Abstract
The advent of immune checkpoint inhibition has revolutionized treatment of advanced melanoma. While most patients derive survival benefit from established immunotherapies, notably monoclonal antibodies blocking cytotoxic T-lymphocyte antigen 4 and programmed cell death protein 1, a subset does not optimally respond due to the manifestation of innate or acquired resistance to these therapies. Combination regimens have proven efficacious relative to single-agent blockade, but also yield high-grade treatment toxicities that are often dose-limiting for patients. In this review, we discuss the significant strides made in the past half-decade toward expanding the melanoma immunotherapy treatment paradigm. These include newly approved therapies, adoption of neoadjuvant immunotherapy, and studies in the clinical trials pipeline targeting alternative immune checkpoints and key immunoregulatory molecules. We then review how developments in molecular and functional diagnostics have furthered our understanding of the tumor-intrinsic and -extrinsic mechanisms driving immunotherapy resistance, as well as highlight novel biomarkers for predicting treatment response. Throughout, we discuss potential approaches for targeting these resistance mechanisms in rational combination with established immunotherapies to improve outcomes for patients with melanoma.
Similar content being viewed by others
References
Pires da Silva I, Ahmed T, Reijers ILM, Weppler AM, Betof Warner A, Patrinely JR, et al. Ipilimumab alone or ipilimumab plus anti-PD-1 therapy in patients with metastatic melanoma resistant to anti-PD-(L)1 monotherapy: a multicentre, retrospective, cohort study. Lancet Oncol. 2021;22:836–47.
Grob JJ, Amonkar MM, Karaszewska B, Schachter J, Dummer R, Mackiewicz A, et al. Comparison of dabrafenib and trametinib combination therapy with vemurafenib monotherapy on health-related quality of life in patients with unresectable or metastatic cutaneous BRAF Val600-mutation-positive melanoma (COMBI-v): results of a phase 3, open-label, randomised trial. Lancet Oncol. 2015;16:1389–98.
Tawbi HA, Schadendorf D, Lipson EJ, Ascierto PA, Matamala L, Castillo Gutiérrez E, et al. Relatlimab and nivolumab versus nivolumab in untreated advanced melanoma. N Engl J Med. 2022;386:24–34.
Weiss SA, Wolchok JD, Sznol M. Immunotherapy of melanoma: facts and hopes. Clin Cancer Res. 2019;25:5191–201.
Long GV, Stroyakovskiy D, Gogas H, Levchenko E, de Braud F, Larkin J, et al. Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. N Engl J Med. 2014;371:1877–88.
Liu D, Jenkins RW, Sullivan RJ. Mechanisms of resistance to immune checkpoint blockade. Am J Clin Dermatol. 2019;20:41–54.
Schadendorf D, Hodi FS, Robert C, Weber JS, Margolin K, Hamid O, et al. Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma. J Clin Oncol. 2015;33:1889–94.
Schachter J, Ribas A, Long GV, Arance A, Grob J-J, Mortier L, et al. Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicentre, randomised, open-label phase 3 study (KEYNOTE-006). Lancet. 2017;390:1853–62.
Ribas A, Hamid O, Daud A, Hodi FS, Wolchok JD, Kefford R, et al. Association of pembrolizumab with tumor response and survival among patients with advanced melanoma. JAMA. 2016;315:1600–9.
Wolchok JD, Chiarion-Sileni V, Gonzalez R, Rutkowski P, Grob J-J, Cowey CL, et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2017;377:1345–56.
Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, et al. Combined nivolumab and ipilimumab or monotherapy in untreated melanoma. N Engl J Med. 2015;373:23–34.
Wolchok JD, Chiarion-Sileni V, Rutkowski P, Cowey CL, Schadendorf D, Wagstaff J, et al. Final, 10-year outcomes with nivolumab plus ipilimumab in advanced melanoma. N Engl J Med. 2024. https://doi.org/10.1056/NEJMoa2407417.
Larkin J, Chiarion-Sileni V, Gonzalez R, Grob J-J, Rutkowski P, Lao CD, et al. Five-year survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med. 2019;381:1535–46.
Wolchok JD, Chiarion-Sileni V, Gonzalez R, Grob J-J, Rutkowski P, Lao CD, et al. Long-term outcomes with nivolumab plus ipilimumab or nivolumab alone versus ipilimumab in patients with advanced melanoma. J Clin Oncol. 2022;40:127–37.
Watson AS, Goutam S, Stukalin I, Ewanchuk BW, Sander M, Meyers DE, et al. Association of immune-related adverse events, hospitalization, and therapy resumption with survival among patients with metastatic melanoma receiving single-agent or combination immunotherapy. JAMA Netw Open. 2022;5: e2245596.
Maruhashi T, Sugiura D, Okazaki I-M, Okazaki T. LAG-3: from molecular functions to clinical applications. J Immunother Cancer. 2020;8: e001014. https://doi.org/10.1136/jitc-2020-001014.
Hemon P, Jean-Louis F, Ramgolam K, Brignone C, Viguier M, Bachelez H, et al. MHC class II engagement by its ligand LAG-3 (CD223) contributes to melanoma resistance to apoptosis. J Immunol. 2011;186:5173–83.
Woo S-R, Turnis ME, Goldberg MV, Bankoti J, Selby M, Nirschl CJ, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res. 2012;72:917–27.
Thudium K, Selby M, Zorn JA, Rak G, Wang X-T, Bunch RT, et al. Preclinical characterization of relatlimab, a human LAG-3-blocking antibody, alone or in combination with nivolumab. Cancer Immunol Res. 2022;10:1175–89.
Menzies AM, Pires da Silva I, Trojaniello C, Vieu E, Amaria RN, Zimmer L, et al. CTLA-4 blockade resistance after relatlimab and nivolumab. N Engl J Med. 2022;386:1668–9.
Dixon KO, Tabaka M, Schramm MA, Xiao S, Tang R, Dionne D, et al. TIM-3 restrains anti-tumour immunity by regulating inflammasome activation. Nature. 2021;595:101–6.
Tang W, Chen J, Ji T, Cong X. TIGIT, a novel immune checkpoint therapy for melanoma. Cell Death Dis. 2023;14:466.
Ascierto PA, Dummer R, Gaudy-Marqueste C, Bowyer S, Lipson EJ, Ghisoni E, et al. Efficacy and safety of triplet nivolumab, relatlimab, and ipilimumab (NIVO + RELA + IPI) in advanced melanoma: results from RELATIVITY-048. J Clin Oncol. 2024;42:9504–9504.
Betof Warner A, Corrie PG, Hamid O. Tumor-infiltrating lymphocyte therapy in melanoma: facts to the future. Clin Cancer Res. 2023;29:1835–54.
Dudley ME, Wunderlich JR, Robbins PF, Yang JC, Hwu P, Schwartzentruber DJ, et al. Cancer regression and autoimmunity in patients after clonal repopulation with antitumor lymphocytes. Science. 2002;298:850–4.
Dudley ME, Yang JC, Sherry R, Hughes MS, Royal R, Kammula U, et al. Adoptive cell therapy for patients with metastatic melanoma: evaluation of intensive myeloablative chemoradiation preparative regimens. J Clin Oncol. 2008;26:5233–9.
Rosenberg SA, Restifo NP, Yang JC, Morgan RA, Dudley ME. Adoptive cell transfer: a clinical path to effective cancer immunotherapy. Nat Rev Cancer. 2008;8:299–308.
Rohaan MW, Borch TH, van den Berg JH, Met Ö, Kessels R, Geukes Foppen MH, et al. Tumor-infiltrating lymphocyte therapy or ipilimumab in advanced melanoma. N Engl J Med. 2022;387:2113–25.
Sarnaik AA, Hamid O, Khushalani NI, Lewis KD, Medina T, Kluger HM, et al. Lifileucel, a tumor-infiltrating lymphocyte therapy, in metastatic melanoma. J Clin Oncol. 2021;39:2656–66.
Chesney J, Lewis KD, Kluger H, Hamid O, Whitman E, Thomas S, et al. Efficacy and safety of lifileucel, a one-time autologous tumor-infiltrating lymphocyte (TIL) cell therapy, in patients with advanced melanoma after progression on immune checkpoint inhibitors and targeted therapies: pooled analysis of consecutive cohorts of the C-144-01 study. J Immunother Cancer. 2022;10: e005755. https://doi.org/10.1136/jitc-2022-005755.
Parums DV. Editorial: first regulatory approval for adoptive cell therapy with autologous tumor-infiltrating lymphocytes (TILs)—lifileucel (Amtagvi). Med Sci Monit. 2024;30: e944927.
Grigoleit G-U, Kluger H, Thomas S, Chesney JA, Domingo-Musibay E, Sanmamed MF, et al. 1086MO Lifileucel tumor-infiltrating lymphocyte (TIL) cell therapy in patients (pts) with advanced mucosal melanoma after progression on immune checkpoint inhibitors (ICI): results from the phase II C-144-01 study. Ann Oncol. 2023;34:S654.
Chandran SS, Somerville RPT, Yang JC, Sherry RM, Klebanoff CA, Goff SL, et al. Treatment of metastatic uveal melanoma with adoptive transfer of tumour-infiltrating lymphocytes: a single-centre, two-stage, single-arm, phase 2 study. Lancet Oncol. 2017;18:792–802.
Middleton MR, McAlpine C, Woodcock VK, Corrie P, Infante JR, Steven NM, et al. Tebentafusp, a TCR/Anti-CD3 Bispecific fusion protein targeting gp100, potently activated antitumor immune responses in patients with metastatic melanoma. Clin Cancer Res. 2020;26:5869–78.
van der Kooij MK, Speetjens FM, van der Burg SH, Kapiteijn E. Uveal versus cutaneous melanoma; same origin, very distinct tumor types. Cancers. 2019;11:845. https://doi.org/10.3390/cancers11060845.
Nathan P, Hassel JC, Rutkowski P, Baurain J-F, Butler MO, Schlaak M, et al. Overall survival benefit with tebentafusp in metastatic uveal melanoma. N Engl J Med. 2021;385:1196–206.
Hassel JC, Piperno-Neumann S, Rutkowski P, Baurain J-F, Schlaak M, Butler MO, et al. Three-year overall survival with tebentafusp in metastatic uveal melanoma. N Engl J Med. 2023;389:2256–66.
Kassner J, Abdellatif B, Yamshon S, Monge J, Kaner J. Current landscape of CD3 bispecific antibodies in hematologic malignancies. Trends Cancer Res. 2024;10:708–32.
Middelburg J, Kemper K, Engelberts P, Labrijn AF, Schuurman J, van Hall T. Overcoming challenges for CD3-bispecific antibody therapy in solid tumors. Cancers. 2021;13(2):287. https://doi.org/10.3390/cancers13020287.
Middelburg J, Sluijter M, Schaap G, Göynük B, Lloyd K, Ovcinnikovs V, et al. T-cell stimulating vaccines empower CD3 bispecific antibody therapy in solid tumors. Nat Commun. 2024;15:48.
Long GV, Weber JS, Infante JR, Kim KB, Daud A, Gonzalez R, et al. Overall survival and durable responses in patients with braf v600-mutant metastatic melanoma receiving dabrafenib combined with trametinib. J Clin Oncol. 2016;34:871–8.
Ascierto PA, Mandalà M, Ferrucci PF, Guidoboni M, Rutkowski P, Ferraresi V, et al. Sequencing of ipilimumab plus nivolumab and encorafenib plus binimetinib for untreated BRAF-mutated metastatic melanoma (SECOMBIT): a randomized, three-arm, open-label phase II trial. J Clin Oncol. 2023. https://doi.org/10.1200/JCO.21.02961.
Ascierto PA, Casula M, Bulgarelli J, Pisano M, Piccinini C, Piccin L, et al. Sequential immunotherapy and targeted therapy for metastatic BRAF V600 mutated melanoma: 4-year survival and biomarkers evaluation from the phase II SECOMBIT trial. Nat Commun. 2024;15:146.
Atkins MB, Lee SJ, Chmielowski B, Tarhini AA, Cohen GI, Truong T-G, et al. Combination dabrafenib and trametinib versus combination nivolumab and ipilimumab for patients with advanced BRAF-mutant melanoma: the DREAMseq trial—ECOG-ACRIN EA6134. J Clin Oncol. 2023. https://doi.org/10.1200/JCO.22.01763.
Ascierto PA, Mandalà M, Ferrucci PF, Guidoboni M, Rutkowski P, Ferraresi V, et al. Sequencing of checkpoint or BRAF/MEK inhibitors on brain metastases in melanoma. NEJM Evid. 2024;3:EVIDoa2400087.
Luke JJ, Rutkowski P, Queirolo P, Del Vecchio M, Mackiewicz J, Chiarion-Sileni V, et al. Pembrolizumab versus placebo as adjuvant therapy in completely resected stage IIB or IIC melanoma (KEYNOTE-716): a randomised, double-blind, phase 3 trial. Lancet. 2022;399:1718–29.
Eggermont AMM, Chiarion-Sileni V, Grob J-J, Dummer R, Wolchok JD, Schmidt H, et al. Prolonged survival in stage III melanoma with ipilimumab adjuvant therapy. N Engl J Med. 2016;375:1845–55.
Amaria RN, Menzies AM, Burton EM, Scolyer RA, Tetzlaff MT, Antdbacka R, et al. Neoadjuvant systemic therapy in melanoma: recommendations of the International Neoadjuvant Melanoma Consortium. Lancet Oncol. 2019;20:e378–89.
Witt RG, Erstad DJ, Wargo JA. Neoadjuvant therapy for melanoma: rationale for neoadjuvant therapy and pivotal clinical trials. Ther Adv Med Oncol. 2022;14:17588359221083052.
Blank CU, Rozeman EA, Fanchi LF, Sikorska K, van de Wiel B, Kvistborg P, et al. Neoadjuvant versus adjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma. Nat Med. 2018;24:1655–61.
Versluis JM, Menzies AM, Sikorska K, Rozeman EA, Saw RPM, van Houdt WJ, et al. Survival update of neoadjuvant ipilimumab plus nivolumab in macroscopic stage III melanoma in the OpACIN and OpACIN-neo trials. Ann Oncol. 2023;34:420–30.
Lebbé C, Meyer N, Mortier L, Marquez-Rodas I, Robert C, Rutkowski P, et al. Evaluation of two dosing regimens for nivolumab in combination with ipilimumab in patients with advanced melanoma: results from the phase IIIb/IV CheckMate 511 trial. J Clin Oncol. 2019;37:867–75.
Menzies AM, Amaria RN, Rozeman EA, Huang AC, Tetzlaff MT, van de Wiel BA, et al. Pathological response and survival with neoadjuvant therapy in melanoma: a pooled analysis from the International Neoadjuvant Melanoma Consortium (INMC). Nat Med. 2021;27:301–9.
Rozeman EA, Hoefsmit EP, Reijers ILM, Saw RPM, Versluis JM, Krijgsman O, et al. Survival and biomarker analyses from the OpACIN-neo and OpACIN neoadjuvant immunotherapy trials in stage III melanoma. Nat Med. 2021;27:256–63.
Amaria RN, Postow M, Burton EM, Tetzlaff MT, Ross MI, Torres-Cabala C, et al. Neoadjuvant relatlimab and nivolumab in resectable melanoma. Nature. 2022;611:155–60.
Reijers ILM, Menzies AM, van Akkooi ACJ, Versluis JM, van den Heuvel NMJ, Saw RPM, et al. Personalized response-directed surgery and adjuvant therapy after neoadjuvant ipilimumab and nivolumab in high-risk stage III melanoma: the PRADO trial. Nat Med. 2022;28:1178–88.
Patel SP, Othus M, Chen Y, Wright GP Jr, Yost KJ, Hyngstrom JR, et al. Neoadjuvant-adjuvant or adjuvant-only pembrolizumab in advanced melanoma. N Engl J Med. 2023;388:813–23.
Patel S, Othus M, Wright P, Hyngstrom J, Lao CD, Truong T-G, et al. LBA48 Pathologic response and exploratory analyses of neoadjuvant-adjuvant versus adjuvant pembrolizumab (PEM) for resectable stage IIIb-IV melanoma from SWOG S1801. Ann Oncol. 2023;34:S1288.
Blank CU, Lucas MW, Scolyer RA, van de Wiel BA, Menzies AM, Lopez-Yurda M, et al. Neoadjuvant nivolumab and ipilimumab in resectable stage III melanoma. N Engl J Med. 2024. https://doi.org/10.1056/NEJMoa2402604.
Rauwerdink DJW, van Not O, de Meza M, van Doorn R, van der Hage J, van den Eertwegh AJM, et al. Adverse events in anti-PD-1-treated adjuvant and first-line advanced melanoma patients. Cancers. 2024;16(15):2656. https://doi.org/10.3390/cancers16152656.
Soldozy S, Mulligan KM, Zheng DX, Levoska MA, Cullison CR, Elarjani T, et al. Oncolytic virotherapy for melanoma brain metastases, a potential new treatment paradigm? Brain Sci. 2021;11:1260. https://doi.org/10.3390/brainsci11101260.
Ferrucci PF, Pala L, Conforti F, Cocorocchio E. Talimogene laherparepvec (T-VEC): an intralesional cancer immunotherapy for advanced melanoma. Cancers. 2021;13(6):1383. https://doi.org/10.3390/cancers13061383.
Chesney JA, Ribas A, Long GV, Kirkwood JM, Dummer R, Puzanov I, et al. Randomized, double-blind, placebo-controlled, global phase III trial of talimogene laherparepvec combined with pembrolizumab for advanced melanoma. J Clin Oncol. 2023;41:528–40.
Diab A, Gogas H, Sandhu S, Long GV, Ascierto PA, Larkin J, et al. Bempegaldesleukin plus nivolumab in untreated advanced melanoma: the open-label, phase III PIVOT IO 001 trial results. J Clin Oncol. 2023;41:4756–67.
Fujiwara Y, Kato S, Nesline MK, Conroy JM, DePietro P, Pabla S, et al. Indoleamine 2,3-dioxygenase (IDO) inhibitors and cancer immunotherapy. Cancer Treat Rev. 2022;110: 102461.
Long GV, Dummer R, Hamid O, Gajewski TF, Caglevic C, Dalle S, et al. Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomised, double-blind study. Lancet Oncol. 2019;20:1083–97.
Odunsi K, Qian F, Lugade AA, Yu H, Geller MA, Fling SP, et al. Metabolic adaptation of ovarian tumors in patients treated with an IDO1 inhibitor constrains antitumor immune responses. Sci Transl Med. 2022;14:eabg8402.
Ascierto PA, Ferrucci PF, Fisher R, Del Vecchio M, Atkinson V, Schmidt H, et al. Dabrafenib, trametinib and pembrolizumab or placebo in BRAF-mutant melanoma. Nat Med. 2019;25:941–6.
Dummer R, Long GV, Robert C, Tawbi HA, Flaherty KT, Ascierto PA, et al. Randomized phase III trial evaluating spartalizumab plus dabrafenib and trametinib for V600-mutant unresectable or metastatic melanoma. J Clin Oncol. 2022;40:1428–38.
Gutzmer R, Stroyakovskiy D, Gogas H, Robert C, Lewis K, Protsenko S, et al. Atezolizumab, vemurafenib, and cobimetinib as first-line treatment for unresectable advanced BRAF mutation-positive melanoma (IMspire150): primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2020;395:1835–44.
Long GV, Carlino MS, Au-Yeung G, Spillane AJ, Shannon KF, Gyorki DE, et al. Neoadjuvant pembrolizumab, dabrafenib and trametinib in BRAF-mutant resectable melanoma: the randomized phase 2 NeoTrio trial. Nat Med. 2024;30:2540–8.
Sade-Feldman M, Jiao YJ, Chen JH, Rooney MS, Barzily-Rokni M, Eliane J-P, et al. Resistance to checkpoint blockade therapy through inactivation of antigen presentation. Nat Commun. 2017;8:1136.
Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med. 2016;375:819–29.
Shin DS, Zaretsky JM, Escuin-Ordinas H, Garcia-Diaz A, Hu-Lieskovan S, Kalbasi A, et al. Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov. 2017;7:188–201.
Benci JL, Xu B, Qiu Y, Wu TJ, Dada H, Twyman-Saint Victor C, et al. Tumor interferon signaling regulates a multigenic resistance program to immune checkpoint blockade. Cell. 2016;167:1540-54.e12.
Lauss M, Phung B, Borch TH, Harbst K, Kaminska K, Ebbesson A, et al. Molecular patterns of resistance to immune checkpoint blockade in melanoma. Nat Commun. 2024;15:3075.
Liu D, Schilling B, Liu D, Sucker A, Livingstone E, Jerby-Arnon L, et al. Integrative molecular and clinical modeling of clinical outcomes to PD1 blockade in patients with metastatic melanoma. Nat Med. 2019;25:1916–27.
Tarantino G, Ricker CA, Wang A, Ge W, Aprati TJ, Huang AY, et al. Genomic heterogeneity and ploidy identify patients with intrinsic resistance to PD-1 blockade in metastatic melanoma. bioRxiv. 2022. https://doi.org/10.1101/2022.12.11.519808.
Wolf Y, Bartok O, Patkar S, Eli GB, Cohen S, Litchfield K, et al. UVB-induced tumor heterogeneity diminishes immune response in melanoma. Cell. 2019;179:219-235.e21.
Tirosh I, Izar B, Prakadan SM, Wadsworth MH 2nd, Treacy D, Trombetta JJ, et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science. 2016;352:189–96.
Landsberg J, Kohlmeyer J, Renn M, Bald T, Rogava M, Cron M, et al. Melanomas resist T-cell therapy through inflammation-induced reversible dedifferentiation. Nature. 2012;490:412–6.
Boiko AD, Razorenova OV, van de Rijn M, Swetter SM, Johnson DL, Ly DP, et al. Human melanoma-initiating cells express neural crest nerve growth factor receptor CD271. Nature. 2010;466:133–7.
Boshuizen J, Vredevoogd DW, Krijgsman O, Ligtenberg MA, Blankenstein S, de Bruijn B, et al. Reversal of pre-existing NGFR-driven tumor and immune therapy resistance. Nat Commun. 2020;11:3946.
Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic β-catenin signalling prevents anti-tumour immunity. Nature. 2015;523:231–5.
Riesenberg S, Groetchen A, Siddaway R, Bald T, Reinhardt J, Smorra D, et al. MITF and c-Jun antagonism interconnects melanoma dedifferentiation with pro-inflammatory cytokine responsiveness and myeloid cell recruitment. Nat Commun. 2015;6:8755.
Fallahi-Sichani M, Becker V, Izar B, Baker GJ, Lin J-R, Boswell SA, et al. Adaptive resistance of melanoma cells to RAF inhibition via reversible induction of a slowly dividing de-differentiated state. Mol Syst Biol. 2017;13:905.
Shaffer SM, Dunagin MC, Torborg SR, Torre EA, Emert B, Krepler C, et al. Rare cell variability and drug-induced reprogramming as a mode of cancer drug resistance. Nature. 2017;546:431–5.
Diazzi S, Tartare-Deckert S, Deckert M. The mechanical phenotypic plasticity of melanoma cell: an emerging driver of therapy cross-resistance. Oncogenesis. 2023;12:7.
Karras P, Bordeu I, Pozniak J, Nowosad A, Pazzi C, Van Raemdonck N, et al. A cellular hierarchy in melanoma uncouples growth and metastasis. Nature. 2022;610:190–8.
Tsoi J, Robert L, Paraiso K, Galvan C, Sheu KM, Lay J, et al. Multi-stage differentiation defines melanoma subtypes with differential vulnerability to drug-induced iron-dependent oxidative stress. Cancer Cell. 2018;33:890-904.e5.
Kebschull JM, Zador AM. Cellular barcoding: lineage tracing, screening and beyond. Nat Methods. 2018;15:871–9.
Goyal Y, Busch GT, Pillai M, Li J, Boe RH, Grody EI, et al. Diverse clonal fates emerge upon drug treatment of homogeneous cancer cells. Nature. 2023;620:651–9.
Biermann J, Melms JC, Amin AD, Wang Y, Caprio LA, Karz A, et al. Dissecting the treatment-naive ecosystem of human melanoma brain metastasis. Cell. 2022;185:2591-608.e30.
Patil NS, Nabet BY, Müller S, Koeppen H, Zou W, Giltnane J, et al. Intratumoral plasma cells predict outcomes to PD-L1 blockade in non-small cell lung cancer. Cancer Cell. 2022;40:289-300.e4.
Li Y-R, Lyu Z, Tian Y, Fang Y, Zhu Y, Chen Y, et al. Advancements in CRISPR screens for the development of cancer immunotherapy strategies. Mol Ther Oncolytics. 2023;31: 100733.
Manguso RT, Pope HW, Zimmer MD, Brown FD, Yates KB, Miller BC, et al. In vivo CRISPR screening identifies Ptpn2 as a cancer immunotherapy target. Nature. 2017;547:413–8.
Baumgartner CK, Ebrahimi-Nik H, Iracheta-Vellve A, Hamel KM, Olander KE, Davis TGR, et al. The PTPN2/PTPN1 inhibitor ABBV-CLS-484 unleashes potent anti-tumour immunity. Nature. 2023;622:850–62.
Ito Y, Pan D, Zhang W, Zhang X, Juan TY, Pyrdol JW, et al. Addressing tumor heterogeneity by sensitizing resistant cancer cells to T cell-secreted cytokines. Cancer Discov. 2023;13:1186–209.
Vredevoogd DW, Kuilman T, Ligtenberg MA, Boshuizen J, Stecker KE, de Bruijn B, et al. Augmenting immunotherapy impact by lowering tumor tnf cytotoxicity threshold. Cell. 2019;178:585-99.e15.
Cucolo L, Chen Q, Qiu J, Yu Y, Klapholz M, Budinich KA, et al. The interferon-stimulated gene RIPK1 regulates cancer cell intrinsic and extrinsic resistance to immune checkpoint blockade. Immunity. 2022;55:671-85.e10.
Xu D, Jin T, Zhu H, Chen H, Ofengeim D, Zou C, et al. TBK1 Suppresses RIPK1-driven apoptosis and inflammation during development and in aging. Cell. 2018;174:1477-1491e19.
Lafont E, Draber P, Rieser E, Reichert M, Kupka S, de Miguel D, et al. TBK1 and IKKepsilon prevent TNF-induced cell death by RIPK1 phosphorylation. Nat Cell Biol. 2018;20:1389–99.
Taft J, Markson M, Legarda D, Patel R, Chan M, Malle L, et al. Human TBK1 deficiency leads to autoinflammation driven by TNF-induced cell death. Cell. 2021;184:4447-4463e20.
Sun Y, Revach O-Y, Anderson S, Kessler EA, Wolfe CH, Jenney A, et al. Targeting TBK1 to overcome resistance to cancer immunotherapy. Nature. 2023;615:158–67.
Aref AR, Campisi M, Ivanova E, Portell A, Larios D, Piel BP, et al. 3D microfluidic ex vivo culture of organotypic tumor spheroids to model immune checkpoint blockade. Lab Chip. 2018;18:3129–43.
Moriceau G, Hugo W, Hong A, Shi H, Kong X, Yu CC, et al. Tunable-combinatorial mechanisms of acquired resistance limit the efficacy of BRAF/MEK cotargeting but result in melanoma drug addiction. Cancer Cell. 2015;27:240–56.
Kong X, Kuilman T, Shahrabi A, Boshuizen J, Kemper K, Song J-Y, et al. Cancer drug addiction is relayed by an ERK2-dependent phenotype switch. Nature. 2017;550:270–4.
Bushweller JH. Targeting transcription factors in cancer—from undruggable to reality. Nat Rev Cancer. 2019;19:611–24.
Lawson KA, Sousa CM, Zhang X, Kim E, Akthar R, Caumanns JJ, et al. Functional genomic landscape of cancer-intrinsic evasion of killing by T cells. Nature. 2020;586:120–6.
Marsh T, Tolani B, Debnath J. The pleiotropic functions of autophagy in metastasis. J Cell Sci. 2021. https://doi.org/10.1242/jcs.247056.
Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, et al. Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 2018;24:541–50.
Wang L, Geng H, Liu Y, Liu L, Chen Y, Wu F, et al. Hot and cold tumors: immunological features and the therapeutic strategies. MedComm. 2020;2023(4): e343.
Huang AC, Zappasodi R. A decade of checkpoint blockade immunotherapy in melanoma: understanding the molecular basis for immune sensitivity and resistance. Nat Immunol. 2022;23:660–70.
Murciano-Goroff YR, Warner AB, Wolchok JD. The future of cancer immunotherapy: microenvironment-targeting combinations. Cell Res. 2020;30:507–19.
Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol. 2020;20:651–68.
Borst J, Ahrends T, Bąbała N, Melief CJM, Kastenmüller W. CD4 T cell help in cancer immunology and immunotherapy. Nat Rev Immunol. 2018;18:635–47.
Alspach E, Lussier DM, Miceli AP, Kizhvatov I, DuPage M, Luoma AM, et al. MHC-II neoantigens shape tumour immunity and response to immunotherapy. Nature. 2019;574:696–701.
Oliveira G, Stromhaug K, Cieri N, Iorgulescu JB, Klaeger S, Wolff JO, et al. Landscape of helper and regulatory antitumour CD4 T cells in melanoma. Nature. 2022;605:532–8.
Bonaventura P, Shekarian T, Alcazer V, Valladeau-Guilemond J, Valsesia-Wittmann S, Amigorena S, et al. Cold tumors: a therapeutic challenge for immunotherapy. Front Immunol. 2019;10:168.
Blank CU, Haining WN, Held W, Hogan PG, Kallies A, Lugli E, et al. Defining “T cell exhaustion.” Nat Rev Immunol. 2019;19:665–74.
Shitara K, Nishikawa H. Regulatory T cells: a potential target in cancer immunotherapy. Ann N Y Acad Sci. 2018;1417:104–15.
Kruse B, Buzzai AC, Shridhar N, Braun AD, Gellert S, Knauth K, et al. CD4 T cell-induced inflammatory cell death controls immune-evasive tumours. Nature. 2023;618:1033–40.
Hunder NN, Wallen H, Cao J, Hendricks DW, Reilly JZ, Rodmyre R, et al. Treatment of metastatic melanoma with autologous CD4+ T cells against NY-ESO-1. N Engl J Med. 2008;358:2698–703.
Lu Y-C, Parker LL, Lu T, Zheng Z, Toomey MA, White DE, et al. Treatment of patients with metastatic cancer using a major histocompatibility complex class II-restricted T-cell receptor targeting the cancer germline antigen MAGE-A3. J Clin Oncol. 2017;35:3322–9.
Hu Z, Ott PA, Wu CJ. Towards personalized, tumour-specific, therapeutic vaccines for cancer. Nat Rev Immunol. 2018;18:168–82.
Sellars MC, Wu CJ, Fritsch EF. Cancer vaccines: building a bridge over troubled waters. Cell. 2022;185:2770–88.
Tarhini AA, Leng S, Moschos SJ, Yin Y, Sander C, Lin Y, et al. Safety and immunogenicity of vaccination with MART-1 (26–35, 27L), gp100 (209–217, 210M), and tyrosinase (368–376, 370D) in adjuvant with PF-3512676 and GM-CSF in metastatic melanoma. J Immunother. 2012;35:359–66.
Dreno B, Thompson JF, Smithers BM, Santinami M, Jouary T, Gutzmer R, et al. MAGE-A3 immunotherapeutic as adjuvant therapy for patients with resected, MAGE-A3-positive, stage III melanoma (DERMA): a double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Oncol. 2018;19:916–29.
D’Alise AM, Scarselli E. Getting personal in metastatic melanoma: neoantigen-based vaccines as a new therapeutic strategy. Curr Opin Oncol. 2023;35:94–9.
Ott PA, Hu Z, Keskin DB, Shukla SA, Sun J, Bozym DJ, et al. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature. 2017;547:217–21.
Sahin U, Derhovanessian E, Miller M, Kloke B-P, Simon P, Löwer M, et al. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature. 2017;547:222–6.
Weber JS, Carlino MS, Khattak A, Meniawy T, Ansstas G, Taylor MH, et al. Individualised neoantigen therapy mRNA-4157 (V940) plus pembrolizumab versus pembrolizumab monotherapy in resected melanoma (KEYNOTE-942): a randomised, phase 2b study. Lancet. 2024;403:632–44.
Hu Z, Leet DE, Allesøe RL, Oliveira G, Li S, Luoma AM, et al. Personal neoantigen vaccines induce persistent memory T cell responses and epitope spreading in patients with melanoma. Nat Med. 2021;27:515–25.
Puig-Saus C, Sennino B, Peng S, Wang CL, Pan Z, Yuen B, et al. Neoantigen-targeted CD8 T cell responses with PD-1 blockade therapy. Nature. 2023;615:697–704.
Lim SY, Shklovskaya E, Lee JH, Pedersen B, Stewart A, Ming Z, et al. The molecular and functional landscape of resistance to immune checkpoint blockade in melanoma. Nat Commun. 2023;14:1516.
Li H, van der Leun AM, Yofe I, Lubling Y, Gelbard-Solodkin D, van Akkooi ACJ, et al. Dysfunctional CD8 T cells form a proliferative, dynamically regulated compartment within human melanoma. Cell. 2019;176:775-89.e18.
Miller BC, Sen DR, Al Abosy R, Bi K, Virkud YV, LaFleur MW, et al. Subsets of exhausted CD8 T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol. 2019;20:326–36.
Schietinger A, Philip M, Krisnawan VE, Chiu EY, Delrow JJ, Basom RS, et al. Tumor-specific T cell dysfunction is a dynamic antigen-driven differentiation program initiated early during tumorigenesis. Immunity. 2016;45:389–401.
Pauken KE, Sammons MA, Odorizzi PM, Manne S, Godec J, Khan O, et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science. 2016;354:1160–5.
Philip M, Fairchild L, Sun L, Horste EL, Camara S, Shakiba M, et al. Chromatin states define tumour-specific T cell dysfunction and reprogramming. Nature. 2017;545:452–6.
Sen DR, Kaminski J, Barnitz RA, Kurachi M, Gerdemann U, Yates KB, et al. The epigenetic landscape of T cell exhaustion. Science. 2016;354:1165–9.
Siddiqui I, Schaeuble K, Chennupati V, Fuertes Marraco SA, Calderon-Copete S, Pais Ferreira D, et al. Intratumoral Tcf1PD-1CD8 T cells with stem-like properties promote tumor control in response to vaccination and checkpoint blockade immunotherapy. Immunity. 2019;50:195-211.e10.
Khan O, Giles JR, McDonald S, Manne S, Ngiow SF, Patel KP, et al. TOX transcriptionally and epigenetically programs CD8 T cell exhaustion. Nature. 2019;571:211–8.
Sade-Feldman M, Yizhak K, Bjorgaard SL, Ray JP, de Boer CG, Jenkins RW, et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell. 2018;175:998-1013.e20.
Yates KB, Tonnerre P, Martin GE, Gerdemann U, Al Abosy R, Comstock DE, et al. Epigenetic scars of CD8 T cell exhaustion persist after cure of chronic infection in humans. Nat Immunol. 2021;22:1020–9.
Rudloff MW, Zumbo P, Favret NR, Roetman JJ, Detrés Román CR, Erwin MM, et al. Hallmarks of CD8 T cell dysfunction are established within hours of tumor antigen encounter before cell division. Nat Immunol. 2023;24:1527–39.
Li K, Shi H, Zhang B, Ou X, Ma Q, Chen Y, et al. Myeloid-derived suppressor cells as immunosuppressive regulators and therapeutic targets in cancer. Signal Transduct Target Ther. 2021;6:362.
Gebhardt C, Sevko A, Jiang H, Lichtenberger R, Reith M, Tarnanidis K, et al. Myeloid cells and related chronic inflammatory factors as novel predictive markers in melanoma treatment with ipilimumab. Clin Cancer Res. 2015;21:5453–9.
Noman MZ, Desantis G, Janji B, Hasmim M, Karray S, Dessen P, et al. PD-L1 is a novel direct target of HIF-1α, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med. 2014;211:781–90.
Pico de Coaña Y, Poschke I, Gentilcore G, Mao Y, Nyström M, Hansson J, et al. Ipilimumab treatment results in an early decrease in the frequency of circulating granulocytic myeloid-derived suppressor cells as well as their Arginase1 production. Cancer Immunol Res. 2013;1:158–62.
Rodriguez PC, Ernstoff MS, Hernandez C, Atkins M, Zabaleta J, Sierra R, et al. Arginase I-producing myeloid-derived suppressor cells in renal cell carcinoma are a subpopulation of activated granulocytes. Cancer Res. 2009;69:1553–60.
Srivastava MK, Sinha P, Clements VK, Rodriguez P, Ostrand-Rosenberg S. Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res. 2010;70:68–77.
Yu J, Du W, Yan F, Wang Y, Li H, Cao S, et al. Myeloid-derived suppressor cells suppress antitumor immune responses through IDO expression and correlate with lymph node metastasis in patients with breast cancer. J Immunol. 2013;190:3783–97.
Mazzoni A, Bronte V, Visintin A, Spitzer JH, Apolloni E, Serafini P, et al. Myeloid suppressor lines inhibit T cell responses by an NO-dependent mechanism. J Immunol. 2002;168:689–95.
Corzo CA, Cotter MJ, Cheng P, Cheng F, Kusmartsev S, Sotomayor E, et al. Mechanism regulating reactive oxygen species in tumor-induced myeloid-derived suppressor cells. J Immunol. 2009;182:5693–701.
Molon B, Ugel S, Del Pozzo F, Soldani C, Zilio S, Avella D, et al. Chemokine nitration prevents intratumoral infiltration of antigen-specific T cells. J Exp Med. 2011;208:1949–62.
Hoechst B, Voigtlaender T, Ormandy L, Gamrekelashvili J, Zhao F, Wedemeyer H, et al. Myeloid derived suppressor cells inhibit natural killer cells in patients with hepatocellular carcinoma via the NKp30 receptor. Hepatology. 2009;50:799–807.
Ugolini A, Tyurin VA, Tyurina YY, Tcyganov EN, Donthireddy L, Kagan VE, et al. Polymorphonuclear myeloid-derived suppressor cells limit antigen cross-presentation by dendritic cells in cancer. JCI Insight. 2020. https://doi.org/10.1172/jci.insight.138581.
Siret C, Collignon A, Silvy F, Robert S, Cheyrol T, André P, et al. Deciphering the crosstalk between myeloid-derived suppressor cells and regulatory t cells in pancreatic ductal adenocarcinoma. Front Immunol. 2019;10:3070.
Ozbay Kurt FG, Lasser S, Arkhypov I, Utikal J, Umansky V. Enhancing immunotherapy response in melanoma: myeloid-derived suppressor cells as a therapeutic target. J Clin Invest. 2023. https://doi.org/10.1172/JCI170762.
Steinberg SM, Shabaneh TB, Zhang P, Martyanov V, Li Z, Malik BT, et al. Myeloid cells that impair immunotherapy are restored in melanomas with acquired resistance to BRAF inhibitors. Cancer Res. 2017;77:1599–610.
De Henau O, Rausch M, Winkler D, Campesato LF, Liu C, Cymerman DH, et al. Overcoming resistance to checkpoint blockade therapy by targeting PI3Kγ in myeloid cells. Nature. 2016;539:443–7.
Hong DS, Postow M, Chmielowski B, Sullivan R, Patnaik A, Cohen EEW, et al. Eganelisib, a first-in-class PI3Kγ inhibitor, in patients with advanced solid tumors: results of the phase 1/1b MARIO-1 trial. Clin Cancer Res. 2023;29:2210–9.
Heras-Murillo I, Adán-Barrientos I, Galán M, Wculek SK, Sancho D. Dendritic cells as orchestrators of anticancer immunity and immunotherapy. Nat Rev Clin Oncol. 2024;21:257–77.
Böttcher JP, Reis e Sousa C. The role of type 1 conventional dendritic cells in cancer immunity. Trends Cancer Res. 2018;4:784–92.
Garris CS, Arlauckas SP, Kohler RH, Trefny MP, Garren S, Piot C, et al. Successful anti-PD-1 cancer immunotherapy requires T cell-dendritic cell crosstalk involving the cytokines IFN-γ and IL-12. Immunity. 2018;49:1148-61.e7.
Lee YS, O’Brien LJ, Walpole CM, Pearson FE, Leal-Rojas IM, Masterman K-A, et al. Human CD141 dendritic cells (cDC1) are impaired in patients with advanced melanoma but can be targeted to enhance anti-PD-1 in a humanized mouse model. J Immunother Cancer. 2021;9:e001963. https://doi.org/10.1136/jitc-2020-001963.
Bol KF, Aarntzen EHJG, ’t Hout FEMI, Schreibelt G, Creemers JHA, Lesterhuis WJ, et al. Favorable overall survival in stage III melanoma patients after adjuvant dendritic cell vaccination. Oncoimmunology. 2016;5: e1057673.
Soruri A, Kiafard Z, Dettmer C, Riggert J, Köhl J, Zwirner J. IL-4 down-regulates anaphylatoxin receptors in monocytes and dendritic cells and impairs anaphylatoxin-induced migration in vivo. J Immunol. 2003;170:3306–14.
Wimmers F, Schreibelt G, Sköld AE, Figdor CG, De Vries IJM. Paradigm shift in dendritic cell-based immunotherapy: from in vitro generated monocyte-derived DCs to naturally circulating DC subsets. Front Immunol. 2014;5:165.
Bol KF, Schreibelt G, Bloemendal M, van Willigen WW, Hins-de Bree S, de Goede AL, et al. Adjuvant dendritic cell therapy in stage IIIB/C melanoma: the MIND-DC randomized phase III trial. Nat Commun. 2024;15:1632.
Sautès-Fridman C, Petitprez F, Calderaro J, Fridman WH. Tertiary lymphoid structures in the era of cancer immunotherapy. Nat Rev Cancer. 2019;19:307–25.
Laumont CM, Banville AC, Gilardi M, Hollern DP, Nelson BH. Tumour-infiltrating B cells: immunological mechanisms, clinical impact and therapeutic opportunities. Nat Rev Cancer. 2022;22:414–30.
Singh S, Roszik J, Saini N, Singh VK, Bavisi K, Wang Z, et al. B cells are required to generate optimal anti-melanoma immunity in response to checkpoint blockade. Front Immunol. 2022;13: 794684.
Cabrita R, Lauss M, Sanna A, Donia M, Skaarup Larsen M, Mitra S, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577:561–5.
Bod L, Kye Y-C, Shi J, Torlai Triglia E, Schnell A, Fessler J, et al. B-cell-specific checkpoint molecules that regulate anti-tumour immunity. Nature. 2023;619:348–56.
Prince EA, Sanzari JK, Pandya D, Huron D, Edwards R. Analytical concordance of PD-L1 assays utilizing antibodies from FDA-approved diagnostics in advanced cancers: a systematic literature review. JCO Precis Oncol. 2021;5:953–73.
Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med. 2015;372:2509–20.
Yamaguchi H, Hsu J-M, Sun L, Wang S-C, Hung M-C. Advances and prospects of biomarkers for immune checkpoint inhibitors. Cell Rep Med. 2024;5: 101621.
Jalal S, Earley JN, Turchi JJ. DNA repair: from genome maintenance to biomarker and therapeutic target. Clin Cancer Res. 2011;17:6973–84.
Rousseau B, Bieche I, Pasmant E, Hamzaoui N, Leulliot N, Michon L, et al. PD-1 Blockade in solid tumors with defects in polymerase epsilon. Cancer Discov. 2022;12:1435–48.
Goodman AM, Castro A, Pyke RM, Okamura R, Kato S, Riviere P, et al. MHC-I genotype and tumor mutational burden predict response to immunotherapy. Genome Med. 2020;12:45.
Marty R, Kaabinejadian S, Rossell D, Slifker MJ, van de Haar J, Engin HB, et al. MHC-I genotype restricts the oncogenic mutational landscape. Cell. 2017;171:1272-83.e15.
Sacco A, Forgione L, Carotenuto M, Luca AD, Ascierto PA, Botti G, et al. Circulating tumor DNA testing opens new perspectives in melanoma management. Cancers. 2020;12:2914. https://doi.org/10.3390/cancers12102914.
Leung AM, Lee AF, Ozao-Choy J, Ramos RI, Hamid O, O’Day SJ, et al. Clinical benefit from ipilimumab therapy in melanoma patients may be associated with serum CTLA4 levels. Front Oncol. 2014;4:110.
Bartlett EK, Flynn JR, Panageas KS, Ferraro RA, Sta Cruz JM, Postow MA, et al. High neutrophil-to-lymphocyte ratio (NLR) is associated with treatment failure and death in patients who have melanoma treated with PD-1 inhibitor monotherapy. Cancer. 2020;126:76–85.
Mallardo D, Fordellone M, White A, Ottaviano M, Sparano F, Bailey M, et al. CD39 and LDHA affects the prognostic role of NLR in metastatic melanoma patients treated with immunotherapy. J Transl Med. 2023;21:610.
Weber R, Groth C, Lasser S, Arkhypov I, Petrova V, Altevogt P, et al. IL-6 as a major regulator of MDSC activity and possible target for cancer immunotherapy. Cell Immunol. 2021;359: 104254.
Cortellini A, D’Alessio A, Cleary S, Buti S, Bersanelli M, Bordi P, et al. Type 2 diabetes mellitus and efficacy outcomes from immune checkpoint blockade in patients with cancer. Clin Cancer Res. 2023;29:2714–24.
Mallardo D, Woodford R, Menzies AM, Zimmer L, Williamson A, Ramelyte E, et al. The role of diabetes in metastatic melanoma patients treated with nivolumab plus relatlimab. J Transl Med. 2023;21:753.
Gopalakrishnan V, Spencer CN, Nezi L, Reuben A, Andrews MC, Karpinets TV, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359:97–103.
McCulloch JA, Davar D, Rodrigues RR, Badger JH, Fang JR, Cole AM, et al. Intestinal microbiota signatures of clinical response and immune-related adverse events in melanoma patients treated with anti-PD-1. Nat Med. 2022;28:545–56.
Limeta A, Ji B, Levin M, Gatto F, Nielsen J. Meta-analysis of the gut microbiota in predicting response to cancer immunotherapy in metastatic melanoma. JCI Insight. 2020. https://doi.org/10.1172/jci.insight.140940.
Matson V, Fessler J, Bao R, Chongsuwat T, Zha Y, Alegre M-L, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359:104–8.
Li H, van der Merwe PA, Sivakumar S. Biomarkers of response to PD-1 pathway blockade. Br J Cancer. 2022;126:1663–75.
Zhang T, Forde PM, Sullivan RJ, Sharon E, Barksdale E, Selig W, et al. Addressing resistance to PD-1/PD-(L)1 pathway inhibition: considerations for combinatorial clinical trial designs. J Immunother Cancer. 2023;11: e006555. https://doi.org/10.1136/jitc-2022-006555.
Author information
Authors and Affiliations
Corresponding author
Ethics declarations
Funding
This paper was supported by the grant National Institutes of Health R38AG070229 to David X. Zheng.
Conflicts of Interest
Russell W. Jenkins is a member of the advisory board for and has a financial interest in Xsphera Biosciences Inc., a company focused on using ex vivo profiling technology to deliver functional, precision immune-oncology solutions for patients, providers, and drug development companies. Russell W. Jenkins has received honoraria from Incyte (invited speaker), G1 Therapeutics (advisory board), and Bioxcel Therapeutics (invited speaker). Russell W. Jenkins has ownership interest in US patents US20200399573A9 and US20210363595A1. Russell W. Jenkins’s interests were reviewed and are managed by Massachusetts General Hospital and Mass General Brigham in accordance with their conflict-of-interest policies. Ryan J. Sullivan has served as a consultant or on an advisory board for BMS, Merck, Pfizer, Marengo, and Replimune, and has received research funding from Merck. David X. Zheng, David J. Bozym, Giuseppe Tarantino, and David Liu declare that they have no conflicts of interest that might be relevant to the contents of this manuscript.
Ethics Approval
Not applicable.
Consent to Participate
Not applicable.
Consent for Publication
Not applicable.
Availability of Data and Material
No datasets were generated or analyzed during the current study.
Code Availability
Not applicable.
Authors’ Contributions
D.X.Z.: conceptualization, writing—original draft preparation, project administration, and funding acquisition. D.J.B.: conceptualization and writing—original draft preparation. G.T.: conceptualization, writing—original draft preparation, and visualization. R.J.S.: conceptualization, writing—reviewing and editing, and supervision. D.L.: conceptualization and writing—reviewing and editing, and supervision. R.W.J.: conceptualization, writing—reviewing and editing, supervision, and project administration.
Rights and permissions
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
About this article
Cite this article
Zheng, D.X., Bozym, D.J., Tarantino, G. et al. Overcoming Resistance Mechanisms to Melanoma Immunotherapy. Am J Clin Dermatol 26, 77–96 (2025). https://doi.org/10.1007/s40257-024-00907-7
Accepted:
Published:
Issue Date:
DOI: https://doi.org/10.1007/s40257-024-00907-7