Targeting Genome Stability in Melanoma—A New Approach to an Old Field
Abstract
:1. Introduction
2. Conventional Chemotherapy and Radiotherapy
3. Mitogen-Activated Protein Kinase Inhibitors
4. Immunotherapy
5. Conclusions and Future Directions
6. Methodology
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Roos, W.P.; Thomas, A.D.; Kaina, B. DNA damage and the balance between survival and death in cancer biology. Nat. Rev. Cancer 2016, 16, 20–33. [Google Scholar] [CrossRef] [PubMed]
- Chatterjee, N.; Walker, G.C. Mechanisms of DNA damage, repair, and mutagenesis. Environ. Mol. Mutagen. 2017, 58, 235–263. [Google Scholar] [CrossRef] [Green Version]
- Yi, C.; He, C. DNA repair by reversal of DNA damage. Cold Spring Harb. Perspect. Biol. 2013, 5, a012575. [Google Scholar] [CrossRef] [PubMed]
- Li, G.M. Mechanisms and functions of DNA mismatch repair. Cell Res. 2008, 18, 85–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Robertson, A.B.; Klungland, A.; Rognes, T.; Leiros, I. DNA repair in mammalian cells: Base excision repair: The long and short of it. Cell. Mol. Life Sci. 2009, 66, 981–993. [Google Scholar] [CrossRef]
- Marteijn, J.A.; Lans, H.; Vermeulen, W.; Hoeijmakers, J.H. Understanding nucleotide excision repair and its roles in cancer and ageing. Nat. Rev. Mol. Cell. Biol. 2014, 15, 465–481. [Google Scholar] [CrossRef] [PubMed]
- Wright, W.D.; Shah, S.S.; Heyer, W.D. Homologous recombination and the repair of DNA double-strand breaks. J. Biol. Chem. 2018, 293, 10524–10535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Davis, A.J.; Chen, D.J. DNA double strand break repair via non-homologous end-joining. Transl. Cancer Res. 2013, 2, 130–143. [Google Scholar] [CrossRef]
- Walden, H.; Deans, A.J. The Fanconi anemia DNA repair pathway: Structural and functional insights into a complex disorder. Annu. Rev. Biophys. 2014, 43, 257–278. [Google Scholar] [CrossRef]
- Friedberg, E.C. DNA damage and repair. Nature 2003, 421, 436–440. [Google Scholar] [CrossRef]
- McKinnon, P.J. DNA repair deficiency and neurological disease. Nat. Rev. Neurosci. 2009, 10, 100–112. [Google Scholar] [CrossRef] [Green Version]
- Tiwari, V.; Wilson, D.M., 3rd. DNA Damage and Associated DNA Repair Defects in Disease and Premature Aging. Am. J. Hum. Genet. 2019, 105, 237–257. [Google Scholar] [CrossRef] [Green Version]
- Dimitriou, F.; Krattinger, R.; Ramelyte, E.; Barysch, M.J.; Micaletto, S.; Dummer, R.; Goldinger, S.M. The World of Melanoma: Epidemiologic, Genetic, and Anatomic Differences of Melanoma across the Globe. Curr. Oncol. Rep. 2018, 20, 87. [Google Scholar] [CrossRef]
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef]
- American Cancer Society. Available online: https://www.cancer.org/cancer/melanoma-skin-cancer.html (accessed on 10 February 2021).
- Cancer Genome Atlas Network. Genomic Classification of Cutaneous Melanoma. Cell 2015, 161, 1681–1696. [Google Scholar] [CrossRef] [Green Version]
- Birkeland, E.; Zhang, S.; Poduval, D.; Geisler, J.; Nakken, S.; Vodak, D.; Meza-Zepeda, L.A.; Hovig, E.; Myklebost, O.; Knappskog, S.; et al. Patterns of genomic evolution in advanced melanoma. Nat. Commun. 2018, 10, 2665. [Google Scholar] [CrossRef] [PubMed]
- Fidler, I.J.; Kripke, M.L. The challenge of targeting metastasis. Cancer Metastasis Rev. 2015, 34, 635–641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bakhoum, S.F.; Ngo, B.; Laughney, A.M.; Cavallo, J.A.; Murphy, C.J.; Ly, P.; Shah, P.; Sriram, R.K.; Watkins, T.B.K.; Taunk, N.K.; et al. Chromosomal instability drives metastasis through a cytosolic DNA response. Nature 2018, 553, 467–472. [Google Scholar] [CrossRef] [Green Version]
- Campbell, P.J.; Yachida, S.; Mudie, L.J.; Stephens, P.J.; Pleasance, E.D.; Stebbings, L.A.; Morsberger, L.A.; Latimer, C.; McLaren, S.; Lin, M.L.; et al. The patterns and dynamics of genomic instability in metastatic pancreatic cancer. Nature 2010, 467, 1109–1113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yachida, S.; Jones, S.; Bozic, I.; Antal, T.; Leary, R.; Fu, B.; Kamiyama, M.; Hruban, R.H.; Eshleman, J.R.; Nowak, M.A.; et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature 2010, 467, 1114–1117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bertucci, F.; Ng, C.K.Y.; Patsouris, A.; Droin, N.; Piscuoglio, S.; Carbuccia, N.; Soria, J.C.; Dien, A.T.; Adnani, Y.; Kamal, M.; et al. Genomic characterization of metastatic breast cancers. Nature 2019, 569, 560–564. [Google Scholar] [CrossRef]
- Yates, L.R.; Knappskog, S.; Wedge, D.; Farmery, J.H.R.; Gonzalez, S.; Martincorena, I.; Alexandrov, L.B.; Van Loo, P.; Haugland, H.K.; Lilleng, P.K.; et al. Genomic Evolution of Breast Cancer Metastasis and Relapse. Cancer Cell. 2017, 32, 169–184.e7. [Google Scholar] [CrossRef] [Green Version]
- Jamal-Hanjani, M.; Wilson, G.A.; McGranahan, N.; Birkbak, N.J.; Watkins, T.B.K.; Veeriah, S.; Shafi, S.; Johnson, D.H.; Mitter, R.; Rosenthal, R.; et al. Tracking the Evolution of Non-Small-Cell Lung Cancer. N. Engl. J. Med. 2017, 376, 2109–2121. [Google Scholar] [CrossRef] [Green Version]
- Turajlic, S.; Xu, H.; Litchfield, K.; Rowan, A.; Chambers, T.; Lopez, J.I.; Nicol, D.; O’Brien, T.; Larkin, J.; Horswell, S.; et al. Tracking Cancer Evolution Reveals Constrained Routes to Metastases: TRACERx Renal. Cell 2018, 173, 581–594.e12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Casuscelli, J.; Weinhold, N.; Gundem, G.; Wang, L.; Zabor, E.C.; Drill, E.; Wang, P.I.; Nanjangud, G.J.; Redzematovic, A.; Nargund, A.M.; et al. Genomic landscape and evolution of metastatic chromophobe renal cell carcinoma. JCI Insight. 2017, 2, e92688. [Google Scholar] [CrossRef] [PubMed]
- Angus, L.; Smid, M.; Wilting, S.M.; van Riet, J.; Van Hoeck, A.; Nguyen, L.; Nik-Zainal, S.; Steenbruggen, T.G.; Tjan-Heijnen, V.C.G.; Labots, M.; et al. The genomic landscape of metastatic breast cancer highlights changes in mutation and signature frequencies. Nat. Genet. 2019, 51, 1450–1458. [Google Scholar] [CrossRef]
- Mei, P.; Freitag, C.E.; Wei, L.; Zhang, Y.; Parwani, A.V.; Li, Z. High tumor mutation burden is associated with DNA damage repair gene mutation in breast carcinomas. Diagn. Pathol. 2020, 15, 50. [Google Scholar] [CrossRef]
- Bashashati, A.; Ha, G.; Tone, A.; Ding, J.; Prentice, L.M.; Roth, A.; Rosner, J.; Shumansky, K.; Kalloger, S.; Senz, J.; et al. Distinct evolutionary trajectories of primary high-grade serous ovarian cancers revealed through spatial mutational profiling. J. Pathol. 2013, 231, 21–34. [Google Scholar] [CrossRef]
- Quigley, D.A.; Dang, H.X.; Zhao, S.G.; Lloyd, P.; Aggarwal, R.; Alumkal, J.J.; Foye, A.; Kothari, V.; Perry, M.D.; Bailey, A.M.; et al. Genomic Hallmarks and Structural Variation in Metastatic Prostate Cancer. Cell 2018, 174, 758–769.e9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barroso-Sousa, R.; Jain, E.; Cohen, O.; Kim, D.; Buendia-Buendia, J.; Winer, E.; Lin, N.; Tolaney, S.M.; Wagle, N. Prevalence and mutational determinants of high tumor mutation burden in breast cancer. Ann. Oncol. 2020, 31, 387–394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lawrence, M.S.; Stojanov, P.; Polak, P.; Kryukov, G.V.; Cibulskis, K.; Sivachenko, A.; Carter, S.L.; Stewart, C.; Mermel, C.H.; Roberts, S.A.; et al. Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 2013, 499, 214–218. [Google Scholar] [CrossRef] [PubMed]
- Makino, E.; Fröhlich, L.M.; Sinnberg, T.; Kosnopfel, C.; Sauer, B.; Garbe, C.; Schittek, B. Targeting Rad51 as a strategy for the treatment of melanoma cells resistant to MAPK pathway inhibition. Cell Death Dis. 2020, 11, 581. [Google Scholar] [CrossRef] [PubMed]
- Kauffmann, A.; Rosselli, F.; Lazar, V.; Winnepenninckx, V.; Mansuet-Lupo, A.; Dessen, P.; van den Oord, J.J.; Spatz, A.; Sarasin, A. High expression of DNA repair pathways is associated with metastasis in melanoma patients. Oncogene 2008, 27, 565–573. [Google Scholar] [CrossRef] [Green Version]
- 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]
- 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]
- Luke, J.J.; Flaherty, K.T.; Ribas, A.; Long, G.V. Targeted agents and immunotherapies: Optimizing outcomes in melanoma. Nat. Rev. Clin. Oncol. 2017, 14, 463–482. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Hong, A.; Moriceau, G.; Sun, L.; Lomeli, S.; Piva, M.; Damoiseaux, R.; Holmen, S.L.; Sharpless, N.E.; Hugo, W.; Lo, R.S. Exploiting Drug Addiction Mechanisms to Select against MAPKi-Resistant Melanoma. Cancer Discov. 2018, 8, 74–93. [Google Scholar] [CrossRef] [Green Version]
- Makino, E.; Gutmann, V.; Kosnopfel, C.; Niessner, H.; Forschner, A.; Garbe, C.; Sinnberg, T.; Schittek, B. Melanoma cells resistant towards MAPK inhibitors exhibit reduced TAp73 expression mediating enhanced sensitivity to platinum-based drugs. Cell Death Dis. 2018, 9, 930. [Google Scholar] [CrossRef]
- Maertens, O.; Kuzmickas, R.; Manchester, H.E.; Emerson, C.E.; Gavin, A.G.; Guild, C.J.; Wong, T.C.; De Raedt, T.; Bowman-Colin, C.; Hatchi, E.; et al. MAPK Pathway Suppression Unmasks Latent DNA Repair Defects and Confers a Chemical Synthetic Vulnerability in BRAF-, NRAS-, and NF1-Mutant Melanomas. Cancer Discov. 2019, 9, 526–545. [Google Scholar] [CrossRef] [Green Version]
- Osrodek, M.; Rozanski, M.; Czyz, M. Insulin Reduces the Efficacy of Vemurafenib and Trametinib in Melanoma Cells. Cancer Manag. Res. 2020, 12, 7231–7250. [Google Scholar] [CrossRef]
- Hiniker, S.M.; Chen, D.S.; Reddy, S.; Chang, D.T.; Jones, J.C.; Mollick, J.A.; Swetter, S.M.; Knox, S.J. A systemic complete response of metastatic melanoma to local radiation and immunotherapy. Transl. Oncol. 2012, 5, 404–407. [Google Scholar] [CrossRef] [Green Version]
- Mohiuddin, M.; Park, H.; Hallmeyer, S.; Richards, J. High-Dose Radiation as a Dramatic, Immunological Primer in Locally Advanced Melanoma. Cureus 2015, 7, e417. [Google Scholar] [CrossRef] [Green Version]
- Haymaker, C.L.; Kim, D.; Uemura, M.; Vence, L.M.; Phillip, A.; McQuail, N.; Brown, P.D.; Fernandez, I.; Hudgens, C.W.; Creasy, C.; et al. Metastatic Melanoma Patient Had a Complete Response with Clonal Expansion after Whole Brain Radiation and PD-1 Blockade. Cancer Immunol. Res. 2017, 5, 100–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hiniker, S.M.; Reddy, S.A.; Maecker, H.T.; Subrahmanyam, P.B.; Rosenberg-Hasson, Y.; Swetter, S.M.; Saha, S.; Shura, L.; Knox, S.J. A Prospective Clinical Trial Combining Radiation Therapy With Systemic Immunotherapy in Metastatic Melanoma. Int. J. Radiat. Oncol. Biol. Phys. 2016, 96, 578–588. [Google Scholar] [CrossRef] [Green Version]
- Bomar, L.; Senithilnathan, A.; Ahn, C. Systemic Therapies for Advanced Melanoma. Dermatol. Clin. 2019, 37, 409–423. [Google Scholar] [CrossRef] [PubMed]
- National Cancer Institute. Available online: https://www.cancer.gov/types/skin/patient/melanoma-treatment-pdq (accessed on 25 February 2021).
- Gogas, H.J.; Kirkwood, J.M.; Sondak, V.K. Chemotherapy for metastatic melanoma: Time for a change? Cancer 2007, 109, 455–464. [Google Scholar] [CrossRef] [PubMed]
- Wilson, M.A.; Schuchter, L.M. Chemotherapy for Melanoma. Cancer Treat. Res. 2016, 167, 209–229. [Google Scholar] [CrossRef]
- Middleton, M.R.; Grob, J.J.; Aaronson, N.; Fierlbeck, G.; Tilgen, W.; Seiter, S.; Gore, M.; Aamdal, S.; Cebon, J.; Coates, A.; et al. Randomized phase III study of temozolomide versus dacarbazine in the treatment of patients with advanced metastatic malignant melanoma. J. Clin. Oncol. 2000, 18, 158–166. [Google Scholar] [CrossRef] [PubMed]
- Kokkinakis, D.M.; Ahmed, M.M.; Chendil, D.; Moschel, R.C.; Pegg, A.E. Sensitization of pancreatic tumor xenografts to carmustine and temozolomide by inactivation of their O6-Methylguanine-DNA methyltransferase with O6-benzylguanine or O6-benzyl-2’-deoxyguanosine. Clin. Cancer Res. 2003, 9, 3801–3807. [Google Scholar] [PubMed]
- Nagane, M.; Kobayashi, K.; Ohnishi, A.; Shimizu, S.; Shiokawa, Y. Prognostic significance of O6-methylguanine-DNA methyltransferase protein expression in patients with recurrent glioblastoma treated with temozolomide. Jpn. J. Clin. Oncol. 2007, 37, 897–906. [Google Scholar] [CrossRef] [Green Version]
- Kitange, G.J.; Carlson, B.L.; Schroeder, M.A.; Grogan, P.T.; Lamont, J.D.; Decker, P.A.; Wu, W.; James, C.D.; Sarkaria, J.N. Induction of MGMT expression is associated with temozolomide resistance in glioblastoma xenografts. Neuro-Oncology 2009, 11, 281–291. [Google Scholar] [CrossRef] [Green Version]
- Ranson, M.; Hersey, P.; Thompson, D.; Beith, J.; McArthur, G.A.; Haydon, A.; Davis, I.D.; Kefford, R.F.; Mortimer, P.; Harris, P.A.; et al. Randomized trial of the combination of lomeguatrib and temozolomide compared with temozolomide alone in chemotherapy naive patients with metastatic cutaneous melanoma. J. Clin. Oncol. 2007, 25, 2540–2545. [Google Scholar] [CrossRef] [Green Version]
- Eggermont, A.M.; Kirkwood, J.M. Re-evaluating the role of dacarbazine in metastatic melanoma: What have we learned in 30 years? Eur. J. Cancer 2004, 40, 1825–1836. [Google Scholar] [CrossRef] [PubMed]
- Luke, J.J.; Schwartz, G.K. Chemotherapy in the management of advanced cutaneous malignant melanoma. Clin. Dermatol. 2013, 31, 290–297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hansson, J.; Aamdal, S.; Bastholt, L.; Brandberg, Y.; Hernberg, M.; Nilsson, B.; Stierner, U.; von der Maase, H.; Nordic Melanoma Cooperative Group. Two different durations of adjuvant therapy with intermediate-dose interferon alfa-2b in patients with high-risk melanoma (Nordic IFN trial): A randomised phase 3 trial. Lancet Oncol. 2011, 12, 144–152. [Google Scholar] [CrossRef]
- Garbe, C.; Radny, P.; Linse, R.; Dummer, R.; Gutzmer, R.; Ulrich, J.; Stadler, R.; Weichenthal, M.; Eigentler, T.; Ellwanger, U.; et al. Adjuvant low-dose interferon α2a with or without dacarbazine compared with surgery alone: A prospective-randomized phase III DeCOG trial in melanoma patients with regional lymph node metastasis. Ann. Oncol. 2008, 19, 1195–1201. [Google Scholar] [CrossRef] [PubMed]
- Cocconi, G.; Bella, M.; Calabresi, F.; Tonato, M.; Canaletti, R.; Boni, C.; Buzzi, F.; Ceci, G.; Corgna, E.; Costa, P. Treatment of metastatic malignant melanoma with dacarbazine plus tamoxifen. N. Engl. J. Med. 1992, 327, 516–523. [Google Scholar] [CrossRef]
- Falkson, C.I.; Ibrahim, J.; Kirkwood, J.M.; Coates, A.S.; Atkins, M.B.; Blum, R.H. Phase III trial of dacarbazine versus dacarbazine with interferon alpha-2b versus dacarbazine with tamoxifen versus dacarbazine with interferon alpha-2b and tamoxifen in patients with metastatic malignant melanoma: An Eastern Cooperative Oncology Group study. J. Clin. Oncol. 1998, 16, 1743–1751. [Google Scholar] [CrossRef]
- Rogers, S.J.; Puric, E.; Eberle, B.; Datta, N.R.; Bodis, S.B. Radiotherapy for Melanoma: More than DNA Damage. Dermatol. Res. Pract. 2019, 2019, 9435389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Farshad, A.; Burg, G.; Panizzon, R.; Dummer, R. A retrospective study of 150 patients with lentigo maligna and lentigo maligna melanoma and the efficacy of radiotherapy using Grenz or soft X-rays. Br. J. Dermatol. 2002, 146, 1042–1046. [Google Scholar] [CrossRef]
- Savoia, P.; Fava, P.; Casoni, F.; Cremona, O. Targeting the ERK Signaling Pathway in Melanoma. Int. J. Mol. Sci. 2019, 20, 1483. [Google Scholar] [CrossRef] [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]
- Flaherty, K.T.; Infante, J.R.; Daud, A.; Gonzalez, R.; Kefford, R.F.; Sosman, J.; Hamid, O.; Schuchter, L.; Cebon, J.; Ibrahim, N.; et al. Combined BRAF and MEK inhibition in melanoma with BRAF V600 mutations. N. Engl. J. Med. 2012, 367, 1694–1703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kozar, I.; Margue, C.; Rothengatter, S.; Haan, C.; Kreis, S. Many ways to resistance: How melanoma cells evade targeted therapies. Biochim. Biophys. Acta Rev. Cancer 2019, 1871, 313–322. [Google Scholar] [CrossRef]
- Romano, E.; Pradervand, S.; Paillusson, A.; Weber, J.; Harshman, K.; Muehlethaler, K.; Speiser, D.; Peters, S.; Rimoldi, D.; Michielin, O. Identification of multiple mechanisms of resistance to vemurafenib in a patient with BRAFV600E-mutated cutaneous melanoma successfully rechallenged after progression. Clin. Cancer Res. 2013, 19, 5749–5757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tirosh, I.; Izar, B.; Prakadan, S.M.; Wadsworth, M.H., 2nd; Treacy, D.; Trombetta, J.J.; Rotem, A.; Rodman, C.; Lian, C.; Murphy, G.; et al. Dissecting the multicellular ecosystem of metastatic melanoma by single-cell RNA-seq. Science 2016, 352, 189–196. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shannan, B.; Perego, M.; Somasundaram, R.; Herlyn, M. Heterogeneity in Melanoma. Cancer Treat. Res. 2016, 167, 1–15. [Google Scholar] [CrossRef] [PubMed]
- Ahn, A.; Chatterjee, A.; Eccles, M.R. The Slow Cycling Phenotype: A Growing Problem for Treatment Resistance in Melanoma. Mol. Cancer Ther. 2017, 16, 1002–1009. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bhatia, R.; Holtz, M.; Niu, N.; Gray, R.; Snyder, D.S.; Sawyers, C.L.; Arber, D.A.; Slovak, M.L.; Forman, S.J. Persistence of malignant hematopoietic progenitors in chronic myelogenous leukemia patients in complete cytogenetic remission following imatinib mesylate treatment. Blood 2003, 101, 4701–4707. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dean, M.; Fojo, T.; Bates, S. Tumour stem cells and drug resistance. Nat. Rev. Cancer 2005, 5, 275–284. [Google Scholar] [CrossRef]
- Dylla, S.J.; Beviglia, L.; Park, I.K.; Chartier, C.; Raval, J.; Ngan, L.; Pickell, K.; Aguilar, J.; Lazetic, S.; Smith-Berdan, S.; et al. Colorectal cancer stem cells are enriched in xenogeneic tumors following chemotherapy. PLoS ONE 2008, 3, e2428. [Google Scholar] [CrossRef]
- Singh, A.; Settleman, J. EMT, cancer stem cells and drug resistance: An emerging axis of evil in the war on cancer. Oncogene 2010, 29, 4741–4751. [Google Scholar] [CrossRef] [Green Version]
- Wu, F.H.; Mu, L.; Li, X.L.; Hu, Y.B.; Liu, H.; Han, L.T.; Gong, J.P. Characterization and functional analysis of a slow-cycling subpopulation in colorectal cancer enriched by cell cycle inducer combined chemotherapy. Oncotarget 2017, 8, 78466–78479. [Google Scholar] [CrossRef] [PubMed]
- Shlush, L.I.; Mitchell, A.; Heisler, L.; Abelson, S.; Ng, S.W.K.; Trotman-Grant, A.; Medeiros, J.J.F.; Rao-Bhatia, A.; Jaciw-Zurakowsky, I.; Marke, R.; et al. Tracing the origins of relapse in acute myeloid leukaemia to stem cells. Nature 2017, 547, 104–108. [Google Scholar] [CrossRef]
- Roesch, A.; Vultur, A.; Bogeski, I.; Wang, H.; Zimmermann, K.M.; Speicher, D.; Körbel, C.; Laschke, M.W.; Gimotty, P.A.; Philipp, S.E.; et al. Overcoming intrinsic multidrug resistance in melanoma by blocking the mitochondrial respiratory chain of slow-cycling JARID1B(high) cells. Cancer Cell 2013, 23, 811–825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hartman, M.L.; Rozanski, M.; Osrodek, M.; Zalesna, I.; Czyz, M. Vemurafenib and trametinib reduce expression of CTGF and IL-8 in V600EBRAF melanoma cells. Lab. Investig. 2017, 97, 217–227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moore, N.; Houghton, J.; Lyle, S. Slow-cycling therapy-resistant cancer cells. Stem Cells Dev. 2012, 21, 1822–1830. [Google Scholar] [CrossRef] [Green Version]
- Desai, A.; Webb, B.; Gerson, S.L. CD133+ cells contribute to radioresistance via altered regulation of DNA repair genes in human lung cancer cells. Radiother. Oncol. 2014, 110, 538–545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lim, Y.C.; Roberts, T.L.; Day, B.W.; Harding, A.; Kozlov, S.; Kijas, A.W.; Ensbey, K.S.; Walker, D.G.; Lavin, M.F. A role for homologous recombination and abnormal cell-cycle progression in radioresistance of glioma-initiating cells. Mol. Cancer Ther. 2012, 11, 1863–1872. [Google Scholar] [CrossRef] [Green Version]
- Srivastava, A.K.; Han, C.; Zhao, R.; Cui, T.; Dai, Y.; Mao, C.; Zhao, W.; Zhang, X.; Yu, J.; Wang, Q.E. Enhanced expression of DNA polymerase eta contributes to cisplatin resistance of ovarian cancer stem cells. Proc. Natl. Acad. Sci. USA 2015, 112, 4411–4416. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; Burness, M.L.; Martin-Trevino, R.; Guy, J.; Bai, S.; Harouaka, R.; Brooks, M.D.; Shang, L.; Fox, A.; Luther, T.K.; et al. RAD51 Mediates Resistance of Cancer Stem Cells to PARP Inhibition in Triple-Negative Breast Cancer. Clin. Cancer Res. 2017, 23, 514–522. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vitale, I.; Manic, G.; De Maria, R.; Kroemer, G.; Galluzzi, L. DNA Damage in Stem Cells. Mol. Cell 2017, 66, 306–319. [Google Scholar] [CrossRef]
- Schulz, A.; Meyer, F.; Dubrovska, A.; Borgmann, K. Cancer Stem Cells and Radioresistance: DNA Repair and Beyond. Cancers 2019, 11, 862. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lehraiki, A.; Cerezo, M.; Rouaud, F.; Abbe, P.; Allegra, M.; Kluza, J.; Marchetti, P.; Imbert, V.; Cheli, Y.; Bertolotto, C.; et al. Increased CD271 expression by the NF-kB pathway promotes melanoma cell survival and drives acquired resistance to BRAF inhibitor vemurafenib. Cell Discov. 2015, 1, 15030. [Google Scholar] [CrossRef] [PubMed]
- Müller, J.; Krijgsman, O.; Tsoi, J.; Robert, L.; Hugo, W.; Song, C.; Kong, X.; Possik, P.A.; Cornelissen-Steijger, P.D.; Geukes Foppen, M.H.; et al. Low MITF/AXL ratio predicts early resistance to multiple targeted drugs in melanoma. Nat. Commun. 2014, 5, 5712. [Google Scholar] [CrossRef]
- Boiko, A.D.; Razorenova, O.V.; van de Rijn, M.; Swetter, S.M.; Johnson, D.L.; Ly, D.P.; Butler, P.D.; Yang, G.P.; Joshua, B.; Kaplan, M.J.; et al. Human melanoma-initiating cells express neural crest nerve growth factor receptor CD271. Nature 2010, 466, 133–137. [Google Scholar] [CrossRef]
- Civenni, G.; Walter, A.; Kobert, N.; Mihic-Probst, D.; Zipser, M.; Belloni, B.; Seifert, B.; Moch, H.; Dummer, R.; van den Broek, M.; et al. Human CD271-positive melanoma stem cells associated with metastasis establish tumor heterogeneity and long-term growth. Cancer Res. 2011, 71, 3098–3109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Redmer, T.; Welte, Y.; Behrens, D.; Fichtner, I.; Przybilla, D.; Wruck, W.; Yaspo, M.L.; Lehrach, H.; Schäfer, R.; Regenbrecht, C.R. The nerve growth factor receptor CD271 is crucial to maintain tumorigenicity and stem-like properties of melanoma cells. PLoS ONE 2014, 9, e92596. [Google Scholar] [CrossRef] [Green Version]
- Redmer, T.; Walz, I.; Klinger, B.; Khouja, S.; Welte, Y.; Schäfer, R.; Regenbrecht, C. The role of the cancer stem cell marker CD271 in DNA damage response and drug resistance of melanoma cells. Oncogenesis 2017, 6, e291. [Google Scholar] [CrossRef] [Green Version]
- Filipp, F.V.; Li, C.; Boiko, A.D. CD271 is a molecular switch with divergent roles in melanoma and melanocyte development. Sci. Rep. 2019, 9, 7696. [Google Scholar] [CrossRef] [Green Version]
- Fang, Y.; Zhang, X. Targeting NEK2 as a promising therapeutic approach for cancer treatment. Cell Cycle 2016, 15, 895–907. [Google Scholar] [CrossRef] [Green Version]
- Zhang, M.; Liang, C.; Chen, Q.; Yan, H.; Xu, J.; Zhao, H.; Yuan, X.; Liu, J.; Lin, S.; Lu, W.; et al. Histone H2A phosphorylation recruits topoisomerase IIα to centromeres to safeguard genomic stability. EMBO J. 2020, 39, e101863. [Google Scholar] [CrossRef]
- Pires, E.; Sung, P.; Wiese, C. Role of RAD51AP1 in homologous recombination DNA repair and carcinogenesis. DNA Repair 2017, 59, 76–81. [Google Scholar] [CrossRef]
- Liang, F.; Miller, A.S.; Longerich, S.; Tang, C.; Maranon, D.; Williamson, E.A.; Hromas, R.; Wiese, C.; Kupfer, G.M.; Sung, P. DNA requirement in FANCD2 deubiquitination by USP1-UAF1-RAD51AP1 in the Fanconi anemia DNA damage response. Nat. Commun. 2019, 10, 2849. [Google Scholar] [CrossRef] [PubMed]
- Roesch, A.; Fukunaga-Kalabis, M.; Schmidt, E.C.; Zabierowski, S.E.; Brafford, P.A.; Vultur, A.; Basu, D.; Gimotty, P.; Vogt, T.; Herlyn, M. A temporarily distinct subpopulation of slow-cycling melanoma cells is required for continuous tumor growth. Cell 2010, 141, 583–594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, X.; Liu, L.; Yang, S.; Song, N.; Zhou, X.; Gao, J.; Yu, N.; Shan, L.; Wang, Q.; Liang, J.; et al. Histone demethylase KDM5B is a key regulator of genome stability. Proc. Natl. Acad. Sci. USA 2014, 111, 7096–7101. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bayo, J.; Tran, T.A.; Wang, L.; Pena-Llopis, S.; Das, A.K.; Martinez, E.D. Jumonji inhibitors overcome radioresistance in cancer through changes in H3K4 methylation at double-strand breaks. Cell. Rep. 2018, 25, 1040–1050. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shannan, B.; Matschke, J.; Chauvistré, H.; Vogel, F.; Klein, D.; Meier, F.; Westphal, D.; Bruns, J.; Rauschenberg, R.; Utikal, J.; et al. Sequence-dependent cross-resistance of combined radiotherapy plus BRAFV600E inhibition in melanoma. Eur. J. Cancer 2019, 109, 137–153. [Google Scholar] [CrossRef]
- Zhu, C.; Wei, Y.; Wei, X. AXL receptor tyrosine kinase as a promising anti-cancer approach: Functions, molecular mechanisms and clinical applications. Mol. Cancer 2019, 18, 153. [Google Scholar] [CrossRef] [Green Version]
- Brand, T.M.; Iida, M.; Stein, A.P.; Corrigan, K.L.; Braverman, C.M.; Coan, J.P.; Pearson, H.E.; Bahrar, H.; Fowler, T.L.; Bednarz, B.P.; et al. AXL Is a Logical Molecular Target in Head and Neck Squamous Cell Carcinoma. Clin. Cancer Res. 2015, 21, 2601–2612. [Google Scholar] [CrossRef] [Green Version]
- Balaji, K.; Vijayaraghavan, S.; Diao, L.; Tong, P.; Fan, Y.; Carey, J.P.; Bui, T.N.; Warner, S.; Heymach, J.V.; Hunt, K.K.; et al. AXL Inhibition Suppresses the DNA Damage Response and Sensitizes Cells to PARP Inhibition in Multiple Cancers. Mol. Cancer Res. 2017, 15, 45–58. [Google Scholar] [CrossRef] [Green Version]
- Kariolis, M.S.; Miao, Y.R.; Diep, A.; Nash, S.E.; Olcina, M.M.; Jiang, D.; Jones, D.S., 2nd; Kapur, S.; Mathews, I.I.; Koong, A.C.; et al. Inhibition of the GAS6/AXL pathway augments the efficacy of chemotherapies. J. Clin. Investig. 2017, 127, 183–198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, C.; Jin, H.; Wang, N.; Fan, S.; Wang, Y.; Zhang, Y.; Wei, L.; Tao, X.; Gu, D.; Zhao, F.; et al. Gas6/Axl Axis Contributes to Chemoresistance and Metastasis in Breast Cancer through Akt/GSK-3β/β-catenin Signaling. Theranostics 2016, 6, 1205–1219. [Google Scholar] [CrossRef]
- Zuo, Q.; Liu, J.; Huang, L.; Qin, Y.; Hawley, T.; Seo, C.; Merlino, G.; Yu, Y. AXL/AKT axis mediated-resistance to BRAF inhibitor depends on PTEN status in melanoma. Oncogene 2018, 37, 3275–3289. [Google Scholar] [CrossRef]
- Sen, T.; Tong, P.; Diao, L.; Li, L.; Fan, Y.; Hoff, J.; Heymach, J.V.; Wang, J.; Byers, L.A. Targeting AXL and mTOR Pathway Overcomes Primary and Acquired Resistance to WEE1 Inhibition in Small-Cell Lung Cancer. Clin. Cancer Res. 2017, 23, 6239–6253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Flem-Karlsen, K.; McFadden, E.; Omar, N.; Haugen, M.H.; Øy, G.F.; Ryder, T.; Gullestad, H.P.; Hermann, R.; Mælandsmo, G.M.; Flørenes, V.A. Targeting AXL and the DNA Damage Response Pathway as a Novel Therapeutic Strategy in Melanoma. Mol. Cancer Ther. 2020, 19, 895–905. [Google Scholar] [CrossRef] [Green Version]
- Smith, J.; Tho, L.M.; Xu, N.; Gillespie, D.A. The ATM-Chk2 and ATR-Chk1 pathways in DNA damage signaling and cancer. Adv. Cancer Res. 2010, 108, 73–112. [Google Scholar] [CrossRef] [PubMed]
- Hartman, M.L.; Sztiller-Sikorska, M.; Gajos-Michniewicz, A.; Czyz, M. Dissecting Mechanisms of Melanoma Resistance to BRAF and MEK Inhibitors Revealed Genetic and Non-Genetic Patient- and Drug-Specific Alterations and Remarkable Phenotypic Plasticity. Cells 2020, 9, 142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pathria, G.; Garg, B.; Borgdorff, V.; Garg, K.; Wagner, C.; Superti-Furga, G.; Wagner, S.N. Overcoming MITF-conferred drug resistance through dual AURKA/MAPK targeting in human melanoma cells. Cell Death Dis. 2016, 7, e2135. [Google Scholar] [CrossRef] [Green Version]
- Arozarena, I.; Wellbrock, C. Overcoming resistance to BRAF inhibitors. Ann. Transl. Med. 2017, 5, 387. [Google Scholar] [CrossRef] [Green Version]
- Hertzman Johansson, C.; Azimi, A.; Frostvik Stolt, M.; Shojaee, S.; Wiberg, H.; Grafström, E.; Hansson, J.; Egyházi Brage, S. Association of MITF and other melanosome-related proteins with chemoresistance in melanoma tumors and cell lines. Melanoma Res. 2013, 23, 360–365. [Google Scholar] [CrossRef] [PubMed]
- Seoane, M.; Buhs, S.; Iglesias, P.; Strauss, J.; Puller, A.C.; Müller, J.; Gerull, H.; Feldhaus, S.; Alawi, M.; Brandner, J.M.; et al. Lineage-specific control of TFIIH by MITF determines transcriptional homeostasis and DNA repair. Oncogene 2019, 38, 3616–3635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Beuret, L.; Ohanna, M.; Strub, T.; Allegra, M.; Davidson, I.; Bertolotto, C.; Ballotti, R. BRCA1 is a new MITF target gene. Pigment Cell Melanoma Res. 2011, 24, 725–727. [Google Scholar] [CrossRef] [PubMed]
- Strub, T.; Giuliano, S.; Ye, T.; Bonet, C.; Keime, C.; Kobi, D.; Le Gras, S.; Cormont, M.; Ballotti, R.; Bertolotto, C.; et al. Essential role of microphthalmia transcription factor for DNA replication, mitosis and genomic stability in melanoma. Oncogene 2011, 30, 2319–2332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Krumm, A.; Barckhausen, C.; Kücük, P.; Tomaszowski, K.H.; Loquai, C.; Fahrer, J.; Krämer, O.H.; Kaina, B.; Roos, W.P. Enhanced Histone Deacetylase Activity in Malignant Melanoma Provokes RAD51 and FANCD2-Triggered Drug Resistance. Cancer Res. 2016, 76, 3067–3077. [Google Scholar] [CrossRef] [Green Version]
- Tuominen, R.; Jewell, R.; van den Oord, J.J.; Wolter, P.; Stierner, U.; Lindholm, C.; Hertzman Johansson, C.; Lindén, D.; Johansson, H.; Frostvik Stolt, M.; et al. MGMT promoter methylation is associated with temozolomide response and prolonged progression-free survival in disseminated cutaneous melanoma. Int. J. Cancer 2015, 136, 2844–2853. [Google Scholar] [CrossRef] [Green Version]
- Weller, M.; Stupp, R.; Reifenberger, G.; Brandes, A.A.; van den Bent, M.J.; Wick, W.; Hegi, M.E. MGMT promoter methylation in malignant gliomas: Ready for personalized medicine? Nat. Rev. Neurol. 2010, 6, 39–51. [Google Scholar] [CrossRef] [Green Version]
- Hoon, D.S.; Spugnardi, M.; Kuo, C.; Huang, S.K.; Morton, D.L.; Taback, B. Profiling epigenetic inactivation of tumor suppressor genes in tumors and plasma from cutaneous melanoma patients. Oncogene 2004, 23, 4014–4022. [Google Scholar] [CrossRef] [Green Version]
- Hassel, J.C.; Sucker, A.; Edler, L.; Kurzen, H.; Moll, I.; Stresemann, C.; Spieth, K.; Mauch, C.; Rass, K.; Dummer, R.; et al. MGMT gene promoter methylation correlates with tolerance of temozolomide treatment in melanoma but not with clinical outcome. Br. J. Cancer 2010, 103, 820–826. [Google Scholar] [CrossRef] [Green Version]
- Schraml, P.; von Teichman, A.; Mihic-Probst, D.; Simcock, M.; Ochsenbein, A.; Dummer, R.; Michielin, O.; Seifert, B.; Schläppi, M.; Moch, H.; et al. Predictive value of the MGMT promoter methylation status in metastatic melanoma patients receiving first-line temozolomide plus bevacizumab in the trial SAKK 50/07. Oncol. Rep. 2012, 28, 654–658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Robb, R.; Yang, L.; Shen, C.; Wolfe, A.R.; Webb, A.; Zhang, X.; Vedaie, M.; Saji, M.; Jhiang, S.; Ringel, M.D.; et al. Inhibiting BRAF Oncogene-Mediated Radioresistance Effectively Radiosensitizes BRAFV600E-Mutant Thyroid Cancer Cells by Constraining DNA Double-Strand Break Repair. Clin. Cancer Res. 2019, 25, 4749–4760. [Google Scholar] [CrossRef] [PubMed]
- Estrada-Bernal, A.; Chatterjee, M.; Haque, S.J.; Yang, L.; Morgan, M.A.; Kotian, S.; Morrell, D.; Chakravarti, A.; Williams, T.M. MEK inhibitor GSK1120212-mediated radiosensitization of pancreatic cancer cells involves inhibition of DNA double-strand break repair pathways. Cell Cycle 2015, 14, 3713–3724. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moriceau, G.; Hugo, W.; Hong, A.; Shi, H.; Kong, X.; Yu, C.C.; Koya, R.C.; Samatar, A.A.; Khanlou, N.; Braun, J.; 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, 240–256. [Google Scholar] [CrossRef] [Green Version]
- Das Thakur, M.; Salangsang, F.; Landman, A.S.; Sellers, W.R.; Pryer, N.K.; Levesque, M.P.; Dummer, R.; McMahon, M.; Stuart, D.D. Modelling vemurafenib resistance in melanoma reveals a strategy to forestall drug resistance. Nature 2013, 494, 251–255. [Google Scholar] [CrossRef] [PubMed]
- Nazarian, R.; Shi, H.; Wang, Q.; Kong, X.; Koya, R.C.; Lee, H.; Chen, Z.; Lee, M.K.; Attar, N.; Sazegar, H.; et al. Melanomas acquire resistance to B-RAF(V600E) inhibition by RTK or N-RAS upregulation. Nature 2010, 468, 973–977. [Google Scholar] [CrossRef] [Green Version]
- Hugo, W.; Shi, H.; Sun, L.; Piva, M.; Song, C.; Kong, X.; Moriceau, G.; Hong, A.; Dahlman, K.B.; Johnson, D.B.; et al. Non-genomic and Immune Evolution of Melanoma Acquiring MAPKi Resistance. Cell 2015, 162, 1271–1285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bhattacharya, S.; Srinivasan, K.; Abdisalaam, S.; Su, F.; Raj, P.; Dozmorov, I.; Mishra, R.; Wakeland, E.K.; Ghose, S.; Mukherjee, S.; et al. RAD51 interconnects between DNA replication, DNA repair and immunity. Nucleic Acids Res. 2017, 45, 4590–4605. [Google Scholar] [CrossRef] [Green Version]
- Gopal, Y.N.; Deng, W.; Woodman, S.E.; Komurov, K.; Ram, P.; Smith, P.D.; Davies, M.A. Basal and treatment-induced activation of AKT mediates resistance to cell death by AZD6244 (ARRY-142886) in Braf-mutant human cutaneous melanoma cells. Cancer Res. 2010, 70, 8736–8747. [Google Scholar] [CrossRef] [Green Version]
- Deng, W.; Gopal, Y.N.; Scott, A.; Chen, G.; Woodman, S.E.; Davies, M.A. Role and therapeutic potential of PI3K-mTOR signaling in de novo resistance to BRAF inhibition. Pigment Cell Melanoma Res. 2012, 25, 248–258. [Google Scholar] [CrossRef]
- Shen, W.H.; Balajee, A.S.; Wang, J.; Wu, H.; Eng, C.; Pandolfi, P.P.; Yin, Y. Essential role for nuclear PTEN in maintaining chromosomal integrity. Cell 2007, 128, 157–170. [Google Scholar] [CrossRef] [Green Version]
- He, J.; Kang, X.; Yin, Y.; Chao, K.S.; Shen, W.H. PTEN regulates DNA replication progression and stalled fork recovery. Nat. Commun. 2015, 6, 7620. [Google Scholar] [CrossRef] [Green Version]
- Kang, X.; Song, C.; Du, X.; Zhang, C.; Liu, Y.; Liang, L.; He, J.; Lamb, K.; Shen, W.H.; Yin, Y. PTEN stabilizes TOP2A and regulates the DNA decatenation. Sci. Rep. 2015, 5, 17873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sulkowski, P.L.; Scanlon, S.E.; Oeck, S.; Glazer, P.M. PTEN Regulates Nonhomologous End Joining By Epigenetic Induction of NHEJ1/XLF. Mol. Cancer Res. 2018, 16, 1241–1254. [Google Scholar] [CrossRef] [Green Version]
- Mendes-Pereira, A.M.; Martin, S.A.; Brough, R.; McCarthy, A.; Taylor, J.R.; Kim, J.S.; Waldman, T.; Lord, C.J.; Ashworth, A. Synthetic lethal targeting of PTEN mutant cells with PARP inhibitors. EMBO Mol. Med. 2009, 1, 315–322. [Google Scholar] [CrossRef] [PubMed]
- Turchick, A.; Liu, Y.; Zhao, W.; Cohen, I.; Glazer, P.M. Synthetic lethality of a cell-penetrating anti-RAD51 antibody in PTEN-deficient melanoma and glioma cells. Oncotarget 2019, 10, 1272–1283. [Google Scholar] [CrossRef]
- Aguissa-Touré, A.H.; Li, G. Genetic alterations of PTEN in human melanoma. Cell. Mol. Life Sci. 2012, 69, 1475–1491. [Google Scholar] [CrossRef]
- Zhou, X.P.; Gimm, O.; Hampel, H.; Niemann, T.; Walker, M.J.; Eng, C. Epigenetic PTEN silencing in malignant melanomas without PTEN mutation. Am. J. Pathol. 2000, 157, 1123–1128. [Google Scholar] [CrossRef] [Green Version]
- Mirmohammadsadegh, A.; Marini, A.; Nambiar, S.; Hassan, M.; Tannapfel, A.; Ruzicka, T.; Hengge, U.R. Epigenetic silencing of the PTEN gene in melanoma. Cancer Res. 2006, 66, 6546–6552. [Google Scholar] [CrossRef] [Green Version]
- Santamaría, P.G.; Floristán, A.; Fontanals-Cirera, B.; Vázquez-Naharro, A.; Santos, V.; Morales, S.; Yuste, L.; Peinado, H.; García-Gómez, A.; Portillo, F.; et al. Lysyl oxidase-like 3 is required for melanoma cell survival by maintaining genomic stability. Cell Death Differ. 2018, 25, 935–950. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mancuso, P.; Tricarico, R.; Bhattacharjee, V.; Cosentino, L.; Kadariya, Y.; Jelinek, J.; Nicolas, E.; Einarson, M.; Beeharry, N.; Devarajan, K.; et al. Thymine DNA glycosylase as a novel target for melanoma. Oncogene 2019, 38, 3710–3728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Burkel, F.; Jost, T.; Hecht, M.; Heinzerling, L.; Fietkau, R.; Distel, L. Dual mTOR/DNA-PK Inhibitor CC-115 Induces Cell Death in Melanoma Cells and Has Radiosensitizing Potential. Int. J. Mol. Sci. 2020, 21, 9321. [Google Scholar] [CrossRef]
- Seidel, J.A.; Otsuka, A.; Kabashima, K. Anti-PD-1 and Anti-CTLA-4 Therapies in Cancer: Mechanisms of Action, Efficacy, and Limitations. Front. Oncol. 2018, 8, 86. [Google Scholar] [CrossRef]
- Rowshanravan, B.; Halliday, N.; Sansom, D.M. CTLA-4: A moving target in immunotherapy. Blood 2018, 131, 58–67. [Google Scholar] [CrossRef] [PubMed]
- Chamoto, K.; Al-Habsi, M.; Honjo, T. Role of PD-1 in Immunity and Diseases. Curr. Top. Microbiol. Immunol. 2017, 410, 75–97. [Google Scholar] [CrossRef]
- 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]
- Schachter, J.; Ribas, A.; Long, G.V.; Arance, A.; Grob, J.J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.; Lotem, M.; 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, 1853–1862. [Google Scholar] [CrossRef]
- Larkin, J.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Dummer, R.; Smylie, M.; Rutkowski, P.; et al. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N. Engl. J. Med. 2015, 373, 23–34. [Google Scholar] [CrossRef] [Green Version]
- Robert, C.; Schachter, J.; Long, G.V.; Arance, A.; Grob, J.J.; Mortier, L.; Daud, A.; Carlino, M.S.; McNeil, C.; Lotem, M.; et al. Pembrolizumab versus Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2015, 372, 2521–2532. [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] [Green Version]
- Wolchok, J.D.; Chiarion-Sileni, V.; Gonzalez, R.; Rutkowski, P.; Grob, J.J.; Cowey, C.L.; Lao, C.D.; Wagstaff, J.; Schadendorf, D.; Ferrucci, P.F.; et al. Overall Survival with Combined Nivolumab and Ipilimumab in Advanced Melanoma. N. Engl. J. Med. 2017, 377, 1345–1356. [Google Scholar] [CrossRef] [PubMed]
- Byrne, E.H.; Fisher, D.E. Immune and molecular correlates in melanoma treated with immune checkpoint blockade. Cancer 2017, 123, 2143–2153. [Google Scholar] [CrossRef] [PubMed] [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]
- Paijens, S.T.; Vledder, A.; de Bruyn, M.; Nijman, H.W. Tumor-infiltrating lymphocytes in the immunotherapy era. Cell. Mol. Immunol. 2020. [Google Scholar] [CrossRef] [PubMed]
- Maibach, F.; Sadozai, H.; Seyed Jafari, S.M.; Hunger, R.E.; Schenk, M. Tumor-Infiltrating Lymphocytes and Their Prognostic Value in Cutaneous Melanoma. Front. Immunol. 2020, 11, 2105. [Google Scholar] [CrossRef]
- 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]
- Galuppini, F.; Dal Pozzo, C.A.; Deckert, J.; Loupakis, F.; Fassan, M.; Baffa, R. Tumor mutation burden: From comprehensive mutational screening to the clinic. Cancer Cell Int. 2019, 19, 209. [Google Scholar] [CrossRef]
- Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.; Behjati, S.; Biankin, A.V.; Bignell, G.R.; Bolli, N.; Borg, A.; Børresen-Dale, A.L.; et al. Signatures of mutational processes in human cancer. Nature. 2013, 500, 415–421. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Chalmers, Z.R.; Greenbowe, J.; Ali, S.M.; Balasubramanian, S.; Sun, J.X.; He, Y.; Frederick, D.T.; Puzanov, I.; Balko, J.M.; Cates, J.M.; et al. Targeted Next Generation Sequencing Identifies Markers of Response to PD-1 Blockade. Cancer Immunol. Res. 2016, 4, 959–967. [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]
- Zaretsky, J.M.; Garcia-Diaz, A.; Shin, D.S.; Escuin-Ordinas, H.; Hugo, W.; Hu-Lieskovan, S.; Torrejon, D.Y.; Abril-Rodriguez, G.; Sandoval, S.; Barthly, L.; et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N. Engl. J. Med. 2016, 375, 819–829. [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.; 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]
- Kümpers, C.; Jokic, M.; Haase, O.; Offermann, A.; Vogel, W.; Grätz, V.; Langan, E.A.; Perner, S.; Terheyden, P. Immune Cell Infiltration of the Primary Tumor, Not PD-L1 Status, Is Associated With Improved Response to Checkpoint Inhibition in Metastatic Melanoma. Front. Med. 2019, 6, 27. [Google Scholar] [CrossRef] [Green Version]
- Thomas, N.E.; Busam, K.J.; From, L.; Kricker, A.; Armstrong, B.K.; Anton-Culver, H.; Gruber, S.B.; Gallagher, R.P.; Zanetti, R.; Rosso, S.; et al. Tumor-infiltrating lymphocyte grade in primary melanomas is independently associated with melanoma-specific survival in the population-based genes, environment and melanoma study. J. Clin. Oncol. 2013, 31, 4252–4259. [Google Scholar] [CrossRef] [Green Version]
- Mandalà, M.; Merelli, B.; Massi, D. PD-L1 in melanoma: Facts and myths. Melanoma Manag. 2016, 3, 187–194. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Deng, W.; Li, N.; Neri, S.; Sharma, A.; Jiang, W.; Lin, S.H. Combining Immunotherapy and Radiotherapy for Cancer Treatment: Current Challenges and Future Directions. Front. Pharmacol. 2018, 9, 185. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Biau, J.; Chautard, E.; Verrelle, P.; Dutreix, M. Altering DNA Repair to Improve Radiation Therapy: Specific and Multiple Pathway Targeting. Front. Oncol. 2019, 9, 1009. [Google Scholar] [CrossRef]
- Demaria, S.; Golden, E.B.; Formenti, S.C. Role of Local Radiation Therapy in Cancer Immunotherapy. JAMA Oncol. 2015, 1, 1325–1332. [Google Scholar] [CrossRef]
- Reits, E.A.; Hodge, J.W.; Herberts, C.A.; Groothuis, T.A.; Chakraborty, M.; Wansley, E.K.; Camphausen, K.; Luiten, R.M.; de Ru, A.H.; Neijssen, J.; et al. Radiation modulates the peptide repertoire, enhances MHC class I expression, and induces successful antitumor immunotherapy. J. Exp. Med. 2006, 203, 1259–1271. [Google Scholar] [CrossRef]
- Corso, C.D.; Ali, A.N.; Diaz, R. Radiation-induced tumor neoantigens: Imaging and therapeutic implications. Am. J. Cancer Res. 2011, 1, 390–412. [Google Scholar] [PubMed]
- Sharabi, A.B.; Lim, M.; DeWeese, T.L.; Drake, C.G. Radiation and checkpoint blockade immunotherapy: Radiosensitisation and potential mechanisms of synergy. Lancet Oncol. 2015, 16, e498–e509. [Google Scholar] [CrossRef]
- Durante, M.; Formenti, S.C. Radiation-Induced Chromosomal Aberrations and Immunotherapy: Micronuclei, Cytosolic DNA, and Interferon-Production Pathway. Front. Oncol. 2018, 8, 192. [Google Scholar] [CrossRef] [PubMed]
- Woo, S.R.; Fuertes, M.B.; Corrales, L.; Spranger, S.; Furdyna, M.J.; Leung, M.Y.; Duggan, R.; Wang, Y.; Barber, G.N.; Fitzgerald, K.A.; et al. STING-dependent cytosolic DNA sensing mediates innate immune recognition of immunogenic tumors. Immunity 2014, 41, 830–842. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hou, Y.; Liang, H.; Rao, E.; Zheng, W.; Huang, X.; Deng, L.; Zhang, Y.; Yu, X.; Xu, M.; Mauceri, H.; et al. Non-canonical NF-κB Antagonizes STING Sensor-Mediated DNA Sensing in Radiotherapy. Immunity 2018, 49, 490–503.e4. [Google Scholar] [CrossRef] [Green Version]
- Hopfner, K.P.; Hornung, V. Molecular mechanisms and cellular functions of cGAS-STING signalling. Nat. Rev. Mol. Cell. Biol. 2020, 21, 501–521. [Google Scholar] [CrossRef]
- Deng, L.; Liang, H.; Xu, M.; Yang, X.; Burnette, B.; Arina, A.; Li, X.D.; Mauceri, H.; Beckett, M.; Darga, T.; et al. STING-Dependent Cytosolic DNA Sensing Promotes Radiation-Induced Type I Interferon-Dependent Antitumor Immunity in Immunogenic Tumors. Immunity 2014, 41, 843–852. [Google Scholar] [CrossRef] [Green Version]
- Strojan, P. Role of radiotherapy in melanoma management. Radiol. Oncol. 2010, 44, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Reijnen, C.; Küsters-Vandevelde, H.V.N.; Prinsen, C.F.; Massuger, L.F.A.G.; Snijders, M.P.M.L.; Kommoss, S.; Brucker, S.Y.; Kwon, J.S.; McAlpine, J.N.; Pijnenborg, J.M.A. Mismatch repair deficiency as a predictive marker for response to adjuvant radiotherapy in endometrial cancer. Gynecol. Oncol. 2019, 154, 124–130. [Google Scholar] [CrossRef]
- Dovedi, S.J.; Adlard, A.L.; Lipowska-Bhalla, G.; McKenna, C.; Jones, S.; Cheadle, E.J.; Stratford, I.J.; Poon, E.; Morrow, M.; Stewart, R.; et al. Acquired resistance to fractionated radiotherapy can be overcome by concurrent PD-L1 blockade. Cancer Res. 2014, 74, 5458–5468. [Google Scholar] [CrossRef] [Green Version]
- Derer, A.; Spiljar, M.; Bäumler, M.; Hecht, M.; Fietkau, R.; Frey, B.; Gaipl, U.S. Chemoradiation Increases PD-L1 Expression in Certain Melanoma and Glioblastoma Cells. Front. Immunol. 2016, 7, 610. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Twyman-Saint Victor, C.; Rech, A.J.; Maity, A.; Rengan, R.; Pauken, K.E.; Stelekati, E.; Benci, J.L.; Xu, B.; Dada, H.; Odorizzi, P.M.; et al. Radiation and dual checkpoint blockade activate non-redundant immune mechanisms in cancer. Nature 2015, 520, 373–377. [Google Scholar] [CrossRef] [Green Version]
- Yilmaz, M.T.; Elmali, A.; Yazici, G. Abscopal Effect, From Myth to Reality: From Radiation Oncologists’ Perspective. Cureus 2019, 11, e3860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Harding, S.M.; Benci, J.L.; Irianto, J.; Discher, D.E.; Minn, A.J.; Greenberg, R.A. Mitotic progression following DNA damage enables pattern recognition within micronuclei. Nature 2017, 548, 466–470. [Google Scholar] [CrossRef] [Green Version]
- Stamell, E.F.; Wolchok, J.D.; Gnjatic, S.; Lee, N.Y.; Brownell, I. The abscopal effect associated with a systemic anti-melanoma immune response. Int. J. Radiat. Oncol. Biol. Phys. 2013, 85, 293–295. [Google Scholar] [CrossRef] [Green Version]
- Postow, M.A.; Callahan, M.K.; Barker, C.A.; Yamada, Y.; Yuan, J.; Kitano, S.; Mu, Z.; Rasalan, T.; Adamow, M.; Ritter, E.; et al. Immunologic correlates of the abscopal effect in a patient with melanoma. N. Engl. J. Med. 2012, 366, 925–931. [Google Scholar] [CrossRef] [Green Version]
- Park, S.S.; Dong, H.; Liu, X.; Harrington, S.M.; Krco, C.J.; Grams, M.P.; Mansfield, A.S.; Furutani, K.M.; Olivier, K.R.; Kwon, E.D. PD-1 Restrains Radiotherapy-Induced Abscopal Effect. Cancer Immunol. Res. 2015, 3, 610–619. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ribeiro Gomes, J.; Schmerling, R.A.; Haddad, C.K.; Racy, D.J.; Ferrigno, R.; Gil, E.; Zanuncio, P.; Buzaid, A.C. Analysis of the Abscopal Effect With Anti-PD1 Therapy in Patients With Metastatic Solid Tumors. J. Immunother. 2016, 39, 367–372. [Google Scholar] [CrossRef] [PubMed]
- Trommer, M.; Yeo, S.Y.; Persigehl, T.; Bunck, A.; Grüll, H.; Schlaak, M.; Theurich, S.; von Bergwelt-Baildon, M.; Morgenthaler, J.; Herter, J.M.; et al. Abscopal Effects in Radio-Immunotherapy-Response Analysis of Metastatic Cancer Patients With Progressive Disease Under Anti-PD-1 Immune Checkpoint. Front. Pharmacol. 2019, 10, 511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- ElJalby, M.; Pannullo, S.C.; Schwartz, T.H.; Parashar, B.; Wernicke, A.G. Optimal Timing and Sequence of Immunotherapy When Combined with Stereotactic Radiosurgery in the Treatment of Brain Metastases. World Neurosurg. 2019, 127, 397–404. [Google Scholar] [CrossRef] [PubMed]
- Qian, J.M.; Yu, J.B.; Kluger, H.M.; Chiang, V.L. Timing and type of immune checkpoint therapy affect the early radiographic response of melanoma brain metastases to stereotactic radiosurgery. Cancer 2016, 122, 3051–3058. [Google Scholar] [CrossRef] [Green Version]
- Patel, K.R.; Shoukat, S.; Oliver, D.E.; Chowdhary, M.; Rizzo, M.; Lawson, D.H.; Khosa, F.; Liu, Y.; Khan, M.K. Ipilimumab and Stereotactic Radiosurgery Versus Stereotactic Radiosurgery Alone for Newly Diagnosed Melanoma Brain Metastases. Am. J. Clin. Oncol. 2017, 40, 444–450. [Google Scholar] [CrossRef]
- Knispel, S.; Stang, A.; Zimmer, L.; Lax, H.; Gutzmer, R.; Heinzerling, L.; Weishaupt, C.; Pföhler, C.; Gesierich, A.; Herbst, R.; et al. Impact of a preceding radiotherapy on the outcome of immune checkpoint inhibition in metastatic melanoma: A multicenter retrospective cohort study of the DeCOG. J. Immunother. Cancer 2020, 8, e000395. [Google Scholar] [CrossRef] [PubMed]
- Mouw, K.W.; Goldberg, M.S.; Konstantinopoulos, P.A.; D’Andrea, A.D. DNA Damage and Repair Biomarkers of Immunotherapy Response. Cancer Discov. 2017, 7, 675–693. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.; Shih, D.J.H.; Lin, S.Y. Role of DNA repair defects in predicting immunotherapy response. Biomark. Res. 2020, 8, 23. [Google Scholar] [CrossRef]
- 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]
- Kelderman, S.; Schumacher, T.N.; Kvistborg, P. Mismatch Repair-Deficient Cancers Are Targets for Anti-PD-1 Therapy. Cancer Cell 2015, 28, 11–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lemery, S.; Keegan, P.; Pazdur, R. First FDA Approval Agnostic of Cancer Site—When a Biomarker Defines the Indication. N. Engl. J. Med. 2017, 377, 1409–1412. [Google Scholar] [CrossRef]
- Conway, J.R.; Dietlein, F.; Taylor-Weiner, A.; AlDubayan, S.; Vokes, N.; Keenan, T.; Reardon, B.; He, M.X.; Margolis, C.A.; Weirather, J.L.; et al. Integrated molecular drivers coordinate biological and clinical states in melanoma. Nat. Genet. 2020, 52, 1373–1383. [Google Scholar] [CrossRef]
- Ponti, G.; Pellacani, G.; Tomasi, A.; Depenni, R.; Maccaferri, M.; Maiorana, A.; Orsi, G.; Giusti, F.; Cascinu, S.; Manfredini, M. Immunohistochemical mismatch repair proteins expression as a tool to predict the melanoma immunotherapy response. Mol. Clin. Oncol. 2020, 12, 3–8. [Google Scholar] [CrossRef] [Green Version]
- Mandal, R.; Samstein, R.M.; Lee, K.W.; Havel, J.J.; Wang, H.; Krishna, C.; Sabio, E.Y.; Makarov, V.; Kuo, F.; Blecua, P.; et al. Genetic diversity of tumors with mismatch repair deficiency influences anti-PD-1 immunotherapy response. Science 2019, 364, 485–491. [Google Scholar] [CrossRef]
- Biau, J.; Devun, F.; Jdey, W.; Kotula, E.; Quanz, M.; Chautard, E.; Sayarath, M.; Sun, J.S.; Verrelle, P.; Dutreix, M. A preclinical study combining the DNA repair inhibitor Dbait with radiotherapy for the treatment of melanoma. Neoplasia 2014, 16, 835–844. [Google Scholar] [CrossRef] [Green Version]
- Le Tourneau, C.; Dreno, B.; Kirova, Y.; Grob, J.J.; Jouary, T.; Dutriaux, C.; Thomas, L.; Lebbé, C.; Mortier, L.; Saiag, P.; et al. First-in-human phase I study of the DNA-repair inhibitor DT01 in combination with radiotherapy in patients with skin metastases from melanoma. Br. J. Cancer 2016, 114, 1199–1205. [Google Scholar] [CrossRef] [PubMed]
- Higuchi, T.; Flies, D.B.; Marjon, N.A.; Mantia-Smaldone, G.; Ronner, L.; Gimotty, P.A.; Adams, S.F. CTLA-4 Blockade Synergizes Therapeutically with PARP Inhibition in BRCA1-Deficient Ovarian Cancer. Cancer Immunol. Res. 2015, 3, 1257–1268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiao, S.; Xia, W.; Yamaguchi, H.; Wei, Y.; Chen, M.K.; Hsu, J.M.; Hsu, J.L.; Yu, W.H.; Du, Y.; Lee, H.H.; et al. PARP Inhibitor Upregulates PD-L1 Expression and Enhances Cancer-Associated Immunosuppression. Clin. Cancer Res. 2017, 23, 3711–3720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jdey, W.; Thierry, S.; Russo, C.; Devun, F.; Al Abo, M.; Noguiez-Hellin, P.; Sun, J.S.; Barillot, E.; Zinovyev, A.; Kuperstein, I.; et al. Drug-Driven Synthetic Lethality: Bypassing Tumor Cell Genetics with a Combination of AsiDNA and PARP Inhibitors. Clin. Cancer Res. 2017, 23, 1001–1011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Clinical Trials. Available online: https://clinicaltrials.gov/ct2/home (accessed on 23 March 2021).
Identifier | Phase | Enrollment | Primary Outcome Measures | Treatment Regimen Description |
---|---|---|---|---|
NCT01676649 1 | 2 | 30 | adverse events | ipilimumab + carboplatin + paclitaxel |
NCT02097732 1 | 2 | 4 | LCR | SRS + ipilimumab |
NCT0239287 1 | 1/2 | 10 | adverse events and radiotherapy associated toxicities | radiotherapy + dabrafenib + trametinib |
NCT02617849 1 | 2 | 30 | ORR | carboplatin + paclitaxel + pembrolizumab |
NCT02718066 2 | 1/2 | 118 | RP2D | HBI-8000 (HDACi) + nivolumab |
NCT0281602 2 | 2 | 71 | ORR | azacytidine(cytidine analog) + pembrolizumab |
NCT02872259 2 | 1/2 | 92 | ORR | BGB324 (AXL inhibitor) + pembrolizumab BGB324 + dabrafenib + trametinib |
NCT02974803 1 | 2 | 6 | intracranial OR | SRS + dabrafenib + trametinib |
NCT02978404 1 | 2 | 26 | intracranial PFS | nivolumab + radiosurgery |
NCT02988817 2 | 1/2 | 374 | DLTs, adverse events | enapotamab vedotin (HuMax-AXL-ADC) |
NCT03050060 2 | 2 | 129 | ORR | nelfinavir mesylate + pembrolizumab, nivolumab, or atezolizumab + hypofractionated radiation therapy |
NCT03278665 2 | 1/2 | 40 | IAE | 4SC-202 (HDACi) + pembrolizumab |
NCT03340129 2 | 2 | 218 | intracranial response to immunotherapy | nivolumab + ipilimumab + SRS |
NCT03425279 2 | 1/2 | 120 | DLTs, MTD, ORR | CAB-AXL-ADC (anti-AXL antibody drug conjugate) |
NCT03430947 2 | 2 | 32 | ORR in brain | radiosurgery + vemurafenib + cobimetinib |
NCT03448666 3 | 2 | 53 | ORR | electrochemotherapy + pembrolizumab |
NCT03474497 2 | 1/2 | 45 | ARR | pembrolizumab + IL-2 + hypofractionated radiotherapy. |
NCT03511391 1 | 2 | 99 | PFS | nivolumab or pembrolizumab or atezolizumab + SBRT |
NCT03646617 2 | 2 | 70 | number of adverse events | ipilumumab + nivolumab + HFRT |
NCT03693014 2 | 2 | 60 | ORR | SBRT + ipilimumab, nivolumab, pembrolizumab or atezolizumab |
NCT03765229 2 | 2 | 14 | ORR, PFS | entinostat (HDACi) + pembrolizumab |
NCT03780608 1 | 2 | 61 | ORR | ceralasertib (ATR inhibitor) + durvalumab (PD-1/PD-L1 inhibitor) |
NCT03898908 2 | 2 | 38 | intracranial ORR | encorafenib + binimetinib + radiation |
NCT03925350 2 | 2 | 41 | ORR | niraparib (PARPi) |
NCT03958383 2 | 1/2 | 61 | IAE, MTD, MAD | radiation + nivolumab + ipilimumab + hu14.18-IL2 |
NCT04017897 2 | 2 | 52 | ORR | pembrolizumab or nivolumab + radiotherapy |
NCT04042506 2 | 2 | 15 | safety of SBRT | SBRT + nivolumab |
NCT04074096 3 | 2 | 150 | intracranial PFS | SRS + encorafenib + binimetinib |
NCT04133948 2 | 1/2 | 45 | safety of patients | domatinostat (HDACi) + nivolumab + ipilimumab |
NCT04187833 2 | 2 | 37 | best overall response (CR + PR) | nivolumab + talazoparib (PARPi) |
NCT04225390 2 | 2 | 38 | CR, PR, SD or PD | DTIC + re-exposure to immunotherapy |
NCT04594187 2 | 3 | 168 | time to regional nodal recurrence | nodal radiation therapy + immunotherapy |
NCT04620603 3 | 1/2 | 15 | tumor response | low dose rate brachytherapy + nivolumab |
NCT04633902 3 | 2 | 41 | ORR | olaparib (PARPi) + pembrolizumab |
NCT04793737 2 | N/A | 27 | ORR | precision radiation in patients on PD-1 inhibitor treatment that have tumor progression |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Osrodek, M.; Wozniak, M. Targeting Genome Stability in Melanoma—A New Approach to an Old Field. Int. J. Mol. Sci. 2021, 22, 3485. https://doi.org/10.3390/ijms22073485
Osrodek M, Wozniak M. Targeting Genome Stability in Melanoma—A New Approach to an Old Field. International Journal of Molecular Sciences. 2021; 22(7):3485. https://doi.org/10.3390/ijms22073485
Chicago/Turabian StyleOsrodek, Marta, and Michal Wozniak. 2021. "Targeting Genome Stability in Melanoma—A New Approach to an Old Field" International Journal of Molecular Sciences 22, no. 7: 3485. https://doi.org/10.3390/ijms22073485
APA StyleOsrodek, M., & Wozniak, M. (2021). Targeting Genome Stability in Melanoma—A New Approach to an Old Field. International Journal of Molecular Sciences, 22(7), 3485. https://doi.org/10.3390/ijms22073485