Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
Add more filters

Country/Region as subject
Publication year range
1.
EMBO J ; 38(15): e95874, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31267558

ABSTRACT

MAPK inhibitors (MAPKi) show outstanding clinical response rates in melanoma patients harbouring BRAF mutations, but resistance is common. The ability of melanoma cells to switch from melanocytic to mesenchymal phenotypes appears to be associated with therapeutic resistance. High-throughput, subcellular proteome analyses and RNAseq on two panels of primary melanoma cells that were either sensitive or resistant to MAPKi revealed that only 15 proteins were sufficient to distinguish between these phenotypes. The two proteins with the highest discriminatory power were PTRF and IGFBP7, which were both highly upregulated in the mesenchymal-resistant cells. Proteomic analysis of CRISPR/Cas-derived PTRF knockouts revealed targets involved in lysosomal activation, endocytosis, pH regulation, EMT, TGFß signalling and cell migration and adhesion, as well as a significantly reduced invasive index and ability to form spheres in 3D culture. Overexpression of PTRF led to MAPKi resistance, increased cell adhesion and sphere formation. In addition, immunohistochemistry of patient samples showed that PTRF expression levels were a significant biomarker of poor progression-free survival, and IGFBP7 levels in patient sera were shown to be higher after relapse.


Subject(s)
Drug Resistance, Neoplasm , Insulin-Like Growth Factor Binding Proteins/metabolism , Melanoma/metabolism , Protein Kinase Inhibitors/pharmacology , Proteomics/methods , RNA-Binding Proteins/metabolism , Adult , Aged , Carbamates/pharmacology , Cell Adhesion , Cell Line, Tumor , Disease Progression , Epithelial-Mesenchymal Transition , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Insulin-Like Growth Factor Binding Proteins/blood , Male , Melanoma/drug therapy , Melanoma/genetics , Middle Aged , Protein Interaction Maps , Sequence Analysis, RNA , Sulfonamides/pharmacology , Survival Analysis , Up-Regulation , Vemurafenib/pharmacology
2.
J Eur Acad Dermatol Venereol ; 37(5): 922-931, 2023 May.
Article in English | MEDLINE | ID: mdl-36785973

ABSTRACT

BACKGROUND: The COVID-19 lockdown had a dramatic impact on primary care access and resulted in postponed skin cancer screenings. This raises concerns for a diagnostic delay on primary cutaneous melanomas, which can subsequently increase morbidity and mortality. OBJECTIVES: The aim of the study was to investigate the impact of the COVID-19-related restrictions on the melanoma diagnosis in five European skin cancer reference centres in Switzerland, Germany, Austria and Italy. METHODS: A total of 7865 cutaneous melanoma cases were collected between 01 September 2018 and 31 August 2021. The time period was stratified into pre-COVID (pre-lockdown) and post-COVID (lockdown and post-lockdown) according to the established restrictions in each country. The data collection included demographic, clinical and histopathological data from histologically confirmed cutaneous melanomas. Personal and family history of melanoma, and presence of immunosuppression were used to assess the diagnosis delay in high-risk individuals. RESULTS: There was an overall increase of the Breslow tumour thickness (mean 1.25 mm vs. 1.02 mm) during the post-COVID period, as well as an increase in the proportion of T3-T4 melanomas, rates of ulceration and the number of mitotic rates ≥2 (all, p < 0.001). Patients with immunosuppression and personal history of melanoma showed a decrease in the mean log10-transformed Breslow during lockdown and post-COVID. In the multivariate analysis, age at melanoma diagnosis (p < 0.01) and personal history of melanoma (p < 0.01) showed significant differences in the mean Breslow thickness. CONCLUSIONS: The study confirms the diagnostic delay in cutaneous melanomas due to the COVID-19 lockdown. High-risk individuals, such as patients with personal history of melanoma and elderly individuals, were more hesitant to restart their regular skin cancer screenings post-COVID. Further studies with longer follow-up are required to evaluate the consequences of this diagnostic delay in long-term outcomes.


Subject(s)
COVID-19 , Melanoma , Skin Neoplasms , Humans , Aged , Skin Neoplasms/diagnosis , Skin Neoplasms/epidemiology , Skin Neoplasms/pathology , Melanoma/diagnosis , Melanoma/epidemiology , Melanoma/pathology , Retrospective Studies , Delayed Diagnosis , Pandemics , COVID-19/epidemiology , Communicable Disease Control , COVID-19 Testing , Melanoma, Cutaneous Malignant
3.
Nat Chem Biol ; 14(1): 94-101, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29083417

ABSTRACT

Wnt (wingless)/ß-catenin signaling is critical for tumor progression and is frequently activated in colorectal cancer as a result of the mutation of adenomatous polyposis coli (APC); however, therapeutic agents targeting this pathway for clinical use are lacking. Here we report that nitazoxanide (NTZ), a clinically approved antiparasitic drug, efficiently inhibits Wnt signaling independent of APC. Using chemoproteomic approaches, we have identified peptidyl arginine deiminase 2 (PAD2) as the functional target of NTZ in Wnt inhibition. By targeting PAD2, NTZ increased the deamination (citrullination) and turnover of ß-catenin in colon cancer cells. Replacement of arginine residues disrupted the transcriptional activity, and NTZ induced degradation of ß-catenin. In Wnt-activated colon cancer cells, knockout of either PAD2 or ß-catenin substantially increased resistance to NTZ treatment. Our data highlight the potential of NTZ as a modulator of ß-catenin citrullination for the treatment of cancer patients with Wnt pathway mutations.


Subject(s)
Antineoplastic Agents/pharmacology , Colonic Neoplasms/metabolism , Protein-Arginine Deiminases/metabolism , Thiazoles/pharmacology , Wnt Signaling Pathway/drug effects , beta Catenin/metabolism , Animals , Cell Line, Tumor , Citrullination , Colonic Neoplasms/pathology , Gene Knockout Techniques , Humans , Nitro Compounds , Protein-Arginine Deiminase Type 2 , Protein-Arginine Deiminases/genetics , Wnt Signaling Pathway/genetics , beta Catenin/genetics
4.
Mol Cancer ; 17(1): 59, 2018 02 17.
Article in English | MEDLINE | ID: mdl-29454361

ABSTRACT

BACKGROUND: During embryonic development Wnt family members and bone morphogenetic proteins (BMPs) cooperatively induce epithelial-mesenchymal transition (EMT) in the neural crest. Wnt and BMPs are reactivated during malignant transformation in melanoma. We previously demonstrated that the BMP-antagonist noggin blocked the EMT phenotype of melanoma cells in the neural crest and malignant invasion of melanoma cells in the chick embryo; vice-versa, malignant invasion was induced in human melanocytes in vivo by pre-treatment with BMP-2. RESULTS: Although there are conflicting results in the literature about the role of ß-catenin for invasion of melanoma cells, we found Wnt/ß-catenin signaling to be analogously important for the EMT-like phenotype of human metastatic melanoma cells in the neural crest and during invasion: ß-catenin was frequently expressed at the invasive front of human primary melanomas and Wnt3a expression was inversely correlated with survival of melanoma patients. Accordingly, cytoplasmic ß-catenin levels were increased during invasion of melanoma cells in the rhombencephalon of the chick embryo. Fibroblast derived Wnt3a reduced melanoma cell adhesion and enhanced migration, while the ß-catenin inhibitor PKF115-584 increased adhesion and reduced migration in vitro and in the chick embryonic neural crest environment in vivo. Similarly, knockdown of ß-catenin impaired intradermal melanoma cell invasion and PKF115-584 efficiently reduced liver metastasis in a chick chorioallantoic membrane model. Our observations were accompanied by specific alterations in gene expression which are linked to overall survival of melanoma patients. CONCLUSION: We present a novel role for Wnt-signaling in neural crest like melanoma cell invasion and metastasis, stressing the crucial role of embryonic EMT-inducing neural crest signaling for the spreading of malignant melanoma.


Subject(s)
Cell Movement , Cell Transformation, Neoplastic/metabolism , Melanoma/etiology , Melanoma/metabolism , Neural Crest/metabolism , Phenotype , Wnt Signaling Pathway , Animals , Biomarkers , Cell Adhesion , Cell Movement/drug effects , Cell Movement/genetics , Chick Embryo , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Gene Knockdown Techniques , Humans , Melanoma/mortality , Melanoma/pathology , Melanoma, Experimental , Mice , Neoplasm Invasiveness , Neoplasm Metastasis , Neural Crest/pathology , Perylene/analogs & derivatives , Perylene/pharmacology , RNA, Small Interfering/genetics , Zebrafish , beta Catenin/genetics , beta Catenin/metabolism
5.
Cancer Metastasis Rev ; 36(1): 7-21, 2017 03.
Article in English | MEDLINE | ID: mdl-28321632

ABSTRACT

Progress in understanding and treating metastatic melanoma is the result of decades of basic and translational research as well as the development of better in vitro tools for modeling the disease. Here, we review the latest therapeutic options for metastatic melanoma and the known genetic and non-genetic mechanisms of resistance to these therapies, as well as the in vitro toolbox that has provided the greatest insights into melanoma progression. These include next-generation sequencing technologies and more complex 2D and 3D cell culture models to functionally test the data generated by genomics approaches. The combination of hypothesis generating and hypothesis testing paradigms reviewed here will be the foundation for the next phase of metastatic melanoma therapies in the coming years.


Subject(s)
Melanoma/pathology , Melanoma/therapy , Precision Medicine/methods , Translational Research, Biomedical/methods , Animals , Humans
6.
Curr Opin Oncol ; 30(2): 113-117, 2018 03.
Article in English | MEDLINE | ID: mdl-29227308

ABSTRACT

PURPOSE OF REVIEW: Bioinformatic insights from next-generation sequencing has been integral in understanding melanoma biology, resistance to treatment and provided new avenues for melanoma treatment. Whole-genome sequencing, whole-exome sequencing and RNA sequencing has redefined the molecular classification of melanoma, revealed distinct genetic aberrations that define clinical subtypes of melanoma and uncovered the diverse heterogeneity that resides in an individual tumor. RECENT FINDINGS: In this review, we will summarize the recent whole-genome study that catalogs the genomic landscape across many melanoma subtypes, the single-cell RNA sequencing studies that interrogates tumor heterogeneity and the personalized vaccine approaches to melanoma treatment. SUMMARY: Whole-genome sequencing of diverse subtypes of melanoma revealed acral and mucosal subtypes to have a different genomic landscape compared with cutaneous melanoma. Acral and mucosal melanomas are characterized by low mutation burden and high structural variants. Single-cell RNA sequencing revealed high intratumoral heterogeneity and the existence of rare intrinsic drug-resistant populations. Lastly, vaccination against tumor neoantigens could be a potential personalized medicine therapy for melanoma patients. In summary, bioinformatics research is deeply ingrained in all aspects of melanoma research and will continue to blossom together for many years to come.


Subject(s)
Melanoma/genetics , Skin Neoplasms/genetics , Computational Biology , Genome-Wide Association Study , Humans , Melanoma/pathology , Mucous Membrane/pathology , Skin Neoplasms/pathology
7.
Clin Proteomics ; 15: 13, 2018.
Article in English | MEDLINE | ID: mdl-29541007

ABSTRACT

BACKGROUND: MAP kinase inhibitor (MAPKi) therapy for BRAF mutated melanoma is characterized by high response rates but development of drug resistance within a median progression-free survival (PFS) of 9-12 months. Understanding mechanisms of resistance and identifying effective therapeutic alternatives is one of the most important scientific challenges in melanoma. Using proteomics, we want to specifically gain insight into the pathophysiological process of cerebral metastases. METHODS: Cerebral metastases from melanoma patients were initially analyzed by a LC-MS shotgun approach performed on a QExactive HF hybrid quadrupole-orbitrap mass spectrometer. For further validation steps after bioinformatics analysis, a targeted LC-QQQ-MS approach, as well as Western blot, immunohistochemistry and immunocytochemistry was performed. RESULTS: In this pilot study, we were able to identify 5977 proteins by LC-MS analysis (data are available via ProteomeXchange with identifier PXD007592). Based on PFS, samples were classified into good responders (PFS ≥ 6 months) and poor responders (PFS [Formula: see text] 3 months). By evaluating these proteomic profiles according to gene ontology (GO) terms, KEGG pathways and gene set enrichment analysis (GSEA), we could characterize differences between the two distinct groups. We detected an EMT feature (up-regulation of N-cadherin) as classifier between the two groups, V-type proton ATPases, cell adhesion proteins and several transporter and exchanger proteins to be significantly up-regulated in poor responding patients, whereas good responders showed an immune activation, among other features. We identified class-discriminating proteins based on nearest shrunken centroids, validated and quantified this signature by a targeted approach and could correlate parts of this signature with resistance using the CPL/MUW proteome database and survival of patients by TCGA analysis. We further validated an EMT-like signature as a major discriminator between good and poor responders on primary melanoma cells derived from cerebral metastases. Higher immune activity is demonstrated in patients with good response to MAPKi by immunohistochemical staining of biopsy samples of cerebral melanoma metastases. CONCLUSIONS: Employing proteomic analysis, we confirmed known extra-cerebral resistance mechanisms in the cerebral metastases and further discovered possible brain specific mechanisms of drug efflux, which might serve as treatment targets or as predictive markers for these kinds of metastasis.

8.
Exp Dermatol ; 27(6): 694-696, 2018 06.
Article in English | MEDLINE | ID: mdl-29577418

ABSTRACT

Methadone (Met) mainly acts as a µ-opioid receptor agonist. Recent evidence pointing towards the role of Met in sensitization of certain cancer cell lines to chemotherapeutic agents has promoted the hypothesis that Met may be a useful adjuvant to cancer chemotherapy. We wanted to address whether Met has, alone or in combination with a chemotherapeutic agent, an effect on melanoma cell viability in vitro. Only a small fraction (4.3%) of our 102 melanoma biobank cell lines with RNA-sequencing data showed expression of the main receptor for Met (OPRM1). We assessed the viability of melanoma cell lines with high, medium or low/no OPRM1 expression (OPRM1high , OPRM1med , OPRM1neg ) 72 hours after treatment with Met alone or combined with cisplatin (Cis). Our analyses show that Met alone did not affect cell viability. While Cis/Met treatment did not have an effect on viability of OPRM1med or OPRM1neg cell lines, it resulted in a slightly decreased cell viability of OPRM1high cells. Clinically, concurrent temozolomide/Met treatment did not have an effect in our single-case report of a patient suffering from uveal melanoma. Taken together, our findings do not provide evidence for recommending Met as an adjuvant to chemotherapy in patients with melanoma.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Survival/drug effects , Cisplatin/pharmacology , Melanoma/drug therapy , Methadone/pharmacology , Skin Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Drug Screening Assays, Antitumor , Fatal Outcome , Gene Expression , Humans , Melanoma/genetics , Methadone/therapeutic use , Middle Aged , Receptors, Opioid, mu/agonists , Receptors, Opioid, mu/genetics , Skin Neoplasms/genetics , Temozolomide/therapeutic use , Uveal Neoplasms/drug therapy
9.
Curr Opin Oncol ; 28(2): 172-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26742019

ABSTRACT

PURPOSE OF REVIEW: BRAF inhibitors achieve outstanding clinical response rates in BRAF-mutated melanoma patients but therapeutic resistance is common. Although combinatorial targeted therapy has recently improved patient survival, resistance still occurs, which might be because of the plasticity and heterogeneity of melanoma. Proteomics complements the mostly genomics-based approaches used so far to gain additional insights into the pathophysiological mechanisms driving melanoma progression under treatment. RECENT FINDINGS: Few proteomics studies have investigated mitogen-activated protein kinase inhibitor (MAPKi) resistance. Three technologies have been described: shotgun analysis, pressure cycling technology-sequential window acquisition of all theoretical masses (which offers an optimized protein extraction by the pressure cycling technology), and selected reaction monitoring for selected candidate evaluation. Preliminary data demonstrate that BRAFi resistance might be associated with enhanced expression of the lysosomal compartment, cell adhesion, and epithelial-mesenchymal transformation. Melanoma cells change their phenotypes in response to targeted therapy with MAPKi from a proliferative to an invasive state gaining epithelial-mesenchymal transformation features, which are associated with drug resistance. SUMMARY: Performing proteomics may lead to an enhanced understanding of the underlying mechanisms of MAPKi resistance and might offer new insights for rational therapies. Selected reaction monitoring can be used to evaluate predictive or pharmacodynamic biomarkers for tracking therapeutic responses and identifying early features of resistance.


Subject(s)
Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm/physiology , Melanoma/drug therapy , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Skin Neoplasms/drug therapy , Antibodies, Monoclonal/therapeutic use , Epithelial-Mesenchymal Transition/drug effects , Humans , Indoles/therapeutic use , Ipilimumab , MAP Kinase Signaling System/drug effects , Mass Spectrometry , Melanoma/pathology , Proteomics/methods , Skin Neoplasms/pathology , Sulfonamides/therapeutic use , Vemurafenib
10.
J Immunother Cancer ; 12(7)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38969522

ABSTRACT

BACKGROUND: Immune-related hepatitis (irHepatitis) is a relatively common immune-related adverse event (irAE) of checkpoint inhibitors. Often, it responds well to steroids; however, in refractory cases, further therapy is needed. Anti-tumor necrosis factor (TNF) antibodies are used for management of multiple irAEs, but there are little data in irHepatitis. Here, we report on safety and efficacy of infliximab in 10 cases of steroid-refractory irHepatitis. METHODS: We retrospectively reviewed patients treated with infliximab for steroid-refractory grade ≥3 irHepatitis at the Department of Dermatology, University Hospital Zurich. The positive response to infliximab was defined as no further increase in alanine aminotransferase (ALT)/aspartate aminotransferase (AST) above 50% than at the time of first infliximab infusion and control of irHepatitis without therapies other than steroids and infliximab. RESULTS: 10 patients with steroid-resistant irHepatitis grade ≥3 were treated with infliximab 5 mg/kg, of whom 7 (70%) responded positively. In two cases, the liver values increased over 50% before the irHepatitis could be controlled. In another case, therapies other than infliximab and steroids were given. At the median follow-up of 487 days, 90% of the patients demonstrated resolved irHepatitis without AST/ALT elevation following infliximab infusions. CONCLUSIONS: Treatment of irHepatitis with infliximab did not result in hepatotoxicity and led to long-lasting positive response in 9 of 10 of the cases. Further research is needed to evaluate the role of anti-TNF antibodies in management of irHepatitis.


Subject(s)
Infliximab , Steroids , Humans , Infliximab/therapeutic use , Infliximab/adverse effects , Male , Female , Retrospective Studies , Middle Aged , Aged , Steroids/therapeutic use , Hepatitis/drug therapy , Hepatitis/etiology , Adult , Chemical and Drug Induced Liver Injury/etiology
11.
Clin Exp Med ; 24(1): 234, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39352553

ABSTRACT

Metastatic uveal melanoma (mUM) is associated with poor prognosis. Ipilimumab/nivolumab has shown antitumor efficacy in phase II studies. Tebentafusp resulted in longer overall survival (OS) compared to investigator`s choice in a phase III study. We sought to describe the radiological response patterns of mUM patients treated with immunotherapy. Patients with mUM treated with ipilimumab/nivolumab and tebentafusp between July 2018 and December 2022, with available radiological assessment per RECISTv1.1 and/or imPERCIST5, were retrospectively identified and included. Progression-free survival (PFS) and OS rates, liver-specific response and pathological assessment in available liver biopsies were evaluated. In the ipilimumab/nivolumab group, median PFS (mPFS) was 2.9 months (95% CI 2.2-28.6) and mOS 28.9 months (95% CI 12.7-NR). Complete (CMR) and partial (PMR) metabolic response per imPERCIST5, and partial response (PR) per RECISTv1.1 were associated with longer PFS and OS by trend, compared to morphologically and metabolically stable or progressive disease. In the tebentafusp group, mPFS was 2.7 months (95% CI 2.2-3) and mOS 18.6 months (95% CI 11.5-NR). PMR and PR were associated with longer PFS by trend. In both treatments, the overall treatment response was associated with the radiological response at the liver site. In available liver tumor biopsies, differences in pathological and radiological responses were noted. ImPERCIST5 and RECIST v1.1 are valuable tools in the radiological response assessment, but both methods display limitations. Accurate biomarkers to stratify patients at risk for disease progression and future translational studies to investigate mechanisms of response and resistance are required.


Subject(s)
Immunotherapy , Ipilimumab , Melanoma , Nivolumab , Uveal Neoplasms , Humans , Uveal Neoplasms/pathology , Uveal Neoplasms/drug therapy , Uveal Neoplasms/mortality , Uveal Neoplasms/therapy , Uveal Neoplasms/immunology , Melanoma/drug therapy , Melanoma/pathology , Melanoma/therapy , Melanoma/immunology , Retrospective Studies , Male , Female , Middle Aged , Aged , Nivolumab/therapeutic use , Ipilimumab/therapeutic use , Immunotherapy/methods , Adult , Treatment Outcome , Drug Resistance, Neoplasm , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Progression-Free Survival
12.
Nat Cancer ; 5(9): 1390-1408, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39210005

ABSTRACT

Immune checkpoint inhibitors are standard-of-care for the treatment of advanced melanoma, but their use is limited by immune-related adverse events. Proteomic analyses and multiplex cytokine and chemokine assays from serum at baseline and at the adverse event onset indicated aberrant T cell activity with differential expression of type I and III immune signatures. This was in line with the finding of an increase in the proportion of CD4+ T cells with IL-17A expression at the adverse event onset in the peripheral blood using flow cytometry. Multiplex immunohistochemistry and spatial transcriptomics on immunotherapy-induced skin rash and colitis showed an increase in the proportion of CD4+ T cells with IL-17A expression. Anti-IL-17A was administered in two patients with mild myocarditis, colitis and skin rash with resolution of the adverse events. This study highlights the potential role of type III CD4+ T cells in adverse event development and provides proof-of-principle evidence for a clinical trial using anti-IL-17A for treating adverse events.


Subject(s)
CD4-Positive T-Lymphocytes , Immunotherapy , Interleukin-17 , Melanoma , Humans , Melanoma/drug therapy , Melanoma/immunology , CD4-Positive T-Lymphocytes/immunology , Immunotherapy/methods , Immunotherapy/adverse effects , Male , Female , Middle Aged , Aged , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/therapeutic use
13.
Clin Cancer Res ; 30(1): 159-175, 2024 01 05.
Article in English | MEDLINE | ID: mdl-37861398

ABSTRACT

PURPOSE: Despite high clinical need, there are no biomarkers that accurately predict the response of patients with metastatic melanoma to anti-PD-1 therapy. EXPERIMENTAL DESIGN: In this multicenter study, we applied protein depletion and enrichment methods prior to various proteomic techniques to analyze a serum discovery cohort (n = 56) and three independent serum validation cohorts (n = 80, n = 12, n = 17). Further validation analyses by literature and survival analysis followed. RESULTS: We identified several significantly regulated proteins as well as biological processes such as neutrophil degranulation, cell-substrate adhesion, and extracellular matrix organization. Analysis of the three independent serum validation cohorts confirmed the significant differences between responders (R) and nonresponders (NR) observed in the initial discovery cohort. In addition, literature-based validation highlighted 30 markers overlapping with previously published signatures. Survival analysis using the TCGA database showed that overexpression of 17 of the markers we identified correlated with lower overall survival in patients with melanoma. CONCLUSIONS: Ultimately, this multilayered serum analysis led to a potential marker signature with 10 key markers significantly altered in at least two independent serum cohorts: CRP, LYVE1, SAA2, C1RL, CFHR3, LBP, LDHB, S100A8, S100A9, and SAA1, which will serve as the basis for further investigation. In addition to patient serum, we analyzed primary melanoma tumor cells from NR and found a potential marker signature with four key markers: LAMC1, PXDN, SERPINE1, and VCAN.


Subject(s)
Melanoma , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Proteomics , Biomarkers, Tumor/metabolism , Survival Analysis
14.
Commun Biol ; 6(1): 830, 2023 08 10.
Article in English | MEDLINE | ID: mdl-37563418

ABSTRACT

Multi-omics profiling by CITE-seq bridges the RNA-protein gap in single-cell analysis but has been largely applied to liquid biopsies. Applying CITE-seq to clinically relevant solid biopsies to characterize healthy tissue and the tumor microenvironment is an essential next step in single-cell translational studies. In this study, gating of cell populations based on their transcriptome signatures for use in cell type-specific ridge plots allowed identification of positive antibody signals and setting of manual thresholds. Next, we compare five skin dissociation protocols by taking into account dissociation efficiency, captured cell type heterogeneity and recovered surface proteome. To assess the effect of enzymatic digestion on transcriptome and epitope expression in immune cell populations, we analyze peripheral blood mononuclear cells (PBMCs) with and without dissociation. To further assess the RNA-protein gap, RNA-protein we perform codetection and correlation analyses on thresholded protein values. Finally, in a proof-of-concept study, using protein abundance analysis on selected surface markers in a cohort of healthy skin, primary, and metastatic melanoma we identify CD56 surface marker expression on metastatic melanoma cells, which was further confirmed by multiplex immunohistochemistry. This work provides practical guidelines for processing and analysis of clinically relevant solid tissue biopsies for biomarker discovery.


Subject(s)
Melanoma , Membrane Proteins , Humans , Leukocytes, Mononuclear/metabolism , Melanoma/genetics , Melanoma/metabolism , Transcriptome , RNA , Tumor Microenvironment/genetics
15.
Cancer Res ; 83(23): 3974-3988, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37729428

ABSTRACT

Metastatic melanoma is either intrinsically resistant or rapidly acquires resistance to targeted therapy treatments, such as MAPK inhibitors (MAPKi). A leading cause of resistance to targeted therapy is a dynamic transition of melanoma cells from a proliferative to a highly invasive state, a phenomenon called phenotype switching. Mechanisms regulating phenotype switching represent potential targets for improving treatment of patients with melanoma. Using a drug screen targeting chromatin regulators in patient-derived three-dimensional MAPKi-resistant melanoma cell cultures, we discovered that PARP inhibitors (PARPi) restore sensitivity to MAPKis, independent of DNA damage repair pathways. Integrated transcriptomic, proteomic, and epigenomic analyses demonstrated that PARPis induce lysosomal autophagic cell death, accompanied by enhanced mitochondrial lipid metabolism that ultimately increases antigen presentation and sensitivity to T-cell cytotoxicity. Moreover, transcriptomic and epigenetic rearrangements induced by PARP inhibition reversed epithelial-mesenchymal transition-like phenotype switching, which redirected melanoma cells toward a proliferative and MAPKi-sensitive state. The combination of PARP and MAPKis synergistically induced cancer cell death both in vitro and in vivo in patient-derived xenograft models. Therefore, this study provides a scientific rationale for treating patients with melanoma with PARPis in combination with MAPKis to abrogate acquired therapy resistance. SIGNIFICANCE: PARP inhibitors can overcome resistance to MAPK inhibitors by activating autophagic cell death and reversing phenotype switching, suggesting that this synergistic combination could help improve the prognosis of patients with melanoma.


Subject(s)
Melanoma , Poly(ADP-ribose) Polymerase Inhibitors , Humans , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Proteomics , Drug Resistance, Neoplasm/genetics , Cell Line, Tumor , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Phenotype
16.
Cancer Res ; 83(7): 1128-1146, 2023 04 04.
Article in English | MEDLINE | ID: mdl-36946761

ABSTRACT

Clinical management of melanomas with NRAS mutations is challenging. Targeting MAPK signaling is only beneficial to a small subset of patients due to resistance that arises through genetic, transcriptional, and metabolic adaptation. Identification of targetable vulnerabilities in NRAS-mutated melanoma could help improve patient treatment. Here, we used multiomics analyses to reveal that NRAS-mutated melanoma cells adopt a mesenchymal phenotype with a quiescent metabolic program to resist cellular stress induced by MEK inhibition. The metabolic alterations elevated baseline reactive oxygen species (ROS) levels, leading these cells to become highly sensitive to ROS induction. In vivo xenograft experiments and single-cell RNA sequencing demonstrated that intratumor heterogeneity necessitates the combination of a ROS inducer and a MEK inhibitor to inhibit both tumor growth and metastasis. Ex vivo pharmacoscopy of 62 human metastatic melanomas confirmed that MEK inhibitor-resistant tumors significantly benefited from the combination therapy. Finally, oxidative stress response and translational suppression corresponded with ROS-inducer sensitivity in 486 cancer cell lines, independent of cancer type. These findings link transcriptional plasticity to a metabolic phenotype that can be inhibited by ROS inducers in melanoma and other cancers. SIGNIFICANCE: Metabolic reprogramming in drug-resistant NRAS-mutated melanoma cells confers sensitivity to ROS induction, which suppresses tumor growth and metastasis in combination with MAPK pathway inhibitors.


Subject(s)
Melanoma , Skin Neoplasms , Humans , Reactive Oxygen Species , Proto-Oncogene Proteins B-raf/genetics , Melanoma/drug therapy , Melanoma/genetics , Melanoma/pathology , Skin Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Mitogen-Activated Protein Kinase Kinases/genetics , Cell Line, Tumor , Mutation , Membrane Proteins/genetics , GTP Phosphohydrolases/genetics
17.
Nat Commun ; 14(1): 4253, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37474523

ABSTRACT

Immune checkpoint blockade therapy is beneficial and even curative for some cancer patients. However, the majority don't respond to immune therapy. Across different tumor types, pre-existing T cell infiltrates predict response to checkpoint-based immunotherapy. Based on in vitro pharmacological studies, mouse models and analyses of human melanoma patients, we show that the cytokine GDF-15 impairs LFA-1/ß2-integrin-mediated adhesion of T cells to activated endothelial cells, which is a pre-requisite of T cell extravasation. In melanoma patients, GDF-15 serum levels strongly correlate with failure of PD-1-based immune checkpoint blockade therapy. Neutralization of GDF-15 improves both T cell trafficking and therapy efficiency in murine tumor models. Thus GDF-15, beside its known role in cancer-related anorexia and cachexia, emerges as a regulator of T cell extravasation into the tumor microenvironment, which provides an even stronger rationale for therapeutic anti-GDF-15 antibody development.


Subject(s)
Melanoma , T-Lymphocytes , Humans , Mice , Animals , T-Lymphocytes/pathology , Lymphocyte Function-Associated Antigen-1 , Endothelial Cells/pathology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/pathology , Immunotherapy , Tumor Microenvironment
18.
Front Cell Dev Biol ; 10: 916033, 2022.
Article in English | MEDLINE | ID: mdl-35693944

ABSTRACT

Deregulation of cellular metabolism through metabolic rewiring and translational reprogramming are considered hallmark traits of tumor development and malignant progression. The transcription factor YY1 is a master regulator of metabolism that we have previously shown to orchestrate a metabolic program required for melanoma formation. In this study, we demonstrate that YY1, while being essential for primary melanoma formation, suppresses metastatic spreading. Its downregulation or loss resulted in the induction of an invasiveness gene program and sensitized melanoma cells for pro-invasive signaling molecules, such as TGF-ß. In addition, NGFR, a key effector in melanoma invasion and phenotype switching, was among the most upregulated genes after YY1 knockdown. High levels of NGFR were also associated with other metabolic stress inducers, further indicating that YY1 knockdown mimics a metabolic stress program associated with an increased invasion potential in melanoma. Accordingly, while counteracting tumor growth, loss of YY1 strongly promoted melanoma cell invasiveness in vitro and metastasis formation in melanoma mouse models in vivo. Thus, our findings show that the metabolic regulator YY1 controls phenotype switching in melanoma.

19.
J Invest Dermatol ; 142(12): 3304-3312.e5, 2022 12.
Article in English | MEDLINE | ID: mdl-35850206

ABSTRACT

Cutaneous squamous cell carcinoma (cSCC) is the second most common skin cancer worldwide. Cancer-associated stroma (CAS) is central to tumor development and strongly influences therapy response. Perineural infiltration (PNI) represents a major risk factor for cSCC and likely influences CAS reprogramming. However, stromal reprogramming in cSCC remains poorly characterized, and it is unknown whether and how PNI influences CAS. To address these questions, we analyzed CAS and matched normal stroma from 20 cSCC cases (11 without PNI and 9 with PNI) by laser-capture microdissection using RNA sequencing. Our analysis reveals extensive stromal reprogramming strongly driven by changes in immune cells, as validated using immunohistochemistry. Furthermore, CAS of cSCC displays markers of immune exhaustion, and multiplex spatial analysis suggests that PD-L1 expression on NK T cells contributes to T-cell exhaustion and immunosuppression. Finally, PNI is characterized by increased IL-17A. In PNI-negative cases, IL-17A derives predominantly from CD3+ cells. However, with PNI, we observe an increased contribution of fibroblasts to high IL-17A, which coincides with a significant increase in FAP+ cells. Our analysis elucidates the molecular landscape of CAS in cSCC and identifies the presence of immunosuppressive mechanisms, supporting further research into immunotherapy and anti‒IL-17A in cSCC, especially for cases with PNI.


Subject(s)
Carcinoma, Squamous Cell , Skin Neoplasms , Humans , Carcinoma, Squamous Cell/metabolism , Skin Neoplasms/metabolism , Immunohistochemistry , Stromal Cells , Laser Capture Microdissection
20.
Oncotarget ; 13: 1370-1379, 2022 12 29.
Article in English | MEDLINE | ID: mdl-36580495

ABSTRACT

The Covid-19 pandemic created new uncertainties in the management of metastatic melanoma patients. In particular, the impact of immunotherapy, targeted therapy, or chemotherapy on the risk of Sars-CoV-2 infection and severity was debated. In this study, we analyzed all patients with metastatic melanoma receiving therapy who developed Covid-19 between February 2020 and February 2022. We retrospectively collected demographic data, cancer-specific parameters, melanoma treatment regimen, comorbidities and Covid-19-specific parameters in these patients. Of the 350 patients with metastatic melanoma, 25 had Covid-19. The median age at the time of Covid-19 diagnosis was 66 years (range 36-86), 10 patients were female, and 15 patients were male. The treatment regimen during infection was immunotherapy in 12 cases, followed by targeted therapy (n = 8), chemotherapy (n = 2), and TVEC injections, follow-up and palliative therapy in 1 case each. The severity was mild in 17 patients and 8 had a moderate to critical course. Patients with a severe Covid-19 course were often older and had more comorbidities than patients with a mild infection. Many of the patients had a mild Covid-19 course despite having metastatic melanoma and systemic therapy. We therefore recommend continuing systemic therapy whenever possible, even in such exceptional situations as the Covid-19 pandemic.


Subject(s)
COVID-19 , Melanoma , Neoplasms, Second Primary , Humans , Male , Female , Adult , Middle Aged , Aged , Aged, 80 and over , Pandemics , Retrospective Studies , COVID-19 Testing , SARS-CoV-2 , Melanoma/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL