Insights and Strategies of Melanoma Immunotherapy: Predictive Biomarkers of Response and Resistance and Strategies to Improve Response Rates
Abstract
:1. Introduction
2. Immune Checkpoint Inhibitors
3. Melanoma Biomarkers of Response to ICI
3.1. Established Clinical Biomarkers
3.2. Emerging Predictive Biomarkers
3.2.1. Gene Expression Signatures
3.2.2. Gene Expression Signatures at Baseline and On-Treatment Tumor Specimens
3.2.3. Pathway Signatures
3.2.4. Tumor Antigens
3.2.5. Genomic Alterations
4. Who Is Responding to ICI?
5. Tumor-Immunity Cycle and Resistance Mechanisms Involving Tumor Immunophenotypes
5.1. Tumor Cell-Intrinsic and Tumor Cell-Extrinsic Resistance Mechanisms
5.2. Immune Resistance Mechanisms in Melanoma
6. Therapeutic Strategies to Turn “Cold Tumors” into “Hot Tumors”
7. Lessons Learned: ICI Therapies in Melanoma
8. Conclusions and Future Perspectives
Author Contributions
Funding
Institutional Review Board Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
Abbreviations
A2aR | adenosine A2a receptor |
ADO | adenosine |
AE | adverse event |
APC | antigen-presenting cell |
ATP | adenosine triphosphate |
B2M | beta-2-microglobulin |
B7RP1 | B7-related protein 1 |
BATF3 | basic leucine zipper ATF-like transcription factor 3 |
BTLA | B and T lymphocyte attenuator |
CAF | cancer-associated fibroblast |
CCR4 | C-C chemokine receptor type 4 |
CD40L | CD40 ligand |
CDN | cyclic dinucleotide |
CRT | Calreticulin |
CSF-1 | colony stimulating factor 1 |
CTCs | circulating tumor cells |
ctDNA | circulating tumor DNA |
CTLA-4 | cytotoxic T-lymphocyte-associated protein 4 |
CXCL/CCL | chemokine motif ligands; |
dMMR | defective mismatch repair |
DNMT | DNA methyltransferase |
ECM | extracellular matrix |
EGFR | epidermal growth factor receptor |
ER | endoplasmic reticulum |
ETBR | endothelin B receptor |
EZH2 | enhancer of zeste homolog 2 |
FDA | Food and Drug Administration |
FLT3L | Fms-like tyrosine kinase 3 ligand |
GAL9 | galectin 9 |
GITR | glucocorticoid-induced tumor necrosis factor receptor |
GM-CSF | granulocyte-macrophage colony-stimulating factor |
HDAC | histone deacetylase |
HER2 | human epidermal growth factor receptor 2 |
HEV | high endothelial venule |
HLA | human leukocyte antigen |
HMGB1 | high mobility family protein B1 |
HR | hormone receptor positive |
HVEM | herpesvirus entry mediator |
ICAM | intercellular adhesion molecule |
ICOS | inducible T cell co-stimulator |
IDO | indoleamine 2,3-dioxygenase |
IFN | interferon |
IL | interleukin |
ITT | intention-to-treat |
KIR | killer cell immunoglobulin-like receptors |
LAG-3 | lymphocyte activation gene 3 |
LFA1 | lymphocyte function-associated antigen-1 |
mAb | monoclonal antibody |
M-CSF | macrophage colony-stimulating factor |
MDSC | myeloid-derived suppressor cell |
MHC | major histocompatibility complex |
MMR | mismatch repair |
MSI | microsatellite instability |
MSI-H | microsatellite instability-high |
MɸII | type II macrophage |
NK | natural killer |
OS | overall survival |
OX40L | OX40 ligand |
PD-1 | programmed cell death protein-1 |
PD-L1 | programmed death-ligand 1 |
PD-L2 | programmed death-ligand 2 |
PFS | progression-free survival |
PI3K | phosphatidyl-inositol 3-kinase |
PTEN | phosphatase and tensin homolog |
STC1 | stanniocalcin 1 |
TAM | tumor-associated macrophage |
TAP | transporter associated with antigen processing |
TCR | T-cell receptor |
TGF | transforming growth factor |
TGFβ | transforming growth factor-beta |
TILs | tumor-infiltrating lymphocytes |
TIM-3 | T cell immunoglobulin and mucin-domain containing-3 |
TLR | Toll-like receptor |
TLS | tertiary lymphoid structure |
TMB | tumor mutation burden |
TME | tumor microenvironment |
TNF | tumor necrosis factor |
TNFRSF14 | tumor necrosis factor receptor superfamily member 14 |
Treg | T-regulatory cell |
VCAM | vascular cell adhesion molecule |
VEGF | vascular endothelial growth factor |
VISTA | V-domain Ig suppressor of T cell activation |
References
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharma, P.; Allison, J.P. Immune checkpoint targeting in cancer therapy: Toward combination strategies with curative potential. Cell 2015, 161, 205–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2018. CA Cancer J. Clin. 2018, 68, 7–30. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Leach, D.R.; Krummel, M.F.; Allison, J.P. Enhancement of antitumor immunity by CTLA-4 blockade. Science 1996, 271, 1734–1736. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Schumacher, T.N.; Schreiber, R.D. Neoantigens in cancer immunotherapy. Science 2015, 348, 69–74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zappasodi, R.; Wolchok, J.D.; Merghoub, T. Correction to: Strategies for Predicting Response to Checkpoint Inhibitors. Curr. Hematol. Malig. Rep. 2019, 14, 62. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Paik, J. Nivolumab Plus Relatlimab: First Approval. Drugs 2022, 82, 925–931. [Google Scholar] [CrossRef] [PubMed]
- Sharma, P.; Hu-Lieskovan, S.; Wargo, J.A.; Ribas, A. Primary, Adaptive, and Acquired Resistance to Cancer Immunotherapy. Cell 2017, 168, 707–723. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Gibney, G.T.; Weiner, L.M.; Atkins, M.B. Predictive biomarkers for checkpoint inhibitor-based immunotherapy. Lancet Oncol. 2016, 17, e542–e551. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef]
- Carlino, M.S.; Larkin, J.; Long, G.V. Immune checkpoint inhibitors in melanoma. Lancet 2021, 398, 1002–1014. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Gajewski, T.F.; Schreiber, H.; Fu, Y.X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 2013, 14, 1014–1022. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pitt, J.M.; Vétizou, M.; Daillère, R.; Roberti, M.P.; Yamazaki, T.; Routy, B.; Lepage, P.; Boneca, I.G.; Chamaillard, M.; Kroemer, G.; et al. Resistance Mechanisms to Immune-Checkpoint Blockade in Cancer: Tumor-Intrinsic and -Extrinsic Factors. Immunity 2016, 44, 1255–1269. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Zhang, N.; Song, J.; Liu, Y.; Liu, M.; Zhang, L.; Sheng, D.; Deng, L.; Yi, H.; Wu, M.; Zheng, Y.; et al. Photothermal therapy mediated by phase-transformation nanoparticles facilitates delivery of anti-PD1 antibody and synergizes with antitumor immunotherapy for melanoma. J. Control. Release 2019, 306, 15–28. [Google Scholar] [CrossRef]
- Khong, H.T.; Restifo, N.P. Natural selection of tumor variants in the generation of "tumor escape" phenotypes. Nat. Immunol. 2002, 3, 999–1005. [Google Scholar] [CrossRef] [Green Version]
- Pitt, J.M.; Marabelle, A.; Eggermont, A.; Soria, J.C.; Kroemer, G.; Zitvogel, L. Targeting the tumor microenvironment: Removing obstruction to anticancer immune responses and immunotherapy. Ann. Oncol. 2016, 27, 1482–1492. [Google Scholar] [CrossRef]
- Restifo, N.P.; Smyth, M.J.; Snyder, A. Acquired resistance to immunotherapy and future challenges. Nat. Rev. Cancer 2016, 16, 121–126. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Iliopoulos, D.; Kavousanaki, M.; Ioannou, M.; Boumpas, D.; Verginis, P. The negative costimulatory molecule PD-1 modulates the balance between immunity and tolerance via miR-21. Eur. J. Immunol. 2011, 41, 1754–1763. [Google Scholar] [CrossRef] [PubMed]
- Han, Y.; Liu, D.; Li, L. PD-1/PD-L1 pathway: Current researches in cancer. Am. J. Cancer Res. 2020, 10, 727–742. [Google Scholar] [PubMed]
- Carlsen, L.; Huntington, K.E.; El-Deiry, W.S. Immunotherapy for Colorectal Cancer: Mechanisms and Predictive Biomarkers. Cancers 2022, 14, 1028. [Google Scholar] [CrossRef] [PubMed]
- Gravbrot, N.; Gilbert-Gard, K.; Mehta, P.; Ghotmi, Y.; Banerjee, M.; Mazis, C.; Sundararajan, S. Therapeutic Monoclonal Antibodies Targeting Immune Checkpoints for the Treatment of Solid Tumors. Antibodies 2019, 8, 51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fan, J.; Shang, D.; Han, B.; Song, J.; Chen, H.; Yang, J.M. Adoptive Cell Transfer: Is it a Promising Immunotherapy for Colorectal Cancer? Theranostics 2018, 8, 5784–5800. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Weinstein, D.; Leininger, J.; Hamby, C.; Safai, B. Diagnostic and prognostic biomarkers in melanoma. J. Clin. Aesthet. Dermatol. 2014, 7, 13–24. [Google Scholar]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Genomic Classification of Cutaneous Melanoma. Cell 2015, 161, 1681–1696. [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Xiao, X.; Xu, C.; Yang, W.; Yu, R. Inconsistent prediction capability of ImmuneCells.Sig across different RNA-seq datasets. Nat. Commun. 2021, 12, 4167. [Google Scholar] [CrossRef]
- Xiong, D.; Wang, Y.; You, M. Reply to: "Inconsistent prediction capability of ImmuneCells.Sig across different RNA-seq datasets". Nat. Commun. 2021, 12, 4168. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vogelstein, B.; Papadopoulos, N.; Velculescu, V.E.; Zhou, S.; Diaz, L.A., Jr.; Kinzler, K.W. Cancer genome landscapes. Science 2013, 339, 1546–1558. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Hogan, S.A.; Levesque, M.P.; Cheng, P.F. Melanoma Immunotherapy: Next-Generation Biomarkers. Front. Oncol. 2018, 8, 178. [Google Scholar] [CrossRef]
- Keenan, T.E.; Burke, K.P.; Van Allen, E.M. Genomic correlates of response to immune checkpoint blockade. Nat. Med. 2019, 25, 389–402. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharma, P.; Allison, J.P. The future of immune checkpoint therapy. Science 2015, 348, 56–61. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.T.; Sun, Z.J. Turning cold tumors into hot tumors by improving T-cell infiltration. Theranostics 2021, 11, 5365–5386. [Google Scholar] [CrossRef] [PubMed]
- Chen, D.S.; Mellman, I. Oncology meets immunology: The cancer-immunity cycle. Immunity 2013, 39, 1–10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, D.S.; Mellman, I. Elements of cancer immunity and the cancer-immune set point. Nature 2017, 541, 321–330. [Google Scholar] [CrossRef] [PubMed]
- Hegde, P.S.; Karanikas, V.; Evers, S. The Where, the When, and the How of Immune Monitoring for Cancer Immunotherapies in the Era of Checkpoint Inhibition. Clin. Cancer Res. 2016, 22, 1865–1874. [Google Scholar] [CrossRef] [Green Version]
- Galon, J.; Bruni, D. Approaches to treat immune hot, altered and cold tumours with combination immunotherapies. Nat. Rev. Drug Discov. 2019, 18, 197–218. [Google Scholar] [CrossRef]
- Hegde, P.S.; Chen, D.S. Top 10 Challenges in Cancer Immunotherapy. Immunity 2020, 52, 17–35. [Google Scholar] [CrossRef]
- Ansell, S.M.; Lesokhin, A.M.; Borrello, I.; Halwani, A.; Scott, E.C.; Gutierrez, M.; Schuster, S.J.; Millenson, M.M.; Cattry, D.; Freeman, G.J.; et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N. Engl. J. Med. 2015, 372, 311–319. [Google Scholar] [CrossRef] [Green Version]
- Gubin, M.M.; Zhang, X.; Schuster, H.; Caron, E.; Ward, J.P.; Noguchi, T.; Ivanova, Y.; Hundal, J.; Arthur, C.D.; Krebber, W.J.; et al. Checkpoint blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 2014, 515, 577–581. [Google Scholar] [CrossRef] [PubMed]
- Marincola, F.M.; Jaffee, E.M.; Hicklin, D.J.; Ferrone, S. Escape of human solid tumors from T-cell recognition: Molecular mechanisms and functional significance. Adv. Immunol. 2000, 74, 181–273. [Google Scholar] [PubMed]
- Sucker, A.; Zhao, F.; Real, B.; Heeke, C.; Bielefeld, N.; Maβen, S.; Horn, S.; Moll, I.; Maltaner, R.; Horn, P.A.; et al. Genetic evolution of T-cell resistance in the course of melanoma progression. Clin. Cancer Res. 2014, 20, 6593–6604. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, C.; Peng, W.; Xu, C.; Lou, Y.; Zhang, M.; Wargo, J.A.; Chen, J.Q.; Li, H.S.; Watowich, S.S.; Yang, Y.; et al. BRAF inhibition increases tumor infiltration by T cells and enhances the antitumor activity of adoptive immunotherapy in mice. Clin. Cancer Res. 2013, 19, 393–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Spranger, S.; Bao, R.; Gajewski, T.F. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature 2015, 523, 231–235. [Google Scholar] [CrossRef]
- Shin, D.S.; Zaretsky, J.M.; Escuin-Ordinas, H.; Garcia-Diaz, A.; Hu-Lieskovan, S.; Kalbasi, A.; Grasso, C.S.; Hugo, W.; Sandoval, S.; Torrejon, D.Y.; et al. Primary Resistance to PD-1 Blockade Mediated by JAK1/2 Mutations. Cancer Discov. 2017, 7, 188–201. [Google Scholar] [CrossRef] [Green Version]
- He, Y.; Cao, J.; Zhao, C.; Li, X.; Zhou, C.; Hirsch, F.R. TIM-3, a promising target for cancer immunotherapy. Onco. Targets Ther. 2018, 11, 7005–7009. [Google Scholar] [CrossRef] [Green Version]
- Kryczek, I.; Zou, L.; Rodriguez, P.; Zhu, G.; Wei, S.; Mottram, P.; Brumlik, M.; Cheng, P.; Curiel, T.; Myers, L.; et al. B7-H4 expression identifies a novel suppressive macrophage population in human ovarian carcinoma. J. Exp. Med. 2006, 203, 871–881. [Google Scholar] [CrossRef]
- Kuang, D.M.; Zhao, Q.; Peng, C.; Xu, J.; Zhang, J.P.; Wu, C.; Zheng, L. Activated monocytes in peritumoral stroma of hepatocellular carcinoma foster immune privilege and disease progression through PD-L1. J. Exp. Med. 2009, 206, 1327–1337. [Google Scholar] [CrossRef] [Green Version]
- Meyer, C.; Cagnon, L.; Costa-Nunes, C.M.; Baumgaertner, P.; Montandon, N.; Leyvraz, L.; Michielin, O.; Romano, E.; Speiser, D.E. Frequencies of circulating MDSC correlate with clinical outcome of melanoma patients treated with ipilimumab. Cancer Immunol. Immunother. 2014, 63, 247–257. [Google Scholar] [CrossRef] [Green Version]
- Şenbabaoğlu, Y.; Gejman, R.S.; Winer, A.G.; Liu, M.; Van Allen, E.M.; de Velasco, G.; Miao, D.; Ostrovnaya, I.; Drill, E.; Luna, A.; et al. Tumor immune microenvironment characterization in clear cell renal cell carcinoma identifies prognostic and immunotherapeutically relevant messenger RNA signatures. Genome Biol. 2016, 17, 231. [Google Scholar] [CrossRef] [PubMed]
- Charoentong, P.; Finotello, F.; Angelova, M.; Mayer, C.; Efremova, M.; Rieder, D.; Hackl, H.; Trajanoski, Z. Pan-cancer Immunogenomic Analyses Reveal Genotype-Immunophenotype Relationships and Predictors of Response to Checkpoint Blockade. Cell Rep. 2017, 18, 248–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prat, A.; Navarro, A.; Paré, L.; Reguart, N.; Galván, P.; Pascual, T.; Martínez, A.; Nuciforo, P.; Comerma, L.; Alos, L.; et al. Immune-Related Gene Expression Profiling After PD-1 Blockade in Non-Small Cell Lung Carcinoma, Head and Neck Squamous Cell Carcinoma, and Melanoma. Cancer Res. 2017, 77, 3540–3550. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Danaher, P.; Warren, S.; Lu, R.; Samayoa, J.; Sullivan, A.; Pekker, I.; Wallden, B.; Marincola, F.M.; Cesano, A. Pan-cancer adaptive immune resistance as defined by the Tumor Inflammation Signature (TIS): Results from The Cancer Genome Atlas (TCGA). J. Immunother. Cancer 2018, 6, 63. [Google Scholar] [CrossRef] [Green Version]
- McDermott, D.F.; Huseni, M.A.; Atkins, M.B.; Motzer, R.J.; Rini, B.I.; Escudier, B.; Fong, L.; Joseph, R.W.; Pal, S.K.; Reeves, J.A.; et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nat. Med. 2018, 24, 749–757. [Google Scholar] [CrossRef]
- Anagnostou, V.; Smith, K.N.; Forde, P.M.; Niknafs, N.; Bhattacharya, R.; White, J.; Zhang, T.; Adleff, V.; Phallen, J.; Wali, N.; et al. Evolution of Neoantigen Landscape during Immune Checkpoint Blockade in Non-Small Cell Lung Cancer. Cancer Discov. 2017, 7, 264–276. [Google Scholar] [CrossRef] [Green Version]
- Im, S.J.; Hashimoto, M.; Gerner, M.Y.; Lee, J.; Kissick, H.T.; Burger, M.C.; Shan, Q.; Hale, J.S.; Lee, J.; Nasti, T.H.; et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 2016, 537, 417–421. [Google Scholar] [CrossRef] [Green Version]
- Seiwert, T.Y.; Burtness, B.; Mehra, R.; Weiss, J.; Berger, R.; Eder, J.P.; Heath, K.; McClanahan, T.; Lunceford, J.; Gause, C.; et al. Safety and clinical activity of pembrolizumab for treatment of recurrent or metastatic squamous cell carcinoma of the head and neck (KEYNOTE-012): An open-label, multicentre, phase 1b trial. Lancet Oncol. 2016, 17, 956–965. [Google Scholar] [CrossRef]
- Fehlings, M.; Simoni, Y.; Penny, H.L.; Becht, E.; Loh, C.Y.; Gubin, M.M.; Ward, J.P.; Wong, S.C.; Schreiber, R.D.; Newell, E.W. Checkpoint blockade immunotherapy reshapes the high-dimensional phenotypic heterogeneity of murine intratumoural neoantigen-specific CD8(+) T cells. Nat. Commun. 2017, 8, 562. [Google Scholar] [CrossRef]
- van Rooij, N.; van Buuren, M.M.; Philips, D.; Velds, A.; Toebes, M.; Heemskerk, B.; van Dijk, L.J.; Behjati, S.; Hilkmann, H.; El Atmioui, D.; et al. Tumor exome analysis reveals neoantigen-specific T-cell reactivity in an ipilimumab-responsive melanoma. J. Clin. Oncol. 2013, 31, e439–e442. [Google Scholar] [CrossRef]
- Le, D.T.; Durham, J.N.; Smith, K.N.; Wang, H.; Bartlett, B.R.; Aulakh, L.K.; Lu, S.; Kemberling, H.; Wilt, C.; Luber, B.S.; et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science 2017, 357, 409–413. [Google Scholar] [CrossRef] [PubMed]
- Le, D.T.; Uram, J.N.; Wang, H.; Bartlett, B.R.; Kemberling, H.; Eyring, A.D.; Skora, A.D.; Luber, B.S.; Azad, N.S.; Laheru, D.; et al. PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N. Engl. J. Med. 2015, 372, 2509–2520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nghiem, P.T.; Bhatia, S.; Lipson, E.J.; Kudchadkar, R.R.; Miller, N.J.; Annamalai, L.; Berry, S.; Chartash, E.K.; Daud, A.; Fling, S.P.; et al. PD-1 Blockade with Pembrolizumab in Advanced Merkel-Cell Carcinoma. N. Engl. J. Med. 2016, 374, 2542–2552. [Google Scholar] [CrossRef] [PubMed]
- Łuksza, M.; Riaz, N.; Makarov, V.; Balachandran, V.P.; Hellmann, M.D.; Solovyov, A.; Rizvi, N.A.; Merghoub, T.; Levine, A.J.; Chan, T.A.; et al. A neoantigen fitness model predicts tumour response to checkpoint blockade immunotherapy. Nature 2017, 551, 517–520. [Google Scholar] [CrossRef]
- Panda, A.; de Cubas, A.A.; Stein, M.; Riedlinger, G.; Kra, J.; Mayer, T.; Smith, C.C.; Vincent, B.G.; Serody, J.S.; Beckermann, K.E.; et al. Endogenous retrovirus expression is associated with response to immune checkpoint blockade in clear cell renal cell carcinoma. JCI Insight 2018, 3, e121522. [Google Scholar] [CrossRef]
- Rosenberg, J.E.; Hoffman-Censits, J.; Powles, T.; van der Heijden, M.S.; Balar, A.V.; Necchi, A.; Dawson, N.; O’Donnell, P.H.; Balmanoukian, A.; Loriot, Y.; et al. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: A single-arm, multicentre, phase 2 trial. Lancet 2016, 387, 1909–1920. [Google Scholar] [CrossRef] [Green Version]
- Hellmann, M.D.; Callahan, M.K.; Awad, M.M.; Calvo, E.; Ascierto, P.A.; Atmaca, A.; Rizvi, N.A.; Hirsch, F.R.; Selvaggi, G.; Szustakowski, J.D.; et al. Tumor Mutational Burden and Efficacy of Nivolumab Monotherapy and in Combination with Ipilimumab in Small-Cell Lung Cancer. Cancer Cell 2018, 33, 853–861.e4. [Google Scholar] [CrossRef] [Green Version]
- Riaz, N.; Havel, J.J.; Kendall, S.M.; Makarov, V.; Walsh, L.A.; Desrichard, A.; Weinhold, N.; Chan, T.A. Recurrent SERPINB3 and SERPINB4 mutations in patients who respond to anti-CTLA4 immunotherapy. Nat. Genet. 2016, 48, 1327–1329. [Google Scholar] [CrossRef]
- Miao, D.; Margolis, C.A.; Vokes, N.I.; Liu, D.; Taylor-Weiner, A.; Wankowicz, S.M.; Adeegbe, D.; Keliher, D.; Schilling, B.; Tracy, A.; et al. Genomic correlates of response to immune checkpoint blockade in microsatellite-stable solid tumors. Nat. Genet. 2018, 50, 1271–1281. [Google Scholar] [CrossRef]
- Wang, S.; Jia, M.; He, Z.; Liu, X.S. APOBEC3B and APOBEC mutational signature as potential predictive markers for immunotherapy response in non-small cell lung cancer. Oncogene 2018, 37, 3924–3936. [Google Scholar] [CrossRef]
- Chen, R.; Zinzani, P.L.; Fanale, M.A.; Armand, P.; Johnson, N.A.; Brice, P.; Radford, J.; Ribrag, V.; Molin, D.; Vassilakopoulos, T.P.; et al. Phase II Study of the Efficacy and Safety of Pembrolizumab for Relapsed/Refractory Classic Hodgkin Lymphoma. J. Clin. Oncol. 2017, 35, 2125–2132. [Google Scholar] [CrossRef] [PubMed]
- Green, M.R.; Monti, S.; Rodig, S.J.; Juszczynski, P.; Currie, T.; O’Donnell, E.; Chapuy, B.; Takeyama, K.; Neuberg, D.; Golub, T.R.; et al. Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood 2010, 116, 3268–3277. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nayak, L.; Iwamoto, F.M.; LaCasce, A.; Mukundan, S.; Roemer, M.G.M.; Chapuy, B.; Armand, P.; Rodig, S.J.; Shipp, M.A. PD-1 blockade with nivolumab in relapsed/refractory primary central nervous system and testicular lymphoma. Blood 2017, 129, 3071–3073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Goodman, A.M.; Piccioni, D.; Kato, S.; Boichard, A.; Wang, H.Y.; Frampton, G.; Lippman, S.M.; Connelly, C.; Fabrizio, D.; Miller, V.; et al. Prevalence of PDL1 Amplification and Preliminary Response to Immune Checkpoint Blockade in Solid Tumors. JAMA Oncol. 2018, 4, 1237–1244. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.; Bu, X.; Wang, H.; Zhu, Y.; Geng, Y.; Nihira, N.T.; Tan, Y.; Ci, Y.; Wu, F.; Dai, X.; et al. Cyclin D-CDK4 kinase destabilizes PD-L1 via cullin 3-SPOP to control cancer immune surveillance. Nature 2018, 553, 91–95. [Google Scholar] [CrossRef] [Green Version]
- Mezzadra, R.; Sun, C.; Jae, L.T.; Gomez-Eerland, R.; de Vries, E.; Wu, W.; Logtenberg, M.E.W.; Slagter, M.; Rozeman, E.A.; Hofland, I.; et al. Identification of CMTM6 and CMTM4 as PD-L1 protein regulators. Nature 2017, 549, 106–110. [Google Scholar] [CrossRef]
- Burr, M.L.; Sparbier, C.E.; Chan, Y.C.; Williamson, J.C.; Woods, K.; Beavis, P.A.; Lam, E.Y.N.; Henderson, M.A.; Bell, C.C.; Stolzenburg, S.; et al. CMTM6 maintains the expression of PD-L1 and regulates anti-tumour immunity. Nature 2017, 549, 101–105. [Google Scholar] [CrossRef] [Green Version]
- Pan, D.; Kobayashi, A.; Jiang, P.; Ferrari de Andrade, L.; Tay, R.E.; Luoma, A.M.; Tsoucas, D.; Qiu, X.; Lim, K.; Rao, P.; et al. A major chromatin regulator determines resistance of tumor cells to T cell-mediated killing. Science 2018, 359, 770–775. [Google Scholar] [CrossRef] [Green Version]
- Shen, J.; Ju, Z.; Zhao, W.; Wang, L.; Peng, Y.; Ge, Z.; Nagel, Z.D.; Zou, J.; Wang, C.; Kapoor, P.; et al. ARID1A deficiency promotes mutability and potentiates therapeutic antitumor immunity unleashed by immune checkpoint blockade. Nat. Med. 2018, 24, 556–562. [Google Scholar] [CrossRef]
- Jelinic, P.; Ricca, J.; Van Oudenhove, E.; Olvera, N.; Merghoub, T.; Levine, D.A.; Zamarin, D. Immune-Active Microenvironment in Small Cell Carcinoma of the Ovary, Hypercalcemic Type: Rationale for Immune Checkpoint Blockade. J. Natl. Cancer Inst. 2018, 110, 787–790. [Google Scholar] [CrossRef] [Green Version]
- Shukla, S.A.; Bachireddy, P.; Schilling, B.; Galonska, C.; Zhan, Q.; Bango, C.; Langer, R.; Lee, P.C.; Gusenleitner, D.; Keskin, D.B.; et al. Cancer-Germline Antigen Expression Discriminates Clinical Outcome to CTLA-4 Blockade. Cell 2018, 173, 624–633.e8. [Google Scholar] [CrossRef] [PubMed]
- Rizvi, H.; Sanchez-Vega, F.; La, K.; Chatila, W.; Jonsson, P.; Halpenny, D.; Plodkowski, A.; Long, N.; Sauter, J.L.; Rekhtman, N.; et al. Molecular Determinants of Response to Anti-Programmed Cell Death (PD)-1 and Anti-Programmed Death-Ligand 1 (PD-L1) Blockade in Patients With Non-Small-Cell Lung Cancer Profiled With Targeted Next-Generation Sequencing. J. Clin. Oncol. 2018, 36, 633–641. [Google Scholar] [CrossRef] [PubMed]
- Koyama, S.; Akbay, E.A.; Li, Y.Y.; Aref, A.R.; Skoulidis, F.; Herter-Sprie, G.S.; Buczkowski, K.A.; Liu, Y.; Awad, M.M.; Denning, W.L.; et al. STK11/LKB1 Deficiency Promotes Neutrophil Recruitment and Proinflammatory Cytokine Production to Suppress T-cell Activity in the Lung Tumor Microenvironment. Cancer Res. 2016, 76, 999–1008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peng, W.; Chen, J.Q.; Liu, C.; Malu, S.; Creasy, C.; Tetzlaff, M.T.; Xu, C.; McKenzie, J.A.; Zhang, C.; Liang, X.; et al. Loss of PTEN Promotes Resistance to T Cell-Mediated Immunotherapy. Cancer Discov. 2016, 6, 202–216. [Google Scholar] [CrossRef] [Green Version]
- George, S.; Miao, D.; Demetri, G.D.; Adeegbe, D.; Rodig, S.J.; Shukla, S.; Lipschitz, M.; Amin-Mansour, A.; Raut, C.P.; Carter, S.L.; et al. Loss of PTEN Is Associated with Resistance to Anti-PD-1 Checkpoint Blockade Therapy in Metastatic Uterine Leiomyosarcoma. Immunity 2017, 46, 197–204. [Google Scholar] [CrossRef] [Green Version]
- Spranger, S.; Dai, D.; Horton, B.; Gajewski, T.F. Tumor-Residing Batf3 Dendritic Cells Are Required for Effector T Cell Trafficking and Adoptive T Cell Therapy. Cancer Cell 2017, 31, 711–723.e4. [Google Scholar] [CrossRef] [Green Version]
- Gainor, J.F.; Shaw, A.T.; Sequist, L.V.; Fu, X.; Azzoli, C.G.; Piotrowska, Z.; Huynh, T.G.; Zhao, L.; Fulton, L.; Schultz, K.R.; et al. EGFR Mutations and ALK Rearrangements Are Associated with Low Response Rates to PD-1 Pathway Blockade in Non-Small Cell Lung Cancer: A Retrospective Analysis. Clin. Cancer Res. 2016, 22, 4585–4593. [Google Scholar] [CrossRef] [Green Version]
- Lee, C.K.; Man, J.; Lord, S.; Cooper, W.; Links, M.; Gebski, V.; Herbst, R.S.; Gralla, R.J.; Mok, T.; Yang, J.C. Clinical and Molecular Characteristics Associated With Survival Among Patients Treated With Checkpoint Inhibitors for Advanced Non-Small Cell Lung Carcinoma: A Systematic Review and Meta-analysis. JAMA Oncol. 2018, 4, 210–216. [Google Scholar] [CrossRef]
- Dong, Z.Y.; Zhong, W.Z.; Zhang, X.C.; Su, J.; Xie, Z.; Liu, S.Y.; Tu, H.Y.; Chen, H.J.; Sun, Y.L.; Zhou, Q.; et al. Potential Predictive Value of TP53 and KRAS Mutation Status for Response to PD-1 Blockade Immunotherapy in Lung Adenocarcinoma. Clin. Cancer Res. 2017, 23, 3012–3024. [Google Scholar] [CrossRef] [Green Version]
- Mariathasan, S.; Turley, S.J.; Nickles, D.; Castiglioni, A.; Yuen, K.; Wang, Y.; Kadel, E.E., III; Koeppen, H.; Astarita, J.L.; Cubas, R.; et al. TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells. Nature 2018, 554, 544–548. [Google Scholar] [CrossRef]
- Chakravarthy, A.; Khan, L.; Bensler, N.P.; Bose, P.; De Carvalho, D.D. TGF-β-associated extracellular matrix genes link cancer-associated fibroblasts to immune evasion and immunotherapy failure. Nat. Commun. 2018, 9, 4692. [Google Scholar] [CrossRef] [PubMed]
- O’Donnell, J.S.; Long, G.V.; Scolyer, R.A.; Teng, M.W.; Smyth, M.J. Resistance to PD1/PDL1 checkpoint inhibition. Cancer Treat. Rev. 2017, 52, 71–81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jenkins, R.W.; Barbie, D.A.; Flaherty, K.T. Mechanisms of resistance to immune checkpoint inhibitors. Br. J. Cancer 2018, 118, 9–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cui, Y.; Guo, G. Immunomodulatory Function of the Tumor Suppressor p53 in Host Immune Response and the Tumor Microenvironment. Int. J. Mol. Sci. 2016, 17, 1942. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.; Veeramachaneni, N. Targeting interleukin-1beta and inflammation in lung cancer. Biomark Res. 2022, 10, 5. [Google Scholar] [CrossRef]
- Zippelius, A.; Batard, P.; Rubio-Godoy, V.; Bioley, G.; Lienard, D.; Lejeune, F.; Rimoldi, D.; Guillaume, P.; Meidenbauer, N.; Mackensen, A.; et al. Effector function of human tumor-specific CD8 T cells in melanoma lesions: A state of local functional tolerance. Cancer Res. 2004, 64, 2865–2873. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Chin, E.N.; Yu, C.; Vartabedian, V.F.; Jia, Y.; Kumar, M.; Gamo, A.M.; Vernier, W.; Ali, S.H.; Kissai, M.; Lazar, D.C.; et al. Antitumor activity of a systemic STING-activating non-nucleotide cGAMP mimetic. Science 2020, 369, 993–999. [Google Scholar] [CrossRef]
- Twumasi-Boateng, K.; Pettigrew, J.L.; Kwok, Y.Y.E.; Bell, J.C.; Nelson, B.H. Oncolytic viruses as engineering platforms for combination immunotherapy. Nat. Rev. Cancer 2018, 18, 419–432. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef]
- Barker, H.E.; Paget, J.T.; Khan, A.A.; Harrington, K.J. The tumour microenvironment after radiotherapy: Mechanisms of resistance and recurrence. Nat. Rev. Cancer 2015, 15, 409–425. [Google Scholar] [CrossRef] [PubMed]
- Yamazaki, T.; Buqué, A.; Ames, T.D.; Galluzzi, L. PT-112 induces immunogenic cell death and synergizes with immune checkpoint blockers in mouse tumor models. Oncoimmunology 2020, 9, 1721810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maatouk, D.M.; Kellam, L.D.; Mann, M.R.; Lei, H.; Li, E.; Bartolomei, M.S.; Resnick, J.L. DNA methylation is a primary mechanism for silencing postmigratory primordial germ cell genes in both germ cell and somatic cell lineages. Development 2006, 133, 3411–3418. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ritter, C.; Fan, K.; Paschen, A.; Reker Hardrup, S.; Ferrone, S.; Nghiem, P.; Ugurel, S.; Schrama, D.; Becker, J.C. Epigenetic priming restores the HLA class-I antigen processing machinery expression in Merkel cell carcinoma. Sci. Rep. 2017, 7, 2290. [Google Scholar] [CrossRef] [Green Version]
- Luo, N.; Nixon, M.J.; Gonzalez-Ericsson, P.I.; Sanchez, V.; Opalenik, S.R.; Li, H.; Zahnow, C.A.; Nickels, M.L.; Liu, F.; Tantawy, M.N.; et al. DNA methyltransferase inhibition upregulates MHC-I to potentiate cytotoxic T lymphocyte responses in breast cancer. Nat. Commun. 2018, 9, 248. [Google Scholar] [CrossRef] [Green Version]
- Seth, P.; Csizmadia, E.; Hedblom, A.; Vuerich, M.; Xie, H.; Li, M.; Longhi, M.S.; Wegiel, B. Deletion of Lactate Dehydrogenase-A in Myeloid Cells Triggers Antitumor Immunity. Cancer Res. 2017, 77, 3632–3643. [Google Scholar] [CrossRef] [Green Version]
- Pilon-Thomas, S.; Kodumudi, K.N.; El-Kenawi, A.E.; Russell, S.; Weber, A.M.; Luddy, K.; Damaghi, M.; Wojtkowiak, J.W.; Mulé, J.J.; Ibrahim-Hashim, A.; et al. Neutralization of Tumor Acidity Improves Antitumor Responses to Immunotherapy. Cancer Res. 2016, 76, 1381–1390. [Google Scholar] [CrossRef] [Green Version]
- Qi, X.; Yang, M.; Ma, L.; Sauer, M.; Avella, D.; Kaifi, J.T.; Bryan, J.; Cheng, K.; Staveley-O’Carroll, K.F.; Kimchi, E.T.; et al. Synergizing sunitinib and radiofrequency ablation to treat hepatocellular cancer by triggering the antitumor immune response. J. Immunother. Cancer 2020, 8, e001038. [Google Scholar] [CrossRef]
- Chen, Q.; He, Y.; Wang, Y.; Li, C.; Zhang, Y.; Guo, Q.; Zhang, Y.; Chu, Y.; Liu, P.; Chen, H.; et al. Penetrable Nanoplatform for "Cold" Tumor Immune Microenvironment Reeducation. Adv. Sci. (Weinh) 2020, 7, 2000411. [Google Scholar] [CrossRef]
- Sun, Y.; Zhang, Y.; Gao, Y.; Wang, P.; He, G.; Blum, N.T.; Lin, J.; Liu, Q.; Wang, X.; Huang, P. Six Birds with One Stone: Versatile Nanoporphyrin for Single-Laser-Triggered Synergistic Phototheranostics and Robust Immune Activation. Adv. Mater. 2020, 32, e2004481. [Google Scholar] [CrossRef]
- Zanganeh, S.; Hutter, G.; Spitler, R.; Lenkov, O.; Mahmoudi, M.; Shaw, A.; Pajarinen, J.S.; Nejadnik, H.; Goodman, S.; Moseley, M.; et al. Iron oxide nanoparticles inhibit tumour growth by inducing pro-inflammatory macrophage polarization in tumour tissues. Nat. Nanotechnol. 2016, 11, 986–994. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Zheng, J.; Sun, W.; Zhao, X.; Li, Y.; Gong, N.; Wang, Y.; Ma, X.; Zhang, T.; Zhao, L.Y.; et al. Ferrimagnetic Vortex Nanoring-Mediated Mild Magnetic Hyperthermia Imparts Potent Immunological Effect for Treating Cancer Metastasis. ACS Nano 2019, 13, 8811–8825. [Google Scholar] [CrossRef] [PubMed]
- Chavez, M.; Silvestrini, M.T.; Ingham, E.S.; Fite, B.Z.; Mahakian, L.M.; Tam, S.M.; Ilovitsh, A.; Monjazeb, A.M.; Murphy, W.J.; Hubbard, N.E.; et al. Distinct immune signatures in directly treated and distant tumors result from TLR adjuvants and focal ablation. Theranostics 2018, 8, 3611–3628. [Google Scholar] [CrossRef] [PubMed]
- Ilovitsh, T.; Feng, Y.; Foiret, J.; Kheirolomoom, A.; Zhang, H.; Ingham, E.S.; Ilovitsh, A.; Tumbale, S.K.; Fite, B.Z.; Wu, B.; et al. Low-frequency ultrasound-mediated cytokine transfection enhances T cell recruitment at local and distant tumor sites. Proc. Natl. Acad. Sci. USA 2020, 117, 12674–12685. [Google Scholar] [CrossRef]
- Lai, J.; Mardiana, S.; House, I.G.; Sek, K.; Henderson, M.A.; Giuffrida, L.; Chen, A.X.Y.; Todd, K.L.; Petley, E.V.; Chan, J.D.; et al. Adoptive cellular therapy with T cells expressing the dendritic cell growth factor Flt3L drives epitope spreading and antitumor immunity. Nat. Immunol. 2020, 21, 914–926. [Google Scholar] [CrossRef]
- Rosenberg, S.A.; Yang, J.C.; Sherry, R.M.; Kammula, U.S.; Hughes, M.S.; Phan, G.Q.; Citrin, D.E.; Restifo, N.P.; Robbins, P.F.; Wunderlich, J.R.; et al. Durable complete responses in heavily pretreated patients with metastatic melanoma using T-cell transfer immunotherapy. Clin. Cancer Res. 2011, 17, 4550–4557. [Google Scholar] [CrossRef] [Green Version]
- Adachi, K.; Kano, Y.; Nagai, T.; Okuyama, N.; Sakoda, Y.; Tamada, K. IL-7 and CCL19 expression in CAR-T cells improves immune cell infiltration and CAR-T cell survival in the tumor. Nat. Biotechnol. 2018, 36, 346–351. [Google Scholar] [CrossRef]
- Ott, P.A.; Hu-Lieskovan, S.; Chmielowski, B.; Govindan, R.; Naing, A.; Bhardwaj, N.; Margolin, K.; Awad, M.M.; Hellmann, M.D.; Lin, J.J.; et al. A Phase Ib Trial of Personalized Neoantigen Therapy Plus Anti-PD-1 in Patients with Advanced Melanoma, Non-small Cell Lung Cancer, or Bladder Cancer. Cell 2020, 183, 347–362.e24. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Sumimoto, H.; Imabayashi, F.; Iwata, T.; Kawakami, Y. The BRAF-MAPK signaling pathway is essential for cancer-immune evasion in human melanoma cells. J. Exp. Med. 2006, 203, 1651–1656. [Google Scholar] [CrossRef] [Green Version]
- Peng, D.; Kryczek, I.; Nagarsheth, N.; Zhao, L.; Wei, S.; Wang, W.; Sun, Y.; Zhao, E.; Vatan, L.; Szeliga, W.; et al. Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature 2015, 527, 249–253. [Google Scholar] [CrossRef] [PubMed]
- Nagarsheth, N.; Peng, D.; Kryczek, I.; Wu, K.; Li, W.; Zhao, E.; Zhao, L.; Wei, S.; Frankel, T.; Vatan, L.; et al. PRC2 Epigenetically Silences Th1-Type Chemokines to Suppress Effector T-Cell Trafficking in Colon Cancer. Cancer Res. 2016, 76, 275–282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Topper, M.J.; Vaz, M.; Chiappinelli, K.B.; DeStefano Shields, C.E.; Niknafs, N.; Yen, R.C.; Wenzel, A.; Hicks, J.; Ballew, M.; Stone, M.; et al. Epigenetic Therapy Ties MYC Depletion to Reversing Immune Evasion and Treating Lung Cancer. Cell 2017, 171, 1284–1300.e21. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tauriello, D.V.F.; Palomo-Ponce, S.; Stork, D.; Berenguer-Llergo, A.; Badia-Ramentol, J.; Iglesias, M.; Sevillano, M.; Ibiza, S.; Cañellas, A.; Hernando-Momblona, X.; et al. TGFβ drives immune evasion in genetically reconstituted colon cancer metastasis. Nature 2018, 554, 538–543. [Google Scholar] [CrossRef] [Green Version]
- Vanpouille-Box, C.; Diamond, J.M.; Pilones, K.A.; Zavadil, J.; Babb, J.S.; Formenti, S.C.; Barcellos-Hoff, M.H.; Demaria, S. TGFβ Is a Master Regulator of Radiation Therapy-Induced Antitumor Immunity. Cancer Res. 2015, 75, 2232–2242. [Google Scholar] [CrossRef] [Green Version]
- Feig, C.; Jones, J.O.; Kraman, M.; Wells, R.J.; Deonarine, A.; Chan, D.S.; Connell, C.M.; Roberts, E.W.; Zhao, Q.; Caballero, O.L.; et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc. Natl. Acad. Sci. USA 2013, 110, 20212–20217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bockorny, B.; Semenisty, V.; Macarulla, T.; Borazanci, E.; Wolpin, B.M.; Stemmer, S.M.; Golan, T.; Geva, R.; Borad, M.J.; Pedersen, K.S.; et al. BL-8040, a CXCR4 antagonist, in combination with pembrolizumab and chemotherapy for pancreatic cancer: The COMBAT trial. Nat. Med. 2020, 26, 878–885. [Google Scholar] [CrossRef]
- Kalbasi, A.; Ribas, A. Tumour-intrinsic resistance to immune checkpoint blockade. Nat. Rev. Immunol. 2020, 20, 25–39. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [Green Version]
- 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. [Google Scholar] [CrossRef]
- 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. [Google Scholar] [CrossRef] [PubMed]
- 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. [Google Scholar] [CrossRef] [Green Version]
- Spranger, S.; Gajewski, T.F. Impact of oncogenic pathways on evasion of antitumour immune responses. Nat. Rev. Cancer 2018, 18, 139–147. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Franco, F.; Tsui, Y.C.; Xie, X.; Trefny, M.P.; Zappasodi, R.; Mohmood, S.R.; Fernández-García, J.; Tsai, C.H.; Schulze, I.; et al. CD36-mediated metabolic adaptation supports regulatory T cell survival and function in tumors. Nat. Immunol. 2020, 21, 298–308. [Google Scholar] [CrossRef]
- Landry, L.G.; Ali, N.; Williams, D.R.; Rehm, H.L.; Bonham, V.L. Lack Of Diversity In Genomic Databases Is A Barrier To Translating Precision Medicine Research Into Practice. Health Aff. (Millwood) 2018, 37, 780–785. [Google Scholar] [CrossRef] [PubMed]
- Berger, T.R.; Wen, P.Y.; Lang-Orsini, M.; Chukwueke, U.N. World Health Organization 2021 Classification of Central Nervous System Tumors and Implications for Therapy for Adult-Type Gliomas: A Review. JAMA Oncol. 2022, 8, 1493–1501. [Google Scholar] [CrossRef] [PubMed]
Tumor Cells | Tumor Microenvironment | Circulating Factors | Host Factors | Immune-Related Adverse Events |
---|---|---|---|---|
PDL-1 expression | PDL-1 expression | Peripheral blood cells, e.g., myelogenous cells, eosinophils, macrophages, CD+ ICSO+ T cells | Age, gender, body fat distribution | Endocrine immune-related adverse events, e.g., thyroid dysfunction Skin immune-related adverse events e.g., vitiligo, pruritus, lichenoid toxicity |
TMB | Tumor-infiltrating lymphocytes, e.g., CD39+ CD8+ T cells. CD4+ T cells, FOXP3+ T cells, TAMs, myeloid cells, NKp46+ cells | Other circulating factors, e.g., PDL-1, soluble proteins, cytokines and inflammatory factors | Host germline mutations, HLA diversity, and other specific mutations | |
DDR pathways: dMMR/MSI | Immune status of tumor microenvironment | Circulating nucleic acids, e.g., ctDNA, RNA (mRNA, miRNA) | Intestinal commensal microbiota | |
Specific gene mutations | Immunologic classification, immunoscore | Circulating tumor cells (CTCs) | ||
Neoantigen load | Diversity of immune cell repertoire | |||
TIL richness and clonality |
Diagnostic Biomarkers | Prognostic Biomarkers | Predictive Biomarkers | On-Treatment Biomarkers | ||
---|---|---|---|---|---|
Purpose | Presence of disease | Overall patient survival independent of therapy | Response to treatment (efficacy or toxicity) | Pharmacodynamic biomarkers (drug interaction with its target) | |
Time of evaluation | Before diagnosis and at diagnosis | At diagnosis | Before treatment selection | During or post-treatment | |
Clinical utility | Before diagnosis: Allows screening of healthy patients At diagnosis: Stratifies benign vs. malignant, classify into subtypes | At diagnosis: Estimate risk of disease Post diagnosis: Allows monitoring disease status, detects recurrence | Identify treatments likely to be effective, guides initial treatment strategy and decision making | Determines degree of drug response, guides treatments decision making during treatment | |
Current state | Validated | Emerging | |||
Human Melanoma Black-45 (HMB-45) | Lactate dehydrogenase | BRAF V600 mutation | TMB | ctDNA profiles | |
Melan-A | M stage | Neoantigen load | Absolute lymphocyte count | ||
Tyrosinase | Disease sites | Molecular alterations | Proliferating CD8+ T cells | ||
Microphthalmia transcription factor (MITF) | PDL-1 expression | Increase of T-cell subsets and checkpoint molecules (PDL-1, LAG-3) | |||
S100 | LAG-3 expression | Granzyme B expression | |||
SM5-1 | CD8+ T cells at tumor invasive margin | T-cell receptor (TCR) signature | |||
Chondroitin sulfate proteoglycan 4 (CSPG4) | IL-17 expression | ||||
Loss of p16 protein expression | Immune-related gene expression signatures | ||||
Biomarker panels and gene arrays | T-cell receptor (TCR) signature |
Primary Location | Response Category | Characteristics or Modality | References |
---|---|---|---|
T cell | Intratumoral infiltration | Transcriptional signatures of cytotoxic lymphocytes infiltrating the tumor core | [67,83,141,142,143,144] |
Enhanced effector function | Increased expression of PRF1, GZMA/B, CD8A, and IFNG | [82,95,145] | |
Increased clonality | Ranging from 0 to 1, with 1 indicating a monoclonal population | [46,121,146] | |
Greater stemness | Express chemokine receptor CXCR5 and transcription factor TCF7; lack TIM-3/CD39 | [98,147] | |
Reduced exhaustion | Express co-inhibitory receptor TIM-3 and ectonucleotidase CD39; lack CXCR5/TCF7 | [98,147] | |
Tumor cell (response mechanisms) | Tumor antigens | Neoantigens, viral antigens | [7,66,69,72,131,148,149,150,151,152,153,154,155] |
Increased tumor mutation burden | Mismatch repair deficiency | [151,156,157] | |
Immunogenic alterations | Inactivating mutations in SERPINB3 and SERPINB4 | [158] | |
Mutational signatures | Smoking, ultraviolet light, alkylating agent therapy, APOBEC | [66,159,160] | |
Genomic upregulation of PD-L1 | PDL1 amplification and loss of CDK4, SPOP, and CMTM4 and CMTM6 | [130,161,162,163,164,165,166,167] | |
Chromatin modifier loss | Inactivating mutations in PBRM1, ARID1A, and SMARCA4 | [159,168,169,170] | |
Tumor cell (resistance mechanisms) | Tumor antigens | Cancer/testis antigens similar to self and less immunogenic | [171] |
Deficient antigen presentation | Inactivating mutations in B2M, HLA, JAK/STAT, and IFN-γ response genes | [151,159] | |
Oncogenic pathways | Inactivating STK11 and PTEN mutations, WNT/β-catenin, EGFR and KRAS mutations | [67,135,172,173,174,175,176,177,178,179] | |
Immune evasion alterations | Increased expression of SERPINB9 | [82] | |
CNAs | High levels of copy-number loss, chromosome arm and whole-chromosome CNAs | [46,73] | |
Tumor microenvironment | Immunosuppressive stromal cells | Transcriptional signatures of fibroblasts, endothelial cells, and TGF-β signaling | [30,67,180,181] |
Immunosuppressive immune cells | Transcriptional signatures of myeloid-derived suppressor cells and regulatory T cells | [82,142] |
Therapeutic Modalities | Primary Effect Site | Main Mechanisms | References |
---|---|---|---|
Immune adjuvants (TLR agonists, STING agonists) | Draining lymph node | Promotes T cell priming by antigen release, antigen processing and presentation, and DC-T cell interaction | [187,188] |
Oncolytic viruses | [189,190] | ||
Chemotherapy and radiotherapy | [191,192] | ||
Epigenetic modification inhibitors (DNMT inhibitor, HDAC inhibitor, EZH2 inhibitor) | [193,194,195] | ||
Metabolic intervention | [196,197] | ||
Local thermal ablation therapy (Radiofrequency ablation) | [198] | ||
Photothermal therapy and photodynamic therapy | [26,199,200] | ||
Magnetic hyperthermia | [201,202] | ||
High-intensity focused ultrasound | [203,204] | ||
Adoptive cellular therapy (TILs, CAR-T cells) | Draining lymph node | Promotes T cell expansion | [205,206,207] |
Vaccines | [208] | ||
Oncogenic pathway inhibitors | Tumor microenvironment | Promotes T cell trafficking and infiltration | [174,209,210] |
Epigenetic modification inhibitors | [211,212,213] | ||
Antiangiogenic therapy (anti-VEGF) | [102] | ||
TGFβ inhibitors | [180,214,215] | ||
CXCR4 inhibitors | [216,217] |
Title | Interventions | Trial No | Conditions | Phase | Setting | Status as of 11-2022 |
---|---|---|---|---|---|---|
Neoantigen vaccines | ||||||
A Phase I Study Combining NeoVax, a Personalized NeoAntigen Cancer Vaccine, With Ipilimumab to Treat High-risk Renal Cell Carcinoma | NeoVax + anti-CTLA-4 ipilimumab | NCT02950766 | Stage III or low-volume stage IV renal cell carcinoma | I | Adjuvant/1st line | Recruiting |
Neoantigen-based Personalized Vaccine Combined with Immune Checkpoint Blockade Therapy in Patients with Newly Diagnosed, Unmethylated Glioblastoma | NeoVax + anti-PD-1 nivolumab ± anti-CTLA-4 ipilimumab | NCT03422094 | Newly diagnosed, unmethylated glioblastoma | I | 1st line | Terminated (Manufacturer changed focus to cell therapy) |
Personalized Neoantigen Cancer Vaccine + Pembrolizumab After Rituximab for Follicular Lymphoma | NeoVax + anti-CD20 rituxumab | NCT03361852 | Follicular lymphoma | I | 1st line | Recruiting |
A Personalized Neoantigen Cancer Vaccine in Treatment Naïve, Asymptomatic Patients with IGHV Unmutated CLL | NeoVax + alkylating agent cyclophosphamide | NCT03219450 | Chronic lymphocytic leukemia | I | 1st line | Recruiting |
Pooled Mutant KRAS-Targeted Long Peptide Vaccine Combined with Nivolumab and Ipilimumab for Patients with Resected MMR-p Colorectal and Pancreatic Cancer | KRAS peptide vaccine, nivolumab, ipilimumab | NCT04117087 | Colorectal cancer, pancreatic cancer | I | Recurrent or metastatic | Recruiting |
NeoVax Plus Ipilimumab in Renal Cell Carcinoma | NeoVax, ipilimumab | NCT02950766 | Kidney cancer | I | 1st line | Recruiting |
Immunotherapy Combined with Radiation and Influenza Vaccine for Pancreatic Cancer | Nivolumab, ipilimumab, influenza vaccine, stereotactic body radiation therapy | NCT05116917 | Pancreatic cancer | II | Recurrent | Recruiting |
Combination Therapy with Nivolumab and PD-L1/IDO Peptide Vaccine to Patients with Metastatic Melanoma | Nivolumab, PD-L1/IDO peptide vaccine | NCT03047928 | Metastatic melanoma | I/II | Recurrent | Recruiting |
A Phase I/II Trial to Evaluate a Peptide Vaccine Plus Ipilimumab in Patients with Melanoma | Ipilimumab, peptide vaccine 6MHP | NCT02385669 | Melanoma | I/II | Stage III or IV, recurrent or metastatic, stage IIA, IIB-IV resected to no evidence of disease | Terminated, has Results |
Vaccine Combining Multiple Class I Peptides and Montanide ISA 51VG With Escalating Doses of Anti-PD-1 Antibody Nivolumab or Ipilimumab with Nivolumab For Patients with Resected Stages IIIC/IV Melanoma | NY-ESO-1 157-165 (165 V), nivolumab, gp100:280-288 (288 V), montanide ISA 51 vegetable grade (VG), ipilimumab | NCT01176474 | Melanoma | I | Stages IIIC/IV melanoma, with no evidence of disease | Active, not recruiting |
Dendritic cell vaccines | ||||||
Combination Immunotherapy-Ipilimumab-Nivolumab-Dendritic Cell p53 Vac - Patients with Small Cell Lung Cancer | Dendritic cell-based p53 vaccine + nivolumab + ipilimumab | NCT03406715 | Small cell lung cancer | II | Recurrent | Active, not recruiting |
Nivolumab with DC Vaccines for Recurrent Brain Tumors | Dentritic cell + nivolumab | NCT02529072 | Malignant glioma, astrocytoma, glioblastoma | I | Recurrent | Completed, has results |
Neoantigen Dendritic Cell Vaccine and Nivolumab in HCC and Liver Metastases from CRC | Neoantigen dendritic cell vaccine + nivolumab | NCT03782064 | Hepatocellular carcinoma, hepatocellular cancer, colorectal cancer, colorectal carcinoma, liver metastases | II | Newly diagnosed or recurrent hepatocellular carcinoma | Recruiting |
Dendritic Cell (DC)/Myeloma Fusions in Combination with Nivolumab in Patients with Relapsed Multiple Myeloma | NCT03782064 | Multiple myeloma | II | Relapsed | Terminated Has Results | |
Polarized Dendritic Cell (aDC1) Vaccine, Interferon Alpha-2, Rintatolimod, and Celecoxib for the Treatment of HLA-A2 + Refractory Melanoma | Alpha-type-1 polarized dendritic cells, celecoxib, PD-L1 inhibitor, PD-1 inhibitor, recombinant Interferon alpha-2b, rintatolimod | NCT04093323 | HLA-A2 positive cells present, refractory melanoma | II | Refractory | Recruiting |
Interferon therapies | ||||||
Testing the Combination of Two Experimental Drugs MK-3475 (Pembrolizumab) and Interferon-gamma for the Treatment of Mycosis Fungoides and Sézary Syndrome and Advanced Synovial Sarcoma | Interferon-γ-1β + anti-PD-1 pembrolizumab | NCT03063632 | Mycosis fungoides and Sézary syndrome | II | Refractory | Active, not recruiting |
Combination of Interferon-gamma and Nivolumab for Advanced Solid Tumors | Interferon-γ + anti-PD-1 nivolumab | NCT02614456 | Advanced solid tumors | I | 2nd line | Completed |
Pembrolizumab Combined with Itacitinib (INCB039110) and/or Pembrolizumab Combined with INCB050465 in Advanced Solid Tumors | JAK1 inhibitor itacitinib or PI3Kδ inhibitor + anti-PD-1 pembrolizumab | NCT02646748 | Advanced solid tumors | I/II | Refractory | Completed |
Pembrolizumab and Ruxolitinib Phosphate in Treating Patients with Metastatic Stage IV Triple Negative Breast Cancer | JAK2 inhibitor ruxolitinib + anti-PD-1 pembrolizumab | NCT03012230 | Metastatic triple-negative breast cancer | I | 2nd line | Recruiting |
Pembrolizumab and Itacitinib (INCB039110) for Non-Small Cell Lung Cancer | JAK1 inhibitor itacitinib + anti-PD-1 pembrolizumab | NCT03425006 | Metastatic PD-L1+ non-small cell lung cancer | II | 1st line | Terminated |
Study of the Safety and Efficacy of MIW815 with PDR001 in Patients with Advanced/Metastatic Solid Tumors or Lymphomas | STING agonist MIW815 + anti-PD-1 spartalizumab | NCT03172936 | Advanced solid tumors or lymphomas | I | Any line | Terminated (Sponsor’s decision) |
Trial of Intratumoral Injections of TTI-621 in Subjects with Relapsed and Refractory Solid Tumors and Mycosis Fungoides | TTI-621 monotherapy, TTI-621 + PD-1/PD-L1 inhibitor, TTI-621 + pegylated interferon-α2aOther: TTI-621 + T-Vec, TTI-621 + radiation TTI-621 (SIRPα-IgG1 Fc) anti- CD47 “don’t eat me” signal | NCT02890368 | Solid tumors, mycosis fungoides, melanoma, merkel-cell carcinoma, squamous cell carcinoma, breast carcinoma, human papillomavirus-related malignant neoplasm, soft tissue sarcoma | I | Relapsed and refractory | Terminated |
A Study to Assess the Safety and Tolerability of Atezolizumab in Combination with Other Immune-Modulating Therapies in Participants with Locally Advanced or Metastatic Solid Tumors | Atezolizumab, bevacizumab, interferon alpha-2b, ipilimumab, obinutuzumab, PEG-interferon alpha-2a | NCT02174172 | Solid tumors | I | Locally advanced or metastatic solid tumors | Completed |
The Study of JS001 Compared to High-Dose Interferon in Patients with Mucosal Melanoma That Has Been Removed by Surgery | Humanized anti-PD-1 monoclonal antibody toripalimab, high-dose recombinant interferon a-2B | NCT03178123 | Mucosal melanoma | II | 1st line after surgery | Active, not recruiting |
Ipilimumab or High-Dose Interferon Alfa-2b in Treating Patients with High-Risk Stage III–IV Melanoma That Has Been Removed by Surgery | Ipilimumab, interferon alpha-2b | NCT01274338 | Melanoma of unknown primary, recurrent melanoma, stage IIIB cutaneous melanoma AJCC v7, stage IIIC cutaneous melanoma AJCC v7, stage IV cutaneous melanoma AJCC v6 and v7 | III | Recurrence after surgery | Active, not recruiting, has results |
Ipilimumab With or Without High-Dose Recombinant Interferon Alfa-2b in Treating Patients with Stage III–IV Melanoma That Cannot Be Removed by Surgery | Ipilimumab, recombinant interferon alpha-2b | NCT01708941 | Recurrent melanoma, stage IIIA cutaneous melanoma AJCC v7, stage IIIB cutaneous melanoma AJCC v7, stage IIIC cutaneous melanoma AJCC v7, stage IV cutaneous melanoma AJCC v6 and v7 | II | Unresectable stage III or stage IV melanoma, either initial presentation or recurrent | Active, not recruiting, has results |
Nivolumab, Fluorouracil, and Interferon Alpha 2B for the Treatment of Unresectable Fibrolamellar Cancer | Fluorouracil, nivolumab, recombinant interferon alpha 2b-like protein | NCT04380545 | Stage III IVB epatocellular carcinoma AJCC v8, unresectable fibrolamellar carcinoma | I/II | 1st line | Recruiting |
High-Dose Recombinant Interferon Alfa-2B, Ipilimumab, or Pembrolizumab in Treating Patients with Stage III–IV High Risk Melanoma That Has Been Removed by Surgery | Ipilimumab, pembrolizumab, recombinant interferon alpha-2b | NCT02506153 | Cutaneous melanoma, metastatic mucosal melanoma, metastatic non-cutaneous melanoma, non-cutaneous melanoma, recurrent cutaneous melanoma, recurrent mucosal melanoma, recurrent non-cutaneous melanoma, and stage III-IVC of all above | III | 1st line, high risk melanoma that has been removed by surgery | Active, not recruiting |
Safety and Tolerability of Pembrolizumab (MK-3475) + Pegylated Interferon Alfa-2b and Pembrolizumab+ Ipilimumab in Participants with Advanced Melanoma or Renal Cell Carcinoma (MK-3475-029/KEYNOTE-29) | Pembrolizumab, PegIFN-2b, Ipilimumab | NCT02089685 | Renal cell carcinoma, melanoma | I/II | Advanced or unresectable or metastatic renal cell carcinoma, melanoma | Completed Has Results |
IPI-Biotherapy for Patients Previously Treated with Cytotoxic Drugs with Metastatic Melanoma | Ipilimumab, interferon, interleukin-2 (aldesleukin) | NCT01409187 | Melanoma | I/II | Recurrent | Withdrawn |
TIL and Anti-PD1 in Metastatic Melanoma | Nivolumab & tumor- infiltrating lymphocytes with/without interferon-alpha | NCT03638375 | Melanoma | I/II | Recurrent | Recruiting |
Trial of Intratumoral Injections of TTI-621 in Subjects with Relapsed and Refractory Solid Tumors and Mycosis Fungoides | TTI-621 (SIRPα-IgG1 Fc) monotherapy TTI-621 + PD-1/PD-L1 inhibitor, TTI-621 + pegylated interferon-α2a, TTI-621 + T-Vec, TTI-621 + radiation | NCT02890368 | Solid tumors, mycosis fungoides Melanoma, merkel-cell carcinoma, squamous cell carcinoma, breast carcinoma, human papillomavirus-related malignant neoplasm, soft tissue sarcoma | I | Recurrent | Terminated |
CDK4/6 Inhibitors | ||||||
Palbociclib After CDK and Endocrine Therapy (PACE) | Fulvestrant ± CDK4/6 inhibitor palbociclib ± anti-PD-L1 avelumab | NCT03147287 | Metastatic HR+HER2− breast cancer | II | 2nd–3rd line | Active, not recruiting |
Neoadjuvant Endocrine Therapy, Palbociclib, Avelumab in Estrogen Receptor Positive Breast Cancer (ImmunoADAPT) | Tamoxifen ± CDK4/6 inhibitor palbociclib followed by anti-PD-L1 avelumab | NCT03573648 | Stage II or III HR+ breast cancer | II | Neoadjuvant | Recruiting |
Ribociclib + PDR001 in Breast Cancer and Ovarian Cancer | CDK4/6 inhibitor ribociclib + anti-PD-1 spartalizumab ( + fulvestrant if breast) | NCT03294694 | Metastatic ovarian cancer or HR+HER2− breast cancer | I | Any line | Terminated (safety implications) |
A Study of Abemaciclib (LY2835219) in Participants with Non-Small Cell Lung Cancer or Breast Cancer | CDK4/6 inhibitor abemaciclib + anti-PD-1 pembrolizumab | NCT02779751 | Metastatic non-small cell lung cancer or HR+HER2− breast cancer | I | 1st–3rd line | Active, not recruiting |
Avelumab, Cetuximab, and Palbociclib in Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma | Anti-PD-L1 avelumab, cetuximab, + CDK4/6 inhibitor palbociclib | NCT03498378 | Recurrent/metastatic head and neck squamous cell carcinoma | I | Any line | Active, not recruiting |
Carboplatin, Etoposide, and Atezolizumab with or Without Trilaciclib (G1T28), a CDK4/6 Inhibitor, in Extensive-Stage SCLC | Anti-PDL-1 atezolizumab, trilaciclib, carboplatin, etoposide, placebo | NCT03041311 | Small cell lung cancer | II | 1st line | Terminated, has results |
Avelumab or Hydroxychloroquine with or Without Palbociclib to Eliminate Dormant Breast Cancer | Anti-PDL-1 avelumab, HCQ, palbociclib | NCT04841148 | Breast cancer | II | Refractory | Recruiting |
Phase Ib Study of TNO155 in Combination with Spartalizumab or Ribociclib in Selected Malignancies | Anti-PD-1 spartalizumab, ribociclib, TNO155 | NCT04000529 | Non-small cell lung cancer, head and neck squamous cell carcinoma, esophageal squamous cell carcinoma, gastrointestinal stromal tumors, colorectal cancer | I | Refractory | Recruiting |
Ribociclib and Spartalizumab in R/M HNSCC | Anti-PD-1 spartalizumab, ribociclib, | NCT04213404 | Head and neck squamous cell carcinoma | I | Recurrent | Active, not recruiting |
Anti-TGF-β therapies | ||||||
Phase I/Ib Study of NIS793 in Combination with PDR001 in Patients with Advanced Malignancies | Anti-TGF-β NIS793 + anti-PD-1 spartalizumab | NCT02947165 | Advanced solid tumors | I | Refractory | Completed |
A Study of Galunisertib (LY2157299) and Durvalumab (MEDI4736) in Participants with Metastatic Pancreatic Cancer | TGF-β receptor 1 inhibitor galunisertib + anti-PD-L1 durvalumab | NCT02734160 | Metastatic pancreatic cancer | I | 3rd line | Completed |
A Study of Galunisertib (LY2157299) in Combination with Nivolumab in Advanced Refractory Solid Tumors and in Recurrent or Refractory NSCLC, or Hepatocellular Carcinoma | TGF-β receptor 1 inhibitor galunisertib + anti-PD-1 nivolumab | NCT02423343 | Recurrent or refractory non-small cell lung cancer or hepatocellular carcinoma | I/II | 2nd line | Completed |
MSB0011359C (M7824) in Metastatic or Locally Advanced Solid Tumors | Anti-PD-L1/TGF-β trap M7824 | NCT02517398 | Metastatic or locally advanced solid tumors | I | Refractory | Completed |
M7824 in Treating Patients with Stage II–III HER2 Positive Breast Cancer | Anti-PD-L1/TGF-β trap M7824 + treatment of physician’s choice | NCT03620201 | Stage II–III HER2+ breast cancer | I | Neoadjuvant | Active, not recruiting |
M7824 vs. Pembrolizumab as a First line (1L) Treatment in Participants with Programmed Death-ligand 1 (PD-L1) Expressing Advanced Non-small Cell Lung Cancer (NSCLC) | Anti-PD-L1/TGF-β trap M7824 vs. anti-PD-1 pembrolizumab | NCT03631706 | PD-L1+ non-small cell lung cancer | II | 1st line | Active, not recruiting |
Antitumor Activity of Vactosertib in Combination with Pembrolizumab in Acral and Mucosal Melanoma Patients Progressed from Prior Immune Check Point Inhibitor | Anti-PD-1 pembrolizumab + anti-TGF-βR1 vactosertib | NCT05436990 | Acral melanoma, mucosal melanoma | II | Relapsed | Not yet recruiting |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Seyhan, A.A.; Carini, C. Insights and Strategies of Melanoma Immunotherapy: Predictive Biomarkers of Response and Resistance and Strategies to Improve Response Rates. Int. J. Mol. Sci. 2023, 24, 41. https://doi.org/10.3390/ijms24010041
Seyhan AA, Carini C. Insights and Strategies of Melanoma Immunotherapy: Predictive Biomarkers of Response and Resistance and Strategies to Improve Response Rates. International Journal of Molecular Sciences. 2023; 24(1):41. https://doi.org/10.3390/ijms24010041
Chicago/Turabian StyleSeyhan, Attila A., and Claudio Carini. 2023. "Insights and Strategies of Melanoma Immunotherapy: Predictive Biomarkers of Response and Resistance and Strategies to Improve Response Rates" International Journal of Molecular Sciences 24, no. 1: 41. https://doi.org/10.3390/ijms24010041
APA StyleSeyhan, A. A., & Carini, C. (2023). Insights and Strategies of Melanoma Immunotherapy: Predictive Biomarkers of Response and Resistance and Strategies to Improve Response Rates. International Journal of Molecular Sciences, 24(1), 41. https://doi.org/10.3390/ijms24010041