Mycosis Fungoides and Sézary Syndrome: Microenvironment and Cancer Progression.

Mycosis fungoides and Sézary syndrome are epidermotropic cutaneous lymphomas, and both of them are rare diseases. Mycosis fungoides is the most frequent primary cutaneous lymphoma. In about 25% of patients with mycosis fungoides, the disease may progress to higher stages. The pathogenesis and risk factors of progression in mycosis fungoides and Sézary syndrome are not yet fully understood. Previous works have investigated inter- and intrapatient tumor cell heterogeneity. Here, we overview the role of the tumor microenvironment of mycosis fungoides and Sézary syndrome by describing its key components and functions. Emphasis is put on the role of the microenvironment in promoting tumor growth or antitumor immune response, as well as possible therapeutic targets. We focus on recent advances in the field and point out treatment-related alterations of the microenvironment. Deciphering the tumor microenvironment may help to develop strategies that lead to long-term disease control and cure.

[1]  Ruoyan Li,et al.  Hyperprogression of cutaneous T cell lymphoma after anti–PD-1 treatment , 2023, JCI insight.

[2]  A. Elsensohn,et al.  Cutaneous T‐cell lymphoma following dupilumab use: a systematic review , 2022, International journal of dermatology.

[3]  D. Weisenburger,et al.  Expression of immune checkpoint molecules programmed death protein 1, programmed death‐ligand 1 and inducible T‐cell co‐stimulator in mycosis fungoides and Sézary syndrome: association with disease stage and clinical outcome , 2022, The British Journal of Dermatology.

[4]  Robert W. Hsieh,et al.  Tiragolumab plus atezolizumab versus placebo plus atezolizumab as a first-line treatment for PD-L1-selected non-small-cell lung cancer (CITYSCAPE): primary and follow-up analyses of a randomised, double-blind, phase 2 study. , 2022, The Lancet. Oncology.

[5]  U. Wehkamp,et al.  The Skin Microbiome and Influencing Elements in Cutaneous T-Cell Lymphomas , 2022, Cancers.

[6]  R. Clark,et al.  CCR8 is a new therapeutic target in cutaneous T-cell lymphomas , 2022, Blood advances.

[7]  J. Dang,et al.  Targeting macrophages for enhancing CD47 blockade-elicited lymphoma clearance and overcoming tumor-induced suppression. , 2022, Blood.

[8]  L. Biard,et al.  Mogamulizumab induces long‐term immune restoration and reshapes tumour heterogeneity in Sézary syndrome * , 2022, The British journal of dermatology.

[9]  P. de la Grange,et al.  Macrophage-derived CXCL9 and CXCL11, T-cell skin homing and disease control in mogamulizumab-treated CTCL patients. , 2021, Blood.

[10]  M. Khodadoust,et al.  Resistance to Mogamulizumab Is Associated with Loss of CCR4 in Cutaneous T Cell Lymphoma , 2021, Blood.

[11]  Jie Liu,et al.  The Role of Tumor Microenvironment in Mycosis Fungoides and Sézary Syndrome , 2021, Annals of Dermatology.

[12]  L. Michel,et al.  Exploring the role of the skin microenvironment in cutaneous T-cell lymphoma using single cell RNA-sequencing. , 2021, European journal of cancer.

[13]  J. Scarisbrick,et al.  Phase II trial of atezolizumab (anti-PD-L1) in the treatment of stage IIb-IVB mycosis fungoides/Sézary syndrome patients relapsed/refractory after a previous systemic treatment (PARCT). , 2021, European journal of cancer.

[14]  A. Pileri,et al.  The Microenvironment’s Role in Mycosis Fungoides and Sézary Syndrome: From Progression to Therapeutic Implications , 2021, Cells.

[15]  G. Nolan,et al.  Highly Multiplexed Phenotyping of Immunoregulatory Proteins in the Tumor Microenvironment by CODEX Tissue Imaging , 2021, Frontiers in Immunology.

[16]  N. Siemers,et al.  Fc-Optimized Anti-CCR8 Antibody Depletes Regulatory T Cells in Human Tumor Models , 2021, Cancer Research.

[17]  F. Tacke,et al.  Therapeutic depletion of CCR8+ tumor-infiltrating regulatory T cells elicits antitumor immunity and synergizes with anti-PD-1 therapy , 2021, Journal for ImmunoTherapy of Cancer.

[18]  Salil S. Bhate,et al.  Immune cell topography predicts response to PD-1 blockade in cutaneous T cell lymphoma , 2020, Nature Communications.

[19]  N. Wald,et al.  Restoration of T-cell Effector Function, Depletion of Tregs, and Direct Killing of Tumor Cells: The Multiple Mechanisms of Action of a-TIGIT Antagonist Antibodies , 2020, Molecular Cancer Therapeutics.

[20]  Sonali M. Smith,et al.  A Phase I Study of Anti-ICOS Antibody MEDI-570 for Relapsed/Refractory (R/R) Peripheral T-Cell Lymphoma (PTCL) and Angioimmunoblastic T-Cell Lymphoma (AITL) (NCI-9930) , 2020 .

[21]  J. Scarisbrick,et al.  Phenotypical Markers, Molecular Mutations, and Immune Microenvironment as Targets for New Treatments in Patients with Mycosis Fungoides and/or Sézary Syndrome. , 2020, The Journal of investigative dermatology.

[22]  P. Gaulard,et al.  ICOS is widely expressed in cutaneous T-cell lymphoma, and its targeting promotes potent killing of malignant cells. , 2020, Blood advances.

[23]  A. Semesi,et al.  Structural characterization of the ICOS/ICOS-L immune complex reveals high molecular mimicry by therapeutic antibodies , 2020, Nature Communications.

[24]  T. Krejsgaard,et al.  Cellular Interactions and Inflammation in the Pathogenesis of Cutaneous T-Cell Lymphoma , 2020, Frontiers in Cell and Developmental Biology.

[25]  Ellen J. Kim,et al.  Cutaneous T-cell lymphoma and concomitant atopic dermatitis responding to dupilumab. , 2020, Cutis.

[26]  M. Bagot,et al.  Dupilumab Treatment in Two Patients with Cutaneous T-cell Lymphomas , 2020, Acta dermato-venereologica.

[27]  Hyun Je Kim,et al.  Single-cell transcriptome analysis of human skin identifies novel fibroblast subpopulation and enrichment of immune subsets in atopic dermatitis. , 2020, The Journal of allergy and clinical immunology.

[28]  C. Solinas,et al.  The rationale behind targeting the ICOS-ICOS ligand costimulatory pathway in cancer immunotherapy , 2020, ESMO Open.

[29]  H. Kohrt,et al.  Pembrolizumab in Relapsed and Refractory Mycosis Fungoides and Sézary Syndrome: A Multicenter Phase II Study. , 2020, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[30]  B. Seliger Basis of PD1/PD-L1 Therapies , 2019, Journal of clinical medicine.

[31]  Xiwei Wu,et al.  Phase 1 Results of Anti-PD-Ligand 1 (Durvalumab) & Lenalidomide in Patients with Cutaneous T Cell Lymphoma and Correlation with Programmed Death Ligand 1 Expression and Gene Expression Profile , 2019, Blood.

[32]  S. Ogino,et al.  Hyperprogressive Disease during Anti-PD-1 (PDCD1) / PD-L1 (CD274) Therapy: A Systematic Review and Meta-Analysis , 2019, Cancers.

[33]  M. Sugaya,et al.  Expression of CCR3 and CCR4 Suggests a Poor Prognosis in Mycosis Fungoides and Sézary Syndrome. , 2019, Acta dermato-venereologica.

[34]  J. Conway,et al.  Single-Cell Lymphocyte Heterogeneity in Advanced Cutaneous T-cell Lymphoma Skin Tumors , 2019, Clinical Cancer Research.

[35]  Z. Zeng,et al.  Role of the tumor microenvironment in PD-L1/PD-1-mediated tumor immune escape , 2019, Molecular Cancer.

[36]  L. French,et al.  Divergent LAG-3 versus BTLA, TIGIT, and FCRL3 expression in Sézary syndrome , 2019, Leukemia & lymphoma.

[37]  R. Clark,et al.  Benign T cells drive clinical skin inflammation in cutaneous T cell lymphoma. , 2019, JCI insight.

[38]  C. Querfeld,et al.  Targeting CD47 as a cancer therapeutic strategy: the cutaneous T-cell lymphoma experience , 2018, Current opinion in oncology.

[39]  C. Elmets,et al.  Mogamulizumab versus vorinostat in previously treated cutaneous T-cell lymphoma (MAVORIC): an international, open-label, randomised, controlled phase 3 trial. , 2018, The Lancet. Oncology.

[40]  R. Sun,et al.  Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity , 2018, Nature Immunology.

[41]  A. Halpern,et al.  Primary T Cells from Cutaneous T-cell Lymphoma Skin Explants Display an Exhausted Immune Checkpoint Profile , 2018, Cancer Immunology Research.

[42]  K. Fujii New Therapies and Immunological Findings in Cutaneous T-Cell Lymphoma , 2018, Front. Oncol..

[43]  Y. Asano,et al.  Interleukin‐25 is involved in cutaneous T‐cell lymphoma progression by establishing a T helper 2‐dominant microenvironment , 2018, The British journal of dermatology.

[44]  R. Clark,et al.  High-throughput sequencing of the T cell receptor β gene identifies aggressive early-stage mycosis fungoides , 2018, Science Translational Medicine.

[45]  Simon C Watkins,et al.  Therapeutic reduction of cell-mediated immunosuppression in mycosis fungoides and Sézary syndrome , 2018, Cancer Immunology, Immunotherapy.

[46]  R. Salgia,et al.  Development of PD-1 and PD-L1 inhibitors as a form of cancer immunotherapy: a comprehensive review of registration trials and future considerations , 2018, Journal of Immunotherapy for Cancer.

[47]  Y. Asano,et al.  Serum vascular endothelial growth factor A levels reflect itch severity in mycosis fungoides and Sézary syndrome , 2018, The Journal of dermatology.

[48]  A. Toubert,et al.  Circulating and skin-derived Sézary cells: clonal but with phenotypic plasticity. , 2017, Blood.

[49]  Miao Xu,et al.  IL‐25 in allergic inflammation , 2017, Immunological reviews.

[50]  W. Dougall,et al.  TIGIT and CD96: new checkpoint receptor targets for cancer immunotherapy , 2017, Immunological reviews.

[51]  J. Szepietowski,et al.  Podoplanin Expression Correlates with Disease Progression in Mycosis Fungoides. , 2017, Acta dermato-venereologica.

[52]  F. Trautinger,et al.  Mast Cells Are Abundant in Primary Cutaneous T-Cell Lymphomas: Results from a Computer-Aided Quantitative Immunohistological Study , 2016, PloS one.

[53]  Shinichi Sato,et al.  Thymic Stromal Chemokine TSLP Acts through Th2 Cytokine Production to Induce Cutaneous T-cell Lymphoma. , 2016, Cancer research.

[54]  N. Mongan,et al.  Malignant inflammation in cutaneous T‐cell lymphoma—a hostile takeover , 2016, Seminars in Immunopathology.

[55]  M. Millenson,et al.  Nivolumab in Patients With Relapsed or Refractory Hematologic Malignancy: Preliminary Results of a Phase Ib Study. , 2016, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[56]  J. Szepietowski,et al.  Expression of CD31 in Mycosis Fungoides. , 2016, Anticancer research.

[57]  U. Wehkamp,et al.  Sézary Syndrome and Atopic Dermatitis: Comparison of Immunological Aspects and Targets , 2016, BioMed research international.

[58]  Jedd D. Wolchok,et al.  PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations , 2016, Science Translational Medicine.

[59]  C. Assaf,et al.  Mac attack: macrophages as key drivers of cutaneous T‐cell lymphoma pathogenesis , 2016, Experimental dermatology.

[60]  S. Aiba,et al.  The possible interaction between periostin expressed by cancer stroma and tumor‐associated macrophages in developing mycosis fungoides , 2016, Experimental dermatology.

[61]  C. Querfeld,et al.  Tumor microenvironment in mycosis fungoides and Sézary syndrome , 2016, Current opinion in oncology.

[62]  R. Johnston,et al.  The checkpoint inhibitor TIGIT limits antitumor and antiviral CD8+ T cell responses , 2015, Oncoimmunology.

[63]  C. M. Eischen,et al.  Genetic markers associated with progression in early mycosis fungoides , 2014, Journal of the European Academy of Dermatology and Venereology : JEADV.

[64]  Ji-Hye Park,et al.  Expression of Follicular Helper T-cell Markers in Primary Cutaneous T-cell Lymphoma , 2014, The American Journal of dermatopathology.

[65]  P. Greenberg,et al.  Tolerance and exhaustion: defining mechanisms of T cell dysfunction. , 2014, Trends in immunology.

[66]  R. Clark,et al.  TH2 Cytokines from Malignant Cells Suppress TH1 Responses and Enforce a Global TH2 Bias in Leukemic Cutaneous T-cell Lymphoma , 2013, Clinical Cancer Research.

[67]  J. Becker,et al.  Vascular endothelial growth factor receptor-3 expression in mycosis fungoides , 2013, Leukemia & lymphoma.

[68]  R. Willemze,et al.  Differential expression of programmed death-1 (PD-1) in Sézary syndrome and mycosis fungoides. , 2012, Archives of dermatology.

[69]  Y. Tada,et al.  Association of the numbers of CD163(+) cells in lesional skin and serum levels of soluble CD163 with disease progression of cutaneous T cell lymphoma. , 2012, Journal of dermatological science.

[70]  R. Büttner,et al.  Mast cells play a protumorigenic role in primary cutaneous lymphoma. , 2012, Blood.

[71]  M. Wasik,et al.  Expression patterns of the immunosuppressive proteins PD-1/CD279 and PD-L1/CD274 at different stages of cutaneous T-cell lymphoma/mycosis fungoides. , 2012, The American Journal of dermatopathology.

[72]  R. Clark,et al.  Skin Effector Memory T Cells Do Not Recirculate and Provide Immune Protection in Alemtuzumab-Treated CTCL Patients , 2012, Science Translational Medicine.

[73]  Y. Tada,et al.  IL-22, but Not IL-17, Dominant Environment in Cutaneous T-cell Lymphoma , 2011, Clinical Cancer Research.

[74]  Katherine G. Evans,et al.  Increased programmed death-1 expression on CD4+ T cells in cutaneous T-cell lymphoma: implications for immune suppression. , 2010, Archives of dermatology.

[75]  H. Rasheed,et al.  Immunohistochemical Study of the Expression of Matrix Metalloproteinase-9 in Skin Lesions of Mycosis Fungoides , 2010, The American Journal of dermatopathology.

[76]  James J. Campbell,et al.  Cutting Edge: Chemokine Receptor CCR4 Is Necessary for Antigen-Driven Cutaneous Accumulation of CD4 T Cells under Physiological Conditions1 , 2007, The Journal of Immunology.

[77]  Juan F. García,et al.  Mycosis fungoides shows concurrent deregulation of multiple genes involved in the TNF signaling pathway: an expression profile study. , 2003, Blood.

[78]  M. G. Lewis,et al.  Target Role of Langerhans Cells in Mycosis Fungoides: Transmission and Immuno‐Electron Microscopic Studies , 1979, Journal of cutaneous pathology.