Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Country/Region as subject
Publication year range
2.
Nature ; 577(7791): 561-565, 2020 01.
Article in English | MEDLINE | ID: mdl-31942071

ABSTRACT

Checkpoint blockade therapies that reactivate tumour-associated T cells can induce durable tumour control and result in the long-term survival of patients with advanced cancers1. Current predictive biomarkers for therapy response include high levels of intratumour immunological activity, a high tumour mutational burden and specific characteristics of the gut microbiota2,3. Although the role of T cells in antitumour responses has thoroughly been studied, other immune cells remain insufficiently explored. Here we use clinical samples of metastatic melanomas to investigate the role of B cells in antitumour responses, and find that the co-occurrence of tumour-associated CD8+ T cells and CD20+ B cells is associated with improved survival, independently of other clinical variables. Immunofluorescence staining of CXCR5 and CXCL13 in combination with CD20 reveals the formation of tertiary lymphoid structures in these CD8+CD20+ tumours. We derived a gene signature associated with tertiary lymphoid structures, which predicted clinical outcomes in cohorts of patients treated with immune checkpoint blockade. Furthermore, B-cell-rich tumours were accompanied by increased levels of TCF7+ naive and/or memory T cells. This was corroborated by digital spatial-profiling data, in which T cells in tumours without tertiary lymphoid structures had a dysfunctional molecular phenotype. Our results indicate that tertiary lymphoid structures have a key role in the immune microenvironment in melanoma, by conferring distinct T cell phenotypes. Therapeutic strategies to induce the formation of tertiary lymphoid structures should be explored to improve responses to cancer immunotherapy.


Subject(s)
Melanoma/immunology , Melanoma/therapy , Tertiary Lymphoid Structures/immunology , Antigens, CD20/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B7-H1 Antigen/antagonists & inhibitors , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Chemokine CXCL13/metabolism , Humans , Immunologic Memory/immunology , Melanoma/genetics , Melanoma/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Phenotype , Prognosis , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Proteomics , RNA-Seq , Receptors, CXCR5/metabolism , Single-Cell Analysis , Survival Rate , T Cell Transcription Factor 1/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tertiary Lymphoid Structures/genetics , Treatment Outcome , Tumor Microenvironment/immunology
3.
Cancer Immunol Immunother ; 71(3): 553-563, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34272988

ABSTRACT

BACKGROUND: Studying tumor cell-T cell interactions in the tumor microenvironment (TME) can elucidate tumor immune escape mechanisms and help predict responses to cancer immunotherapy. METHODS: We selected 14 pairs of highly tumor-reactive tumor-infiltrating lymphocytes (TILs) and autologous short-term cultured cell lines, covering four distinct tumor types, and co-cultured TILs and tumors at sub-lethal ratios in vitro to mimic the interactions occurring in the TME. We extracted gene signatures associated with a tumor-directed T cell attack based on transcriptomic data of tumor cells. RESULTS: An autologous T cell attack induced pronounced transcriptomic changes in the attacked tumor cells, partially independent of IFN-γ signaling. Transcriptomic changes were mostly independent of the tumor histological type and allowed identifying common gene expression changes, including a shared gene set of 55 transcripts influenced by T cell recognition (Tumors undergoing T cell attack, or TuTack, focused gene set). TuTack scores, calculated from tumor biopsies, predicted the clinical outcome after anti-PD-1/anti-PD-L1 therapy in multiple tumor histologies. Notably, the TuTack scores did not correlate to the tumor mutational burden, indicating that these two biomarkers measure distinct biological phenomena. CONCLUSIONS: The TuTack scores measure the effects on tumor cells of an anti-tumor immune response and represent a comprehensive method to identify immunologically responsive tumors. Our findings suggest that TuTack may allow patient selection in immunotherapy clinical trials and warrant its application in multimodal biomarker strategies.


Subject(s)
Biomarkers, Tumor , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Neoplasms/etiology , Transcriptome , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Cell Line, Tumor , Coculture Techniques , Computational Biology/methods , DNA Contamination , Gene Expression Profiling/methods , Gene Expression Profiling/standards , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immune Checkpoint Inhibitors , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Organ Specificity , ROC Curve , Tumor Cells, Cultured
4.
Brief Bioinform ; 21(2): 729-740, 2020 03 23.
Article in English | MEDLINE | ID: mdl-30721923

ABSTRACT

The development of multigene classifiers for cancer prognosis, treatment prediction, molecular subtypes or clinicopathological groups has been a cornerstone in transcriptomic analyses of human malignancies for nearly two decades. However, many reported classifiers are critically limited by different preprocessing needs like normalization and data centering. In response, a new breed of classifiers, single sample predictors (SSPs), has emerged. SSPs classify samples in an N-of-1 fashion, relying on, e.g. gene rules comparing expression values within a sample. To date, several methods have been reported, but there is a lack of head-to-head performance comparison for typical cancer classification problems, representing an unmet methodological need in cancer bioinformatics. To resolve this need, we performed an evaluation of two SSPs [k-top-scoring pair classifier (kTSP) and absolute intrinsic molecular subtyping (AIMS)] for two case examples of different magnitude of difficulty in non-small cell lung cancer: gene expression-based classification of (i) tumor histology and (ii) molecular subtype. Through the analysis of ~2000 lung cancer samples for each case example (n = 1918 and n = 2106, respectively), we compared the performance of the methods for different sample compositions, training data set sizes, gene expression platforms and gene rule selections. Three main conclusions are drawn from the comparisons: both methods are platform independent, they select largely overlapping gene rules associated with actual underlying tumor biology and, for large training data sets, they behave interchangeably performance-wise. While SSPs like AIMS and kTSP offer new possibilities to move gene expression signatures/predictors closer to a clinical context, they are still importantly limited by the difficultness of the classification problem at hand.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Gene Expression Regulation, Neoplastic , Lung Neoplasms/pathology , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/classification , Carcinoma, Non-Small-Cell Lung/genetics , Case-Control Studies , Gene Expression Profiling/methods , Humans , Lung Neoplasms/classification , Lung Neoplasms/genetics
5.
J Med Genet ; 57(5): 316-321, 2020 05.
Article in English | MEDLINE | ID: mdl-30291219

ABSTRACT

BACKGROUND: Inherited CDKN2A mutation is a strong risk factor for cutaneous melanoma. Moreover, carriers have been found to have poor melanoma-specific survival. In this study, responses to novel immunotherapy agents in CDKN2A mutation carriers with metastatic melanoma were evaluated. METHODS: CDKN2A mutation carriers that have developed metastatic melanoma and undergone immunotherapy treatments were identified among carriers enrolled in follow-up studies for familial melanoma. The carriers' responses were compared with responses reported in phase III clinical trials for CTLA-4 and PD-1 inhibitors. From publicly available data sets, melanomas with somatic CDKN2A mutation were analysed for association with tumour mutational load. RESULTS: Eleven of 19 carriers (58%) responded to the therapy, a significantly higher frequency than observed in clinical trials (p=0.03, binomial test against an expected rate of 37%). Further, 6 of the 19 carriers (32%) had complete response, a significantly higher frequency than observed in clinical trials (p=0.01, binomial test against an expected rate of 7%). In 118 melanomas with somatic CDKN2A mutations, significantly higher total numbers of mutations were observed compared with 761 melanomas without CDKN2A mutation (Wilcoxon test, p<0.001). CONCLUSION: Patients with CDKN2A mutated melanoma may have improved immunotherapy responses due to increased tumour mutational load, resulting in more neoantigens and stronger antitumorous immune responses.


Subject(s)
CTLA-4 Antigen/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , Melanoma/drug therapy , Programmed Cell Death 1 Receptor/genetics , Adult , Aged , CTLA-4 Antigen/antagonists & inhibitors , Clinical Trials as Topic , Female , Germ-Line Mutation/genetics , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/adverse effects , Immunotherapy/adverse effects , Ipilimumab/administration & dosage , Ipilimumab/adverse effects , Male , Melanoma/genetics , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Programmed Cell Death 1 Receptor/antagonists & inhibitors
6.
Breast Cancer Res ; 18(1): 27, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26923702

ABSTRACT

BACKGROUND: Aberrant DNA methylation is frequently observed in breast cancer. However, the relationship between methylation patterns and the heterogeneity of breast cancer has not been comprehensively characterized. METHODS: Whole-genome DNA methylation analysis using Illumina Infinium HumanMethylation450 BeadChip arrays was performed on 188 human breast tumors. Unsupervised bootstrap consensus clustering was performed to identify DNA methylation epigenetic subgroups (epitypes). The Cancer Genome Atlas data, including methylation profiles of 669 human breast tumors, was used for validation. The identified epitypes were characterized by integration with publicly available genome-wide data, including gene expression levels, DNA copy numbers, whole-exome sequencing data, and chromatin states. RESULTS: We identified seven breast cancer epitypes. One epitype was distinctly associated with basal-like tumors and with BRCA1 mutations, one epitype contained a subset of ERBB2-amplified tumors characterized by multiple additional amplifications and the most complex genomes, and one epitype displayed a methylation profile similar to normal epithelial cells. Luminal tumors were stratified into the remaining four epitypes, with differences in promoter hypermethylation, global hypomethylation, proliferative rates, and genomic instability. Specific hyper- and hypomethylation across the basal-like epitype was rare. However, we observed that the candidate genomic instability drivers BRCA1 and HORMAD1 displayed aberrant methylation linked to gene expression levels in some basal-like tumors. Hypomethylation in luminal tumors was associated with DNA repeats and subtelomeric regions. We observed two dominant patterns of aberrant methylation in breast cancer. One pattern, constitutively methylated in both basal-like and luminal breast cancer, was linked to genes with promoters in a Polycomb-repressed state in normal epithelial cells and displayed no correlation with gene expression levels. The second pattern correlated with gene expression levels and was associated with methylation in luminal tumors and genes with active promoters in normal epithelial cells. CONCLUSIONS: Our results suggest that hypermethylation patterns across basal-like breast cancer may have limited influence on tumor progression and instead reflect the repressed chromatin state of the tissue of origin. On the contrary, hypermethylation patterns specific to luminal breast cancer influence gene expression, may contribute to tumor progression, and may present an actionable epigenetic alteration in a subset of luminal breast cancers.


Subject(s)
Breast Neoplasms/genetics , Chromatin/genetics , DNA Methylation/genetics , Epigenesis, Genetic/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , CpG Islands/genetics , Female , Gene Expression Regulation, Neoplastic , Genome, Human , Humans , Mammary Glands, Human/metabolism , Neoplasm Proteins/genetics , Oligonucleotide Array Sequence Analysis/methods , Promoter Regions, Genetic
7.
J Pathol ; 233(1): 39-50, 2014 May.
Article in English | MEDLINE | ID: mdl-24399611

ABSTRACT

Diversity between metastatic melanoma tumours in individual patients is known; however, the molecular and genetic differences remain unclear. To examine the molecular and genetic differences between metastatic tumours, we performed gene-expression profiling of 63 melanoma tumours obtained from 28 patients (two or three tumours/patient), followed by analysis of their mutational landscape, using targeted deep sequencing of 1697 cancer genes and DNA copy number analysis. Gene-expression signatures revealed discordant phenotypes between tumour lesions within a patient in 50% of the cases. In 18 of 22 patients (where matched normal tissue was available), we found that the multiple lesions within a patient were genetically divergent, with one or more melanoma tumours harbouring 'private' somatic mutations. In one case, the distant subcutaneous metastasis of one patient occurring 3 months after an earlier regional lymph node metastasis had acquired 37 new coding sequence mutations, including mutations in PTEN and CDH1. However, BRAF and NRAS mutations, when present in the first metastasis, were always preserved in subsequent metastases. The patterns of nucleotide substitutions found in this study indicate an influence of UV radiation but possibly also DNA alkylating agents. Our results clearly demonstrate that metastatic melanoma is a molecularly highly heterogeneous disease that continues to progress throughout its clinical course. The private aberrations observed on a background of shared aberrations within a patient provide evidence of continued evolution of individual tumours following divergence from a common parental clone, and might have implications for personalized medicine strategies in melanoma treatment.


Subject(s)
Biomarkers, Tumor/genetics , Melanoma/genetics , Melanoma/secondary , Mutation , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Antigens, CD , Cadherins/genetics , Chromosomes, Human , DNA Copy Number Variations , DNA Mutational Analysis/methods , Disease Progression , Female , GTP Phosphohydrolases/genetics , Gene Dosage , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Rearrangement , Genetic Predisposition to Disease , High-Throughput Nucleotide Sequencing , Humans , Lymphatic Metastasis , Male , Membrane Proteins/genetics , Middle Aged , PTEN Phosphohydrolase/genetics , Phenotype , Proto-Oncogene Proteins B-raf/genetics , Transcriptome
8.
Am J Pathol ; 183(3): 681-91, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23827819

ABSTRACT

We recently defined molecular subtypes of urothelial carcinomas according to whole genome gene expression. Herein we describe molecular pathologic characterization of the subtypes using 20 genes and IHC of 237 tumors. In addition to differences in expression levels, the subtypes show important differences in stratification of protein expression. The selected genes included biological features central to bladder cancer biology, eg, cell cycle activity, cellular architecture, cell-cell interactions, and key receptor tyrosine kinases. We show that the urobasal (Uro) A subtype shares features with normal urothelium such as keratin 5 (KRT5), P-cadherin (P-Cad), and epidermal growth factor receptor (EGFR) expression confined to basal cells, and cell cycle activity (CCNB1) restricted to the tumor-stroma interface. In contrast, the squamous cell cancer-like (SCCL) subtype uniformly expresses KRT5, P-Cad, EGFR, KRT14, and cell cycle genes throughout the tumor parenchyma. The genomically unstable subtype shows proliferation throughout the tumor parenchyma and high ERBB2 and E-Cad expression but absence of KRT5, P-Cad, and EGFR expression. UroB tumors demonstrate features shared by both UroA and SCCL subtypes. A major transition in tumor progression seems to be loss of dependency of stromal interaction for proliferation. We present a simple IHC/histology-based classifier that is easy to implement as a standard pathologic evaluation to differentiate the three major subtypes: urobasal, genomically unstable, and SCCL. These three major subtypes exhibit important prognostic differences.


Subject(s)
Urinary Bladder Neoplasms/classification , Urinary Bladder Neoplasms/pathology , Urothelium/pathology , Biomarkers, Tumor/metabolism , Carcinoma, Squamous Cell/pathology , Cell Proliferation , Gene Expression Regulation, Neoplastic , Genome, Human/genetics , Humans , Immunohistochemistry , Kaplan-Meier Estimate , RNA, Messenger/genetics , RNA, Messenger/metabolism , Urinary Bladder Neoplasms/genetics , Urothelium/metabolism
9.
Nat Commun ; 15(1): 3075, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38594286

ABSTRACT

Immune checkpoint blockade (ICB) has improved outcome for patients with metastatic melanoma but not all benefit from treatment. Several immune- and tumor intrinsic features are associated with clinical response at baseline. However, we need to further understand the molecular changes occurring during development of ICB resistance. Here, we collect biopsies from a cohort of 44 patients with melanoma after progression on anti-CTLA4 or anti-PD1 monotherapy. Genetic alterations of antigen presentation and interferon gamma signaling pathways are observed in approximately 25% of ICB resistant cases. Anti-CTLA4 resistant lesions have a sustained immune response, including immune-regulatory features, as suggested by multiplex spatial and T cell receptor (TCR) clonality analyses. One anti-PD1 resistant lesion harbors a distinct immune cell niche, however, anti-PD1 resistant tumors are generally immune poor with non-expanded TCR clones. Such immune poor microenvironments are associated with melanoma cells having a de-differentiated phenotype lacking expression of MHC-I molecules. In addition, anti-PD1 resistant tumors have reduced fractions of PD1+ CD8+ T cells as compared to ICB naïve metastases. Collectively, these data show the complexity of ICB resistance and highlight differences between anti-CTLA4 and anti-PD1 resistance that may underlie differential clinical outcomes of therapy sequence and combination.


Subject(s)
Melanoma , Humans , Melanoma/drug therapy , Melanoma/genetics , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , CD8-Positive T-Lymphocytes , Programmed Cell Death 1 Receptor , Receptors, Antigen, T-Cell , Tumor Microenvironment
10.
Breast Cancer Res ; 14(1): R31, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22333393

ABSTRACT

INTRODUCTION: Male breast cancer (MBC) is a rare and inadequately characterized disease. The aim of the present study was to characterize MBC tumors transcriptionally, to classify them into comprehensive subgroups, and to compare them with female breast cancer (FBC). METHODS: A total of 66 clinicopathologically well-annotated fresh frozen MBC tumors were analyzed using Illumina Human HT-12 bead arrays, and a tissue microarray with 220 MBC tumors was constructed for validation using immunohistochemistry. Two external gene expression datasets were used for comparison purposes: 37 MBCs and 359 FBCs. RESULTS: Using an unsupervised approach, we classified the MBC tumors into two subgroups, luminal M1 and luminal M2, respectively, with differences in tumor biological features and outcome, and which differed from the intrinsic subgroups described in FBC. The two subgroups were recapitulated in the external MBC dataset. Luminal M2 tumors were characterized by high expression of immune response genes and genes associated with estrogen receptor (ER) signaling. Luminal M1 tumors, on the other hand, despite being ER positive by immunohistochemistry showed a lower correlation to genes associated with ER signaling and displayed a more aggressive phenotype and worse prognosis. Validation of two of the most differentially expressed genes, class 1 human leukocyte antigen (HLA) and the metabolizing gene N-acetyltransferase-1 (NAT1), respectively, revealed significantly better survival associated with high expression of both markers (HLA, hazard ratio (HR) 3.6, P = 0.002; NAT1, HR 2.5, P = 0.033). Importantly, NAT1 remained significant in a multivariate analysis (HR 2.8, P = 0.040) and may thus be a novel prognostic marker in MBC. CONCLUSIONS: We have detected two unique and stable subgroups of MBC with differences in tumor biological features and outcome. They differ from the widely acknowledged intrinsic subgroups of FBC. As such, they may constitute two novel subgroups of breast cancer, occurring exclusively in men, and which may consequently require novel treatment approaches. Finally, we identified NAT1 as a possible prognostic biomarker for MBC, as suggested by NAT1 positivity corresponding to better outcome.


Subject(s)
Arylamine N-Acetyltransferase/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms, Male/enzymology , Carcinoma, Ductal, Breast/enzymology , Carcinoma, Intraductal, Noninfiltrating/enzymology , Isoenzymes/metabolism , Transcriptome , Adult , Aged , Aged, 80 and over , Arylamine N-Acetyltransferase/genetics , Biomarkers, Tumor/genetics , Breast Neoplasms, Male/classification , Breast Neoplasms, Male/diagnosis , Breast Neoplasms, Male/mortality , Carcinoma, Ductal, Breast/classification , Carcinoma, Ductal, Breast/diagnosis , Carcinoma, Ductal, Breast/mortality , Carcinoma, Intraductal, Noninfiltrating/classification , Carcinoma, Intraductal, Noninfiltrating/diagnosis , Carcinoma, Intraductal, Noninfiltrating/mortality , Cluster Analysis , Female , Gene Expression Profiling , Humans , Isoenzymes/genetics , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Oligonucleotide Array Sequence Analysis , Principal Component Analysis , Prognosis , Statistics, Nonparametric , Tissue Array Analysis , Young Adult
11.
Clin Cancer Res ; 28(9): 1751-1758, 2022 05 02.
Article in English | MEDLINE | ID: mdl-34965949

ABSTRACT

Tumor cells pose a challenge to the adaptive immune system, and its key cell types, T and B cells, have frequently been associated with an improved prognosis. The success of immune checkpoint blockade has confirmed the relevance of T cells. However, the role of B cells is increasingly recognized, and highlighted in this review. Recent data suggest that tumors contain a diverse set of B cells reflecting different developmental states and exerting functions such as antigen presentation, antibody production, and regulatory effects. Further, B cells are frequently located in tertiary lymphoid structures (TLS), which are immune cell niches that sustain an immune response at sites of chronic inflammation. TLSs in tumors display substantial heterogeneity, ranging from cell aggregates to mature structures with an active germinal center. Recent studies have provided insights into initiation, cellular and spatial composition, and function of TLS in a variety of cancer types; however, several critical issues still need to be resolved. Currently, initial reports are discerning the role of TLSs in immunotherapy, with the majority of studies observing TLSs to confer favorable patient outcome. Finally, TLS induction in tumors is evaluated, with the therapeutic aim to reactivate the host immune response.


Subject(s)
Neoplasms , Tertiary Lymphoid Structures , B-Lymphocytes , Germinal Center/metabolism , Humans , Immunotherapy , Neoplasms/pathology , Tumor Microenvironment
12.
JCI Insight ; 7(19)2022 10 10.
Article in English | MEDLINE | ID: mdl-36040798

ABSTRACT

Cellular stress contributes to the capacity of melanoma cells to undergo phenotype switching into highly migratory and drug-tolerant dedifferentiated states. Such dedifferentiated melanoma cell states are marked by loss of melanocyte-specific gene expression and increase of mesenchymal markers. Two crucial transcription factors, microphthalmia-associated transcription factor (MITF) and SRY-box transcription factor 10 (SOX10), important in melanoma development and progression, have been implicated in this process. In this study we describe that loss of MITF is associated with a distinct transcriptional program, MITF promoter hypermethylation, and poor patient survival in metastatic melanoma. From a comprehensive collection of melanoma cell lines, we observed that MITF-methylated cultures were subdivided in 2 distinct subtypes. Examining mRNA levels of neural crest-associated genes, we found that 1 subtype had lost the expression of several lineage genes, including SOX10. Intriguingly, SOX10 loss was associated with SOX10 gene promoter hypermethylation and distinct phenotypic and metastatic properties. Depletion of SOX10 in MITF-methylated melanoma cells using CRISPR/Cas9 supported these findings. In conclusion, this study describes the significance of melanoma state and the underlying functional properties explaining the aggressiveness of such states.


Subject(s)
Melanoma , Microphthalmia-Associated Transcription Factor , DNA/metabolism , Humans , Melanocytes/pathology , Melanoma/pathology , Microphthalmia-Associated Transcription Factor/genetics , Microphthalmia-Associated Transcription Factor/metabolism , Phenotype , RNA, Messenger/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism
13.
Front Immunol ; 12: 705422, 2021.
Article in English | MEDLINE | ID: mdl-34707600

ABSTRACT

Detecting the entire repertoire of tumor-specific reactive tumor-infiltrating lymphocytes (TILs) is essential for investigating their immunological functions in the tumor microenvironment. Current in vitro assays identifying tumor-specific functional activation measure the upregulation of surface molecules, de novo production of antitumor cytokines, or mobilization of cytotoxic granules following recognition of tumor-antigens, yet there is no widely adopted standard method. Here we established an enhanced, yet simple, method for identifying simultaneously CD8+ and CD4+ tumor-specific reactive TILs in vitro, using a combination of widely known and available flow cytometry assays. By combining the detection of intracellular CD137 and de novo production of TNF and IFNγ after recognition of naturally-presented tumor antigens, we demonstrate that a larger fraction of tumor-specific and reactive CD8+ TILs can be detected in vitro compared to commonly used assays. This assay revealed multiple polyfunctionality-based clusters of both CD4+ and CD8+ tumor-specific reactive TILs. In situ, the combined detection of TNFRSF9, TNF, and IFNG identified most of the tumor-specific reactive TIL repertoire. In conclusion, we describe a straightforward method for efficient identification of the tumor-specific reactive TIL repertoire in vitro, which can be rapidly adopted in most cancer immunology laboratories.


Subject(s)
Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/chemistry , CD8-Positive T-Lymphocytes/chemistry , Interferon-gamma/analysis , Lymphocytes, Tumor-Infiltrating/chemistry , Neoplasm Proteins/analysis , Tumor Necrosis Factor Receptor Superfamily, Member 9/analysis , Tumor Necrosis Factor-alpha/analysis , Antigens, CD/analysis , Apyrase/analysis , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Datasets as Topic , Flow Cytometry , Humans , Integrin alpha Chains/analysis , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Lymphocyte Activation/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Single-Cell Analysis , Transcriptome , Tumor Microenvironment/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
14.
J Immunother Cancer ; 9(7)2021 07.
Article in English | MEDLINE | ID: mdl-34210820

ABSTRACT

PURPOSE: Despite impressive response rates following adoptive transfer of autologous tumor-infiltrating lymphocytes (TILs) in patients with metastatic melanoma, improvement is needed to increase the efficacy and broaden the applicability of this treatment. We evaluated the use of vemurafenib, a small-molecule BRAF inhibitor with immunomodulatory properties, as priming before TIL harvest and adoptive T cell therapy in a phase I/II clinical trial. METHODS: 12 patients were treated with vemurafenib for 7 days before tumor excision and during the following weeks until TIL infusion. TILs were grown from tumor fragments, expanded in vitro and reinfused to the patient preceded by a lymphodepleting chemotherapy regimen and followed by interleukin-2 infusion. Extensive immune monitoring, tumor profiling and T cell receptor sequencing were performed. RESULTS: No unexpected toxicity was observed, and treatment was well tolerated. Of 12 patients, 1 achieved a complete response, 8 achieved partial response and 3 achieved stable disease. A PR and the CR are ongoing for 23 and 43 months, respectively. In vitro anti-tumor reactivity was found in TILs from 10 patients, including all patients achieving objective response. Serum and tumor biomarker analyses indicate that baseline cytokine levels and the number of T cell clones may predict response to TIL therapy. Further, TCR sequencing suggested skewing of TCR repertoire during in vitro expansion, promoting certain low frequency clonotypes. CONCLUSIONS: Priming with vemurafenib before infusion of TILs was safe and feasible, and induced objective clinical responses in this cohort of patients with checkpoint inhibitor-resistant metastatic melanoma. In this trial, vemurafenib treatment seemed to decrease attrition and could be considered to bridge the waiting time while TILs are prepared.


Subject(s)
Immunotherapy/methods , Melanoma/drug therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , T-Lymphocytes/metabolism , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Protein Kinase Inhibitors/pharmacology , Young Adult
15.
Nat Commun ; 12(1): 3707, 2021 06 17.
Article in English | MEDLINE | ID: mdl-34140478

ABSTRACT

While the major drivers of melanoma initiation, including activation of NRAS/BRAF and loss of PTEN or CDKN2A, have been identified, the role of key transcription factors that impose altered transcriptional states in response to deregulated signaling is not well understood. The POU domain transcription factor BRN2 is a key regulator of melanoma invasion, yet its role in melanoma initiation remains unknown. Here, in a BrafV600E PtenF/+ context, we show that BRN2 haplo-insufficiency promotes melanoma initiation and metastasis. However, metastatic colonization is less efficient in the absence of Brn2. Mechanistically, BRN2 directly induces PTEN expression and in consequence represses PI3K signaling. Moreover, MITF, a BRN2 target, represses PTEN transcription. Collectively, our results suggest that on a PTEN heterozygous background somatic deletion of one BRN2 allele and temporal regulation of the other allele elicits melanoma initiation and progression.


Subject(s)
Carcinogenesis/metabolism , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Genes, Tumor Suppressor , Homeodomain Proteins/metabolism , Melanoma/metabolism , POU Domain Factors/metabolism , Skin Neoplasms/metabolism , Animals , Carcinogenesis/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , Cohort Studies , DNA Copy Number Variations , Disease Progression , Gene Knockdown Techniques , Haploinsufficiency , Homeodomain Proteins/genetics , Humans , Immunohistochemistry , Melanoma/genetics , Melanoma/mortality , Melanoma/secondary , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microarray Analysis , Microphthalmia-Associated Transcription Factor/metabolism , Mutation , POU Domain Factors/genetics , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins B-raf/genetics , RNA, Small Interfering , Skin Neoplasms/genetics , Skin Neoplasms/mortality , Skin Neoplasms/secondary , Melanoma, Cutaneous Malignant
16.
Nat Commun ; 12(1): 1137, 2021 02 18.
Article in English | MEDLINE | ID: mdl-33602918

ABSTRACT

Adjuvant systemic therapies are now routinely used following resection of stage III melanoma, however accurate prognostic information is needed to better stratify patients. We use differential expression analyses of primary tumours from 204 RNA-sequenced melanomas within a large adjuvant trial, identifying a 121 metastasis-associated gene signature. This signature strongly associated with progression-free (HR = 1.63, p = 5.24 × 10-5) and overall survival (HR = 1.61, p = 1.67 × 10-4), was validated in 175 regional lymph nodes metastasis as well as two externally ascertained datasets. The machine learning classification models trained using the signature genes performed significantly better in predicting metastases than models trained with clinical covariates (pAUROC = 7.03 × 10-4), or published prognostic signatures (pAUROC < 0.05). The signature score negatively correlated with measures of immune cell infiltration (ρ = -0.75, p < 2.2 × 10-16), with a higher score representing reduced lymphocyte infiltration and a higher 5-year risk of death in stage II melanoma. Our expression signature identifies melanoma patients at higher risk of metastases and warrants further evaluation in adjuvant clinical trials.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Databases, Genetic , Humans , Machine Learning , Multivariate Analysis , Neoplasm Staging , Prognosis , Progression-Free Survival , Proportional Hazards Models , Reproducibility of Results , Time Factors , Treatment Outcome
17.
BMC Cancer ; 10: 532, 2010 Oct 06.
Article in English | MEDLINE | ID: mdl-20925936

ABSTRACT

BACKGROUND: Genome wide gene expression data is a rich source for the identification of gene signatures suitable for clinical purposes and a number of statistical algorithms have been described for both identification and evaluation of such signatures. Some employed algorithms are fairly complex and hence sensitive to over-fitting whereas others are more simple and straight forward. Here we present a new type of simple algorithm based on ROC analysis and the use of metagenes that we believe will be a good complement to existing algorithms. RESULTS: The basis for the proposed approach is the use of metagenes, instead of collections of individual genes, and a feature selection using AUC values obtained by ROC analysis. Each gene in a data set is assigned an AUC value relative to the tumor class under investigation and the genes are ranked according to these values. Metagenes are then formed by calculating the mean expression level for an increasing number of ranked genes, and the metagene expression value that optimally discriminates tumor classes in the training set is used for classification of new samples. The performance of the metagene is then evaluated using LOOCV and balanced accuracies. CONCLUSIONS: We show that the simple uni-variate gene expression average algorithm performs as well as several alternative algorithms such as discriminant analysis and the more complex approaches such as SVM and neural networks. The R package rocc is freely available at http://cran.r-project.org/web/packages/rocc/index.html.


Subject(s)
Computational Biology/methods , Genome-Wide Association Study , Genomics/methods , Urinary Bladder Neoplasms/genetics , Algorithms , Area Under Curve , Artificial Intelligence , Databases, Genetic , Gene Expression Regulation , Humans , Models, Statistical , Neural Networks, Computer , Pattern Recognition, Automated , ROC Curve , Reproducibility of Results
18.
Mol Oncol ; 14(5): 933-950, 2020 05.
Article in English | MEDLINE | ID: mdl-32147909

ABSTRACT

The presence of immune cells in the tumor microenvironment has been associated with response to immunotherapies across several cancer types, including melanoma. Despite its therapeutic relevance, characterization of the melanoma immune microenvironments remains insufficiently explored. To distinguish the immune microenvironment in a cohort of 180 metastatic melanoma clinical specimens, we developed a method using promoter CpG methylation of immune cell type-specific genes extracted from genome-wide methylation arrays. Unsupervised clustering identified three immune methylation clusters with varying levels of immune CpG methylation that are related to patient survival. Matching protein and gene expression data further corroborated the identified epigenetic characterization. Exploration of the possible immune exclusion mechanisms at play revealed likely dependency on MITF protein level and PTEN loss-of-function events for melanomas unresponsive to immunotherapies (immune-low). To understand whether melanoma tumors resemble other solid tumors in terms of immune methylation characteristics, we explored 15 different solid tumor cohorts from TCGA. Low-dimensional projection based on immune cell type-specific methylation revealed grouping of the solid tumors in line with melanoma immune methylation clusters rather than tumor types. Association of survival outcome with immune cell type-specific methylation differed across tumor and cell types. However, in melanomas immune cell type-specific methylation was associated with inferior patient survival. Exploration of the immune methylation patterns in a pan-cancer context suggested that specific immune microenvironments might occur across the cancer spectrum. Together, our findings underscore the existence of diverse immune microenvironments, which may be informative for future immunotherapeutic applications.


Subject(s)
Gene Expression Regulation, Neoplastic/immunology , Lymphocytes/cytology , Melanoma/immunology , Melanoma/metabolism , Myeloid Cells/cytology , Skin Neoplasms/metabolism , Tumor Microenvironment/immunology , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Carcinoma/genetics , Carcinoma/immunology , Carcinoma/metabolism , Cell Line, Tumor , Cohort Studies , CpG Islands , DNA Methylation , Databases, Genetic , Dendritic Cells/cytology , Dendritic Cells/metabolism , Epigenesis, Genetic , Glioma/genetics , Glioma/immunology , Glioma/metabolism , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Lymphocytes/metabolism , Macrophages/cytology , Macrophages/metabolism , Melanoma/genetics , Melanoma/secondary , Mesothelioma/genetics , Mesothelioma/immunology , Mesothelioma/metabolism , Microphthalmia-Associated Transcription Factor/metabolism , Myeloid Cells/metabolism , Neoplasm Metastasis/genetics , Neoplasm Metastasis/immunology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Promoter Regions, Genetic , Skin Neoplasms/genetics , Skin Neoplasms/mortality , Skin Neoplasms/pathology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
19.
Cancers (Basel) ; 12(3)2020 Mar 21.
Article in English | MEDLINE | ID: mdl-32245160

ABSTRACT

Checkpoint blockade therapies have changed the clinical management of metastatic melanoma patients considerably, showing survival benefits. Despite the clinical success, not all patients respond to treatment or they develop resistance. Although there are several treatment predictive biomarkers, understanding therapy resistance and the mechanisms of tumor immune evasion is crucial to increase the frequency of patients benefiting from treatment. The PTEN gene is thought to promote immune evasion and is frequently mutated in cancer and melanoma. Another feature of melanoma tumors that may affect the capacity of escaping T-cell recognition is melanoma cell dedifferentiation characterized by decreased expression of the microphtalmia-associated transcription factor (MITF) gene. In this study, we have explored the role of PTEN in prognosis, therapy response, and immune escape in the context of MITF expression using immunostaining and genomic data from a large cohort of metastatic melanoma. We confirmed in our cohort that PTEN alterations promote immune evasion highlighted by decreased frequency of T-cell infiltration in such tumors, resulting in a worse patient survival. More importantly, our results suggest that dedifferentiated PTEN negative melanoma tumors have poor patient outcome, no T-cell infiltration, and transcriptional properties rendering them resistant to targeted- and immuno-therapy.

SELECTION OF CITATIONS
SEARCH DETAIL