Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Gynecol Oncol ; 186: 26-34, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38555766

ABSTRACT

OBJECTIVES: Alterations in the tumor suppressor TP53 gene are the most common mutations in high grade serous ovarian carcinoma. The impact of TP53 mutations on clinical outcomes and platinum resistance is controversial. We sought to evaluate the genomic profile of high grade serous ovarian carcinoma and explore the association of TP53 mutations with platinum resistance. METHODS: Next generation sequencing data was obtained from our institutional database for patients with high grade serous ovarian carcinoma undergoing primary treatment. Sequencing data, demographic, and clinical information was reviewed. The primary outcome analyzed was time to recurrence or refractory diagnosis. Associations between the primary outcome and different classification schemes for TP53 mutations (structural, functional, hot spot, pathogenicity scores, immunohistochemical staining patterns) were performed. RESULTS: 209 patients met inclusion criteria. TP53 mutations were the most common mutation. There were no differences in platinum response with TP53 hotspot mutations or high pathogenicity scores. Presence of TP53 gain-of-function mutations or measure of TP53 gain-of function activity were not associated with platinum resistance. Immunohistochemical staining patterns correlated with expected TP53 protein function and were not associated with platinum resistance. CONCLUSIONS: TP53 hotspot mutations or high pathogenicity scores were not associated with platinum resistance or refractory disease. Contrary to prior studies, TP53 gain-of-function mutations were not associated with platinum resistance. Estimation of TP53 gain-of-function effect using missense mutation phenotype scores was not associated with platinum resistance. The polymorphic nature of TP53 mutations may be too complex to demonstrate effect using simple models, or response to platinum therapy may be independent of initiating TP53 mutation.


Subject(s)
Cystadenocarcinoma, Serous , Drug Resistance, Neoplasm , Mutation , Ovarian Neoplasms , Tumor Suppressor Protein p53 , Humans , Female , Ovarian Neoplasms/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Tumor Suppressor Protein p53/genetics , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/drug therapy , Middle Aged , Aged , Adult , Neoplasm Grading , Aged, 80 and over , Gain of Function Mutation , Retrospective Studies
2.
PLoS Pathog ; 19(9): e1011676, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37747933

ABSTRACT

Sustainable HIV remission after antiretroviral therapy (ART) withdrawal, or post-treatment control (PTC), remains a top priority for HIV treatment. We observed surprising PTC in an MHC-haplomatched cohort of MHC-M3+ SIVmac239+ Mauritian cynomolgus macaques (MCMs) initiated on ART at two weeks post-infection (wpi). None of the MCMs possessed MHC haplotypes previously associated with SIV control. For six months after ART withdrawal, we observed undetectable or transient viremia in seven of the eight MCMs, despite detecting replication competent SIV using quantitative viral outgrowth assays. In vivo depletion of CD8α+ cells induced rebound in all animals, indicating the observed PTC was mediated, at least in part, by CD8α+ cells. With intact proviral DNA assays, we found that MCMs had significantly smaller viral reservoirs two wpi than a cohort of identically infected rhesus macaques, a population that rarely develops PTC. We found a similarly small viral reservoir among six additional SIV+ MCMs in which ART was initiated at eight wpi, some of whom exhibited viral rebound. These results suggest that an unusually small viral reservoir is a hallmark among SIV+ MCMs. By evaluating immunological differences between MCMs that did and did not rebound, we identified that PTC was associated with a reduced frequency of CD4+ and CD8+ lymphocyte subsets expressing exhaustion markers. Together, these results suggest a combination of small reservoirs and immune-mediated virus suppression contribute to PTC in MCMs. Further, defining the immunologic mechanisms that engender PTC in this model may identify therapeutic targets for inducing durable HIV remission in humans.


Subject(s)
HIV Infections , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus , Humans , Animals , Macaca mulatta , CD8-Positive T-Lymphocytes , HIV Infections/drug therapy , Macaca fascicularis , Viral Load , Virus Replication , Anti-Retroviral Agents/therapeutic use , Anti-Retroviral Agents/pharmacology
3.
Biomark Res ; 11(1): 73, 2023 Jul 25.
Article in English | MEDLINE | ID: mdl-37491309

ABSTRACT

BACKGROUND: A subset of triple-negative breast cancers (TNBCs) have homologous recombination deficiency with upregulation of compensatory DNA repair pathways. PIKTOR, a combination of TAK-228 (TORC1/2 inhibitor) and TAK-117 (PI3Kα inhibitor), is hypothesized to increase genomic instability and increase DNA damage repair (DDR) deficiency, leading to increased sensitivity to DNA-damaging chemotherapy and to immune checkpoint blockade inhibitors. METHODS: 10 metastatic TNBC patients received 4 mg TAK-228 and 200 mg TAK-117 (PIKTOR) orally each day for 3 days followed by 4 days off, weekly, until disease progression (PD), followed by intravenous cisplatin 75 mg/m2 plus nab paclitaxel 220 mg/m2 every 3 weeks for up to 6 cycles. Patients received subsequent treatment with pembrolizumab and/or chemotherapy. Primary endpoints were objective response rate with cisplatin/nab paclitaxel and safety. Biopsies of a metastatic lesion were collected prior to and at PD on PIKTOR. Whole exome and RNA-sequencing and reverse phase protein arrays (RPPA) were used to phenotype tumors pre- and post-PIKTOR for alterations in DDR, proliferation, and immune response. RESULTS: With cisplatin/nab paclitaxel (cis/nab pac) therapy post PIKTOR, 3 patients had clinical benefit (1 partial response (PR) and 2 stable disease (SD) ≥ 6 months) and continued to have durable benefit in progression-free survival with pembrolizumab post-cis/nab pac for 1.2, 2, and 3.6 years. Their post-PIKTOR metastatic tissue displayed decreased mismatch repair (MMR), increased tumor mutation burden, and significantly lower levels of 53BP1, DAG Lipase ß, GCN2, AKT Ser473, and PKCzeta Thr410/403 compared to pre-PIKTOR tumor tissue. CONCLUSIONS: Priming patients' chemotherapy-pretreated metastatic TNBC with PIKTOR led to very prolonged response/disease control with subsequent cis/nab pac, followed by pembrolizumab, in 3 of 10 treated patients. Our multi-omics approach revealed a higher number of genomic alterations, reductions in MMR, and alterations in immune and stress response pathways post-PIKTOR in patients who had durable responses. TRIAL REGISTRATION: This clinical trial was registered on June 21, 2017, at ClinicalTrials.gov using identifier NCT03193853.

4.
bioRxiv ; 2023 Mar 02.
Article in English | MEDLINE | ID: mdl-36909458

ABSTRACT

Sustainable HIV remission after antiretroviral therapy (ART) withdrawal, or post-treatment control (PTC), remains a top priority for HIV treatment. We observed surprising PTC in an MHC-haplomatched cohort of MHC-M3+ SIVmac239+ Mauritian cynomolgus macaques (MCMs) initiated on ART at two weeks post-infection (wpi). For six months after ART withdrawal, we observed undetectable or transient viremia in seven of eight MCMs. In vivo depletion of CD8α+ cells induced rebound in all animals, indicating the PTC was mediated, at least in part, by CD8α+ cells. We found that MCMs had smaller acute viral reservoirs than a cohort of identically infected rhesus macaques, a population that rarely develops PTC. The mechanisms by which unusually small viral reservoirs and CD8α+ cell-mediated virus suppression enable PTC can be investigated using this MHC-haplomatched MCM model. Further, defining the immunologic mechanisms that engender PTC in this model may identify therapeutic targets for inducing durable HIV remission in humans.

5.
Cancer Prev Res (Phila) ; 15(5): 285-296, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35121582

ABSTRACT

The mechanisms underlying the regulation of a checkpoint receptor, PD-1, in tumor-infiltrating immune cells during the development of colorectal cancer are not fully understood. Here we demonstrate that COX-2-derived PGE2, an inflammatory mediator and tumor promoter, induces PD-1 expression by enhancing NFκB's binding to the PD-1 promoter via an EP4-PI3K-Akt signaling pathway in both CD8+ T cells and macrophages. Moreover, PGE2 suppresses CD8+ T-cell proliferation and cytotoxicity against tumor cells and impairs macrophage phagocytosis of cancer cells via an EP4-PI3K-Akt-NFκB-PD-1 signaling pathway. In contrast, inhibiting the COX-2-PGE2-EP4 pathway increases intestinal CD8+ T-cell activation and proliferation and enhances intestinal macrophage phagocytosis of carcinoma cells accompanied by reduction of PD-1 expression in intestinal CD8+ T cells and macrophages in ApcMin/+ mice. PD-1 expression correlates well with COX-2 levels in human colorectal cancer specimens. Both elevated PD-1 and COX-2 are associated with poorer overall survival in patients with colorectal cancer. Our results uncover a novel role of PGE2 in tumor immune evasion. They may provide the rationale for developing new therapeutic approaches to subvert this process by targeting immune checkpoint pathways using EP4 antagonists. In addition, our findings reveal a novel mechanism explaining how NSAIDs reduce colorectal cancer risk by suppressing tumor immune evasion. PREVENTION RELEVANCE: These findings provide a potential explanation underlying the chemopreventive effect of NSAIDs on reducing colorectal cancer incidence during premalignancy and provide a rationale for developing EP4 antagonists for colorectal cancer prevention and treatment. Simply targeting PGE2 signaling alone may be efficacious in colorectal cancer prevention and treatment, avoiding side effects associated with NSAIDs.


Subject(s)
Adenoma , Colorectal Neoplasms , Animals , Anti-Inflammatory Agents, Non-Steroidal , Colorectal Neoplasms/pathology , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Humans , Mice , Phosphatidylinositol 3-Kinases/metabolism , Programmed Cell Death 1 Receptor , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Prostaglandin E, EP4 Subtype/metabolism
6.
Med ; 2(6): 736-754, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34223403

ABSTRACT

BACKGROUND: Upregulated glucose metabolism is a common feature of tumors. Glucose can be broken down by either glycolysis or the oxidative pentose phosphate pathway (oxPPP). The relative usage within tumors of these catabolic pathways remains unclear. Similarly, the extent to which tumors make biomass precursors from glucose, versus take them up from the circulation, is incompletely defined. METHODS: We explore human triple negative breast cancer (TNBC) metabolism by isotope tracing with [1,2-13C]glucose, a tracer that differentiates glycolytic versus oxPPP catabolism and reveals glucose-driven anabolism. Patients enrolled in clinical trial NCT03457779 and received IV infusion of [1,2-13C]glucose during core biopsy of their primary TNBC. Tumor samples were analyzed for metabolite labeling by liquid chromatography-mass spectrometry (LC-MS). Genomic and proteomic analyses were performed and related to observed metabolic fluxes. FINDINGS: TNBC ferments glucose to lactate, with glycolysis dominant over the oxPPP. Most ribose phosphate is nevertheless produced by oxPPP. Glucose also feeds amino acid synthesis, including of serine, glycine, aspartate, glutamate, proline and glutamine (but not asparagine). Downstream in glycolysis, tumor pyruvate and lactate labeling exceeds that found in serum, indicating that lactate exchange via monocarboxylic transporters is less prevalent in human TNBC compared with most normal tissues or non-small cell lung cancer. CONCLUSIONS: Glucose directly feeds ribose phosphate, amino acid synthesis, lactate, and the TCA cycle locally within human breast tumors.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Triple Negative Breast Neoplasms , Amino Acids , Glucose/metabolism , Humans , Lactic Acid/metabolism , Proteomics , Ribosemonophosphates
7.
Sci Rep ; 11(1): 10740, 2021 05 24.
Article in English | MEDLINE | ID: mdl-34031440

ABSTRACT

The robust detection of disease-associated splice events from RNAseq data is challenging due to the potential confounding effect of gene expression levels and the often limited number of patients with relevant RNAseq data. Here we present a novel statistical approach to splicing outlier detection and differential splicing analysis. Our approach tests for differences in the percentages of sequence reads representing local splice events. We describe a software package called Bisbee which can predict the protein-level effect of splice alterations, a key feature lacking in many other splicing analysis resources. We leverage Bisbee's prediction of protein level effects as a benchmark of its capabilities using matched sets of RNAseq and mass spectrometry data from normal tissues. Bisbee exhibits improved sensitivity and specificity over existing approaches and can be used to identify tissue-specific splice variants whose protein-level expression can be confirmed by mass spectrometry. We also applied Bisbee to assess evidence for a pathogenic splicing variant contributing to a rare disease and to identify tumor-specific splice isoforms associated with an oncogenic mutation. Bisbee was able to rediscover previously validated results in both of these cases and also identify common tumor-associated splice isoforms replicated in two independent melanoma datasets.


Subject(s)
Alternative Splicing , Melanoma/genetics , Proto-Oncogene Proteins/metabolism , Sequence Analysis, RNA/methods , Computational Biology/methods , Gene Expression Profiling , Humans , Mass Spectrometry , Melanoma/metabolism , Mutation , Organ Specificity , Proto-Oncogene Proteins/genetics , Software
8.
Elife ; 92020 12 23.
Article in English | MEDLINE | ID: mdl-33355532

ABSTRACT

Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare and aggressive form of ovarian cancer. SCCOHT tumors have inactivating mutations in SMARCA4 (BRG1), one of the two mutually exclusive ATPases of the SWI/SNF chromatin remodeling complex. To address the role that BRG1 loss plays in SCCOHT tumorigenesis, we performed integrative multi-omic analyses in SCCOHT cell lines +/- BRG1 reexpression. BRG1 reexpression induced a gene and protein signature similar to an epithelial cell and gained chromatin accessibility sites correlated with other epithelial originating TCGA tumors. Gained chromatin accessibility and BRG1 recruited sites were strongly enriched for transcription-factor-binding motifs of AP-1 family members. Furthermore, AP-1 motifs were enriched at the promoters of highly upregulated epithelial genes. Using a dominant-negative AP-1 cell line, we found that both AP-1 DNA-binding activity and BRG1 reexpression are necessary for the gene and protein expression of epithelial genes. Our study demonstrates that BRG1 reexpression drives an epithelial-like gene and protein signature in SCCOHT cells that depends upon by AP-1 activity.


Subject(s)
Carcinoma, Small Cell/pathology , DNA Helicases/genetics , Hypercalcemia/pathology , Nuclear Proteins/genetics , Ovarian Neoplasms/metabolism , Transcription Factor AP-1/metabolism , Transcription Factors/genetics , Biomarkers, Tumor/analysis , Carcinoma, Small Cell/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , DNA Helicases/metabolism , Female , Humans , Hypercalcemia/genetics , Mutation/genetics , Nuclear Proteins/metabolism , Ovarian Neoplasms/pathology , Ovary/metabolism , Ovary/pathology , Transcription Factor AP-1/genetics , Transcription Factors/metabolism
9.
Clin Cancer Res ; 26(15): 3908-3917, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32156746

ABSTRACT

Small-cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare and highly aggressive ovarian malignancy. In almost all cases, it is associated with somatic and often germline pathogenic variants in SMARCA4, which encodes for the SMARCA4 protein (BRG1), a subunit of the SWI/SNF chromatin remodeling complex. Approximately 20% of human cancers possess pathogenic variants in at least one SWI/SNF subunit. Because of their role in regulating many important cellular processes including transcriptional control, DNA repair, differentiation, cell division, and DNA replication, SWI/SNF complexes with mutant subunits are thought to contribute to cancer initiation and progression. Fewer than 500 cases of SCCOHT have been reported in the literature and approximately 60% are associated with hypercalcemia. SCCOHT primarily affects females under 40 years of age who usually present with symptoms related to a pelvic mass. SCCOHT is an aggressive cancer, with long-term survival rates of 30% in early-stage cases. Although various treatment approaches have been proposed, there is no consensus on surveillance and therapeutic strategy. An international group of multidisciplinary clinicians and researchers recently formed the International SCCOHT Consortium to evaluate current knowledge and propose consensus surveillance and therapeutic recommendations, with the aim of improving outcomes. Here, we present an overview of the genetics of this cancer, provide updates on new treatment targets, and propose management guidelines for this challenging cancer.


Subject(s)
Carcinoma, Small Cell/genetics , DNA Helicases/genetics , Hypercalcemia/genetics , Nuclear Proteins/genetics , Ovarian Neoplasms/genetics , Transcription Factors/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Small Cell/blood , Carcinoma, Small Cell/mortality , Carcinoma, Small Cell/therapy , Chemotherapy, Adjuvant/methods , Chemotherapy, Adjuvant/standards , Chromatin Assembly and Disassembly/genetics , Female , Gynecology/standards , Humans , Hypercalcemia/blood , Hypercalcemia/pathology , Hypercalcemia/therapy , Medical Oncology/standards , Mutation , Ovarian Neoplasms/blood , Ovarian Neoplasms/mortality , Ovarian Neoplasms/therapy , Ovariectomy/standards , Ovary/pathology , Ovary/surgery , Practice Guidelines as Topic , Radiotherapy, Adjuvant/standards , Stem Cell Transplantation/standards , Survival Rate , Treatment Outcome
10.
Gastroenterology ; 158(4): 971-984.e10, 2020 03.
Article in English | MEDLINE | ID: mdl-31734182

ABSTRACT

BACKGROUND & AIMS: Prostaglandin E2 (PGE2) promotes colorectal tumor formation and progression by unknown mechanisms. We sought to identify microRNAs (miRNAs) that might mediate the effects of PGE2 on colorectal cancer (CRC) development. METHODS: We incubated LS174T colorectal cancer cells with PGE2 or without (control) and used miRNA-sequencing technology to compare expression patterns of miRNAs. We knocked down levels of specific miRNAs or proteins in cells using small interfering RNAs or genome editing. Cells were analyzed by immunoblot, quantitative polymerase chain reaction, chromosome immunoprecipitation, cell invasion, and luciferase reporter assays; we measured gene expression, binding activity, cell migration and invasion, and transcriptional activity of transcription factors. NOD-scidIL-2Rg-/- mice were given injections of LS174T cells, and growth of primary tumors and numbers of liver and lung metastases were quantified and analyzed by histology. We used public databases to identify correlations in gene expression pattern with patient outcomes. RESULTS: We identified miRNA 675-5p (miR675-5p) as the miRNA most highly up-regulated by incubation of colorectal cancer cells with PGE2. PGE2 increased expression of miR675-5p by activating expression of Myc, via activation of protein kinase B, also known as (AKT), nuclear factor κB, and ß-catenin. PGE2 increased the invasive activities of cultured CRC cells. LS174T cells incubated with PGE2 formed more liver and lung metastases in mice than control LS174T cells. We identified a 3' untranslated region in the TP53 messenger RNA that bound miR675-5p; binding resulted in loss of the p53 protein. Expression of miR675-5p or its precursor RNA, H19, correlated with expression of cyclooxygenase-1 and cyclooxygenase-2 and shorter survival times of patients with CRC. CONCLUSIONS: We found that treatment of mice with PGE2 increased CRC cells invasive activity and ability to form liver and lung metastases. PGE2 down-regulates expression of p53 by increasing expression of miR675-5p, which binds to and prevents translation of TP53 messenger RNA. These findings provide insight into the mechanisms by which PGE2 promotes tumor development and progression. Strategies to target PGE2 might be developed for treatment of CRC.


Subject(s)
Colorectal Neoplasms/metabolism , Dinoprostone/genetics , MicroRNAs/metabolism , Neoplasm Metastasis/genetics , Tumor Suppressor Protein p53/metabolism , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Humans , RNA, Long Noncoding/metabolism , Tumor Cells, Cultured , Up-Regulation/genetics
11.
Mol Cancer Ther ; 17(12): 2767-2779, 2018 12.
Article in English | MEDLINE | ID: mdl-30232145

ABSTRACT

Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare but extremely lethal malignancy that mainly impacts young women. SCCOHT is characterized by a diploid genome with loss of SMARCA4 and lack of SMARCA2 expression, two mutually exclusive ATPases of the SWI/SNF chromatin-remodeling complex. We and others have identified the histone methyltransferase EZH2 as a promising therapeutic target for SCCOHT, suggesting that SCCOHT cells depend on the alternation of epigenetic pathways for survival. In this study, we found that SCCOHT cells were more sensitive to pan-HDAC inhibitors compared with other ovarian cancer lines or immortalized cell lines tested. Pan-HDAC inhibitors, such as quisinostat, reversed the expression of a group of proteins that were deregulated in SCCOHT cells due to SMARCA4 loss, leading to growth arrest, apoptosis, and differentiation in vitro and suppressed tumor growth of xenografted tumors of SCCOHT cells. Moreover, combined treatment of HDAC inhibitors and EZH2 inhibitors at sublethal doses synergistically induced histone H3K27 acetylation and target gene expression, leading to rapid induction of apoptosis and growth suppression of SCCOHT cells and xenografted tumors. Therefore, our preclinical study highlighted the therapeutic potential of combined treatment of HDAC inhibitors with EZH2 catalytic inhibitors to treat SCCOHT.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Small Cell/drug therapy , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Histone Deacetylase Inhibitors/therapeutic use , Hypercalcemia/drug therapy , Ovarian Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Biocatalysis/drug effects , Carcinoma, Small Cell/complications , Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/pathology , Cell Cycle Checkpoints/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cell Lineage/drug effects , DNA Helicases/metabolism , Drug Synergism , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Hypercalcemia/complications , Mice , Nuclear Proteins/metabolism , Ovarian Neoplasms/complications , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Proteome/metabolism , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
12.
Clin Cancer Res ; 24(8): 1932-1943, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29440177

ABSTRACT

Purpose: Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is a rare, aggressive ovarian cancer in young women that is universally driven by loss of the SWI/SNF ATPase subunits SMARCA4 and SMARCA2. A great need exists for effective targeted therapies for SCCOHT.Experimental Design: To identify underlying therapeutic vulnerabilities in SCCOHT, we conducted high-throughput siRNA and drug screens. Complementary proteomics approaches profiled kinases inhibited by ponatinib. Ponatinib was tested for efficacy in two patient-derived xenograft (PDX) models and one cell-line xenograft model of SCCOHT.Results: The receptor tyrosine kinase (RTK) family was enriched in siRNA screen hits, with FGFRs and PDGFRs being overlapping hits between drug and siRNA screens. Of multiple potent drug classes in SCCOHT cell lines, RTK inhibitors were only one of two classes with selectivity in SCCOHT relative to three SWI/SNF wild-type ovarian cancer cell lines. We further identified ponatinib as the most effective clinically approved RTK inhibitor. Reexpression of SMARCA4 was shown to confer a 1.7-fold increase in resistance to ponatinib. Subsequent proteomic assessment of ponatinib target modulation in SCCOHT cell models confirmed inhibition of nine known ponatinib target kinases alongside 77 noncanonical ponatinib targets in SCCOHT. Finally, ponatinib delayed tumor doubling time 4-fold in SCCOHT-1 xenografts while reducing final tumor volumes in SCCOHT PDX models by 58.6% and 42.5%.Conclusions: Ponatinib is an effective agent for SMARCA4-mutant SCCOHT in both in vitro and in vivo preclinical models through its inhibition of multiple kinases. Clinical investigation of this FDA-approved oncology drug in SCCOHT is warranted. Clin Cancer Res; 24(8); 1932-43. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/pathology , Imidazoles/pharmacology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Pyridazines/pharmacology , Animals , Carcinoma, Small Cell/drug therapy , Cell Line, Tumor , Computational Biology/methods , Disease Models, Animal , Female , Humans , Mice , Ovarian Neoplasms/drug therapy , Protein Interaction Mapping , Protein Interaction Maps , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays
13.
Methods Mol Biol ; 1706: 367-379, 2018.
Article in English | MEDLINE | ID: mdl-29423809

ABSTRACT

Cancer is a complex genetic disease that can arise through the stepwise accumulation of mutations in oncogenes and tumor suppressor genes in a variety of different tissues. While the varied landscapes of mutations driving common cancer types such as lung, breast, and colorectal cancer have been comprehensively charted, the genetic underpinnings of many rare cancers remain poorly defined. Study of rare cancers faces unique methodological challenges, but collaborative enterprises that incorporate next generation sequencing, reach across disciplines (i.e., pathology, genetic epidemiology, genomics, functional biology, and preclinical modeling), engage advocacy groups, tumor registries, and clinical specialists are adding increasing resolution to the genomic landscapes of rare cancers. Here we describe the approaches and methods used to identify SMARCA4 mutations, which drive development of the rare ovarian cancer, small cell carcinoma of the ovary, hypercalcemic type (SCCOHT), and point to the broader relevance of this paradigm for future research in rare cancers.


Subject(s)
Carcinoma, Small Cell/genetics , DNA Helicases/genetics , Mutation , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Ovarian Neoplasms/genetics , Rare Diseases/genetics , Transcription Factors/genetics , Animals , Carcinoma, Small Cell/metabolism , Carcinoma, Small Cell/pathology , DNA Helicases/metabolism , Female , Humans , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Rare Diseases/metabolism , Rare Diseases/pathology , Transcription Factors/metabolism
14.
J Pathol ; 242(3): 371-383, 2017 07.
Article in English | MEDLINE | ID: mdl-28444909

ABSTRACT

Small cell carcinoma of the ovary, hypercalcaemic type (SCCOHT) is a rare but aggressive and untreatable malignancy affecting young women. We and others recently discovered that SMARCA4, a gene encoding the ATPase of the SWI/SNF chromatin-remodelling complex, is the only gene recurrently mutated in the majority of SCCOHT. The low somatic complexity of SCCOHT genomes and the prominent role of the SWI/SNF chromatin-remodelling complex in transcriptional control of genes suggest that SCCOHT cells may rely on epigenetic rewiring for oncogenic transformation. Herein, we report that approximately 80% (19/24) of SCCOHT tumour samples have strong expression of the histone methyltransferase EZH2 by immunohistochemistry, with the rest expressing variable amounts of EZH2. Re-expression of SMARCA4 suppressed the expression of EZH2 in SCCOHT cells. In comparison to other ovarian cell lines, SCCOHT cells displayed hypersensitivity to EZH2 shRNAs and two selective EZH2 inhibitors, GSK126 and EPZ-6438. EZH2 inhibitors induced cell cycle arrest, apoptosis, and cell differentiation in SCCOHT cells, along with the induction of genes involved in cell cycle regulation, apoptosis, and neuron-like differentiation. EZH2 inhibitors suppressed tumour growth and improved the survival of mice bearing SCCOHT xenografts. Therefore, our data suggest that loss of SMARCA4 creates a dependency on the catalytic activity of EZH2 in SCCOHT cells and that pharmacological inhibition of EZH2 is a promising therapeutic strategy for treating this disease. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Carcinoma, Small Cell/enzymology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Hypercalcemia/enzymology , Ovarian Neoplasms/enzymology , Animals , Apoptosis/physiology , Carcinoma, Ovarian Epithelial , Cell Cycle Checkpoints/physiology , Cell Line, Tumor , Cell Transformation, Neoplastic , DNA Helicases/deficiency , Down-Regulation , Enhancer of Zeste Homolog 2 Protein/metabolism , Female , Histone Methyltransferases , Humans , Neoplasm Transplantation , Neoplasms, Glandular and Epithelial/enzymology , Nuclear Proteins/deficiency , Transcription Factors/deficiency , Transplantation, Heterologous , Tumor Cells, Cultured , Up-Regulation
15.
Integr Biol (Camb) ; 6(2): 224-31, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24413730

ABSTRACT

Co-culture of multiple cell types within a single device enables the study of paracrine signaling events. However, extracting gene expression endpoints from co-culture experiments is laborious, due in part to pre-PCR processing of the sample (i.e., post-culture cell sorting and nucleic acid purification). Also, a significant loss of nucleic acid may occur during these steps, especially with microfluidic cell culture where lysate volumes are small and difficult to access. Here, we describe an integrated platform for performing microfluidic cell culture and extraction of mRNA for gene expression analysis. This platform was able to recover 30-fold more mRNA than a similar, non-integrated system. Additionally, using a breast cancer/bone marrow stroma co-culture, we recapitulated stromal-dependent, estrogen-independent growth of the breast cancer cells, coincident with transcriptional changes. We anticipate that this platform will be used for streamlined analysis of paracrine signaling events as well as for screening potential drugs and/or patient samples.


Subject(s)
Breast Neoplasms/genetics , Coculture Techniques/methods , Gene Expression Profiling/methods , Microfluidics/methods , Paracrine Communication/genetics , Stromal Cells/cytology , Cell Line, Tumor , Cell Proliferation , Coculture Techniques/instrumentation , Female , Humans , Microfluidics/instrumentation , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
16.
Integr Biol (Camb) ; 5(5): 807-16, 2013 May.
Article in English | MEDLINE | ID: mdl-23559098

ABSTRACT

Breast cancer cell growth and therapeutic response are manipulated extrinsically by microenvironment signals. Despite recognition of the importance of the microenvironment in a variety of tumor processes, predictive measures that incorporate the activity of the surrounding cellular environment are lacking. In contrast, tumor cell biomarkers are well established in the clinic. Expression of Estrogen Receptor-alpha (ERα) is the primary defining feature of hormonally responsive tumors and is the molecular target of therapy in the most commonly diagnosed molecular subtype of breast cancer. While a number of soluble factors have been implicated in ERα activation, the complexity of signaling between the cellular microenvironment and the cancer cell implies multivariate control. The cumulative impact of the microenvironment signaling, which we define as microenvironmental activity, is more difficult to predict than the sum of its parts. Here we tested the impact of an array of microenvironments on ERα signaling utilizing a microfluidic co-culture model. Quantitative immunofluorescence was employed to assess changes in ERα protein levels, combined with gene expression and phosphorylation status, as measures of activation. Analysis of microenvironment-induced growth under the same conditions revealed a previously undescribed correlation between growth and ERα protein down-regulation. These data suggest an expanded utility for the tumor biomarker ERα, in which the combination of dynamic regulation of ERα protein and growth in a breast cancer biosensor cell become a read-out of the microenvironmental activity.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Estrogen Receptor alpha/metabolism , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Breast Neoplasms/genetics , Cell Line, Tumor , Coculture Techniques , Female , Gene Expression Regulation, Neoplastic , Humans , Microfluidics , Microscopy, Fluorescence , Neoplasms, Hormone-Dependent/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...