1
|
Yang Q, Falahati A, Khosh A, Lastra RR, Boyer TG, Al-Hendy A. Unraveling the Role of Bromodomain and Extra-Terminal Proteins in Human Uterine Leiomyosarcoma. Cells 2024; 13:1443. [PMID: 39273015 PMCID: PMC11394028 DOI: 10.3390/cells13171443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 08/17/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Uterine leiomyosarcoma (uLMS) is the most common type of uterine sarcoma, associated with poor prognosis, high rates of recurrence, and metastasis. Currently, the molecular mechanism of the origin and development of uLMS is limited. Bromodomain and extra-terminal (BET) proteins are involved in both physiological and pathological events. However, the role of BET proteins in the pathogenesis of uLMS is unknown. Here, we show for the first time that BET protein family members, BRD2, BRD3, and BRD4, are aberrantly overexpressed in uLMS tissues compared to the myometrium, with a significant change by histochemical scoring assessment. Furthermore, inhibiting BET proteins with their small, potent inhibitors (JQ1 and I-BET 762) significantly inhibited the uLMS proliferation dose-dependently via cell cycle arrest. Notably, RNA-sequencing analysis revealed that the inhibition of BET proteins with JQ1 and I-BET 762 altered several critical pathways, including the hedgehog pathway, EMT, and transcription factor-driven pathways in uLMS. In addition, the targeted inhibition of BET proteins altered several other epigenetic regulators, including DNA methylases, histone modification, and m6A regulators. The connections between BET proteins and crucial biological pathways provide a fundamental structure to better understand uterine diseases, particularly uLMS pathogenesis. Accordingly, targeting the vulnerable epigenome may provide an additional regulatory mechanism for uterine cancer treatment.
Collapse
Affiliation(s)
- Qiwei Yang
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA;
| | - Ali Falahati
- Poundbury Cancer Institute for Personalised Medicine, Dorchester DT1 3BJ, UK;
- DNA GTx LAB, Dubai Healthcare City, Dubai 505262, United Arab Emirates
| | - Azad Khosh
- Department of Molecular Medicine, Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.K.); (T.G.B.)
| | - Ricardo R. Lastra
- Department of Pathology, University of Chicago, Chicago, IL 60637, USA;
| | - Thomas G. Boyer
- Department of Molecular Medicine, Institute of Biotechnology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (A.K.); (T.G.B.)
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA;
| |
Collapse
|
2
|
Ahmadpour Youshanlui M, Yari A, Bahojb Mahdavi SZ, Amini M, Baradaran B, Ahangar R, Pourbagherian O, Mokhtarzadeh AA. BRD4 expression and its regulatory interaction with miR-26a-3p, DLG5-AS1, and JMJD1C-AS1 lncRNAs in gastric cancer progression. Discov Oncol 2024; 15:356. [PMID: 39152304 PMCID: PMC11329449 DOI: 10.1007/s12672-024-01230-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/12/2024] [Indexed: 08/19/2024] Open
Abstract
Gastric cancer remains a significant health challenge despite advancements in diagnosis and treatment. Early detection is critical to reducing mortality, necessitating the investigation of molecular mechanisms underlying gastric cancer progression. This study focuses on BRD4 expression and its correlation with miR-26a-3p, DLG5-AS1, and JMJD1C-AS1 lncRNAs in gastric cancer. Analysis of The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) datasets revealed significant upregulation of BRD4 in gastric cancer tissues compared to normal tissues, correlating negatively with miR-26a-3p and positively with DLG5-AS1 and JMJD1C-AS1 lncRNAs. Quantitative RT-PCR confirmed these findings in 25 gastric cancer tissue samples and 25 normal samples. BRD4's overexpression was associated with reduced survival rates and older patient age. MiR-26a-3p, a known tumor suppressor, showed decreased expression in gastric cancer tissues, with ROC analysis suggesting it, alongside BRD4, as a potential diagnostic biomarker. Additionally, bioinformatics predicted miR-26a-3p's interaction with BRD4 mRNA. Upregulated lncRNAs DLG5-AS1 and JMJD1C-AS1 likely act as competing endogenous RNAs, sponging miR-26a-3p, thus promoting BRD4 dysregulation. These lncRNAs have not been previously studied in gastric cancer. The findings propose a novel BRD4/lncRNA/miRNA regulatory axis in gastric cancer, highlighting the potential of BRD4, DLG5-AS1, and JMJD1C-AS1 as biomarkers for early diagnosis. Further studies with larger sample sizes and in vivo and in vitro experiments are needed to elucidate this regulatory mechanism's role in gastric cancer progression.
Collapse
Affiliation(s)
| | - Amirhossein Yari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Ahangar
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Omid Pourbagherian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
3
|
Stiegeler N, Garsed DW, Au-Yeung G, Bowtell DDL, Heinzelmann-Schwarz V, Zwimpfer TA. Homologous recombination proficient subtypes of high-grade serous ovarian cancer: treatment options for a poor prognosis group. Front Oncol 2024; 14:1387281. [PMID: 38894867 PMCID: PMC11183307 DOI: 10.3389/fonc.2024.1387281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/15/2024] [Indexed: 06/21/2024] Open
Abstract
Approximately 50% of tubo-ovarian high-grade serous carcinomas (HGSCs) have functional homologous recombination-mediated (HR) DNA repair, so-called HR-proficient tumors, which are often associated with primary platinum resistance (relapse within six months after completion of first-line therapy), minimal benefit from poly(ADP-ribose) polymerase (PARP) inhibitors, and shorter survival. HR-proficient tumors comprise multiple molecular subtypes including cases with CCNE1 amplification, AKT2 amplification or CDK12 alteration, and are often characterized as "cold" tumors with fewer infiltrating lymphocytes and decreased expression of PD-1/PD-L1. Several new treatment approaches aim to manipulate these negative prognostic features and render HR-proficient tumors more susceptible to treatment. Alterations in multiple different molecules and pathways in the DNA damage response are driving new drug development to target HR-proficient cancer cells, such as inhibitors of the CDK or P13K/AKT pathways, as well as ATR inhibitors. Treatment combinations with chemotherapy or PARP inhibitors and agents targeting DNA replication stress have shown promising preclinical and clinical results. New approaches in immunotherapy are also being explored, including vaccines or antibody drug conjugates. Many approaches are still in the early stages of development and further clinical trials will determine their clinical relevance. There is a need to include HR-proficient tumors in ovarian cancer trials and to analyze them in a more targeted manner to provide further evidence for their specific therapy, as this will be crucial in improving the overall prognosis of HGSC and ovarian cancer in general.
Collapse
Affiliation(s)
| | - Dale W. Garsed
- Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - George Au-Yeung
- Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - David D. L. Bowtell
- Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | | | - Tibor A. Zwimpfer
- Cancer Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Department of Gynecological Oncology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
4
|
Kourtis S, Cianferoni D, Serrano L, Sdelci S. Detection of differential bait proteoforms through immunoprecipitation-mass spectrometry data analysis. Sci Data 2024; 11:551. [PMID: 38811611 PMCID: PMC11137132 DOI: 10.1038/s41597-024-03394-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024] Open
Abstract
Proteins are often referred to as the workhorses of cells, and their interactions are necessary to facilitate specific cellular functions. Despite the recognition that protein-protein interactions, and thus protein functions, are determined by proteoform states, such as mutations and post-translational modifications (PTMs), methods for determining the differential abundance of proteoforms across conditions are very limited. Classically, immunoprecipitation coupled with mass spectrometry (IP-MS) has been used to understand how the interactome (preys) of a given protein (bait) changes between conditions to elicit specific cellular functions. Reversing this concept, we present here a new workflow for IP-MS data analysis that focuses on identifying the differential peptidoforms of the bait protein between conditions. This method can provide detailed information about specific bait proteoforms, potentially revealing pathogenic protein states that can be exploited for the development of targeted therapies.
Collapse
Affiliation(s)
- Savvas Kourtis
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - Damiano Cianferoni
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Luis Serrano
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Sara Sdelci
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain.
| |
Collapse
|
5
|
Andrikopoulou A, Bletsa G, Rouvalis A, Tsakogiannis D, Kaparelou M, Papatheodoridi A, Haidopoulos D, Liontos M, Dimopoulos MA, Zagouri F. The Prognostic Role of BRD4 Expression in High-Grade Serous Ovarian Cancer. Cancers (Basel) 2024; 16:1962. [PMID: 38893083 PMCID: PMC11171195 DOI: 10.3390/cancers16111962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/12/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Bromodomain and extra-terminal (BET) domain proteins that bind to acetylated lysine residues of histones serve as the "readers" of DNA acetylation. BRD4 is the most thoroughly studied member of the BET family and regulates the expression of key oncogenes. BRD4 gene amplification has been identified in ovarian cancer (~18-19%) according to The Cancer Genome Atlas (TCGA) analysis. BET inhibitors are novel small molecules that displace BET proteins from acetylated histones and are currently tested in Phase I/II trials. We here aim to explore the prognostic role of the BRD4 gene and protein expression in the ascitic fluid of patients with advanced FIGO III/IV high-grade serous ovarian carcinoma (HGSC). METHODS Ascitic fluid was obtained from 28 patients with advanced stage (FIGO III/IV) HGSC through diagnostic/therapeutic paracentesis or laparoscopy before the initiation of chemotherapy. An amount of ~200 mL of ascitic fluid was collected from each patient and peripheral blood mononuclear cells (PBMCs) were isolated. Each sample was evaluated for BRD4 and GAPDH gene expression through RT-qPCR and BRD4 protein levels through enzyme-linked immunosorbent assay (ELISA). The study protocol was approved by the Institutional Review Board of Alexandra University Hospital and the Committee on Ethics and Good Practice (CEGP) of the National and Kapodistrian University of Athens (NKUA). RESULTS Low BRD4 gene expression was associated with worse prognosis at 12 months compared to intermediate/high expression (95% CI; 1.75-30.49; p = 0.008). The same association was observed at 24 months although this association was not statistically significant (95% CI; 0.96-9.2; p = 0.065). Progression-free survival was shorter in patients with low BRD4 gene expression at 12 months (5.6 months; 95% CI; 2.6-8.6) compared to intermediate/high expression (9.8 months; 95% CI; 8.3-11.3) (95% CI; 1.2-16.5; p = 0.03). The same association was confirmed at 24 months (6.9 months vs. 13.1 months) (95% CI; 1.1-8.6; p = 0.048). There was a trend for worse prognosis in patients with high BRD4 protein levels versus intermediate/low BRD4 protein expression both at 12 months (9.8 months vs. 7.6 months; p = 0.3) and at 24 months (14.2 months vs. 16.6 months; p = 0.56) although not statistically significant. Again, there was a trend for shorter PFS in patients with high BRD4 protein expression although not statistically significant both at 12 months (p = 0.29) and at 24 months (p = 0.47). CONCLUSIONS There are contradictory data in the literature over the prognostic role of BRD4 gene expression in solid tumors. In our study, intermediate/high BRD4 gene expression was associated with a favorable prognosis in terms of overall survival and progression-free survival compared to low BRD4 gene expression.
Collapse
Affiliation(s)
- Angeliki Andrikopoulou
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.K.); (A.P.); (M.L.); (M.-A.D.); (F.Z.)
| | - Garyfalia Bletsa
- Research Center, Hellenic Anticancer Institute, 10680 Athens, Greece; (G.B.); (D.T.)
| | - Angeliki Rouvalis
- Obstetrics and Gynecology, 1st Obstetrics and Gynecology Clinic, National and Kapodistrian University of Athens, 10509 Athens, Greece; (A.R.); (D.H.)
| | - Dimitris Tsakogiannis
- Research Center, Hellenic Anticancer Institute, 10680 Athens, Greece; (G.B.); (D.T.)
| | - Maria Kaparelou
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.K.); (A.P.); (M.L.); (M.-A.D.); (F.Z.)
| | - Alkistis Papatheodoridi
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.K.); (A.P.); (M.L.); (M.-A.D.); (F.Z.)
| | - Dimitrios Haidopoulos
- Obstetrics and Gynecology, 1st Obstetrics and Gynecology Clinic, National and Kapodistrian University of Athens, 10509 Athens, Greece; (A.R.); (D.H.)
| | - Michalis Liontos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.K.); (A.P.); (M.L.); (M.-A.D.); (F.Z.)
| | - Meletios-Athanasios Dimopoulos
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.K.); (A.P.); (M.L.); (M.-A.D.); (F.Z.)
| | - Flora Zagouri
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece; (M.K.); (A.P.); (M.L.); (M.-A.D.); (F.Z.)
| |
Collapse
|
6
|
Weidle UH, Birzele F. Deregulated circRNAs in Epithelial Ovarian Cancer With Activity in Preclinical In Vivo Models: Identification of Targets and New Modalities for Therapeutic Intervention. Cancer Genomics Proteomics 2024; 21:213-237. [PMID: 38670587 PMCID: PMC11059596 DOI: 10.21873/cgp.20442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 04/28/2024] Open
Abstract
Epithelial ovarian cancer (EOC) is associated with a dismal prognosis due to development of resistance to chemotherapy and metastasis in the peritoneal cavity and distant organs. In order to identify new targets and treatment modalities we searched the literature for up- and and down-regulated circRNAs with efficacy in preclinical EOC-related in vivo systems. Our search yielded circRNAs falling into the following categories: cisplatin and paclitaxel resistance, transmembrane receptors, secreted factors, transcription factors, RNA splicing and processing factors, RAS pathway-related components, proteolysis and cell-cycle regulation, signaling-related proteins, and circRNAs regulating proteins in additional categories. These findings can be potentially translated by validation and manipulation of the corresponding targets, inhibition of circRNAs with antisense oligonucleotides (ASO), small interfering RNAs (siRNA) or small hairpin RNA (shRNA) or by reconstituting their activity.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany;
| | - Fabian Birzele
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, Basel, Switzerland
| |
Collapse
|
7
|
Kim Y, Park WH, Suh DH, Kim K, No JH, Kim YB. Anticancer Effects of BRD4 Inhibitor in Epithelial Ovarian Cancer. Cancers (Basel) 2024; 16:959. [PMID: 38473320 DOI: 10.3390/cancers16050959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 02/23/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Efforts have been made to develop bromodomain inhibitors as cancer treatments. Sub-pathways, particularly in ovarian cancer, affected by bromodomain-containing protein (BRD) remain unclear. This study verified the antitumor effects of a new drug that can overcome OPT-0139-chemoresistance to treat ovarian cancer. A mouse xenograft model of human ovarian cancer cells, SKOV3 and OVCAR3, was used in this study. Cell viability and proliferation were assessed using MTT and ATP assays. Cell cycle arrest and apoptosis were determined using flow cytometry. BRD4 and c-Myc expression and apoptosis-related molecules were detected using RT-PCR and real-time PCR and Western blot. We confirmed the OPT-0139 effect and mechanism of action in epithelial ovarian cancer. OPT-0139 significantly reduced cell viability and proliferation and induced apoptosis and cell cycle arrest. In the mouse xenograft model, significant changes in tumor growth, volume, weight, and BRD4-related gene expression were observed, suggesting the antitumor effects of BRD4 inhibitors. Combination therapy with cisplatin promoted apoptosis and suppressed tumor growth in vitro and in vivo. Our results suggest OPT-0139, a BRD4 inhibitor, as a promising anticancer drug for the treatment of ovarian cancer by inhibiting cell proliferation, decreasing cell viability, arresting cell cycle, and inducing apoptosis.
Collapse
Affiliation(s)
- Yeorae Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
| | - Wook-Ha Park
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
| | - Dong-Hoon Suh
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Jongno-gu, Seoul 03080, Republic of Korea
| | - Kidong Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Jongno-gu, Seoul 03080, Republic of Korea
| | - Jae-Hong No
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Jongno-gu, Seoul 03080, Republic of Korea
| | - Yong-Beom Kim
- Department of Obstetrics and Gynecology, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam 13620, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 103 Jongno-gu, Seoul 03080, Republic of Korea
| |
Collapse
|
8
|
Esplen HP, Yang RK, Kalia A, Tang Z, Tang G, Medeiros LJ, Toruner GA. Recurrent Somatic Copy Number Alterations and Their Association with Oncogene Expression Levels in High-Grade Ovarian Serous Carcinoma. Life (Basel) 2023; 13:2192. [PMID: 38004332 PMCID: PMC10672014 DOI: 10.3390/life13112192] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Somatic copy number alterations (SCNAs) are frequently observed in high-grade ovarian serous carcinoma (HGOSC). However, their impact on gene expression levels has not been systematically assessed. In this study, we explored the relationship between recurrent SCNA and gene expression using The Cancer Genome Atlas Pan Cancer dataset (OSC, TCGA, PanCancer Atlas) to identify cancer-related genes in HGOSC. We then investigated any association between highly correlated cancer genes and clinicopathological parameters, including age of diagnosis, disease stage, overall survival (OS), and progression-free survival (PFS). A total of 772 genes with recurrent SCNAs were observed. SCNA and mRNA expression levels were highly correlated for 274 genes; 24 genes were classified as a Tier 1 gene in the Cancer Gene Census in the Catalogue of Somatic Mutations in Cancer (CGC-COSMIC). Of these, 11 Tier 1 genes had highly correlated SCNA and mRNA expression levels: TBL1XR1, PIK3CA, UBR5, EIF3E, RAD21, EXT1, RECQL4, KRAS, PRKACA, BRD4, and TPM4. There was no association between gene amplification and disease stage or PFS. EIF3E, RAD21, and EXT1 were more frequently amplified in younger patients, specifically those under the age of 55 years. Patients with tumors carrying PRKACA, BRD4, or TPM4 amplification were associated with a significantly shorter OS. RECQL4 amplification was more frequent in younger patients, and tumors with this amplification were associated with a significantly better OS.
Collapse
Affiliation(s)
- Hillary P. Esplen
- Graduate Program in Diagnostic Genetics and Genomics, School of Health Professions, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | - Richard K. Yang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | - Awdhesh Kalia
- Graduate Program in Diagnostic Genetics and Genomics, School of Health Professions, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | - Zhenya Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Avenue, Houston, TX 77030-4009, USA
- Department of Pathology and Microbiology, College of Medicine, University of Nebraska Medical Center, Omaha, NE 68198-7815, USA
| | - Guilin Tang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Avenue, Houston, TX 77030-4009, USA
| | - L. Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Avenue, Houston, TX 77030-4009, USA
| | - Gokce A. Toruner
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, 6565 MD Anderson Avenue, Houston, TX 77030-4009, USA
| |
Collapse
|
9
|
Wang L, Chen X, Song L, Zou H. Machine Learning Developed a Programmed Cell Death Signature for Predicting Prognosis, Ecosystem, and Drug Sensitivity in Ovarian Cancer. Anal Cell Pathol (Amst) 2023; 2023:7365503. [PMID: 37868825 PMCID: PMC10586435 DOI: 10.1155/2023/7365503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 08/14/2023] [Accepted: 09/07/2023] [Indexed: 10/24/2023] Open
Abstract
Background Ovarian cancer (OC) is the leading cause of gynecological cancer death and the fifth most common cause of cancer-related death in women in America. Programmed cell death played a vital role in tumor progression and immunotherapy response in cancer. Methods The prognostic cell death signature (CDS) was constructed with an integrative machine learning procedure, including 10 methods, using TCGA, GSE14764, GSE26193, GSE26712, GSE63885, and GSE140082 datasets. Several methods and single-cell analysis were used to explore the correlation between CDS and the ecosystem and therapy response of OC patients. Results The prognostic CDS constructed by the combination of StepCox (n = both) + Enet (alpha = 0.2) acted as an independent risk factor for the overall survival (OS) of OC patients and showed stable and powerful performance in predicting the OS rate of OC patients. Compared with tumor grade, clinical stage, and many developed signatures, the CDS had a higher C-index. OC patients with low CDS score had a higher level of CD8+ cytotoxic T, B cell, and M1-like macrophage, representing a related immunoactivated ecosystem. A low CDS score indicated a higher PD1 and CTLA4 immunophenoscore, higher tumor mutation burden score, lower tumor immune dysfunction and exclusion score, and lower tumor escape score in OC, demonstrating a better immunotherapy response. OC patients with high CDS score had a higher gene set score of cancer-related hallmarks, including angiogenesis, epithelial-mesenchymal transition, hypoxia, glycolysis, and notch signaling. Conclusion The current study constructed a novel CDS for OC, which could serve as an indicator for predicting the prognosis, ecosystem, and immunotherapy benefits of OC patients.
Collapse
Affiliation(s)
- Le Wang
- Department of Blood Transfusion, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Xi Chen
- Department of Emergency, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Lei Song
- Department of General Practice, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Hua Zou
- Department of Organ Transplantation, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| |
Collapse
|
10
|
Hamilton EP, Wang JS, Oza AM, Patel MR, Ulahannan SV, Bauer T, Karlix JL, Zeron-Medina J, Fabbri G, Marco-Casanova P, Moorthy G, Hattersley MM, Littlewood GM, Mitchell P, Saeh J, Pouliot GP, Moore KN. First-in-human Study of AZD5153, A Small-molecule Inhibitor of Bromodomain Protein 4, in Patients with Relapsed/Refractory Malignant Solid Tumors and Lymphoma. Mol Cancer Ther 2023; 22:1154-1165. [PMID: 37486983 PMCID: PMC10544002 DOI: 10.1158/1535-7163.mct-23-0065] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/12/2023] [Accepted: 07/18/2023] [Indexed: 07/26/2023]
Abstract
AZD5153, a reversible, bivalent inhibitor of the bromodomain and extraterminal family protein BRD4, has preclinical activity in multiple tumors. This first-in-human, phase I study investigated AZD5153 alone or with olaparib in patients with relapsed/refractory solid tumors or lymphoma. Adults with relapsed tumors intolerant of, or refractory to, prior therapies received escalating doses of oral AZD5153 once daily or twice daily continuously (21-day cycles), or AZD5153 once daily/twice daily continuously or intermittently plus olaparib 300 mg twice daily, until disease progression or unacceptable toxicity. Between June 30, 2017 and April 19, 2021, 34 patients received monotherapy and 15 received combination therapy. Dose-limiting toxicities were thrombocytopenia/platelet count decreased (n = 4/n = 2) and diarrhea (n = 1). The recommended phase II doses (RP2D) were AZD5153 30 mg once daily or 15 mg twice daily (monotherapy) and 10 mg once daily (intermittent schedule) with olaparib. With AZD5153 monotherapy, common treatment-emergent adverse events (TEAE) included fatigue (38.2%), thrombocytopenia, and diarrhea (each 32.4%); common grade ≥ 3 TEAEs were thrombocytopenia (14.7%) and anemia (8.8%). With the combination, common TEAEs included nausea (66.7%) and fatigue (53.3%); the most common grade ≥ 3 TEAE was thrombocytopenia (26.7%). AZD5153 had dose-dependent pharmacokinetics, with minimal accumulation, and demonstrated dose-dependent modulation of peripheral biomarkers, including upregulation of HEXIM1. One patient with metastatic pancreatic cancer receiving combination treatment had a partial response lasting 4.2 months. These results show AZD5153 was tolerable as monotherapy and in combination at the RP2Ds; common toxicities were fatigue, hematologic AEs, and gastrointestinal AEs. Strong evidence of peripheral target engagement was observed.
Collapse
Affiliation(s)
- Erika P. Hamilton
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, Tennessee
| | - Judy S. Wang
- Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota, Florida
| | - Amit M. Oza
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre/University Health Network/Sinai Health Systems, Toronto, Ontario, Canada
| | - Manish R. Patel
- Florida Cancer Specialists/Sarah Cannon Research Institute, Sarasota, Florida
| | - Susanna V. Ulahannan
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, Tennessee
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Todd Bauer
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, Tennessee
| | | | | | | | | | - Ganesh Moorthy
- Clinical Pharmacology and Quantitative Pharmacology, R&D, AstraZeneca, Boston, Massachusetts
| | | | | | | | - Jamal Saeh
- Oncology R&D, AstraZeneca, Waltham, Massachusetts
| | | | - Kathleen N. Moore
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, Tennessee
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
11
|
Drumond-Bock AL, Wang L, Wang L, Cybula M, Rostworowska M, Kinter M, Bieniasz M. Increased expression of BRD4 isoforms long (BRD4-L) and short (BRD4-S) promotes chemotherapy resistance in high-grade serous ovarian carcinoma. Genes Cancer 2023; 14:56-76. [PMID: 37705995 PMCID: PMC10496930 DOI: 10.18632/genesandcancer.233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023] Open
Abstract
Chemoresistance in ovarian carcinoma is a puzzling issue that urges understanding of strategies used by cancer cells to survive DNA damage and to escape cell death. Expanding efforts to understand mechanisms driving chemoresistance and to develop alternative therapies targeting chemoresistant tumors are critical. Amplification of BRD4 is frequently associated with chemoresistant ovarian carcinoma, but little is known about the biological effects of the overexpression of BRD4 isoforms in this malignancy. Here, we described the consequences of BRD4-L and BRD4-S overexpression in ovarian carcinoma shedding a light on a complex regulation of BRD4 isoforms. We demonstrated that the BRD4-L transcript expression is required to generate both isoforms, BRD4-L and BRD4-S. We showed that the BRD4-S mRNA expression positively correlated with BRD4-S protein levels, while BRD4-L isoform showed negative correlation between mRNA and protein levels. Moreover, we demonstrated that an overexpression of BRD4 isoforms is associated with chemoresistance in ovarian cancer.
Collapse
Affiliation(s)
- Ana Luiza Drumond-Bock
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Luyao Wang
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Lin Wang
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | | | - Maria Rostworowska
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Michael Kinter
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| | - Magdalena Bieniasz
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA
| |
Collapse
|
12
|
Jeong S, Kim HR, Shin JH, Son MH, Lee IH, Roe JS. Streamlined DNA-encoded small molecule library screening and validation for the discovery of novel chemotypes targeting BET proteins. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 32:637-649. [PMID: 37207130 PMCID: PMC10189352 DOI: 10.1016/j.omtn.2023.04.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 04/20/2023] [Indexed: 05/21/2023]
Abstract
Targeting aberrant epigenetic programs that drive tumorigenesis is a promising approach to cancer therapy. DNA-encoded library (DEL) screening is a core platform technology increasingly used to identify drugs that bind to protein targets. Here, we use DEL screening against bromodomain and extra-terminal motif (BET) proteins to identify inhibitors with new chemotypes, and successfully identified BBC1115 as a selective BET inhibitor. While BBC1115 does not structurally resemble OTX-015, a clinically active pan-BET inhibitor, our intensive biological characterization revealed that BBC1115 binds to BET proteins, including BRD4, and suppresses aberrant cell fate programs. Phenotypically, BBC1115-mediated BET inhibition impaired proliferation in acute myeloid leukemia, pancreatic, colorectal, and ovarian cancer cells in vitro. Moreover, intravenous administration of BBC1115 inhibited subcutaneous tumor xenograft growth with minimal toxicity and favorable pharmacokinetic properties in vivo. Since epigenetic regulations are ubiquitously distributed across normal and malignant cells, it will be critical to evaluate if BBC1115 affects normal cell function. Nonetheless, our study shows integrating DEL-based small-molecule compound screening and multi-step biological validation represents a reliable strategy to discover new chemotypes with selectivity, efficacy, and safety profiles for targeting proteins involved in epigenetic regulation in human malignancies.
Collapse
Affiliation(s)
- Seoyeon Jeong
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Hwa-Ryeon Kim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - June-Ha Shin
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | | | | | - Jae-Seok Roe
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
- Corresponding author: Jae-Seok Roe, PhD, Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea.
| |
Collapse
|
13
|
Quintela M, James DW, Pociute A, Powell L, Edwards K, Coombes Z, Garcia J, Garton N, Das N, Lutchman-Singh K, Margarit L, Beynon AL, Rioja I, Prinjha RK, Harker NR, Gonzalez D, Conlan RS, Francis LW. Bromodomain inhibitor i-BET858 triggers a unique transcriptional response coupled to enhanced DNA damage, cell cycle arrest and apoptosis in high-grade ovarian carcinoma cells. Clin Epigenetics 2023; 15:63. [PMID: 37060086 PMCID: PMC10105475 DOI: 10.1186/s13148-023-01477-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 03/29/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND Ovarian cancer has a specific unmet clinical need, with a persistently poor 5-year survival rate observed in women with advanced stage disease warranting continued efforts to develop new treatment options. The amplification of BRD4 in a significant subset of high-grade serous ovarian carcinomas (HGSC) has led to the development of BET inhibitors (BETi) as promising antitumour agents that have subsequently been evaluated in phase I/II clinical trials. Here, we describe the molecular effects and ex vivo preclinical activities of i-BET858, a bivalent pan-BET inhibitor with proven in vivo BRD inhibitory activity. RESULTS i-BET858 demonstrates enhanced cytotoxic activity compared with earlier generation BETis both in cell lines and primary cells derived from clinical samples of HGSC. At molecular level, i-BET858 triggered a bipartite transcriptional response, comprised of a 'core' network of genes commonly associated with BET inhibition in solid tumours, together with a unique i-BET858 gene signature. Mechanistically, i-BET858 elicited enhanced DNA damage, cell cycle arrest and apoptotic cell death compared to its predecessor i-BET151. CONCLUSIONS Overall, our ex vivo and in vitro studies indicate that i-BET858 represents an optimal candidate to pursue further clinical validation for the treatment of HGSC.
Collapse
Affiliation(s)
- Marcos Quintela
- Swansea University Medical School, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - David W James
- Swansea University Medical School, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Agne Pociute
- Swansea University Medical School, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Lydia Powell
- Swansea University Medical School, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Kadie Edwards
- Swansea University Medical School, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Zoe Coombes
- Swansea University Medical School, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Jetzabel Garcia
- Swansea University Medical School, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Neil Garton
- Immunology Research Unit, GlaxoSmithKline, Medicines Research Centre, Stevenage, SG1 2NY, UK
| | - Nagindra Das
- Swansea Bay University Health Board, Swansea, SA12 7BR, UK
| | | | - Lavinia Margarit
- Swansea University Medical School, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
- Cwm Taf Morgannwg University Health Board, Swansea, SA2 8QA, UK
| | | | - Inmaculada Rioja
- Immunology Research Unit, GlaxoSmithKline, Medicines Research Centre, Stevenage, SG1 2NY, UK
| | - Rab K Prinjha
- Immunology Research Unit, GlaxoSmithKline, Medicines Research Centre, Stevenage, SG1 2NY, UK
| | - Nicola R Harker
- Immunology Research Unit, GlaxoSmithKline, Medicines Research Centre, Stevenage, SG1 2NY, UK
| | - Deyarina Gonzalez
- Swansea University Medical School, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - R Steven Conlan
- Swansea University Medical School, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - Lewis W Francis
- Swansea University Medical School, Swansea University, Singleton Park, Swansea, SA2 8PP, UK.
| |
Collapse
|
14
|
Dickmann CGF, McDonald AF, Huynh N, Rigopoulos A, Liu Z, Guo N, Osellame LD, Gorman MA, Parker MW, Gan HK, Scott AM, Ackermann U, Burvenich IJG, White JM. Bromodomain and extraterminal protein-targeted probe enables tumour visualisation in vivo using positron emission tomography. Chem Commun (Camb) 2023; 59:3126-3129. [PMID: 36809538 DOI: 10.1039/d2cc04813b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Bromodomain and extraterminal (BET) proteins, a family of epigenetic regulators, have emerged as important oncology drug targets. BET proteins have not been targeted for molecular imaging of cancer. Here, we report the development of a novel molecule radiolabelled with positron emitting fluorine-18, [18F]BiPET-2, and its in vitro and preclinical evaluation in glioblastoma models.
Collapse
Affiliation(s)
- Catherine G Fitzgerald Dickmann
- Bio21 Institute, The University of Melbourne, Parkville, VIC 3010, Australia. .,Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, and School of Cancer Medicine, La Trobe University, Heidelberg, Australia. .,Department of Molecular Imaging and Therapy, Austin Hospital, Heidelberg, Australia
| | - Alexander F McDonald
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, and School of Cancer Medicine, La Trobe University, Heidelberg, Australia. .,Department of Molecular Imaging and Therapy, Austin Hospital, Heidelberg, Australia
| | - Nhi Huynh
- Department of Molecular Imaging and Therapy, Austin Hospital, Heidelberg, Australia
| | - Angela Rigopoulos
- Department of Molecular Imaging and Therapy, Austin Hospital, Heidelberg, Australia
| | - Zhanqi Liu
- Department of Molecular Imaging and Therapy, Austin Hospital, Heidelberg, Australia
| | - Nancy Guo
- Department of Molecular Imaging and Therapy, Austin Hospital, Heidelberg, Australia
| | - Laura D Osellame
- Department of Molecular Imaging and Therapy, Austin Hospital, Heidelberg, Australia
| | - Michael A Gorman
- Bio21 Institute, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Michael W Parker
- Bio21 Institute, The University of Melbourne, Parkville, VIC 3010, Australia. .,ACRF Facility for Innovative Cancer Drug Discovery, Bio21 Institute, The University of Melbourne, Parkville, VIC 3010, Australia.,ACRF Rational Drug Discovery Centre, St. Vincent's Institute of Medical Research, VIC 3065, Australia
| | - Hui K Gan
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, and School of Cancer Medicine, La Trobe University, Heidelberg, Australia.
| | - Andrew M Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, and School of Cancer Medicine, La Trobe University, Heidelberg, Australia. .,Department of Molecular Imaging and Therapy, Austin Hospital, Heidelberg, Australia.,Faculty of Medicine, University of Melbourne, Parkville, Australia
| | - Uwe Ackermann
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, and School of Cancer Medicine, La Trobe University, Heidelberg, Australia. .,Department of Molecular Imaging and Therapy, Austin Hospital, Heidelberg, Australia
| | - Ingrid J G Burvenich
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, and School of Cancer Medicine, La Trobe University, Heidelberg, Australia.
| | - Jonathan M White
- Bio21 Institute, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
15
|
Design, synthesis and anti-ovarian cancer activities of thieno[2,3-d]pyrimidine based chimeric BRD4 inhibitor/nitric oxide-donator. Eur J Med Chem 2023; 246:114970. [PMID: 36470106 DOI: 10.1016/j.ejmech.2022.114970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/22/2022] [Accepted: 11/23/2022] [Indexed: 12/03/2022]
Abstract
Bromodomain protein 4 (BRD4) is an attractive epigenetic target that regulating diverse cellular processes, and the discovery of dual-target inhibitors including BRD4 is an effective approach in cancer treatment to increase potency and reduce drug resistance. Based on the multifunctional drug development strategy, a series of new derivatives of nitrooxy (ONO2) or furoxan (1,2,5-oxadiazole 2-oxide) with BRD4 inhibitor capable of inhibiting BRD4 and simultaneously releasing NO were designed and synthesized. When NO concentrations were measured with Griess reagent under physiological conditions, all compounds released NO at micromolar levels, reaching effective antitumor concentrations. Biological studies showed that the most potent BRD4/NO hybrid 11a exhibited good BRD4 inhibitory activity and selectivity. Further mechanistic studies revealed that 11a significantly decreased the expression of BRD4 and c-Myc, as well as induced cellular apoptosis and autophagic cell death both in vitro and in vivo. In summary, we optimized the chimeric BRD4-inhibitor/NO-donor based on our previous studies, and it should be a lead compound for targeted therapy of OC (ovarian cancer) in the future. This interesting strategy could expand the usage of BRDi in human malignancies and endogenous gastro-transmitters.
Collapse
|
16
|
Shen H, Hu X, Yang X, Chen J, Fu Y, He H, Shi Y, Zeng R, Chang W, Zheng S. Inhibition of BRD4 enhanced the tumor suppression effect of dasatinib in gastric cancer. Med Oncol 2023; 40:9. [PMID: 36352160 PMCID: PMC9646567 DOI: 10.1007/s12032-022-01831-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 08/20/2022] [Indexed: 11/10/2022]
Abstract
BRD4, a member of the bromodomain and extraterminal (BET) family, is elevated in multiple cancer tissues, including gastric cancer (GC). Targeted therapy with BRD4 may help improve the overall survival of patients with GC. Meanwhile, the approved multi-target kinase inhibitor, dasatinib, was recently reported to show varied tumor-suppressive effects in GC cells. This study investigated BRD4 expression in vivo and in vitro using immunohistochemistry and western blotting, respectively. We discussed the relationship between BRD4 expression and patient prognosis. Next, the antitumor efficacy of dasatinib was measured in BRD4-knockdown GC cells to determine the role of BRD4 blockage in dasatinib treatment. Finally, molibresib, a BET inhibitor, was used to measure the cooperative function of BRD4 inhibition and dasatinib treatment in three GC cell lines. Epithelial BRD4 expression was higher in tumoral and metastatic tissues and was strongly associated with unfavorable tumor, node, and metastasis stages and survival. BRD4 expression was heterogeneous in the three GC cell lines tested in vitro. In SGC7901, a BRD4-high GC cell line, knockdown of BRD4 using specific siRNAs suppressed cell growth individually and cooperatively with dasatinib. Moreover, molibresib and dasatinib showed a cooperative effect in suppressing the proliferation of BRD4-high GC cells. In conclusion, we confirmed that increased epithelial BRD4 expression is associated with poor disease stage and prognosis in GC and BRD4 blockage might be a valuable strategy to improve the sensitivity of dasatinib and other drugs in the chemotherapy of advanced GC.
Collapse
Affiliation(s)
- Hao Shen
- Department of Navy Environmental and Occupational Health, Faculty of Naval Medicine, Navy Military Medical University, Shanghai, People's Republic of China
- School of Medicine, Yunnan University, Kunming, Yunnan, People's Republic of China
| | - Xuefei Hu
- Department of Navy Environmental and Occupational Health, Faculty of Naval Medicine, Navy Military Medical University, Shanghai, People's Republic of China
| | - Xinrui Yang
- School of Medicine, Yunnan University, Kunming, Yunnan, People's Republic of China
| | - Jiahui Chen
- Department of Navy Environmental and Occupational Health, Faculty of Naval Medicine, Navy Military Medical University, Shanghai, People's Republic of China
| | - Yating Fu
- Department of Navy Environmental and Occupational Health, Faculty of Naval Medicine, Navy Military Medical University, Shanghai, People's Republic of China
| | - Hongwei He
- School of Medicine, Yunnan University, Kunming, Yunnan, People's Republic of China
| | - Yongkang Shi
- School of Medicine, Yunnan University, Kunming, Yunnan, People's Republic of China
| | - Rong Zeng
- Department of Medical Oncology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, People's Republic of China.
| | - Wenjun Chang
- Department of Navy Environmental and Occupational Health, Faculty of Naval Medicine, Navy Military Medical University, Shanghai, People's Republic of China.
| | - Shangyong Zheng
- School of Medicine, Yunnan University, Kunming, Yunnan, People's Republic of China.
| |
Collapse
|
17
|
Yan G, Luna A, Wang H, Bozorgui B, Li X, Sanchez M, Dereli Z, Kahraman N, Kara G, Chen X, Zheng C, McGrail D, Sahni N, Lu Y, Babur O, Cokol M, Lim B, Ozpolat B, Sander C, Mills GB, Korkut A. BET inhibition induces vulnerability to MCL1 targeting through upregulation of fatty acid synthesis pathway in breast cancer. Cell Rep 2022; 40:111304. [PMID: 36103824 PMCID: PMC9523722 DOI: 10.1016/j.celrep.2022.111304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 05/06/2022] [Accepted: 08/10/2022] [Indexed: 11/12/2022] Open
Abstract
Therapeutic options for treatment of basal-like breast cancers remain limited. Here, we demonstrate that bromodomain and extra-terminal (BET) inhibition induces an adaptive response leading to MCL1 protein-driven evasion of apoptosis in breast cancer cells. Consequently, co-targeting MCL1 and BET is highly synergistic in breast cancer models. The mechanism of adaptive response to BET inhibition involves the upregulation of lipid synthesis enzymes including the rate-limiting stearoyl-coenzyme A (CoA) desaturase. Changes in lipid synthesis pathway are associated with increases in cell motility and membrane fluidity as well as re-localization and activation of HER2/EGFR. In turn, the HER2/EGFR signaling results in the accumulation of and vulnerability to the inhibition of MCL1. Drug response and genomics analyses reveal that MCL1 copy-number alterations are associated with effective BET and MCL1 co-targeting. The high frequency of MCL1 chromosomal amplifications (>30%) in basal-like breast cancers suggests that BET and MCL1 co-targeting may have therapeutic utility in this aggressive subtype of breast cancer.
Collapse
Affiliation(s)
- Gonghong Yan
- Department of Bioinformatics and Computational Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Augustin Luna
- cBio Center, Department of Data Sciences, Dana Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Heping Wang
- Department of Bioinformatics and Computational Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Behnaz Bozorgui
- Department of Bioinformatics and Computational Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xubin Li
- Department of Bioinformatics and Computational Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Maga Sanchez
- Department of Bioinformatics and Computational Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zeynep Dereli
- Department of Bioinformatics and Computational Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nermin Kahraman
- Department of Experimental Therapeutics, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Goknur Kara
- Department of Experimental Therapeutics, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaohua Chen
- Department of Bioinformatics and Computational Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Caishang Zheng
- Department of Bioinformatics and Computational Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Daniel McGrail
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nidhi Sahni
- Department of Bioinformatics and Computational Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Epigenetics and Molecular Carcinogenesis, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yiling Lu
- Department of Genomic Medicine, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ozgun Babur
- Computer Science, College of Science and Mathematics, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Murat Cokol
- Axcella Therapeutics, Cambridge, MA 02139, USA
| | - Bora Lim
- Breast Cancer Research Program, Dan L Duncan Comprehensive Cancer Center, Houston, TX 77030, USA
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, UT MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chris Sander
- cBio Center, Department of Data Sciences, Dana Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Gordon B Mills
- Department of Cell, Development and Cancer Biology, Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, USA
| | - Anil Korkut
- Department of Bioinformatics and Computational Biology, UT MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
18
|
van der Noord VE, van de Water B, Le Dévédec SE. Targeting the Heterogeneous Genomic Landscape in Triple-Negative Breast Cancer through Inhibitors of the Transcriptional Machinery. Cancers (Basel) 2022; 14:4353. [PMID: 36139513 PMCID: PMC9496798 DOI: 10.3390/cancers14184353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 08/28/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer defined by lack of the estrogen, progesterone and human epidermal growth factor receptor 2. Although TNBC tumors contain a wide variety of oncogenic mutations and copy number alterations, the direct targeting of these alterations has failed to substantially improve therapeutic efficacy. This efficacy is strongly limited by interpatient and intratumor heterogeneity, and thereby a lack in uniformity of targetable drivers. Most of these genetic abnormalities eventually drive specific transcriptional programs, which may be a general underlying vulnerability. Currently, there are multiple selective inhibitors, which target the transcriptional machinery through transcriptional cyclin-dependent kinases (CDKs) 7, 8, 9, 12 and 13 and bromodomain extra-terminal motif (BET) proteins, including BRD4. In this review, we discuss how inhibitors of the transcriptional machinery can effectively target genetic abnormalities in TNBC, and how these abnormalities can influence sensitivity to these inhibitors. These inhibitors target the genomic landscape in TNBC by specifically suppressing MYC-driven transcription, inducing further DNA damage, improving anti-cancer immunity, and preventing drug resistance against MAPK and PI3K-targeted therapies. Because the transcriptional machinery enables transcription and propagation of multiple cancer drivers, it may be a promising target for (combination) treatment, especially of heterogeneous malignancies, including TNBC.
Collapse
Affiliation(s)
| | | | - Sylvia E. Le Dévédec
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, 2333 CC Leiden, The Netherlands
| |
Collapse
|
19
|
Guo X, Geng X, Chu Y, Gao J, Jiang L. MiR-204-5p Alleviates Neuropathic Pain by Targeting BRD4 in a Rat Chronic Constrictive Injury Model. J Pain Res 2022; 15:2427-2435. [PMID: 36003288 PMCID: PMC9394659 DOI: 10.2147/jpr.s371616] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 08/08/2022] [Indexed: 12/05/2022] Open
Abstract
Purpose The pathogenesis of neuropathic pain is complex, and previous studies have found that microRNAs are important regulators of neuropathic pain and are associated with the progression of neuropathic pain. This study aims to explore the level and role of miR-204-5p in the chronic constrictive injury (CCI) model of rats. Patients and Methods The CCI rat model was constructed to evaluate paw withdrawal threshold (PWT), paw withdrawal latency (PWL), the expressions of miR-204-5p, and the contents of inflammatory factors in the model. Overexpression of miR-204-5p in rat spinal cord was induced by intrathecal injection of miR-204-5p mimics. PWT and PWL were used to estimate mechanical and thermal pain thresholds. IL-6 and TNF-α were determined by ELISA. Luciferase reporter gene was conducted to verify the targeting relationship between miR-204-5p and BRD4. Results miR-204-5p was abnormally down-regulated in the CCI group. The thresholds of mechanical and thermal pain stimulation in the CCI group were lower, and the levels of inflammatory factors were higher than those in the sham group. Overexpression of miR-204-5p alleviated PWT, PWL and inflammatory factors. Besides, the luciferase reporter gene showed that BRD4 was a target gene of miR-204-5p. Conclusion These results suggested that miR-204-5p may alleviate neuropathic pain and inflammation through targeted regulation of BRD4 expression.
Collapse
Affiliation(s)
- Xiaona Guo
- Pain Department, Dongying People's Hospital, Dongying, Shandong, People's Republic of China
| | - Xia Geng
- Pain Department, Dongying People's Hospital, Dongying, Shandong, People's Republic of China
| | - Yunchao Chu
- Pain Department, Shengli Oilfield Central Hospital, Dongying, Shandong, People's Republic of China
| | - Jianfei Gao
- Pain Department, Dongying People's Hospital, Dongying, Shandong, People's Republic of China
| | - Linkai Jiang
- Pain Department, Dongying People's Hospital, Dongying, Shandong, People's Republic of China
| |
Collapse
|
20
|
Kelly MR, Wisniewska K, Regner MJ, Lewis MW, Perreault AA, Davis ES, Phanstiel DH, Parker JS, Franco HL. A multi-omic dissection of super-enhancer driven oncogenic gene expression programs in ovarian cancer. Nat Commun 2022; 13:4247. [PMID: 35869079 PMCID: PMC9307778 DOI: 10.1038/s41467-022-31919-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/08/2022] [Indexed: 01/14/2023] Open
Abstract
The human genome contains regulatory elements, such as enhancers, that are often rewired by cancer cells for the activation of genes that promote tumorigenesis and resistance to therapy. This is especially true for cancers that have little or no known driver mutations within protein coding genes, such as ovarian cancer. Herein, we utilize an integrated set of genomic and epigenomic datasets to identify clinically relevant super-enhancers that are preferentially amplified in ovarian cancer patients. We systematically probe the top 86 super-enhancers, using CRISPR-interference and CRISPR-deletion assays coupled to RNA-sequencing, to nominate two salient super-enhancers that drive proliferation and migration of cancer cells. Utilizing Hi-C, we construct chromatin interaction maps that enable the annotation of direct target genes for these super-enhancers and confirm their activity specifically within the cancer cell compartment of human tumors using single-cell genomics data. Together, our multi-omic approach examines a number of fundamental questions about how regulatory information encoded into super-enhancers drives gene expression networks that underlie the biology of ovarian cancer.
Collapse
Affiliation(s)
- Michael R Kelly
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Bioinformatics and Computational Biology Graduate Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Kamila Wisniewska
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Matthew J Regner
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Bioinformatics and Computational Biology Graduate Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Michael W Lewis
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Andrea A Perreault
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Eric S Davis
- Bioinformatics and Computational Biology Graduate Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Douglas H Phanstiel
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Thurston Arthritis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Cell Biology & Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Joel S Parker
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Bioinformatics and Computational Biology Graduate Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Hector L Franco
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Bioinformatics and Computational Biology Graduate Program, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
21
|
Wang Y, Wu Y, Jiang J, Zhang Y, Fu Y, Zheng M, Tao X, Yi J, Mu D, Cao X. The prognostic significance of bromodomain protein 4 expression in solid tumor patients: A meta-analysis. Pathol Res Pract 2022; 234:153918. [PMID: 35561521 DOI: 10.1016/j.prp.2022.153918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 04/15/2022] [Accepted: 04/24/2022] [Indexed: 11/19/2022]
Abstract
BACKGROUND Cancer is a leading cause of death worldwide. At present, several inhibitors of bromodomain protein 4 have shown promising anti-tumor responses in clinical trials. Numerous studies have reported the value of bromodomain protein 4 expression in predicting the prognosis of patients with cancers, but their conclusions remain controversial. Therefore, we conducted a meta-analysis to explore the association between bromodomain protein 4 and patient prognosis with the aim to provide new directions for the development of strategies for targeted cancer therapy. METHODS The meta-analysis was registered in the International Prospective Register of Systematic Reviews (https://www.crd.york.ac.uk/prospero/; Registration No. CRD42020184948) and followed the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) statement. PubMed Central, PubMed, Cochrane Library and Embase were thoroughly searched to identify eligible studies published through March 31, 2021. Odds ratios with 95% confidence intervals were calculated to demonstrate the relationship between bromodomain protein 4 expression and clinicopathological features. We computed pooled estimated hazard ratios with 95% confidence intervals using Stata 12.0 software to clarify the relationship between bromodomain protein 4 expression and overall survival of various cancers. A quality assessment of the eligible articles was performed based on the Newcastle-Ottawa scale. RESULTS A total of 974 patients from 10 studies were enrolled in the meta-analysis. Our results revealed that compared to low bromodomain protein 4 expression, high bromodomain protein 4 expression in cancer tissues was significantly associated with lymph node metastasis (Odds ratio = 3.59, 95% confidence interval: 2.62-4.91), distant metastasis (Odds ratio = 4.22, 95% confidence interval: 2.40-7.45), advanced TNM stage (III+IV vs. I+II: Odds ratio = 3.23, 95% confidence interval: 1.29-8.08), and poorly differentiated tumors (Odds ratio = 1.87, 95% confidence interval: 1.33-2.63). In addition, an elevated expression of bromodomain protein 4 tended to shorten survival time (Hazard ratio = 2.23, 95% confidence interval: 1.62-3.07). The subgroup analysis results showed that bromodomain protein 4 upregulation was related to poor prognosis in patients with digestive system cancers (Hazard ratio = 2.54, 95% confidence interval: 1.85-3.50). CONCLUSION This meta-analysis indicated that bromodomain protein 4 may serve as a promising prognostic biomarker for cancers and a direct effective cancer treatment target.
Collapse
Affiliation(s)
- Yueqi Wang
- Division of Clinical Research, First Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Yanhua Wu
- Division of Clinical Research, First Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Jing Jiang
- Division of Clinical Research, First Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Yangyu Zhang
- Division of Clinical Research, First Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Yingli Fu
- Division of Clinical Research, First Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Min Zheng
- Division of Clinical Research, First Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Xuerong Tao
- Division of Clinical Research, First Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Jiaxin Yi
- Division of Clinical Research, First Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Dongmei Mu
- Division of Clinical Research, First Hospital of Jilin University, Changchun, Jilin Province, China.
| | - Xueyuan Cao
- Department of Gastrointestinal Surgery, First Hospital of Jilin University, Changchun, Jilin Province, China.
| |
Collapse
|
22
|
Lewis MW, Wisniewska K, King CM, Li S, Coffey A, Kelly MR, Regner MJ, Franco HL. Enhancer RNA Transcription Is Essential for a Novel CSF1 Enhancer in Triple-Negative Breast Cancer. Cancers (Basel) 2022; 14:1852. [PMID: 35406623 PMCID: PMC8997997 DOI: 10.3390/cancers14071852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/24/2022] [Accepted: 03/29/2022] [Indexed: 12/11/2022] Open
Abstract
Enhancers are critical regulatory elements in the genome that help orchestrate spatiotemporal patterns of gene expression during development and normal physiology. In cancer, enhancers are often rewired by various genetic and epigenetic mechanisms for the activation of oncogenes that lead to initiation and progression. A key feature of active enhancers is the production of non-coding RNA molecules called enhancer RNAs, whose functions remain unknown but can be used to specify active enhancers de novo. Using a combination of eRNA transcription and chromatin modifications, we have identified a novel enhancer located 30 kb upstream of Colony Stimulating Factor 1 (CSF1). Notably, CSF1 is implicated in the progression of breast cancer, is overexpressed in triple-negative breast cancer (TNBC) cell lines, and its enhancer is primarily active in TNBC patient tumors. Genomic deletion of the enhancer (via CRISPR/Cas9) enabled us to validate this regulatory element as a bona fide enhancer of CSF1 and subsequent cell-based assays revealed profound effects on cancer cell proliferation, colony formation, and migration. Epigenetic silencing of the enhancer via CRISPR-interference assays (dCas9-KRAB) coupled to RNA-sequencing, enabled unbiased identification of additional target genes, such as RSAD2, that are predictive of clinical outcome. Additionally, we repurposed the RNA-guided RNA-targeting CRISPR-Cas13 machinery to specifically degrade the eRNAs transcripts produced at this enhancer to determine the consequences on CSF1 mRNA expression, suggesting a post-transcriptional role for these non-coding transcripts. Finally, we test our eRNA-dependent model of CSF1 enhancer function and demonstrate that our results are extensible to other forms of cancer. Collectively, this work describes a novel enhancer that is active in the TNBC subtype, which is associated with cellular growth, and requires eRNA transcripts for proper enhancer function. These results demonstrate the significant impact of enhancers in cancer biology and highlight their potential as tractable targets for therapeutic intervention.
Collapse
Affiliation(s)
- Michael W. Lewis
- The Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.W.L.); (K.W.); (C.M.K.); (S.L.); (A.C.); (M.R.K.); (M.J.R.)
| | - Kamila Wisniewska
- The Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.W.L.); (K.W.); (C.M.K.); (S.L.); (A.C.); (M.R.K.); (M.J.R.)
| | - Caitlin M. King
- The Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.W.L.); (K.W.); (C.M.K.); (S.L.); (A.C.); (M.R.K.); (M.J.R.)
| | - Shen Li
- The Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.W.L.); (K.W.); (C.M.K.); (S.L.); (A.C.); (M.R.K.); (M.J.R.)
| | - Alisha Coffey
- The Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.W.L.); (K.W.); (C.M.K.); (S.L.); (A.C.); (M.R.K.); (M.J.R.)
| | - Michael R. Kelly
- The Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.W.L.); (K.W.); (C.M.K.); (S.L.); (A.C.); (M.R.K.); (M.J.R.)
- Bioinformatics and Computational Biology Graduate Program, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Matthew J. Regner
- The Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.W.L.); (K.W.); (C.M.K.); (S.L.); (A.C.); (M.R.K.); (M.J.R.)
- Bioinformatics and Computational Biology Graduate Program, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hector L. Franco
- The Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (M.W.L.); (K.W.); (C.M.K.); (S.L.); (A.C.); (M.R.K.); (M.J.R.)
- Bioinformatics and Computational Biology Graduate Program, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- The Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
23
|
Lin CH, Kuo JCT, Li D, Koenig AB, Pan A, Yan P, Bai XF, Lee RJ, Ghoshal K. AZD5153, a Bivalent BRD4 Inhibitor, Suppresses Hepatocarcinogenesis by Altering BRD4 Chromosomal Landscape and Modulating the Transcriptome of HCC Cells. Front Cell Dev Biol 2022; 10:853652. [PMID: 35399501 PMCID: PMC8987780 DOI: 10.3389/fcell.2022.853652] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/09/2022] [Indexed: 12/13/2022] Open
Abstract
BRD4, a chromatin modifier frequently upregulated in a variety of neoplasms including hepatocellular cancer (HCC), promotes cancer cell growth by activating oncogenes through its interaction with acetylated histone tails of nucleosomes. Here, we determined the anti-HCC efficacy of AZD5153, a potent bivalent BRD4 inhibitor, and elucidated its underlying molecular mechanism of action. AZD5153 treatment inhibited HCC cell proliferation, clonogenic survival and induced apoptosis in HCC cells. In vivo, AZD5153-formulated lipid nanoemulsions inhibited both orthotopic and subcutaneous HCCLM3 xenograft growth in NSG mice. Mapping of BRD4- chromosomal targets by ChIP-seq analysis identified the occupancy of BRD4 with the promoters, gene bodies, and super-enhancers of both mRNA and noncoding RNA genes, which were disrupted upon AZD5153 treatment. RNA-seq analysis of polyadenylated RNAs showed several BRD4 target genes involved in DNA replication, cell proliferation, and anti-apoptosis were repressed in AZD5153-treated HCC cells. In addition to known tumor-promoting genes, e.g., c-MYC, YAP1, RAD51B, TRIB3, SLC17A9, JADE1, we found that NAPRT, encoding a key enzyme for NAD+ biosynthesis from nicotinic acid, was also suppressed in HCC cells by the BRD4 inhibitor. Interestingly, AZD5153 treatment upregulated NAMPT, whose product is the rate-limiting enzyme for NAD+ synthesis from nicotinamide. This may explain why AZD5153 acted in concert with FK866, a potent NAMPT inhibitor, in reducing HCC cell proliferation and clonogenic survival. In conclusion, our results identified novel targets of BRD4 in the HCCLM3 cell genome and demonstrated anti-HCC efficacy of AZD5153, which was potentiated in combination with an NAMPT inhibitor.
Collapse
Affiliation(s)
- Cho-Hao Lin
- Department of Pathology, College of Medicine, Columbus, OH, United States
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Jimmy Chun-Tien Kuo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, Columbus, OH, United States
| | - Ding Li
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, Columbus, OH, United States
| | - Aaron B. Koenig
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Alexander Pan
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Pearlly Yan
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Xue-Feng Bai
- Department of Pathology, College of Medicine, Columbus, OH, United States
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Robert J. Lee
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, Columbus, OH, United States
- *Correspondence: Robert J. Lee, ; Kalpana Ghoshal,
| | - Kalpana Ghoshal
- Department of Pathology, College of Medicine, Columbus, OH, United States
- Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
- *Correspondence: Robert J. Lee, ; Kalpana Ghoshal,
| |
Collapse
|
24
|
Vieito M, Simonelli M, de Vos F, Moreno V, Geurts M, Lorenzi E, Macchini M, van den Bent MJ, Del Conte G, de Jonge M, Martín-Soberón MC, Amoroso B, Sanchez-Perez T, Zuraek M, Hanna B, Aronchik I, Filvaroff E, Chang H, Mendez C, Arias Parro M, Wei X, Nikolova Z, Sepulveda JM. Trotabresib (CC-90010) in combination with adjuvant temozolomide or concomitant temozolomide plus radiotherapy in patients with newly diagnosed glioblastoma. Neurooncol Adv 2022; 4:vdac146. [PMID: 36382109 PMCID: PMC9653173 DOI: 10.1093/noajnl/vdac146] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Background Standard-of-care treatment for newly diagnosed glioblastoma (ndGBM), consisting of surgery followed by radiotherapy (RT) and temozolomide (TMZ), has improved outcomes compared with RT alone; however, prognosis remains poor. Trotabresib, a novel bromodomain and extraterminal inhibitor, has demonstrated antitumor activity in patients with high-grade gliomas. Methods In this phase Ib, dose-escalation study (NCT04324840), we investigated trotabresib 15, 30, and 45 mg combined with TMZ in the adjuvant setting and trotabresib 15 and 30 mg combined with TMZ+RT in the concomitant setting in patients with ndGBM. Primary endpoints were to determine safety, tolerability, maximum tolerated dose, and/or recommended phase II dose (RP2D) of trotabresib. Secondary endpoints were assessment of preliminary efficacy and pharmacokinetics. Pharmacodynamics were investigated as an exploratory endpoint. Results The adjuvant and concomitant cohorts enrolled 18 and 14 patients, respectively. Trotabresib in combination with TMZ or TMZ+RT was well tolerated; most treatment-related adverse events were mild or moderate. Trotabresib pharmacokinetics and pharmacodynamics in both settings were consistent with previous data for trotabresib monotherapy. The RP2D of trotabresib was selected as 30 mg 4 days on/24 days off in both settings. At last follow-up, 5 (28%) and 6 (43%) patients remain on treatment in the adjuvant and concomitant settings, respectively, with 1 patient in the adjuvant cohort achieving complete response. Conclusions Trotabresib combined with TMZ in the adjuvant setting and with TMZ+RT in the concomitant setting was safe and well tolerated in patients with ndGBM, with encouraging treatment durations. Trotabresib 30 mg was established as the RP2D in both settings.
Collapse
Affiliation(s)
- Maria Vieito
- Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Universidad Autonoma de Barcelona, Barcelona, Spain
| | - Matteo Simonelli
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- IRCCS Humanitas Research Hospital, Milan, Italy
| | - Filip de Vos
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Victor Moreno
- START Madrid-FJD, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | | | | | - Marina Macchini
- Department of Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Gianluca Del Conte
- Department of Oncology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maja de Jonge
- Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Maria Cruz Martín-Soberón
- Neuro-Oncology Unit, Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Barbara Amoroso
- Centre for Innovation and Translational Research Europe, A Bristol Myers Squibb Company, Seville, Spain
| | - Tania Sanchez-Perez
- Centre for Innovation and Translational Research Europe, A Bristol Myers Squibb Company, Seville, Spain
| | | | | | | | | | - Henry Chang
- Bristol Myers Squibb, San Francisco, CA, USA
| | - Cristina Mendez
- Centre for Innovation and Translational Research Europe, A Bristol Myers Squibb Company, Seville, Spain
| | | | - Xin Wei
- Bristol Myers Squibb, Princeton, NJ, USA
| | - Zariana Nikolova
- Centre for Innovation and Translational Research Europe, A Bristol Myers Squibb Company, Seville, Spain
| | | |
Collapse
|
25
|
Drumond-Bock AL, Bieniasz M. The role of distinct BRD4 isoforms and their contribution to high-grade serous ovarian carcinoma pathogenesis. Mol Cancer 2021; 20:145. [PMID: 34758842 PMCID: PMC8579545 DOI: 10.1186/s12943-021-01424-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 09/11/2021] [Indexed: 12/13/2022] Open
Abstract
High-grade serous ovarian carcinoma (HGSOC) is the most aggressive type of ovarian cancer, often diagnosed at advanced stages. Molecularly, HGSOC shows high degree of genomic instability associated with large number of genetic alterations. BRD4 is the 4th most amplified gene in HGSOC, which correlates with poor patients' prognosis. BRD4 is constitutively expressed and generates two proteins, BRD4 long (BRD4-L) and BRD4 short (BRD4-S). Both isoforms contain bromodomains that bind to lysine-acetylated histones. Amongst other functions, BRD4 participates in chromatin organization, acetylation of histones, transcriptional control and DNA damage repair. In cancer patients with amplified BRD4, the increased activity of BRD4 is associated with higher expression of oncogenes, such as MYC, NOTCH3 and NRG1. BRD4-driven oncogenes promote increased tumor cells proliferation, genetic instability, epithelial-mesenchymal transition, metastasis and chemoresistance. Ablation of BRD4 activity can be successfully achieved with bromodomain inhibitors (BETi) and degraders, and it has been applied in pre-clinical and clinical settings. Inhibition of BRD4 function has an effective anti-cancer effect, reducing tumor growth whether ablated by single agents or in combination with other drugs. When combined with standard chemotherapy, BETi are capable of sensitizing highly resistant ovarian cancer cell lines to platinum drugs. Despite the evidence that BRD4 amplification in ovarian cancer contributes to poor patient prognosis, little is known about the specific mechanisms by which BRD4 drives tumor progression. In addition, newly emerging data revealed that BRD4 isoforms exhibit contradicting functions in cancer. Therefore, it is paramount to expand studies elucidating distinct roles of BRD4-L and BRD4-S in HGSOC, which has important implications on development of therapeutic approaches targeting BRD4.
Collapse
Affiliation(s)
- Ana Luiza Drumond-Bock
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA.
| | - Magdalena Bieniasz
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| |
Collapse
|
26
|
Niu H, Song F, Wei H, Li Y, Huang H, Wu C. Inhibition of BRD4 Suppresses the Growth of Esophageal Squamous Cell Carcinoma. Cancer Invest 2021; 39:826-841. [PMID: 34519605 DOI: 10.1080/07357907.2021.1975736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Bromodomain-containing protein 4 (BRD4) binds acetylated lysine residues on histones to facilitate the epigenetic regulation of many genes, and it plays a key role in many cancer types. Despite many prior reports that have explored the importance of BRD4 in oncogenesis and the regulation of epigenetic memory, its role in esophageal squamous cell carcinoma (ESCC) progression is poorly understood. Here, we investigated BRD4 expression in human ESCC tissues to understand how it regulates the biology of these tumor cells. METHODS BRD4 expression in ESCC tissues was measured via immunohistochemical staining. BRD4 inhibition in the Eca-109 and KYSE-150 ESCC cell lines was conducted to explore its functional role in these tumor cells. RESULTS BRD4 overexpression was observed in ESCC tissues and cells, and inhibiting the function of the gene impaired the proliferative, invasive, and migratory activity of these cells while promoting their apoptosis. Cyclin D1 and c-Myc expression were also suppressed by BRD4 inhibition, and the expression of key epithelial-mesenchymal transition markers including E-cadherin and Vimentin was markedly altered by such inhibition. CONCLUSIONS BRD4 plays key functional roles in the biology of ESCC, proposing that it could be a viable therapeutic target for treating this cancer type.
Collapse
Affiliation(s)
- Haiyu Niu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Department of Oncology, Lanzhou University Second Hospital, Lanzhou, China.,Institute of Cell Therapy, Soochow University, Changzhou, China
| | - Feixue Song
- Department of Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Hanwen Wei
- Department of Cardiology, The First People's Hospital of Lanzhou, Lanzhou, China
| | - Yuan Li
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Institute of Cell Therapy, Soochow University, Changzhou, China
| | - Hao Huang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Institute of Cell Therapy, Soochow University, Changzhou, China
| | - Changping Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Institute of Cell Therapy, Soochow University, Changzhou, China.,Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
27
|
Coughlan AY, Testa G. Exploiting epigenetic dependencies in ovarian cancer therapy. Int J Cancer 2021; 149:1732-1743. [PMID: 34213777 PMCID: PMC9292863 DOI: 10.1002/ijc.33727] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 01/02/2023]
Abstract
Ovarian cancer therapy has remained fundamentally unchanged for 50 years, with surgery and chemotherapy still the frontline treatments. Typically asymptomatic until advanced stages, ovarian cancer is known as “the silent killer.” Consequently, it has one of the worst 5‐year survival rates, as low as 30%. The most frequent driver mutations are found in well‐defined tumor suppressors, such as p53 and BRCA1/2. In recent years, it has become clear that, like the majority of other cancers, many epigenetic regulators are altered in ovarian cancer, including EZH2, SMARCA2/4 and ARID1A. Disruption of epigenetic regulators often leads to loss of transcriptional control, aberrant cell fate trajectories and disruption of senescence, apoptotic and proliferation pathways. These mitotically inherited epigenetic alterations are particularly promising targets for therapy as they are largely reversible. Consequently, many drugs targeting chromatin modifiers and other epigenetic regulators are at various stages of clinical trials for other cancers. Understanding the mechanisms by which ovarian cancer‐specific epigenetic processes are disrupted in patients can allow for informed targeting of epigenetic pathways tailored for each patient. In recent years, there have been groundbreaking new advances in disease modeling through ovarian cancer organoids; these models, alongside single‐cell transcriptomic and epigenomic technologies, allow the elucidation of the epigenetic pathways deregulated in ovarian cancer. As a result, ovarian cancer therapy may finally be ready to advance to next‐generation treatments. Here, we review the major developments in ovarian cancer, including genetics, model systems and technologies available for their study and the implications of applying epigenetic therapies to ovarian cancer.
Collapse
Affiliation(s)
- Aisling Y Coughlan
- Department of Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Giuseppe Testa
- Department of Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy.,Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| |
Collapse
|
28
|
Goel N, Foxall ME, Scalise CB, Wall JA, Arend RC. Strategies in Overcoming Homologous Recombination Proficiency and PARP Inhibitor Resistance. Mol Cancer Ther 2021; 20:1542-1549. [PMID: 34172532 DOI: 10.1158/1535-7163.mct-20-0992] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/21/2021] [Accepted: 06/23/2021] [Indexed: 11/16/2022]
Abstract
Ovarian cancer is the second most common gynecologic malignancy in the United States and the most common cause of gynecologic cancer-related death. The majority of ovarian cancers ultimately recur despite excellent response rates to upfront platinum- and taxane-based chemotherapy. Maintenance therapy after frontline treatment has emerged in recent years as an effective tool for extending the platinum-free interval of these patients. Maintenance therapy with PARP inhibitors (PARPis), in particular, has become part of standard of care in the upfront setting and in patients with platinum-sensitive disease. Homologous recombination deficient (HRD) tumors have a nonfunctioning homologous recombination repair (HRR) pathway and respond well to PARPis, which takes advantage of synthetic lethality by concomitantly impairing DNA repair mechanisms. Conversely, patients with a functioning HRR pathway, that is, HR-proficient tumors, can still elicit benefit from PARPi, but the efficacy is not as remarkable as what is seen in HRD tumors. PARPis are ineffective in some patients due to HR proficiency, which is either inherent to the tumor or potentially acquired as a method of therapeutic resistance. This review seeks to outline current strategies employed by clinicians and scientists to overcome PARPi resistance-either acquired or inherent to the tumor.
Collapse
Affiliation(s)
- Nidhi Goel
- University of Alabama School of Medicine, Birmingham, Alabama
| | - McKenzie E Foxall
- Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Carly Bess Scalise
- Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jaclyn A Wall
- Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Rebecca C Arend
- Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
29
|
Harachi M, Masui K, Cavenee WK, Mischel PS, Shibata N. Protein Acetylation at the Interface of Genetics, Epigenetics and Environment in Cancer. Metabolites 2021; 11:216. [PMID: 33916219 PMCID: PMC8066013 DOI: 10.3390/metabo11040216] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 03/25/2021] [Accepted: 03/31/2021] [Indexed: 02/07/2023] Open
Abstract
Metabolic reprogramming is an emerging hallmark of cancer and is driven by abnormalities of oncogenes and tumor suppressors. Accelerated metabolism causes cancer cell aggression through the dysregulation of rate-limiting metabolic enzymes as well as by facilitating the production of intermediary metabolites. However, the mechanisms by which a shift in the metabolic landscape reshapes the intracellular signaling to promote the survival of cancer cells remain to be clarified. Recent high-resolution mass spectrometry-based proteomic analyses have spotlighted that, unexpectedly, lysine residues of numerous cytosolic as well as nuclear proteins are acetylated and that this modification modulates protein activity, sublocalization and stability, with profound impact on cellular function. More importantly, cancer cells exploit acetylation as a post-translational protein for microenvironmental adaptation, nominating it as a means for dynamic modulation of the phenotypes of cancer cells at the interface between genetics and environments. The objectives of this review were to describe the functional implications of protein lysine acetylation in cancer biology by examining recent evidence that implicates oncogenic signaling as a strong driver of protein acetylation, which might be exploitable for novel therapeutic strategies against cancer.
Collapse
Affiliation(s)
- Mio Harachi
- Department of Pathology, Division of Pathological Neuroscience, Tokyo Women’s Medical University, Tokyo 162-8666, Japan; (M.H.); (N.S.)
| | - Kenta Masui
- Department of Pathology, Division of Pathological Neuroscience, Tokyo Women’s Medical University, Tokyo 162-8666, Japan; (M.H.); (N.S.)
| | - Webster K. Cavenee
- Ludwig Institute for Cancer Research, University of California San Diego, La Jolla, CA 92093, USA;
| | - Paul S. Mischel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA;
| | - Noriyuki Shibata
- Department of Pathology, Division of Pathological Neuroscience, Tokyo Women’s Medical University, Tokyo 162-8666, Japan; (M.H.); (N.S.)
| |
Collapse
|
30
|
Megiorni F, Camero S, Pontecorvi P, Camicia L, Marampon F, Ceccarelli S, Anastasiadou E, Bernabò N, Perniola G, Pizzuti A, Benedetti Panici P, Tombolini V, Marchese C. OTX015 Epi-Drug Exerts Antitumor Effects in Ovarian Cancer Cells by Blocking GNL3-Mediated Radioresistance Mechanisms: Cellular, Molecular and Computational Evidence. Cancers (Basel) 2021; 13:cancers13071519. [PMID: 33806232 PMCID: PMC8059141 DOI: 10.3390/cancers13071519] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/10/2021] [Accepted: 03/19/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The outcome for women diagnosed with ovarian cancer (OC), the most aggressive gynecological tumor worldwide, remains very poor. Encouraging therapeutic impact of epigenetic drugs has been suggested in a wide range of human solid tumors, including OC. The present study assessed the in vitro cytostatic and cytotoxic effects of OTX015, a pan Bromodomain and Extra-Terminal motif inhibitor, in human OC cells, both as single treatment and in combination with radiotherapy. Cellular, molecular and computational network analyses indicated the centrality of GNL3 downregulation in mediating the OTX015-related antitumor efficacy that blocks disease progression/maintenance and radioresistance acquisition. Our preclinical results confirm that targeted and combinatorial treatments represent effective anticancer strategies to be translated in the clinical research for improving OC patient care. Abstract Ovarian cancer (OC) is the most aggressive gynecological tumor worldwide and, notwithstanding the increment in conventional treatments, many resistance mechanisms arise, this leading to cure failure and patient death. So, the use of novel adjuvant drugs able to counteract these pathways is urgently needed to improve patient overall survival. A growing interest is focused on epigenetic drugs for cancer therapy, such as Bromodomain and Extra-Terminal motif inhibitors (BETi). Here, we investigate the antitumor effects of OTX015, a novel BETi, as a single agent or in combination with ionizing radiation (IR) in OC cellular models. OTX015 treatment significantly reduced tumor cell proliferation by triggering cell cycle arrest and apoptosis that were linked to nucleolar stress and DNA damage. OTX015 impaired migration capacity and potentiated IR effects by reducing the expression of different drivers of cancer resistance mechanisms, including GNL3 gene, whose expression was found to be significantly higher in OC biopsies than in normal ovarian tissues. Gene specific knocking down and computational network analysis confirmed the centrality of GNL3 in OTX015-mediated OC antitumor effects. Altogether, our findings suggest OTX015 as an effective option to improve therapeutic strategies and overcome the development of resistant cancer cells in patients with OC.
Collapse
Affiliation(s)
- Francesca Megiorni
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (E.A.); (A.P.); (C.M.)
- Correspondence: ; Tel.: +39-06-4997-8272
| | - Simona Camero
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (S.C.); (L.C.); (G.P.); (P.B.P.)
| | - Paola Pontecorvi
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (E.A.); (A.P.); (C.M.)
| | - Lucrezia Camicia
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (S.C.); (L.C.); (G.P.); (P.B.P.)
| | - Francesco Marampon
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (F.M.); (V.T.)
| | - Simona Ceccarelli
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (E.A.); (A.P.); (C.M.)
| | - Eleni Anastasiadou
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (E.A.); (A.P.); (C.M.)
| | - Nicola Bernabò
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, 64100 Teramo, Italy;
| | - Giorgia Perniola
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (S.C.); (L.C.); (G.P.); (P.B.P.)
| | - Antonio Pizzuti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (E.A.); (A.P.); (C.M.)
| | - Pierluigi Benedetti Panici
- Department of Maternal and Child Health and Urological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (S.C.); (L.C.); (G.P.); (P.B.P.)
| | - Vincenzo Tombolini
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (F.M.); (V.T.)
| | - Cinzia Marchese
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy; (P.P.); (S.C.); (E.A.); (A.P.); (C.M.)
| |
Collapse
|
31
|
Zhao X, Zhou HB, Liu J, Xie J, Hu R. Apigenin suppresses proliferation, invasion, and epithelial-mesenchymal transition of cervical carcinoma cells by regulation of miR-152/BRD4 axis. Kaohsiung J Med Sci 2021; 37:583-593. [PMID: 33611824 DOI: 10.1002/kjm2.12370] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/04/2021] [Accepted: 01/07/2021] [Indexed: 01/01/2023] Open
Abstract
The present study aimed to investigate the role of apigenin and the molecular mechanism of miR-152-5p and bromodomain containing 4 (BRD4) in the proliferation, invasion, and epithelial-mesenchymal transition (EMT) of cervical carcinoma cells. Quantitative real-time PCR was used to detect the transfection efficiency and the expression of miR-152-5p and BRD4. Western blotting was conducted to evaluate the protein level of BRD4, E-cadherin, N-cadherin, and MMP9. Luciferase reporter assay was performed to confirm whether miR-152-5p bound to BRD4. MTT and Transwell invasion assay were applied to determine the cell proliferation and invasion, respectively. MiR-152-5p was downregulated and BRD4 was upregulated in cervical carcinoma tissue. Besides, miR-152-5p could directly bind to BRD4 in Hela and CaSki cells. In addition, apigenin inhibited proliferation, invasion, and EMT of Hela and CaSki cells by regulating miR-152-5p/BRD4 axis. Apigenin suppresses proliferation, invasion, and induced EMT of cervical carcinoma cells by regulation of miR-152-5p/BRD4 axis.
Collapse
Affiliation(s)
- Xia Zhao
- Department of Obstetrics and Gynecology, The first Affiliated Hospital of University of South China, Hengyang, China
| | - Hua-Bo Zhou
- Department of Intensive Care Unit, Hengyang City Central Hospital, Hengyang, China
| | - Jie Liu
- Department of Obstetrics and Gynecology, The first Affiliated Hospital of University of South China, Hengyang, China
| | - Jing Xie
- Department of Obstetrics and Gynecology, The first Affiliated Hospital of University of South China, Hengyang, China
| | - Rong Hu
- Department of Radiology, The first Affiliated Hospital of University of South China, Hengyang, China
| |
Collapse
|
32
|
Boija A, Klein IA, Young RA. Biomolecular Condensates and Cancer. Cancer Cell 2021; 39:174-192. [PMID: 33417833 PMCID: PMC8721577 DOI: 10.1016/j.ccell.2020.12.003] [Citation(s) in RCA: 153] [Impact Index Per Article: 51.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/02/2020] [Accepted: 12/04/2020] [Indexed: 12/14/2022]
Abstract
Malignant transformation is characterized by dysregulation of diverse cellular processes that have been the subject of detailed genetic, biochemical, and structural studies, but only recently has evidence emerged that many of these processes occur in the context of biomolecular condensates. Condensates are membrane-less bodies, often formed by liquid-liquid phase separation, that compartmentalize protein and RNA molecules with related functions. New insights from condensate studies portend a profound transformation in our understanding of cellular dysregulation in cancer. Here we summarize key features of biomolecular condensates, note where they have been implicated-or will likely be implicated-in oncogenesis, describe evidence that the pharmacodynamics of cancer therapeutics can be greatly influenced by condensates, and discuss some of the questions that must be addressed to further advance our understanding and treatment of cancer.
Collapse
Affiliation(s)
- Ann Boija
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.
| | - Isaac A Klein
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA.
| | - Richard A Young
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
33
|
Pistoni M, Rossi T, Donati B, Torricelli F, Polano M, Ciarrocchi A. Long Noncoding RNA NEAT1 Acts as a Molecular Switch for BRD4 Transcriptional Activity and Mediates Repression of BRD4/WDR5 Target Genes. Mol Cancer Res 2021; 19:799-811. [PMID: 33547232 DOI: 10.1158/1541-7786.mcr-20-0324] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 11/18/2020] [Accepted: 02/03/2021] [Indexed: 11/16/2022]
Abstract
BRD4 is an epigenome reader known to exert key roles at the interface between chromatin remodeling and transcriptional regulation, and is primarily known for its role in promoting gene expression. In selective contexts, however, BRD4 may work as negative regulator of transcription. Here, we reported that BRD4 binds several long noncoding RNAs (lncRNA). Among these, the lncRNA NEAT1 was found to interfere with BRD4 transcriptional activity. Mechanistically, lncNEAT1 forms a complex with BRD4 and WDR5 and maintains them in a low-activity state. Treatment with Bromodomains and Extraterminal (BET) inhibitor caused the lncRNA NEAT1 to dissociate from the BRD4/WDR5 complex, restored the acetyl-transferase capacity of BRD4, and restored the availability of WDR5 to promote histone trimethylation, thereby promoting BRD4/WDR5 transcriptional activity and activation of target gene expression. In addition, the lncRNA NEAT1 then became available to bind and to inhibit EZH2, cooperatively increasing transcriptional activation. IMPLICATIONS: Our results revealed an epigenetic program that involves the interaction between the lncRNA NEAT1 and BRD4, functioning as a molecular switch between BRD4's activator and repressor chromatin complexes.
Collapse
Affiliation(s)
- Mariaelena Pistoni
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy.
| | - Teresa Rossi
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Benedetta Donati
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Federica Torricelli
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Maurizio Polano
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda USL-IRCCS di Reggio Emilia, Reggio Emilia, Italy.
| |
Collapse
|
34
|
Abstract
BACKGROUND Bromodomain and extra-terminal (BET) proteins are epigenetic readers that bind to acetylated lysines of histones and regulate gene transcription. BET protein family members mediate the expression of various oncogenic drivers in ovarian cancer, such as the MYC and Neuregulin 1 (NRG1) genes. BRD4, the most thoroughly studied member of the BET family, is amplified in a significant subset of high-grade serous carcinomas (HGSC) of the ovary. It has been reported that BET inhibitors can attenuate the proliferation and dissemination of ovarian cancer cells by inhibiting oncogenic pathways, such as the FOXM1 and JAK/STAT pathways. BET inhibition can re-sensitize resistant ovarian cancer cells to already approved anticancer agents, including cisplatin and PARP inhibitors. This synergism was also confirmed in vivo in animal models. These and other preclinical results provide a promising basis for the application of BET inhibitors in ovarian cancer treatment. Currently, Phase I/II clinical trials explore the safety and efficacy profiles of BET inhibitors in various solid tumors, including ovarian tumors. Here, we review current knowledge on the molecular effects and preclinical activities of BET inhibitors in ovarian tumors. CONCLUSIONS BET proteins have emerged as new druggable targets for ovarian cancer. BET inhibitors may enhance antitumor activity when co-administered with conventional treatment regimens. Results from ongoing Phase I/II studies are anticipated to confirm this notion.
Collapse
|
35
|
Manasa P, Sidhanth C, Krishnapriya S, Vasudevan S, Ganesan TS. Oncogenes in high grade serous adenocarcinoma of the ovary. Genes Cancer 2020; 11:122-136. [PMID: 33488950 PMCID: PMC7805537 DOI: 10.18632/genesandcancer.206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 10/15/2020] [Indexed: 12/19/2022] Open
Abstract
High grade serous ovarian cancer is characterized by relatively few mutations occurring at low frequency, except in TP53. However other genetic aberrations such as copy number variation alter numerous oncogenes and tumor suppressor genes. Oncogenes are positive regulators of tumorigenesis and play a critical role in cancer cell growth, proliferation, and survival. Accumulating evidence suggests that they are crucial for the development and the progression of high grade serous ovarian carcinoma (HGSOC). Though many oncogenes have been identified, no successful inhibitors targeting these molecules and their associated pathways are available. This review discusses oncogenes that have been identified recently in HGSOC using different screening strategies. All the genes discussed in this review have been functionally characterized both in vitro and in vivo and some of them are able to transform immortalized ovarian surface epithelial and fallopian tube cells upon overexpression. However, it is necessary to delineate the molecular pathways affected by these oncogenes for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Pacharla Manasa
- Laboratory for Cancer Biology, Department of Medical Oncology and Clinical Research Cancer Institute (WIA), Chennai, India
| | - Chirukandath Sidhanth
- Laboratory for Cancer Biology, Department of Medical Oncology and Clinical Research Cancer Institute (WIA), Chennai, India
| | - Syama Krishnapriya
- Laboratory for Cancer Biology, Department of Medical Oncology and Clinical Research Cancer Institute (WIA), Chennai, India
| | - Sekar Vasudevan
- Laboratory for Cancer Biology, Department of Medical Oncology and Clinical Research Cancer Institute (WIA), Chennai, India
| | - Trivadi S Ganesan
- Laboratory for Cancer Biology, Department of Medical Oncology and Clinical Research Cancer Institute (WIA), Chennai, India
| |
Collapse
|
36
|
Nameki R, Chang H, Reddy J, Corona RI, Lawrenson K. Transcription factors in epithelial ovarian cancer: histotype-specific drivers and novel therapeutic targets. Pharmacol Ther 2020; 220:107722. [PMID: 33137377 DOI: 10.1016/j.pharmthera.2020.107722] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 10/26/2020] [Indexed: 02/06/2023]
Abstract
Transcription factors (TFs) are major contributors to cancer risk and somatic development. In preclinical and clinical studies, direct or indirect inhibition of TF-mediated oncogenic gene expression profiles have proven to be effective in many tumor types, highlighting this group of proteins as valuable therapeutic targets. In spite of this, our understanding of TFs in epithelial ovarian cancer (EOC) is relatively limited. EOC is a heterogeneous disease composed of five major histologic subtypes; high-grade serous, low-grade serous, endometrioid, clear cell and mucinous. Each histology is associated with unique clinical etiologies, sensitivity to therapies, and molecular signatures - including diverse transcriptional regulatory programs. While some TFs are shared across EOC subtypes, a set of TFs are expressed in a histotype-specific manner and likely explain part of the histologic diversity of EOC subtypes. Targeting TFs present with unique opportunities for development of novel precision medicine strategies for ovarian cancer. This article reviews the critical TFs in EOC subtypes and highlights the potential of exploiting TFs as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Robbin Nameki
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Heidi Chang
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jessica Reddy
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Rosario I Corona
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Kate Lawrenson
- Women's Cancer Research Program at the Samuel Oschin Comprehensive Cancer Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Center for Bioinformatics and Functional Genomics, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
37
|
Ray-Coquard I, Vanacker H, Le Saux O, Tredan O. Overcoming resistance to PARP inhibitor in epithelial ovarian cancer, are we ready? EBioMedicine 2020; 61:103046. [PMID: 33038760 PMCID: PMC7553990 DOI: 10.1016/j.ebiom.2020.103046] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 09/21/2020] [Indexed: 10/29/2022] Open
Affiliation(s)
- Isabelle Ray-Coquard
- Centre Léon Bérard, 28 Prom. Léa et Napoléon Bullukian, Lyon 69008, France; University Claude Bernard Lyon 1, Lyon, France.
| | - Hélène Vanacker
- Centre Léon Bérard, 28 Prom. Léa et Napoléon Bullukian, Lyon 69008, France; University Claude Bernard Lyon 1, Lyon, France
| | - Olivia Le Saux
- Centre Léon Bérard, 28 Prom. Léa et Napoléon Bullukian, Lyon 69008, France; Centre de Recherche en Cancérologie de Lyon, UMR Inserm 1052, CNRS 5286, Centre Léon-Bérard, Lyon, France
| | - Olivier Tredan
- Centre Léon Bérard, 28 Prom. Léa et Napoléon Bullukian, Lyon 69008, France
| |
Collapse
|
38
|
Hsu JY, Major JL, Riching AS, Sen R, Pires da Silva J, Bagchi RA. Beyond the genome: challenges and potential for epigenetics-driven therapeutic approaches in pulmonary arterial hypertension. Biochem Cell Biol 2020; 98:631-646. [PMID: 32706995 DOI: 10.1139/bcb-2020-0039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a devastating disease of the cardiopulmonary system caused by the narrowing of the pulmonary arteries, leading to increased vascular resistance and pressure. This leads to right ventricle remodeling, dysfunction, and eventually, death. While conventional therapies have largely focused on targeting vasodilation, other pathological features of PAH including aberrant inflammation, mitochondrial dynamics, cell proliferation, and migration have not been well explored. Thus, despite some recent improvements in PAH treatment, the life expectancy and quality of life for patients with PAH remains poor. Showing many similarities to cancers, PAH is characterized by increased pulmonary arterial smooth muscle cell proliferation, decreased apoptotic signaling pathways, and changes in metabolism. The recent successes of therapies targeting epigenetic modifiers for the treatment of cancer has prompted epigenetic research in PAH, revealing many new potential therapeutic targets. In this minireview we discuss the emergence of epigenetic dysregulation in PAH and highlight epigenetic-targeting compounds that may be effective for the treatment of PAH.
Collapse
Affiliation(s)
- Jessica Y Hsu
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jennifer L Major
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Andrew S Riching
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA.,Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rwik Sen
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Julie Pires da Silva
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Rushita A Bagchi
- Department of Medicine, Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
39
|
Li X, Fu Y, Yang B, Guo E, Wu Y, Huang J, Zhang X, Xiao R, Li K, Wang B, Hu J, Sun C, Chen G. BRD4 Inhibition by AZD5153 Promotes Antitumor Immunity via Depolarizing M2 Macrophages. Front Immunol 2020; 11:89. [PMID: 32184777 PMCID: PMC7058627 DOI: 10.3389/fimmu.2020.00089] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 01/14/2020] [Indexed: 01/13/2023] Open
Abstract
High-grade serous ovarian cancer (HGSOC), with its high recurrence rates, urges for reasonable therapeutic strategies that can prolong overall survival. A tumor microenvironment (TME) discloses prognostic and prospective information on cancer, such as the expression level of PD-1 or PD-L1. However, in HGSOC, the impact of the therapies aiming at these targets remains unsatisfying. Tumor-associated macrophages (TAMs) in HGSOC make up a large part of the TMEs and transform between diverse phenotypes under different treatments. AZD5153 inhibiting BRD4, as a potential therapeutic strategy for HGSOC, was demonstrated to confer controversial plasticity on TAMs, which shows the need to uncover its impact on TAMs in HGSOC. Therefore, we established models for TAMs and TAMs co-culturing with T lymphocytes in vitro. Via RT-PCR and flow cytometry, we find that AZD5153 resets TAMs from M2-type macrophages to M1-like macrophages, consequently promoting pro-inflammatory cytokine secretion and thus activating CD8+ cytotoxic T lymphocytes (CTLs) in vitro. This modification occurs on MAF transcripts in TAMs and modified by BRD4, which is the target of AZD5153. Importantly, the 3-D microfluid model demonstrates that AZD5153 sensitizes ovarian cancer to anti-PD-L1 therapy. Our results clarify that besides eliminating tumor cells directly, AZD5153 works as a regulator of the TAM phenotype, providing a novel strategy combining AZD5153 and PD-1/PD-L1 antibody that could benefit HGSOC patients.
Collapse
Affiliation(s)
- Xi Li
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Fu
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Yang
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ensong Guo
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yifan Wu
- Department of Gynecology and Obstetrics, The Central Hospital of Wuhan, Wuhan, China
| | - Jia Huang
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxiao Zhang
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Rourou Xiao
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Kezhen Li
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Beibei Wang
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Junbo Hu
- Department of Gastrointestinal Surgery, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Chaoyang Sun
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Chen
- Department of Gynecology and Obstetrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
40
|
Morel D, Jeffery D, Aspeslagh S, Almouzni G, Postel-Vinay S. Combining epigenetic drugs with other therapies for solid tumours - past lessons and future promise. Nat Rev Clin Oncol 2019; 17:91-107. [PMID: 31570827 DOI: 10.1038/s41571-019-0267-4] [Citation(s) in RCA: 256] [Impact Index Per Article: 51.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2019] [Indexed: 12/16/2022]
Abstract
Epigenetic dysregulation has long been recognized as a key factor contributing to tumorigenesis and tumour maintenance that can influence all of the recognized hallmarks of cancer. Despite regulatory approvals for the treatment of certain haematological malignancies, the efficacy of the first generation of epigenetic drugs (epi-drugs) in patients with solid tumours has been disappointing; however, successes have now been achieved in selected solid tumour subtypes, thanks to the development of novel compounds and a better understanding of cancer biology that have enabled precision medicine approaches. Several lines of evidence support that, beyond their potential as monotherapies, epigenetic drugs could have important roles in synergy with other anticancer therapies or in reversing acquired therapy resistance. Herein, we review the mechanisms by which epi-drugs can modulate the sensitivity of cancer cells to other forms of anticancer therapy, including chemotherapy, radiation therapy, hormone therapy, molecularly targeted therapy and immunotherapy. We provide a critical appraisal of the preclinical rationale, completed clinical studies and ongoing clinical trials relating to combination therapies incorporating epi-drugs. Finally, we propose and discuss rational clinical trial designs and drug development strategies, considering key factors including patient selection, tumour biomarker evaluation, drug scheduling and response assessment and study end points, with the aim of optimizing the development of such combinations.
Collapse
Affiliation(s)
- Daphné Morel
- ATIP-Avenir Group, UMR981, INSERM (French National Institute of Health and Medical Research), Gustave Roussy Cancer Campus, Villejuif, France
| | - Daniel Jeffery
- Nuclear Dynamics Unit - UMR3664, National Centre for Scientific Research, Institut Curie, Paris, France
| | | | - Geneviève Almouzni
- Nuclear Dynamics Unit - UMR3664, National Centre for Scientific Research, Institut Curie, Paris, France.
| | - Sophie Postel-Vinay
- ATIP-Avenir Group, UMR981, INSERM (French National Institute of Health and Medical Research), Gustave Roussy Cancer Campus, Villejuif, France. .,Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Paris-Saclay University, Villejuif, France.
| |
Collapse
|
41
|
Systematic Identification of Characteristic Genes of Ovarian Clear Cell Carcinoma Compared with High-Grade Serous Carcinoma Based on RNA-Sequencing. Int J Mol Sci 2019; 20:ijms20184330. [PMID: 31487856 PMCID: PMC6770582 DOI: 10.3390/ijms20184330] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/25/2019] [Accepted: 09/02/2019] [Indexed: 12/28/2022] Open
Abstract
Objective: Ovarian cancer has the highest mortality among gynecological cancers. High-grade serous carcinoma (HGSC) is the most common histotype of ovarian cancer regardless of ethnicity, whereas clear cell carcinoma (CCC) is more common in East Asians than Caucasians. The elucidation of predominant signaling pathways in these cancers is the first step towards understanding their molecular mechanisms and developing their clinical management. Methods: RNA sequencing was performed for 27 clinical ovarian specimens from Japanese women. Principal component analysis (PCA) was conducted on the sequence data mapped on RefSeq with normalized read counts, and functional annotation analysis was performed on genes with substantial weights in PCA. Knockdown experiments were conducted on the selected genes on the basis of PCA. Results: Functional annotation analysis of PCA-defined genes showed predominant pathways, such as cell growth regulators and blood coagulators in CCC and transcription regulators in HGSC. Knockdown experiments showed that the inhibition of the calcium-dependent protein copine 8 (CPNE8) and the transcription factor basic helix-loop-helix family member e 41 (BHLHE41) repressed the proliferation of CCC- and HGSC-derived cells, respectively. Conclusions: This study identified CPNE8 and BHLHE41 as characteristic genes for CCC and HGSC, respectively. The systemic identification of differentially expressed genes in CCC and HGSC will provide useful information to understand transcriptomic differences in these ovarian cancers and to further develop potential diagnostic and therapeutic options for advanced disease.
Collapse
|
42
|
Washington CR, Richardson DL, Moore KN. Olaparib in the treatment of ovarian cancer. Future Oncol 2019; 15:3435-3449. [PMID: 31478762 DOI: 10.2217/fon-2019-0271] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The poly ADP ribose polymerase olaparib is currently approved in front line BRCA-associated epithelial ovarian cancer (EOC), platinum-sensitive recurrence agnostic to BRCA status and for gBRCA as treatment in the fourth line and beyond. Women who are diagnosed with advanced stage EOC face a formidable challenge in overcoming their disease and achieving long-term, disease-free survival. The qualifier here is disease free. EOC is largely exquisitely chemosensitive, especially in the treatment naive (first line) setting and the expectation is that the vast majority of women will complete front line platinum-based chemotherapy with a response. When unselected (not selected by BRCA) women are enrolled on clinical trials, the response rate among those who have measurable disease at the time of chemotherapy initiation is 48% for carboplatin/paclitaxel and 67% for carboplatin/paclitaxel plus bevacizumab. When one considers the addition of women who start chemotherapy without measurable disease, they will likely also end chemotherapy without measurable disease and the overall rate of no evidence of disease at conclusion of chemotherapy approaches 80%. Despite this, the majority of women will suffer relapse of their disease, typically within the first 3 years following completion of therapy. Once recurrent, the disease is highly treatable for many years but no longer considered curable. This review will cover indications for olaparib in ovarian cancer as well as ongoing combination trials and rationale for these combinations.
Collapse
Affiliation(s)
- Christina R Washington
- Division of Gynecologic Oncology, Stephenson Cancer Center at the University of Oklahoma HSC Oklahoma City, OK 73121, USA
| | - Debra L Richardson
- Division of Gynecologic Oncology, Stephenson Cancer Center at the University of Oklahoma HSC Oklahoma City, OK 73121, USA
| | - Kathleen N Moore
- Division of Gynecologic Oncology, Stephenson Cancer Center at the University of Oklahoma HSC Oklahoma City, OK 73121, USA
| |
Collapse
|
43
|
Tang L, Wang P, Wang Q, Zhong L. Correlation of LAMA3 with onset and prognosis of ovarian cancer. Oncol Lett 2019; 18:2813-2818. [PMID: 31402958 PMCID: PMC6676679 DOI: 10.3892/ol.2019.10600] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 05/06/2019] [Indexed: 01/29/2023] Open
Abstract
Correlation of laminin subunit α3 (LAMA3) gene with onset and prognosis of ovarian cancer was investigated. In total, 210 ovarian cancer patients who received surgical resection in West China Second Hospital from March 2011 to March 2013 were randomly selected, and another 160 non-ovarian cancer patients who needed ovariectomy were also selected. The relative expression of LAMA3 gene was compared via quantitative polymerase chain reaction (qPCR) in carcinoma tissues, para-carcinoma tissues and non-carcinoma normal tissues in ovarian cancer patients. The methylation level was compared among the above three tissue types. The correlation between the mutation site rs12373237 in LAMA3 gene and onset was analyzed. The expression of laminin in ovarian cancer was detected using immunohistochemistry. Moreover, the 5-year survival rate after operation was recorded and the survival curve was plotted. The expression level of LAMA3 was lower in carcinoma tissues than those in normal tissues and para-carcinoma tissues (P<0.05). The methylation degree was lower in para-carcinoma tissues and normal tissues than that in carcinoma tissues (P<0.05). The CC homozygous mutation of rs12373237 was highly correlated with the onset of ovarian cancer (OR=4.333, P=0.028). The expression of LAMA3 was classified via immunohistochemistry, and the number in high-expression group (63.8%) was larger than that in low-expression group (36.2%) (P<0.05). According to the analysis of 5-year survival rate, the recurrence-free survival rate and overall survival rate in LAMA3 high-expression group were significantly higher than those in LAMA3 low-expression group (P<0.05). The expression level and base mutation of LAMA3 gene can change the level of laminin, which have a certain influence on the onset and prognosis of ovarian cancer.
Collapse
Affiliation(s)
- Lin Tang
- Emergency Obstetrics and Gynecology Department, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Pin Wang
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Qilin Wang
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Lan Zhong
- Department of Obstetrics and Gynecology, West China Second Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
44
|
Song H, Shi L, Xu Y, Xu T, Fan R, Cao M, Xu W, Song J. BRD4 promotes the stemness of gastric cancer cells via attenuating miR-216a-3p-mediated inhibition of Wnt/β-catenin signaling. Eur J Pharmacol 2019; 852:189-197. [PMID: 30876979 DOI: 10.1016/j.ejphar.2019.03.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/01/2019] [Accepted: 03/11/2019] [Indexed: 02/02/2023]
Abstract
The bromodomain and extra-terminal domain (BET) protein BRD4 is emerging as a potential target for cancer therapy. However, BRD4 roles in regulating the stemness of gastric cancer cells are unclear. Here, we demonstrated that BRD4 expression was significantly increased in gastric cancer tissues, cell spheroids, and BRD4 knockdown attenuated the stemness of gastric cancer cells characterized as the decrease of stemness markers expression, capacity of cells spheroids formation and ALDH1 activity. Importantly, BRD4 expression was negatively correlated with overall survival, first progression survival and post progression survival of gastric cancer patients. Mechanistic investigations revealed that miR-216a-3p was the most remarkably upregulated miRNA in response to BRD4 knockdown and Wnt/β-catenin signaling was necessary for BRD4-mediated promotion on the stemness of gastric cancer cells. Additionally, BRD4 directly bound to the promoter and promoted the methylation level of MIR216A promoter, thus decreasing miR-216a-3p level. Notably, Wnt3a was identified as the direct target of miR-216a-3p in gastric cancer cells. Therefore, our results defined a BRD4/miR-216a-3p/Wnt/β-catenin pathway in regulating the stemness of gastric cancer cells.
Collapse
Affiliation(s)
- Hu Song
- Department of General Surgery, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu 221002, PR China; Institute of Digestive Disease, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, Jiangsu 221002, PR China
| | - Linseng Shi
- Department of General Surgery, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu 221002, PR China; Institute of Digestive Disease, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, Jiangsu 221002, PR China
| | - Yixin Xu
- Department of General Surgery, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu 221002, PR China; Institute of Digestive Disease, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, Jiangsu 221002, PR China
| | - Teng Xu
- Department of General Surgery, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu 221002, PR China; Institute of Digestive Disease, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, Jiangsu 221002, PR China
| | - Ruizhi Fan
- Department of General Surgery, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu 221002, PR China; Institute of Digestive Disease, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, Jiangsu 221002, PR China
| | - Meng Cao
- Department of General Surgery, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu 221002, PR China; Institute of Digestive Disease, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, Jiangsu 221002, PR China
| | - Wei Xu
- Department of General Surgery, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu 221002, PR China; Institute of Digestive Disease, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, Jiangsu 221002, PR China
| | - Jun Song
- Department of General Surgery, the Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, Jiangsu 221002, PR China; Institute of Digestive Disease, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, Jiangsu 221002, PR China.
| |
Collapse
|
45
|
Li C, Bonazzoli E, Bellone S, Choi J, Dong W, Menderes G, Altwerger G, Han C, Manzano A, Bianchi A, Pettinella F, Manara P, Lopez S, Yadav G, Riccio F, Zammataro L, Zeybek B, Yang-Hartwich Y, Buza N, Hui P, Wong S, Ravaggi A, Bignotti E, Romani C, Todeschini P, Zanotti L, Zizioli V, Odicino F, Pecorelli S, Ardighieri L, Silasi DA, Litkouhi B, Ratner E, Azodi M, Huang GS, Schwartz PE, Lifton RP, Schlessinger J, Santin AD. Mutational landscape of primary, metastatic, and recurrent ovarian cancer reveals c-MYC gains as potential target for BET inhibitors. Proc Natl Acad Sci U S A 2019; 116:619-624. [PMID: 30584090 PMCID: PMC6329978 DOI: 10.1073/pnas.1814027116] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Ovarian cancer remains the most lethal gynecologic malignancy. We analyzed the mutational landscape of 64 primary, 41 metastatic, and 17 recurrent fresh-frozen tumors from 77 patients along with matched normal DNA, by whole-exome sequencing (WES). We also sequenced 13 pairs of synchronous bilateral ovarian cancer (SBOC) to evaluate the evolutionary history. Lastly, to search for therapeutic targets, we evaluated the activity of the Bromodomain and Extra-Terminal motif (BET) inhibitor GS-626510 on primary tumors and xenografts harboring c-MYC amplifications. In line with previous studies, the large majority of germline and somatic mutations were found in BRCA1/2 (21%) and TP53 (86%) genes, respectively. Among mutations in known cancer driver genes, 77% were transmitted from primary tumors to metastatic tumors, and 80% from primary to recurrent tumors, indicating that driver mutations are commonly retained during ovarian cancer evolution. Importantly, the number, mutation spectra, and signatures in matched primary-metastatic tumors were extremely similar, suggesting transcoelomic metastases as an early dissemination process using preexisting metastatic ability rather than an evolution model. Similarly, comparison of SBOC showed extensive sharing of somatic mutations, unequivocally indicating a common ancestry in all cases. Among the 17 patients with matched tumors, four patients gained PIK3CA amplifications and two patients gained c-MYC amplifications in the recurrent tumors, with no loss of amplification or gain of deletions. Primary cell lines and xenografts derived from chemotherapy-resistant tumors demonstrated sensitivity to JQ1 and GS-626510 (P = 0.01), suggesting that oral BET inhibitors represent a class of personalized therapeutics in patients harboring recurrent/chemotherapy-resistant disease.
Collapse
Affiliation(s)
- Charles Li
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510
| | - Elena Bonazzoli
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Stefania Bellone
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Jungmin Choi
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510
| | - Weilai Dong
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510
| | - Gulden Menderes
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Gary Altwerger
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Chanhee Han
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Aranzazu Manzano
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Anna Bianchi
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Francesca Pettinella
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Paola Manara
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Salvatore Lopez
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Ghanshyam Yadav
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Francesco Riccio
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Luca Zammataro
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Burak Zeybek
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Yang Yang-Hartwich
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Natalia Buza
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510
| | - Pei Hui
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510
| | - Serena Wong
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510
| | - Antonella Ravaggi
- Department of Obstetrics & Gynecology, Angelo Nocivelli Institute of Molecular Medicine, University of Brescia, 25100 Brescia, Italy
| | - Eliana Bignotti
- Department of Obstetrics & Gynecology, Angelo Nocivelli Institute of Molecular Medicine, University of Brescia, 25100 Brescia, Italy
| | - Chiara Romani
- Department of Obstetrics & Gynecology, Angelo Nocivelli Institute of Molecular Medicine, University of Brescia, 25100 Brescia, Italy
| | - Paola Todeschini
- Department of Obstetrics & Gynecology, Angelo Nocivelli Institute of Molecular Medicine, University of Brescia, 25100 Brescia, Italy
| | - Laura Zanotti
- Department of Obstetrics & Gynecology, Angelo Nocivelli Institute of Molecular Medicine, University of Brescia, 25100 Brescia, Italy
| | - Valentina Zizioli
- Department of Obstetrics & Gynecology, Angelo Nocivelli Institute of Molecular Medicine, University of Brescia, 25100 Brescia, Italy
| | - Franco Odicino
- Department of Obstetrics & Gynecology, Angelo Nocivelli Institute of Molecular Medicine, University of Brescia, 25100 Brescia, Italy
| | - Sergio Pecorelli
- Department of Obstetrics & Gynecology, Angelo Nocivelli Institute of Molecular Medicine, University of Brescia, 25100 Brescia, Italy
| | - Laura Ardighieri
- Department of Pathology, University of Brescia, 25100 Brescia, Italy
| | - Dan-Arin Silasi
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Babak Litkouhi
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Elena Ratner
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Masoud Azodi
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Gloria S Huang
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Peter E Schwartz
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| | - Richard P Lifton
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510
- Laboratory of Human Genetics and Genomics, The Rockefeller University, New York, NY 10065
| | | | - Alessandro D Santin
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale School of Medicine, New Haven, CT 06510
| |
Collapse
|