Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 164
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
Mol Cancer ; 23(1): 64, 2024 03 26.
Article in English | MEDLINE | ID: mdl-38532456

ABSTRACT

Previous studies have shown the clinical benefit of rechallenging the RAF pathway in melanoma patients previously treated with BRAF inhibitors. 44 patients with multiple tumors harboring RAF alterations were rechallenged with a second RAF inhibitor, either as monotherapy or in combination with other therapies, after prior therapy with a first RAF inhibitor. This retrospective observational study results showed that rechallenging with RAFi(s) led to an overall response rate of 18.1% [PR in thyroid (1 anaplastic; 3 papillary), 1 ovarian, 2 melanoma, 1 cholangiocarcinoma, and 1 anaplastic astrocytoma]. The clinical benefit rate was 54.5%; more than 30% of patients had durable responses with PR and SD lasting > 6 months. The median progression-free survival on therapy with second RAF inhibitor in the rechallenge setting either as monotherapy or combination was shorter at 2.7 months (0.9-30.1 m) compared to 8.6 months (6.5-11.5 m) with RAF-1i. However, the median PFS with RAF-2i responders (PFS-2) improved at 12.8 months compared to 11.4 months with RAF-1i responders. The median OS from retreatment with RAF-2i was 15.5 months (11.1-30.8 m). Further prospective studies are needed to validate these results and expand targeted therapy options for RAF-aberrant cancers.


Subject(s)
Melanoma , Humans , Melanoma/pathology , Proto-Oncogene Proteins B-raf/genetics , Protein Kinase Inhibitors/therapeutic use , Retrospective Studies , Prospective Studies , Mutation
3.
Gastroenterology ; 162(4): 1303-1318.e18, 2022 04.
Article in English | MEDLINE | ID: mdl-34973294

ABSTRACT

BACKGROUND & AIMS: RNF43 is an E3 ubiquitin ligase that is recurrently mutated in pancreatic ductal adenocarcinoma (PDAC) and precursor cystic neoplasms of the pancreas. The impact of RNF43 mutations on PDAC is poorly understood and autochthonous models have not been characterized sufficiently. In this study, we describe a genetically engineered mouse model (GEMM) of PDAC with conditional expression of oncogenic Kras and deletion of the catalytic domain of Rnf43 in exocrine cells. METHODS: We generated Ptf1a-Cre;LSL-KrasG12D;Rnf43flox/flox (KRC) and Ptf1a-Cre; LSL-KrasG12D (KC) mice and animal survival was assessed. KRC mice were sacrificed at 2 months, 4 months, and at moribund status followed by analysis of pancreata by single-cell RNA sequencing. Comparative analyses between moribund KRC and a moribund Kras/Tp53-driven PDAC GEMM (KPC) was performed. Cell lines were isolated from KRC and KC tumors and interrogated by cytokine array analyses, ATAC sequencing, and in vitro drug assays. KRC GEMMs were also treated with an anti-CTLA4 neutralizing antibody with treatment response measured by magnetic response imaging. RESULTS: We demonstrate that KRC mice display a marked increase in incidence of high-grade cystic lesions of the pancreas and PDAC compared with KC. Importantly, KRC mice have a significantly decreased survival compared with KC mice. Using single-cell RNA sequencing, we demonstrated that KRC tumor progression is accompanied by a decrease in macrophages, as well as an increase in T and B lymphocytes, with evidence of increased immune checkpoint molecule expression and affinity maturation, respectively. This was in stark contrast to the tumor immune microenvironment observed in the KPC PDAC GEMM. Furthermore, expression of the chemokine CXCL5 was found to be specifically decreased in KRC cancer cells by means of epigenetic regulation and emerged as a putative candidate for mediating the unique KRC immune landscape. CONCLUSIONS: The KRC GEMM establishes RNF43 as a bona fide tumor suppressor gene in PDAC. This GEMM features a markedly different immune microenvironment compared with previously reported PDAC GEMMs and puts forth a rationale for an immunotherapy approach in this subset of PDAC cases.


Subject(s)
Adenocarcinoma , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Ubiquitin-Protein Ligases , Adenocarcinoma/genetics , Animals , Carcinoma, Pancreatic Ductal/pathology , Disease Models, Animal , Epigenesis, Genetic , Humans , Mice , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Tumor Microenvironment , Ubiquitin-Protein Ligases/genetics , Pancreatic Neoplasms
4.
J Immunol ; 206(8): 1966-1975, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33722878

ABSTRACT

Inflammation has long been associated with cancer initiation and progression; however, how inflammation causes immune suppression in the tumor microenvironment and resistance to immunotherapy is not well understood. In this study, we show that both innate proinflammatory cytokine IL-1α and immunotherapy-induced IL-1α make melanoma resistant to immunotherapy. In a mouse melanoma model, we found that tumor size was inversely correlated with response to immunotherapy. Large tumors had higher levels of IL-1α, Th2 cytokines, polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs), and regulatory T cells but lower levels of IL-12, Th1 cytokines, and activated T cells. We found that therapy with adenovirus-encoded CD40L (rAd.CD40L) increased tumor levels of IL-1α and PMN-MDSCs. Blocking the IL-1 signaling pathway significantly decreased rAd.CD40L-induced PMN-MDSCs and their associated PD-L1 expression in the tumor microenvironment and enhanced tumor-specific immunity. Similarly, blocking the IL-1 signaling pathway improved the antimelanoma activity of anti-PD-L1 Ab therapy. Our study suggests that blocking the IL-1α signaling pathway may increase the efficacy of immunotherapies against melanoma.


Subject(s)
Drug Resistance, Neoplasm/immunology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Interleukin-1alpha/immunology , Melanoma, Experimental/therapy , Animals , Cell Line, Tumor , Cytokines/immunology , Cytokines/metabolism , Humans , Immune Checkpoint Inhibitors/immunology , Interleukin-1alpha/metabolism , Kaplan-Meier Estimate , Melanoma, Experimental/immunology , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
5.
Gastroenterology ; 161(1): 196-210, 2021 07.
Article in English | MEDLINE | ID: mdl-33745946

ABSTRACT

BACKGROUND & AIMS: Understanding the mechanisms by which tumors adapt to therapy is critical for developing effective combination therapeutic approaches to improve clinical outcomes for patients with cancer. METHODS: To identify promising and clinically actionable targets for managing colorectal cancer (CRC), we conducted a patient-centered functional genomics platform that includes approximately 200 genes and paired this with a high-throughput drug screen that includes 262 compounds in four patient-derived xenografts (PDXs) from patients with CRC. RESULTS: Both screening methods identified exportin 1 (XPO1) inhibitors as drivers of DNA damage-induced lethality in CRC. Molecular characterization of the cellular response to XPO1 inhibition uncovered an adaptive mechanism that limited the duration of response in TP53-mutated, but not in TP53-wild-type CRC models. Comprehensive proteomic and transcriptomic characterization revealed that the ATM/ATR-CHK1/2 axes were selectively engaged in TP53-mutant CRC cells upon XPO1 inhibitor treatment and that this response was required for adapting to therapy and escaping cell death. Administration of KPT-8602, an XPO1 inhibitor, followed by AZD-6738, an ATR inhibitor, resulted in dramatic antitumor effects and prolonged survival in TP53-mutant models of CRC. CONCLUSIONS: Our findings anticipate tremendous therapeutic benefit and support the further evaluation of XPO1 inhibitors, especially in combination with DNA damage checkpoint inhibitors, to elicit an enduring clinical response in patients with CRC harboring TP53 mutations.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Biomarkers, Tumor/genetics , Colorectal Neoplasms/drug therapy , Karyopherins/antagonists & inhibitors , Mutation , Protein Kinase Inhibitors/administration & dosage , Receptors, Cytoplasmic and Nuclear/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Databases, Genetic , HCT116 Cells , HT29 Cells , Humans , Indoles/administration & dosage , Karyopherins/metabolism , Mice , Morpholines/administration & dosage , Piperazines/administration & dosage , Pyridines/administration & dosage , Pyrimidines/administration & dosage , Receptors, Cytoplasmic and Nuclear/metabolism , Sulfonamides/administration & dosage , Xenograft Model Antitumor Assays , Exportin 1 Protein
6.
Hepatology ; 73(5): 1701-1716, 2021 05.
Article in English | MEDLINE | ID: mdl-32779242

ABSTRACT

BACKGROUND AND AIMS: 17-Beta hydroxysteroid dehydrogenase 13 (HSD17B13) is genetically associated with human nonalcoholic fatty liver disease (NAFLD). Inactivating mutations in HSD17B13 protect humans from NAFLD-associated and alcohol-associated liver injury, fibrosis, cirrhosis, and hepatocellular carcinoma, leading to clinical trials of anti-HSD17B13 therapeutic agents in humans. We aimed to study the in vivo function of HSD17B13 using a mouse model. APPROACH AND RESULTS: Single-cell RNA-sequencing and quantitative RT-PCR data revealed that hepatocytes are the main HSD17B13-expressing cells in mice and humans. We compared Hsd17b13 whole-body knockout (KO) mice and wild-type (WT) littermate controls fed regular chow (RC), a high-fat diet (HFD), a Western diet (WD), or the National Institute on Alcohol Abuse and Alcoholism model of alcohol exposure. HFD and WD induced significant weight gain, hepatic steatosis, and inflammation. However, there was no difference between genotypes with regard to body weight, liver weight, hepatic triglycerides (TG), histological inflammatory scores, expression of inflammation-related and fibrosis-related genes, and hepatic retinoid levels. Compared to WT, KO mice on the HFD had hepatic enrichment of most cholesterol esters, monoglycerides, and certain sphingolipid species. Extended feeding with the WD for 10 months led to extensive liver injury, fibrosis, and hepatocellular carcinoma, with no difference between genotypes. Under alcohol exposure, KO and WT mice showed similar hepatic TG and liver enzyme levels. Interestingly, chow-fed KO mice showed significantly higher body and liver weights compared to WT mice, while KO mice on obesogenic diets had a shift toward larger lipid droplets. CONCLUSIONS: Extensive evaluation of Hsd17b13 deficiency in mice under several fatty liver-inducing dietary conditions did not reproduce the protective role of HSD17B13 loss-of-function mutants in human NAFLD. Moreover, mouse Hsd17b13 deficiency induces weight gain under RC. It is crucial to understand interspecies differences prior to leveraging HSD17B13 therapies.


Subject(s)
17-Hydroxysteroid Dehydrogenases/deficiency , Diet, High-Fat/adverse effects , 17-Hydroxysteroid Dehydrogenases/metabolism , Animals , Diet, Western/adverse effects , Ethanol/adverse effects , Fatty Liver/etiology , Lipids/analysis , Liver/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Weight Gain
7.
Invest New Drugs ; 40(1): 157-162, 2022 02.
Article in English | MEDLINE | ID: mdl-34341905

ABSTRACT

BACKGROUND: Several TRK inhibitors have demonstrated clinical efficacy in patients with solid tumors harboring NTRK gene fusions. However, the natural history and prognostic implications of NTRK fusions in solid tumors remain unknown. METHODS: A cohort of 77 MD Anderson Cancer Center patients (MDACC) with NTRK gene fusions was identified and retrospectively compared to a second cohort from the Cancer Genome Atlas (TCGA) database. Due to paucity of events in early stage cancers and lack of TCGA data in rare tumors, 25 randomly selected MDACC patients were matched to 122 TCGA patients without NTRK gene fusion. Next we assessed the associations between NTRK gene fusion and overall (OS) and progression-free survivals (PFS). RESULTS: Among the 77 MDACC patients with NTRK gene fusions, 18 NTRK fusion partners were identified. There were insufficient OS events for analysis in the matched cohort. PFS was not significantly different (p = 0.49) between the NTRK-fusion positive MDACC patients (median PFS 786 weeks, 95% CI 317-NE) and the NTRK-fusion negative TCGA patients (median PFS NE). The adjusted hazard ratio comparing TCGA patients to MDACC patients was HR = 0.72 (95% CI: 0.23-2.33), which trended towards a reduced rate of progression or death experienced by TCGA patients. CONCLUSIONS: This study did not identify statistically significant associations between NTRK fusion and PFS. Nonsignificant trends estimated increases in the risk of progression or death events for patients with NTRK fusions when compared to matched controls. Our findings help illuminate the influence of NTRK fusions on the natural history of a variety of solid tumors.


Subject(s)
Gene Fusion , Neoplasms/genetics , Receptor Protein-Tyrosine Kinases/genetics , Humans , Neoplasms/mortality , Retrospective Studies , Survival Analysis
8.
Cancer Immunol Immunother ; 70(4): 1101-1113, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33123754

ABSTRACT

Although immunotherapy has achieved impressive durable clinical responses, many cancers respond only temporarily or not at all to immunotherapy. To find novel, targetable mechanisms of resistance to immunotherapy, patient-derived melanoma cell lines were transduced with 576 open reading frames, or exposed to arrayed libraries of 850 bioactive compounds, prior to co-culture with autologous tumor-infiltrating lymphocytes (TILs). The synergy between the targets and TILs to induce apoptosis, and the mechanisms of inhibiting resistance to TILs were interrogated. Gene expression analyses were performed on tumor samples from patients undergoing immunotherapy for metastatic melanoma. Finally, the effect of inhibiting the top targets on the efficacy of immunotherapy was investigated in multiple preclinical models. Aurora kinase was identified as a mediator of melanoma cell resistance to T-cell-mediated cytotoxicity in both complementary screens. Aurora kinase inhibitors were validated to synergize with T-cell-mediated cytotoxicity in vitro. The Aurora kinase inhibition-mediated sensitivity to T-cell cytotoxicity was shown to be partially driven by p21-mediated induction of cellular senescence. The expression levels of Aurora kinase and related proteins were inversely correlated with immune infiltration, response to immunotherapy and survival in melanoma patients. Aurora kinase inhibition showed variable responses in combination with immunotherapy in vivo, suggesting its activity is modified by other factors in the tumor microenvironment. These data suggest that Aurora kinase inhibition enhances T-cell cytotoxicity in vitro and can potentiate antitumor immunity in vivo in some but not all settings. Further studies are required to determine the mechanism of primary resistance to this therapeutic intervention.


Subject(s)
Aurora Kinase A/metabolism , Aurora Kinase B/metabolism , Drug Resistance, Neoplasm/immunology , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , T-Lymphocytes, Cytotoxic/transplantation , Animals , Apoptosis , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Aurora Kinase B/antagonists & inhibitors , Aurora Kinase B/genetics , Cell Proliferation , Female , Humans , Melanoma/genetics , Melanoma/metabolism , Melanoma/therapy , Mice , Prognosis , Survival Rate , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
10.
Proc Natl Acad Sci U S A ; 115(8): 1913-1918, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29434041

ABSTRACT

Polo-like kinase 4 (PLK4) is a serine/threonine kinase regulating centriole duplication. CFI-400945 is a highly selective PLK4 inhibitor that deregulates centriole duplication, causing mitotic defects and death of aneuploid cancers. Prior work was substantially extended by showing CFI-400945 causes polyploidy, growth inhibition, and apoptotic death of murine and human lung cancer cells, despite expression of mutated KRAS or p53. Analysis of DNA content by propidium iodide (PI) staining revealed cells with >4N DNA content (polyploidy) markedly increased after CFI-400945 treatment. Centrosome numbers and mitotic spindles were scored. CFI-400945 treatment produced supernumerary centrosomes and mitotic defects in lung cancer cells. In vivo antineoplastic activity of CFI-400945 was established in mice with syngeneic lung cancer xenografts. Lung tumor growth was significantly inhibited at well-tolerated dosages. Phosphohistone H3 staining of resected lung cancers following CFI-400945 treatment confirmed the presence of aberrant mitosis. PLK4 expression profiles in human lung cancers were explored using The Cancer Genome Atlas (TCGA) and RNA in situ hybridization (RNA ISH) of microarrays containing normal and malignant lung tissues. PLK4 expression was significantly higher in the malignant versus normal lung and conferred an unfavorable survival (P < 0.05). Intriguingly, cyclin dependent kinase 2 (CDK2) antagonism cooperated with PLK4 inhibition. Taken together, PLK4 inhibition alone or as part of a combination regimen is a promising way to combat lung cancer.


Subject(s)
Apoptosis/drug effects , Indazoles/pharmacology , Indoles/pharmacology , Polyploidy , Protein Serine-Threonine Kinases/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Centrosome , Gene Expression Regulation, Neoplastic , Humans , Indazoles/therapeutic use , Indoles/therapeutic use , Mice , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism
11.
Cancer Immunol Immunother ; 68(9): 1515-1526, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31515669

ABSTRACT

OBJECTIVE: To investigate the prognostic and biologic significance of immune-related gene expression in high grade serous ovarian cancer (HGSOC). METHODS: Gene expression dependent survival analyses for a panel of immune related genes were evaluated in HGSOC utilizing The Cancer Genome Atlas (TCGA). Prognostic value of LCK was validated using IHC in an independent set of 72 HGSOC. Prognostic performance of LCK was compared to cytolytic score (CYT) using RNAseq across multiple tumor types. Differentially expressed genes in LCK high samples and gene ontology enrichment were analyzed. RESULTS: High pre-treatment LCK mRNA expression was found to be a strong predictor of survival in a set of 535 ovarian cancers. Patients with high LCK mRNA expression had a longer median progression free survival (PFS) of 29.4 months compared to 16.9 months in those without LCK high expression (p = 0.003), and longer median overall survival (OS) of 95.1 months versus 44.5 months (p = 0.001), which was confirmed in an independent cohort by IHC (p = 0.04). LCK expression was compared to CYT across tumor types available in the TCGA and was a significant predictor of prognosis in HGSOC where CYT was not predictive. Unexpectedly, LCK high samples also were enriched in numerous immunoglobulin-related and other B cell transcripts. CONCLUSIONS: LCK is a better prognostic factor than CYT in ovarian cancer. In HGSOC, LCK high samples were characterized by higher expression of immunoglobulin and B-cell related genes suggesting that a cooperative interaction between tumor infiltrating T and B cells may correlate with better survival in this disease.


Subject(s)
B-Lymphocytes/physiology , Biomarkers, Tumor/metabolism , Cystadenocarcinoma, Serous/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Ovarian Neoplasms/immunology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/genetics , Cystadenocarcinoma, Serous/diagnosis , Cystadenocarcinoma, Serous/mortality , Cytotoxicity, Immunologic , Female , Gene Expression Regulation, Neoplastic , Gene Ontology , Humans , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Middle Aged , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/mortality , Predictive Value of Tests , Prognosis , Survival Analysis , Transcriptome
12.
Cancer Immunol Immunother ; 68(9): 1493-1500, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31501955

ABSTRACT

Immunotherapy with checkpoint inhibitors revolutionized melanoma treatment in both the adjuvant and metastatic setting, yet not all metastatic patients respond, and metastatic disease still often recurs among immunotherapy-treated patients with locally advanced disease. TNFSF4 is a co-stimulatory checkpoint protein expressed by several types of immune and non-immune cells, and was shown in the past to enhance the anti-neoplastic activity of T cells. Here, we assessed its expression in melanoma and its association with outcome in locally advanced and metastatic disease. We used publicly available data from The Cancer Genome Atlas (TCGA) and the Cancer Cell Line Encyclopedia (CCLE), and RNA sequencing data from anti-PD1-treated patients at Sheba medical center. TNFSF4 mRNA is expressed in melanoma cell lines and melanoma samples, including those with low lymphocytic infiltrates, and is not associated with the ulceration status of the primary tumor. Low expression of TNFSF4 mRNA is associated with worse prognosis in all melanoma patients and in the cohorts of stage III and stage IIIc-IV patients. Low expression of TNFSF4 mRNAs is also associated with worse prognosis in the subgroup of patients with low lymphocytic infiltrates, suggesting that tumoral TNFSF4 is associated with outcome. TNFSF4 expression was not correlated with the expression of other known checkpoint mRNAs. Last, metastatic patients with TNFSF4 mRNA expression within the lowest quartile have significantly worse outcome on anti-PD1 treatment, and a significantly lower response rate to these agents. Our current work points to TNFSF4 expression in melanoma as a potential determinant of prognosis, and warrants further translational and clinical research.


Subject(s)
Immunotherapy/methods , Melanoma/metabolism , OX40 Ligand/metabolism , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cohort Studies , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Melanoma/drug therapy , Melanoma/mortality , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Nivolumab/pharmacology , Nivolumab/therapeutic use , OX40 Ligand/genetics , Prognosis , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Survival Analysis , Treatment Outcome
13.
Proc Natl Acad Sci U S A ; 113(21): 5999-6004, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27162338

ABSTRACT

Cancer cells develop under immune surveillance, thus necessitating immune escape for successful growth. Loss of MHC class I expression provides a key immune evasion strategy in many cancers, although the molecular mechanisms remain elusive. MHC class I transactivator (CITA), known as "NLRC5" [NOD-like receptor (NLR) family, caspase recruitment (CARD) domain containing 5], has recently been identified as a critical transcriptional coactivator of MHC class I gene expression. Here we show that the MHC class I transactivation pathway mediated by CITA/NLRC5 constitutes a target for cancer immune evasion. In all the 21 tumor types we examined, NLRC5 expression was highly correlated with the expression of MHC class I, with cytotoxic T-cell markers, and with genes in the MHC class I antigen-presentation pathway, including LMP2/LMP7, TAP1, and ß2-microglobulin. Epigenetic and genetic alterations in cancers, including promoter methylation, copy number loss, and somatic mutations, were most prevalent in NLRC5 among all MHC class I-related genes and were associated with the impaired expression of components of the MHC class I pathway. Strikingly, NLRC5 expression was significantly associated with the activation of CD8(+) cytotoxic T cells and patient survival in multiple cancer types. Thus, NLRC5 constitutes a novel prognostic biomarker and potential therapeutic target of cancers.


Subject(s)
Antigen Presentation , Biomarkers, Tumor/immunology , Gene Expression Regulation, Neoplastic/immunology , Histocompatibility Antigens Class I/immunology , Intracellular Signaling Peptides and Proteins/immunology , Neoplasm Proteins/immunology , Neoplasms/immunology , Trans-Activators/immunology , Transcriptional Activation/immunology , Tumor Escape , ATP Binding Cassette Transporter, Subfamily B, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 2/immunology , Biomarkers, Tumor/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cysteine Endopeptidases/genetics , Cysteine Endopeptidases/immunology , Female , Histocompatibility Antigens Class I/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Male , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/immunology , Trans-Activators/genetics , beta 2-Microglobulin/genetics , beta 2-Microglobulin/immunology
14.
Cancer Metastasis Rev ; 36(2): 273-288, 2017 06.
Article in English | MEDLINE | ID: mdl-28681242

ABSTRACT

The consensus molecular subtypes (CMS) in colorectal cancer (CRC) represent distinct molecular subcategories of disease as reflected by comprehensive molecular profiling. The four CMS subtypes represent unique biology. CMS1 represents high immune infiltration. CMS2 demonstrates upregulation of canonical pathways such as WNT signaling. Widespread metabolic changes are seen in CMS3. CMS4 represents a mesenchymal phenotype with hallmark features including complement activation, matrix remodeling, angiogenesis, epithelial-mesechymal transition (EMT), integrin upregulation and stromal infiltration. In contrast to this new paradigm, a number of observations regarding CRC remain disconnected. Cancers are associated with thrombocytosis. Venous thromboembolic events are more likely in malignancy and may signify worse prognosis. Aspirin, an anti-platelet agent, has been linked in large observational studies to decrease incidence of adenocarcinoma and less advanced presentations of cancer, in particular CRC. Inflammatory bowel disease is a risk factor for CRC. Gross markers to recognize the immunothrombotic link such as the platelet to lymphocyte ratio are associated with poorer outcomes in many cancers. Platelets are increasingly recognized for their dual roles in coordinating the immune response in addition to hemostasis. Here, we explore how these different but related observations coalesce. Platelets, as first responders to pathogens and injury, form the link between hemostasis and immunity. We outline how platelets contribute to tumorigenesis and how some disconnected ideas may be linked through inflammation. CMS4 through its shared mechanisms has predicted platelet activation as a hallmark feature. We demonstrate a platelet gene expression signature that predicts platelet presence within CMS4 tumors.


Subject(s)
Blood Platelets/physiology , Colorectal Neoplasms/blood , Animals , Blood Platelets/pathology , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/pathology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Humans , Transcriptome
15.
J Pathol ; 243(3): 271-272, 2017 11.
Article in English | MEDLINE | ID: mdl-28816351

ABSTRACT

Adenosquamous carcinoma of the pancreas (ASCP) is a mixed tumor type which contains squamous cell carcinoma and also ductal adenocarcinoma components. Due to the rarity of this malignancy, only very limited genomic profiling has been performed. A recent paper by Fang et al. published in The Journal of Pathology contributed to our knowledge of genomic alterations by performing whole-genome and -exome sequencing of 17 ASCP tumors. They found major genomic similarities to pancreatic ductal adenocarcinoma; however, the p53 pathway was altered in a greater proportion of cases, while a high frequency of 3p loss was a distinct copy number alteration pattern observed in ASCP. Laser capture microdissection revealed that adenocarcinoma and squamous carcinoma components of ASCP harbor similar genomic variations, indicating that the origin of tumor components is the same or similar. Although the study published by Fang et al. increases our knowledge of this rare mixed tumor type, further investigation, including RNA sequencing, will be needed to fully characterize this malignancy and to aid the development of novel treatment approaches. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Carcinoma, Adenosquamous/genetics , Genomics , Mutation/genetics , Pancreas/pathology , Pancreatic Neoplasms/genetics , Animals , High-Throughput Nucleotide Sequencing/methods , Humans , Pancreatic Neoplasms
16.
Curr Oncol Rep ; 20(5): 37, 2018 03 26.
Article in English | MEDLINE | ID: mdl-29582189

ABSTRACT

Mesenchymal chondrosarcoma is a rare but deadly form of chondrosarcoma that typically affects adolescents and young adults. While curative intent is possible for patients with localized disease, few options exist for patients in the unresectable/metastatic setting. Thus, it is imperative to understand the fusion-driven biology of this rare malignant neoplasm so as to lead to the future development of better therapeutics for this disease. This manuscript will briefly review the clinical and pathologic features of mesenchymal chondrosarcoma followed by an appraisal of existing data linked to the fusions, HEY1-NCOA2 and IRF2BP2-CDX1, and the associated downstream pathways.


Subject(s)
Bone Neoplasms/pathology , Chondrosarcoma, Mesenchymal/pathology , Oncogene Proteins, Fusion/genetics , Bone Neoplasms/genetics , Chondrosarcoma, Mesenchymal/genetics , Humans , Prognosis
17.
Cancer ; 123(8): 1372-1381, 2017 04 15.
Article in English | MEDLINE | ID: mdl-27911979

ABSTRACT

BACKGROUND: BRAFV600 , NRAS, TP53, and BRAFNon-V600 are among the most common mutations detected in non-acral cutaneous melanoma patients. Although several studies have identified clinical and pathological features associated with BRAFV600 and NRAS mutations, limited data are available regarding the correlates and significance of TP53 and BRAFNon-V600 mutations. METHODS: This study analyzed the patient demographics, primary tumor features, and clinical outcomes of a large cohort of non-acral cutaneous melanoma patients who had undergone clinically indicated molecular testing (n = 926). RESULTS: The prevalence of BRAFV600 , NRAS, TP53, and BRAFNon-V600 mutations was 43%, 21%, 19%, and 7%, respectively. The presence of a TP53 mutation was associated with older age (P = .019), a head and neck primary tumor site (P = .0001), and longer overall survival (OS) from the diagnosis of stage IV disease in univariate (P = .039) and multivariate analyses (P = .015). BRAFNon-V600 mutations were associated with older age (P = .005) but not with primary tumor features or OS from stage IV. Neither TP53 nor BRAFNon-V600 mutations correlated significantly with OS with frontline ipilimumab treatment, and the TP53 status was not significantly associated with outcomes with frontline BRAF inhibitor therapy. Eleven patients with BRAFNon-V600 mutations were treated with a BRAF inhibitor. Three patients were not evaluable for a response because of treatment cessation for toxicities; the remaining patients had disease progression as the best response to therapy. CONCLUSIONS: These results add to the understanding of the clinical features associated with TP53 and BRAFNon-V600 mutations in advanced cutaneous melanoma patients, and they support the rationale for evaluating the prognostic significance of TP53 in other cohorts of melanoma patients. Cancer 2017;123:1372-1381. © 2016 American Cancer Society.


Subject(s)
Melanoma/diagnosis , Melanoma/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Skin Neoplasms/diagnosis , Skin Neoplasms/genetics , Tumor Suppressor Protein p53/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Child , DNA Mutational Analysis , Female , Humans , Kaplan-Meier Estimate , Male , Melanoma/drug therapy , Melanoma/mortality , Middle Aged , Molecular Targeted Therapy , Mutation Rate , Neoplasm Staging , Phenotype , Prognosis , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Skin Neoplasms/drug therapy , Skin Neoplasms/mortality , Tumor Burden , Young Adult , Melanoma, Cutaneous Malignant
18.
Biochem Biophys Res Commun ; 490(3): 868-875, 2017 08 26.
Article in English | MEDLINE | ID: mdl-28647357

ABSTRACT

Lysine 27 to methionine (K27 M) mutation of the histone variant H3.3 drives the formation of an aggressive glioblastoma multiforme tumor in infants. Here we analyzed how the methionine substitution alters the stability of H3.3 nucleosomes in vitro and modifies its kinetic properties in live cells. We also determined whether the presence of mutant nucleosomes perturbed the mobility of the PRC2 subunit Ezh2 (enhancer-of-zeste homolog 2). We found that K27 M nucleosomes maintained the wild-type molecular architecture both at the level of bulk histones and single nucleosomes and followed similar diffusion kinetics to wild-type histones in live cells. Nevertheless, we observed a remarkable differential recovery of Ezh2 in response to transcriptional stress that was accompanied by a faster diffusion rate of the mobile fraction of Ezh2 and a significantly increased immobile fraction, suggesting tighter chromatin binding of Ezh2 upon transcription inhibition. The differential recovery of Ezh2 was dependent on transcription, however, it was independent from K27 M mutation status. These biophysical characteristics shed more light on the mechanism of histone H3.3 K27M in glioma genesis in relation to the kinetic properties of Ezh2.


Subject(s)
Histones/genetics , Point Mutation , Animals , Enhancer of Zeste Homolog 2 Protein/analysis , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Fluorescence Resonance Energy Transfer , Glioblastoma/genetics , Glioblastoma/metabolism , HeLa Cells , Histones/analysis , Histones/metabolism , Humans , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcriptional Activation , Xenopus laevis
19.
Curr Oncol Rep ; 19(12): 78, 2017 Oct 13.
Article in English | MEDLINE | ID: mdl-29030741

ABSTRACT

PURPOSE OF REVIEW: Sarcomas are rare, heterogeneous group of soft tissue and bone tumors. Precise diagnosis of specific subtypes is challenging using conventional methods. Herein, we review the role of next-generation sequencing (NGS) technology that is used for rapid sequencing of DNA and RNA. RECENT FINDINGS: Recent sarcoma specific studies recommend that molecular genetic testing should be added at diagnosis for appropriate clinical management in addition to diagnosis by expert pathologists. NGS has already been used to identify potentially actionable mutations, copy number alterations, and gene fusions. Rationally, choosing a drug based on an individual patient profile aka: "precision oncology" has been so far limited to few case reports in sarcomas. As we improve our ability to deliver personalized medicine using all modalities including conventional therapy, more patients may eventually benefit. As the cost and capacity of NGS outpace Moore's law, so does the probability of success.


Subject(s)
High-Throughput Nucleotide Sequencing , Precision Medicine , Sarcoma/diagnosis , Sarcoma/genetics , Humans , Mutation , Sarcoma/pathology
20.
Proc Natl Acad Sci U S A ; 111(29): 10660-5, 2014 Jul 22.
Article in English | MEDLINE | ID: mdl-25002471

ABSTRACT

Clinical-grade T cells are genetically modified ex vivo to express chimeric antigen receptors (CARs) to redirect their specificity to target tumor-associated antigens in vivo. We now have developed this molecular strategy to render cytotoxic T cells specific for fungi. We adapted the pattern-recognition receptor Dectin-1 to activate T cells via chimeric CD28 and CD3-ζ (designated "D-CAR") upon binding with carbohydrate in the cell wall of Aspergillus germlings. T cells genetically modified with the Sleeping Beauty system to express D-CAR stably were propagated selectively on artificial activating and propagating cells using an approach similar to that approved by the Food and Drug Administration for manufacturing CD19-specific CAR(+) T cells for clinical trials. The D-CAR(+) T cells exhibited specificity for ß-glucan which led to damage and inhibition of hyphal growth of Aspergillus in vitro and in vivo. Treatment of D-CAR(+) T cells with steroids did not compromise antifungal activity significantly. These data support the targeting of carbohydrate antigens by CAR(+) T cells and provide a clinically appealing strategy to enhance immunity for opportunistic fungal infections using T-cell gene therapy.


Subject(s)
Aspergillosis/immunology , Aspergillosis/therapy , Bioengineering/methods , Carbohydrates/antagonists & inhibitors , Opportunistic Infections/immunology , Opportunistic Infections/therapy , T-Lymphocytes/immunology , Animals , Antigens, CD19/metabolism , Aspergillosis/microbiology , Aspergillosis/pathology , Aspergillus/drug effects , Aspergillus/physiology , Dexamethasone/pharmacology , Humans , Hyphae/drug effects , Hyphae/physiology , Immunophenotyping , Lectins, C-Type/metabolism , Lymphocyte Activation/drug effects , Mice , Opportunistic Infections/pathology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL