Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 15.785
Filter
Add more filters

Publication year range
1.
Cell ; 185(12): 2071-2085.e12, 2022 06 09.
Article in English | MEDLINE | ID: mdl-35561684

ABSTRACT

Giant congenital melanocytic nevi are NRAS-driven proliferations that may cover up to 80% of the body surface. Their most dangerous consequence is progression to melanoma. This risk often triggers preemptive extensive surgical excisions in childhood, producing severe lifelong challenges. We have presented preclinical models, including multiple genetically engineered mice and xenografted human lesions, which enabled testing locally applied pharmacologic agents to avoid surgery. The murine models permitted the identification of proliferative versus senescent nevus phases and treatments targeting both. These nevi recapitulated the histologic and molecular features of human giant congenital nevi, including the risk of melanoma transformation. Cutaneously delivered MEK, PI3K, and c-KIT inhibitors or proinflammatory squaric acid dibutylester (SADBE) achieved major regressions. SADBE triggered innate immunity that ablated detectable nevocytes, fully prevented melanoma, and regressed human giant nevus xenografts. These findings reveal nevus mechanistic vulnerabilities and suggest opportunities for topical interventions that may alter the therapeutic options for children with congenital giant nevi.


Subject(s)
Melanoma , Nevus, Pigmented , Skin Neoplasms , Animals , Heterografts , Humans , Melanoma/drug therapy , Melanoma/pathology , Mice , Neoplasm Transplantation , Nevus, Pigmented/congenital , Nevus, Pigmented/drug therapy , Nevus, Pigmented/pathology , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Skin Neoplasms/prevention & control
2.
Nat Immunol ; 22(7): 851-864, 2021 07.
Article in English | MEDLINE | ID: mdl-34099918

ABSTRACT

Group 2 innate lymphoid cells (ILC2s) are essential to maintain tissue homeostasis. In cancer, ILC2s can harbor both pro-tumorigenic and anti-tumorigenic functions, but we know little about their underlying mechanisms or whether they could be clinically relevant or targeted to improve patient outcomes. Here, we found that high ILC2 infiltration in human melanoma was associated with a good clinical prognosis. ILC2s are critical producers of the cytokine granulocyte-macrophage colony-stimulating factor, which coordinates the recruitment and activation of eosinophils to enhance antitumor responses. Tumor-infiltrating ILC2s expressed programmed cell death protein-1, which limited their intratumoral accumulation, proliferation and antitumor effector functions. This inhibition could be overcome in vivo by combining interleukin-33-driven ILC2 activation with programmed cell death protein-1 blockade to significantly increase antitumor responses. Together, our results identified ILC2s as a critical immune cell type involved in melanoma immunity and revealed a potential synergistic approach to harness ILC2 function for antitumor immunotherapies.


Subject(s)
Antibodies/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Interleukin-33/pharmacology , Lymphocytes/drug effects , Melanoma, Experimental/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Neoplasms/drug therapy , Animals , Cell Line, Tumor , Chemotaxis, Leukocyte/drug effects , Cytotoxicity, Immunologic/drug effects , Eosinophils/drug effects , Eosinophils/immunology , Eosinophils/metabolism , Female , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Melanoma, Experimental/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/immunology , Skin Neoplasms/metabolism
3.
Cell ; 173(6): 1413-1425.e14, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29754815

ABSTRACT

BRAF(V600E) mutant melanomas treated with inhibitors of the BRAF and MEK kinases almost invariably develop resistance that is frequently caused by reactivation of the mitogen activated protein kinase (MAPK) pathway. To identify novel treatment options for such patients, we searched for acquired vulnerabilities of MAPK inhibitor-resistant melanomas. We find that resistance to BRAF+MEK inhibitors is associated with increased levels of reactive oxygen species (ROS). Subsequent treatment with the histone deacetylase inhibitor vorinostat suppresses SLC7A11, leading to a lethal increase in the already-elevated levels of ROS in drug-resistant cells. This causes selective apoptotic death of only the drug-resistant tumor cells. Consistently, treatment of BRAF inhibitor-resistant melanoma with vorinostat in mice results in dramatic tumor regression. In a study in patients with advanced BRAF+MEK inhibitor-resistant melanoma, we find that vorinostat can selectively ablate drug-resistant tumor cells, providing clinical proof of concept for the novel therapy identified here.


Subject(s)
Drug Resistance, Neoplasm , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Amino Acid Transport System y+/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System , Melanoma/genetics , Mice , Mutation , Neoplasm Transplantation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Reactive Oxygen Species/metabolism , Skin Neoplasms/genetics , Treatment Outcome , Vorinostat/pharmacology
4.
Nat Immunol ; 20(2): 206-217, 2019 02.
Article in English | MEDLINE | ID: mdl-30664764

ABSTRACT

Immune checkpoint blockade therapy has shifted the paradigm for cancer treatment. However, the majority of patients lack effective responses due to insufficient T cell infiltration in tumors. Here we show that expression of mitochondrial uncoupling protein 2 (UCP2) in tumor cells determines the immunostimulatory feature of the tumor microenvironment (TME) and is positively associated with prolonged survival. UCP2 reprograms the immune state of the TME by altering its cytokine milieu in an interferon regulatory factor 5-dependent manner. Consequently, UCP2 boosts the conventional type 1 dendritic cell- and CD8+ T cell-dependent anti-tumor immune cycle and normalizes the tumor vasculature. Finally we show, using either a genetic or pharmacological approach, that induction of UCP2 sensitizes melanomas to programmed cell death protein-1 blockade treatment and elicits effective anti-tumor responses. Together, this study demonstrates that targeting the UCP2 pathway is a potent strategy for alleviating the immunosuppressive TME and overcoming the primary resistance of programmed cell death protein-1 blockade.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Melanoma, Experimental/immunology , Skin Neoplasms/immunology , Tumor Microenvironment/immunology , Uncoupling Protein 2/immunology , Animals , Antineoplastic Agents, Immunological/pharmacology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Drug Resistance, Neoplasm/immunology , Female , Humans , Immunotherapy/methods , Interferon Regulatory Factors/immunology , Interferon Regulatory Factors/metabolism , Melanoma, Experimental/blood supply , Melanoma, Experimental/drug therapy , Melanoma, Experimental/mortality , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Skin Neoplasms/blood supply , Skin Neoplasms/drug therapy , Skin Neoplasms/mortality , Survival Analysis , Treatment Outcome , Uncoupling Protein 2/genetics , Uncoupling Protein 2/metabolism
5.
Cell ; 164(5): 831, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26919418

ABSTRACT

The hedgehog (Hh) signaling pathway is aberrantly activated in a majority of basal cell carcinomas (BCC). Vismodegib and sonidegib are targeted inhibitors of Smoothened (SMO). Both drugs are approved for use in locally advanced BCC (laBCC), with vismodegib also approved for metastatic BCC (mBCC).


Subject(s)
Anilides/therapeutic use , Antineoplastic Agents/therapeutic use , Biphenyl Compounds/therapeutic use , Carcinoma, Basal Cell/drug therapy , Hedgehog Proteins/metabolism , Pyridines/therapeutic use , Signal Transduction/drug effects , Skin Neoplasms/drug therapy , Humans , Receptors, G-Protein-Coupled/antagonists & inhibitors , Smoothened Receptor , Translational Research, Biomedical
6.
Cell ; 167(2): 397-404.e9, 2016 Oct 06.
Article in English | MEDLINE | ID: mdl-27667683

ABSTRACT

Antibody blockade of the inhibitory CTLA-4 pathway has led to clinical benefit in a subset of patients with metastatic melanoma. Anti-CTLA-4 enhances T cell responses, including production of IFN-γ, which is a critical cytokine for host immune responses. However, the role of IFN-γ signaling in tumor cells in the setting of anti-CTLA-4 therapy remains unknown. Here, we demonstrate that patients identified as non-responders to anti-CTLA-4 (ipilimumab) have tumors with genomic defects in IFN-γ pathway genes. Furthermore, mice bearing melanoma tumors with knockdown of IFN-γ receptor 1 (IFNGR1) have impaired tumor rejection upon anti-CTLA-4 therapy. These data highlight that loss of the IFN-γ signaling pathway is associated with primary resistance to anti-CTLA-4 therapy. Our findings demonstrate the importance of tumor genomic data, especially IFN-γ related genes, as prognostic information for patients selected to receive treatment with immune checkpoint therapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CTLA-4 Antigen/antagonists & inhibitors , Drug Resistance, Neoplasm/genetics , Interferon-gamma/genetics , Melanoma/drug therapy , Receptors, Interferon/genetics , Skin Neoplasms/drug therapy , Animals , Cell Line, Tumor , Cytokines/immunology , Gene Knockdown Techniques , Humans , Ipilimumab , Melanoma/genetics , Melanoma, Experimental/drug therapy , Melanoma, Experimental/genetics , Mice , Mice, Inbred C57BL , Skin Neoplasms/genetics , T-Lymphocytes/immunology , Interferon gamma Receptor
7.
Cell ; 162(5): 937, 2015 Aug 27.
Article in English | MEDLINE | ID: mdl-26317459

ABSTRACT

Nivolumab and pembrolizumab are monoclonal antibodies that block the programmed death-1 receptor (PD-1, CD279), resulting in dis-inhibition of tumor-specific immune responses. Both are recently approved for use in the treatment of metastatic melanoma, and nivolumab as well for non-small cell lung cancer.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Melanoma/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Neoplasms/drug therapy , Clinical Trials as Topic , Humans , Nivolumab
8.
Cell ; 160(5): 963-976, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25723170

ABSTRACT

Subsets of long-lived, tumor-initiating stem cells often escape cancer therapies. However, sources and mechanisms that generate tumor heterogeneity and drug-resistant cell population are still unfolding. Here, we devise a functional reporter system to lineage trace and/or genetic ablate signaling in TGF-ß-activated squamous cell carcinoma stem cells (SCC-SCs). Dissecting TGF-ß's impact on malignant progression, we demonstrate that TGF-ß concentrating near tumor-vasculature generates heterogeneity in TGF-ß signaling at tumor-stroma interface and bestows slower-cycling properties to neighboring SCC-SCs. While non-responding progenies proliferate faster and accelerate tumor growth, TGF-ß-responding progenies invade, aberrantly differentiate, and affect gene expression. Intriguingly, TGF-ß-responding SCC-SCs show increased protection against anti-cancer drugs, but slower-cycling alone does not confer survival. Rather, TGF-ß transcriptionally activates p21, which stabilizes NRF2, thereby markedly enhancing glutathione metabolism and diminishing effectiveness of anti-cancer therapeutics. Together, these findings establish a surprising non-genetic paradigm for TGF-ß signaling in fueling heterogeneity in SCC-SCs, tumor characteristics, and drug resistance.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Drug Resistance, Neoplasm , Signal Transduction , Skin Neoplasms/metabolism , Transforming Growth Factor beta/metabolism , 9,10-Dimethyl-1,2-benzanthracene , Animals , Carcinoma, Squamous Cell/drug therapy , Cisplatin/therapeutic use , Female , Gene Expression Profiling , Glutathione/metabolism , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Heterografts , Humans , Mice , Mice, Nude , NF-E2-Related Factor 2 , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Skin Neoplasms/drug therapy , Tetradecanoylphorbol Acetate
9.
Cell ; 156(5): 986-1001, 2014 Feb 27.
Article in English | MEDLINE | ID: mdl-24581497

ABSTRACT

Melanoma metastasis is a devastating outcome lacking an effective preventative therapeutic. We provide pharmacologic, molecular, and genetic evidence establishing the liver-X nuclear hormone receptor (LXR) as a therapeutic target in melanoma. Oral administration of multiple LXR agonists suppressed melanoma invasion, angiogenesis, tumor progression, and metastasis. Molecular and genetic experiments revealed these effects to be mediated by LXRß, which elicits these outcomes through transcriptional induction of tumoral and stromal apolipoprotein-E (ApoE). LXRß agonism robustly suppressed tumor growth and metastasis across a diverse mutational spectrum of melanoma lines. LXRß targeting significantly prolonged animal survival, suppressed the progression of established metastases, and inhibited brain metastatic colonization. Importantly, LXRß activation displayed melanoma-suppressive cooperativity with the frontline regimens dacarbazine, B-Raf inhibition, and the anti-CTLA-4 antibody and robustly inhibited melanomas that had acquired resistance to B-Raf inhibition or dacarbazine. We present a promising therapeutic approach that uniquely acts by transcriptionally activating a metastasis suppressor gene.


Subject(s)
Melanoma/drug therapy , Melanoma/secondary , Neoplasm Metastasis/drug therapy , Orphan Nuclear Receptors/agonists , Skin Neoplasms/drug therapy , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Benzoates/administration & dosage , Benzylamines/administration & dosage , Cells, Cultured , Disease Models, Animal , Humans , Hydrocarbons, Fluorinated/administration & dosage , Liver X Receptors , Melanoma/pathology , Mice , Neoplasm Metastasis/pathology , Signal Transduction , Skin Neoplasms/pathology , Sulfonamides/administration & dosage , Transcription, Genetic
10.
Nature ; 616(7955): 168-175, 2023 04.
Article in English | MEDLINE | ID: mdl-36949199

ABSTRACT

The resistance of cancer cells to therapy is responsible for the death of most patients with cancer1. Epithelial-to-mesenchymal transition (EMT) has been associated with resistance to therapy in different cancer cells2,3. However, the mechanisms by which EMT mediates resistance to therapy remain poorly understood. Here, using a mouse model of skin squamous cell carcinoma undergoing spontaneous EMT during tumorigenesis, we found that EMT tumour cells are highly resistant to a wide range of anti-cancer therapies both in vivo and in vitro. Using gain and loss of function studies in vitro and in vivo, we found that RHOJ-a small GTPase that is preferentially expressed in EMT cancer cells-controls resistance to therapy. Using genome-wide transcriptomic and proteomic profiling, we found that RHOJ regulates EMT-associated resistance to chemotherapy by enhancing the response to replicative stress and activating the DNA-damage response, enabling tumour cells to rapidly repair DNA lesions induced by chemotherapy. RHOJ interacts with proteins that regulate nuclear actin, and inhibition of actin polymerization sensitizes EMT tumour cells to chemotherapy-induced cell death in a RHOJ-dependent manner. Together, our study uncovers the role and the mechanisms through which RHOJ acts as a key regulator of EMT-associated resistance to chemotherapy.


Subject(s)
Carcinoma, Squamous Cell , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Skin Neoplasms , rho GTP-Binding Proteins , Actins/drug effects , Actins/metabolism , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Proteomics , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , Animals , Mice , Skin Neoplasms/drug therapy , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Gene Expression Profiling , Genome
11.
Nature ; 620(7973): 402-408, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37532929

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) regulates tumour initiation, progression, metastasis and resistance to anti-cancer therapy1-7. Although great progress has been made in understanding the role of EMT and its regulatory mechanisms in cancer, no therapeutic strategy to pharmacologically target EMT has been identified. Here we found that netrin-1 is upregulated in a primary mouse model of skin squamous cell carcinoma (SCC) exhibiting spontaneous EMT. Pharmacological inhibition of netrin-1 by administration of NP137, a netrin-1-blocking monoclonal antibody currently used in clinical trials in human cancer (ClinicalTrials.gov identifier NCT02977195 ), decreased the proportion of EMT tumour cells in skin SCC, decreased the number of metastases and increased the sensitivity of tumour cells to chemotherapy. Single-cell RNA sequencing revealed the presence of different EMT states, including epithelial, early and late hybrid EMT, and full EMT states, in control SCC. By contrast, administration of NP137 prevented the progression of cancer cells towards a late EMT state and sustained tumour epithelial states. Short hairpin RNA knockdown of netrin-1 and its receptor UNC5B in EPCAM+ tumour cells inhibited EMT in vitro in the absence of stromal cells and regulated a common gene signature that promotes tumour epithelial state and restricts EMT. To assess the relevance of these findings to human cancers, we treated mice transplanted with the A549 human cancer cell line-which undergoes EMT following TGFß1 administration8,9-with NP137. Netrin-1 inhibition decreased EMT in these transplanted A549 cells. Together, our results identify a pharmacological strategy for targeting EMT in cancer, opening up novel therapeutic interventions for anti-cancer therapy.


Subject(s)
Antibodies, Monoclonal , Carcinoma, Squamous Cell , Epithelial-Mesenchymal Transition , Netrin-1 , Skin Neoplasms , Animals , Humans , Mice , A549 Cells , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Netrin Receptors/antagonists & inhibitors , Netrin Receptors/deficiency , Netrin Receptors/genetics , Netrin-1/antagonists & inhibitors , Netrin-1/deficiency , Netrin-1/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Disease Models, Animal , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Neoplasm Metastasis/drug therapy , Single-Cell Gene Expression Analysis , RNA-Seq , Epithelial Cell Adhesion Molecule/metabolism , Xenograft Model Antitumor Assays , Transforming Growth Factor beta1/pharmacology
12.
Nature ; 606(7915): 797-803, 2022 06.
Article in English | MEDLINE | ID: mdl-35705814

ABSTRACT

Treatment with therapy targeting BRAF and MEK (BRAF/MEK) has revolutionized care in melanoma and other cancers; however, therapeutic resistance is common and innovative treatment strategies are needed1,2. Here we studied a group of patients with melanoma who were treated with neoadjuvant BRAF/MEK-targeted therapy ( NCT02231775 , n = 51) and observed significantly higher rates of major pathological response (MPR; ≤10% viable tumour at resection) and improved recurrence-free survival (RFS) in female versus male patients (MPR, 66% versus 14%, P = 0.001; RFS, 64% versus 32% at 2 years, P = 0.021). The findings were validated in several additional cohorts2-4 of patients with unresectable metastatic melanoma who were treated with BRAF- and/or MEK-targeted therapy (n = 664 patients in total), demonstrating improved progression-free survival and overall survival in female versus male patients in several of these studies. Studies in preclinical models demonstrated significantly impaired anti-tumour activity in male versus female mice after BRAF/MEK-targeted therapy (P = 0.006), with significantly higher expression of the androgen receptor in tumours of male and female BRAF/MEK-treated mice versus the control (P = 0.0006 and P = 0.0025). Pharmacological inhibition of androgen receptor signalling improved responses to BRAF/MEK-targeted therapy in male and female mice (P = 0.018 and P = 0.003), whereas induction of androgen receptor signalling (through testosterone administration) was associated with a significantly impaired response to BRAF/MEK-targeted therapy in male and female patients (P = 0.021 and P < 0.0001). Together, these results have important implications for therapy.


Subject(s)
Androgen Receptor Antagonists , Melanoma , Mitogen-Activated Protein Kinase Kinases , Molecular Targeted Therapy , Proto-Oncogene Proteins B-raf , Receptors, Androgen , Animals , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Female , Humans , Male , Melanoma/drug therapy , Melanoma/pathology , Mice , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Receptors, Androgen/metabolism , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Survival Analysis
13.
Proc Natl Acad Sci U S A ; 121(14): e2316303121, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38551838

ABSTRACT

Photodynamic therapy (PDT) relies on a series of photophysical and photochemical reactions leading to cell death. While effective for various cancers, PDT has been less successful in treating pigmented melanoma due to high light absorption by melanin. Here, this limitation is addressed by 2-photon excitation of the photosensitizer (2p-PDT) using ~100 fs pulses of near-infrared laser light. A critical role of melanin in enabling rather than hindering 2p-PDT is elucidated using pigmented and non-pigmented murine melanoma clonal cell lines in vitro. The photocytotoxicities were compared between a clinical photosensitizer (Visudyne) and a porphyrin dimer (Oxdime) with ~600-fold higher σ2p value. Unexpectedly, while the 1p-PDT responses are similar in both cell lines, 2p activation is much more effective in killing pigmented than non-pigmented cells, suggesting a dominant role of melanin 2p-PDT. The potential for clinical translational is demonstrated in a conjunctival melanoma model in vivo, where complete eradication of small tumors was achieved. This work elucidates the melanin contribution in multi-photon PDT enabling significant advancement of light-based treatments that have previously been considered unsuitable in pigmented tumors.


Subject(s)
Melanoma , Photochemotherapy , Skin Neoplasms , Mice , Humans , Animals , Photosensitizing Agents/pharmacology , Melanoma/drug therapy , Melanoma/pathology , Melanins/metabolism , Skin Neoplasms/drug therapy
14.
N Engl J Med ; 388(9): 813-823, 2023 Mar 02.
Article in English | MEDLINE | ID: mdl-36856617

ABSTRACT

BACKGROUND: Whether pembrolizumab given both before surgery (neoadjuvant therapy) and after surgery (adjuvant therapy), as compared with pembrolizumab given as adjuvant therapy alone, would increase event-free survival among patients with resectable stage III or IV melanoma is unknown. METHODS: In a phase 2 trial, we randomly assigned patients with clinically detectable, measurable stage IIIB to IVC melanoma that was amenable to surgical resection to three doses of neoadjuvant pembrolizumab, surgery, and 15 doses of adjuvant pembrolizumab (neoadjuvant-adjuvant group) or to surgery followed by pembrolizumab (200 mg intravenously every 3 weeks for a total of 18 doses) for approximately 1 year or until disease recurred or unacceptable toxic effects developed (adjuvant-only group). The primary end point was event-free survival in the intention-to-treat population. Events were defined as disease progression or toxic effects that precluded surgery; the inability to resect all gross disease; disease progression, surgical complications, or toxic effects of treatment that precluded the initiation of adjuvant therapy within 84 days after surgery; recurrence of melanoma after surgery; or death from any cause. Safety was also evaluated. RESULTS: At a median follow-up of 14.7 months, the neoadjuvant-adjuvant group (154 patients) had significantly longer event-free survival than the adjuvant-only group (159 patients) (P = 0.004 by the log-rank test). In a landmark analysis, event-free survival at 2 years was 72% (95% confidence interval [CI], 64 to 80) in the neoadjuvant-adjuvant group and 49% (95% CI, 41 to 59) in the adjuvant-only group. The percentage of patients with treatment-related adverse events of grades 3 or higher during therapy was 12% in the neoadjuvant-adjuvant group and 14% in the adjuvant-only group. CONCLUSIONS: Among patients with resectable stage III or IV melanoma, event-free survival was significantly longer among those who received pembrolizumab both before and after surgery than among those who received adjuvant pembrolizumab alone. No new toxic effects were identified. (Funded by the National Cancer Institute and Merck Sharp and Dohme; S1801 ClinicalTrials.gov number, NCT03698019.).


Subject(s)
Antineoplastic Agents, Immunological , Melanoma , Neoadjuvant Therapy , Skin Neoplasms , Humans , Adjuvants, Immunologic , Disease Progression , Melanoma/drug therapy , Melanoma/pathology , Melanoma/surgery , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Skin Neoplasms/surgery , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/therapeutic use , Chemotherapy, Adjuvant
15.
Blood ; 143(2): 178-182, 2024 Jan 11.
Article in English | MEDLINE | ID: mdl-37963262

ABSTRACT

ABSTRACT: Nonmelanoma skin cancers (NMSCs) in ruxolitinib-treated patients with myeloproliferative neoplasms behave aggressively, with adverse features and high recurrence. In our cohort, mortality from metastatic NMSC exceeded that from myelofibrosis. Vigilant skin assessment, counseling on NMSC risks, and prospective ruxolitinib-NMSC studies are crucial.


Subject(s)
Myeloproliferative Disorders , Pyrazoles , Pyrimidines , Skin Neoplasms , Humans , Prospective Studies , Myeloproliferative Disorders/drug therapy , Nitriles , Skin Neoplasms/drug therapy
16.
Mol Cell ; 69(6): 923-937.e8, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29547721

ABSTRACT

Dietary supplements such as vitamins and minerals are widely used in the hope of improving health but may have unidentified risks and side effects. In particular, a pathogenic link between dietary supplements and specific oncogenes remains unknown. Here we report that chondroitin-4-sulfate (CHSA), a natural glycosaminoglycan approved as a dietary supplement used for osteoarthritis, selectively promotes the tumor growth potential of BRAF V600E-expressing human melanoma cells in patient- and cell line-derived xenograft mice and confers resistance to BRAF inhibitors. Mechanistically, chondroitin sulfate glucuronyltransferase (CSGlcA-T) signals through its product CHSA to enhance casein kinase 2 (CK2)-PTEN binding and consequent phosphorylation and inhibition of PTEN, which requires CHSA chains and is essential to sustain AKT activation in BRAF V600E-expressing melanoma cells. However, this CHSA-dependent PTEN inhibition is dispensable in cancer cells expressing mutant NRAS or PI3KCA, which directly activate the PI3K-AKT pathway. These results suggest that dietary supplements may exhibit oncogene-dependent pro-tumor effects.


Subject(s)
Carcinogens/toxicity , Cell Transformation, Neoplastic/genetics , Chondroitin Sulfates/toxicity , Dietary Supplements/toxicity , Melanoma/chemically induced , Mutation , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/chemically induced , Animals , Antinematodal Agents/pharmacology , Casein Kinase II/metabolism , Cell Proliferation/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , GTP Phosphohydrolases/genetics , HEK293 Cells , HT29 Cells , Humans , Melanoma/drug therapy , Melanoma/enzymology , Melanoma/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred NOD , Mice, Nude , Mice, Transgenic , NIH 3T3 Cells , Nuclear Proteins/genetics , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/metabolism , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Skin Neoplasms/drug therapy , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Transcription Factors/genetics , Xenograft Model Antitumor Assays
17.
Lancet ; 403(10427): 632-644, 2024 02 17.
Article in English | MEDLINE | ID: mdl-38246194

ABSTRACT

BACKGROUND: Checkpoint inhibitors are standard adjuvant treatment for stage IIB-IV resected melanoma, but many patients recur. Our study aimed to evaluate whether mRNA-4157 (V940), a novel mRNA-based individualised neoantigen therapy, combined with pembrolizumab, improved recurrence-free survival and distant metastasis-free survival versus pembrolizumab monotherapy in resected high-risk melanoma. METHODS: We did an open-label, randomised, phase 2b, adjuvant study of mRNA-4157 plus pembrolizumab versus pembrolizumab monotherapy in patients, enrolled from sites in the USA and Australia, with completely resected high-risk cutaneous melanoma. Patients with completely resected melanoma (stage IIIB-IV) were assigned 2:1 to receive open-label mRNA-4157 plus pembrolizumab or pembrolizumab monotherapy. mRNA-4157 was administered intramuscularly (maximum nine doses) and pembrolizumab intravenously (maximum 18 doses) in 3-week cycles. The primary endpoint was recurrence-free survival in the intention-to-treat population. This ongoing trial is registered at ClinicalTrials.gov, NCT03897881. FINDINGS: From July 18, 2019, to Sept 30, 2021, 157 patients were assigned to mRNA-4157 plus pembrolizumab combination therapy (n=107) or pembrolizumab monotherapy (n=50); median follow-up was 23 months and 24 months, respectively. Recurrence-free survival was longer with combination versus monotherapy (hazard ratio [HR] for recurrence or death, 0·561 [95% CI 0·309-1·017]; two-sided p=0·053), with lower recurrence or death event rate (24 [22%] of 107 vs 20 [40%] of 50); 18-month recurrence-free survival was 79% (95% CI 69·0-85·6) versus 62% (46·9-74·3). Most treatment-related adverse events were grade 1-2. Grade ≥3 treatment-related adverse events occurred in 25% of patients in the combination group and 18% of patients in the monotherapy group, with no mRNA-4157-related grade 4-5 events. Immune-mediated adverse event frequency was similar for the combination (37 [36%]) and monotherapy (18 [36%]) groups. INTERPRETATION: Adjuvant mRNA-4157 plus pembrolizumab prolonged recurrence-free survival versus pembrolizumab monotherapy in patients with resected high-risk melanoma and showed a manageable safety profile. These results provide evidence that an mRNA-based individualised neoantigen therapy might be beneficial in the adjuvant setting. FUNDING: Moderna in collaboration with Merck Sharp & Dohme, a subsidiary of Merck & Co, Rahway, NJ, USA.


Subject(s)
Melanoma , Skin Neoplasms , Humans , Adjuvants, Immunologic/therapeutic use , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Melanoma/drug therapy , Melanoma/genetics , Melanoma/surgery , Skin Neoplasms/drug therapy , Skin Neoplasms/genetics , Skin Neoplasms/surgery
18.
N Engl J Med ; 387(17): 1557-1568, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36094839

ABSTRACT

BACKGROUND: In a pilot study involving patients with cutaneous squamous-cell carcinoma, a high percentage of patients had a pathological complete response with the use of two doses of neoadjuvant cemiplimab before surgery. Data from a phase 2 study are needed to confirm these findings. METHODS: We conducted a phase 2, confirmatory, multicenter, nonrandomized study to evaluate cemiplimab as neoadjuvant therapy in patients with resectable stage II, III, or IV (M0) cutaneous squamous-cell carcinoma. Patients received cemiplimab, administered at a dose of 350 mg every 3 weeks for up to four doses, before undergoing surgery with curative intent. The primary end point was a pathological complete response (the absence of viable tumor cells in the surgical specimen) on independent review at a central laboratory, with a null hypothesis that a pathological complete response would be observed in 25% of patients. Key secondary end points included a pathological major response (the presence of viable tumor cells that constitute ≤10% of the surgical specimen) on independent review, a pathological complete response and a pathological major response on investigator assessment at a local laboratory, an objective response on imaging, and adverse events. RESULTS: A total of 79 patients were enrolled and received neoadjuvant cemiplimab. On independent review, a pathological complete response was observed in 40 patients (51%; 95% confidence interval [CI], 39 to 62) and a pathological major response in 10 patients (13%; 95% CI, 6 to 22). These results were consistent with the pathological responses determined on investigator assessment. An objective response on imaging was observed in 54 patients (68%; 95% CI, 57 to 78). Adverse events of any grade that occurred during the study period, regardless of whether they were attributed to the study treatment, were observed in 69 patients (87%). Grade 3 or higher adverse events that occurred during the study period were observed in 14 patients (18%). CONCLUSIONS: Neoadjuvant therapy with cemiplimab was associated with a pathological complete response in a high percentage of patients with resectable cutaneous squamous-cell carcinoma. (Funded by Regeneron Pharmaceuticals and Sanofi; ClinicalTrials.gov number, NCT04154943.).


Subject(s)
Carcinoma, Squamous Cell , Neoadjuvant Therapy , Skin Neoplasms , Humans , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/surgery , Carcinoma, Squamous Cell/pathology , Neoplasm Staging , Pilot Projects , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Skin Neoplasms/surgery , Remission Induction , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/therapeutic use
19.
Blood ; 141(7): 695-703, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36379025

ABSTRACT

Agents targeting the unique biology of mycosis fungoides and Sézary syndrome are quickly being incorporated into clinical management. With these new therapies, we are now capable of inducing more durable responses and even complete remissions in advanced disease, outcomes which were exceedingly rare with prior therapies. Yet, even this new generation of therapies typically produce objective responses in only a minority of patients. As our therapeutic options increase, we are now challenged with selecting treatments from a growing list of options. To gain the full benefit of these novel agents, we must develop strategies to match treatments for the patients most likely to benefit from them. Here, we consider both the current approaches to treatment selection based on clinical features and the future of molecular biomarker-guided therapy for patients with this heterogeneous disease.


Subject(s)
Mycosis Fungoides , Sezary Syndrome , Skin Neoplasms , Humans , Sezary Syndrome/drug therapy , Skin Neoplasms/drug therapy , Mycosis Fungoides/drug therapy
20.
Blood ; 142(9): 794-805, 2023 08 31.
Article in English | MEDLINE | ID: mdl-37217183

ABSTRACT

Targeted therapies for cutaneous T-cell lymphoma (CTCL) are limited and curative approaches are lacking. Furthermore, relapses and drug induced side effects are major challenges in the therapeutic management of patients with CTCL, creating an urgent need for new and effective therapies. Pathologic constitutive NF-κB activity leads to apoptosis resistance in CTCL cells and, thus, represents a promising therapeutic target in CTCL. In a preclinical study we showed the potential of dimethyl fumarate (DMF) to block NF-κB and, specifically, kill CTCL cells. To translate these findings to applications in a clinical setting, we performed a multicentric phase 2 study evaluating oral DMF therapy in 25 patients with CTCL stages Ib to IV over 24 weeks (EudraCT number 2014-000924-11/NCT number NCT02546440). End points were safety and efficacy. We evaluated skin involvement (using a modified severity weighted assessment tool [mSWAT]), pruritus, quality of life, and blood involvement, if applicable, as well as translational data. Upon skin analysis, 7 of 23 (30.4%) patients showed a response with >50% reduction in the mSWAT score. Patients with high tumor burden in the skin and blood responded best to DMF therapy. Although not generally significant, DMF also improved pruritus in several patients. Response in the blood was mixed, but we confirmed the NF-κB-inhibiting mechanism of DMF in the blood. The overall tolerability of the DMF therapy was very favorable, with mostly mild side effects. In conclusion, our study presents DMF as an effective and excellently tolerable therapeutic option in CTCL to be further evaluated in a phase 3 study or real-life patient care as well as in combination therapies. This trial was registered at www.clinicaltrials.gov as #NCT02546440.


Subject(s)
Lymphoma, T-Cell, Cutaneous , Skin Neoplasms , Humans , Dimethyl Fumarate/therapeutic use , NF-kappa B , Quality of Life , Skin Neoplasms/drug therapy , Skin Neoplasms/pathology , Neoplasm Recurrence, Local/drug therapy , Lymphoma, T-Cell, Cutaneous/drug therapy , Lymphoma, T-Cell, Cutaneous/pathology , Pruritus/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL