151
|
Cummin TEC, Cox KL, Murray TD, Turaj AH, Dunning L, English VL, Fell R, Packham G, Ma Y, Powell B, Johnson PWM, Cragg MS, Carter MJ. BET inhibitors synergize with venetoclax to induce apoptosis in MYC-driven lymphomas with high BCL-2 expression. Blood Adv 2020; 4:3316-3328. [PMID: 32717030 PMCID: PMC7391160 DOI: 10.1182/bloodadvances.2020002231] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/13/2020] [Indexed: 12/12/2022] Open
Abstract
Although the MYC oncogenic network represents an attractive therapeutic target for lymphoma, MYC inhibitors have been difficult to develop. Alternatively, inhibitors of epigenetic/ transcriptional regulators, particularly the bromodomain and extraterminal (BET) family, have been used to modulate MYC. However, current benzodiazepine-derivative BET inhibitors (BETi) elicit disappointing responses and dose-limiting toxicity in relapsed/refractory lymphoma, potentially because of enrichment of high-risk molecular features and chemical backbone-associated toxicities. Consequently, novel nonbenzodiazepine BETi and improved mechanistic understanding are required. Here we characterize the responses of aggressive MYC-driven lymphomas to 2 nonbenzodiazepine BETi: PLX51107 and PLX2853. Both invoked BIM-dependent apoptosis and in vivo therapy, associated with miR-17∼92 repression, in murine Eµ-myc lymphomas, with PLX2853 exhibiting enhanced potency. Accordingly, exogenous BCL-2 expression abrogated these effects. Because high BCL-2 expression is common in diffuse large B-cell lymphoma (DLBCL), BETi were ineffective in driving apoptosis and in vivo therapy of DLBCL cell lines, mirroring clinical results. However, BETi-mediated BIM upregulation and miR-17∼92 repression remained intact. Consequently, coadministration of BETi and ABT199/venetoclax restored cell death and in vivo therapy. Collectively, these data identify BIM-dependent apoptosis as a critical mechanism of action for this class of BETi that, via coadministration of BH3 mimetics, can deliver effective tumor control in DLBCL.
Collapse
Affiliation(s)
| | - Kerry L Cox
- Antibody and Vaccine Group, Centre for Cancer Immunology
| | - Tom D Murray
- Antibody and Vaccine Group, Centre for Cancer Immunology
| | - Anna H Turaj
- Antibody and Vaccine Group, Centre for Cancer Immunology
| | - Lisa Dunning
- Preclinical Unit, Centre for Cancer Immunology, and
| | | | - Rachel Fell
- Cancer Research UK Centre, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom; and
| | - Graham Packham
- Cancer Research UK Centre, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom; and
| | - Yan Ma
- Plexxikon Inc., Berkeley, CA
| | | | - Peter W M Johnson
- Cancer Research UK Centre, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom; and
| | - Mark S Cragg
- Antibody and Vaccine Group, Centre for Cancer Immunology
| | | |
Collapse
|
152
|
Sanders YY, Lyv X, Zhou QJ, Xiang Z, Stanford D, Bodduluri S, Rowe SM, Thannickal VJ. Brd4-p300 inhibition downregulates Nox4 and accelerates lung fibrosis resolution in aged mice. JCI Insight 2020; 5:137127. [PMID: 32544088 DOI: 10.1172/jci.insight.137127] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 06/10/2020] [Indexed: 12/13/2022] Open
Abstract
Tissue regeneration capacity declines with aging in association with heightened oxidative stress. Expression of the oxidant-generating enzyme, NADPH oxidase 4 (Nox4), is elevated in aged mice with diminished capacity for fibrosis resolution. Bromodomain-containing protein 4 (Brd4) is a member of the bromodomain and extraterminal (BET) family of proteins that function as epigenetic "readers" of acetylated lysine groups on histones. In this study, we explored the role of Brd4 and its interaction with the p300 acetyltransferase in the regulation of Nox4 and the in vivo efficacy of a BET inhibitor to reverse established age-associated lung fibrosis. BET inhibition interferes with the association of Brd4, p300, and acetylated histone H4K16 with the Nox4 promoter in lung fibroblasts stimulated with the profibrotic cytokine, TGF-β1. A number of BET inhibitors, including I-BET-762, JQ1, and OTX015, downregulate Nox4 gene expression and activity. Aged mice with established and persistent lung fibrosis recover capacity for fibrosis resolution with OTX015 treatment. This study implicates epigenetic regulation of Nox4 by Brd4 and p300 and supports BET/Brd4 inhibition as an effective strategy for the treatment of age-related fibrotic lung disease.
Collapse
|
153
|
Spriano F, Stathis A, Bertoni F. Targeting BET bromodomain proteins in cancer: The example of lymphomas. Pharmacol Ther 2020; 215:107631. [PMID: 32693114 DOI: 10.1016/j.pharmthera.2020.107631] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/15/2020] [Indexed: 12/12/2022]
Abstract
The Bromo- and Extra-Terminal domain (BET) family proteins act as "readers" of acetylated histones and they are important transcription regulators. BRD2, BRD3, BRD4 and BRDT, part of the BET family, are important in different tumors, where upregulation or translocation often occurs. The potential of targeting BET proteins as anti-cancer treatment originated with data obtained with a first series of compounds, and there are now several data supporting BET inhibition in both solid tumors and hematological malignancies. Despite very positive preclinical data in different tumor types, the clinical results have been so far moderate. Using lymphoma as an example to review the data produced in the laboratory and in the context of the early clinical trials, we discuss the modalities to make BET targeting more efficient both generating novel generation of compounds and by exploring the combination with small molecules affecting various signaling pathways, BCL2, or DNA damage response signaling, but also with additional epigenetic agents and with immunotherapy. We also discuss the mechanisms of resistance and the toxicity profiles so far reported.
Collapse
Affiliation(s)
- Filippo Spriano
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland
| | - Anastasios Stathis
- Oncology Institute of Southern Switzerland, Bellinzona, Switzerland; Faculty of Biomedical Sciences, USI, Lugano, Switzerland
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland; Oncology Institute of Southern Switzerland, Bellinzona, Switzerland.
| |
Collapse
|
154
|
Epigenetic Mechanisms of Resistance to Immune Checkpoint Inhibitors. Biomolecules 2020; 10:biom10071061. [PMID: 32708698 PMCID: PMC7407667 DOI: 10.3390/biom10071061] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/13/2020] [Accepted: 07/13/2020] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have demonstrated to be highly efficient in treating solid tumors; however, many patients have limited benefits in terms of response and survival. This rapidly led to the investigation of combination therapies to enhance response rates. Moreover, predictive biomarkers were assessed to better select patients. Although PD-L1 expression remains the only validated marker in clinics, molecular profiling has brought valuable information, showing that the tumor mutation load and microsatellite instability (MSI) status were associated to higher response rates in nearly all cancer types. Moreover, in lung cancer, EGFR and MET mutations, oncogene fusions or STK11 inactivating mutations were associated with low response rates. Cancer progression towards invasive phenotypes that impede immune surveillance relies on complex regulatory networks and cell interactions within the tumor microenvironment. Epigenetic modifications, such as the alteration of histone patterns, chromatin structure, DNA methylation status at specific promoters and changes in microRNA levels, may alter the cell phenotype and reshape the tumor microenvironment, allowing cells to grow and escape from immune surveillance. The objective of this review is to make an update on the identified epigenetic changes that target immune surveillance and, ultimately, ICI responses, such as histone marks, DNA methylation and miR signatures. Translational studies or clinical trials, when available, and potential epigenetic biomarkers will be discussed as perspectives in the context of combination treatment strategies to enhance ICI responses in patients with solid tumors.
Collapse
|
155
|
Zheng C, Liu M, Fan H. Targeting complexes of super-enhancers is a promising strategy for cancer therapy. Oncol Lett 2020; 20:2557-2566. [PMID: 32782573 PMCID: PMC7400756 DOI: 10.3892/ol.2020.11855] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 05/27/2020] [Indexed: 12/16/2022] Open
Abstract
The hyperactivation and overexpression of critical oncogenes is a common occurrence in multiple types of malignant tumors. Recently, the abnormal activation mechanism of an oncogene by a super-enhancer (SE) has attracted significant attention. A series of changes (insertion, deletion, translocation and rearrangement) in the genome occurring in cancer cells may generate new SEs, leading to the overexpression of SE-driven oncogenes. SEs are composed of typical enhancers densely loaded with mediator complexes, transcription factors, and chromatin regulators, and drive the overexpression of oncogenes associated with cellular identity and disease. Cyclin-dependent kinase 7 (CDK7) and bromodomain protein 4 (BRD4) are critical mediator complexes associated with SE-mediated transcription. Clinical trials have shown that emerging small-molecule inhibitors (CDK7 and BRD4 inhibitor), targeting the SE exert a notable effect on cancer treatment. Increasing evidences has illustrated that the SE and its associated complexes play a critical role in the development of various types of cancer. The present review discusses the composition, function and regulation of SEs and their contribution to oncogenic transcription. In addition, creative therapeutic approaches that target SE, their advantages and disadvantages, as well as the problems with their clinical application are discussed. It was found that targeting SE may be used in conventional treatment and establish more access for patients with cancer.
Collapse
Affiliation(s)
- Chuqian Zheng
- Department of Medical Genetics and Developmental Biology, School of Medicine, The Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Min Liu
- Department of Medical Genetics and Developmental Biology, School of Medicine, The Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, P.R. China.,School of Life Science and Technology, Southeast University, Nanjing, Jiangsu 210018, P.R. China
| | - Hong Fan
- Department of Medical Genetics and Developmental Biology, School of Medicine, The Key Laboratory of Developmental Genes and Human Diseases, Ministry of Education, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
156
|
Booth S, Collins G. Epigenetic targeting in lymphoma. Br J Haematol 2020; 192:50-61. [PMID: 32609383 DOI: 10.1111/bjh.16914] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 06/01/2020] [Indexed: 12/19/2022]
Abstract
Despite considerable progress in the treatment of patients with lymphoid malignancies in recent decades, the prognosis of patients with relapsed or refractory lymphomas often remains disappointing. Increasing evidence has established the relevance of epigenetic alterations in the pathogenesis of lymphoid malignancies, and a succession of agents has been evaluated in clinical studies with varying efficacy. In the present review, we outline the importance of epigenetic modifications in lymphoma biology and discuss the published experience with epigenetic modifying agents by lymphoma subtype before considering ongoing clinical studies in this area.
Collapse
Affiliation(s)
- Stephen Booth
- Early Phase Clinical Trials Unit, Department of Oncology, University of Oxford, Oxford, UK
| | - Graham Collins
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| |
Collapse
|
157
|
Funck-Brentano E, Vizlin-Hodzic D, Nilsson JA, Nilsson LM. BET bromodomain inhibitor HMBA synergizes with MEK inhibition in treatment of malignant glioma. Epigenetics 2020; 16:54-63. [PMID: 32603264 PMCID: PMC7889204 DOI: 10.1080/15592294.2020.1786319] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
(1) Background: BET bromodomain proteins regulate transcription by binding acetylated histones and attracting key factors for, e.g., transcriptional elongation. BET inhibitors have been developed to block pathogenic processes such as cancer and inflammation. Despite having potent biological activities, BET inhibitors have still not made a breakthrough in clinical use for treating cancer. Multiple resistance mechanisms have been proposed but thus far no attempts to block this in glioma has been made. (2) Methods: Here, we have conducted a pharmacological synergy screen in glioma cells to search for possible combination treatments augmenting the apoptotic response to BET inhibitors. We first used HMBA, a compound that was developed as a differentiation therapy four decades ago but more recently was shown to primarily inhibit BET bromodomain proteins. Data was also generated using other BET inhibitors. (3) Results: In the synergy screen, we discovered that several MEK inhibitors can enhance apoptosis in response to HMBA in rat and human glioma cells in vitro as well as in vivo xenografts. The combination is not unique to HMBA but also other BET inhibitors such as JQ1 and I-BET-762 can synergize with MEK inhibitors. (4) Conclusions: Our findings validate a combination therapy previously demonstrated to exhibit anti-cancer activities in multiple other tumour types but which appears to have been lost in translation to the clinic.
Collapse
Affiliation(s)
- Elisa Funck-Brentano
- From Sahlgrenska Cancer Center, Department of Surgery, Institute of Clinical Sciences, University of Gothenburg , Gothenburg, Sweden
| | - Dzeneta Vizlin-Hodzic
- From Sahlgrenska Cancer Center, Department of Surgery, Institute of Clinical Sciences, University of Gothenburg , Gothenburg, Sweden
| | - Jonas A Nilsson
- From Sahlgrenska Cancer Center, Department of Surgery, Institute of Clinical Sciences, University of Gothenburg , Gothenburg, Sweden
| | - Lisa M Nilsson
- From Sahlgrenska Cancer Center, Department of Surgery, Institute of Clinical Sciences, University of Gothenburg , Gothenburg, Sweden
| |
Collapse
|
158
|
Brd2 haploinsufficiency extends lifespan and healthspan in C57B6/J mice. PLoS One 2020; 15:e0234910. [PMID: 32559200 PMCID: PMC7304595 DOI: 10.1371/journal.pone.0234910] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 06/04/2020] [Indexed: 11/19/2022] Open
Abstract
Aging in mammals is the gradual decline of an organism's physical, mental, and physiological capacity. Aging leads to increased risk for disease and eventually to death. Here, we show that Brd2 haploinsufficiency (Brd2+/-) extends lifespan and increases healthspan in C57B6/J mice. In Brd2+/- mice, longevity is increased by 23% (p<0.0001), and, relative to wildtype animals (Brd2+/+), cancer incidence is reduced by 43% (p<0.001). In addition, relative to age-matched wildtype mice, Brd2 heterozygotes show healthier aging including: improved grooming, extended period of fertility, and lack of age-related decline in kidney function and morphology. Our data support a role for haploinsufficiency of Brd2 in promoting healthy aging. We hypothesize that Brd2 affects aging by protecting against the accumulation of molecular and cellular damage. Given the recent advances in the development of BET inhibitors, our research provides impetus to test drugs that target BRD2 as a way to understand and treat/prevent age-related diseases.
Collapse
|
159
|
Unzue A, Jessen-Trefzer C, Spiliotopoulos D, Gaudio E, Tarantelli C, Dong J, Zhao H, Pachmayr J, Zahler S, Bernasconi E, Sartori G, Cascione L, Bertoni F, Śledź P, Caflisch A, Nevado C. Understanding the mechanism of action of pyrrolo[3,2- b]quinoxaline-derivatives as kinase inhibitors. RSC Med Chem 2020; 11:665-675. [PMID: 33479666 PMCID: PMC7557569 DOI: 10.1039/d0md00049c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/16/2020] [Indexed: 12/02/2022] Open
Abstract
Two novel quinoxaline-based EphA3 tyrosine kinase inhibitors have been designed and characterized in vivo in a relevant lymphoma model, showing high efficacy in the control of tumor size.
The X-ray structure of the catalytic domain of the EphA3 tyrosine kinase in complex with a previously reported type II inhibitor was used to design two novel quinoxaline derivatives, inspired by kinase inhibitors that have reached clinical development. These two new compounds were characterized by an array of cell-based assays and gene expression profiling experiments. A global chemical proteomics approach was used to generate the drug-protein interaction profile, which suggested suitable therapeutic indications. Both inhibitors, studied in the context of angiogenesis and in vivo in a relevant lymphoma model, showed high efficacy in the control of tumor size.
Collapse
Affiliation(s)
- Andrea Unzue
- Department of Chemistry , University of Zurich , Winterthurerstrasse 190 , CH-8057 , Zürich , Switzerland . ; ; Tel: (+41) 446353945
| | - Claudia Jessen-Trefzer
- Department of Chemistry , University of Zurich , Winterthurerstrasse 190 , CH-8057 , Zürich , Switzerland . ; ; Tel: (+41) 446353945
| | - Dimitrios Spiliotopoulos
- Department of Biochemistry , University of Zurich , Winterthurerstrasse 190 , CH-8057 , Zürich , Switzerland
| | - Eugenio Gaudio
- Institute of Oncology Research , Faculty of Biomedical Sciences , USI , Bellinzona , Switzerland
| | - Chiara Tarantelli
- Institute of Oncology Research , Faculty of Biomedical Sciences , USI , Bellinzona , Switzerland
| | - Jing Dong
- Department of Biochemistry , University of Zurich , Winterthurerstrasse 190 , CH-8057 , Zürich , Switzerland
| | - Hongtao Zhao
- Department of Biochemistry , University of Zurich , Winterthurerstrasse 190 , CH-8057 , Zürich , Switzerland
| | - Johanna Pachmayr
- Department of Pharmacy , University of Munich , Butenandstrasse 5-13 , 81377 Munich , Germany
| | - Stefan Zahler
- Department of Pharmacy , University of Munich , Butenandstrasse 5-13 , 81377 Munich , Germany
| | - Elena Bernasconi
- Institute of Oncology Research , Faculty of Biomedical Sciences , USI , Bellinzona , Switzerland
| | - Giulio Sartori
- Institute of Oncology Research , Faculty of Biomedical Sciences , USI , Bellinzona , Switzerland
| | - Luciano Cascione
- Institute of Oncology Research , Faculty of Biomedical Sciences , USI , Bellinzona , Switzerland.,SIB Swiss Institute of Bioinformatics , Lausanne , Switzerland
| | - Francesco Bertoni
- Institute of Oncology Research , Faculty of Biomedical Sciences , USI , Bellinzona , Switzerland.,Oncology Institute of Southern Switzerland (IOSI) , Bellinzona , Switzerland
| | - Paweł Śledź
- Department of Biochemistry , University of Zurich , Winterthurerstrasse 190 , CH-8057 , Zürich , Switzerland
| | - Amedeo Caflisch
- Department of Biochemistry , University of Zurich , Winterthurerstrasse 190 , CH-8057 , Zürich , Switzerland
| | - Cristina Nevado
- Department of Chemistry , University of Zurich , Winterthurerstrasse 190 , CH-8057 , Zürich , Switzerland . ; ; Tel: (+41) 446353945
| |
Collapse
|
160
|
Tian XP, Cai J, Ma SY, Fang Y, Huang HQ, Lin TY, Rao HL, Li M, Xia ZJ, Kang TB, Xie D, Cai QQ. BRD2 induces drug resistance through activation of the RasGRP1/Ras/ERK signaling pathway in adult T-cell lymphoblastic lymphoma. Cancer Commun (Lond) 2020; 40:245-259. [PMID: 32459053 PMCID: PMC7307265 DOI: 10.1002/cac2.12039] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/06/2020] [Accepted: 05/14/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Adult patients with T-cell lymphoblastic lymphoma (T-LBL) are treated with high-intensity chemotherapy regimens, but the response rate is still unsatisfactory because of frequent drug resistance. We aimed to investigate the potential mechanisms of drug resistance in adults with T-LBL. METHODS Gene expression microarray was used to identify differential mRNA expression profiles between chemotherapy-resistant and chemotherapy-sensitive adult T-LBL tissues. Real-time PCR and immunohistochemistry were performed to detect the expression of bromodomain-containing protein 2 (BRD2) and c-Myc in fresh-frozen T-LBL tissues from 85 adult patients. The Ras pull-down assay was performed to monitor Ras activation. Chromatin immunoprecipitation assays were used to analyze the binding of E2F transcription factor 1 (E2F1)/BRD2 to the RAS guanyl releasing protein 1 (RasGRP1) promoter region. The drug resistance effect and mechanism of BRD2 were determined by both in vivo and in vitro studies. RESULTS A total of 86 chemotherapy resistance-related genes in adult T-LBL were identified by gene expression microarray. Among them, BRD2 was upregulated in chemotherapy-resistant adult T-LBL tissues and associated with worse progression-free survival and overall survival of 85 adult T-LBL patients. Furthermore, BRD2 suppressed doxorubicin (Dox)-induced cell apoptosis both in vitro and in vivo. The activation of RasGRP1/Ras/ERK signaling might contribute to the Dox resistance effect of BRD2. Besides, OTX015, a bromodomain and extra-terminal (BET) inhibitor, reversed the Dox resistance effect of BRD2. Patient-derived tumor xenograft demonstrated that the sequential use of OTX015 after Dox showed superior therapeutic effects. CONCLUSIONS Our data showed that BRD2 promotes drug resistance in adult T-LBL through the RasGRP1/Ras/ERK signaling pathway. Targeting BRD2 may be a novel strategy to improve the therapeutic efficacy and prolong survival of adults with T-LBL.
Collapse
Affiliation(s)
- Xiao-Peng Tian
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China.,Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| | - Jun Cai
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| | - Shu-Yun Ma
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| | - Yu Fang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| | - Hui-Qiang Huang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| | - Tong-Yu Lin
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| | - Hui-Lan Rao
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| | - Mei Li
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| | - Zhong-Jun Xia
- Department of Hematology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| | - Tie-Bang Kang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| | - Dan Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| | - Qing-Qing Cai
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China.,Department of Medical Oncology, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, 510060, P. R. China
| |
Collapse
|
161
|
Guerra SL, Maertens O, Kuzmickas R, De Raedt T, Adeyemi RO, Guild CJ, Guillemette S, Redig AJ, Chambers ES, Xu M, Tiv H, Santagata S, Jänne PA, Elledge SJ, Cichowski K. A Deregulated HOX Gene Axis Confers an Epigenetic Vulnerability in KRAS-Mutant Lung Cancers. Cancer Cell 2020; 37:705-719.e6. [PMID: 32243838 PMCID: PMC10805385 DOI: 10.1016/j.ccell.2020.03.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/12/2019] [Accepted: 03/03/2020] [Indexed: 01/02/2023]
Abstract
While KRAS mutations are common in non-small cell lung cancer (NSCLC), effective treatments are lacking. Here, we report that half of KRAS-mutant NSCLCs aberrantly express the homeobox protein HOXC10, largely due to unappreciated defects in PRC2, which confers sensitivity to combined BET/MEK inhibitors in xenograft and PDX models. Efficacy of the combination is dependent on suppression of HOXC10 by BET inhibitors. We further show that HOXC10 regulates the expression of pre-replication complex (pre-RC) proteins in sensitive tumors. Accordingly, BET/MEK inhibitors suppress pre-RC proteins in cycling cells, triggering stalled replication, DNA damage, and death. These studies reveal a promising therapeutic strategy for KRAS-mutant NSCLCs, identify a predictive biomarker of response, and define a subset of NSCLCs with a targetable epigenetic vulnerability.
Collapse
Affiliation(s)
- Stephanie L Guerra
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Ophélia Maertens
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Ryan Kuzmickas
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Thomas De Raedt
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Richard O Adeyemi
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Caroline J Guild
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Shawna Guillemette
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Amanda J Redig
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Emily S Chambers
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Man Xu
- Belfer Center for Applied Cancer Science, Boston, MA 02115, USA
| | - Hong Tiv
- Experimental Therapeutic Core, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Sandro Santagata
- Harvard Medical School, Boston, MA 02115, USA; Departments of Pathology, Brigham and Women's Hospital, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Ludwig Center at Harvard, Harvard Medical School, Boston, MA 02115, USA
| | - Pasi A Jänne
- Harvard Medical School, Boston, MA 02115, USA; Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Belfer Center for Applied Cancer Science, Boston, MA 02115, USA
| | - Stephen J Elledge
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Ludwig Center at Harvard, Harvard Medical School, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; Howard Hughes Medical Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Karen Cichowski
- Genetics Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Harvard Medical School, Boston, MA 02115, USA; Ludwig Center at Harvard, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
162
|
Gilan O, Rioja I, Knezevic K, Bell MJ, Yeung MM, Harker NR, Lam EYN, Chung CW, Bamborough P, Petretich M, Urh M, Atkinson SJ, Bassil AK, Roberts EJ, Vassiliadis D, Burr ML, Preston AGS, Wellaway C, Werner T, Gray JR, Michon AM, Gobbetti T, Kumar V, Soden PE, Haynes A, Vappiani J, Tough DF, Taylor S, Dawson SJ, Bantscheff M, Lindon M, Drewes G, Demont EH, Daniels DL, Grandi P, Prinjha RK, Dawson MA. Selective targeting of BD1 and BD2 of the BET proteins in cancer and immunoinflammation. Science 2020; 368:387-394. [PMID: 32193360 PMCID: PMC7610820 DOI: 10.1126/science.aaz8455] [Citation(s) in RCA: 260] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 03/06/2020] [Indexed: 12/12/2022]
Abstract
The two tandem bromodomains of the BET (bromodomain and extraterminal domain) proteins enable chromatin binding to facilitate transcription. Drugs that inhibit both bromodomains equally have shown efficacy in certain malignant and inflammatory conditions. To explore the individual functional contributions of the first (BD1) and second (BD2) bromodomains in biology and therapy, we developed selective BD1 and BD2 inhibitors. We found that steady-state gene expression primarily requires BD1, whereas the rapid increase of gene expression induced by inflammatory stimuli requires both BD1 and BD2 of all BET proteins. BD1 inhibitors phenocopied the effects of pan-BET inhibitors in cancer models, whereas BD2 inhibitors were predominantly effective in models of inflammatory and autoimmune disease. These insights into the differential requirement of BD1 and BD2 for the maintenance and induction of gene expression may guide future BET-targeted therapies.
Collapse
Affiliation(s)
- Omer Gilan
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Inmaculada Rioja
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | - Kathy Knezevic
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Matthew J Bell
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | - Miriam M Yeung
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Nicola R Harker
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | - Enid Y N Lam
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Chun-Wa Chung
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | - Paul Bamborough
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | - Massimo Petretich
- Cellzome GmbH, Functional Genomics R&D, GlaxoSmithKline, Heidelberg, Germany
| | | | - Stephen J Atkinson
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | - Anna K Bassil
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | - Emma J Roberts
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | - Dane Vassiliadis
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Marian L Burr
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Alex G S Preston
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | | | - Thilo Werner
- Cellzome GmbH, Functional Genomics R&D, GlaxoSmithKline, Heidelberg, Germany
| | - James R Gray
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | - Anne-Marie Michon
- Cellzome GmbH, Functional Genomics R&D, GlaxoSmithKline, Heidelberg, Germany
| | - Thomas Gobbetti
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | - Vinod Kumar
- Computational Biology, GlaxoSmithKline, Collegeville, PA, USA
| | - Peter E Soden
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | - Andrea Haynes
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | - Johanna Vappiani
- Cellzome GmbH, Functional Genomics R&D, GlaxoSmithKline, Heidelberg, Germany
| | - David F Tough
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | - Simon Taylor
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | - Sarah-Jane Dawson
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| | - Marcus Bantscheff
- Cellzome GmbH, Functional Genomics R&D, GlaxoSmithKline, Heidelberg, Germany
| | - Matthew Lindon
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | - Gerard Drewes
- Cellzome GmbH, Functional Genomics R&D, GlaxoSmithKline, Heidelberg, Germany
| | - Emmanuel H Demont
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK
| | | | - Paola Grandi
- Cellzome GmbH, Functional Genomics R&D, GlaxoSmithKline, Heidelberg, Germany
| | - Rab K Prinjha
- Epigenetics RU, GlaxoSmithKline Medicines Research Centre, Stevenage, UK.
| | - Mark A Dawson
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Centre for Cancer Research, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
163
|
Dzobo K. Epigenomics-Guided Drug Development: Recent Advances in Solving the Cancer Treatment "jigsaw puzzle". OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2020; 23:70-85. [PMID: 30767728 DOI: 10.1089/omi.2018.0206] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The human epigenome plays a key role in determining cellular identity and eventually function. Drug discovery undertakings have focused mainly on the role of genomics in carcinogenesis, with the focus turning to the epigenome recently. Drugs targeting DNA and histone modifications are under development with some such as 5-azacytidine, decitabine, vorinostat, and panobinostat already approved by the Food and Drug Administration (FDA) and the European Medicines Agency (EMA). This expert review offers a critical analysis of the epigenomics-guided drug discovery and development and the opportunities and challenges for the next decade. Importantly, the coupling of epigenetic editing techniques, such as clustered regularly interspersed short palindromic repeat (CRISPR)-CRISPR-associated protein-9 (Cas9) and APOBEC-coupled epigenetic sequencing (ACE-seq) with epigenetic drug screens, will allow the identification of small-molecule inhibitors or drugs able to reverse epigenetic changes responsible for many diseases. In addition, concrete and sustainable innovation in cancer treatment ought to integrate epigenome targeting drugs with classic therapies such as chemotherapy and immunotherapy.
Collapse
Affiliation(s)
- Kevin Dzobo
- 1 International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Cape Town, South Africa.,2 Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
164
|
Piha-Paul SA, Hann CL, French CA, Cousin S, Braña I, Cassier PA, Moreno V, de Bono JS, Harward SD, Ferron-Brady G, Barbash O, Wyce A, Wu Y, Horner T, Annan M, Parr NJ, Prinjha RK, Carpenter CL, Hilton J, Hong DS, Haas NB, Markowski MC, Dhar A, O’Dwyer PJ, Shapiro GI. Phase 1 Study of Molibresib (GSK525762), a Bromodomain and Extra-Terminal Domain Protein Inhibitor, in NUT Carcinoma and Other Solid Tumors. JNCI Cancer Spectr 2020; 4:pkz093. [PMID: 32328561 PMCID: PMC7165800 DOI: 10.1093/jncics/pkz093] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Revised: 08/21/2019] [Accepted: 10/31/2019] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Bromodomain and extra-terminal domain proteins are promising epigenetic anticancer drug targets. This first-in-human study evaluated the safety, recommended phase II dose, pharmacokinetics, pharmacodynamics, and preliminary antitumor activity of the bromodomain and extra-terminal domain inhibitor molibresib (GSK525762) in patients with nuclear protein in testis (NUT) carcinoma (NC) and other solid tumors. METHODS This was a phase I and II, open-label, dose-escalation study. Molibresib was administered orally once daily. Single-patient dose escalation (from 2 mg/d) was conducted until the first instance of grade 2 or higher drug-related toxicity, followed by a 3 + 3 design. Pharmacokinetic parameters were obtained during weeks 1 and 3. Circulating monocyte chemoattractant protein-1 levels were measured as a pharmacodynamic biomarker. RESULTS Sixty-five patients received molibresib. During dose escalation, 11% experienced dose-limiting toxicities, including six instances of grade 4 thrombocytopenia, all with molibresib 60-100 mg. The most frequent treatment-related adverse events of any grade were thrombocytopenia (51%) and gastrointestinal events, including nausea, vomiting, diarrhea, decreased appetite, and dysgeusia (22%-42%), anemia (22%), and fatigue (20%). Molibresib demonstrated an acceptable safety profile up to 100 mg; 80 mg once daily was selected as the recommended phase II dose. Following single and repeat dosing, molibresib showed rapid absorption and elimination (maximum plasma concentration: 2 hours; t1/2: 3-7 hours). Dose-dependent reductions in circulating monocyte chemoattractant protein-1 levels were observed. Among 19 patients with NC, four achieved either confirmed or unconfirmed partial response, eight had stable disease as best response, and four were progression-free for more than 6 months. CONCLUSIONS Once-daily molibresib was tolerated at doses demonstrating target engagement. Preliminary data indicate proof-of-concept in NC.
Collapse
Affiliation(s)
| | | | - Christopher A French
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| | - Sophie Cousin
- Medical Oncology, Institute Bergonié, Bordeaux, France
| | - Irene Braña
- Medical Oncology Department, Vall d’Hebron University Hospital, Barcelona, Spain
| | | | - Victor Moreno
- Medical Oncology, START Madrid-FJD, Fundación Jiménez Díaz Hospital, Madrid, Spain
| | - Johann S de Bono
- The Institute of Cancer Research and Royal Marsden Hospital, London, UK
| | | | | | | | | | | | | | | | | | - Rabinder K Prinjha
- Division of Medical Oncology, Ottawa Hospital Cancer Centre, Ottawa, ON, Canada
| | | | - John Hilton
- Division of Medical Oncology, Ottawa Hospital Cancer Centre, Ottawa, ON, Canada
| | | | - Naomi B Haas
- Abramson Cancer Center at University of Pennsylvania, Philadelphia, PA
| | | | | | | | - Geoffrey I Shapiro
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
165
|
Martin-Romano P, Baldini C, Postel-Vinay S. How Much Can We Bet on Activity of BET Inhibitors Beyond NUT-Midline Carcinoma? JNCI Cancer Spectr 2020; 4:pkz092. [PMID: 32328560 PMCID: PMC7165801 DOI: 10.1093/jncics/pkz092] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 10/29/2019] [Accepted: 10/31/2019] [Indexed: 11/13/2022] Open
Affiliation(s)
- Patricia Martin-Romano
- DITEP (Département d’Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif, France
| | - Capucine Baldini
- DITEP (Département d’Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif, France
| | - Sophie Postel-Vinay
- DITEP (Département d’Innovations Thérapeutiques et Essais Précoces), Gustave Roussy, Villejuif, France
- Université Paris Saclay, Université Paris-Sud, Faculté de médicine, Le Kremlin Bicêtre, France
- ATIP-Avenir group, Inserm Unit U981, Gustave Roussy, Villejuif, France
| |
Collapse
|
166
|
Moreno V, Sepulveda JM, Vieito M, Hernández-Guerrero T, Doger B, Saavedra O, Ferrero O, Sarmiento R, Arias M, De Alvaro J, Di Martino J, Zuraek M, Sanchez-Pérez T, Aronchik I, Filvaroff EH, Lamba M, Hanna B, Nikolova Z, Braña I. Phase I study of CC-90010, a reversible, oral BET inhibitor in patients with advanced solid tumors and relapsed/refractory non-Hodgkin's lymphoma. Ann Oncol 2020; 31:780-788. [PMID: 32240793 DOI: 10.1016/j.annonc.2020.03.294] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 02/13/2020] [Accepted: 03/18/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Bromodomain and extra-terminal (BET) proteins are epigenetic readers that regulate expression of genes involved in oncogenesis. CC-90010 is a novel, oral, reversible, small-molecule BET inhibitor. PATIENTS AND METHODS CC-90010-ST-001 (NCT03220347; 2015-004371-79) is a phase I dose-escalation and expansion study of CC-90010 in patients with advanced or unresectable solid tumors and relapsed/refractory (R/R) non-Hodgkin's lymphoma (NHL). We report results from the dose escalation phase, which explored 11 dose levels and four dosing schedules, two weekly (2 days on/5 days off; 3 days on/4 days off), one biweekly (3 days on/11 days off), and one monthly (4 days on/24 days off). The primary objectives were to determine the safety, maximum tolerated dose (MTD) and/or recommended phase II dose (RP2D) and schedule. Secondary objectives were to evaluate signals of early antitumor activity, pharmacokinetics, and pharmacodynamics. RESULTS This study enrolled 69 patients, 67 with solid tumors and two with diffuse large B-cell lymphoma (DLBCL). The median age was 57 years (range, 21-80) and the median number of prior regimens was four (range, 1-9). Treatment-related adverse events (TRAEs) were mostly mild and manageable; grade 3/4 TRAEs reported in more than two patients were thrombocytopenia (13%), anemia, and fatigue (4% each). Six patients had dose-limiting toxicities. MTDs were 15 mg (2 days on/5 days off), 30 mg (3 days on/11 days off), and 45 mg (4 days on/24 days off). The RP2D and schedule selected for expansion was 45 mg (4 days on/24 days off). As of 8 October 2019, one patient with grade 2 astrocytoma achieved a complete response, one patient with endometrial carcinoma had a partial response, and six patients had prolonged stable disease ≥11 months. CONCLUSIONS CC-90010 is well tolerated, with single-agent activity in patients with heavily pretreated, advanced solid tumors.
Collapse
Affiliation(s)
- V Moreno
- START Madrid-FJD, Hospital Fundación Jimenez Diaz, Madrid, Spain.
| | - J M Sepulveda
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - M Vieito
- Department of Gene Expression and Cancer, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - B Doger
- START Madrid-FJD, Hospital Fundación Jimenez Diaz, Madrid, Spain
| | - O Saavedra
- Department of Gene Expression and Cancer, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - O Ferrero
- START Madrid-FJD, Hospital Fundación Jimenez Diaz, Madrid, Spain
| | - R Sarmiento
- Celgene Institute for Translational Research Europe, a Bristol Myers Squibb Company, Seville, Spain
| | - M Arias
- Celgene Institute for Translational Research Europe, a Bristol Myers Squibb Company, Seville, Spain
| | - J De Alvaro
- Celgene Institute for Translational Research Europe, a Bristol Myers Squibb Company, Seville, Spain
| | | | - M Zuraek
- Bristol Myers Squibb, San Francisco, USA
| | - T Sanchez-Pérez
- Celgene Institute for Translational Research Europe, a Bristol Myers Squibb Company, Seville, Spain
| | - I Aronchik
- Bristol Myers Squibb, San Francisco, USA
| | | | - M Lamba
- Bristol Myers Squibb, Summit, USA
| | - B Hanna
- Bristol Myers Squibb, Summit, USA
| | - Z Nikolova
- Celgene Institute for Translational Research Europe, a Bristol Myers Squibb Company, Seville, Spain
| | - I Braña
- Department of Gene Expression and Cancer, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| |
Collapse
|
167
|
Role of BET Inhibitors in Triple Negative Breast Cancers. Cancers (Basel) 2020; 12:cancers12040784. [PMID: 32218352 PMCID: PMC7226117 DOI: 10.3390/cancers12040784] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 12/20/2022] Open
Abstract
Bromodomain and extraterminal domain (BET) proteins have evolved as key multifunctional super-regulators that control gene expression. These proteins have been shown to upregulate transcriptional machinery leading to over expression of genes involved in cell proliferation and carcinogenesis. Based on favorable preclinical evidence of BET inhibitors in various cancer models; currently, 26 clinical trials are underway in various stages of study on various hematological and solid organ cancers. Unfortunately, preliminary evidence for these clinical studies does not support the application of BET inhibitors as monotherapy in cancer treatment. Furthermore, the combinatorial efficiency of BET inhibitors with other chemo-and immunotherapeutic agents remain elusive. In this review, we will provide a concise summary of the molecular basis and preliminary clinical outcomes of BET inhibitors in cancer therapy, with special focus on triple negative breast cancer.
Collapse
|
168
|
Camero S, Camicia L, Marampon F, Ceccarelli S, Shukla R, Mannarino O, Pizer B, Schiavetti A, Pizzuti A, Tombolini V, Marchese C, Dominici C, Megiorni F. BET inhibition therapy counteracts cancer cell survival, clonogenic potential and radioresistance mechanisms in rhabdomyosarcoma cells. Cancer Lett 2020; 479:71-88. [PMID: 32200036 DOI: 10.1016/j.canlet.2020.03.011] [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: 01/17/2020] [Revised: 03/04/2020] [Accepted: 03/10/2020] [Indexed: 12/15/2022]
Abstract
The antitumour effects of OTX015, a first-in-class BET inhibitor (BETi), were investigated as a single agent or in combination with ionizing radiation (IR) in preclinical in vitro models of rhabdomyosarcoma (RMS), the most common childhood soft tissue sarcoma. Herein, we demonstrated the upregulation of BET Bromodomain gene expression in RMS tumour biopsies and cell lines compared to normal skeletal muscle. In vitro experiments showed that OTX015 significantly reduced RMS cell proliferation by altering cell cycle modulators and apoptotic related proteins due to the accumulation of DNA breaks that cells are unable to repair. Interestingly, OTX015 also impaired migration capacity and tumour-sphere architecture by downregulating pro-stemness genes and was able to potentiate ionizing radiation effects by reducing the expression of different drivers of tumour dissemination and resistance mechanisms, including the GNL3 gene, that we correlated for the first time with the RMS phenotype. In conclusion, our research sheds further light on the molecular events of OTX015 action against RMS cells and indicates this novel BETi as an effective option to improve therapeutic strategies and overcome the development of resistant cancer cells in patients with RMS.
Collapse
Affiliation(s)
- Simona Camero
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Lucrezia Camicia
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Francesco Marampon
- Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Simona Ceccarelli
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | - Rajeev Shukla
- Department of Perinatal and Paediatric Pathology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK.
| | - Olga Mannarino
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Barry Pizer
- Department of Oncology, Alder Hey Children's NHS Foundation Trust, Eaton Road, Liverpool, L12 2AP, UK.
| | - Amalia Schiavetti
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Antonio Pizzuti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | - Vincenzo Tombolini
- Department of Radiological, Oncological and Pathological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Cinzia Marchese
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | - Carlo Dominici
- Department of Maternal, Infantile, and Urological Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Francesca Megiorni
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| |
Collapse
|
169
|
Katavolos P, Cain G, Farman C, Romero FA, Magnuson S, Ly JQ, Choo EF, Katakam AK, Andaya R, Maher J. Preclinical Safety Assessment of a Highly Selective and Potent Dual Small-Molecule Inhibitor of CBP/P300 in Rats and Dogs. Toxicol Pathol 2020; 48:465-480. [PMID: 32124659 DOI: 10.1177/0192623319898469] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cyclic adenosine monophosphate-response element (CREB)-binding protein (CBP) and EP300E1A-binding protein (p300) are members of the bromodomain and extraterminal motif (BET) family. These highly homologous proteins have a key role in modulating transcription, including altering the status of chromatin or through interactions with or posttranslational modifications of transcription factors. As CBP and p300 have known roles for stimulating c-Myc oncogenic activity, a small-molecule inhibitor, GNE-781, was developed to selectively and potently inhibit the CBP/p300 bromodomains (BRDs). Genetic models have been challenging to develop due to embryonic lethality arising from germline homozygous mutations in either CBP or P300. Hence, the purpose of this study was to characterize the role of dual inhibition of these proteins in adult rats and dogs. Repeat dose toxicity studies were conducted, and toxicologic and pathologic end points were assessed. GNE-781 was generally tolerated; however, marked effects on thrombopoiesis occurred in both species. Evidence of inhibition of erythroid, granulocytic, and lymphoid cell differentiation was also present, as well as deleterious changes in gastrointestinal and reproductive tissues. These findings are consistent with many preclinical (and clinical) effects reported with BET inhibitors targeting BRD proteins; thus, the current study findings indicate a likely important role for CBP/p300 in stem cell differentiation.
Collapse
Affiliation(s)
- Paula Katavolos
- Safety Assessment, Genentech, Inc, South San Francisco, CA, USA
| | - Gary Cain
- Safety Assessment, Genentech, Inc, South San Francisco, CA, USA
| | - Cindy Farman
- Safety Assessment, Genentech, Inc, South San Francisco, CA, USA.,Vet Path Services, Inc, Mason, OH, USA
| | - F Anthony Romero
- Medicinal Chemistry, Genentech, Inc, South San Francisco, CA, USA.,Terns Pharmaceuticals, San Mateo, CA, USA
| | - Steven Magnuson
- Medicinal Chemistry, Genentech, Inc, South San Francisco, CA, USA
| | - Justin Q Ly
- Department of Drug Metabolism and Pharmacology, Genentech, Inc, South San Francisco, CA, USA
| | - Edna F Choo
- Department of Drug Metabolism and Pharmacology, Genentech, Inc, South San Francisco, CA, USA
| | | | - Roxanne Andaya
- Safety Assessment, Genentech, Inc, South San Francisco, CA, USA
| | - Jonathan Maher
- Safety Assessment, Genentech, Inc, South San Francisco, CA, USA
| |
Collapse
|
170
|
Werner MT, Wang H, Hamagami N, Hsu SC, Yano JA, Stonestrom AJ, Behera V, Zong Y, Mackay JP, Blobel GA. Comparative structure-function analysis of bromodomain and extraterminal motif (BET) proteins in a gene-complementation system. J Biol Chem 2020; 295:1898-1914. [PMID: 31792058 PMCID: PMC7029111 DOI: 10.1074/jbc.ra119.010679] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/17/2019] [Indexed: 11/06/2022] Open
Abstract
The widely expressed bromodomain and extraterminal motif (BET) proteins bromodomain-containing protein 2 (BRD2), BRD3, and BRD4 are multifunctional transcriptional regulators that bind acetylated chromatin via their conserved tandem bromodomains. Small molecules that target BET bromodomains are being tested for various diseases but typically do not discern between BET family members. Genomic distributions and protein partners of BET proteins have been described, but the basis for differences in BET protein function within a given lineage remains unclear. By establishing a gene knockout-rescue system in a Brd2-null erythroblast cell line, here we compared a series of mutant and chimeric BET proteins for their ability to modulate cell growth, differentiation, and gene expression. We found that the BET N-terminal halves bearing the bromodomains convey marked differences in protein stability but do not account for specificity in BET protein function. Instead, when BET proteins were expressed at comparable levels, their specificity was largely determined by the C-terminal half. Remarkably, a chimeric BET protein comprising the N-terminal half of the structurally similar short BRD4 isoform (BRD4S) and the C-terminal half of BRD2 functioned similarly to intact BRD2. We traced part of the BRD2-specific activity to a previously uncharacterized short segment predicted to harbor a coiled-coil (CC) domain. Deleting the CC segment impaired BRD2's ability to restore growth and differentiation, and the CC region functioned in conjunction with the adjacent ET domain to impart BRD2-like activity onto BRD4S. In summary, our results identify distinct BET protein domains that regulate protein turnover and biological activities.
Collapse
Affiliation(s)
- Michael T Werner
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104.
| | - Hongxin Wang
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104
| | - Nicole Hamagami
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104
| | - Sarah C Hsu
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Jennifer A Yano
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104
| | - Aaron J Stonestrom
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Vivek Behera
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Yichen Zong
- School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Joel P Mackay
- School of Life and Environmental Sciences, University of Sydney, Sydney, New South Wales 2006, Australia
| | - Gerd A Blobel
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104.
| |
Collapse
|
171
|
Nguyen MV, Loof L, Falchook GS. Bromodomain and Extra-Terminal (BET) Domain Protein Inhibitors for Solid Tumor Cancers. ACTA ACUST UNITED AC 2020; 3:16-22. [PMID: 35756176 PMCID: PMC9208386 DOI: 10.4103/jipo.jipo_2_20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/02/2022]
Abstract
The bromodomain and extraterminal (BET) domain protein family is involved in the process of transcription of genetic information. The BET protein family includes BRD2, BRD3, BRD4, and bromodomain testis-specific protein. BET protein alterations are associated with some solid tumor cancers, including nuclear protein in testis midline carcinoma. BET protein has a role in carcinogenesis and in the regulation of the cell cycle. A number of BET inhibitors have entered clinical trials. This review discusses the results of BET inhibitor clinical trials in solid tumor cancers.
Collapse
Affiliation(s)
| | - Lydia Loof
- Sarah Cannon Research Institute at HealthONE, Denver, CO, USA
| | | |
Collapse
|
172
|
Selective inhibition of the BD2 bromodomain of BET proteins in prostate cancer. Nature 2020; 578:306-310. [PMID: 31969702 DOI: 10.1038/s41586-020-1930-8] [Citation(s) in RCA: 245] [Impact Index Per Article: 61.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 11/25/2019] [Indexed: 12/24/2022]
Abstract
Proteins of the bromodomain and extra-terminal (BET) domain family are epigenetic readers that bind acetylated histones through their bromodomains to regulate gene transcription. Dual-bromodomain BET inhibitors (DbBi) that bind with similar affinities to the first (BD1) and second (BD2) bromodomains of BRD2, BRD3, BRD4 and BRDt have displayed modest clinical activity in monotherapy cancer trials. A reduced number of thrombocytes in the blood (thrombocytopenia) as well as symptoms of gastrointestinal toxicity are dose-limiting adverse events for some types of DbBi1-5. Given that similar haematological and gastrointestinal defects were observed after genetic silencing of Brd4 in mice6, the platelet and gastrointestinal toxicities may represent on-target activities associated with BET inhibition. The two individual bromodomains in BET family proteins may have distinct functions7-9 and different cellular phenotypes after pharmacological inhibition of one or both bromodomains have been reported10,11, suggesting that selectively targeting one of the bromodomains may result in a different efficacy and tolerability profile compared with DbBi. Available compounds that are selective to individual domains lack sufficient potency and the pharmacokinetics properties that are required for in vivo efficacy and tolerability assessment10-13. Here we carried out a medicinal chemistry campaign that led to the discovery of ABBV-744, a highly potent and selective inhibitor of the BD2 domain of BET family proteins with drug-like properties. In contrast to the broad range of cell growth inhibition induced by DbBi, the antiproliferative activity of ABBV-744 was largely, but not exclusively, restricted to cell lines of acute myeloid leukaemia and prostate cancer that expressed the full-length androgen receptor (AR). ABBV-744 retained robust activity in prostate cancer xenografts, and showed fewer platelet and gastrointestinal toxicities than the DbBi ABBV-07514. Analyses of RNA expression and chromatin immunoprecipitation followed by sequencing revealed that ABBV-744 displaced BRD4 from AR-containing super-enhancers and inhibited AR-dependent transcription, with less impact on global transcription compared with ABBV-075. These results underscore the potential value of selectively targeting the BD2 domain of BET family proteins for cancer therapy.
Collapse
|
173
|
Mancarella C, Scotlandi K. IGF2BP3 From Physiology to Cancer: Novel Discoveries, Unsolved Issues, and Future Perspectives. Front Cell Dev Biol 2020; 7:363. [PMID: 32010687 PMCID: PMC6974587 DOI: 10.3389/fcell.2019.00363] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/12/2019] [Indexed: 12/24/2022] Open
Abstract
RNA network control is a key aspect of proper cellular homeostasis. In this context, RNA-binding proteins (RBPs) play a major role as regulators of the RNA life cycle due to their capability to bind to RNA sequences and precisely direct nuclear export, translation/degradation rates, and the intracellular localization of their target transcripts. Alterations in RBP expression or functions result in aberrant RNA translation and may drive the emergence and progression of several pathological conditions, including cancer. Among the RBPs, insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) is of particular interest in tumorigenesis and tumor progression. This review highlights the molecular mechanisms underlying the oncogenic functions of IGF2BP3, summarizes the therapeutic potential related to its inhibition and notes the fundamental issues that remain unanswered. To fully exploit IGF2BP3 for tumor diagnosis and therapy, it is crucial to dissect the mechanisms governing IGF2BP3 re-expression and to elucidate the complex interactions between IGF2BP3 and its target mRNAs as normal cells become tumor cells.
Collapse
Affiliation(s)
- Caterina Mancarella
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Katia Scotlandi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| |
Collapse
|
174
|
Bechter O, Schöffski P. Make your best BET: The emerging role of BET inhibitor treatment in malignant tumors. Pharmacol Ther 2020; 208:107479. [PMID: 31931101 DOI: 10.1016/j.pharmthera.2020.107479] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 11/15/2019] [Indexed: 12/17/2022]
Abstract
Bromodomains are protein-protein interaction modules with a great diversity in terms of number of proteins and their function. The bromodomain and extraterminal protein (BET) represents a distinct subclass of bromodomain proteins mainly involved in transcriptional regulation via their interaction with acetylated chromatin. In cancer cells BET proteins are found to be altered in many ways such as overexpression, mutations and fusions of BET proteins or their interference with cancer relevant signaling pathways and transcriptional programs in order to sustain cancer growth and viability. Blocking BET protein function with small molecules is associated with therapeutic activity. Consequently, a variety of small molecules have been developed and a number of phase I clinical trials have explored their tolerability and efficacy in patients with solid tumors and hematological malignancies. We will review the rational for applying BET inhibitors in the clinic and we will discuss the toxicity profile as well as efficacy of this new class of protein inhibitors. We will also highlight the emerging problem of treatment resistance and the potential these drugs might have when combined with other anti-cancer therapies.
Collapse
Affiliation(s)
- Oliver Bechter
- Leuven Cancer Institute, Department of General Medical Oncology, University Hospitals Leuven, Belgium; Department of Oncology, KU, Leuven, Belgium.
| | - Patrick Schöffski
- Leuven Cancer Institute, Department of General Medical Oncology, University Hospitals Leuven, Belgium; Department of Oncology, KU, Leuven, Belgium.
| |
Collapse
|
175
|
Phillipou AN, Lay CS, Carver CE, Messenger C, Evans JP, Lewis AJ, Gordon LJ, Mahmood M, Greenhough LA, Sammon D, Cheng AT, Chakraborty S, Jones EJ, Lucas SCC, Gatfield KM, Brierley DJ, Craggs PD. Cellular Target Engagement Approaches to Monitor Epigenetic Reader Domain Interactions. SLAS DISCOVERY 2019; 25:163-175. [PMID: 31875412 DOI: 10.1177/2472555219896278] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Malfunctions in the basic epigenetic mechanisms such as histone modifications, DNA methylation, and chromatin remodeling are implicated in a number of cancers and immunological and neurodegenerative conditions. Within GlaxoSmithKline (GSK) we have utilized a number of variations of the NanoBRET technology for the direct measurement of compound-target engagement within native cellular environments to drive high-throughput, routine structure-activity relationship (SAR) profiling across differing epigenetic targets. NanoBRET is a variation of the bioluminescence resonance energy transfer (BRET) methodology utilizing proteins of interest fused to either NanoLuc, a small, high-emission-intensity luciferase, or HaloTag, a modified dehalogenase enzyme that can be selectively labeled with a fluorophore. The combination of these two technologies has enabled the application of NanoBRET to biological systems such as epigenetic protein-protein interactions, which have previously been challenging. By synergizing target engagement assays with more complex primary cell phenotypic assays, we have been able to demonstrate compound-target selectivity profiles to enhance cellular potency and offset potential liability risks. Additionally, we have shown that in the absence of a robust, cell phenotypic assay, it is possible to utilize NanoBRET target engagement assays to aid chemistry in progressing at a higher scale than would have otherwise been achievable. The NanoBRET target engagement assays utilized have further shown an excellent correlation with more reductionist biochemical and biophysical assay systems, clearly demonstrating the possibility of using such assay systems at scale, in tandem with, or in preference to, lower-throughput cell phenotypic approaches.
Collapse
Affiliation(s)
- Alexander N Phillipou
- Medicine Design, Medicinal Science and Technology, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Charles S Lay
- Medicine Design, Medicinal Science and Technology, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Charlotte E Carver
- Medicine Design, Medicinal Science and Technology, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Cassie Messenger
- Medicine Design, Medicinal Science and Technology, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - John P Evans
- Medicine Design, Medicinal Science and Technology, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Antonia J Lewis
- Medicine Design, Medicinal Science and Technology, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Laurie J Gordon
- Medicine Design, Medicinal Science and Technology, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Mahnoor Mahmood
- Medicine Design, Medicinal Science and Technology, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Luke A Greenhough
- Medicine Design, Medicinal Science and Technology, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Douglas Sammon
- Medicine Design, Medicinal Science and Technology, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Aaron T Cheng
- Functional Genomics, Medicinal Science and Technology, GlaxoSmithKline, Collegeville, PA, USA
| | - Syandan Chakraborty
- Functional Genomics, Medicinal Science and Technology, GlaxoSmithKline, Collegeville, PA, USA
| | - Emma J Jones
- Medicine Design, Medicinal Science and Technology, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Simon C C Lucas
- Epigenetics Research Unit, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Kelly M Gatfield
- Medicine Design, Medicinal Science and Technology, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - David J Brierley
- Medicine Design, Medicinal Science and Technology, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| | - Peter D Craggs
- Medicine Design, Medicinal Science and Technology, GlaxoSmithKline, Stevenage, Hertfordshire, UK
| |
Collapse
|
176
|
The BET inhibitor GS-5829 targets chronic lymphocytic leukemia cells and their supportive microenvironment. Leukemia 2019; 34:1588-1598. [PMID: 31862959 PMCID: PMC7272263 DOI: 10.1038/s41375-019-0682-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 11/18/2019] [Accepted: 12/05/2019] [Indexed: 12/11/2022]
Abstract
Despite major improvements in treatment outcome with novel targeted therapies, such as the Bruton tyrosine kinase (BTK) inhibitor ibrutinib, chronic lymphocytic leukemia (CLL) remains incurable in the majority of patients. Activation of PI3K, NF-κB, and/or MYC has been linked to residual disease and/or resistance in ibrutinib-treated patients. These pathways can be targeted by inhibitors of bromodomain and extra-terminal (BET) proteins. Here we report about the preclinical activity of GS-5829, a novel BET inhibitor, in CLL. GS-5829 inhibited CLL cell proliferation and induced leukemia cell apoptosis through deregulation of key signaling pathways, such as BLK, AKT, ERK1/2, and MYC. IκBα modulation indicates that GS-5829 also inhibited NF-κB signaling. GS-5829-induced apoptosis resulted from an imbalance between positive (BIM) and negative regulators (BCL-XL) of the intrinsic apoptosis pathway. The antileukemia activity of GS-5829 increased synergistically in combinations with B-cell receptor signaling inhibitors, the BTK inhibitor ibrutinib, the PI3Kδ inhibitor idelalisib, and the SYK inhibitor entospletinib. In cocultures that mimic the lymph node microenvironment, GS-5829 inhibited signaling pathways within nurselike cells and their growth, indicating that BET inhibitors also can target the supportive CLL microenvironment. Collectively, these data provide a rationale for the clinical evaluation of BET inhibitors in CLL.
Collapse
|
177
|
Challenges and Opportunities for High-grade B-Cell Lymphoma With MYC and BCL2 and/or BCL6 Rearrangement (Double-hit Lymphoma). Am J Clin Oncol 2019; 42:304-316. [PMID: 29419530 DOI: 10.1097/coc.0000000000000427] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The most common subtype of non-Hodgkin lymphoma, diffuse large B-cell lymphoma, is cured in approximately two thirds of patients after initial therapy. The remaining one-third of patients who suffer relapse or become refractory have very poor survival outcomes despite salvage chemotherapy with or without stem cell transplantation. A considerable proportion of relapsed or refractory large B cells belong to the WHO subtype known as high-grade B-cell lymphoma with rearrangement of MYC and BCL2 and/or BCL6, also known as double-hit lymphoma (DHL). Most DHL patients present with Ann Arbor's stage III/IV, a comparatively higher rate of extranodal involvement including bone marrow and central nervous system infiltration, high levels of lactate dehydrogenase, and an elevated Ki67 expression in the tumor cells. Newer therapeutic approaches, including targeted therapy against BCL2, MYC, or other associated pathways, are needed. In addition, recent therapies that harness the immune system, such as checkpoint inhibitors and chimeric antigen receptor T-cell therapy, are changing the paradigm of treatment for non-Hodgkin lymphoma and could impact the outcome of DHL.
Collapse
|
178
|
Santos-de-Frutos K, Segrelles C, Lorz C. Hippo Pathway and YAP Signaling Alterations in Squamous Cancer of the Head and Neck. J Clin Med 2019; 8:jcm8122131. [PMID: 31817001 PMCID: PMC6947155 DOI: 10.3390/jcm8122131] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/26/2019] [Accepted: 11/29/2019] [Indexed: 12/20/2022] Open
Abstract
Head and neck cancer affects the upper aerodigestive tract and is the sixth leading cancer worldwide by incidence and the seventh by cause of death. Despite significant advances in surgery and chemotherapy, molecularly targeted therapeutic options for this type of cancer are scarce and long term survival rates remain low. Recently, comprehensive genomic studies have highlighted the most commonly altered genes and signaling pathways in this cancer. The Hippo-YAP pathway has been identified as a key oncogenic pathway in multiple tumors. Expression of genes controlled by the Hippo downstream transcriptional coactivators YAP (Yes-associated protein 1) and TAZ (WWTR1, WW domain containing transcription regulator 1) is widely deregulated in human cancer including head and neck squamous cell carcinoma (HNSCC). Interestingly, YAP/TAZ signaling might not be as essential for the normal homeostasis of adult tissues as for oncogenic growth, altogether making the pathway an amenable therapeutic target in cancer. Recent advances in the role of Hippo-YAP pathway in HNSCC have provided evidence that genetic alterations frequent in this type of cancer such as PIK3CA (phosphatidylinositide 3-kinase catalytic subunit alpha) overexpression or FAT1 (FAT atypical cadherin 1) functional loss can result in YAP activation. We discuss current therapeutic options targeting this pathway which are currently in use for other tumor types.
Collapse
Affiliation(s)
- Karla Santos-de-Frutos
- Molecular Oncology Unit, CIEMAT (ed 70A), Ave Complutense 40, 28040 Madrid, Spain; (K.S.-d.-F.); (C.S.)
- Molecular Oncology, Research Institute 12 de Octubre i+12, University Hospital 12 de Octubre, Ave Córdoba s/n, 28041 Madrid, Spain
| | - Carmen Segrelles
- Molecular Oncology Unit, CIEMAT (ed 70A), Ave Complutense 40, 28040 Madrid, Spain; (K.S.-d.-F.); (C.S.)
- Molecular Oncology, Research Institute 12 de Octubre i+12, University Hospital 12 de Octubre, Ave Córdoba s/n, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Ave Monforte de Lemos 3-5, 28029 Madrid, Spain
| | - Corina Lorz
- Molecular Oncology Unit, CIEMAT (ed 70A), Ave Complutense 40, 28040 Madrid, Spain; (K.S.-d.-F.); (C.S.)
- Molecular Oncology, Research Institute 12 de Octubre i+12, University Hospital 12 de Octubre, Ave Córdoba s/n, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Ave Monforte de Lemos 3-5, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-91-4962-521; Fax: +34-91-3466-484
| |
Collapse
|
179
|
Phuoc V, Sandoval-Sus J, Chavez JC. Drug therapy for double-hit lymphoma. Drugs Context 2019; 8:dic-8-2019-8-1. [PMID: 31844420 PMCID: PMC6905641 DOI: 10.7573/dic.2019-8-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/15/2019] [Accepted: 10/17/2019] [Indexed: 12/19/2022] Open
Abstract
Double-hit lymphoma (DHL) is a rare type of aggressive B-cell lymphoma defined as a high-grade B-cell lymphoma (HGBCL) with the presence of MYC, BCL2 and/or BCL6 rearrangements. Patients usually present with rapidly progressive and advanced stage of disease and, commonly, with extranodal involvement. Typically, patients become refractory to standard R-CHOP, and more aggressive regimens such as DA-EPOCH-R, R-hyperCVAD or CODOX-R regimens are typically needed. MYC is considered an “undruggable” mutation. Recent evidence suggests that pathogenic mechanisms associated with MYC could be potential targets. In this review, we also discuss the role of hematopoietic stem cell transplantation (HCT) and chimeric antigen receptor (CAR) T-cell therapy in DHL. We also discuss the role of potential novel agents such as BCL2 inhibitors, checkpoint inhibitors, bromodomain and extraterminal (BET) family inhibitors, Pi3K inhibitors, and others.
Collapse
Affiliation(s)
- Vania Phuoc
- Division Hematology/Oncology, University of South Florida, Tampa, FL, USA
| | - Jose Sandoval-Sus
- Department of Malignant Hematology and Cellular Therapy at Memorial Healthcare System, Pembroke Pines, FL, USA
| | - Julio C Chavez
- Department of Malignant Hematology, Moffitt Cancer Center, Tampa FL, USA
| |
Collapse
|
180
|
Han F, Zhang L, Chen C, Wang Y, Zhang Y, Qian L, Sun W, Zhou D, Yang B, Zhang H, Lai M. GLTSCR1 Negatively Regulates BRD4-Dependent Transcription Elongation and Inhibits CRC Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1901114. [PMID: 31832310 PMCID: PMC6891902 DOI: 10.1002/advs.201901114] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/30/2019] [Indexed: 05/08/2023]
Abstract
Frameshift mutations frequently occur in colorectal cancer (CRC) with microsatellite instability (MSI), but the nature and biological function of many MSI-associated mutations remain elusive. Here, an MSI frameshift mutation is identified in glioma tumor suppressor candidate region gene 1 (GLTSCR1) that produces two C-terminal-truncated proteins. Additionally, GLTSCR1 is verified as a tumor suppressor that inhibits CRC metastasis. Through binding to bromodomains and the phosphorylation-dependent interaction domain of bromodomain protein 4 (BRD4) via the C-terminus, GLTSCR1 blocks oncogenic transcriptional elongation. However, truncated GLTSCR1 translocates into the cytoplasm and loses BRD4 binding domain, which induces the phosphorylation of RNA Pol II at Ser2 and dephosphorylation at Ser5, then increases oncogenic transcriptional elongation. Importantly, GLTSCR1 deficiency decreases sensitivity to bromodomain and extra terminal domain inhibitors. This study highlights the molecular mechanism of the GLTSCR1-BRD4 interaction, which is a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Fengyan Han
- Department of PathologyKey Laboratory of Disease Proteomics of Zhejiang ProvinceResearch unit of intelligence classification of tumor pathology and precision therapy Chinese Academy of Medical Sciences (2019RU042)School of MedicineZhejiang UniversityHangzhou310058China
| | - Lei Zhang
- Department of PharmacologyChina Pharmaceutical UniversityNanjing210009China
| | - Chaoyi Chen
- Department of PathologyKey Laboratory of Disease Proteomics of Zhejiang ProvinceResearch unit of intelligence classification of tumor pathology and precision therapy Chinese Academy of Medical Sciences (2019RU042)School of MedicineZhejiang UniversityHangzhou310058China
| | - Yan Wang
- Department of PathologyKey Laboratory of Disease Proteomics of Zhejiang ProvinceResearch unit of intelligence classification of tumor pathology and precision therapy Chinese Academy of Medical Sciences (2019RU042)School of MedicineZhejiang UniversityHangzhou310058China
| | - Yi Zhang
- Department of PathologyKey Laboratory of Disease Proteomics of Zhejiang ProvinceResearch unit of intelligence classification of tumor pathology and precision therapy Chinese Academy of Medical Sciences (2019RU042)School of MedicineZhejiang UniversityHangzhou310058China
| | - Lili Qian
- Department of PathologyKey Laboratory of Disease Proteomics of Zhejiang ProvinceResearch unit of intelligence classification of tumor pathology and precision therapy Chinese Academy of Medical Sciences (2019RU042)School of MedicineZhejiang UniversityHangzhou310058China
| | - Wenjie Sun
- Department of PathologyKey Laboratory of Disease Proteomics of Zhejiang ProvinceResearch unit of intelligence classification of tumor pathology and precision therapy Chinese Academy of Medical Sciences (2019RU042)School of MedicineZhejiang UniversityHangzhou310058China
| | - Dan Zhou
- Department of PathologyKey Laboratory of Disease Proteomics of Zhejiang ProvinceResearch unit of intelligence classification of tumor pathology and precision therapy Chinese Academy of Medical Sciences (2019RU042)School of MedicineZhejiang UniversityHangzhou310058China
| | - Beibei Yang
- Department of PathologyKey Laboratory of Disease Proteomics of Zhejiang ProvinceResearch unit of intelligence classification of tumor pathology and precision therapy Chinese Academy of Medical Sciences (2019RU042)School of MedicineZhejiang UniversityHangzhou310058China
| | - Honghe Zhang
- Department of PathologyKey Laboratory of Disease Proteomics of Zhejiang ProvinceResearch unit of intelligence classification of tumor pathology and precision therapy Chinese Academy of Medical Sciences (2019RU042)School of MedicineZhejiang UniversityHangzhou310058China
| | - Maode Lai
- Department of PathologyKey Laboratory of Disease Proteomics of Zhejiang ProvinceResearch unit of intelligence classification of tumor pathology and precision therapy Chinese Academy of Medical Sciences (2019RU042)School of MedicineZhejiang UniversityHangzhou310058China
- Department of PharmacologyChina Pharmaceutical UniversityNanjing210009China
| |
Collapse
|
181
|
Sermer D, Pasqualucci L, Wendel HG, Melnick A, Younes A. Emerging epigenetic-modulating therapies in lymphoma. Nat Rev Clin Oncol 2019; 16:494-507. [PMID: 30837715 DOI: 10.1038/s41571-019-0190-8] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Despite considerable advances in the treatment of lymphoma, the prognosis of patients with relapsed and/or refractory disease continues to be poor; thus, a continued need exists for the development of novel approaches and therapies. Epigenetic dysregulation might drive and/or promote tumorigenesis in various types of malignancies and is prevalent in both B cell and T cell lymphomas. Over the past decade, a large number of epigenetic-modifying agents have been developed and introduced into the clinical management of patients with haematological malignancies. In this Review, we provide a concise overview of the most promising epigenetic therapies for the treatment of lymphomas, including inhibitors of histone deacetylases (HDACs), DNA methyltransferases (DNMTs), enhancer of zeste homologue 2 (EZH2), bromodomain and extra-terminal domain proteins (BETs), protein arginine N-methyltransferases (PRMTs) and isocitrate dehydrogenases (IDHs), and highlight the most promising future directions of research in this area.
Collapse
Affiliation(s)
- David Sermer
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laura Pasqualucci
- Institute for Cancer Genetics, Columbia University, New York, NY, USA
| | - Hans-Guido Wendel
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ari Melnick
- Weill-Cornell Medical College, New York, NY, USA
| | - Anas Younes
- Department of Medicine, Lymphoma Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
182
|
Miao Y, Medeiros LJ, Li Y, Li J, Young KH. Genetic alterations and their clinical implications in DLBCL. Nat Rev Clin Oncol 2019; 16:634-652. [PMID: 31127191 DOI: 10.1038/s41571-019-0225-1] [Citation(s) in RCA: 119] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Diffuse large B cell lymphoma (DLBCL) is a highly heterogeneous lymphoid neoplasm with variations in gene expression profiles and genetic alterations, which lead to substantial variations in clinical course and response to therapy. The advent of high-throughput genome sequencing platforms, and especially whole-exome sequencing, has helped to define the genetic landscape of DLBCL. In the past 10 years, these studies have identified many genetic alterations in DLBCL, some of which are specific to B cell lymphomas, whereas others can also be observed in other types of cancer. These aberrations result in altered activation of a wide range of signalling pathways and other cellular processes, including those involved in B cell differentiation, B cell receptor signalling, activation of the NF-κB pathway, apoptosis and epigenetic regulation. Further elaboration of the genetics of DLBCL will not only improve our understanding of disease pathogenesis but also provide further insight into disease classification, prognostication and therapeutic targets. In this Review, we describe the current understanding of the prevalence and causes of specific genetic alterations in DLBCL and their role in disease development and progression. We also summarize the available clinical data on therapies designed to target the aberrant pathways driven by these alterations.
Collapse
Affiliation(s)
- Yi Miao
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yong Li
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jianyong Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, China
| | - Ken H Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Graduate School of Biomedical Sciences, University of Texas Health Science Center, Houston, TX, USA.
| |
Collapse
|
183
|
Pillow TH, Adhikari P, Blake RA, Chen J, Del Rosario G, Deshmukh G, Figueroa I, Gascoigne KE, Kamath AV, Kaufman S, Kleinheinz T, Kozak KR, Latifi B, Leipold DD, Sing Li C, Li R, Mulvihill MM, O'Donohue A, Rowntree RK, Sadowsky JD, Wai J, Wang X, Wu C, Xu Z, Yao H, Yu S, Zhang D, Zang R, Zhang H, Zhou H, Zhu X, Dragovich PS. Antibody Conjugation of a Chimeric BET Degrader Enables
in vivo
Activity. ChemMedChem 2019; 15:17-25. [DOI: 10.1002/cmdc.201900497] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/11/2019] [Indexed: 12/12/2022]
Affiliation(s)
| | | | | | - Jinhua Chen
- Wuxi Apptec 288 Fute Zhong Road Waigaoqiao Free Trade Zone Shanghai 200131 China
| | | | - Gauri Deshmukh
- Genentech Inc. 1 DNA Way South San Francisco CA 94080 USA
| | | | | | | | - Susan Kaufman
- Genentech Inc. 1 DNA Way South San Francisco CA 94080 USA
| | | | | | - Brandon Latifi
- Genentech Inc. 1 DNA Way South San Francisco CA 94080 USA
| | | | - Chun Sing Li
- Wuxi Apptec 288 Fute Zhong Road Waigaoqiao Free Trade Zone Shanghai 200131 China
| | - Ruina Li
- Genentech Inc. 1 DNA Way South San Francisco CA 94080 USA
| | | | | | | | | | - John Wai
- Wuxi Apptec 288 Fute Zhong Road Waigaoqiao Free Trade Zone Shanghai 200131 China
| | - Xinxin Wang
- Wuxi Apptec 288 Fute Zhong Road Waigaoqiao Free Trade Zone Shanghai 200131 China
| | - Cong Wu
- Genentech Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Zijin Xu
- Wuxi Apptec 288 Fute Zhong Road Waigaoqiao Free Trade Zone Shanghai 200131 China
| | - Hui Yao
- Wuxi Apptec 288 Fute Zhong Road Waigaoqiao Free Trade Zone Shanghai 200131 China
| | - Shang‐Fan Yu
- Genentech Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Donglu Zhang
- Genentech Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Richard Zang
- Genentech Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Hongyan Zhang
- Wuxi Apptec 288 Fute Zhong Road Waigaoqiao Free Trade Zone Shanghai 200131 China
| | - Hao Zhou
- Wuxi Apptec 288 Fute Zhong Road Waigaoqiao Free Trade Zone Shanghai 200131 China
| | - Xiaoyu Zhu
- Wuxi Apptec 288 Fute Zhong Road Waigaoqiao Free Trade Zone Shanghai 200131 China
| | | |
Collapse
|
184
|
Huang QW, He LJ, Zheng S, Liu T, Peng BN. An Overview of Molecular Mechanism, Clinicopathological Factors, and Treatment in NUT Carcinoma. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1018439. [PMID: 31815119 PMCID: PMC6877965 DOI: 10.1155/2019/1018439] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 10/15/2019] [Indexed: 12/13/2022]
Abstract
NUT carcinoma (NC) is a rare and poorly differentiated tumor, with highly aggressive and fatal neoplasm. NC is characterized by chromosomal rearrangement involving NUTM1 gene, but lack of specific clinical and histomorphological features. It is more common in midline anatomic sites, such as head and neck, mediastinum, and other midline organs. NC may occur at any age, but mainly in children and young adults. In addition, male and female are equally affected. Most clinicians lack a clear understanding of the disease, and NC diagnostic reagents are still not widely used; therefore, misdiagnosis often occurs in clinic. Due to the highly aggressive nature of the disease and the insensitivity to nonspecific chemotherapy or radiotherapy, many patients have died before the confirmation of NC. In fact, the true incidence of NC is much higher than the current statistics. In recent years, targeted therapy for NC has also made some progress. This article aims to summarize the molecular mechanisms, clinicopathological characteristics, and treatment of NC.
Collapse
Affiliation(s)
- Qian W. Huang
- Department of Medical Oncology, People's Hospital of Boluo County, Huizhou 516000, China
| | - Li J. He
- Department of Medical Oncology, People's Hospital of Liaoning Province, Shenyang 110000, China
| | - Shuang Zheng
- Department of Medical Oncology, People's Hospital of Liaoning Province, Shenyang 110000, China
| | - Tao Liu
- Department of Orthopaedics, People's Hospital of Gaotang County, Liaocheng 252000, China
| | - Bei N. Peng
- Dalian Medical University, Dalian 116000, China
| |
Collapse
|
185
|
Morgado-Pascual JL, Rayego-Mateos S, Tejedor L, Suarez-Alvarez B, Ruiz-Ortega M. Bromodomain and Extraterminal Proteins as Novel Epigenetic Targets for Renal Diseases. Front Pharmacol 2019; 10:1315. [PMID: 31780938 PMCID: PMC6857099 DOI: 10.3389/fphar.2019.01315] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022] Open
Abstract
Epigenetic mechanisms, especially DNA methylation and histone modifications, are dynamic processes that regulate the gene expression transcriptional program in normal and diseased states. The bromodomain and extraterminal (BET) protein family (BRD2, BRD3, BRD4, and BRDT) are epigenetic readers that, via bromodomains, regulate gene transcription by binding to acetylated lysine residues on histones and master transcriptional factors. Experimental data have demonstrated the involvement of some BET proteins in many pathological conditions, including tumor development, infections, autoimmunity, and inflammation. Selective bromodomain inhibitors are epigenetic drugs that block the interaction between BET proteins and acetylated proteins, thus exerting beneficial effects. Recent data have described the beneficial effect of BET inhibition on experimental renal diseases. Emerging evidence underscores the importance of environmental modifications in the origin of pathological features in chronic kidney diseases (CKD). Several cellular processes such as oxidation, metabolic disorders, cytokines, inflammation, or accumulated uremic toxins may induce epigenetic modifications that regulate key processes involved in renal damage and in other pathological conditions observed in CKD patients. Here, we review how targeting bromodomains in BET proteins may regulate essential processes involved in renal diseases and in associated complications found in CKD patients, such as cardiovascular damage, highlighting the potential of epigenetic therapeutic strategies against BET proteins for CKD treatment and associated risks.
Collapse
Affiliation(s)
- Jose Luis Morgado-Pascual
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Sandra Rayego-Mateos
- Red de Investigación Renal (REDinREN), Madrid, Spain.,Vascular and Renal Translational Research Group, Institut de Recerca Biomèdica de Lleida (IRBLleida), Lleida, Spain
| | - Lucia Tejedor
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| | - Beatriz Suarez-Alvarez
- Red de Investigación Renal (REDinREN), Madrid, Spain.,Translational Immunology Laboratory, Health Research Institute of the Principality of Asturias (ISPA), Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, IIS-Fundación Jiménez Díaz, Universidad Autónoma Madrid, Madrid, Spain.,Red de Investigación Renal (REDinREN), Madrid, Spain
| |
Collapse
|
186
|
Ribeiro ML, Reyes-Garau D, Armengol M, Fernández-Serrano M, Roué G. Recent Advances in the Targeting of Epigenetic Regulators in B-Cell Non-Hodgkin Lymphoma. Front Genet 2019; 10:986. [PMID: 31681423 PMCID: PMC6807552 DOI: 10.3389/fgene.2019.00986] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 09/17/2019] [Indexed: 12/13/2022] Open
Abstract
In the last 10 years, major advances have been made in the diagnosis and development of selective therapies for several blood cancers, including B-cell non-Hodgkin lymphoma (B-NHL), a heterogeneous group of malignancies arising from the mature B lymphocyte compartment. However, most of these entities remain incurable and current treatments are associated with variable efficacy, several adverse events, and frequent relapses. Thus, new diagnostic paradigms and novel therapeutic options are required to improve the prognosis of patients with B-NHL. With the recent deciphering of the mutational landscapes of B-cell disorders by high-throughput sequencing, it came out that different epigenetic deregulations might drive and/or promote B lymphomagenesis. Consistently, over the last decade, numerous epigenetic drugs (or epidrugs) have emerged in the clinical management of B-NHL patients. In this review, we will present an overview of the most relevant epidrugs tested and/or used so far for the treatment of different subtypes of B-NHL, from first-generation epigenetic therapies like histone acetyl transferases (HDACs) or DNA-methyl transferases (DNMTs) inhibitors to new agents showing selectivity for proteins that are mutated, translocated, and/or overexpressed in these diseases, including EZH2, BET, and PRMT. We will dissect the mechanisms of action of these epigenetic inhibitors, as well as the molecular processes underlying their lack of efficacy in refractory patients. This review will also provide a summary of the latest strategies being employed in preclinical and clinical settings, and will point out the most promising lines of investigation in the field.
Collapse
Affiliation(s)
- Marcelo L. Ribeiro
- Laboratory of Experimental Hematology, Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University Medical School, Braganca Paulista, São Paulo, Brazil
| | - Diana Reyes-Garau
- Laboratory of Experimental Hematology, Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
| | - Marc Armengol
- Laboratory of Experimental Hematology, Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
| | - Miranda Fernández-Serrano
- Laboratory of Experimental Hematology, Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
| | - Gaël Roué
- Laboratory of Experimental Hematology, Department of Hematology, Vall d’Hebron Institute of Oncology (VHIO), Vall d’Hebron University Hospital, Autonomous University of Barcelona, Barcelona, Spain
| |
Collapse
|
187
|
Reyes-Garau D, Ribeiro ML, Roué G. Pharmacological Targeting of BET Bromodomain Proteins in Acute Myeloid Leukemia and Malignant Lymphomas: From Molecular Characterization to Clinical Applications. Cancers (Basel) 2019; 11:cancers11101483. [PMID: 31581671 PMCID: PMC6826405 DOI: 10.3390/cancers11101483] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/16/2022] Open
Abstract
Alterations in protein-protein and DNA-protein interactions and abnormal chromatin remodeling are a major cause of uncontrolled gene transcription and constitutive activation of critical signaling pathways in cancer cells. Multiple epigenetic regulators are known to be deregulated in several hematologic neoplasms, by somatic mutation, amplification, or deletion, allowing the identification of specific epigenetic signatures, but at the same time providing new therapeutic opportunities. While these vulnerabilities have been traditionally addressed by hypomethylating agents or histone deacetylase inhibitors, pharmacological targeting of bromodomain-containing proteins has recently emerged as a promising approach in a number of lymphoid and myeloid malignancies. Indeed, preclinical and clinical studies highlight the relevance of targeting the bromodomain and extra-terminal (BET) family as an efficient strategy of target transcription irrespective of the presence of epigenetic mutations. Here we will summarize the main advances achieved in the last decade regarding the preclinical and clinical evaluation of BET bromodomain inhibitors in hematologic cancers, either as monotherapies or in combinations with standard and/or experimental agents. A mention will finally be given to the new concept of the protein degrader, and the perspective it holds for the design of bromodomain-based therapies.
Collapse
Affiliation(s)
- Diana Reyes-Garau
- Laboratory of Experimental Hematology, Department of Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Autonomous University of Barcelona, 08035 Barcelona, Spain.
| | - Marcelo L Ribeiro
- Laboratory of Experimental Hematology, Department of Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Autonomous University of Barcelona, 08035 Barcelona, Spain.
- Laboratory of Immunopharmacology and Molecular Biology, Sao Francisco University Medical School, Braganca Paulista, São Paulo 12916-900, Brazil.
| | - Gaël Roué
- Laboratory of Experimental Hematology, Department of Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Autonomous University of Barcelona, 08035 Barcelona, Spain.
| |
Collapse
|
188
|
Falchook G, Rosen S, LoRusso P, Watts J, Gupta S, Coombs CC, Talpaz M, Kurzrock R, Mita M, Cassaday R, Harb W, Peguero J, Smith DC, Piha-Paul SA, Szmulewitz R, Noel MS, Yeleswaram S, Liu P, Switzky J, Zhou G, Zheng F, Mehta A. Development of 2 Bromodomain and Extraterminal Inhibitors With Distinct Pharmacokinetic and Pharmacodynamic Profiles for the Treatment of Advanced Malignancies. Clin Cancer Res 2019; 26:1247-1257. [PMID: 31527168 DOI: 10.1158/1078-0432.ccr-18-4071] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 06/13/2019] [Accepted: 09/11/2019] [Indexed: 01/17/2023]
Abstract
PURPOSE Bromodomain and extraterminal (BET) proteins are key epigenetic transcriptional regulators, inhibition of which may suppress oncogene expression. We report results from 2 independent first-in-human phase 1/2 dose-escalation and expansion, safety and tolerability studies of BET inhibitors INCB054329 (study INCB 54329-101; NCT02431260) and INCB057643 (study INCB 57643-101; NCT02711137). PATIENTS AND METHODS Patients (≥18 years) with advanced malignancies, ≥1 prior therapy, and adequate organ functions received oral INCB054329 (monotherapy) or INCB057643 (monotherapy or in combination with standard-of-care) in 21-day cycles (or 28-day cycles depending on standard-of-care combination). Primary endpoints were safety and tolerability. RESULTS Sixty-nine and 134 patients received INCB054329 and INCB057643, respectively. Study INCB 54329-101 has been completed; INCB 57643-101 is currently active, but not recruiting (no patients were receiving treatment as of January 8, 2019). Terminal elimination half-life was shorter for INCB054329 versus INCB057643 (mean [SD], 2.24 [2.03] vs. 11.1 [8.27] hours). INCB054329 demonstrated higher interpatient variability in oral clearance versus INCB057643 (CV%, 142% vs. 45.5%). Most common (>20%) any-grade treatment-related adverse events were similar for both drugs (INCB054329; INCB057643): nausea (35%; 30%), thrombocytopenia (33%; 32%), fatigue (29%; 30%), decreased appetite (26%; 22%). Two confirmed complete responses and 4 confirmed partial responses with INCB057643 were reported as best responses. CONCLUSIONS INCB057643 exhibited a more favorable PK profile versus INCB054329; exposure-dependent thrombocytopenia was observed with both drugs which limited the target inhibition that could be safely maintained. Further efforts are required to identify patient populations that can benefit most, and an optimal dosing scheme to maximize therapeutic index.
Collapse
Affiliation(s)
- Gerald Falchook
- Sarah Cannon Research Institute at HealthONE, Denver, Colorado.
| | - Seth Rosen
- Hematology-Oncology Associates of Treasure Coast, Port St Lucie, Florida
| | | | - Justin Watts
- Sylvester Comprehensive Cancer Center, Miami, Florida
| | | | - Catherine C Coombs
- University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | | | - Monica Mita
- Cedars-Sinai Medical Center, Los Angeles, California
| | - Ryan Cassaday
- University of Washington and Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Wael Harb
- Horizon Oncology Center, Lafeyette, Indiana
| | | | | | | | | | - Marcus S Noel
- University of Rochester, Wilmot Cancer Center, Rochester, New York
| | - Swamy Yeleswaram
- Incyte Research Institute, Incyte Corporation, Wilmington, Delaware
| | - Phillip Liu
- Incyte Research Institute, Incyte Corporation, Wilmington, Delaware
| | | | | | - Fred Zheng
- Incyte Corporation, Wilmington, Delaware
| | | |
Collapse
|
189
|
Wingelhofer B, Somervaille TCP. Emerging Epigenetic Therapeutic Targets in Acute Myeloid Leukemia. Front Oncol 2019; 9:850. [PMID: 31552175 PMCID: PMC6743337 DOI: 10.3389/fonc.2019.00850] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 08/19/2019] [Indexed: 01/23/2023] Open
Abstract
Acute myeloid leukemia (AML) is a genetically heterogeneous malignancy for which treatment options have been largely limited to cytotoxic chemotherapy for the past four decades. Next-generation sequencing and other approaches have identified a spectrum of genomic and epigenomic alterations that contribute to AML initiation and maintenance. The key role of epigenetic modifiers and the reversibility of epigenetic changes have paved the way for evaluation of a new set of drug targets, and facilitated the design of novel candidate treatment strategies. More recently, seven new targeted therapies have been FDA-approved demonstrating successful implementation of the past decades' research. In this review, we will summarize the most recent advances in targeted therapeutics designed for a focused group of key epigenetic regulators in AML, outline their mechanism of action and their current status in clinical development. Furthermore, we will discuss promising new approaches for epigenetic targeted treatment in AML which are currently being tested in pre-clinical trials.
Collapse
Affiliation(s)
| | - Tim C. P. Somervaille
- Leukaemia Biology Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
190
|
Erkes DA, Field CO, Capparelli C, Tiago M, Purwin TJ, Chervoneva I, Berger AC, Hartsough EJ, Villanueva J, Aplin AE. The next-generation BET inhibitor, PLX51107, delays melanoma growth in a CD8-mediated manner. Pigment Cell Melanoma Res 2019; 32:687-696. [PMID: 31063649 PMCID: PMC6697571 DOI: 10.1111/pcmr.12788] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/26/2019] [Accepted: 04/28/2019] [Indexed: 12/30/2022]
Abstract
Epigenetic agents such as bromodomain and extra-terminal region inhibitors (BETi) slow tumor growth via tumor intrinsic alterations; however, their effects on antitumor immunity remain unclear. A recent advance is the development of next-generation BETi that are potent and display a favorable half-life. Here, we tested the BETi, PLX51107, for immune-based effects on tumor growth in BRAF V600E melanoma syngeneic models. PLX51107 delayed melanoma tumor growth and increased activated, proliferating, and functional CD8+ T cells in tumors leading to CD8+ T-cell-mediated tumor growth delay. PLX51107 decreased Cox2 expression, increased dendritic cells, and lowered PD-L1, FasL, and IDO-1 expression in the tumor microenvironment. Importantly, PLX51107 delayed the growth of tumors that progressed on anti-PD-1 therapy; a response associated with decreased Cox2 levels, decreased PD-L1 expression on non-immune cells, and increased intratumoral CD8+ T cells. Thus, next-generation BETi represent a potential first-line and secondary treatment strategy for metastatic melanoma by eliciting effects, at least in part, on antitumor CD8+ T cells.
Collapse
Affiliation(s)
- Dan A. Erkes
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Conroy O. Field
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Claudia Capparelli
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Manoela Tiago
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Timothy J. Purwin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Inna Chervoneva
- Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Adam C. Berger
- Department of Surgical Oncology, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Edward J. Hartsough
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19107
| | - Jessie Villanueva
- Molecular and Cellular Oncogenesis Program, Melanoma Research Center, The Wistar Institute, PA 19104, USA
| | - Andrew E. Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
191
|
Tsai DY, Hung KH, Chang CW, Lin KI. Regulatory mechanisms of B cell responses and the implication in B cell-related diseases. J Biomed Sci 2019; 26:64. [PMID: 31472685 PMCID: PMC6717636 DOI: 10.1186/s12929-019-0558-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 08/22/2019] [Indexed: 12/13/2022] Open
Abstract
Terminally differentiated B cell, the plasma cell, is the sole cell type capable of producing antibodies in our body. Over the past 30 years, the identification of many key molecules controlling B cell activation and differentiation has elucidated the molecular pathways for generating antibody-producing plasma cells. Several types of regulation modulating the functions of the important key molecules in B cell activation and differentiation add other layers of complexity in shaping B cell responses following antigen exposure in the absence or presence of T cell help. Further understanding of the mechanisms contributing to the proper activation and differentiation of B cells into antibody-secreting plasma cells may enable us to develop new strategies for managing antibody humoral responses during health and disease. Herein, we reviewed the effect of different types of regulation, including transcriptional regulation, post-transcriptional regulation and epigenetic regulation, on B cell activation, and on mounting memory B cell and antibody responses. We also discussed the link between the dysregulation of the abovementioned regulatory mechanisms and B cell-related disorders.
Collapse
Affiliation(s)
- Dong-Yan Tsai
- Genomics Research Center, Academia Sinica, 128 Academia Road, Sec. 2, Nankang Dist, Taipei, 115, Taiwan
| | - Kuo-Hsuan Hung
- Genomics Research Center, Academia Sinica, 128 Academia Road, Sec. 2, Nankang Dist, Taipei, 115, Taiwan
| | - Chia-Wei Chang
- Genomics Research Center, Academia Sinica, 128 Academia Road, Sec. 2, Nankang Dist, Taipei, 115, Taiwan.,Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, 110, Taiwan
| | - Kuo-I Lin
- Genomics Research Center, Academia Sinica, 128 Academia Road, Sec. 2, Nankang Dist, Taipei, 115, Taiwan. .,Graduate Institute of Immunology, College of Medicine, National Taiwan University, Taipei, 110, Taiwan.
| |
Collapse
|
192
|
Lai H, Wang R, Li S, Shi Q, Cai Z, Li Y, Liu Y. LIN9 confers paclitaxel resistance in triple negative breast cancer cells by upregulating CCSAP. SCIENCE CHINA-LIFE SCIENCES 2019; 63:419-428. [DOI: 10.1007/s11427-019-9581-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 06/16/2019] [Indexed: 12/17/2022]
|
193
|
Gobbi G, Donati B, Do Valle IF, Reggiani F, Torricelli F, Remondini D, Castellani G, Ambrosetti DC, Ciarrocchi A, Sancisi V. The Hippo pathway modulates resistance to BET proteins inhibitors in lung cancer cells. Oncogene 2019; 38:6801-6817. [PMID: 31406246 DOI: 10.1038/s41388-019-0924-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 04/19/2019] [Accepted: 05/13/2019] [Indexed: 11/09/2022]
Abstract
Inhibitors of BET proteins (BETi) are anti-cancer drugs that have shown efficacy in pre-clinical settings and are currently in clinical trials for different types of cancer, including non-small cell lung cancer (NSCLC). Currently, no predictive biomarker is available to identify patients that may benefit from this treatment. To uncover the mechanisms of resistance to BETi, we performed a genome-scale CRISPR/Cas9 screening in lung cancer cells. We identified three Hippo pathway genes, LATS2, TAOK1, and NF2, as key determinants for sensitivity to BETi. The knockout of these genes induces resistance to BETi, by promoting TAZ nuclear localization and transcriptional activity. Conversely, TAZ expression promotes resistance to these drugs. We also showed that TAZ, YAP, and their partner TEAD are direct targets of BRD4 and that treatment with BETi downregulates their expression. Noticeably, molecular alterations in one or more of these genes are present in a large fraction of NSCLC patients and TAZ amplification or overexpression correlates with a worse outcome in lung adenocarcinoma. Our data define the central role of Hippo pathway in mediating resistance to BETi and provide a rationale for using BETi to counter-act YAP/TAZ-mediated pro-oncogenic activity.
Collapse
Affiliation(s)
- Giulia Gobbi
- Laboratory of Translational Research, Azienda USL di Reggio Emilia - IRCCS, Reggio Emilia, Italy
| | - Benedetta Donati
- Laboratory of Translational Research, Azienda USL di Reggio Emilia - IRCCS, Reggio Emilia, Italy
| | - Italo Faria Do Valle
- Department of Physics, Center for Complex Network Research, Northeastern University, Boston, MA, USA
| | - Francesca Reggiani
- Laboratory of Translational Research, Azienda USL di Reggio Emilia - IRCCS, Reggio Emilia, Italy
| | - Federica Torricelli
- Laboratory of Translational Research, Azienda USL di Reggio Emilia - IRCCS, Reggio Emilia, Italy
| | - Daniel Remondini
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | - Gastone Castellani
- Department of Physics and Astronomy, University of Bologna, Bologna, Italy
| | | | - Alessia Ciarrocchi
- Laboratory of Translational Research, Azienda USL di Reggio Emilia - IRCCS, Reggio Emilia, Italy
| | - Valentina Sancisi
- Laboratory of Translational Research, Azienda USL di Reggio Emilia - IRCCS, Reggio Emilia, Italy.
| |
Collapse
|
194
|
Chandhok NS, Prebet T. Insights into novel emerging epigenetic drugs in myeloid malignancies. Ther Adv Hematol 2019; 10:2040620719866081. [PMID: 31431820 PMCID: PMC6685116 DOI: 10.1177/2040620719866081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 06/10/2019] [Indexed: 12/15/2022] Open
Abstract
Epigenetics has been defined as ‘a stably heritable phenotype resulting from changes in a chromosome without alterations in the DNA sequence’ and several epigenetic regulators are recurrently mutated in hematological malignancies. Epigenetic modifications include changes such as DNA methylation, histone modifications and RNA associated gene silencing. Transcriptional regulation, chromosome stability, DNA replication and DNA repair are all controlled by these modifications. Mutations in genes encoding epigenetic modifiers are a frequent occurrence in hematologic malignancies and important in both the initiation and progression of cancer. Epigenetic modifications are also frequently reversible, allowing excellent opportunities for therapeutic intervention. The goal of epigenetic therapies is to reverse epigenetic dysregulation, restore the epigenetic balance, and revert malignant cells to a more normal condition. The role of epigenetic therapies thus far is most established in hematologic malignancies, with several agents already approved by the US Food and Drug Administration. In this review, we discuss pharmacological agents targeting epigenetic regulators.
Collapse
Affiliation(s)
- Namrata S Chandhok
- Division of Hematology/Oncology, Smilow Cancer Center at Yale New Haven Hospital, New Haven, CT, USA
| | - Thomas Prebet
- Division of Hematology/Oncology, Smilow Cancer Center at Yale New Haven Hospital, 35 Park Street, New Haven, CT 06511, USA
| |
Collapse
|
195
|
Civenni G, Bosotti R, Timpanaro A, Vàzquez R, Merulla J, Pandit S, Rossi S, Albino D, Allegrini S, Mitra A, Mapelli SN, Vierling L, Giurdanella M, Marchetti M, Paganoni A, Rinaldi A, Losa M, Mira-Catò E, D'Antuono R, Morone D, Rezai K, D'Ambrosio G, Ouafik L, Mackenzie S, Riveiro ME, Cvitkovic E, Carbone GM, Catapano CV. Epigenetic Control of Mitochondrial Fission Enables Self-Renewal of Stem-like Tumor Cells in Human Prostate Cancer. Cell Metab 2019; 30:303-318.e6. [PMID: 31130467 DOI: 10.1016/j.cmet.2019.05.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 12/06/2018] [Accepted: 04/30/2019] [Indexed: 01/16/2023]
Abstract
Cancer stem cells (CSCs) contribute to disease progression and treatment failure in human cancers. The balance among self-renewal, differentiation, and senescence determines the expansion or progressive exhaustion of CSCs. Targeting these processes might lead to novel anticancer therapies. Here, we uncover a novel link between BRD4, mitochondrial dynamics, and self-renewal of prostate CSCs. Targeting BRD4 by genetic knockdown or chemical inhibitors blocked mitochondrial fission and caused CSC exhaustion and loss of tumorigenic capability. Depletion of CSCs occurred in multiple prostate cancer models, indicating a common vulnerability and dependency on mitochondrial dynamics. These effects depended on rewiring of the BRD4-driven transcription and repression of mitochondrial fission factor (Mff). Knockdown of Mff reproduced the effects of BRD4 inhibition, whereas ectopic Mff expression rescued prostate CSCs from exhaustion. This novel concept of targeting mitochondrial plasticity in CSCs through BRD4 inhibition provides a new paradigm for developing more effective treatment strategies for prostate cancer.
Collapse
Affiliation(s)
- Gianluca Civenni
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Roberto Bosotti
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Andrea Timpanaro
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Ramiro Vàzquez
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Jessica Merulla
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Shusil Pandit
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Simona Rossi
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Domenico Albino
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Sara Allegrini
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Abhishek Mitra
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Sarah N Mapelli
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland; Institute of Computational Science, Università della Svizzera Italiana (USI), Lugano 6900, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne 1015, Switzerland
| | - Luca Vierling
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Martina Giurdanella
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Martina Marchetti
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Alyssa Paganoni
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Andrea Rinaldi
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Marco Losa
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Enrica Mira-Catò
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Rocco D'Antuono
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Diego Morone
- Institute for Research in Biomedicine (IRB), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Keyvan Rezai
- Institute Curie-Hospital René Huguenin, Saint Cloud 92210, France
| | | | | | - Sarah Mackenzie
- Oncology Therapeutic Development (OTD), Clichy 92110, France
| | - Maria E Riveiro
- Oncology Therapeutic Development (OTD), Clichy 92110, France
| | - Esteban Cvitkovic
- Oncology Therapeutic Development (OTD), Clichy 92110, France; Oncoethix GmbH, Merck Sharp and Dohme Corp., Lucerne 6006, Switzerland
| | - Giuseppina M Carbone
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland
| | - Carlo V Catapano
- Institute of Oncology Research (IOR), Università della Svizzera Italiana (USI), Bellinzona 6500, Switzerland; Swiss Institute of Bioinformatics (SIB), Lausanne 1015, Switzerland; Department of Oncology, Faculty of Biology and Medicine, University of Lausanne, Lausanne 1005, Switzerland.
| |
Collapse
|
196
|
Jia Y, Chng WJ, Zhou J. Super-enhancers: critical roles and therapeutic targets in hematologic malignancies. J Hematol Oncol 2019; 12:77. [PMID: 31311566 PMCID: PMC6636097 DOI: 10.1186/s13045-019-0757-y] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 06/14/2019] [Indexed: 12/11/2022] Open
Abstract
Super-enhancers (SEs) in a broad range of human cell types are large clusters of enhancers with aberrant high levels of transcription factor binding, which are central to drive expression of genes in controlling cell identity and stimulating oncogenic transcription. Cancer cells acquire super-enhancers at oncogene and cancerous phenotype relies on these abnormal transcription propelled by SEs. Furthermore, specific inhibitors targeting SEs assembly and activation have offered potential targets for treating various tumors including hematological malignancies. Here, we first review the identification, functional significance of SEs. Next, we summarize recent findings of SEs and SE-driven gene regulation in normal hematopoiesis and hematologic malignancies. The importance and various modes of SE-mediated MYC oncogene amplification are illustrated. Finally, we highlight the progress of SEs as selective therapeutic targets in basic research and clinical trials. Some open questions regarding functional significance and future directions of targeting SEs in the clinic will be discussed too.
Collapse
Affiliation(s)
- Yunlu Jia
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, 117599, Republic of Singapore.,Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, 310016, Zhejiang, China
| | - Wee-Joo Chng
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, 117599, Republic of Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Republic of Singapore.,Department of Hematology-Oncology, National University Cancer Institute of Singapore (NCIS), The National University Health System (NUHS), 1E, Kent Ridge Road, Singapore, 119228, Republic of Singapore
| | - Jianbiao Zhou
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, Centre for Translational Medicine, Singapore, 117599, Republic of Singapore. .,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Republic of Singapore.
| |
Collapse
|
197
|
Abstract
Less than a decade ago, it was shown that bromodomains, acetyl lysine 'reader' modules found in proteins with varied functions, were highly tractable small-molecule targets. This is an unusual property for protein-protein or protein-peptide interaction domains, and it prompted a wave of chemical probe discovery to understand the biological potential of new agents that targeted bromodomains. The original examples, inhibitors of the bromodomain and extra-terminal (BET) class of bromodomains, showed enticing anti-inflammatory and anticancer activities, and several compounds have since advanced to human clinical trials. Here, we review the current state of BET inhibitor biology in relation to clinical development, and we discuss the next wave of bromodomain inhibitors with clinical potential in oncology and non-oncology indications. The lessons learned from BET inhibitor programmes should affect efforts to develop drugs that target non-BET bromodomains and other epigenetic readers.
Collapse
|
198
|
Damaneh MS, Hu JP, Huan XJ, Song SS, Tian CQ, Chen DQ, Meng T, Chen YL, Shen JK, Xiong B, Miao ZH, Wang YQ. A new BET inhibitor, 171, inhibits tumor growth through cell proliferation inhibition more than apoptosis induction. Invest New Drugs 2019; 38:700-713. [DOI: 10.1007/s10637-019-00818-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 06/14/2019] [Indexed: 01/06/2023]
|
199
|
Anwer F, Gee KM, Iftikhar A, Baig M, Russ AD, Saeed S, Zar MA, Razzaq F, Carew J, Nawrocki S, Al-Kateb H, Cavalcante Parr NN, McBride A, Valent J, Samaras C. Future of Personalized Therapy Targeting Aberrant Signaling Pathways in Multiple Myeloma. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2019; 19:397-405. [PMID: 31036508 PMCID: PMC6626550 DOI: 10.1016/j.clml.2019.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 02/19/2019] [Accepted: 03/17/2019] [Indexed: 12/19/2022]
Abstract
Multiple myeloma (MM) is a genetically complex disease. Identification of mutations and aberrant signaling pathways that contribute to the progression of MM and drug resistance has potential to lead to specific targets and personalized treatment. Aberrant signal pathways include RAS pathway activation due to RAS or BRAF mutations (targeted by vemurafenib alone or combined with cobimetinib), BCL-2 overexpression in t(11:14) (targeted by venetoclax), JAK2 pathway activation (targeted by ruxolitinib), NF-κB pathway activation (treated with DANFIN combined with bortezomib), MDM2 overexpression, and PI3K/mTOR pathway activation (targeted by BEZ235). Cyclin D1 (CCND1) and MYC are also emerging as key potential targets. In addition, histone deacetylase inhibitors are already in use for the treatment of MM in combination therapy, and targeted inhibition of FGFR3 (AZD4547) is effective in myeloma cells with t(4;14) translocation. Bromodomain and extra terminal (BET) protein antagonists decrease the expression of MYC and have displayed promising antimyeloma activity. A better understanding of the alterations in signaling pathways that promote MM progression will further inform the development of precision therapy for patients.
Collapse
Affiliation(s)
- Faiz Anwer
- Taussig Cancer Center, Department of Hematology, Medical Oncology, Cleveland Clinic, Cleveland, OH.
| | - Kevin Mathew Gee
- Department of Molecular and Cellular Biology, The University of Arizona, Tucson, AZ
| | - Ahmad Iftikhar
- Department of Medicine, The University of Arizona, Tucson, AZ
| | - Mirza Baig
- Department of Medicine, Summit Medical Group, Summit, NJ
| | | | - Sabina Saeed
- College of Public Health, The University of Arizona, Tucson, AZ
| | - Muhammad Abu Zar
- Department of Medicine, Division of Hematology & Oncology, The University of Arizona, Tucson, AZ
| | - Faryal Razzaq
- Department of Medicine, Division of Hematology & Oncology, The University of Arizona, Tucson, AZ
| | - Jennifer Carew
- Department of Medicine, Division of Hematology & Oncology, The University of Arizona, Tucson, AZ
| | - Steffan Nawrocki
- Department of Medicine, Division of Hematology & Oncology, The University of Arizona, Tucson, AZ
| | - Hussam Al-Kateb
- Division of Human Genetics, Children's Hospital, Cincinnati, OH
| | | | - Ali McBride
- College of Pharmacy, The University of Arizona, Tucson, AZ
| | - Jason Valent
- Taussig Cancer Center, Department of Hematology, Medical Oncology, Cleveland Clinic, Cleveland, OH
| | - Christy Samaras
- Taussig Cancer Center, Department of Hematology, Medical Oncology, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
200
|
Astorgues-Xerri L, Vázquez R, Odore E, Rezai K, Kahatt C, Mackenzie S, Bekradda M, Coudé MM, Dombret H, Gardin C, Lokiec F, Raymond E, Noel K, Cvitkovic E, Herait P, Bertoni F, Riveiro ME. Insights into the cellular pharmacological properties of the BET-inhibitor OTX015/MK-8628 (birabresib), alone and in combination, in leukemia models. Leuk Lymphoma 2019; 60:3067-3070. [DOI: 10.1080/10428194.2019.1617860] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
| | - Ramiro Vázquez
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Elodie Odore
- RadioPharmacology Department, Curie Institute–Rene Huguenin Hospital, Saint Cloud, France
| | - Keyvan Rezai
- RadioPharmacology Department, Curie Institute–Rene Huguenin Hospital, Saint Cloud, France
| | | | | | | | | | - Herve Dombret
- Laboratoire de Transfert des Leucémies, Université Paris Diderot, Paris, France
| | - Claude Gardin
- Laboratoire de Transfert des Leucémies, Université Paris Diderot, Paris, France
| | - Francois Lokiec
- RadioPharmacology Department, Curie Institute–Rene Huguenin Hospital, Saint Cloud, France
| | - Eric Raymond
- Medical Oncology Department, CHUV, Lausanne, Switzerland
| | - Kay Noel
- Oncoethix SA, Lucerne, Switzerland
| | | | | | - Francesco Bertoni
- Institute of Oncology Research, Università della Svizzera Italiana, Bellinzona, Switzerland
| | | |
Collapse
|