Overcoming Immune Evasion in Melanoma
Abstract
:1. Introduction to Melanoma
1.1. Melanocyte Biology and its Role in Melanoma Etiology
1.2. Melanoma Statistics and Risk Factors
1.3. Melanoma Diagnosis and Staging
1.4. Melanoma Subtypes and Their Molecular Abnormalities
2. Conventional Melanoma Therapies
2.1. Surgery
2.2. Radiation Therapy
2.3. Chemotherapy
2.4. Targeted Therapy
3. Mechanisms of Immune Evasion in Melanoma
3.1. T-Cell Dysfunction
3.2. Melanoma Microenvironment Contains Many Immune Suppressive Immune Cells: Regulatory T-cells, Myeloid Derived Suppressor Cells, and Tumor Associated Macrophages
3.2.1. Role of Regulatory T-cells in Melanoma Immune Evasion
3.2.2. Role of Myeloid Derived Suppressor Cells in Melanoma Immune Evasion
3.2.3. Tumor Associated Macrophages in Melanoma Immune Evasion
3.3. Defective Immune Recognition of Melanomas by the Immune System
3.4. Spontaneous Melanoma-Prone Mouse Model Mimics Immune Dysfunction in Humans
3.5. Ultraviolet (UV) Radiation-Induced Immune Suppression in Melanomagenesis
3.6. Exosomes
4. Understanding Melanoma Subtype Etiology and Biology to Better Treat Patients with Immunotherapies: Identification of Patient Biomarkers, Characteristics, and Combination Therapies to Improve Response Rates and Survival
4.1. Immune Checkpoint Blockade Therapy
Patient Characteristics that can Improve Response Rates to Immune Checkpoint Blockade Therapy
4.2. Adoptive T Cell Therapy
4.2.1. Tumor Infiltrating Lymphocyte Therapy
4.2.2. Engineered TCR Therapy
4.2.3. Chimeric Antigen Receptor Therapy
4.2.4. Patient Characteristics that can Improve Response Rates to Adoptive T cell Therapy
4.3. Oncolytic Viruses for the Treatment of Malignant Melanoma
5. Mechanistic Driven Design of Combination Therapies with Immunotherapy
6. Conclusions and Future Directions
Author Contributions
Funding
Conflicts of Interest
Abbreviations
ABCDEF | Asymmetry, Border irregularity, Color variegation, Diameter larger than 6 mm, Evolution of a lesion, Funny looking |
TNM | Tumor thickness, Nodal Involvement, Metastasis |
CSID | Chronically Sun Induced Melanoma |
Non-CSID | Non- Chronically Sun Induced Melanoma |
mGluR | Metabotropic Glutamate Receptor |
PD-1 | Program Cell Death Protein 1 |
PD-L1 | Programmed Death Ligand 1 |
PD-L2 | Programmed Death Ligand 2 |
CTLA-4 | Cytotoxic T-Lymphocyte Associated Protein- 4 |
Tregs | Regulatory T-cells |
MDSC | Myeloid Derived Suppressor Cells |
TAMs | Tumor Associated Macrophages |
Th | Helper T-cells |
MHC I | Major Histocompatibility I |
MHC II | Major Histocompatibility II |
IFN | Interferon |
IL-2 | Interleukin-2 |
BCG | Bacillus-Calmette-Guerin |
RECIST | Response Evaluation Criteria in Solid Tumors |
iRECIST | Immune related Response Evaluation Criteria in Solid Tumors |
iCR | Immune Complete Response |
iPR | Immune Partial Response |
iSD | Immune Stable Disease |
iUPD | Immune Unconfirmed Progressive Disease |
iCPD | Immune Confirmed Progressive Disease |
NLR | Neutrophil to Lymphocyte Ratio |
iCAM | Immune Checkpoint Activating Mutation |
CAR | Chimeric Antigen Receptor |
TIL | Tumor Infiltrating Lymphocytes |
T-VEC | Talimogene Laherparepevec |
TCR | T-Cell Receptor |
References
- Mort, R.L.; Jackson, I.J.; Patton, E.E. The melanocyte lineage in development and disease. Development 2015, 142, 620–632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shain, A.H.; Bastian, B.C. From melanocytes to melanomas. Nat. Rev. Cancer 2016, 16, 345–358. [Google Scholar] [CrossRef] [PubMed]
- Seiji, M.; Fitzpatrick, T.B. The Reciprocal Relationship between Melanization and Tyrosinase Activity in Melanosomes (Melanin Granules). J. Biochem. 1961, 49, 700–706. [Google Scholar] [CrossRef] [PubMed]
- Brenner, M.; Hearing, V.J. The Protective Role of Melanin Against UV Damage in Human Skin†. Photochem. Photobiol. 2008, 84, 539–549. [Google Scholar] [CrossRef] [Green Version]
- Isola, A.L.; Eddy, K.; Chen, S. Biology, Therapy and Implications of Tumor Exosomes in the Progression of Melanoma. Cancers 2016, 8, 110. [Google Scholar] [CrossRef] [Green Version]
- Seiji, M.; Fitzpatrick, T.; Birbeck, M. The Melanosome: A Distinctive Subcellular Particle of Mammalian Melanocytes and the Site of Melanogenesis1. J. Investig. Dermatol. 1961, 36, 243–252. [Google Scholar] [CrossRef] [Green Version]
- Fitzpatrick, T.B.; Breathnach, A.S. The Epidermal Melanin Unit System. Dermatol. Wochenschr. 1963, 147, 481–489. [Google Scholar]
- Alaluf, S.; Atkins, D.; Barrett, K.; Blount, M.; Carter, N.; Heath, A. Ethnic Variation in Melanin Content and Composition in Photoexposed and Photoprotected Human Skin. Pigment. Cell Res. 2002, 15, 112–118. [Google Scholar] [CrossRef]
- Crawford, N.; Kelly, D.E.; Hansen, M.E.B.; Beltrame, M.H.; Fan, S.; Bowman, S.L.; Jewett, E.; Ranciaro, A.; Thompson, S.; Lo, Y.; et al. Loci associated with skin pigmentation identified in African populations. Science 2017, 358, eaan8433. [Google Scholar] [CrossRef] [Green Version]
- Liu, F.; Visser, M.; Duffy, D.L.; Hysi, P.G.; Jacobs, L.C.; Lao, O.; Zhong, K.; Walsh, S.; Chaitanya, L.; Wollstein, A.; et al. Genetics of skin color variation in Europeans: Genome-wide association studies with functional follow-up. Hum. Genet. 2015, 134, 823–835. [Google Scholar] [CrossRef] [Green Version]
- Sturm, R.A. Molecular genetics of human pigmentation diversity. Hum. Mol. Genet. 2009, 18, R9–R17. [Google Scholar] [CrossRef] [PubMed]
- Thody, A.J.; Higgins, E.M.; Wakamatsu, K.; Ito, S.; Burchill, S.A.; Marks, J.M. Pheomelanin as well as Eumelanin Is Present in Human Epidermis. J. Investig. Dermatol. 1991, 97, 340–344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hennessy, A.; Oh, C.; Diffey, B.; Wakamatsu, K.; Ito, S.; Rees, J. Eumelanin and pheomelanin concentrations in human epidermis before and after UVB irradiation. Pigment Cell Res. 2005, 18, 220–223. [Google Scholar] [CrossRef] [PubMed]
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 7–30. [Google Scholar] [CrossRef] [PubMed]
- Yamaguchi, Y.; Takahashi, K.; Zmudzka, B.Z.; Kornhauser, A.; Miller, S.A.; Tadokoro, T.; Berens, W.; Beer, J.Z.; Hearing, V.J. Human skin responses to UV radiation: Pigment in the upper epidermis protects against DNA damage in the lower epidermis and facilitates apoptosis. FASEB J. 2006, 20, 1486–1488. [Google Scholar] [CrossRef]
- Tadokoro, T.; Kobayashi, N.; Zmudzka, B.Z.; Ito, S.; Wakamatsu, K.; Yamaguchi, Y.; Korossy, K.S.; Miller, S.A.; Beer, J.Z.; Hearing, V.J. UV-induced DNA damage and melanin content in human skin differing in racial/ethnic origin. FASEB J. 2003, 17, 1177–1179. [Google Scholar] [CrossRef] [Green Version]
- Scoggins, C.R.; Ross, M.I.; Reintgen, D.S.; Noyes, R.D.; Goydos, J.S.; Beitsch, P.D.; Urist, M.M.; Ariyan, S.; Sussman, J.J.; Edwards, M.J.; et al. Gender-Related Differences in Outcome for Melanoma Patients. Ann. Surg. 2006, 243, 693–700. [Google Scholar] [CrossRef]
- Shaw, H.M.; McGovern, V.J.; Milton, G.W.; Farago, G.A.; McCarthy, W.H. Histologic features of tumors and the female superiority in survival from malignant melanoma. Cancer 1980, 45, 1604–1608. [Google Scholar] [CrossRef]
- Natale, C.A.; Duperret, E.K.; Zhang, J.; Sadeghi, R.; Dahal, A.; O’Brien, K.T.; Cookson, R.; Winkler, J.D.; Ridky, T.W. Sex steroids regulate skin pigmentation through nonclassical membrane-bound receptors. eLife 2016, 5. [Google Scholar] [CrossRef]
- White, L.P. Studies on Melanoma. N. Engl. J. Med. 1959, 260, 789–797. [Google Scholar] [CrossRef]
- Natale, C.A.; Li, J.; Zhang, J.; Dahal, A.; Dentchev, T.; Stanger, B.Z.; Ridky, T.W. Activation of G protein-coupled estrogen receptor signaling inhibits melanoma and improves response to immune checkpoint blockade. eLife 2018, 7, e31770. [Google Scholar] [CrossRef] [PubMed]
- Wu, X.-C.; Eide, M.J.; King, J.; Saraiya, M.; Huang, Y.; Wiggins, C.; Barnholtz-Sloan, J.S.; Martin, N.; Cokkinides, V.; Miller, J.; et al. Racial and ethnic variations in incidence and survival of cutaneous melanoma in the United States, 1999-2006. J. Am. Acad. Dermatol. 2011, 65, S26.e1–S26.e13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kemeny, M.; Busch, E.; Stewart, A.K.; Menck, H.R. Superior Survival of Young Women with Malignant Melanoma. Am. J. Surg. 1998, 175, 437–445. [Google Scholar] [CrossRef]
- Roh, M.R.; Eliades, P.; Gupta, S.; Grant-Kels, J.M.; Tsao, H. Cutaneous melanoma in women. Int. J. Women’s Dermatol. 2017, 3, S11–S15. [Google Scholar] [CrossRef]
- Shitara, D.; Nascimento, M.M.; Puig, S.; Yamada, S.; Enokihara, M.M.; Michalany, N.; Bagatin, E. Nevus-associated melanomas: Clinicopathologic features. Am. J. Clin. Pathol. 2014, 142, 485–491. [Google Scholar] [CrossRef] [Green Version]
- Elder, D.E.; Bastian, B.C.; Cree, I.A.; Massi, D.; Scolyer, R.A. The 2018 World Health Organization Classification of Cutaneous, Mucosal, and Uveal Melanoma: Detailed Analysis of 9 Distinct Subtypes Defined by Their Evolutionary Pathway. Arch. Pathol. Lab. Med. 2020, 144, 500–522. [Google Scholar] [CrossRef] [Green Version]
- Holly, E.A.; Kelly, J.W.; Shpall, S.N.; Chiu, S.-H. Number of melanocytic nevi as a major risk factor for malignant melanoma. J. Am. Acad. Dermatol. 1987, 17, 459–468. [Google Scholar] [CrossRef]
- Harrison, S.L.; Speare, R.; Wronski, I.; MacLennan, R. Sun exposure and melanocytic naevi in young Australian children. Lancet 1994, 344, 1529–1532. [Google Scholar] [CrossRef]
- Aalborg, J.; Morelli, J.G.; Mokrohisky, S.T.; Asdigian, N.L.; Byers, T.E.; Dellavalle, R.P.; Box, N.F.; Crane, L.A. Tanning and Increased Nevus Development in Very-Light-Skinned Children Without Red Hair. Arch. Dermatol. 2009, 145, 989–996. [Google Scholar] [CrossRef] [Green Version]
- Alexandrov, L.B.; Nik-Zainal, S.; Wedge, D.C.; Aparicio, S.A.J.R.; 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]
- Shain, A.H.; Yeh, I.; Kovalyshyn, I.; Sriharan, A.; Talevich, E.; Gagnon, A.; Dummer, R.; North, J.; Pincus, L.; Ruben, B.; et al. The Genetic Evolution of Melanoma from Precursor Lesions. N. Engl. J. Med. 2015, 373, 1926–1936. [Google Scholar] [CrossRef] [PubMed]
- Elwood, J.M. Melanoma and sun exposure: Contrasts between intermittent and chronic exposure. World J. Surg. 1992, 16, 157–165. [Google Scholar] [CrossRef] [PubMed]
- Matthews, N.H.; Li, W.Q.; Qureshi, A.A.; Weinstock, M.A.; Cho, E. Epidemiology of Melanoma. In Cutaneous Melanoma: Etiology and Therapy; Ward, W.H., Farma, J.M., Eds.; Codon Publications: Brisbane, Australia, 2017. [Google Scholar] [CrossRef]
- Akbani, R.; Akdemir, K.C.; Aksoy, B.A.; Albert, M.; Ally, A.; Amin, S.B.; Arachchi, H.M.; Arora, A.; Auman, J.T.; Ayala, B.; et al. Genomic Classification of Cutaneous Melanoma. Cell 2015, 161, 1681–1696. [Google Scholar] [CrossRef] [PubMed]
- Eckerle Mize, D.; Bishop, M.; Resse, E.; Sluzevich, J. Familial Atypical Multiple Mole Melanoma Syndrome. In Cancer Syndromes; Riegert-Johnson, D.L., Boardman, L.A., Hefferon, T., Roberts, M., Eds.; National Center for Biotechnology Information (US): Bethesda, MD, USA, 2009. [Google Scholar]
- Ford, D.; Bliss, J.M.; Swerdlow, A.J.; Armstrong, B.K.; Franceschi, S.; Green, A.; Holly, E.A.; Mack, T.; MacKie, R.M.; Osterlind, A.; et al. Risk of cutaneous melanoma associated with a family history of the disease. The International Melanoma Analysis Group (IMAGE). Int. J. Cancer 1995, 62, 377–381. [Google Scholar] [CrossRef] [Green Version]
- Hussussian, C.J.; Struewing, J.P.; Goldstein, A.M.; Higgins, P.A.; Ally, D.S.; Sheahan, M.D.; Clark, W.H., Jr.; Tucker, M.A.; Dracopoli, N.C. Germline p16 mutations in familial melanoma. Nat. Genet. 1994, 8, 15–21. [Google Scholar] [CrossRef]
- Kamb, A.; Shattuck-Eidens, D.; Eeles, R.; Liu, Q.; Gruis, N.A.; Ding, W.; Hussey, C.; Tran, T.; Miki, Y.; Weaver-Feldhaus, J.; et al. Analysis of the p16 gene (CDKN2) as a candidate for the chromosome 9p melanoma susceptibility locus. Nat. Genet. 1994, 8, 22–26. [Google Scholar] [CrossRef]
- Zuo, L.; Weger, J.; Yang, Q.; Goldstein, A.M.; Tucker, M.A.; Walker, G.J.; Hayward, N.; Dracopoli, N.C. Germline mutations in the p16INK4a binding domain of CDK4 in familial melanoma. Nat. Genet. 1996, 12, 97–99. [Google Scholar] [CrossRef]
- Soufir, N.; Avril, M.-F.; Chompret, A.; Demenais, F.; Bombled, J.; Spatz, A.; Stoppa-Lyonnet, D.; Bénard, J.; Paillerets, B.B.-D. Prevalence of p16 and CDK4 germline mutations in 48 melanoma-prone families in France. Hum. Mol. Genet. 1998, 7, 209–216. [Google Scholar] [CrossRef] [Green Version]
- Molven, A.; Grimstvedt, M.B.; Steine, S.J.; Harland, M.; Avril, M.-F.; Hayward, N.K.; Akslen, L.A. A large Norwegian family with inherited malignant melanoma, multiple atypical nevi, and CDK4 mutation. Genes Chromosom. Cancer 2005, 44, 10–18. [Google Scholar] [CrossRef]
- Goldstein, A.M.; Chan, M.; Harland, M.; Gillanders, E.M.; Hayward, N.K.; Avril, M.-F.; Azizi, E.; Bianchi-Scarra, G.; Newton-Bishop, J.; Paillerets, B.B.-D.; et al. High-risk Melanoma Susceptibility Genes and Pancreatic Cancer, Neural System Tumors, and Uveal Melanoma across GenoMEL. Cancer Res. 2006, 66, 9818–9828. [Google Scholar] [CrossRef] [Green Version]
- Black, J.O. Xeroderma Pigmentosum. Head Neck Pathol. 2016, 10, 139–144. [Google Scholar] [CrossRef] [Green Version]
- Tagliabue, E.; Gandini, S.; Bellocco, R.; Maisonneuve, P.; Newton-Bishop, J.; Polsky, D.; Lazovich, D.; Kanetsky, P.A.; Ghiorzo, P.; Gruis, N.A.; et al. MC1R variants as melanoma risk factors independent of at-risk phenotypic characteristics: A pooled analysis from the M-SKIP project. Cancer Manag. Res. 2018, 10, 1143–1154. [Google Scholar] [CrossRef] [Green Version]
- Puntervoll, H.E.; Yang, X.R.; Vetti, H.H.; Bachmann, I.M.; Avril, M.F.; Benfodda, M.; Catricalà, C.; Dalle, S.; Duval-Modeste, A.B.; Ghiorzo, P.; et al. Melanoma prone families with CDK4 germline mutation: Phenotypic profile and associations with MC1R variants. J. Med. Genet. 2013, 50, 264–270. [Google Scholar] [CrossRef] [Green Version]
- Daniel Jensen, J.; Elewski, B.E. The ABCDEF Rule: Combining the “ABCDE Rule” and the “Ugly Duckling Sign” in an Effort to Improve Patient Self-Screening Examinations. J. Clin. Aesthet. Dermatol. 2015, 8, 15. [Google Scholar]
- Friedman, R.J.; Rigel, D.S.; Kopf, A.W. Early detection of malignant melanoma: The role of physician examination and self-examination of the skin. CA Cancer J. Clin. 1985, 35, 130–151. [Google Scholar] [CrossRef]
- Abbasi, N.R.; Shaw, H.M.; Rigel, D.S.; Friedman, R.J.; McCarthy, W.H.; Osman, I.; Kopf, A.W.; Polsky, D. Early diagnosis of cutaneous melanoma: Revisiting the ABCD criteria. JAMA 2004, 292, 2771–2776. [Google Scholar] [CrossRef]
- Grob, J.J.; Bonerandi, J.J. The ‘ugly duckling’ sign: Identification of the common characteristics of nevi in an individual as a basis for melanoma screening. Arch. Dermatol. 1998, 134, 103–104. [Google Scholar] [CrossRef] [PubMed]
- Ward, W.H.; Lambreton, F.; Goel, N.; Yu, J.Q.; Farma, J.M. Clinical Presentation and Staging of Melanoma. In Cutaneous Melanoma: Etiology and Therapy; Ward, W.H., Farma, J.M., Eds.; Codon Publications: Brisbane, Australia, 2017. [Google Scholar] [CrossRef]
- Dinnes, J.; Deeks, J.J.; Chuchu, N.; Ferrante di Ruffano, L.; Matin, R.N.; Thomson, D.R.; Wong, K.Y.; Aldridge, R.B.; Abbott, R.; Fawzy, M.; et al. Dermoscopy, with and without visual inspection, for diagnosing melanoma in adults. Cochrane Database Syst. Rev. 2018, 12, CD011902. [Google Scholar] [CrossRef] [PubMed]
- Herschorn, A. Dermoscopy for melanoma detection in family practice. Can. Fam. Physician 2012, 58, 740–745. [Google Scholar] [PubMed]
- Holmes, G.A.; Vassantachart, J.M.; Limone, B.A.; Zumwalt, M.; Hirokane, J.; Jacob, S.E. Using Dermoscopy to Identify Melanoma and Improve Diagnostic Discrimination. Fed. Pract. 2018, 35, S39–S45. [Google Scholar]
- Sonthalia, S.; Kaliyadan, F. Dermoscopy Overview and Extradiagnostic Applications. In StatPearls; StatPearls Publishing LLC.: Treasure Island, FL, USA, 2020. [Google Scholar]
- Davis, L.E.; Shalin, S.C.; Tackett, A.J. Current state of melanoma diagnosis and treatment. Cancer Biol. Ther. 2019, 20, 1366–1379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gershenwald, J.E.; Scolyer, R.A.; Hess, K.R.; Sondak, V.K.; Long, G.V.; Ross, M.I.; Lazar, A.J.; Faries, M.B.; Kirkwood, J.M.; McArthur, G.A.; et al. Melanoma staging: Evidence-based changes in the American Joint Committee on Cancer eighth edition cancer staging manual. CA Cancer J. Clin. 2017, 67, 472–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Breslow, A. Thickness, cross-sectional areas and depth of invasion in the prognosis of cutaneous melanoma. Ann. Surg. 1970, 172, 902–908. [Google Scholar] [CrossRef] [PubMed]
- Bønnelykke-Behrndtz, M.L.; Steiniche, T. Ulcerated Melanoma: Aspects and Prognostic Impact. In Cutaneous Melanoma: Etiology and Therapy; Ward, W.H., Farma, J.M., Eds.; Codon Publications: Brisbane, Australia, 2017. [Google Scholar] [CrossRef] [Green Version]
- Urso, C. Are growth phases exclusive to cutaneous melanoma? J. Clin. Pathol. 2004, 57, 560. [Google Scholar] [CrossRef] [Green Version]
- Clark, W.H., Jr.; Elder, D.E.; Guerry, D.T.; Epstein, M.N.; Greene, M.H.; Van Horn, M. A study of tumor progression: The precursor lesions of superficial spreading and nodular melanoma. Hum. Pathol. 1984, 15, 1147–1165. [Google Scholar] [CrossRef]
- Damsky, W.E.; Rosenbaum, L.E.; Bosenberg, M. Decoding melanoma metastasis. Cancers 2010, 3, 126–163. [Google Scholar] [CrossRef]
- Thompson, J.F.; Soong, S.J.; Balch, C.M.; Gershenwald, J.E.; Ding, S.; Coit, D.G.; Flaherty, K.T.; Gimotty, P.A.; Johnson, T.; Johnson, M.M.; et al. Prognostic significance of mitotic rate in localized primary cutaneous melanoma: An analysis of patients in the multi-institutional American Joint Committee on Cancer melanoma staging database. J. Clin. Oncol. 2011, 29, 2199–2205. [Google Scholar] [CrossRef] [Green Version]
- Wilkins, D.K.; Nathan, P.D. Therapeutic opportunities in noncutaneous melanoma. Ther. Adv. Med. Oncol. 2009, 1, 29–36. [Google Scholar] [CrossRef] [Green Version]
- Curtin, J.A.; Fridlyand, J.; Kageshita, T.; Patel, H.N.; Busam, K.J.; Kutzner, H.; Cho, K.-H.; Aiba, S.; Bröcker, E.-B.; LeBoit, P.E.; et al. Distinct Sets of Genetic Alterations in Melanoma. N. Engl. J. Med. 2005, 353, 2135–2147. [Google Scholar] [CrossRef]
- Kuk, D.; Shoushtari, A.N.; Barker, C.A.; Panageas, K.S.; Munhoz, R.R.; Momtaz, P.; Ariyan, C.E.; Brady, M.S.; Coit, D.G.; Bogatch, K.; et al. Prognosis of Mucosal, Uveal, Acral, Nonacral Cutaneous, and Unknown Primary Melanoma from the Time of First Metastasis. Oncologist 2016, 21, 848–854. [Google Scholar] [CrossRef] [Green Version]
- Bennett, D.R.; Wasson, D.; MacArthur, J.D.; McMillen, M.A. The effect of misdiagnosis and delay in diagnosis on clinical outcome in melanomas of the foot. J. Am. Coll. Surg. 1994, 179, 279–284. [Google Scholar] [PubMed]
- Cress, R.D.; Holly, E.A. Incidence of cutaneous melanoma among non-Hispanic whites, Hispanics, Asians, and blacks: An analysis of california cancer registry data, 1988–1993. Cancer Causes Control 1997, 8, 246–252. [Google Scholar] [CrossRef] [PubMed]
- Cormier, J.N.; Xing, Y.; Ding, M.; Lee, J.E.; Mansfield, P.F.; Gershenwald, J.E.; Ross, M.I.; Du, X.L. Ethnic Differences Among Patients with Cutaneous Melanoma. Arch. Intern. Med. 2006, 166, 1907–1914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hayward, N.K.; Wilmott, J.S.; Waddell, N.; Johansson, P.A.; Field, M.A.; Nones, K.; Patch, A.-M.; Kakavand, H.; Alexandrov, L.B.; Burke, H.; et al. Whole-genome landscapes of major melanoma subtypes. Nature 2017, 545, 175–180. [Google Scholar] [CrossRef] [PubMed]
- Krauthammer, M.; Kong, Y.; Ha, B.H.; Evans, P.; Bacchiocchi, A.; McCusker, J.P.; Cheng, E.; Davis, M.J.; Goh, G.; Choi, M.; et al. Exome sequencing identifies recurrent somatic RAC1 mutations in melanoma. Nat. Genet. 2012, 44, 1006–1014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pollock, P.M.; Harper, U.L.; Hansen, K.S.; Yudt, L.M.; Stark, M.; Robbins, C.M.; Moses, T.Y.; Hostetter, G.; Wagner, U.; Kakareka, J.; et al. High frequency of BRAF mutations in nevi. Nat. Genet. 2003, 33, 19–20. [Google Scholar] [CrossRef]
- Menzies, A.M.; Haydu, L.E.; Visintin, L.; Carlino, M.S.; Howle, J.R.; Thompson, J.F.; Kefford, R.F.; Scolyer, R.A.; Long, G.V. Distinguishing clinicopathologic features of patients with V600E and V600K BRAF-mutant metastatic melanoma. Clin. Cancer Res. 2012, 18, 3242–3249. [Google Scholar] [CrossRef] [Green Version]
- Teh, J.L.; Chen, S. Glutamatergic signaling in cellular transformation. Pigment Cell Melanoma Res. 2012, 25, 331–342. [Google Scholar] [CrossRef]
- Horn, S.; Figl, A.; Rachakonda, P.S.; Fischer, C.; Sucker, A.; Gast, A.; Kadel, S.; Moll, I.; Nagore, E.; Hemminki, K.; et al. TERT promoter mutations in familial and sporadic melanoma. Science 2013, 339, 959–961. [Google Scholar] [CrossRef] [Green Version]
- Chiba, K.; Lorbeer, F.K.; Shain, A.H.; McSwiggen, D.T.; Schruf, E.; Oh, A.; Ryu, J.; Darzacq, X.; Bastian, B.C.; Hockemeyer, D. Mutations in the promoter of the telomerase gene TERT contribute to tumorigenesis by a two-step mechanism. Science 2017, 357, 1416–1420. [Google Scholar] [CrossRef] [Green Version]
- Bradford, P.T.; Goldstein, A.M.; McMaster, M.L.; Tucker, M.A. Acral lentiginous melanoma: Incidence and survival patterns in the United States, 1986–2005. Arch. Dermatol. 2009, 145, 427–434. [Google Scholar] [CrossRef] [PubMed]
- Weis, E.; Shah, C.P.; Lajous, M.; Shields, J.A.; Shields, C.L. The Association Between Host Susceptibility Factors and Uveal Melanoma: A Meta-analysis. Arch. Ophthalmol. 2006, 124, 54–60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shah, C.P.; Weis, E.; Lajous, M.; Shields, J.A.; Shields, C.L. Intermittent and Chronic Ultraviolet Light Exposure and Uveal Melanoma: A Meta-analysis. Ophthalmology 2005, 112, 1599–1607. [Google Scholar] [CrossRef] [PubMed]
- Kaliki, S.; Shields, C.L. Uveal melanoma: Relatively rare but deadly cancer. Eye 2017, 31, 241–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McLaughlin, C.C.; Wu, X.C.; Jemal, A.; Martin, H.J.; Roche, L.M.; Chen, V.W. Incidence of noncutaneous melanomas in the U.S. Cancer 2005, 103, 1000–1007. [Google Scholar] [CrossRef]
- Rossi, E.; Schinzari, G.; Zizzari, I.G.; Maiorano, B.A.; Pagliara, M.M.; Sammarco, M.G.; Fiorentino, V.; Petrone, G.; Cassano, A.; Rindi, G.; et al. Immunological Backbone of Uveal Melanoma: Is There a Rationale for Immunotherapy? Cancers 2019, 11, 1055. [Google Scholar] [CrossRef] [Green Version]
- Rabbie, R.; Ferguson, P.; Molina-Aguilar, C.; Adams, D.J.; Robles-Espinoza, C.D. Melanoma subtypes: Genomic profiles, prognostic molecular markers and therapeutic possibilities. J. Pathol. 2019, 247, 539–551. [Google Scholar] [CrossRef]
- Johansson, P.; Aoude, L.G.; Wadt, K.; Glasson, W.J.; Warrier, S.K.; Hewitt, A.W.; Kiilgaard, J.F.; Heegaard, S.; Isaacs, T.; Franchina, M.; et al. Deep sequencing of uveal melanoma identifies a recurrent mutation in PLCB4. Oncotarget 2016, 7, 4624–4631. [Google Scholar] [CrossRef] [Green Version]
- Van Raamsdonk, C.D.; Griewank, K.G.; Crosby, M.B.; Garrido, M.C.; Vemula, S.; Wiesner, T.; Obenauf, A.C.; Wackernagel, W.; Green, G.; Bouvier, N.; et al. Mutations in GNA11 in uveal melanoma. N. Engl. J. Med. 2010, 363, 2191–2199. [Google Scholar] [CrossRef] [Green Version]
- Moore, A.R.; Ceraudo, E.; Sher, J.J.; Guan, Y.; Shoushtari, A.N.; Chang, M.T.; Zhang, J.Q.; Walczak, E.G.; Kazmi, M.A.; Taylor, B.S.; et al. Recurrent activating mutations of G-protein-coupled receptor CYSLTR2 in uveal melanoma. Nat. Genet. 2016, 48, 675–680. [Google Scholar] [CrossRef] [Green Version]
- Van Raamsdonk, C.D.; Bezrookove, V.; Green, G.; Bauer, J.; Gaugler, L.; O’Brien, J.M.; Simpson, E.M.; Barsh, G.S.; Bastian, B.C. Frequent somatic mutations of GNAQ in uveal melanoma and blue naevi. Nature 2009, 457, 599–602. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Khalili, J.S.; Yu, X.; Wang, J.; Hayes, B.C.; Davies, M.A.; Lizee, G.; Esmaeli, B.; Woodman, S.E. Combination small molecule MEK and PI3K inhibition enhances uveal melanoma cell death in a mutant GNAQ- and GNA11-dependent manner. Clin. Cancer Res. 2012, 18, 4345–4355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, F.X.; Luo, J.; Mo, J.S.; Liu, G.; Kim, Y.C.; Meng, Z.; Zhao, L.; Peyman, G.; Ouyang, H.; Jiang, W.; et al. Mutant Gq/11 promote uveal melanoma tumorigenesis by activating YAP. Cancer Cell 2014, 25, 822–830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Feng, X.; Degese, M.S.; Iglesias-Bartolome, R.; Vaque, J.P.; Molinolo, A.A.; Rodrigues, M.; Zaidi, M.R.; Ksander, B.R.; Merlino, G.; Sodhi, A.; et al. Hippo-independent activation of YAP by the GNAQ uveal melanoma oncogene through a trio-regulated rho GTPase signaling circuitry. Cancer Cell 2014, 25, 831–845. [Google Scholar] [CrossRef] [Green Version]
- Toshiaki, S.H.K.; Yoriko, Y.; Masaru, T. Acral Melanoma In Color Atlas of Melanocytic Lesions of the Skin; Soyer, H.P., Argenziano, G., Hofmann-Wellenhof, R., Johr, R.H., Eds.; Springer: Berlin, Germany; New York, NY, USA, 2007; Volume 1, pp. 196–203. [Google Scholar]
- Huang, K.; Fan, J.; Misra, S. Acral Lentiginous Melanoma: Incidence and Survival in the United States, 2006–2015, an Analysis of the SEER Registry. J. Surg. Res. 2020, 251, 329–339. [Google Scholar] [CrossRef]
- Jung, H.J.; Kweon, S.-S.; Lee, J.-B.; Lee, S.-C.; Yun, S.J. A Clinicopathologic Analysis of 177 Acral Melanomas in Koreans: Relevance of Spreading Pattern and Physical Stress. JAMA Dermatol. 2013, 149, 1281–1288. [Google Scholar] [CrossRef] [Green Version]
- Feibleman, C.E.; Stoll, H.; Maize, J.C. Melanomas of the palm, sole, and nailbed: A clinicopathologic study. Cancer 1980, 46, 2492–2504. [Google Scholar] [CrossRef]
- Moon, K.R.; Choi, Y.D.; Kim, J.M.; Jin, S.; Shin, M.H.; Shim, H.J.; Lee, J.B.; Yun, S.J. Genetic Alterations in Primary Acral Melanoma and Acral Melanocytic Nevus in Korea: Common Mutated Genes Show Distinct Cytomorphological Features. J. Investig. Dermatol. 2018, 138, 933–945. [Google Scholar] [CrossRef] [Green Version]
- Liang, W.S.; Hendricks, W.; Kiefer, J.; Schmidt, J.; Sekar, S.; Carpten, J.; Craig, D.W.; Adkins, J.; Cuyugan, L.; Manojlovic, Z.; et al. Integrated genomic analyses reveal frequent TERT aberrations in acral melanoma. Genome Res. 2017, 27, 524–532. [Google Scholar] [CrossRef] [Green Version]
- De Lima Vazquez, V.; Vicente, A.L.; Carloni, A.; Berardinelli, G.; Soares, P.; Scapulatempo, C.; Martinho, O.; Reis, R.M. Molecular profiling, including TERT promoter mutations, of acral lentiginous melanomas. Melanoma Res. 2016, 26, 93–99. [Google Scholar] [CrossRef] [Green Version]
- Curtin, J.A.; Busam, K.; Pinkel, D.; Bastian, B.C. Somatic activation of KIT in distinct subtypes of melanoma. J. Clin. Oncol. 2006, 24, 4340–4346. [Google Scholar] [CrossRef] [PubMed]
- Mihajlovic, M.; Vlajkovic, S.; Jovanovic, P.; Stefanovic, V. Primary mucosal melanomas: A comprehensive review. Int. J. Clin. Exp. Pathol. 2012, 5, 739–753. [Google Scholar]
- Altieri, L.; Eguchi, M.; Peng, D.H.; Cockburn, M. Predictors of mucosal melanoma survival in a population-based setting. J. Am. Acad. Dermatol. 2019, 81, 136–142.e132. [Google Scholar] [CrossRef] [PubMed]
- Lerner, B.A.; Stewart, L.A.; Horowitz, D.P.; Carvajal, R.D. Mucosal Melanoma: New Insights and Therapeutic Options for a Unique and Aggressive Disease. Oncology 2017, 31, e23–e32. [Google Scholar] [PubMed]
- Kabbarah, O.; Chin, L. Revealing the genomic heterogeneity of melanoma. Cancer Cell 2005, 8, 439–441. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hintzsche, J.D.; Gorden, N.T.; Amato, C.M.; Kim, J.; Wuensch, K.E.; Robinson, S.E.; Applegate, A.J.; Couts, K.L.; Medina, T.M.; Wells, K.R.; et al. Whole-exome sequencing identifies recurrent SF3B1 R625 mutation and comutation of NF1 and KIT in mucosal melanoma. Melanoma Res. 2017, 27, 189–199. [Google Scholar] [CrossRef] [PubMed]
- Furney, S.J.; Turajlic, S.; Stamp, G.; Nohadani, M.; Carlisle, A.; Thomas, J.M.; Hayes, A.; Strauss, D.; Gore, M.; van den Oord, J.; et al. Genome sequencing of mucosal melanomas reveals that they are driven by distinct mechanisms from cutaneous melanoma. J. Pathol. 2013, 230, 261–269. [Google Scholar] [CrossRef]
- Sheng, X.; Kong, Y.; Li, Y.; Zhang, Q.; Si, L.; Cui, C.; Chi, Z.; Tang, B.; Mao, L.; Lian, B.; et al. GNAQ and GNA11 mutations occur in 9.5% of mucosal melanoma and are associated with poor prognosis. Eur. J. Cancer 2016, 65, 156–163. [Google Scholar] [CrossRef]
- Si, L.; Wang, X.; Guo, J. Genotyping of mucosal melanoma. Chin. Clin. Oncol. 2014, 3, 34. [Google Scholar] [CrossRef]
- Chen, S.; Zhu, H.; Wetzel, W.J.; Philbert, M.A. Spontaneous melanocytosis in transgenic mice. J. Investig. Dermatol. 1996, 106, 1145–1151. [Google Scholar] [CrossRef] [Green Version]
- Zhu, H.; Reuhl, K.; Zhang, X.; Botha, R.; Ryan, K.; Wei, J.; Chen, S. Development of heritable melanoma in transgenic mice. J. Investig. Dermatol. 1998, 110, 247–252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pollock, P.M.; Cohen-Solal, K.; Sood, R.; Namkoong, J.; Martino, J.J.; Koganti, A.; Zhu, H.; Robbins, C.; Makalowska, I.; Shin, S.S.; et al. Melanoma mouse model implicates metabotropic glutamate signaling in melanocytic neoplasia. Nat. Genet. 2003, 34, 108–112. [Google Scholar] [CrossRef] [PubMed]
- Ohtani, Y.; Harada, T.; Funasaka, Y.; Nakao, K.; Takahara, C.; Abdel-Daim, M.; Sakai, N.; Saito, N.; Nishigori, C.; Aiba, A. Metabotropic glutamate receptor subtype-1 is essential for in vivo growth of melanoma. Oncogene 2008, 27, 7162–7170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wangari-Talbot, J.; Wall, B.A.; Goydos, J.S.; Chen, S. Functional effects of GRM1 suppression in human melanoma cells. Mol. Cancer Res. 2012, 10, 1440–1450. [Google Scholar] [CrossRef] [Green Version]
- Zhu, H.; Reuhl, K.; Botha, R.; Ryan, K.; Wei, J.; Chen, S. Development of early melanocytic lesions in transgenic mice predisposed to melanoma. Pigment Cell Res. 2000, 13, 158–164. [Google Scholar] [CrossRef]
- Shin, S.S.; Namkoong, J.; Wall, B.A.; Gleason, R.; Lee, H.J.; Chen, S. Oncogenic activities of metabotropic glutamate receptor 1 (Grm1) in melanocyte transformation. Pigment. Cell Melanoma Res. 2008, 21, 368–378. [Google Scholar] [CrossRef]
- Namkoong, J.; Shin, S.S.; Lee, H.J.; Marin, Y.E.; Wall, B.A.; Goydos, J.S.; Chen, S. Metabotropic glutamate receptor 1 and glutamate signaling in human melanoma. Cancer Res. 2007, 67, 2298–2305. [Google Scholar] [CrossRef] [Green Version]
- Shin, S.S.; Wall, B.A.; Goydos, J.S.; Chen, S. AKT2 is a downstream target of metabotropic glutamate receptor 1 (Grm1). Pigment. Cell Melanoma Res. 2010, 23, 103–111. [Google Scholar] [CrossRef] [Green Version]
- Marín, Y.E.; Namkoong, J.; Cohen-Solal, K.; Shin, S.S.; Martino, J.J.; Oka, M.; Chen, S. Stimulation of oncogenic metabotropic glutamate receptor 1 in melanoma cells activates ERK1/2 via PKCepsilon. Cell. Signal. 2006, 18, 1279–1286. [Google Scholar] [CrossRef]
- Rutkowski, P.; Zdzienicki, M.; Nowecki, Z.I.; Van Akkooi, A.C. Surgery of primary melanomas. Cancers 2010, 2, 824–841. [Google Scholar] [CrossRef]
- Tyrell, R.; Antia, C.; Stanley, S.; Deutsch, G.B. Surgical resection of metastatic melanoma in the era of immunotherapy and targeted therapy. Melanoma Manag. 2017, 4, 61–68. [Google Scholar] [CrossRef] [PubMed]
- Leung, A.M.; Hari, D.M.; Morton, D.L. Surgery for distant melanoma metastasis. Cancer J. 2012, 18, 176–184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, D.; Dong, Y.; Sun, X.; Yuan, S.; Yu, J. Surgery of primary tumor improves the survival of newly diagnosed metastatic melanoma: A population-based, propensity-matched study. Cancer Manag. Res. 2019, 11, 339–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gannon, C.J.; Rousseau, D.L., Jr.; Ross, M.I.; Johnson, M.M.; Lee, J.E.; Mansfield, P.F.; Cormier, J.N.; Prieto, V.G.; Gershenwald, J.E. Accuracy of lymphatic mapping and sentinel lymph node biopsy after previous wide local excision in patients with primary melanoma. Cancer 2006, 107, 2647–2652. [Google Scholar] [CrossRef] [PubMed]
- Swetter, S.M.; Tsao, H.; Bichakjian, C.K.; Curiel-Lewandrowski, C.; Elder, D.E.; Gershenwald, J.E.; Guild, V.; Grant-Kels, J.M.; Halpern, A.C.; Johnson, T.M.; et al. Guidelines of care for the management of primary cutaneous melanoma. J. Am. Acad. Dermatol. 2019, 80, 208–250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Smyth, E.C.; Flavin, M.; Pulitzer, M.P.; Gardner, G.J.; Costantino, P.D.; Chi, D.S.; Bogatch, K.; Chapman, P.B.; Wolchok, J.D.; Schwartz, G.K.; et al. Treatment of locally recurrent mucosal melanoma with topical imiquimod. J. Clin. Oncol. 2011, 29, e809–e811. [Google Scholar] [CrossRef] [PubMed]
- Turza, K.; Dengel, L.T.; Harris, R.C.; Patterson, J.W.; White, K.; Grosh, W.W.; Slingluff, C.L., Jr. Effectiveness of imiquimod limited to dermal melanoma metastases, with simultaneous resistance of subcutaneous metastasis. J. Cutan. Pathol. 2010, 37, 94–98. [Google Scholar] [CrossRef] [Green Version]
- Nanda, J.; Bermudez, R. Imiquimod. In StatPearls; StatPearls Publishing LLC.: Treasure Island, FL, USA, 2020. [Google Scholar]
- Schön, M.; Bong, A.B.; Drewniok, C.; Herz, J.; Geilen, C.C.; Reifenberger, J.; Benninghoff, B.; Slade, H.B.; Gollnick, H.; Schön, M.P. Tumor-selective induction of apoptosis and the small-molecule immune response modifier imiquimod. J. Natl. Cancer Inst. 2003, 95, 1138–1149. [Google Scholar] [CrossRef] [Green Version]
- Shi, W. Radiation Therapy for Melanoma. In Cutaneous Melanoma: Etiology and Therapy; Ward, W.H., Farma, J.M., Eds.; Codon Publication: Brisbane, Australia, 2017. [Google Scholar] [CrossRef] [Green Version]
- Strojan, P. Role of radiotherapy in melanoma management. Radiol. Oncol. 2010, 44, 1–12. [Google Scholar] [CrossRef]
- 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]
- D’Andrea, M.A.; Reddy, G.K. Systemic Antitumor Effects and Abscopal Responses in Melanoma Patients Receiving Radiation Therapy. Oncology 2020, 98, 202–215. [Google Scholar] [CrossRef] [PubMed]
- Mole, R.H. Whole body irradiation; radiobiology or medicine? Br. J. Radiol. 1953, 26, 234–241. [Google Scholar] [CrossRef] [PubMed]
- Lo, J.A.; Fisher, D.E. The melanoma revolution: From UV carcinogenesis to a new era in therapeutics. Science 2014, 346, 945–949. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wilson, M.A.; Schuchter, L.M. Chemotherapy for Melanoma. Cancer Treat. Res. 2016, 167, 209–229. [Google Scholar] [CrossRef]
- Chang, W.; Lee, S.J.; Park, S.; Choi, M.K.; Hong, J.Y.; Kim, Y.S.; Maeng, C.H.; Jung, H.A.; Kim, S.; Lee, J. Effect of paclitaxel/carboplatin salvage chemotherapy in noncutaneous versus cutaneous metastatic melanoma. Melanoma Res. 2013, 23, 147–151. [Google Scholar] [CrossRef]
- Vakifahmetoglu, H.; Olsson, M.; Zhivotovsky, B. Death through a tragedy: Mitotic catastrophe. Cell Death Differ. 2008, 15, 1153–1162. [Google Scholar] [CrossRef] [Green Version]
- Morse, D.L.; Gray, H.; Payne, C.M.; Gillies, R.J. Docetaxel induces cell death through mitotic catastrophe in human breast cancer cells. Mol. Cancer Ther. 2005, 4, 1495–1504. [Google Scholar] [CrossRef] [Green Version]
- Guerriero, J.L.; Ditsworth, D.; Fan, Y.; Zhao, F.; Crawford, H.C.; Zong, W.X. Chemotherapy induces tumor clearance independent of apoptosis. Cancer Res. 2008, 68, 9595–9600. [Google Scholar] [CrossRef] [Green Version]
- Ricci, M.S.; Zong, W.X. Chemotherapeutic approaches for targeting cell death pathways. Oncologist 2006, 11, 342–357. [Google Scholar] [CrossRef] [Green Version]
- Shtivelman, E.; Davies, M.Q.; Hwu, P.; Yang, J.; Lotem, M.; Oren, M.; Flaherty, K.T.; Fisher, D.E. Pathways and therapeutic targets in melanoma. Oncotarget 2014, 5, 1701–1752. [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]
- Tsai, J.; Lee, J.T.; Wang, W.; Zhang, J.; Cho, H.; Mamo, S.; Bremer, R.; Gillette, S.; Kong, J.; Haass, N.K.; et al. Discovery of a selective inhibitor of oncogenic B-Raf kinase with potent antimelanoma activity. Proc. Natl. Acad. Sci. USA 2008, 105, 3041–3046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Søndergaard, J.N.; Nazarian, R.; Wang, Q.; Guo, D.; Hsueh, T.; Mok, S.; Sazegar, H.; MacConaill, L.E.; Barretina, J.G.; Kehoe, S.M.; et al. Differential sensitivity of melanoma cell lines with BRAFV600E mutation to the specific Raf inhibitor PLX4032. J. Transl. Med. 2010, 8, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Flaherty, K.T.; Puzanov, I.; Kim, K.B.; Ribas, A.; McArthur, G.A.; Sosman, J.A.; O’Dwyer, P.J.; Lee, R.J.; Grippo, J.F.; Nolop, K.; et al. Inhibition of mutated, activated BRAF in metastatic melanoma. N. Engl. J. Med. 2010, 363, 809–819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Falchook, G.S.; Long, G.V.; Kurzrock, R.; Kim, K.B.; Arkenau, T.H.; Brown, M.P.; Hamid, O.; Infante, J.R.; Millward, M.; Pavlick, A.C.; et al. Dabrafenib in patients with melanoma, untreated brain metastases, and other solid tumours: A phase 1 dose-escalation trial. Lancet 2012, 379, 1893–1901. [Google Scholar] [CrossRef] [Green Version]
- Ascierto, P.A.; Minor, D.; Ribas, A.; Lebbe, C.; O’Hagan, A.; Arya, N.; Guckert, M.; Schadendorf, D.; Kefford, R.F.; Grob, J.J.; et al. Phase II trial (BREAK-2) of the BRAF inhibitor dabrafenib (GSK2118436) in patients with metastatic melanoma. J. Clin. Oncol. 2013, 31, 3205–3211. [Google Scholar] [CrossRef]
- 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]
- Falchook, G.S.; Lewis, K.D.; Infante, J.R.; Gordon, M.S.; Vogelzang, N.J.; DeMarini, D.J.; Sun, P.; Moy, C.; Szabo, S.A.; Roadcap, L.T.; et al. Activity of the oral MEK inhibitor trametinib in patients with advanced melanoma: A phase 1 dose-escalation trial. Lancet Oncol. 2012, 13, 782–789. [Google Scholar] [CrossRef] [Green Version]
- Flaherty, K.T.; Robert, C.; Hersey, P.; Nathan, P.; Garbe, C.; Milhem, M.; Demidov, L.V.; Hassel, J.C.; Rutkowski, P.; Mohr, P.; et al. Improved survival with MEK inhibition in BRAF-mutated melanoma. N. Engl. J. Med. 2012, 367, 107–114. [Google Scholar] [CrossRef] [Green Version]
- Eroglu, Z.; Ribas, A. Combination therapy with BRAF and MEK inhibitors for melanoma: Latest evidence and place in therapy. Ther. Adv. Med. Oncol. 2016, 8, 48–56. [Google Scholar] [CrossRef] [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]
- 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. Combined BRAF and MEK inhibition versus BRAF inhibition alone in melanoma. N. Engl. J. Med. 2014, 371, 1877–1888. [Google Scholar] [CrossRef] [PubMed] [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] [PubMed] [Green Version]
- Larkin, J.; Ascierto, P.A.; Dréno, B.; Atkinson, V.; Liszkay, G.; Maio, M.; Mandalà, M.; Demidov, L.; Stroyakovskiy, D.; Thomas, L.; et al. Combined vemurafenib and cobimetinib in BRAF-mutated melanoma. N. Engl. J. Med. 2014, 371, 1867–1876. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- 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]
- Dummer, R.; Ascierto, P.A.; Gogas, H.J.; Arance, A.; Mandala, M.; Liszkay, G.; Garbe, C.; Schadendorf, D.; Krajsova, I.; Gutzmer, R.; et al. Overall survival in patients with BRAF-mutant melanoma receiving encorafenib plus binimetinib versus vemurafenib or encorafenib (COLUMBUS): A multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2018, 19, 1315–1327. [Google Scholar] [CrossRef]
- Beadling, C.; Jacobson-Dunlop, E.; Hodi, F.S.; Le, C.; Warrick, A.; Patterson, J.; Town, A.; Harlow, A.; Cruz, F., III; Azar, S.; et al. KIT gene mutations and copy number in melanoma subtypes. Clin. Cancer Res. 2008, 14, 6821–6828. [Google Scholar] [CrossRef] [Green Version]
- Lebbe, C.; Chevret, S.; Jouary, T.; Dalac, S.; Dalle, S.; Guillot, B.; Arnault, J.-P.; Avril, M.-F.; Bedane, C.; Bens, G.; et al. Phase II multicentric uncontrolled national trial assessing the efficacy of nilotinib in the treatment of advanced melanomas with c-KIT mutation or amplification. J. Clin. Oncol. 2014, 32, 9032. [Google Scholar] [CrossRef]
- Hodi, F.S.; Corless, C.L.; Giobbie-Hurder, A.; Fletcher, J.A.; Zhu, M.; Marino-Enriquez, A.; Friedlander, P.; Gonzalez, R.; Weber, J.S.; Gajewski, T.F.; et al. Imatinib for melanomas harboring mutationally activated or amplified KIT arising on mucosal, acral, and chronically sun-damaged skin. J. Clin. Oncol. 2013, 31, 3182–3190. [Google Scholar] [CrossRef] [Green Version]
- Guo, J.; Carvajal, R.D.; Dummer, R.; Hauschild, A.; Daud, A.; Bastian, B.C.; Markovic, S.N.; Queirolo, P.; Arance, A.; Berking, C.; et al. Efficacy and safety of nilotinib in patients with KIT-mutated metastatic or inoperable melanoma: Final results from the global, single-arm, phase II TEAM trial. Ann. Oncol. 2017, 28, 1380–1387. [Google Scholar] [CrossRef] [PubMed]
- Slipicevic, A.; Herlyn, M. KIT in melanoma: Many shades of gray. J. Investig. Dermatol. 2015, 135, 337–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dahl, C.; Abildgaard, C.; Riber-Hansen, R.; Steiniche, T.; Lade-Keller, J.; Guldberg, P. KIT is a frequent target for epigenetic silencing in cutaneous melanoma. J. Investig. Dermatol. 2015, 135, 516–524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shen, L.; Shi, Q.; Wang, W. Double agents: Genes with both oncogenic and tumor-suppressor functions. Oncogenesis 2018, 7, 25. [Google Scholar] [CrossRef]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
- Nishimura, H.; Nose, M.; Hiai, H.; Minato, N.; Honjo, T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity 1999, 11, 141–151. [Google Scholar] [CrossRef] [Green Version]
- Freeman, G.J.; Long, A.J.; Iwai, Y.; Bourque, K.; Chernova, T.; Nishimura, H.; Fitz, L.J.; Malenkovich, N.; Okazaki, T.; Byrne, M.C.; et al. Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J. Exp. Med. 2000, 192, 1027–1034. [Google Scholar] [CrossRef] [Green Version]
- Nishimura, H.; Honjo, T. PD-1: An inhibitory immunoreceptor involved in peripheral tolerance. Trends Immunol. 2001, 22, 265–268. [Google Scholar] [CrossRef]
- Nishimura, H.; Okazaki, T.; Tanaka, Y.; Nakatani, K.; Hara, M.; Matsumori, A.; Sasayama, S.; Mizoguchi, A.; Hiai, H.; Minato, N.; et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 2001, 291, 319–322. [Google Scholar] [CrossRef]
- Barber, D.L.; Wherry, E.J.; Masopust, D.; Zhu, B.; Allison, J.P.; Sharpe, A.H.; Freeman, G.J.; Ahmed, R. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 2006, 439, 682–687. [Google Scholar] [CrossRef]
- Chemnitz, J.M.; Parry, R.V.; Nichols, K.E.; June, C.H.; Riley, J.L. SHP-1 and SHP-2 associate with immunoreceptor tyrosine-based switch motif of programmed death 1 upon primary human T cell stimulation, but only receptor ligation prevents T cell activation. J. Immunol. 2004, 173, 945–954. [Google Scholar] [CrossRef] [PubMed]
- Blackburn, S.D.; Shin, H.; Haining, W.N.; Zou, T.; Workman, C.J.; Polley, A.; Betts, M.R.; Freeman, G.J.; Vignali, D.A.; Wherry, E.J. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat. Immunol. 2009, 10, 29–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baumeister, S.H.; Freeman, G.J.; Dranoff, G.; Sharpe, A.H. Coinhibitory Pathways in Immunotherapy for Cancer. Annu. Rev. Immunol. 2016, 34, 539–573. [Google Scholar] [CrossRef] [PubMed]
- Ribas, A.; Wolchok, J.D. Cancer immunotherapy using checkpoint blockade. Science 2018, 359, 1350–1355. [Google Scholar] [CrossRef] [Green Version]
- Latchman, Y.; Wood, C.R.; Chernova, T.; Chaudhary, D.; Borde, M.; Chernova, I.; Iwai, Y.; Long, A.J.; Brown, J.A.; Nunes, R.; et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat. Immunol. 2001, 2, 261–268. [Google Scholar] [CrossRef]
- Ghoneim, H.E.; Fan, Y.; Moustaki, A.; Abdelsamed, H.A.; Dash, P.; Dogra, P.; Carter, R.; Awad, W.; Neale, G.; Thomas, P.G.; et al. De Novo Epigenetic Programs Inhibit PD-1 Blockade-Mediated T Cell Rejuvenation. Cell 2017, 170, 142–157. [Google Scholar] [CrossRef] [Green Version]
- Pauken, K.E.; Sammons, M.A.; Odorizzi, P.M.; Manne, S.; Godec, J.; Khan, O.; Drake, A.M.; Chen, Z.; Sen, D.R.; Kurachi, M.; et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 2016, 354, 1160–1165. [Google Scholar] [CrossRef] [Green Version]
- Pereira, R.M.; Hogan, P.G.; Rao, A.; Martinez, G.J. Transcriptional and epigenetic regulation of T cell hyporesponsiveness. J. Leukoc. Biol. 2017, 102, 601–615. [Google Scholar] [CrossRef] [Green Version]
- Dong, W.; Wu, X.; Ma, S.; Wang, Y.; Nalin, A.P.; Zhu, Z.; Zhang, J.; Benson, D.M.; He, K.; Caligiuri, M.A.; et al. The Mechanism of Anti-PD-L1 Antibody Efficacy against PD-L1-Negative Tumors Identifies NK Cells Expressing PD-L1 as a Cytolytic Effector. Cancer Discov. 2019, 9, 1422–1437. [Google Scholar] [CrossRef] [Green Version]
- Hartley, G.; Regan, D.; Guth, A.; Dow, S. Regulation of PD-L1 expression on murine tumor-associated monocytes and macrophages by locally produced TNF-α. Cancer Immunol. Immunother. 2017, 66, 523–535. [Google Scholar] [CrossRef]
- 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]
- Hartley, G.P.; Chow, L.; Ammons, D.T.; Wheat, W.H.; Dow, S.W. Programmed Cell Death Ligand 1 (PD-L1) Signaling Regulates Macrophage Proliferation and Activation. Cancer Immunol. Res. 2018, 6, 1260–1273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ahmadzadeh, M.; Johnson, L.A.; Heemskerk, B.; Wunderlich, J.R.; Dudley, M.E.; White, D.E.; Rosenberg, S.A. Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood 2009, 114, 1537–1544. [Google Scholar] [CrossRef] [PubMed]
- Curiel, T.J.; Wei, S.; Dong, H.; Alvarez, X.; Cheng, P.; Mottram, P.; Krzysiek, R.; Knutson, K.L.; Daniel, B.; Zimmermann, M.C.; et al. Blockade of B7-H1 improves myeloid dendritic cell-mediated antitumor immunity. Nat. Med. 2003, 9, 562–567. [Google Scholar] [CrossRef] [PubMed]
- Dammeijer, F.; van Gulijk, M.; Mulder, E.E.; Lukkes, M.; Klaase, L.; van den Bosch, T.; van Nimwegen, M.; Lau, S.P.; Latupeirissa, K.; Schetters, S.; et al. The PD-1/PD-L1-Checkpoint Restrains T cell Immunity in Tumor-Draining Lymph Nodes. Cancer Cell 2020, 38, 685–700.e8. [Google Scholar] [CrossRef]
- Yearley, J.H.; Gibson, C.; Yu, N.; Moon, C.; Murphy, E.; Juco, J.; Lunceford, J.; Cheng, J.; Chow, L.Q.M.; Seiwert, T.Y.; et al. PD-L2 Expression in Human Tumors: Relevance to Anti-PD-1 Therapy in Cancer. Clin. Cancer Res. 2017, 23, 3158–3167. [Google Scholar] [CrossRef] [Green Version]
- Obeid, J.M.; Erdag, G.; Smolkin, M.E.; Deacon, D.H.; Patterson, J.W.; Chen, L.; Bullock, T.N.; Slingluff, C.L. PD-L1, PD-L2 and PD-1 expression in metastatic melanoma: Correlation with tumor-infiltrating immune cells and clinical outcome. Oncoimmunology 2016, 5, e1235107. [Google Scholar] [CrossRef] [Green Version]
- Rodig, N.; Ryan, T.; Allen, J.A.; Pang, H.; Grabie, N.; Chernova, T.; Greenfield, E.A.; Liang, S.C.; Sharpe, A.H.; Lichtman, A.H.; et al. Endothelial expression of PD-L1 and PD-L2 down-regulates CD8+ T cell activation and cytolysis. Eur. J. Immunol. 2003, 33, 3117–3126. [Google Scholar] [CrossRef]
- Taube, J.M.; Anders, R.A.; Young, G.D.; Xu, H.; Sharma, R.; McMiller, T.L.; Chen, S.; Klein, A.P.; Pardoll, D.M.; Topalian, S.L.; et al. Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci. Transl. Med. 2012, 4, 127ra137. [Google Scholar] [CrossRef] [Green Version]
- Kaunitz, G.J.; Cottrell, T.R.; Lilo, M.; Muthappan, V.; Esandrio, J.; Berry, S.; Xu, H.; Ogurtsova, A.; Anders, R.A.; Fischer, A.H.; et al. Melanoma subtypes demonstrate distinct PD-L1 expression profiles. Lab. Investig. 2017, 97, 1063–1071. [Google Scholar] [CrossRef] [Green Version]
- Morales-Betanzos, C.A.; Lee, H.; Gonzalez Ericsson, P.I.; Balko, J.M.; Johnson, D.B.; Zimmerman, L.J.; Liebler, D.C. Quantitative Mass Spectrometry Analysis of PD-L1 Protein Expression, N-glycosylation and Expression Stoichiometry with PD-1 and PD-L2 in Human Melanoma. Mol. Cell. Proteomics 2017, 16, 1705–1717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Youngnak, P.; Kozono, Y.; Kozono, H.; Iwai, H.; Otsuki, N.; Jin, H.; Omura, K.; Yagita, H.; Pardoll, D.M.; Chen, L.; et al. Differential binding properties of B7-H1 and B7-DC to programmed death-1. Biochem. Biophys. Res. Commun. 2003, 307, 672–677. [Google Scholar] [CrossRef]
- Li, C.W.; Lim, S.O.; Xia, W.; Lee, H.H.; Chan, L.C.; Kuo, C.W.; Khoo, K.H.; Chang, S.S.; Cha, J.H.; Kim, T.; et al. Glycosylation and stabilization of programmed death ligand-1 suppresses T-cell activity. Nat. Commun. 2016, 7, 12632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maher, C.M.; Thomas, J.D.; Haas, D.A.; Longen, C.G.; Oyer, H.M.; Tong, J.Y.; Kim, F.J. Small-Molecule Sigma1 Modulator Induces Autophagic Degradation of PD-L1. Mol. Cancer Res. 2018, 16, 243–255. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hsu, J.M.; Li, C.W.; Lai, Y.J.; Hung, M.C. Posttranslational Modifications of PD-L1 and Their Applications in Cancer Therapy. Cancer Res. 2018, 78, 6349–6353. [Google Scholar] [CrossRef] [Green Version]
- Zerdes, I.; Matikas, A.; Bergh, J.; Rassidakis, G.Z.; Foukakis, T. Genetic, transcriptional and post-translational regulation of the programmed death protein ligand 1 in cancer: Biology and clinical correlations. Oncogene 2018, 37, 4639–4661. [Google Scholar] [CrossRef] [Green Version]
- Dong, H.; Strome, S.E.; Salomao, D.R.; Tamura, H.; Hirano, F.; Flies, D.B.; Roche, P.C.; Lu, J.; Zhu, G.; Tamada, K.; et al. Tumor-associated B7-H1 promotes T-cell apoptosis: A potential mechanism of immune evasion. Nat. Med. 2002, 8, 793–800. [Google Scholar] [CrossRef]
- Garcia-Diaz, A.; Shin, D.S.; Moreno, B.H.; Saco, J.; Escuin-Ordinas, H.; Rodriguez, G.A.; Zaretsky, J.M.; Sun, L.; Hugo, W.; Wang, X.; et al. Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression. Cell Rep. 2017, 19, 1189–1201. [Google Scholar] [CrossRef] [Green Version]
- Thiem, A.; Hesbacher, S.; Kneitz, H.; Di Primio, T.; Heppt, M.V.; Hermanns, H.M.; Goebeler, M.; Meierjohann, S.; Houben, R.; Schrama, D. IFN-gamma-induced PD-L1 expression in melanoma depends on p53 expression. J. Exp. Clin. Cancer Res. 2019, 38, 397. [Google Scholar] [CrossRef] [Green Version]
- Casey, S.C.; Tong, L.; Li, Y.; Do, R.; Walz, S.; Fitzgerald, K.N.; Gouw, A.M.; Baylot, V.; Gütgemann, I.; Eilers, M.; et al. MYC regulates the antitumor immune response through CD47 and PD-L1. Science 2016, 352, 227–231. [Google Scholar] [CrossRef] [Green Version]
- Atefi, M.; Avramis, E.; Lassen, A.; Wong, D.J.; Robert, L.; Foulad, D.; Cerniglia, M.; Titz, B.; Chodon, T.; Graeber, T.G.; et al. Effects of MAPK and PI3K pathways on PD-L1 expression in melanoma. Clin. Cancer Res. 2014, 20, 3446–3457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, X.; Zhou, J.; Giobbie-Hurder, A.; Wargo, J.; Hodi, F.S. The activation of MAPK in melanoma cells resistant to BRAF inhibition promotes PD-L1 expression that is reversible by MEK and PI3K inhibition. Clin. Cancer Res. 2013, 19, 598–609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alcázar, I.; Marqués, M.; Kumar, A.; Hirsch, E.; Wymann, M.; Carrera, A.C.; Barber, D.F. Phosphoinositide 3-kinase gamma participates in T cell receptor-induced T cell activation. J. Exp. Med. 2007, 204, 2977–2987. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- D’Souza, W.N.; Chang, C.F.; Fischer, A.M.; Li, M.; Hedrick, S.M. The Erk2 MAPK regulates CD8 T cell proliferation and survival. J. Immunol. 2008, 181, 7617–7629. [Google Scholar] [CrossRef]
- Adachi, K.; Davis, M.M. T-cell receptor ligation induces distinct signaling pathways in naive vs. antigen-experienced T cells. Proc. Natl. Acad. Sci. USA 2011, 108, 1549–1554. [Google Scholar] [CrossRef] [Green Version]
- Boussiotis, V.A.; Chatterjee, P.; Li, L. Biochemical signaling of PD-1 on T cells and its functional implications. Cancer J. 2014, 20, 265–271. [Google Scholar] [CrossRef]
- Arasanz, H.; Gato-Cañas, M.; Zuazo, M.; Ibañez-Vea, M.; Breckpot, K.; Kochan, G.; Escors, D. PD1 signal transduction pathways in T cells. Oncotarget 2017, 8, 51936–51945. [Google Scholar] [CrossRef] [Green Version]
- Parry, R.V.; Chemnitz, J.M.; Frauwirth, K.A.; Lanfranco, A.R.; Braunstein, I.; Kobayashi, S.V.; Linsley, P.S.; Thompson, C.B.; Riley, J.L. CTLA-4 and PD-1 receptors inhibit T-cell activation by distinct mechanisms. Mol. Cell. Biol. 2005, 25, 9543–9553. [Google Scholar] [CrossRef] [Green Version]
- Patsoukis, N.; Duke-Cohan, J.S.; Chaudhri, A.; Aksoylar, H.I.; Wang, Q.; Council, A.; Berg, A.; Freeman, G.J.; Boussiotis, V.A. Interaction of SHP-2 SH2 domains with PD-1 ITSM induces PD-1 dimerization and SHP-2 activation. Commun. Biol. 2020, 3, 128. [Google Scholar] [CrossRef]
- Quigley, M.; Pereyra, F.; Nilsson, B.; Porichis, F.; Fonseca, C.; Eichbaum, Q.; Julg, B.; Jesneck, J.L.; Brosnahan, K.; Imam, S.; et al. Transcriptional analysis of HIV-specific CD8+ T cells shows that PD-1 inhibits T cell function by upregulating BATF. Nat. Med. 2010, 16, 1147–1151. [Google Scholar] [CrossRef]
- Gibbons, R.M.; Liu, X.; Pulko, V.; Harrington, S.M.; Krco, C.J.; Kwon, E.D.; Dong, H. B7-H1 limits the entry of effector CD8(+) T cells to the memory pool by upregulating Bim. Oncoimmunology 2012, 1, 1061–1073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fife, B.T.; Pauken, K.E.; Eagar, T.N.; Obu, T.; Wu, J.; Tang, Q.; Azuma, M.; Krummel, M.F.; Bluestone, J.A. Interactions between PD-1 and PD-L1 promote tolerance by blocking the TCR-induced stop signal. Nat. Immunol. 2009, 10, 1185–1192. [Google Scholar] [CrossRef] [PubMed]
- Patsoukis, N.; Bardhan, K.; Chatterjee, P.; Sari, D.; Liu, B.; Bell, L.N.; Karoly, E.D.; Freeman, G.J.; Petkova, V.; Seth, P.; et al. PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat. Commun. 2015, 6, 6692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yokosuka, T.; Takamatsu, M.; Kobayashi-Imanishi, W.; Hashimoto-Tane, A.; Azuma, M.; Saito, T. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J. Exp. Med. 2012, 209, 1201–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Okazaki, T.; Maeda, A.; Nishimura, H.; Kurosaki, T.; Honjo, T. PD-1 immunoreceptor inhibits B cell receptor-mediated signaling by recruiting src homology 2-domain-containing tyrosine phosphatase 2 to phosphotyrosine. Proc. Natl. Acad. Sci. USA 2001, 98, 13866–13871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Patsoukis, N.; Li, L.; Sari, D.; Petkova, V.; Boussiotis, V.A. PD-1 increases PTEN phosphatase activity while decreasing PTEN protein stability by inhibiting casein kinase 2. Mol. Cell. Biol. 2013, 33, 3091–3098. [Google Scholar] [CrossRef] [Green Version]
- Qin, W.; Hu, L.; Zhang, X.; Jiang, S.; Li, J.; Zhang, Z.; Wang, X. The Diverse Function of PD-1/PD-L Pathway Beyond Cancer. Front. Immunol. 2019, 10, 2298. [Google Scholar] [CrossRef]
- Leclerc, M.; Voilin, E.; Gros, G.; Corgnac, S.; de Montpréville, V.; Validire, P.; Bismuth, G.; Mami-Chouaib, F. Regulation of antitumour CD8 T-cell immunity and checkpoint blockade immunotherapy by Neuropilin-1. Nat. Commun. 2019, 10, 3345. [Google Scholar] [CrossRef] [Green Version]
- Tivol, E.A.; Borriello, F.; Schweitzer, A.N.; Lynch, W.P.; Bluestone, J.A.; Sharpe, A.H. Loss of CTLA-4 leads to massive lymphoproliferation and fatal multiorgan tissue destruction, revealing a critical negative regulatory role of CTLA-4. Immunity 1995, 3, 541–547. [Google Scholar] [CrossRef] [Green Version]
- Waterhouse, P.; Penninger, J.M.; Timms, E.; Wakeham, A.; Shahinian, A.; Lee, K.P.; Thompson, C.B.; Griesser, H.; Mak, T.W. Lymphoproliferative disorders with early lethality in mice deficient in Ctla-4. Science 1995, 270, 985–988. [Google Scholar] [CrossRef]
- 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] [PubMed] [Green Version]
- Sakuishi, K.; Apetoh, L.; Sullivan, J.M.; Blazar, B.R.; Kuchroo, V.K.; Anderson, A.C. Targeting Tim-3 and PD-1 pathways to reverse T cell exhaustion and restore anti-tumor immunity. J. Exp. Med. 2010, 207, 2187–2194. [Google Scholar] [CrossRef] [PubMed]
- Fourcade, J.; Sun, Z.; Benallaoua, M.; Guillaume, P.; Luescher, I.F.; Sander, C.; Kirkwood, J.M.; Kuchroo, V.; Zarour, H.M. Upregulation of Tim-3 and PD-1 expression is associated with tumor antigen-specific CD8+ T cell dysfunction in melanoma patients. J. Exp. Med. 2010, 207, 2175–2186. [Google Scholar] [CrossRef] [PubMed]
- Grosso, J.F.; Kelleher, C.C.; Harris, T.J.; Maris, C.H.; Hipkiss, E.L.; De Marzo, A.; Anders, R.; Netto, G.; Getnet, D.; Bruno, T.C.; et al. LAG-3 regulates CD8+ T cell accumulation and effector function in murine self- and tumor-tolerance systems. J. Clin. Investig. 2007, 117, 3383–3392. [Google Scholar] [CrossRef] [Green Version]
- Huang, C.T.; Workman, C.J.; Flies, D.; Pan, X.; Marson, A.L.; Zhou, G.; Hipkiss, E.L.; Ravi, S.; Kowalski, J.; Levitsky, H.I.; et al. Role of LAG-3 in regulatory T cells. Immunity 2004, 21, 503–513. [Google Scholar] [CrossRef] [Green Version]
- Lines, J.L.; Pantazi, E.; Mak, J.; Sempere, L.F.; Wang, L.; O’Connell, S.; Ceeraz, S.; Suriawinata, A.A.; Yan, S.; Ernstoff, M.S.; et al. VISTA is an immune checkpoint molecule for human T cells. Cancer Res. 2014, 74, 1924–1932. [Google Scholar] [CrossRef] [Green Version]
- Sansom, D.M. CD28, CTLA-4 and their ligands: Who does what and to whom? Immunology 2000, 101, 169–177. [Google Scholar] [CrossRef]
- Azuma, M.; Ito, D.; Yagita, H.; Okumura, K.; Phillips, J.H.; Lanier, L.L.; Somoza, C. B70 antigen is a second ligand for CTLA-4 and CD28. Nature 1993, 366, 76–79. [Google Scholar] [CrossRef]
- Greene, J.L.; Leytze, G.M.; Emswiler, J.; Peach, R.; Bajorath, J.; Cosand, W.; Linsley, P.S. Covalent dimerization of CD28/CTLA-4 and oligomerization of CD80/CD86 regulate T cell costimulatory interactions. J. Biol. Chem. 1996, 271, 26762–26771. [Google Scholar] [CrossRef] [Green Version]
- Linsley, P.S.; Greene, J.L.; Brady, W.; Bajorath, J.; Ledbetter, J.A.; Peach, R. Human B7-1 (CD80) and B7-2 (CD86) bind with similar avidities but distinct kinetics to CD28 and CTLA-4 receptors. Immunity 1994, 1, 793–801. [Google Scholar] [CrossRef]
- van der Merwe, P.A.; Bodian, D.L.; Daenke, S.; Linsley, P.; Davis, S.J. CD80 (B7-1) binds both CD28 and CTLA-4 with a low affinity and very fast kinetics. J. Exp. Med. 1997, 185, 393–403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Linsley, P.S.; Brady, W.; Urnes, M.; Grosmaire, L.S.; Damle, N.K.; Ledbetter, J.A. CTLA-4 is a second receptor for the B cell activation antigen B7. J. Exp. Med. 1991, 174, 561–569. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kleffel, S.; Posch, C.; Barthel, S.R.; Mueller, H.; Schlapbach, C.; Guenova, E.; Elco, C.P.; Lee, N.; Juneja, V.R.; Zhan, Q.; et al. Melanoma Cell-Intrinsic PD-1 Receptor Functions Promote Tumor Growth. Cell 2015, 162, 1242–1256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Clark, C.A.; Gupta, H.B.; Sareddy, G.; Pandeswara, S.; Lao, S.; Yuan, B.; Drerup, J.M.; Padron, A.; Conejo-Garcia, J.; Murthy, K.; et al. Tumor-Intrinsic PD-L1 Signals Regulate Cell Growth, Pathogenesis, and Autophagy in Ovarian Cancer and Melanoma. Cancer Res. 2016, 76, 6964–6974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mo, X.; Zhang, H.; Preston, S.; Martin, K.; Zhou, B.; Vadalia, N.; Gamero, A.M.; Soboloff, J.; Tempera, I.; Zaidi, M.R. Interferon-γ Signaling in Melanocytes and Melanoma Cells Regulates Expression of CTLA-4. Cancer Res. 2018, 78, 436–450. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Contardi, E.; Palmisano, G.L.; Tazzari, P.L.; Martelli, A.M.; Falà, F.; Fabbi, M.; Kato, T.; Lucarelli, E.; Donati, D.; Polito, L.; et al. CTLA-4 is constitutively expressed on tumor cells and can trigger apoptosis upon ligand interaction. Int. J. Cancer 2005, 117, 538–550. [Google Scholar] [CrossRef]
- Seliger, B.; Maio, M.; Cuaia, O.; Calabro, L. Expression and function of CTLA4 in melanoma. J. Clin. Oncol. 2013, 31, e20040. [Google Scholar] [CrossRef]
- Pistillo, M.P.; Carosio, R.; Grillo, F.; Fontana, V.; Mastracci, L.; Morabito, A.; Banelli, B.; Tanda, E.; Cecchi, F.; Dozin, B.; et al. Phenotypic characterization of tumor CTLA-4 expression in melanoma tissues and its possible role in clinical response to Ipilimumab. Clin. Immunol. 2020, 215, 108428. [Google Scholar] [CrossRef]
- Vignali, D.A.; Collison, L.W.; Workman, C.J. How regulatory T cells work. Nat. Rev. Immunol. 2008, 8, 523–532. [Google Scholar] [CrossRef] [Green Version]
- Viguier, M.; Lemaître, F.; Verola, O.; Cho, M.S.; Gorochov, G.; Dubertret, L.; Bachelez, H.; Kourilsky, P.; Ferradini, L. Foxp3 expressing CD4+CD25(high) regulatory T cells are overrepresented in human metastatic melanoma lymph nodes and inhibit the function of infiltrating T cells. J. Immunol. 2004, 173, 1444–1453. [Google Scholar] [CrossRef] [Green Version]
- Gray, C.P.; Arosio, P.; Hersey, P. Association of increased levels of heavy-chain ferritin with increased CD4+ CD25+ regulatory T-cell levels in patients with melanoma. Clin. Cancer Res. 2003, 9, 2551–2559. [Google Scholar] [PubMed]
- Fujii, H.; Josse, J.; Tanioka, M.; Miyachi, Y.; Husson, F.; Ono, M. Regulatory T Cells in Melanoma Revisited by a Computational Clustering of FOXP3+ T Cell Subpopulations. J. Immunol. 2016, 196, 2885–2892. [Google Scholar] [CrossRef] [PubMed]
- Magnuson, A.M.; Kiner, E.; Ergun, A.; Park, J.S.; Asinovski, N.; Ortiz-Lopez, A.; Kilcoyne, A.; Paoluzzi-Tomada, E.; Weissleder, R.; Mathis, D.; et al. Identification and validation of a tumor-infiltrating Treg transcriptional signature conserved across species and tumor types. Proc. Natl. Acad. Sci. USA 2018, 115, E10672–E10681. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ladányi, A.; Mohos, A.; Somlai, B.; Liszkay, G.; Gilde, K.; Fejos, Z.; Gaudi, I.; Tímár, J. FOXP3+ cell density in primary tumor has no prognostic impact in patients with cutaneous malignant melanoma. Pathol. Oncol. Res. 2010, 16, 303–309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Leslie, C.; Bowyer, S.E.; White, A.; Grieu-Iacopetta, F.; Trevenen, M.; Iacopetta, B.; Amanuel, B.; Millward, M. FOXP3+ T regulatory lymphocytes in primary melanoma are associated with BRAF mutation but not with response to BRAF inhibitor. Pathology 2015, 47, 557–563. [Google Scholar] [CrossRef]
- Klages, K.; Mayer, C.T.; Lahl, K.; Loddenkemper, C.; Teng, M.W.; Ngiow, S.F.; Smyth, M.J.; Hamann, A.; Huehn, J.; Sparwasser, T. Selective depletion of Foxp3+ regulatory T cells improves effective therapeutic vaccination against established melanoma. Cancer Res. 2010, 70, 7788–7799. [Google Scholar] [CrossRef] [Green Version]
- Rasku, M.A.; Clem, A.L.; Telang, S.; Taft, B.; Gettings, K.; Gragg, H.; Cramer, D.; Lear, S.C.; McMasters, K.M.; Miller, D.M.; et al. Transient T cell depletion causes regression of melanoma metastases. J. Transl. Med. 2008, 6, 12. [Google Scholar] [CrossRef] [Green Version]
- Klarquist, J.; Tobin, K.; Farhangi Oskuei, P.; Henning, S.W.; Fernandez, M.F.; Dellacecca, E.R.; Navarro, F.C.; Eby, J.M.; Chatterjee, S.; Mehrotra, S.; et al. Ccl22 Diverts T Regulatory Cells and Controls the Growth of Melanoma. Cancer Res. 2016, 76, 6230–6240. [Google Scholar] [CrossRef] [Green Version]
- Ahmadzadeh, M.; Felipe-Silva, A.; Heemskerk, B.; Powell, D.J., Jr.; Wunderlich, J.R.; Merino, M.J.; Rosenberg, S.A. FOXP3 expression accurately defines the population of intratumoral regulatory T cells that selectively accumulate in metastatic melanoma lesions. Blood 2008, 112, 4953–4960. [Google Scholar] [CrossRef]
- Alb, M.; Sie, C.; Adam, C.; Chen, S.; Becker, J.C.; Schrama, D. Cellular and cytokine-dependent immunosuppressive mechanisms of grm1-transgenic murine melanoma. Cancer Immunol. Immunother. 2012, 61, 2239–2249. [Google Scholar] [CrossRef] [Green Version]
- Gray, C.P.; Franco, A.V.; Arosio, P.; Hersey, P. Immunosuppressive effects of melanoma-derived heavy-chain ferritin are dependent on stimulation of IL-10 production. Int. J. Cancer 2001, 92, 843–850. [Google Scholar] [CrossRef] [PubMed]
- Gray, C.P.; Arosio, P.; Hersey, P. Heavy chain ferritin activates regulatory T cells by induction of changes in dendritic cells. Blood 2002, 99, 3326–3334. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deng, G. Tumor-infiltrating regulatory T cells: Origins and features. Am. J. Clin. Exp. Immunol. 2018, 7, 81–87. [Google Scholar] [PubMed]
- Fan, X.; Allison, J.P. Chemokines and recruitment of regulatory T cells to the tumor. J. Immunol. 2009, 182, 40.14. [Google Scholar]
- Chaudhary, B.; Elkord, E. Regulatory T Cells in the Tumor Microenvironment and Cancer Progression: Role and Therapeutic Targeting. Vaccines 2016, 4, 28. [Google Scholar] [CrossRef] [Green Version]
- Baumgartner, J.; Wilson, C.; Palmer, B.; Richter, D.; Banerjee, A.; McCarter, M. Melanoma induces immunosuppression by up-regulating FOXP3(+) regulatory T cells. J. Surg. Res. 2007, 141, 72–77. [Google Scholar] [CrossRef] [Green Version]
- Shevach, E.M. Foxp3(+) T Regulatory Cells: Still Many Unanswered Questions-A Perspective After 20 Years of Study. Front. Immunol. 2018, 9, 1048. [Google Scholar] [CrossRef]
- Ataera, H.; Hyde, E.; Price, K.M.; Stoitzner, P.; Ronchese, F. Murine melanoma-infiltrating dendritic cells are defective in antigen presenting function regardless of the presence of CD4CD25 regulatory T cells. PLoS ONE 2011, 6, e17515. [Google Scholar] [CrossRef] [Green Version]
- Kawamoto, H.; Minato, N. Myeloid cells. Int. J. Biochem. Cell Biol. 2004, 36, 1374–1379. [Google Scholar] [CrossRef]
- Gabrilovich, D.I. Myeloid-Derived Suppressor Cells. Cancer Immunol. Res. 2017, 5, 3–8. [Google Scholar] [CrossRef] [Green Version]
- Condamine, T.; Ramachandran, I.; Youn, J.I.; Gabrilovich, D.I. Regulation of tumor metastasis by myeloid-derived suppressor cells. Annu. Rev. Med. 2015, 66, 97–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- mansky, V.; Sevko, A.; Gebhardt, C.; Utikal, J. Myeloid-derived suppressor cells in malignant melanoma. J. Dtsch. Dermatol. Ges. 2014, 12, 1021–1027. [Google Scholar] [CrossRef] [PubMed]
- Schlecker, E.; Stojanovic, A.; Eisen, C.; Quack, C.; Falk, C.S.; Umansky, V.; Cerwenka, A. Tumor-infiltrating monocytic myeloid-derived suppressor cells mediate CCR5-dependent recruitment of regulatory T cells favoring tumor growth. J. Immunol. 2012, 189, 5602–5611. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mairhofer, D.G.; Ortner, D.; Tripp, C.H.; Schaffenrath, S.; Fleming, V.; Heger, L.; Komenda, K.; Reider, D.; Dudziak, D.; Chen, S.; et al. Impaired gp100-Specific CD8(+) T-Cell Responses in the Presence of Myeloid-Derived Suppressor Cells in a Spontaneous Mouse Melanoma Model. J. Investig. Dermatol. 2015, 135, 2785–2793. [Google Scholar] [CrossRef] [Green Version]
- Weide, B.; Martens, A.; Zelba, H.; Stutz, C.; Derhovanessian, E.; Di Giacomo, A.M.; Maio, M.; Sucker, A.; Schilling, B.; Schadendorf, D.; et al. Myeloid-derived suppressor cells predict survival of patients with advanced melanoma: Comparison with regulatory T cells and NY-ESO-1- or melan-A-specific T cells. Clin. Cancer Res. 2014, 20, 1601–1609. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jordan, K.R.; Amaria, R.N.; Ramirez, O.; Callihan, E.B.; Gao, D.; Borakove, M.; Manthey, E.; Borges, V.F.; McCarter, M.D. Myeloid-derived suppressor cells are associated with disease progression and decreased overall survival in advanced-stage melanoma patients. Cancer Immunol. Immunother. 2013, 62, 1711–1722. [Google Scholar] [CrossRef] [Green Version]
- Meyer, C.; Sevko, A.; Ramacher, M.; Bazhin, A.V.; Falk, C.S.; Osen, W.; Borrello, I.; Kato, M.; Schadendorf, D.; Baniyash, M.; et al. Chronic inflammation promotes myeloid-derived suppressor cell activation blocking antitumor immunity in transgenic mouse melanoma model. Proc. Natl. Acad. Sci. USA 2011, 108, 17111–17116. [Google Scholar] [CrossRef] [Green Version]
- Poschke, I.; Mougiakakos, D.; Hansson, J.; Masucci, G.V.; Kiessling, R. Immature immunosuppressive CD14+HLA-DR-/low cells in melanoma patients are Stat3hi and overexpress CD80, CD83, and DC-sign. Cancer Res. 2010, 70, 4335–4345. [Google Scholar] [CrossRef] [Green Version]
- Raber, P.L.; Thevenot, P.; Sierra, R.; Wyczechowska, D.; Halle, D.; Ramirez, M.E.; Ochoa, A.C.; Fletcher, M.; Velasco, C.; Wilk, A.; et al. Subpopulations of myeloid-derived suppressor cells impair T cell responses through independent nitric oxide-related pathways. Int. J. Cancer 2014, 134, 2853–2864. [Google Scholar] [CrossRef]
- Kusmartsev, S.; Gabrilovich, D.I. STAT1 signaling regulates tumor-associated macrophage-mediated T cell deletion. J. Immunol. 2005, 174, 4880–4891. [Google Scholar] [CrossRef] [Green Version]
- Mazzoni, A.; Bronte, V.; Visintin, A.; Spitzer, J.H.; Apolloni, E.; Serafini, P.; Zanovello, P.; Segal, D.M. Myeloid suppressor lines inhibit T cell responses by an NO-dependent mechanism. J. Immunol. 2002, 168, 689–695. [Google Scholar] [CrossRef] [PubMed]
- Apolloni, E.; Bronte, V.; Mazzoni, A.; Serafini, P.; Cabrelle, A.; Segal, D.M.; Young, H.A.; Zanovello, P. Immortalized myeloid suppressor cells trigger apoptosis in antigen-activated T lymphocytes. J. Immunol. 2000, 165, 6723–6730. [Google Scholar] [CrossRef] [PubMed]
- Gabrilovich, D.I.; Nagaraj, S. Myeloid-derived suppressor cells as regulators of the immune system. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef] [PubMed]
- Kusmartsev, S.; Nefedova, Y.; Yoder, D.; Gabrilovich, D.I. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. J. Immunol. 2004, 172, 989–999. [Google Scholar] [CrossRef] [Green Version]
- Jayasingam, S.D.; Citartan, M.; Thang, T.H.; Mat Zin, A.A.; Ang, K.C.; Ch’ng, E.S. Evaluating the Polarization of Tumor-Associated Macrophages Into M1 and M2 Phenotypes in Human Cancer Tissue: Technicalities and Challenges in Routine Clinical Practice. Front. Oncol. 2019, 9, 1512. [Google Scholar] [CrossRef] [Green Version]
- Fujimura, T.; Kambayashi, Y.; Fujisawa, Y.; Hidaka, T.; Aiba, S. Tumor-Associated Macrophages: Therapeutic Targets for Skin Cancer. Front. Oncol. 2018, 8, 3. [Google Scholar] [CrossRef] [Green Version]
- Tcyganov, E.; Mastio, J.; Chen, E.; Gabrilovich, D.I. Plasticity of myeloid-derived suppressor cells in cancer. Curr. Opin. Immunol. 2018, 51, 76–82. [Google Scholar] [CrossRef]
- Movahedi, K.; Laoui, D.; Gysemans, C.; Baeten, M.; Stangé, G.; Van den Bossche, J.; Mack, M.; Pipeleers, D.; In’t Veld, P.; De Baetselier, P.; et al. Different tumor microenvironments contain functionally distinct subsets of macrophages derived from Ly6C(high) monocytes. Cancer Res. 2010, 70, 5728–5739. [Google Scholar] [CrossRef] [Green Version]
- Laoui, D.; Van Overmeire, E.; Di Conza, G.; Aldeni, C.; Keirsse, J.; Morias, Y.; Movahedi, K.; Houbracken, I.; Schouppe, E.; Elkrim, Y.; et al. Tumor hypoxia does not drive differentiation of tumor-associated macrophages but rather fine-tunes the M2-like macrophage population. Cancer Res. 2014, 74, 24–30. [Google Scholar] [CrossRef] [Green Version]
- Corzo, C.A.; Condamine, T.; Lu, L.; Cotter, M.J.; Youn, J.I.; Cheng, P.; Cho, H.I.; Celis, E.; Quiceno, D.G.; Padhya, T.; et al. HIF-1α regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J. Exp. Med. 2010, 207, 2439–2453. [Google Scholar] [CrossRef]
- Palazón, A.; Aragonés, J.; Morales-Kastresana, A.; de Landázuri, M.O.; Melero, I. Molecular pathways: Hypoxia response in immune cells fighting or promoting cancer. Clin. Cancer Res. 2012, 18, 1207–1213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Salmi, S.; Siiskonen, H.; Sironen, R.; Tyynelä-Korhonen, K.; Hirschovits-Gerz, B.; Valkonen, M.; Auvinen, P.; Pasonen-Seppänen, S. The number and localization of CD68+ and CD163+ macrophages in different stages of cutaneous melanoma. Melanoma Res. 2019, 29, 237–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Falleni, M.; Savi, F.; Tosi, D.; Agape, E.; Cerri, A.; Moneghini, L.; Bulfamante, G.P. M1 and M2 macrophages’ clinicopathological significance in cutaneous melanoma. Melanoma Res. 2017, 27, 200–210. [Google Scholar] [CrossRef] [PubMed]
- Herwig, M.C.; Bergstrom, C.; Wells, J.R.; Höller, T.; Grossniklaus, H.E. M2/M1 ratio of tumor associated macrophages and PPAR-gamma expression in uveal melanomas with class 1 and class 2 molecular profiles. Exp. Eye Res. 2013, 107, 52–58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bronkhorst, I.H.; Ly, L.V.; Jordanova, E.S.; Vrolijk, J.; Versluis, M.; Luyten, G.P.; Jager, M.J. Detection of M2-macrophages in uveal melanoma and relation with survival. Invest. Ophthalmol. Vis. Sci. 2011, 52, 643–650. [Google Scholar] [CrossRef] [Green Version]
- Mäkitie, T.; Summanen, P.; Tarkkanen, A.; Kivelä, T. Tumor-infiltrating macrophages (CD68(+) cells) and prognosis in malignant uveal melanoma. Invest. Ophthalmol. Vis. Sci. 2001, 42, 1414–1421. [Google Scholar] [PubMed]
- Jensen, T.O.; Schmidt, H.; Møller, H.J.; Høyer, M.; Maniecki, M.B.; Sjoegren, P.; Christensen, I.J.; Steiniche, T. Macrophage markers in serum and tumor have prognostic impact in American Joint Committee on Cancer stage I/II melanoma. J. Clin. Oncol. 2009, 27, 3330–3337. [Google Scholar] [CrossRef]
- Ley, K. M1 Means Kill; M2 Means Heal. J. Immunol. 2017, 199, 2191–2193. [Google Scholar] [CrossRef]
- Buscher, K.; Ehinger, E.; Gupta, P.; Pramod, A.B.; Wolf, D.; Tweet, G.; Pan, C.; Mills, C.D.; Lusis, A.J.; Ley, K. Natural variation of macrophage activation as disease-relevant phenotype predictive of inflammation and cancer survival. Nat. Commun. 2017, 8, 16041. [Google Scholar] [CrossRef]
- Van Overmeire, E.; Stijlemans, B.; Heymann, F.; Keirsse, J.; Morias, Y.; Elkrim, Y.; Brys, L.; Abels, C.; Lahmar, Q.; Ergen, C.; et al. M-CSF and GM-CSF Receptor Signaling Differentially Regulate Monocyte Maturation and Macrophage Polarization in the Tumor Microenvironment. Cancer Res. 2016, 76, 35–42. [Google Scholar] [CrossRef] [Green Version]
- Georgoudaki, A.M.; Prokopec, K.E.; Boura, V.F.; Hellqvist, E.; Sohn, S.; Östling, J.; Dahan, R.; Harris, R.A.; Rantalainen, M.; Klevebring, D.; et al. Reprogramming Tumor-Associated Macrophages by Antibody Targeting Inhibits Cancer Progression and Metastasis. Cell Rep. 2016, 15, 2000–2011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kaneda, M.M.; Messer, K.S.; Ralainirina, N.; Li, H.; Leem, C.J.; Gorjestani, S.; Woo, G.; Nguyen, A.V.; Figueiredo, C.C.; Foubert, P.; et al. PI3Kγ is a molecular switch that controls immune suppression. Nature 2016, 539, 437–442. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Vergadi, E.; Ieronymaki, E.; Lyroni, K.; Vaporidi, K.; Tsatsanis, C. Akt Signaling Pathway in Macrophage Activation and M1/M2 Polarization. J. Immunol. 2017, 198, 1006–1014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mouchemore, K.A.; Sampaio, N.G.; Murrey, M.W.; Stanley, E.R.; Lannutti, B.J.; Pixley, F.J. Specific inhibition of PI3K p110δ inhibits CSF-1-induced macrophage spreading and invasive capacity. FEBS J. 2013, 280, 5228–5236. [Google Scholar] [CrossRef] [Green Version]
- Huang, S.C.; Smith, A.M.; Everts, B.; Colonna, M.; Pearce, E.L.; Schilling, J.D.; Pearce, E.J. Metabolic Reprogramming Mediated by the mTORC2-IRF4 Signaling Axis Is Essential for Macrophage Alternative Activation. Immunity 2016, 45, 817–830. [Google Scholar] [CrossRef] [Green Version]
- Arredouani, M.S. Is the scavenger receptor MARCO a new immune checkpoint? Oncoimmunology 2014, 3, e955709. [Google Scholar] [CrossRef] [Green Version]
- Alberts, B.; Johnson, A.; Lewis, J.; Raff, M.; Roberts, K.; Walter, P. Helper T Cells and Lymphocyte Activation. In Molecular Biology of the Cell, 4th ed.; Garland Science: New York, NY, USA, 2002. [Google Scholar]
- Mills, C.D.; Kincaid, K.; Alt, J.M.; Heilman, M.J.; Hill, A.M. M-1/M-2 macrophages and the Th1/Th2 paradigm. J. Immunol. 2000, 164, 6166–6173. [Google Scholar] [CrossRef] [Green Version]
- Nevala, W.K.; Vachon, C.M.; Leontovich, A.A.; Scott, C.G.; Thompson, M.A.; Markovic, S.N. Evidence of systemic Th2-driven chronic inflammation in patients with metastatic melanoma. Clin. Cancer Res. 2009, 15, 1931–1939. [Google Scholar] [CrossRef] [Green Version]
- Lauerova, L.; Dusek, L.; Simickova, M.; Kocák, I.; Vagundová, M.; Zaloudík, J.; Kovarík, J. Malignant melanoma associates with Th1/Th2 imbalance that coincides with disease progression and immunotherapy response. Neoplasma 2002, 49, 159–166. [Google Scholar]
- Enninga, E.A.; Nevala, W.K.; Holtan, S.G.; Leontovich, A.A.; Markovic, S.N. Galectin-9 modulates immunity by promoting Th2/M2 differentiation and impacts survival in patients with metastatic melanoma. Melanoma Res. 2016, 26, 429–441. [Google Scholar] [CrossRef] [PubMed]
- Biswas, S.K.; Mantovani, A. Macrophage plasticity and interaction with lymphocyte subsets: Cancer as a paradigm. Nat. Immunol. 2010, 11, 889–896. [Google Scholar] [CrossRef] [PubMed]
- Massi, D.; Marconi, C.; Franchi, A.; Bianchini, F.; Paglierani, M.; Ketabchi, S.; Miracco, C.; Santucci, M.; Calorini, L. Arginine metabolism in tumor-associated macrophages in cutaneous malignant melanoma: Evidence from human and experimental tumors. Hum. Pathol. 2007, 38, 1516–1525. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Yang, L.; Wang, D.; Zhang, Q.; Zhang, L. Pro-tumor activities of macrophages in the progression of melanoma. Hum. Vaccines Immunother. 2017, 13, 1556–1562. [Google Scholar] [CrossRef]
- Chen, P.; Huang, Y.; Bong, R.; Ding, Y.; Song, N.; Wang, X.; Song, X.; Luo, Y. Tumor-associated macrophages promote angiogenesis and melanoma growth via adrenomedullin in a paracrine and autocrine manner. Clin. Cancer Res. 2011, 17, 7230–7239. [Google Scholar] [CrossRef] [Green Version]
- Tucci, M.; Passarelli, A.; Mannavola, F.; Felici, C.; Stucci, L.S.; Cives, M.; Silvestris, F. Immune System Evasion as Hallmark of Melanoma Progression: The Role of Dendritic Cells. Front. Oncol. 2019, 9, 1148. [Google Scholar] [CrossRef] [Green Version]
- Escors, D. Tumour immunogenicity, antigen presentation and immunological barriers in cancer immunotherapy. New J. Sci. 2014, 2014. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stoitzner, P.; Green, L.K.; Jung, J.Y.; Price, K.M.; Atarea, H.; Kivell, B.; Ronchese, F. Inefficient presentation of tumor-derived antigen by tumor-infiltrating dendritic cells. Cancer Immunol. Immunother. 2008, 57, 1665–1673. [Google Scholar] [CrossRef]
- Itakura, E.; Huang, R.R.; Wen, D.R.; Paul, E.; Wünsch, P.H.; Cochran, A.J. IL-10 expression by primary tumor cells correlates with melanoma progression from radial to vertical growth phase and development of metastatic competence. Mod. Pathol. 2011, 24, 801–809. [Google Scholar] [CrossRef] [PubMed]
- Chattopadhyay, G.; Shevach, E.M. Antigen-specific induced T regulatory cells impair dendritic cell function via an IL-10/MARCH1-dependent mechanism. J. Immunol. 2013, 191, 5875–5884. [Google Scholar] [CrossRef] [Green Version]
- Mittal, S.K.; Roche, P.A. Suppression of antigen presentation by IL-10. Curr. Opin. Immunol. 2015, 34, 22–27. [Google Scholar] [CrossRef] [Green Version]
- Groux, H.; Bigler, M.; de Vries, J.E.; Roncarolo, M.G. Inhibitory and stimulatory effects of IL-10 on human CD8+ T cells. J. Immunol. 1998, 160, 3188–3193. [Google Scholar] [PubMed]
- Laurent, S.; Carrega, P.; Saverino, D.; Piccioli, P.; Camoriano, M.; Morabito, A.; Dozin, B.; Fontana, V.; Simone, R.; Mortara, L.; et al. CTLA-4 is expressed by human monocyte-derived dendritic cells and regulates their functions. Hum. Immunol. 2010, 71, 934–941. [Google Scholar] [CrossRef] [PubMed]
- Yao, S.; Wang, S.; Zhu, Y.; Luo, L.; Zhu, G.; Flies, S.; Xu, H.; Ruff, W.; Broadwater, M.; Choi, I.H.; et al. PD-1 on dendritic cells impedes innate immunity against bacterial infection. Blood 2009, 113, 5811–5818. [Google Scholar] [CrossRef] [PubMed]
- Hobo, W.; Maas, F.; Adisty, N.; De Witte, T.; Schaap, N.; Van der Voort, R.; Dolstra, H. siRNA silencing of PD-L1 and PD-L2 on dendritic cells augments expansion and function of minor histocompatibility antigen-specific CD8+ T cells. Blood 2010, 116, 4501–4511. [Google Scholar] [CrossRef] [Green Version]
- Oh, S.A.; Wu, D.-C.; Cheung, J.; Navarro, A.; Xiong, H.; Cubas, R.; Totpal, K.; Chiu, H.; Wu, Y.; Comps-Agrar, L.; et al. PD-L1 expression by dendritic cells is a key regulator of T-cell immunity in cancer. Nat. Cancer 2020, 1, 681–691. [Google Scholar] [CrossRef]
- Wang, X.B.; Fan, Z.Z.; Anton, D.; Vollenhoven, A.V.; Ni, Z.H.; Chen, X.F.; Lefvert, A.K. CTLA4 is expressed on mature dendritic cells derived from human monocytes and influences their maturation and antigen presentation. BMC Immunol. 2011, 12, 21. [Google Scholar] [CrossRef] [Green Version]
- Zhang, W.; Song, Z.; Xiao, J.; Liu, X.; Luo, Y.; Yang, Z.; Luo, R.; Li, A. Blocking the PD-1/PD-L1 axis in dendritic cell-stimulated Cytokine-Induced Killer Cells with pembrolizumab enhances their therapeutic effects against hepatocellular carcinoma. J. Cancer 2019, 10, 2578–2587. [Google Scholar] [CrossRef] [Green Version]
- Alexander, M.A.; Bennicelli, J.; Guerry, D.T. Defective antigen presentation by human melanoma cell lines cultured from advanced, but not biologically early, disease. J. Immunol. 1989, 142, 4070–4078. [Google Scholar]
- Kageshita, T.; Hirai, S.; Ono, T.; Hicklin, D.J.; Ferrone, S. Down-regulation of HLA class I antigen-processing molecules in malignant melanoma: Association with disease progression. Am. J. Pathol. 1999, 154, 745–754. [Google Scholar] [CrossRef]
- Chang, C.C.; Pirozzi, G.; Wen, S.H.; Chung, I.H.; Chiu, B.L.; Errico, S.; Luongo, M.; Lombardi, M.L.; Ferrone, S. Multiple structural and epigenetic defects in the human leukocyte antigen class I antigen presentation pathway in a recurrent metastatic melanoma following immunotherapy. J. Biol. Chem. 2015, 290, 26562–26575. [Google Scholar] [CrossRef] [Green Version]
- Khan, A.N.; Gregorie, C.J.; Tomasi, T.B. Histone deacetylase inhibitors induce TAP, LMP, Tapasin genes and MHC class I antigen presentation by melanoma cells. Cancer Immunol. Immunother. 2008, 57, 647–654. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Serrano, A.; Tanzarella, S.; Lionello, I.; Mendez, R.; Traversari, C.; Ruiz-Cabello, F.; Garrido, F. Rexpression of HLA class I antigens and restoration of antigen-specific CTL response in melanoma cells following 5-aza-2’-deoxycytidine treatment. Int. J. Cancer 2001, 94, 243–251. [Google Scholar] [CrossRef] [PubMed]
- Del Campo, A.B.; Kyte, J.A.; Carretero, J.; Zinchencko, S.; Méndez, R.; González-Aseguinolaza, G.; Ruiz-Cabello, F.; Aamdal, S.; Gaudernack, G.; Garrido, F.; et al. Immune escape of cancer cells with beta2-microglobulin loss over the course of metastatic melanoma. Int. J. Cancer 2014, 134, 102–113. [Google Scholar] [CrossRef] [PubMed]
- Hicklin, D.J.; Wang, Z.; Arienti, F.; Rivoltini, L.; Parmiani, G.; Ferrone, S. beta2-Microglobulin mutations, HLA class I antigen loss, and tumor progression in melanoma. J. Clin. Investig. 1998, 101, 2720–2729. [Google Scholar] [CrossRef] [PubMed]
- Cormier, J.N.; Hijazi, Y.M.; Abati, A.; Fetsch, P.; Bettinotti, M.; Steinberg, S.M.; Rosenberg, S.A.; Marincola, F.M. Heterogeneous expression of melanoma-associated antigens and HLA-A2 in metastatic melanoma in vivo. Int. J. Cancer 1998, 75, 517–524. [Google Scholar] [CrossRef]
- Marincola, F.M.; Hijazi, Y.M.; Fetsch, P.; Salgaller, M.L.; Rivoltini, L.; Cormier, J.; Simonis, T.B.; Duray, P.H.; Herlyn, M.; Kawakami, Y.; et al. Analysis of expression of the melanoma-associated antigens MART-1 and gp100 in metastatic melanoma cell lines and in in situ lesions. J. Immunother. Emphasis Tumor Immunol. 1996, 19, 192–205. [Google Scholar] [CrossRef] [PubMed]
- Jäger, E.; Ringhoffer, M.; Karbach, J.; Arand, M.; Oesch, F.; Knuth, A. Inverse relationship of melanocyte differentiation antigen expression in melanoma tissues and CD8+ cytotoxic-T-cell responses: Evidence for immunoselection of antigen-loss variants in vivo. Int. J. Cancer 1996, 66, 470–476. [Google Scholar] [CrossRef]
- Cormier, J.N.; Abati, A.; Fetsch, P.; Hijazi, Y.M.; Rosenberg, S.A.; Marincola, F.M.; Topalian, S.L. Comparative analysis of the in vivo expression of tyrosinase, MART-1/Melan-A, and gp100 in metastatic melanoma lesions: Implications for immunotherapy. J. Immunother. 1998, 21, 27–31. [Google Scholar] [CrossRef]
- Donia, M.; Andersen, R.; Kjeldsen, J.W.; Fagone, P.; Munir, S.; Nicoletti, F.; Andersen, M.H.; Thor Straten, P.; Svane, I.M. Aberrant Expression of MHC Class II in Melanoma Attracts Inflammatory Tumor-Specific CD4+ T- Cells, Which Dampen CD8+ T-cell Antitumor Reactivity. Cancer Res. 2015, 75, 3747–3759. [Google Scholar] [CrossRef] [Green Version]
- Cohen-Solal, K.A.; Reuhl, K.R.; Ryan, K.B.; Roberts, K.G.; Chen, S. Development of cutaneous amelanotic melanoma in the absence of a functional tyrosinase. Pigment Cell Res. 2001, 14, 466–474. [Google Scholar] [CrossRef]
- Schiffner, S.; Chen, S.; Becker, J.C.; Bosserhoff, A.K. Highly pigmented Tg(Grm1) mouse melanoma develops non-pigmented melanoma cells in distant metastases. Exp. Dermatol. 2012, 21, 786–788. [Google Scholar] [CrossRef]
- Shah, R.; Singh, S.J.; Eddy, K.; Filipp, F.V.; Chen, S. Concurrent Targeting of Glutaminolysis and Metabotropic Glutamate Receptor 1 (GRM1) Reduces Glutamate Bioavailability in GRM1+ Melanoma. Cancer Res. 2019, 79, 1799–1809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Prokopi, A.; Tripp, C.H.; Tummers, B.; Komenda, K.; Hutter, K.; Cappellano, G.; Bellmann, L.; Efremova, M.; Trajanoski, Z.; Chen, S.; et al. Abstract A102: Rescue of lost skin dendritic cells in melanoma is key for the resuscitation of antitumor T-cell responses. Cancer Immunol. Res 2019, 7, A102. [Google Scholar] [CrossRef]
- Schwarz, T. Mechanisms of UV-induced immunosuppression. Keio J. Med. 2005, 54, 165–171. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kripke, M.L.; Fisher, M.S. Immunologic parameters of ultraviolet carcinogenesis. J. Natl. Cancer Inst. 1976, 57, 211–215. [Google Scholar] [CrossRef]
- Kripke, M.L.; Fisher, M.S. Immunologic aspects of tumor induction by ultraviolet radiation. Natl. Cancer Inst. Monogr. 1978, 50, 179–183. [Google Scholar]
- Hart, P.H.; Norval, M. Ultraviolet radiation-induced immunosuppression and its relevance for skin carcinogenesis. Photochem. Photobiol. Sci. 2018, 17, 1872–1884. [Google Scholar] [CrossRef]
- Fortner, G.W.; Kripke, M.L. In vitro reactivity of splenic lymphocytes from normal and UV-irradiated mice against syngeneic UV-induced tumors. J. Immunol. 1977, 118, 1483–1487. [Google Scholar]
- Sun, X.; Zhang, N.; Yin, C.; Zhu, B.; Li, X. Ultraviolet Radiation and Melanomagenesis: From Mechanism to Immunotherapy. Front. Oncol. 2020, 10, 951. [Google Scholar] [CrossRef]
- Schwarz, A.; Noordegraaf, M.; Maeda, A.; Torii, K.; Clausen, B.E.; Schwarz, T. Langerhans cells are required for UVR-induced immunosuppression. J. Investig. Dermatol. 2010, 130, 1419–1427. [Google Scholar] [CrossRef] [Green Version]
- Shreedhar, V.K.; Pride, M.W.; Sun, Y.; Kripke, M.L.; Strickland, F.M. Origin and characteristics of ultraviolet-B radiation-induced suppressor T lymphocytes. J. Immunol. 1998, 161, 1327–1335. [Google Scholar] [PubMed]
- Rivas, J.M.; Ullrich, S.E. The role of IL-4, IL-10, and TNF-alpha in the immune suppression induced by ultraviolet radiation. J. Leukoc. Biol. 1994, 56, 769–775. [Google Scholar] [CrossRef] [PubMed]
- Granstein, R.D.; Matsui, M.S. UV radiation-induced immunosuppression and skin cancer. Cutis 2004, 74, 4–9. [Google Scholar] [PubMed]
- Ullrich, S.E. Does exposure to UV radiation induce a shift to a Th-2-like immune reaction? Photochem. Photobiol. 1996, 64, 254–258. [Google Scholar] [CrossRef]
- Schmitt, D.A.; Walterscheid, J.P.; Ullrich, S.E. Reversal of ultraviolet radiation-induced immune suppression by recombinant interleukin-12: Suppression of cytokine production. Immunology 2000, 101, 90–96. [Google Scholar] [CrossRef]
- Hood, J.L.; San, R.S.; Wickline, S.A. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 2011, 71, 3792–3801. [Google Scholar] [CrossRef] [Green Version]
- Hu, L.; Wickline, S.A.; Hood, J.L. Magnetic resonance imaging of melanoma exosomes in lymph nodes. Magn. Reson. Med. 2015, 74, 266–271. [Google Scholar] [CrossRef]
- Düchler, M.; Czernek, L.; Peczek, L.; Cypryk, W.; Sztiller-Sikorska, M.; Czyz, M. Melanoma-Derived Extracellular Vesicles Bear the Potential for the Induction of Antigen-Specific Tolerance. Cells 2019, 8, 665. [Google Scholar] [CrossRef] [Green Version]
- Taylor, D.D.; Gerçel-Taylor, C. Tumour-derived exosomes and their role in cancer-associated T-cell signalling defects. Br. J. Cancer 2005, 92, 305–311. [Google Scholar] [CrossRef]
- Muller, L.; Mitsuhashi, M.; Simms, P.; Gooding, W.E.; Whiteside, T.L. Tumor-derived exosomes regulate expression of immune function-related genes in human T cell subsets. Sci. Rep. 2016, 6, 20254. [Google Scholar] [CrossRef] [Green Version]
- Sharma, P.; Diergaarde, B.; Ferrone, S.; Kirkwood, J.M.; Whiteside, T.L. Melanoma cell-derived exosomes in plasma of melanoma patients suppress functions of immune effector cells. Sci. Rep. 2020, 10, 92. [Google Scholar] [CrossRef] [PubMed]
- Liu, C.; Yu, S.; Zinn, K.; Wang, J.; Zhang, L.; Jia, Y.; Kappes, J.C.; Barnes, S.; Kimberly, R.P.; Grizzle, W.E.; et al. Murine mammary carcinoma exosomes promote tumor growth by suppression of NK cell function. J. Immunol. 2006, 176, 1375–1385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiang, X.; Poliakov, A.; Liu, C.; Liu, Y.; Deng, Z.B.; Wang, J.; Cheng, Z.; Shah, S.V.; Wang, G.J.; Zhang, L.; et al. Induction of myeloid-derived suppressor cells by tumor exosomes. Int. J. Cancer 2009, 124, 2621–2633. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gerloff, D.; Lützkendorf, J.; Moritz, R.K.C.; Wersig, T.; Mäder, K.; Müller, L.P.; Sunderkötter, C. Melanoma-Derived Exosomal miR-125b-5p Educates Tumor Associated Macrophages (TAMs) by Targeting Lysosomal Acid Lipase A (LIPA). Cancers 2020, 12, 464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bardi, G.T.; Smith, M.A.; Hood, J.L. Melanoma exosomes promote mixed M1 and M2 macrophage polarization. Cytokine 2018, 105, 63–72. [Google Scholar] [CrossRef] [PubMed]
- Chen, G.; Huang, A.C.; Zhang, W.; Zhang, G.; Wu, M.; Xu, W.; Yu, Z.; Yang, J.; Wang, B.; Sun, H.; et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature 2018, 560, 382–386. [Google Scholar] [CrossRef] [PubMed]
- Poggio, M.; Hu, T.; Pai, C.C.; Chu, B.; Belair, C.D.; Chang, A.; Montabana, E.; Lang, U.E.; Fu, Q.; Fong, L.; et al. Suppression of Exosomal PD-L1 Induces Systemic Anti-tumor Immunity and Memory. Cell 2019, 177, 414–427.e413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Isola, A.L.; Eddy, K.; Zembrzuski, K.; Goydos, J.S.; Chen, S. Exosomes released by metabotropic glutamate receptor 1 (GRM1) expressing melanoma cells increase cell migration and invasiveness. Oncotarget 2017, 9, 1187–1199. [Google Scholar] [CrossRef] [Green Version]
- Green, J.; Ariyan, C. Update on immunotherapy in melanoma. Surg. Oncol. Clin. N. Am. 2015, 24, 337–346. [Google Scholar] [CrossRef]
- Di Trolio, R.; Simeone, E.; Di Lorenzo, G.; Buonerba, C.; Ascierto, P.A. The use of interferon in melanoma patients: A systematic review. Cytokine Growth Factor Rev. 2015, 26, 203–212. [Google Scholar] [CrossRef]
- Morton, D.L.; Eilber, F.R.; Holmes, E.C.; Hunt, J.S.; Ketcham, A.S.; Silverstein, M.J.; Sparks, F.C. BCG immunotherapy of malignant melanoma: Summary of a seven-year experience. Ann. Surg. 1974, 180, 635–643. [Google Scholar] [CrossRef]
- Choudhry, H.; Helmi, N.; Abdulaal, W.H.; Zeyadi, M.; Zamzami, M.A.; Wu, W.; Mahmoud, M.M.; Warsi, M.K.; Rasool, M.; Jamal, M.S. Prospects of IL-2 in Cancer Immunotherapy. BioMed Res. Int. 2018, 2018, 9056173. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.; Margolin, K. Tumor-infiltrating lymphocytes in melanoma. Curr. Oncol. Rep. 2012, 14, 468–474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hou, B.; Tang, Y.; Li, W.; Zeng, Q.; Chang, D. Efficiency of CAR-T Therapy for Treatment of Solid Tumor in Clinical Trials: A Meta-Analysis. Dis. Markers 2019, 2019, 3425291. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morgan, R.A.; Dudley, M.E.; Rosenberg, S.A. Adoptive cell therapy: Genetic modification to redirect effector cell specificity. Cancer J. 2010, 16, 336–341. [Google Scholar] [CrossRef] [PubMed]
- Eisenhauer, E.A.; Therasse, P.; Bogaerts, J.; Schwartz, L.H.; Sargent, D.; Ford, R.; Dancey, J.; Arbuck, S.; Gwyther, S.; Mooney, M.; et al. New response evaluation criteria in solid tumours: Revised RECIST guideline (version 1.1). Eur. J. Cancer 2009, 45, 228–247. [Google Scholar] [CrossRef] [PubMed]
- Wolchok, J.D.; Hoos, A.; O’Day, S.; Weber, J.S.; Hamid, O.; Lebbé, C.; Maio, M.; Binder, M.; Bohnsack, O.; Nichol, G.; et al. Guidelines for the evaluation of immune therapy activity in solid tumors: Immune-related response criteria. Clin. Cancer Res. 2009, 15, 7412–7420. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seymour, L.; Bogaerts, J.; Perrone, A.; Ford, R.; Schwartz, L.H.; Mandrekar, S.; Lin, N.U.; Litière, S.; Dancey, J.; Chen, A.; et al. iRECIST: Guidelines for response criteria for use in trials testing immunotherapeutics. Lancet Oncol. 2017, 18, e143–e152. [Google Scholar] [CrossRef] [Green Version]
- Klocke, K.; Sakaguchi, S.; Holmdahl, R.; Wing, K. Induction of autoimmune disease by deletion of CTLA-4 in mice in adulthood. Proc. Natl. Acad. Sci. USA 2016, 113, E2383–E2392. [Google Scholar] [CrossRef] [Green Version]
- Zhang, J.; Medeiros, L.J.; Young, K.H. Cancer Immunotherapy in Diffuse Large B-Cell Lymphoma. Front. Oncol. 2018, 8, 351. [Google Scholar] [CrossRef] [Green Version]
- Guntermann, C.; Alexander, D.R. CTLA-4 suppresses proximal TCR signaling in resting human CD4(+) T cells by inhibiting ZAP-70 Tyr(319) phosphorylation: A potential role for tyrosine phosphatases. J. Immunol. 2002, 168, 4420–4429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chambers, C.A.; Kuhns, M.S.; Egen, J.G.; Allison, J.P. CTLA-4-mediated inhibition in regulation of T cell responses: Mechanisms and manipulation in tumor immunotherapy. Annu. Rev. Immunol. 2001, 19, 565–594. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- He, M.; Chai, Y.; Qi, J.; Zhang, C.W.H.; Tong, Z.; Shi, Y.; Yan, J.; Tan, S.; Gao, G.F. Remarkably similar CTLA-4 binding properties of therapeutic ipilimumab and tremelimumab antibodies. Oncotarget 2017, 8, 67129–67139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kvistborg, P.; Philips, D.; Kelderman, S.; Hageman, L.; Ottensmeier, C.; Joseph-Pietras, D.; Welters, M.J.; Van der Burg, S.; Kapiteijn, E.; Michielin, O.; et al. Anti-CTLA-4 therapy broadens the melanoma-reactive CD8+ T cell response. Sci. Transl. Med. 2014, 6, 254ra128. [Google Scholar] [CrossRef]
- Cha, E.; Klinger, M.; Hou, Y.; Cummings, C.; Ribas, A.; Faham, M.; Fong, L. Improved survival with T cell clonotype stability after anti-CTLA-4 treatment in cancer patients. Sci. Transl. Med. 2014, 6, 238ra270. [Google Scholar] [CrossRef] [Green Version]
- Shin, J.H.; Park, H.B.; Oh, Y.M.; Lim, D.P.; Lee, J.E.; Seo, H.H.; Lee, S.J.; Eom, H.S.; Kim, I.H.; Lee, S.H.; et al. Positive conversion of negative signaling of CTLA4 potentiates antitumor efficacy of adoptive T-cell therapy in murine tumor models. Blood 2012, 119, 5678–5687. [Google Scholar] [CrossRef] [Green Version]
- Friese, C.; Harbst, K.; Borch, T.H.; Westergaard, M.C.W.; Pedersen, M.; Kverneland, A.; Jönsson, G.; Donia, M.; Svane, I.M.; Met, Ö. CTLA-4 blockade boosts the expansion of tumor-reactive CD8(+) tumor-infiltrating lymphocytes in ovarian cancer. Sci. Rep. 2020, 10, 3914. [Google Scholar] [CrossRef] [Green Version]
- Kearney, E.R.; Walunas, T.L.; Karr, R.W.; Morton, P.A.; Loh, D.Y.; Bluestone, J.A.; Jenkins, M.K. Antigen-dependent clonal expansion of a trace population of antigen-specific CD4+ T cells in vivo is dependent on CD28 costimulation and inhibited by CTLA-4. J. Immunol. 1995, 155, 1032–1036. [Google Scholar]
- Ng Tang, D.; Shen, Y.; Sun, J.; Wen, S.; Wolchok, J.D.; Yuan, J.; Allison, J.P.; Sharma, P. Increased frequency of ICOS+ CD4 T cells as a pharmacodynamic biomarker for anti-CTLA-4 therapy. Cancer Immunol. Res. 2013, 1, 229–234. [Google Scholar] [CrossRef] [Green Version]
- Simpson, T.R.; Li, F.; Montalvo-Ortiz, W.; Sepulveda, M.A.; Bergerhoff, K.; Arce, F.; Roddie, C.; Henry, J.Y.; Yagita, H.; Wolchok, J.D.; et al. Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. J. Exp. Med. 2013, 210, 1695–1710. [Google Scholar] [CrossRef]
- Romano, E.; Kusio-Kobialka, M.; Foukas, P.G.; Baumgaertner, P.; Meyer, C.; Ballabeni, P.; Michielin, O.; Weide, B.; Romero, P.; Speiser, D.E. Ipilimumab-dependent cell-mediated cytotoxicity of regulatory T cells ex vivo by nonclassical monocytes in melanoma patients. Proc. Natl. Acad. Sci. USA 2015, 112, 6140–6145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ingram, J.R.; Blomberg, O.S.; Rashidian, M.; Ali, L.; Garforth, S.; Fedorov, E.; Fedorov, A.A.; Bonanno, J.B.; Le Gall, C.; Crowley, S.; et al. Anti-CTLA-4 therapy requires an Fc domain for efficacy. Proc. Natl. Acad. Sci. USA 2018, 115, 3912–3917. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arce Vargas, F.; Furness, A.J.S.; Litchfield, K.; Joshi, K.; Rosenthal, R.; Ghorani, E.; Solomon, I.; Lesko, M.H.; Ruef, N.; Roddie, C.; et al. Fc Effector Function Contributes to the Activity of Human Anti-CTLA-4 Antibodies. Cancer Cell 2018, 33, 649–663.e644. [Google Scholar] [CrossRef] [PubMed]
- Subudhi, S.K.; Aparicio, A.; Gao, J.; Zurita, A.J.; Araujo, J.C.; Logothetis, C.J.; Tahir, S.A.; Korivi, B.R.; Slack, R.S.; Vence, L.; et al. Clonal expansion of CD8 T cells in the systemic circulation precedes development of ipilimumab-induced toxicities. Proc. Natl. Acad. Sci. USA 2016, 113, 11919–11924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nishida, N.; Kudo, M. Liver damage related to immune checkpoint inhibitors. Hepatol. Int. 2019, 13, 248–252. [Google Scholar] [CrossRef]
- Ramos-Casals, M.; Brahmer, J.R.; Callahan, M.K.; Flores-Chávez, A.; Keegan, N.; Khamashta, M.A.; Lambotte, O.; Mariette, X.; Prat, A.; Suárez-Almazor, M.E. Immune-related adverse events of checkpoint inhibitors. Nat. Rev. Dis. Prim. 2020, 6, 38. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Eroglu, Z.; Kim, D.W.; Wang, X.; Camacho, L.H.; Chmielowski, B.; Seja, E.; Villanueva, A.; Ruchalski, K.; Glaspy, J.A.; Kim, K.B.; et al. Long term survival with cytotoxic T lymphocyte-associated antigen 4 blockade using tremelimumab. Eur. J. Cancer 2015, 51, 2689–2697. [Google Scholar] [CrossRef] [Green Version]
- Gutzmer, R.; Stroyakovskiy, D.; Gogas, H.; Robert, C.; Lewis, K.; Protsenko, S.; Pereira, R.P.; Eigentler, T.; Rutkowski, P.; Demidov, L.; et al. Atezolizumab, vemurafenib, and cobimetinib as first-line treatment for unresectable advanced BRAF(V600) mutation-positive melanoma (IMspire150): Primary analysis of the randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2020, 395, 1835–1844. [Google Scholar] [CrossRef]
- Okazaki, T.; Honjo, T. PD-1 and PD-1 ligands: From discovery to clinical application. Int. Immunol. 2007, 19, 813–824. [Google Scholar] [CrossRef] [Green Version]
- Alsaab, H.O.; Sau, S.; Alzhrani, R.; Tatiparti, K.; Bhise, K.; Kashaw, S.K.; Iyer, A.K. PD-1 and PD-L1 Checkpoint Signaling Inhibition for Cancer Immunotherapy: Mechanism, Combinations, and Clinical Outcome. Front. Pharmacol. 2017, 8, 561. [Google Scholar] [CrossRef] [PubMed]
- Haanen, J.B. Immunotherapy of melanoma. Eur. J. Cancer Suppl. 2013, 11, 97–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, H.T.; Lee, S.H.; Heo, Y.S. Molecular Interactions of Antibody Drugs Targeting PD-1, PD-L1, and CTLA-4 in Immuno-Oncology. Molecules 2019, 24, 1190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Iwai, Y.; Ishida, M.; Tanaka, Y.; Okazaki, T.; Honjo, T.; Minato, N. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc. Natl. Acad. Sci. USA 2002, 99, 12293–12297. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Iwai, Y.; Terawaki, S.; Honjo, T. PD-1 blockade inhibits hematogenous spread of poorly immunogenic tumor cells by enhanced recruitment of effector T cells. Int. Immunol. 2005, 17, 133–144. [Google Scholar] [CrossRef] [Green Version]
- Hirano, F.; Kaneko, K.; Tamura, H.; Dong, H.; Wang, S.; Ichikawa, M.; Rietz, C.; Flies, D.B.; Lau, J.S.; Zhu, G.; et al. Blockade of B7-H1 and PD-1 by monoclonal antibodies potentiates cancer therapeutic immunity. Cancer Res. 2005, 65, 1089–1096. [Google Scholar]
- Li, J.; Gu, J. Efficacy and safety of ipilimumab for treating advanced melanoma: A systematic review and meta-analysis. J. Clin. Pharm. Ther. 2019, 44, 420–429. [Google Scholar] [CrossRef]
- De Sousa Linhares, A.; Battin, C.; Jutz, S.; Leitner, J.; Hafner, C.; Tobias, J.; Wiedermann, U.; Kundi, M.; Zlabinger, G.J.; Grabmeier-Pfistershammer, K.; et al. Therapeutic PD-L1 antibodies are more effective than PD-1 antibodies in blocking PD-1/PD-L1 signaling. Sci. Rep. 2019, 9, 11472. [Google Scholar] [CrossRef]
- Carretero-González, A.; Lora, D.; Ghanem, I.; Zugazagoitia, J.; Castellano, D.; Sepúlveda, J.M.; López-Martin, J.A.; Paz-Ares, L.; de Velasco, G. Analysis of response rate with ANTI PD1/PD-L1 monoclonal antibodies in advanced solid tumors: A meta-analysis of randomized clinical trials. Oncotarget 2018, 9, 8706–8715. [Google Scholar] [CrossRef] [Green Version]
- Kitano, S.; Nakayama, T.; Yamashita, M. Biomarkers for Immune Checkpoint Inhibitors in Melanoma. Front. Oncol. 2018, 8, 270. [Google Scholar] [CrossRef]
- Ye, Y.; Jing, Y.; Li, L.; Mills, G.B.; Diao, L.; Liu, H.; Han, L. Sex-associated molecular differences for cancer immunotherapy. Nat. Commun. 2020, 11, 1779. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Castro, A.; Pyke, R.M.; Zhang, X.; Thompson, W.K.; Day, C.P.; Alexandrov, L.B.; Zanetti, M.; Carter, H. Strength of immune selection in tumors varies with sex and age. Nat. Commun. 2020, 11, 4128. [Google Scholar] [CrossRef]
- Nakamura, Y. Biomarkers for Immune Checkpoint Inhibitor-Mediated Tumor Response and Adverse Events. Front. Med. 2019, 6, 119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liakou, C.I.; Kamat, A.; Tang, D.N.; Chen, H.; Sun, J.; Troncoso, P.; Logothetis, C.; Sharma, P. CTLA-4 blockade increases IFNgamma-producing CD4+ICOShi cells to shift the ratio of effector to regulatory T cells in cancer patients. Proc. Natl. Acad. Sci. USA 2008, 105, 14987–14992. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carthon, B.C.; Wolchok, J.D.; Yuan, J.; Kamat, A.; Ng Tang, D.S.; Sun, J.; Ku, G.; Troncoso, P.; Logothetis, C.J.; Allison, J.P.; et al. Preoperative CTLA-4 blockade: Tolerability and immune monitoring in the setting of a presurgical clinical trial. Clin. Cancer Res. 2010, 16, 2861–2871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tallerico, R.; Cristiani, C.M.; Staaf, E.; Garofalo, C.; Sottile, R.; Capone, M.; Pico de Coaña, Y.; Madonna, G.; Palella, E.; Wolodarski, M.; et al. IL-15, TIM-3 and NK cells subsets predict responsiveness to anti-CTLA-4 treatment in melanoma patients. Oncoimmunology 2017, 6, e1261242. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hamid, O.; Schmidt, H.; Nissan, A.; Ridolfi, L.; Aamdal, S.; Hansson, J.; Guida, M.; Hyams, D.M.; Gómez, H.; Bastholt, L.; et al. A prospective phase II trial exploring the association between tumor microenvironment biomarkers and clinical activity of ipilimumab in advanced melanoma. J. Transl. Med. 2011, 9, 204. [Google Scholar] [CrossRef] [Green Version]
- Pistillo, M.P.; Fontana, V.; Morabito, A.; Dozin, B.; Laurent, S.; Carosio, R.; Banelli, B.; Ferrero, F.; Spano, L.; Tanda, E.; et al. Soluble CTLA-4 as a favorable predictive biomarker in metastatic melanoma patients treated with ipilimumab: An Italian melanoma intergroup study. Cancer Immunol. Immunother. 2019, 68, 97–107. [Google Scholar] [CrossRef]
- Ji, R.R.; Chasalow, S.D.; Wang, L.; Hamid, O.; Schmidt, H.; Cogswell, J.; Alaparthy, S.; Berman, D.; Jure-Kunkel, M.; Siemers, N.O.; et al. An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer Immunol. Immunother. 2012, 61, 1019–1031. [Google Scholar] [CrossRef]
- Sunshine, J.C.; Nguyen, P.L.; Kaunitz, G.J.; Cottrell, T.R.; Berry, S.; Esandrio, J.; Xu, H.; Ogurtsova, A.; Bleich, K.B.; Cornish, T.C.; et al. PD-L1 Expression in Melanoma: A Quantitative Immunohistochemical Antibody Comparison. Clin. Cancer Res. 2017, 23, 4938–4944. [Google Scholar] [CrossRef] [Green Version]
- Daud, A.I.; Wolchok, J.D.; Robert, C.; Hwu, W.J.; Weber, J.S.; Ribas, A.; Hodi, F.S.; Joshua, A.M.; Kefford, R.; Hersey, P.; et al. Programmed Death-Ligand 1 Expression and Response to the Anti-Programmed Death 1 Antibody Pembrolizumab in Melanoma. J. Clin. Oncol. 2016, 34, 4102–4109. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed] [Green Version]
- Cottrell, T.R.; Taube, J.M. PD-L1 and Emerging Biomarkers in Immune Checkpoint Blockade Therapy. Cancer J. 2018, 24, 41–46. [Google Scholar] [CrossRef] [PubMed]
- Madore, J.; Vilain, R.E.; Menzies, A.M.; Kakavand, H.; Wilmott, J.S.; Hyman, J.; Yearley, J.H.; Kefford, R.F.; Thompson, J.F.; Long, G.V.; et al. PD-L1 expression in melanoma shows marked heterogeneity within and between patients: Implications for anti-PD-1/PD-L1 clinical trials. Pigment Cell Melanoma Res. 2015, 28, 245–253. [Google Scholar] [CrossRef] [PubMed]
- Wolf, Y.; Bartok, O.; Patkar, S.; Eli, G.B.; Cohen, S.; Litchfield, K.; Levy, R.; Jiménez-Sánchez, A.; Trabish, S.; Lee, J.S.; et al. UVB-Induced Tumor Heterogeneity Diminishes Immune Response in Melanoma. Cell 2019, 179, 219–235.e221. [Google Scholar] [CrossRef] [Green Version]
- Gros, A.; Parkhurst, M.R.; Tran, E.; Pasetto, A.; Robbins, P.F.; Ilyas, S.; Prickett, T.D.; Gartner, J.J.; Crystal, J.S.; Roberts, I.M.; et al. Prospective identification of neoantigen-specific lymphocytes in the peripheral blood of melanoma patients. Nat. Med. 2016, 22, 433–438. [Google Scholar] [CrossRef]
- Goodman, A.M.; Kato, S.; Bazhenova, L.; Patel, S.P.; Frampton, G.M.; Miller, V.; Stephens, P.J.; Daniels, G.A.; Kurzrock, R. Tumor Mutational Burden as an Independent Predictor of Response to Immunotherapy in Diverse Cancers. Mol. Cancer Ther. 2017, 16, 2598–2608. [Google Scholar] [CrossRef] [Green Version]
- Chalmers, Z.R.; Connelly, C.F.; Fabrizio, D.; Gay, L.; Ali, S.M.; Ennis, R.; Schrock, A.; Campbell, B.; Shlien, A.; Chmielecki, J.; et al. Analysis of 100,000 human cancer genomes reveals the landscape of tumor mutational burden. Genome Med. 2017, 9, 34. [Google Scholar] [CrossRef]
- Panda, A.; Betigeri, A.; Subramanian, K.; Ross, J.S.; Pavlick, D.C.; Ali, S.; Markowski, P.; Silk, A.; Kaufman, H.L.; Lattime, E.; et al. Identifying a Clinically Applicable Mutational Burden Threshold as a Potential Biomarker of Response to Immune Checkpoint Therapy in Solid Tumors. JCO Precis. Oncol. 2017, 1, 1–13. [Google Scholar] [CrossRef]
- Plesca, I.; Tunger, A.; Müller, L.; Wehner, R.; Lai, X.; Grimm, M.O.; Rutella, S.; Bachmann, M.; Schmitz, M. Characteristics of Tumor-Infiltrating Lymphocytes Prior to and During Immune Checkpoint Inhibitor Therapy. Front. Immunol. 2020, 11, 364. [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]
- Weide, B.; Martens, A.; Hassel, J.C.; Berking, C.; Postow, M.A.; Bisschop, K.; Simeone, E.; Mangana, J.; Schilling, B.; Di Giacomo, A.M.; et al. Baseline Biomarkers for Outcome of Melanoma Patients Treated with Pembrolizumab. Clin. Cancer Res. 2016, 22, 5487–5496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, J.; Mahoney, K.M.; Giobbie-Hurder, A.; Zhao, F.; Lee, S.; Liao, X.; Rodig, S.; Li, J.; Wu, X.; Butterfield, L.H.; et al. Soluble PD-L1 as a Biomarker in Malignant Melanoma Treated with Checkpoint Blockade. Cancer Immunol. Res. 2017, 5, 480–492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103. [Google Scholar] [CrossRef] [Green Version]
- Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.L.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef] [Green Version]
- Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [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. [Google Scholar] [CrossRef] [Green Version]
- Maleki Vareki, S. High and low mutational burden tumors versus immunologically hot and cold tumors and response to immune checkpoint inhibitors. J. Immunother. Cancer 2018, 6, 157. [Google Scholar] [CrossRef]
- Huang, A.C.; Postow, M.A.; Orlowski, R.J.; Mick, R.; Bengsch, B.; Manne, S.; Xu, W.; Harmon, S.; Giles, J.R.; Wenz, B.; et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 2017, 545, 60–65. [Google Scholar] [CrossRef] [Green Version]
- Rossi, E.; Schinzari, G.; Maiorano, B.A.; Indellicati, G.; Di Stefani, A.; Pagliara, M.M.; Fragomeni, S.M.; De Luca, E.V.; Sammarco, M.G.; Garganese, G.; et al. Efficacy of immune checkpoint inhibitors in different types of melanoma. Hum. Vaccin. Immunother. 2020, 1–10. [Google Scholar] [CrossRef]
- 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] [PubMed] [Green Version]
- Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef] [PubMed]
- Perica, K.; Varela, J.C.; Oelke, M.; Schneck, J.P. Adoptive T Cell Immunotherapy for Cancer. Rambam Maimonides Med. J. 2015, 6, e0004. [Google Scholar] [CrossRef] [PubMed]
- Rosenberg, S.A.; Packard, B.S.; Aebersold, P.M.; Solomon, D.; Topalian, S.L.; Toy, S.T.; Simon, P.; Lotze, M.T.; Yang, J.C.; Seipp, C.A.; et al. Use of tumor-infiltrating lymphocytes and interleukin-2 in the immunotherapy of patients with metastatic melanoma. A preliminary report. N. Engl. J. Med. 1988, 319, 1676–1680. [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]
- Besser, M.J.; Shapira-Frommer, R.; Treves, A.J.; Zippel, D.; Itzhaki, O.; Hershkovitz, L.; Levy, D.; Kubi, A.; Hovav, E.; Chermoshniuk, N.; et al. Clinical responses in a phase II study using adoptive transfer of short-term cultured tumor infiltration lymphocytes in metastatic melanoma patients. Clin. Cancer Res. 2010, 16, 2646–2655. [Google Scholar] [CrossRef] [Green Version]
- Rohaan, M.W.; Van den Berg, J.H.; Kvistborg, P.; Haanen, J. Adoptive transfer of tumor-infiltrating lymphocytes in melanoma: A viable treatment option. J. Immunother. Cancer 2018, 6, 102. [Google Scholar] [CrossRef]
- Morgan, R.A.; Dudley, M.E.; Wunderlich, J.R.; Hughes, M.S.; Yang, J.C.; Sherry, R.M.; Royal, R.E.; Topalian, S.L.; Kammula, U.S.; Restifo, N.P.; et al. Cancer regression in patients after transfer of genetically engineered lymphocytes. Science 2006, 314, 126–129. [Google Scholar] [CrossRef] [Green Version]
- Ott, P.A.; Dotti, G.; Yee, C.; Goff, S.L. An Update on Adoptive T-Cell Therapy and Neoantigen Vaccines. Am. Soc. Clin. Oncol. Educ. Book 2019, 39, e70–e78. [Google Scholar] [CrossRef]
- Johnson, L.A.; Morgan, R.A.; Dudley, M.E.; Cassard, L.; Yang, J.C.; Hughes, M.S.; Kammula, U.S.; Royal, R.E.; Sherry, R.M.; Wunderlich, J.R.; et al. Gene therapy with human and mouse T-cell receptors mediates cancer regression and targets normal tissues expressing cognate antigen. Blood 2009, 114, 535–546. [Google Scholar] [CrossRef] [Green Version]
- Pfefferle, A.; Huntington, N.D. You Have Got a Fast CAR: Chimeric Antigen Receptor NK Cells in Cancer Therapy. Cancers 2020, 12, 706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lauss, M.; Donia, M.; Harbst, K.; Andersen, R.; Mitra, S.; Rosengren, F.; Salim, M.; Vallon-Christersson, J.; Törngren, T.; Kvist, A.; et al. Mutational and putative neoantigen load predict clinical benefit of adoptive T cell therapy in melanoma. Nat. Commun. 2017, 8, 1738. [Google Scholar] [CrossRef] [Green Version]
- Pol, J.; Kroemer, G.; Galluzzi, L. First oncolytic virus approved for melanoma immunotherapy. Oncoimmunology 2016, 5, e1115641. [Google Scholar] [CrossRef] [Green Version]
- Bommareddy, P.K.; Shettigar, M.; Kaufman, H.L. Integrating oncolytic viruses in combination cancer immunotherapy. Nat. Rev. Immunol. 2018, 18, 498–513. [Google Scholar] [CrossRef] [PubMed]
- Zamarin, D.; Holmgaard, R.B.; Subudhi, S.K.; Park, J.S.; Mansour, M.; Palese, P.; Merghoub, T.; Wolchok, J.D.; Allison, J.P. Localized oncolytic virotherapy overcomes systemic tumor resistance to immune checkpoint blockade immunotherapy. Sci. Transl. Med. 2014, 6, 226ra232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Orloff, M. Spotlight on talimogene laherparepvec for the treatment of melanoma lesions in the skin and lymph nodes. Oncolytic Virother. 2016, 5, 91–98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Andtbacka, R.H.; Kaufman, H.L.; Collichio, F.; Amatruda, T.; Senzer, N.; Chesney, J.; Delman, K.A.; Spitler, L.E.; Puzanov, I.; Agarwala, S.S.; et al. Talimogene Laherparepvec Improves Durable Response Rate in Patients with Advanced Melanoma. J. Clin. Oncol. 2015, 33, 2780–2788. [Google Scholar] [CrossRef] [PubMed]
- Masoud, S.J.; Hu, J.B.; Beasley, G.M.; Stewart, J.H.t.; Mosca, P.J. Efficacy of Talimogene Laherparepvec (T-VEC) Therapy in Patients with In-Transit Melanoma Metastasis Decreases with Increasing Lesion Size. Ann. Surg. Oncol. 2019, 26, 4633–4641. [Google Scholar] [CrossRef] [PubMed]
- Andtbacka, R.H.; Ross, M.; Puzanov, I.; Milhem, M.; Collichio, F.; Delman, K.A.; Amatruda, T.; Zager, J.S.; Cranmer, L.; Hsueh, E.; et al. Patterns of Clinical Response with Talimogene Laherparepvec (T-VEC) in Patients with Melanoma Treated in the OPTiM Phase III Clinical Trial. Ann. Surg. Oncol. 2016, 23, 4169–4177. [Google Scholar] [CrossRef] [Green Version]
- Achard, C.; Surendran, A.; Wedge, M.E.; Ungerechts, G.; Bell, J.; Ilkow, C.S. Lighting a Fire in the Tumor Microenvironment Using Oncolytic Immunotherapy. EBioMedicine 2018, 31, 17–24. [Google Scholar] [CrossRef] [Green Version]
- Eggermont, A.M.M.; Crittenden, M.; Wargo, J. Combination Immunotherapy Development in Melanoma. Am. Soc. Clin. Oncol. Educ. Book 2018, 38, 197–207. [Google Scholar] [CrossRef]
- Wolchok, J.D.; Kluger, H.; Callahan, M.K.; Postow, M.A.; Rizvi, N.A.; Lesokhin, A.M.; Segal, N.H.; Ariyan, C.E.; Gordon, R.A.; Reed, K.; et al. Nivolumab plus ipilimumab in advanced melanoma. N. Engl. J. Med. 2013, 369, 122–133. [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]
- 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]
- Hodi, F.S.; Chiarion-Sileni, V.; Gonzalez, R.; Grob, J.J.; Rutkowski, P.; Cowey, C.L.; Lao, C.D.; Schadendorf, D.; Wagstaff, J.; Dummer, R.; et al. Nivolumab plus ipilimumab or nivolumab alone versus ipilimumab alone in advanced melanoma (CheckMate 067): 4-year outcomes of a multicentre, randomised, phase 3 trial. Lancet Oncol. 2018, 19, 1480–1492. [Google Scholar] [CrossRef]
- Carlino, M.S.; Menzies, A.M.; Atkinson, V.; Cebon, J.S.; Jameson, M.B.; Fitzharris, B.M.; McNeil, C.M.; Hill, A.G.; Ribas, A.; Atkins, M.B.; et al. Long-term Follow-up of Standard-Dose Pembrolizumab Plus Reduced-Dose Ipilimumab in Patients with Advanced Melanoma: KEYNOTE-029 Part 1B. Clin. Cancer Res. 2020, 26, 5086–5091. [Google Scholar] [CrossRef]
- Das, R.; Verma, R.; Sznol, M.; Boddupalli, C.S.; Gettinger, S.N.; Kluger, H.; Callahan, M.; Wolchok, J.D.; Halaban, R.; Dhodapkar, M.V.; et al. Combination therapy with anti-CTLA-4 and anti-PD-1 leads to distinct immunologic changes in vivo. J. Immunol. 2015, 194, 950–959. [Google Scholar] [CrossRef] [Green Version]
- 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]
- Shi, L.Z.; Goswami, S.; Fu, T.; Guan, B.; Chen, J.; Xiong, L.; Zhang, J.; Ng Tang, D.; Zhang, X.; Vence, L.; et al. Blockade of CTLA-4 and PD-1 Enhances Adoptive T-cell Therapy Efficacy in an ICOS-Mediated Manner. Cancer Immunol. Res. 2019, 7, 1803–1812. [Google Scholar] [CrossRef] [Green Version]
- Kverneland, A.H.; Pedersen, M.; Westergaard, M.C.W.; Nielsen, M.; Borch, T.H.; Olsen, L.R.; Aasbjerg, G.; Santegoets, S.J.; van der Burg, S.H.; Milne, K.; et al. Adoptive cell therapy in combination with checkpoint inhibitors in ovarian cancer. Oncotarget 2020, 11, 2092–2105. [Google Scholar] [CrossRef]
- Koya, R.C.; Mok, S.; Otte, N.; Blacketor, K.J.; Comin-Anduix, B.; Tumeh, P.C.; Minasyan, A.; Graham, N.A.; Graeber, T.G.; Chodon, T.; et al. BRAF inhibitor vemurafenib improves the antitumor activity of adoptive cell immunotherapy. Cancer Res. 2012, 72, 3928–3937. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 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
Eddy, K.; Chen, S. Overcoming Immune Evasion in Melanoma. Int. J. Mol. Sci. 2020, 21, 8984. https://doi.org/10.3390/ijms21238984
Eddy K, Chen S. Overcoming Immune Evasion in Melanoma. International Journal of Molecular Sciences. 2020; 21(23):8984. https://doi.org/10.3390/ijms21238984
Chicago/Turabian StyleEddy, Kevinn, and Suzie Chen. 2020. "Overcoming Immune Evasion in Melanoma" International Journal of Molecular Sciences 21, no. 23: 8984. https://doi.org/10.3390/ijms21238984
APA StyleEddy, K., & Chen, S. (2020). Overcoming Immune Evasion in Melanoma. International Journal of Molecular Sciences, 21(23), 8984. https://doi.org/10.3390/ijms21238984