Rejection of benign melanocytic nevi by nevus-resident CD4+ T cells

Nevi (moles) harbor resident immune cells that can be activated to eradicate moles and, potentially, to prevent melanoma. Melanoma and melanocytic nevi harbor shared lineage-specific antigens and oncogenic mutations. Yet, the relationship between the immune system and melanocytic nevi is unclear. Using a patient-derived xenograft (PDX) model, we found that 81.8% of the transplanted nevi underwent spontaneous regression, while peripheral skin remained intact. Nevus-resident CD4+ T helper 1 cells, which exhibited a massive clonal expansion to melanocyte-specific antigens, were responsible for nevus rejection. Boosting regulatory T cell suppressive function with low-dose exogenous human interleukin-2 injection or treatment with a human leukocyte antigen (HLA) class II–blocking antibody prevented nevus rejection. Notably, mice with rejected nevus PDXs were protected from melanoma tumor growth. We detected a parallel CD4+ T cell–dominant immunity in clinically regressing melanocytic nevi. These findings reveal a mechanistic explanation for spontaneous nevus regression in humans and posit the activation of nevus-resident CD4+ effector T cells as a novel strategy for melanoma immunoprevention and treatment.

[1]  Qianya Su,et al.  IFN-γ induces apoptosis in human melanocytes by activating the JAK1/STAT1 signaling pathway , 2020, Molecular medicine reports.

[2]  Chun Jimmie Ye,et al.  Intratumoral CD4+ T Cells Mediate Anti-tumor Cytotoxicity in Human Bladder Cancer , 2020, Cell.

[3]  E. Richardson,et al.  Revisiting the role of CD4+ T cells in cancer immunotherapy—new insights into old paradigms , 2020, Cancer Gene Therapy.

[4]  M. Lenardo,et al.  A guide to cancer immunotherapy: from T cell basic science to clinical practice , 2020, Nature Reviews Immunology.

[5]  George Coukos,et al.  Robust prediction of HLA class II epitopes by deep motif deconvolution of immunopeptidomes , 2019, Nature Biotechnology.

[6]  Cheryl F. Lichti,et al.  MHC-II neoantigens shape tumor immunity and response to immunotherapy , 2019, Nature.

[7]  R. L. Riding,et al.  The Role of Memory CD8+ T Cells in Vitiligo , 2019, The Journal of Immunology.

[8]  S. Snapper,et al.  Low-Dose Interleukin-2 Ameliorates Colitis in a Preclinical Humanized Mouse Model , 2019, Cellular and molecular gastroenterology and hepatology.

[9]  C. Cetrulo,et al.  Chimeric antigen receptor costimulation domains modulate human regulatory T cell function. , 2019, JCI insight.

[10]  A. Chinnaiyan,et al.  CD8+ T cells regulate tumor ferroptosis during cancer immunotherapy , 2019, Nature.

[11]  H. Mitsui,et al.  Novel immune signatures associated with dysplastic naevi and primary cutaneous melanoma in human skin , 2018, Experimental dermatology.

[12]  J. Pearson,et al.  Whole genome sequencing of melanomas in adolescent and young adults reveals distinct mutation landscapes and the potential role of germline variants in disease susceptibility , 2018, International journal of cancer.

[13]  C. Hsieh,et al.  Central CD4+ T cell tolerance: deletion versus regulatory T cell differentiation , 2018, Nature Reviews Immunology.

[14]  R. Hargest,et al.  The nature of the human T cell response to the cancer antigen 5T4 is determined by the balance of regulatory and inflammatory T cells of the same antigen-specificity: implications for vaccine design , 2018, Cancer Immunology, Immunotherapy.

[15]  S. Wingett,et al.  FastQ Screen: A tool for multi-genome mapping and quality control , 2018, F1000Research.

[16]  J. Borst,et al.  CD4+ T cell help in cancer immunology and immunotherapy , 2018, Nature Reviews Immunology.

[17]  Robert L. Judson,et al.  Genomic and Transcriptomic Analysis Reveals Incremental Disruption of Key Signaling Pathways during Melanoma Evolution. , 2018, Cancer cell.

[18]  H. Soyer,et al.  Whole-Exome Sequencing of Acquired Nevi Identifies Mechanisms for Development and Maintenance of Benign Neoplasms. , 2018, The Journal of investigative dermatology.

[19]  Ling-hua Meng,et al.  Oncogene-induced senescence: a double edged sword in cancer , 2018, Acta Pharmacologica Sinica.

[20]  E. Unni,et al.  17-AAG inhibits vemurafenib-associated MAP kinase activation and is synergistic with cellular immunotherapy in a murine melanoma model , 2018, PloS one.

[21]  Charles H. Yoon,et al.  An immunogenic personal neoantigen vaccine for patients with melanoma , 2017, Nature.

[22]  M. Bosenberg,et al.  Melanocytic nevi and melanoma: unraveling a complex relationship , 2017, Oncogene.

[23]  E. Jordá,et al.  Differential Clinicopathological Features in Spontaneous Regression of Melanomas and Melanocytic Naevi. , 2017, Acta dermato-venereologica.

[24]  Catherine A. Shang,et al.  Whole-genome landscapes of major melanoma subtypes , 2017, Nature.

[25]  William S. DeWitt,et al.  Immunosequencing identifies signatures of cytomegalovirus exposure history and HLA-mediated effects on the T cell repertoire , 2017, Nature Genetics.

[26]  M. Suárez-Fariñas,et al.  Discrimination of Dysplastic Nevi from Common Melanocytic Nevi by Cellular and Molecular Criteria. , 2016, The Journal of investigative dermatology.

[27]  G. Ogg,et al.  Enhanced isolation of lymphoid cells from human skin , 2016, Clinical and experimental dermatology.

[28]  T. Mariani,et al.  CD4+ T Cell Tolerance to Tissue-Restricted Self Antigens Is Mediated by Antigen-Specific Regulatory T Cells Rather Than Deletion. , 2015, Immunity.

[29]  R. Dummer,et al.  The Genetic Evolution of Melanoma from Precursor Lesions. , 2015, The New England journal of medicine.

[30]  Ana Conesa,et al.  Qualimap 2: advanced multi-sample quality control for high-throughput sequencing data , 2015, Bioinform..

[31]  J. Malvehy,et al.  Mutational status of naevus‐associated melanomas , 2015, The British journal of dermatology.

[32]  A. Conesa,et al.  Data quality aware analysis of differential expression in RNA-seq with NOISeq R/Bioc package , 2015, Nucleic acids research.

[33]  D. Klatzmann,et al.  The promise of low-dose interleukin-2 therapy for autoimmune and inflammatory diseases , 2015, Nature Reviews Immunology.

[34]  Rei Watanabe,et al.  Human skin is protected by four functionally and phenotypically discrete populations of resident and recirculating memory T cells , 2015, Science Translational Medicine.

[35]  W. Huber,et al.  Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2 , 2014, Genome Biology.

[36]  Y. Shimojima,et al.  Regulatory T Cells and the Immune Aging Process: A Mini-Review , 2013, Gerontology.

[37]  Mark M Davis,et al.  Virus-specific CD4(+) memory-phenotype T cells are abundant in unexposed adults. , 2013, Immunity.

[38]  L. Zender,et al.  T-helper-1-cell cytokines drive cancer into senescence , 2013, Nature.

[39]  J. Lambert,et al.  Melanocyte-specific immune response in a patient with multiple regressing nevi and a history of melanoma. , 2011, Anticancer research.

[40]  Colin N. Dewey,et al.  RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome , 2011, BMC Bioinformatics.

[41]  J. Reis-Filho,et al.  Oncogenic Braf induces melanocyte senescence and melanoma in mice. , 2009, Cancer cell.

[42]  S. Jones,et al.  Many roads lead to oncogene-induced senescence , 2008, Oncogene.

[43]  R. Kennedy,et al.  Multiple roles for CD4+ T cells in anti‐tumor immune responses , 2008, Immunological reviews.

[44]  D. Fisher,et al.  Melanocyte biology and skin pigmentation , 2007, Nature.

[45]  S. Gad,et al.  Nonclinical Vehicle Use in Studies by Multiple Routes in Multiple Species , 2006, International journal of toxicology.

[46]  D. Fisher,et al.  Topical drug rescue strategy and skin protection based on the role of Mc1r in UV-induced tanning , 2006, Nature.

[47]  J. Shay,et al.  BRAFE600-associated senescence-like cell cycle arrest of human naevi , 2005, Nature.

[48]  C. Harding,et al.  The Mycobacterium tuberculosis 19-Kilodalton Lipoprotein Inhibits Gamma Interferon-Regulated HLA-DR and FcγR1 on Human Macrophages through Toll-Like Receptor 2 , 2003, Infection and Immunity.

[49]  W. Goggins,et al.  The transformation rate of moles (melanocytic nevi) into cutaneous melanoma: a population-based estimate. , 2003, Archives of dermatology.

[50]  V. Brusic,et al.  Melan-A/MART-151–73 represents an immunogenic HLA-DR4-restricted epitope recognized by melanoma-reactive CD4+ T cells , 2000 .

[51]  C. Grin,et al.  The immune response in halo nevi. , 1997, Journal of the American Academy of Dermatology.

[52]  S. Wagner,et al.  Analysis of Pmel17/gp100 expression in primary human tissue specimens: implications for melanoma immuno- and gene-therapy , 1997, Cancer Immunology, Immunotherapy.

[53]  P. Erb,et al.  Mechanism and Biological Significance of CD4‐mediated Cytotoxicity , 1995, Immunological reviews.

[54]  C. Figdor,et al.  Melanocyte lineage-specific antigen gp100 is recognized by melanoma- derived tumor-infiltrating lymphocytes , 1994, The Journal of experimental medicine.

[55]  B. Smoller,et al.  HMB‐45 recognizes stimulated melanocytes , 1989, Journal of cutaneous pathology.

[56]  R. MacKie,et al.  The number and distribution of benign pigmented moles (melanocytic naevi) in a healthy British population , 1985, The British journal of dermatology.

[57]  G. Klaus,et al.  Effects of cyclosporine on the immune system of the mouse. II. Cyclosporine inhibits the effector function of primary T helper cells, but not helper cell priming. , 1983, Transplantation.

[58]  Thomas R. Gingeras,et al.  STAR: ultrafast universal RNA-seq aligner , 2013, Bioinform..

[59]  D. Mueller Mechanisms maintaining peripheral tolerance , 2010, Nature Immunology.

[60]  M. Okada,et al.  Targeting of CD4+CD25high cells while preserving CD4+CD25low cells with low-dose chimeric anti-CD25 antibody in adoptive immunotherapy of cancer. , 2009, International journal of oncology.

[61]  A. Abbas,et al.  The enemy within: keeping self-reactive T cells at bay in the periphery , 2002, Nature Reviews Immunology.