (1) complement cascade; (2) regulation of insulin-like growth factor IGF transport and uptake by insulin-like growth factor binding proteins IGFBPS; (3) binding and uptake of ligands by scavenger receptors; (4) plasma lipoprotein remodeling; (5) IL2 family signaling; and (6) retinoic acid (RA) biosynthesis pathways. Complement cascade, binding, and uptake of ligands by scavenger receptors and IL2 family signaling pathways are related to immune and inflammation, whereas plasma lipoprotein remodeling and the RA biosynthesis pathways are related to metabolism.
In contrast to the pathway-based signature analysis of on-treatment samples, Du et al. [85] identified four pathways, including: (1) peroxisomal lipid metabolism; (2) generation of second messenger molecules; (3) fatty acid metabolism; and (4) PD1 signaling. Of note, peroxisomal lipid metabolism and fatty acid metabolism are related to fatty acid and lipid metabolism [90]. Generation of second messenger molecules is a central signaling pathway in T-cell receptor (TCR) stimulation. Likewise, PD1 signaling plays an important role in immunoregulation as an immunoregulatory signaling pathway.
To further validate the predictive performance of a pathway-based super signature for on-treatment samples (PASS-ON), Du et al. [85] tested three independent datasets with RNA-seq data available for on-treatment samples, including those of Gide et al. and Lee et al. [82][91], and the MGH cohort demonstrated the effectiveness of PASS-ON in predicting patient response to anti-PD1. Patients with high PASS-ON signature scores were associated with significantly improved PFS compared to those with low signature scores in all tested patients.
Furthermore, Du et al. [85] demonstrated that the time-response interaction pathway-based super signature for pre- and on-treatment samples had reasonable predictive power. The study suggested that pathway-based biomarker signatures derived from on-treatment tumor specimens compared to pretreatment tumor specimens were better predictors of response to anti-PD1 therapies in metastatic melanoma patients.

2.2.4. Tumor Antigens

Huang et al. [92] investigated several melanoma-relevant tumor-specific antigens, cancer germline genes, melanocyte differentiation antigens, overexpressed antigens, neoantigens, neuropeptides, and other sources of immunogenic antigens, such as immunogenic epitopes as novel predictive biomarkers of ICI response to melanoma.
Neoantigens are derived from tumor-specific somatic mutations and are exclusively expressed in cancer cells and absent in normal human tissue. The majority (95%) of somatic mutations are single-nucleotide variants (SNVs), which lead to aberrant protein and peptide expression with single amino acid substitutions [93].
Neopeptides also arise from nucleotide insertions or deletions (indels), leading to the expression of aberrant proteins and peptides with frameshift or non-frameshift sequences depending on the number of nucleotides added and deleted. While the minority of mutations are indels (<5% for melanoma) [94][95], frameshift mutations can generate a number of immunogenic neoepitopes that are highly distinct from the self.
Other sources of immunogenic antigens, including immunogenic epitopes, can also derive from mutations associated with gene fusion, aberrant messenger-RNA splicing with retained introns, or aberrant translation resulting in cryptic antigens, and genomically integrated endogenous retroviral sequences as a result of previous retroviral infections, although they are epigenetically silenced, can be reactivated in tumors [94], as in the case of cancer germline antigens.
Furthermore, tumors often present aberrant patterns of DNA methylation, resulting in the demethylation, ectopic expression, and presentation of cancer germline genes to T cells relevant in immune recognition [94][96].
For example, cancer germline genes such as MAGEA1 and NY-ESO-1 are silenced epigenetically through methylation in human tissue, with the exception of male germ cells and trophoblastic cells, which lack MHC-I molecules.
PRAME (preferentially expressed antigen in melanoma), a member of the cancer-testis antigen family, has been reported to be frequently overexpressed in many cancers, including melanoma, which indicates advanced cancer stages and poor clinical prognosis [97][98]. As such, overexpressed PRAME is a potential immunotherapy target. PRAME-specific immunotherapies are currently in development for many cancers, including melanoma. For example, a recent study demonstrated that uterine carcinosarcoma, synovial sarcoma, and leiomyosarcoma patients would potentially benefit from PRAME-specific immunotherapies [97].

2.2.5. Genomic Alterations

Considerable effort has been made to identify genomic alterations and transcriptome profiles as predictive biomarkers of ICI response. Numerous studies have identified distinct stages of CD8+ T cells linked to positive response or failure to ICI treatment [84].
Moreover, tumors from patients responding to ICI showed a higher number of cancer-associated somatic mutations (i.e., mutated antigens or neoantigens) targeted by T cells [44]. The IFN-γ signature has also been shown to predict the response to ICI (e.g., anti-PD-1) in melanoma [99] and in other types of cancers [6][77]. Gene expression signatures obtained from bulk melanoma tumor or single-cell profiling and the TME have been shown to be correlated with sensitivity and resistance to several ICIs [70][72][84][100][101][102].
Collectively, the gene expression signatures associated with response to ICI in metastatic melanoma represent distinct characteristics and play an important function in different signaling pathways, including the inflammatory response, type I interferon signaling pathway, cytokines, and others.

3. Who Is Responding to ICI?

Recent data have demonstrated that immunotherapies against immune checkpoints (e.g., CTLA-4 or PD-1) downregulate two main negative regulators of the anti-tumor immune response [77][103][104][105], resulting in durable anti-tumor responses in a subset of cancer patients, including those with melanoma [2][106].
Another key factor contributing to anti-tumor immune response following ICI treatment [77] is the pre-existing level of T cell infiltration of the tumor [107][108][109], representing the immunogenicity of the cancer cells.
Analysis of tumor biopsies from ICI-treated patients showed that clinical responses associated with ICI were mediated by tumor-infiltrating T cells reactivated following ICI treatment [109][110].
A combination of immunohistochemical (IHC) analysis with RNA-seq performed on cancer biopsies from patients treated with anti-CTLA-4 antibody (ipilimumab) before or after treatment with anti-PD-1 antibody (nivolumab) demonstrated that a major response to anti-CTLA-4 requires cancer cells with high levels of MHC-I expression at baseline, whereas the response to anti-PD-1 was more strongly associated with a pre-existing interferon-γ gene expression signature [72].
Liu et al. demonstrated that the response to anti-PD-1 therapy (with or without prior anti-CTLA-4 treatment) was associated with increased MHC-I and MHC-II expression [69].
Biopsies from patients with metastatic melanoma treated with anti-PD-1 monotherapy (nivolumab) in part 1 of the CheckMate 038 study showed an increase in immune cell subtypes with elevated immune activation gene signatures seen in responders to therapy [78].
The transcriptome analysis of tumor biopsies from patients treated with anti-PD-1 monotherapy (nivolumab) or in combination anti-PD1 plus anti-CTLA-4 therapy (ipilimumab) correlated well with the in vitro analysis of gene expression signatures of melanoma cell lines following exposure to interferon-γ [77].
It appears that cancer cells become enablers of the immune response via the expression of IFN-γ-response genes, triggering the upregulation of antigen presentation, amplification of the interferon response, and induction of chemokines (i.e., CXCL9 and CXCL10) to entice immune cells to the TME. Thus, T cell-induced IFN-γ correlates with ICI therapy response [77]. Collectively, the degree of the anti-tumor T cell response and downstream IFN-γ signaling are the main drivers of response or resistance to ICI therapy [77].
However, little is known about how tumor-intrinsic loss of IFN-γ signaling impacts TILs. The question remains whether tumor-intrinsic IFN-γ signaling actively regulates the infiltration or function of TILs?
Shen et al. [111] demonstrated that IFN-γR1 knockout melanomas and IFN-γR1 knockout melanomas in B6 mice had reduced infiltration and function of tumor-infiltrating lymphocytes (TILs). Furthermore, long-distance effects of IFN-γ on tumor cells also play a crucial role in anti-tumor immunity [111]. These recent findings revealed an important role of tumor-intrinsic IFN-γ signaling and IFN-γ-response genes in shaping TILs.

4. Therapeutic Strategies to Turn “Cold Tumors” into “Hot Tumors”

Several strategies have been investigated to elucidate the mechanisms underlying how T cells are driven into “hot tumors” in order to improve the efficacy of ICI therapy [112]. Several clinical trials have tested these novel therapeutic modalities as interventions in combination with ICI to overcome ICI monotherapy resistance and attempt to turn “cold tumors’ into “hot tumors”.
Grasso et al. [77] showed that a robust anti-tumor immune response relies on the interplay of key factors that can be modulated with innovative interventions.
Recent preclinical and clinical findings have provided insight into the immunological implications of canonical cancer signaling pathways (e.g., WNT-beta-catenin signaling, cell cycle regulatory signaling, mitogen-activated protein kinase signaling, and pathways activated by loss of the tumor suppressor phosphoinositide phosphatase PTEN), thus providing new opportunities for the development of new treatments for those patients who do not respond to ICI monotherapies [113].
Combined therapeutic strategies from preclinical [103][105][114] and clinical studies [14] have shown that anti-PD-1 plus anti-CTLA-4 (nivolumab and ipilimumab) treatments elicit stronger immune stimulation in stage IV melanoma than monotherapy alone, resulting in a favorable anti-tumor immune response. Response to ICI, either following anti-CTLA-4 monotherapy or in combination with anti-PD-1 therapy, triggers a robust T cell response that generated an appreciable antitumor response [113].
Another approach to turn “cold tumors” into “hot tumors” is through the intra-tumoral delivery of oncolytic viruses or Toll-like receptor agonists capable of inducing intra-tumoral interferon production, which triggers the pattern recognition pathways with consequent boosting of the anti-PD1 immune response rate [115][116][117].
Recently, different approaches have been used to boost the response to ICI. Activation of the STING pathway [118], inhibition of immune suppressive factors (e.g., WNT signaling or the adenosine pathway) [99][119][120][121], as well as the release of other immune checkpoints (e.g., LAG-3, TIM-3, or TIGIT, etc.) in T cells [99] have been explored, but no favorable clinical outcomes have yet been reported in patients.
Altogether, these new therapeutic strategies provide new opportunities for cancer immunotherapy for patients who do not respond to ICI [99].

5. Lessons Learned: ICI Therapies in Melanoma

Huang et al. [92] reported that several factors, including the immune TME, tumor-associated immune cells, and different host factors, contribute to the ICI resistance. Melanoma resistance has armed people with a handful of information that can be applied to other types of cancers, such as: (1) the ability to present cancer antigens through MHC-I and elevated TMB; (2) tumor-antigen-specific T cells play a crucial role in the response to ICI; (3) reactivation of terminally exhausted T cells could be considered a biomarker for PD-1 blockade, which is detectable as early as one week after ICI dosing; (4) melanoma immunosuppressive mechanisms are complicated and need additional research to remodel their interaction, cooperation, and dynamics during tumor progression and in immunotherapy resistance; (5) Treg cells are emerging as a key mechanism of resistance to PD-1 blockade, but not necessarily CTLA-4 blockade; (6) emerging neoadjuvant immunotherapy trials are anticipated to provide new insight into pharmacodynamic immune responses and advance the development of rational immunotherapy and neoadjuvant combination regimens while avoiding toxicity and significantly improving patient management; (7) longitudinal assessment of pre- and on-treatment patient specimens is required to determine prognostic vs. predictive use of immune and other parameters, including genomic parameters correlating with patient outcomes, and deduce their biologic role in response to ICI therapy based on their modulation during treatment; and last but not least (8) melanoma-specific oncogenic programs supporting metabolic plasticity and fitness, together with clinical and preclinical evidence of differential activity of ICI therapy depending on the tumor metabolic state, should provide new research opportunities to evaluate these relationships as potential biomarkers for patient stratification and treatment allocation, and formulate novel precision-medicine combinations depending on metabolic and immune therapies.

References

  1. Sharma, P.; Wagner, K.; Wolchok, J.D.; Allison, J.P. Novel cancer immunotherapy agents with survival benefit: Recent successes and next steps. Nat. Rev. Cancer 2011, 11, 805–812.
  2. Sharma, P.; Allison, J.P. Immune checkpoint targeting in cancer therapy: Toward combination strategies with curative potential. Cell 2015, 161, 205–214.
  3. Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264.
  4. Darvin, P.; Toor, S.M.; Sasidharan Nair, V.; Elkord, E. Immune checkpoint inhibitors: Recent progress and potential biomarkers. Exp. Mol. Med. 2018, 50, 1–11.
  5. Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2018. CA Cancer J. Clin. 2018, 68, 7–30.
  6. Ayers, M.; Lunceford, J.; Nebozhyn, M.; Murphy, E.; Loboda, A.; Albright, A.; Cheng, J.; Kang, S.P.; Ebbinghaus, S.; Yearley, J.; et al. Relationship between immune gene signatures and clinical response to PD-1 blockade with pembrolizumab (MK-3475) in patients with advanced solid tumors. J. ImmunoTherapy Cancer 2015, 3, P80.
  7. Van Allen, E.M.; Miao, D.; Schilling, B.; Shukla, S.A.; Blank, C.; Zimmer, L.; Sucker, A.; Hillen, U.; Foppen, M.H.G.; Goldinger, S.M.; et al. Genomic correlates of response to CTLA-4 blockade in metastatic melanoma. Science 2015, 350, 207–211.
  8. Leach, D.R.; Krummel, M.F.; Allison, J.P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 1996, 271, 1734–1736.
  9. Hodi, F.S.; O’Day, S.J.; McDermott, D.F.; Weber, R.W.; Sosman, J.A.; Haanen, J.B.; Gonzalez, R.; Robert, C.; Schadendorf, D.; Hassel, J.C.; et al. Improved survival with ipilimumab in patients with metastatic melanoma. N. Engl. J. Med. 2010, 363, 711–723.
  10. Robert, C.; Thomas, L.; Bondarenko, I.; O’Day, S.; Weber, J.; Garbe, C.; Lebbe, C.; Baurain, J.F.; Testori, A.; Grob, J.J.; et al. Ipilimumab plus dacarbazine for previously untreated metastatic melanoma. N. Engl. J. Med. 2011, 364, 2517–2526.
  11. Pauken, K.E.; Torchia, J.A.; Chaudhri, A.; Sharpe, A.H.; Freeman, G.J. Emerging concepts in PD-1 checkpoint biology. Semin. Immunol. 2021, 52, 101480.
  12. Schumacher, T.N.; Schreiber, R.D. Neoantigens in cancer immunotherapy. Science 2015, 348, 69–74.
  13. Zappasodi, R.; Wolchok, J.D.; Merghoub, T. Correction to: Strategies for Predicting Response to Checkpoint Inhibitors. Curr. Hematol. Malig. Rep. 2019, 14, 62.
  14. Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Five-Year Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2019, 381, 1535–1546.
  15. Paik, J. Nivolumab Plus Relatlimab: First Approval. Drugs 2022, 82, 925–931.
  16. Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723.
  17. Prieto, P.A.; Yang, J.C.; Sherry, R.M.; Hughes, M.S.; Kammula, U.S.; White, D.E.; Levy, C.L.; Rosenberg, S.A.; Phan, G.Q. CTLA-4 blockade with ipilimumab: Long-term follow-up of 177 patients with metastatic melanoma. Clin. Cancer Res. 2012, 18, 2039–2047.
  18. Di Giacomo, A.M.; Calabro, L.; Danielli, R.; Fonsatti, E.; Bertocci, E.; Pesce, I.; Fazio, C.; Cutaia, O.; Giannarelli, D.; Miracco, C.; et al. Long-term survival and immunological parameters in metastatic melanoma patients who responded to ipilimumab 10 mg/kg within an expanded access programme. Cancer Immunol. Immunother. 2013, 62, 1021–1028.
  19. Gibney, G.T.; Weiner, L.M.; Atkins, M.B. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016, 17, e542–e551.
  20. Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Rutkowski, P.; Lao, C.D.; Cowey, C.L.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Long-Term Outcomes With Nivolumab Plus Ipilimumab or Nivolumab Alone Versus Ipilimumab in Patients With Advanced Melanoma. J. Clin. Oncol. 2022, 40, 127–137.
  21. Franzke, A.; Probst-Kepper, M.; Buer, J.; Duensing, S.; Hoffmann, R.; Wittke, F.; Volkenandt, M.; Ganser, A.; Atzpodien, J. Elevated pretreatment serum levels of soluble vascular cell adhesion molecule 1 and lactate dehydrogenase as predictors of survival in cutaneous metastatic malignant melanoma. Br. J. Cancer 1998, 78, 40–45.
  22. Long, G.V.; Eroglu, Z.; Infante, J.; Patel, S.; Daud, A.; Johnson, D.B.; Gonzalez, R.; Kefford, R.; Hamid, O.; Schuchter, L.; et al. Long-Term Outcomes in Patients With BRAF V600-Mutant Metastatic Melanoma Who Received Dabrafenib Combined With Trametinib. J. Clin. Oncol. 2018, 36, 667–673.
  23. Long, G.V.; Grob, J.J.; Nathan, P.; Ribas, A.; Robert, C.; Schadendorf, D.; Lane, S.R.; Mak, C.; Legenne, P.; Flaherty, K.T.; et al. Factors predictive of response, disease progression, and overall survival after dabrafenib and trametinib combination treatment: A pooled analysis of individual patient data from randomised trials. Lancet Oncol. 2016, 17, 1743–1754.
  24. Larkin, J.M.G.; McArthur, G.A.; Ribas, A.; Ascierto, P.A.; Gallo, J.D.; Rooney, I.A.; Chang, I.; Dréno, B. Clinical predictors of response for coBRIM: A phase III study of cobimetinib (C) in combination with vemurafenib (V) in advanced BRAF-mutated melanoma (MM). J. Clin. Oncol. 2016, 34, 9528.
  25. Long, G.V.; Blank, C.; Ribas, A.; Mortier, L.; Carlino, M.S.; Lotem, M.; Lorigan, P.; Neyns, B.; Petrella, T.M.; Puzanov, I.; et al. 1141 Impact of baseline serum lactate dehydrogenase concentration on the efficacy of pembrolizumab and ipilimumab in patients with advanced melanoma: Data from KEYNOTE-006. Eur. J. Cancer 2017, 72, s122–s123.
  26. Roh, W.; Chen, P.L.; Reuben, A.; Spencer, C.N.; Prieto, P.A.; Miller, J.P.; Gopalakrishnan, V.; Wang, F.; Cooper, Z.A.; Reddy, S.M.; et al. Integrated molecular analysis of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals markers of response and resistance. Sci. Transl. Med. 2017, 9, eaah3560.
  27. Zager, J.S.; Gastman, B.R.; Leachman, S.; Gonzalez, R.C.; Fleming, M.D.; Ferris, L.K.; Ho, J.; Miller, A.R.; Cook, R.W.; Covington, K.R.; et al. Performance of a prognostic 31-gene expression profile in an independent cohort of 523 cutaneous melanoma patients. BMC Cancer 2018, 18, 130.
  28. Hauschild, A.; Michaelsen, J.; Brenner, W.; Glaser, R.; Henze, E.; Christophers, E. Prognostic significance of serum S100B detection compared with routine blood parameters in advanced metastatic melanoma patients. Melanoma Res. 1999, 9, 155–161.
  29. Hauschild, A.; Engel, G.; Brenner, W.; Glaser, R.; Monig, H.; Henze, E.; Christophers, E. S100B protein detection in serum is a significant prognostic factor in metastatic melanoma. Oncology 1999, 56, 338–344.
  30. Tarhini, A.A.; Stuckert, J.; Lee, S.; Sander, C.; Kirkwood, J.M. Prognostic significance of serum S100B protein in high-risk surgically resected melanoma patients participating in Intergroup Trial ECOG 1694. J. Clin. Oncol. 2009, 27, 38–44.
  31. Gerami, P.; Cook, R.W.; Russell, M.C.; Wilkinson, J.; Amaria, R.N.; Gonzalez, R.; Lyle, S.; Jackson, G.L.; Greisinger, A.J.; Johnson, C.E.; et al. Gene expression profiling for molecular staging of cutaneous melanoma in patients undergoing sentinel lymph node biopsy. J. Am. Acad. Dermatol. 2015, 72, 780–785.e3.
  32. Gerami, P.; Cook, R.W.; Wilkinson, J.; Russell, M.C.; Dhillon, N.; Amaria, R.N.; Gonzalez, R.; Lyle, S.; Johnson, C.E.; Oelschlager, K.M.; et al. Development of a prognostic genetic signature to predict the metastatic risk associated with cutaneous melanoma. Clin. Cancer Res. 2015, 21, 175–183.
  33. Chapman, P.B.; Hauschild, A.; Robert, C.; Haanen, J.B.; Ascierto, P.; Larkin, J.; Dummer, R.; Garbe, C.; Testori, A.; Maio, M.; et al. Improved survival with vemurafenib in melanoma with BRAF V600E mutation. N. Engl. J. Med. 2011, 364, 2507–2516.
  34. Hauschild, A.; Grob, J.J.; Demidov, L.V.; Jouary, T.; Gutzmer, R.; Millward, M.; Rutkowski, P.; Blank, C.U.; Miller, W.H., Jr.; Kaempgen, E.; et al. Dabrafenib in BRAF-mutated metastatic melanoma: A multicentre, open-label, phase 3 randomised controlled trial. Lancet 2012, 380, 358–365.
  35. Long, G.V.; Stroyakovskiy, D.; Gogas, H.; Levchenko, E.; de Braud, F.; Larkin, J.; Garbe, C.; Jouary, T.; Hauschild, A.; Grob, J.J.; et al. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: A multicentre, double-blind, phase 3 randomised controlled trial. Lancet 2015, 386, 444–451.
  36. Ascierto, P.A.; McArthur, G.A.; Dréno, B.; Atkinson, V.; Liszkay, G.; Di Giacomo, A.M.; Mandalà, M.; Demidov, L.; Stroyakovskiy, D.; Thomas, L.; et al. Cobimetinib combined with vemurafenib in advanced BRAF(V600)-mutant melanoma (coBRIM): Updated efficacy results from a randomised, double-blind, phase 3 trial. Lancet Oncol. 2016, 17, 1248–1260.
  37. Dummer, R.; Ascierto, P.A.; Gogas, H.J.; Arance, A.; Mandala, M.; Liszkay, G.; Garbe, C.; Schadendorf, D.; Krajsova, I.; Gutzmer, R.; et al. Encorafenib plus binimetinib versus vemurafenib or encorafenib in patients with BRAF-mutant melanoma (COLUMBUS): A multicentre, open-label, randomised phase 3 trial. Lancet Oncol. 2018, 19, 603–615.
  38. Robert, C.; Karaszewska, B.; Schachter, J.; Rutkowski, P.; Mackiewicz, A.; Stroiakovski, D.; Lichinitser, M.; Dummer, R.; Grange, F.; Mortier, L.; et al. Improved overall survival in melanoma with combined dabrafenib and trametinib. N. Engl. J. Med. 2015, 372, 30–39.
  39. Genomic Classification of Cutaneous Melanoma. Cell 2015, 161, 1681–1696.
  40. Dummer, R.; Schadendorf, D.; Ascierto, P.A.; Arance, A.; Dutriaux, C.; Di Giacomo, A.M.; Rutkowski, P.; Del Vecchio, M.; Gutzmer, R.; Mandala, M.; et al. Binimetinib versus dacarbazine in patients with advanced NRAS-mutant melanoma (NEMO): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2017, 18, 435–445.
  41. Lebbé, C.; Dutriaux, C.; Lesimple, T.; Kruit, W.; Kerger, J.; Thomas, L.; Guillot, B.; Braud, F.; Garbe, C.; Grob, J.J.; et al. Pimasertib Versus Dacarbazine in Patients With Unresectable NRAS-Mutated Cutaneous Melanoma: Phase II, Randomized, Controlled Trial with Crossover. Cancers 2020, 12, 1727.
  42. Maertens, O.; Johnson, B.; Hollstein, P.; Frederick, D.T.; Cooper, Z.A.; Messiaen, L.; Bronson, R.T.; McMahon, M.; Granter, S.; Flaherty, K.; et al. Elucidating distinct roles for NF1 in melanomagenesis. Cancer Discov. 2013, 3, 338–349.
  43. Guo, J.; Si, L.; Kong, Y.; Flaherty, K.T.; Xu, X.; Zhu, Y.; Corless, C.L.; Li, L.; Li, H.; Sheng, X.; 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. 2011, 29, 2904–2909.
  44. Snyder, A.; Makarov, V.; Merghoub, T.; Yuan, J.; Zaretsky, J.M.; Desrichard, A.; Walsh, L.A.; Postow, M.A.; Wong, P.; Ho, T.S.; et al. Genetic basis for clinical response to CTLA-4 blockade in melanoma. N. Engl. J. Med. 2014, 371, 2189–2199.
  45. Rizvi, N.A.; Hellmann, M.D.; Snyder, A.; Kvistborg, P.; Makarov, V.; Havel, J.J.; Lee, W.; Yuan, J.; Wong, P.; Ho, T.S.; et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015, 348, 124–128.
  46. Cristescu, R.; Mogg, R.; Ayers, M.; Albright, A.; Murphy, E.; Yearley, J.; Sher, X.; Liu, X.Q.; Lu, H.; Nebozhyn, M.; et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 2018, 362, eaar3593.
  47. Topalian, S.L.; Taube, J.M.; Anders, R.A.; Pardoll, D.M. Mechanism-driven biomarkers to guide immune checkpoint blockade in cancer therapy. Nat. Rev. Cancer 2016, 16, 275–287.
  48. McGranahan, N.; Furness, A.J.; Rosenthal, R.; Ramskov, S.; Lyngaa, R.; Saini, S.K.; Jamal-Hanjani, M.; Wilson, G.A.; Birkbak, N.J.; Hiley, C.T.; et al. Clonal neoantigens elicit T cell immunoreactivity and sensitivity to immune checkpoint blockade. Science 2016, 351, 1463–1469.
  49. Chowell, D.; Morris, L.G.T.; Grigg, C.M.; Weber, J.K.; Samstein, R.M.; Makarov, V.; Kuo, F.; Kendall, S.M.; Requena, D.; Riaz, N.; et al. Patient HLA class I genotype influences cancer response to checkpoint blockade immunotherapy. Science 2018, 359, 582–587.
  50. Paulson, K.G.; Voillet, V.; McAfee, M.S.; Hunter, D.S.; Wagener, F.D.; Perdicchio, M.; Valente, W.J.; Koelle, S.J.; Church, C.D.; Vandeven, N.; et al. Acquired cancer resistance to combination immunotherapy from transcriptional loss of class I HLA. Nat. Commun. 2018, 9, 3868.
  51. Rooney, M.S.; Shukla, S.A.; Wu, C.J.; Getz, G.; Hacohen, N. Molecular and genetic properties of tumors associated with local immune cytolytic activity. Cell 2015, 160, 48–61.
  52. Davoli, T.; Uno, H.; Wooten, E.C.; Elledge, S.J. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science 2017, 355, eaaf8399.
  53. Tarhini, A.; Kudchadkar, R.R. Predictive and on-treatment monitoring biomarkers in advanced melanoma: Moving toward personalized medicine. Cancer Treat Rev. 2018, 71, 8–18.
  54. Weinstein, D.; Leininger, J.; Hamby, C.; Safai, B. Diagnostic and prognostic biomarkers in melanoma. J. Clin. Aesthet. Dermatol. 2014, 7, 13–24.
  55. Louie, A.D.; Huntington, K.; Carlsen, L.; Zhou, L.; El-Deiry, W.S. Integrating Molecular Biomarker Inputs Into Development and Use of Clinical Cancer Therapeutics. Front. Pharmacol. 2021, 12, 747194.
  56. Deacon, D.C.; Smith, E.A.; Judson-Torres, R.L. Molecular Biomarkers for Melanoma Screening, Diagnosis and Prognosis: Current State and Future Prospects. Front. Med. (Lausanne) 2021, 8, 642380.
  57. Carter, J.A.; Gilbo, P.; Atwal, G.S. IMPRES does not reproducibly predict response to immune checkpoint blockade therapy in metastatic melanoma. Nat. Med. 2019, 25, 1833–1835.
  58. Auslander, N.; Lee, J.S.; Ruppin, E. Reply to: ‘IMPRES does not reproducibly predict response to immune checkpoint blockade therapy in metastatic melanoma’. Nat. Med. 2019, 25, 1836–1838.
  59. Auslander, N.; Zhang, G.; Lee, J.S.; Frederick, D.T.; Miao, B.; Moll, T.; Tian, T.; Wei, Z.; Madan, S.; Sullivan, R.J.; et al. Robust prediction of response to immune checkpoint blockade therapy in metastatic melanoma. Nat. Med. 2018, 24, 1545–1549.
  60. Xiao, X.; Xu, C.; Yang, W.; Yu, R. Inconsistent prediction capability of ImmuneCells.Sig across different RNA-seq datasets. Nat. Commun. 2021, 12, 4167.
  61. Xiong, D.; Wang, Y.; You, M. Reply to: "Inconsistent prediction capability of ImmuneCells.Sig across different RNA-seq datasets". Nat. Commun. 2021, 12, 4168.
  62. Xiong, D.; Wang, Y.; You, M. A gene expression signature of TREM2(hi) macrophages and gammadelta T cells predicts immunotherapy response. Nat. Commun. 2020, 11, 5084.
  63. Hugo, W.; Zaretsky, J.M.; Sun, L.; Song, C.; Moreno, B.H.; Hu-Lieskovan, S.; Berent-Maoz, B.; Pang, J.; Chmielowski, B.; Cherry, G.; et al. Genomic and Transcriptomic Features of Response to Anti-PD-1 Therapy in Metastatic Melanoma. Cell 2016, 165, 35–44.
  64. Chen, P.L.; Roh, W.; Reuben, A.; Cooper, Z.A.; Spencer, C.N.; Prieto, P.A.; Miller, J.P.; Bassett, R.L.; Gopalakrishnan, V.; Wani, K.; et al. Analysis of Immune Signatures in Longitudinal Tumor Samples Yields Insight into Biomarkers of Response and Mechanisms of Resistance to Immune Checkpoint Blockade. Cancer Discov. 2016, 6, 827–837.
  65. Huang, A.C.; Orlowski, R.J.; Xu, X.; Mick, R.; George, S.M.; Yan, P.K.; Manne, S.; Kraya, A.A.; Wubbenhorst, B.; Dorfman, L.; et al. A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes in resectable melanoma. Nat. Med 2019, 25, 454–461.
  66. Jiang, P.; Gu, S.; Pan, D.; Fu, J.; Sahu, A.; Hu, X.; Li, Z.; Traugh, N.; Bu, X.; Li, B.; et al. Signatures of T cell dysfunction and exclusion predict cancer immunotherapy response. Nat. Med. 2018, 24, 1550–1558.
  67. Ayers, M.; Lunceford, J.; Nebozhyn, M.; Murphy, E.; Loboda, A.; Kaufman, D.R.; Albright, A.; Cheng, J.D.; Kang, S.P.; Shankaran, V.; et al. IFN-gamma-related mRNA profile predicts clinical response to PD-1 blockade. J. Clin. Invest. 2017, 127, 2930–2940.
  68. Ock, C.Y.; Hwang, J.E.; Keam, B.; Kim, S.B.; Shim, J.J.; Jang, H.J.; Park, S.; Sohn, B.H.; Cha, M.; Ajani, J.A.; et al. Genomic landscape associated with potential response to anti-CTLA-4 treatment in cancers. Nat. Commun. 2017, 8, 1050.
  69. Liu, D.; Schilling, B.; Liu, D.; Sucker, A.; Livingstone, E.; Jerby-Arnon, L.; Zimmer, L.; Gutzmer, R.; Satzger, I.; Loquai, C.; et al. Integrative molecular and clinical modeling of clinical outcomes to PD1 blockade in patients with metastatic melanoma. Nat. Med. 2019, 25, 1916–1927.
  70. Jerby-Arnon, L.; Shah, P.; Cuoco, M.S.; Rodman, C.; Su, M.J.; Melms, J.C.; Leeson, R.; Kanodia, A.; Mei, S.; Lin, J.R.; et al. A Cancer Cell Program Promotes T Cell Exclusion and Resistance to Checkpoint Blockade. Cell 2018, 175, 984–997.e24.
  71. Johnson, D.B.; Estrada, M.V.; Salgado, R.; Sanchez, V.; Doxie, D.B.; Opalenik, S.R.; Vilgelm, A.E.; Feld, E.; Johnson, A.S.; Greenplate, A.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. 2016, 7, 10582.
  72. Rodig, S.J.; Gusenleitner, D.; Jackson, D.G.; Gjini, E.; Giobbie-Hurder, A.; Jin, C.; Chang, H.; Lovitch, S.B.; Horak, C.; Weber, J.S.; et al. MHC proteins confer differential sensitivity to CTLA-4 and PD-1 blockade in untreated metastatic melanoma. Sci. Transl. Med. 2018, 10, eaar3342.
  73. Ben-Hamo, R.; Jacob Berger, A.; Gavert, N.; Miller, M.; Pines, G.; Oren, R.; Pikarsky, E.; Benes, C.H.; Neuman, T.; Zwang, Y.; et al. Predicting and affecting response to cancer therapy based on pathway-level biomarkers. Nat. Commun. 2020, 11, 3296.
  74. Bild, A.H.; Yao, G.; Chang, J.T.; Wang, Q.; Potti, A.; Chasse, D.; Joshi, M.B.; Harpole, D.; Lancaster, J.M.; Berchuck, A.; et al. Oncogenic pathway signatures in human cancers as a guide to targeted therapies. Nature 2006, 439, 353–357.
  75. Tian, S.; Simon, I.; Moreno, V.; Roepman, P.; Tabernero, J.; Snel, M.; van’t Veer, L.; Salazar, R.; Bernards, R.; Capella, G. A combined oncogenic pathway signature of BRAF, KRAS and PI3KCA mutation improves colorectal cancer classification and cetuximab treatment prediction. Gut 2013, 62, 540–549.
  76. Haider, S.; Yao, C.Q.; Sabine, V.S.; Grzadkowski, M.; Stimper, V.; Starmans, M.H.W.; Wang, J.; Nguyen, F.; Moon, N.C.; Lin, X.; et al. Pathway-based subnetworks enable cross-disease biomarker discovery. Nat. Commun. 2018, 9, 4746.
  77. Grasso, C.S.; Tsoi, J.; Onyshchenko, M.; Abril-Rodriguez, G.; Ross-Macdonald, P.; Wind-Rotolo, M.; Champhekar, A.; Medina, E.; Torrejon, D.Y.; Shin, D.S.; et al. Conserved Interferon-gamma Signaling Drives Clinical Response to Immune Checkpoint Blockade Therapy in Melanoma. Cancer Cell 2020, 38, 500–515.e3.
  78. Riaz, N.; Havel, J.J.; Makarov, V.; Desrichard, A.; Urba, W.J.; Sims, J.S.; Hodi, F.S.; Martin-Algarra, S.; Mandal, R.; Sharfman, W.H.; et al. Tumor and Microenvironment Evolution during Immunotherapy with Nivolumab. Cell 2017, 171, 934–949.e16.
  79. Helmink, B.A.; Reddy, S.M.; Gao, J.; Zhang, S.; Basar, R.; Thakur, R.; Yizhak, K.; Sade-Feldman, M.; Blando, J.; Han, G.; et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature 2020, 577, 549–555.
  80. Cabrita, R.; Lauss, M.; Sanna, A.; Donia, M.; Skaarup Larsen, M.; Mitra, S.; Johansson, I.; Phung, B.; Harbst, K.; Vallon-Christersson, J.; et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature 2020, 577, 561–565.
  81. Fehrenbacher, L.; Spira, A.; Ballinger, M.; Kowanetz, M.; Vansteenkiste, J.; Mazieres, J.; Park, K.; Smith, D.; Artal-Cortes, A.; Lewanski, C.; et al. Atezolizumab versus docetaxel for patients with previously treated non-small-cell lung cancer (POPLAR): A multicentre, open-label, phase 2 randomised controlled trial. Lancet 2016, 387, 1837–1846.
  82. Gide, T.N.; Quek, C.; Menzies, A.M.; Tasker, A.T.; Shang, P.; Holst, J.; Madore, J.; Lim, S.Y.; Velickovic, R.; Wongchenko, M.; et al. Distinct Immune Cell Populations Define Response to Anti-PD-1 Monotherapy and Anti-PD-1/Anti-CTLA-4 Combined Therapy. Cancer Cell 2019, 35, 238–255.e6.
  83. Petitprez, F.; de Reyniès, A.; Keung, E.Z.; Chen, T.W.; Sun, C.M.; Calderaro, J.; Jeng, Y.M.; Hsiao, L.P.; Lacroix, L.; Bougoüin, A.; et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature 2020, 577, 556–560.
  84. Sade-Feldman, M.; Yizhak, K.; Bjorgaard, S.L.; Ray, J.P.; de Boer, C.G.; Jenkins, R.W.; Lieb, D.J.; Chen, J.H.; Frederick, D.T.; Barzily-Rokni, M.; et al. Defining T Cell States Associated with Response to Checkpoint Immunotherapy in Melanoma. Cell 2018, 175, 998–1013.e20.
  85. Du, K.; Wei, S.; Wei, Z.; Frederick, D.T.; Miao, B.; Moll, T.; Tian, T.; Sugarman, E.; Gabrilovich, D.I.; Sullivan, R.J.; et al. Pathway signatures derived from on-treatment tumor specimens predict response to anti-PD1 blockade in metastatic melanoma. Nat. Commun. 2021, 12, 6023.
  86. Bownes, R.J.; Turnbull, A.K.; Martinez-Perez, C.; Cameron, D.A.; Sims, A.H.; Oikonomidou, O. On-treatment biomarkers can improve prediction of response to neoadjuvant chemotherapy in breast cancer. Breast Cancer Res. 2019, 21, 73.
  87. Turnbull, A.K.; Arthur, L.M.; Renshaw, L.; Larionov, A.A.; Kay, C.; Dunbier, A.K.; Thomas, J.S.; Dowsett, M.; Sims, A.H.; Dixon, J.M. Accurate Prediction and Validation of Response to Endocrine Therapy in Breast Cancer. J. Clin. Oncol. 2015, 33, 2270–2278.
  88. Ellis, M.J.; Suman, V.J.; Hoog, J.; Goncalves, R.; Sanati, S.; Creighton, C.J.; DeSchryver, K.; Crouch, E.; Brink, A.; Watson, M.; et al. Ki67 Proliferation Index as a Tool for Chemotherapy Decisions During and After Neoadjuvant Aromatase Inhibitor Treatment of Breast Cancer: Results From the American College of Surgeons Oncology Group Z1031 Trial (Alliance). J. Clin. Oncol. 2017, 35, 1061–1069.
  89. Wallin, J.J.; Bendell, J.C.; Funke, R.; Sznol, M.; Korski, K.; Jones, S.; Hernandez, G.; Mier, J.; He, X.; Hodi, F.S.; et al. Atezolizumab in combination with bevacizumab enhances antigen-specific T-cell migration in metastatic renal cell carcinoma. Nat. Commun. 2016, 7, 12624.
  90. Harel, M.; Ortenberg, R.; Varanasi, S.K.; Mangalhara, K.C.; Mardamshina, M.; Markovits, E.; Baruch, E.N.; Tripple, V.; Arama-Chayoth, M.; Greenberg, E.; et al. Proteomics of Melanoma Response to Immunotherapy Reveals Mitochondrial Dependence. Cell 2019, 179, 236–250.e18.
  91. Lee, J.H.; Shklovskaya, E.; Lim, S.Y.; Carlino, M.S.; Menzies, A.M.; Stewart, A.; Pedersen, B.; Irvine, M.; Alavi, S.; Yang, J.Y.H.; et al. Transcriptional downregulation of MHC class I and melanoma de-differentiation in resistance to PD-1 inhibition. Nat. Commun. 2020, 11, 1897.
  92. Huang, A.C.; Zappasodi, R. A decade of checkpoint blockade immunotherapy in melanoma: Understanding the molecular basis for immune sensitivity and resistance. Nat. Immunol. 2022, 23, 660–670.
  93. Vogelstein, B.; Papadopoulos, N.; Velculescu, V.E.; Zhou, S.; Diaz, L.A., Jr.; Kinzler, K.W. Cancer genome landscapes. Science 2013, 339, 1546–1558.
  94. Leko, V.; Rosenberg, S.A. Identifying and Targeting Human Tumor Antigens for T Cell-Based Immunotherapy of Solid Tumors. Cancer Cell 2020, 38, 454–472.
  95. Turajlic, S.; Litchfield, K.; Xu, H.; Rosenthal, R.; McGranahan, N.; Reading, J.L.; Wong, Y.N.S.; Rowan, A.; Kanu, N.; Al Bakir, M.; et al. Insertion-and-deletion-derived tumour-specific neoantigens and the immunogenic phenotype: A pan-cancer analysis. Lancet Oncol. 2017, 18, 1009–1021.
  96. Coulie, P.G.; Van den Eynde, B.J.; van der Bruggen, P.; Boon, T. Tumour antigens recognized by T lymphocytes: At the core of cancer immunotherapy. Nat. Rev. Cancer 2014, 14, 135–146.
  97. Roszik, J.; Wang, W.L.; Livingston, J.A.; Roland, C.L.; Ravi, V.; Yee, C.; Hwu, P.; Futreal, A.; Lazar, A.J.; Patel, S.R.; et al. Overexpressed PRAME is a potential immunotherapy target in sarcoma subtypes. Clin. Sarcoma Res. 2017, 7, 11.
  98. Zhang, W.; Li, L.; Cai, L.; Liang, Y.; Xu, J.; Liu, Y.; Zhou, L.; Ding, C.; Zhang, Y.; Zhao, H.; et al. Tumor-associated antigen Prame targets tumor suppressor p14/ARF for degradation as the receptor protein of CRL2(Prame) complex. Cell Death Differ. 2021, 28, 1926–1940.
  99. Smyth, M.J.; Ngiow, S.F.; Ribas, A.; Teng, M.W. Combination cancer immunotherapies tailored to the tumour microenvironment. Nat. Rev. Clin. Oncol. 2016, 13, 143–158.
  100. Hogan, S.A.; Levesque, M.P.; Cheng, P.F. Melanoma Immunotherapy: Next-Generation Biomarkers. Front. Oncol. 2018, 8, 178.
  101. Keenan, T.E.; Burke, K.P.; Van Allen, E.M. Genomic correlates of response to immune checkpoint blockade. Nat. Med. 2019, 25, 389–402.
  102. Trujillo, J.A.; Luke, J.J.; Zha, Y.; Segal, J.P.; Ritterhouse, L.L.; Spranger, S.; Matijevich, K.; Gajewski, T.F. Secondary resistance to immunotherapy associated with beta-catenin pathway activation or PTEN loss in metastatic melanoma. J. Immunother. Cancer 2019, 7, 295.
  103. Curran, M.A.; Montalvo, W.; Yagita, H.; Allison, J.P. PD-1 and CTLA-4 combination blockade expands infiltrating T cells and reduces regulatory T and myeloid cells within B16 melanoma tumors. Proc. Natl. Acad. Sci. USA 2010, 107, 4275–4280.
  104. Sharma, P.; Allison, J.P. The future of immune checkpoint therapy. Science 2015, 348, 56–61.
  105. Wei, S.C.; Levine, J.H.; Cogdill, A.P.; Zhao, Y.; Anang, N.A.S.; Andrews, M.C.; Sharma, P.; Wang, J.; Wargo, J.A.; Pe’er, D.; et al. Distinct Cellular Mechanisms Underlie Anti-CTLA-4 and Anti-PD-1 Checkpoint Blockade. Cell 2017, 170, 1120–1133.e17.
  106. Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355.
  107. Herbst, R.S.; Soria, J.C.; Kowanetz, M.; Fine, G.D.; Hamid, O.; Gordon, M.S.; Sosman, J.A.; McDermott, D.F.; Powderly, J.D.; Gettinger, S.N.; et al. Predictive correlates of response to the anti-PD-L1 antibody MPDL3280A in cancer patients. Nature 2014, 515, 563–567.
  108. Taube, J.M.; Klein, A.; Brahmer, J.R.; Xu, H.; Pan, X.; Kim, J.H.; Chen, L.; Pardoll, D.M.; Topalian, S.L.; Anders, R.A. 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. 2014, 20, 5064–5074.
  109. Tumeh, P.C.; Harview, C.L.; Yearley, J.H.; Shintaku, I.P.; Taylor, E.J.; Robert, L.; Chmielowski, B.; Spasic, M.; Henry, G.; Ciobanu, V.; et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014, 515, 568–571.
  110. Sharma, A.; Subudhi, S.K.; Blando, J.; Vence, L.; Wargo, J.; Allison, J.P.; Ribas, A.; Sharma, P. Anti-CTLA-4 Immunotherapy Does Not Deplete FOXP3(+) Regulatory T Cells (Tregs) in Human Cancers-Response. Clin. Cancer Res. 2019, 25, 3469–3470.
  111. Shen, H.; Huang, F.; Zhang, X.; Ojo, O.A.; Li, Y.; Trummell, H.Q.; Anderson, J.C.; Fiveash, J.; Bredel, M.; Yang, E.S.; et al. Selective suppression of melanoma lacking IFN-gamma pathway by JAK inhibition depends on T cells and host TNF signaling. Nat. Commun. 2022, 13, 5013.
  112. Liu, Y.T.; Sun, Z.J. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics 2021, 11, 5365–5386.
  113. Kalbasi, A.; Ribas, A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat. Rev. Immunol. 2020, 20, 25–39.
  114. Wei, S.C.; Anang, N.A.S.; Sharma, R.; Andrews, M.C.; Reuben, A.; Levine, J.H.; Cogdill, A.P.; Mancuso, J.J.; Wargo, J.A.; Pe’er, D.; et al. Combination anti-CTLA-4 plus anti-PD-1 checkpoint blockade utilizes cellular mechanisms partially distinct from monotherapies. Proc. Natl. Acad. Sci. USA 2019, 116, 22699–22709.
  115. Ribas, A.; Medina, T.; Kummar, S.; Amin, A.; Kalbasi, A.; Drabick, J.J.; Barve, M.; Daniels, G.A.; Wong, D.J.; Schmidt, E.V.; et al. SD-101 in Combination with Pembrolizumab in Advanced Melanoma: Results of a Phase Ib, Multicenter Study. Cancer Discov. 2018, 8, 1250–1257.
  116. Ribas, A.; Dummer, R.; Puzanov, I.; VanderWalde, A.; Andtbacka, R.H.I.; Michielin, O.; Olszanski, A.J.; Malvehy, J.; Cebon, J.; Fernandez, E.; et al. Oncolytic Virotherapy Promotes Intratumoral T Cell Infiltration and Improves Anti-PD-1 Immunotherapy. Cell 2018, 174, 1031–1032.
  117. Vanpouille-Box, C.; Hoffmann, J.A.; Galluzzi, L. Pharmacological modulation of nucleic acid sensors - therapeutic potential and persisting obstacles. Nat. Rev. Drug Discov. 2019, 18, 845–867.
  118. Li, T.; Chen, Z.J. The cGAS-cGAMP-STING pathway connects DNA damage to inflammation, senescence, and cancer. J. Exp. Med. 2018, 215, 1287–1299.
  119. Abril-Rodriguez, G.; Torrejon, D.Y.; Liu, W.; Zaretsky, J.M.; Nowicki, T.S.; Tsoi, J.; Puig-Saus, C.; Baselga-Carretero, I.; Medina, E.; Quist, M.J.; et al. PAK4 inhibition improves PD-1 blockade immunotherapy. Nat. Cancer 2020, 1, 46–58.
  120. Grasso, C.S.; Giannakis, M.; Wells, D.K.; Hamada, T.; Mu, X.J.; Quist, M.; Nowak, J.A.; Nishihara, R.; Qian, Z.R.; Inamura, K.; et al. Genetic Mechanisms of Immune Evasion in Colorectal Cancer. Cancer Discov. 2018, 8, 730–749.
  121. Spranger, S.; Gajewski, T.F. Impact of oncogenic pathways on evasion of antitumour immune responses. Nat. Rev. Cancer 2018, 18, 139–147.
More
Information
Subjects: Biology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 380
Entry Collection: Biopharmaceuticals Technology
Revisions: 2 times (View History)
Update Date: 31 Jan 2023
1000/1000
Video Production Service