The Combination of Chidamide with the CHOEP Regimen in Previously

Total Page:16

File Type:pdf, Size:1020Kb

The Combination of Chidamide with the CHOEP Regimen in Previously Cancer Biol Med 2021. doi: 10.20892/j.issn.2095-3941.2020.0413 ORIGINAL ARTICLE The combination of chidamide with the CHOEP regimen in previously untreated patients with peripheral T-cell lymphoma: a prospective, multicenter, single arm, phase 1b/2 study Wei Zhang1, Liping Su2, Lihong Liu3, Yuhuan Gao3, Quanshun Wang4, Hang Su5, Yuhuan Song6, Huilai Zhang7, Jing Shen8, Hongmei Jing9, Shuye Wang10, Xinan Cen11, Hui Liu12, Aichun Liu13, Zengjun Li14, Jianmin Luo15, Jianxia He16, Jingwen Wang17, O. A. O’Connor18, Daobin Zhou1 1Department of Hematology, Peking Union Medical College Hospital, Beijing 100730, China; 2Department of Hematology, Shanxi Provincial Cancer Hospital, Taiyuan 030013, China; 3Department of Hematology, Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China; 4Department of Hematology, Chinese PLA General Hospital, Beijing 100039, China; 5Department of Lymphoma, the 307 Hospital of PLA, Beijing 100071, China; 6Department of Lymphoma, Peking University Cancer Hospital and Institute, Beijing 100142, China; 7Department of Lymphoma, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China; 8Department of Hematology, Beijing Friendship Hospital, Beijing 100050, China; 9Department of Hematology, Peking University Third Hospital, Beijing 100191, China; 10Department of Hematology, the First Affiliated Hospital of Harbin Medical University, Harbin 150081, China; 11Department of Hematology, Peking University First Hospital, Beijing 100034, China; 12Department of Hematology, Beijing Hospital, Beijing 100730, China; 13Department of Lymphoma, Harbin Medical University Cancer Hospital, Harbin 150081, China; 14Lymphoma Diagnosis and Treatment Center, Institute of Hematology and Blood Diseases Hospital, Tianjin 300052, China; 15Department of Hematology, Second Hospital of Hebei Medical University, Shijiazhuang 050000, China; 16Department of Hematology, Shanxi Provincial People’s Hospital, Taiyuan 030012, China; 17Department of Hematology, Beijing Tongren Hospital, Beijing 100730, China; 18Columbia University Medical Center, New York 10032-3784, NY, USA ABSTRACT Objective: To assess the efficacy and safety of the novel histone deacetylase inhibitor, chidamide, in combination with cyclophosphamide, doxorubicin, vincristine, etoposide, and prednisone (Chi-CHOEP) for untreated peripheral T-cell lymphoma (PTCL). Methods: A prospective, multicenter, single arm, phase 1b/2 study was conducted. A total of 128 patients with untreated PTCL (18–70 years of age) were enrolled between March 2016 and November 2019, and treated with up to 6 cycles with the Chi-CHOEP regimen. In the phase 1b study, 3 dose levels of chidamide were evaluated and the primary endpoint was determination of the maximum- tolerated dose and recommended phase 2 dose (RP2D). The primary endpoint of the phase 2 study was 2-year progression-free survival (PFS). Results: Fifteen patients were enrolled in the phase 1b study and the RP2D for chidamide was determined to be 20 mg, twice a week. A total of 113 patients were treated at the RP2D in the phase 2 study, and the overall response rate was 60.2%, with a complete response rate of 40.7%. At a median follow-up of 36 months, the median PFS was 10.7 months, with 1-, 2-, and 3-year PFS rates of 49.9%, 38.0%, and 32.8%, respectively. The Chi-CHOEP regimen was well-tolerated, with grade 3/4 neutropenia occurring in approximately two-thirds of the patients. No unexpected adverse events (AEs) were reported and the observed AEs were manageable. Conclusions: This large cohort phase 1b/2 study showed that Chi-CHOEP was well-tolerated with modest efficacy in previously untreated PTCL patients. KEYWORDS Peripheral T-cell lymphoma; chidamide; histone deacetylase inhibitor; epigenetic Introduction Correspondence to: Daobin Zhou E-mail: [email protected] ORCID ID: https://orcid.org/0000-0002-1592-1932 Peripheral T-cell lymphomas (PTCLs) constitute a group Received August 07, 2020; accepted October 26, 2020 of heterogeneous lymphomas, mainly including PTCL-not Available at www.cancerbiomed.org ©2021 Cancer Biology & Medicine. Creative Commons otherwise specified (PTCL-NOS), angioimmunoblastic T-cell Attribution-NonCommercial 4.0 International License lymphoma (AITL), anaplastic large cell lymphoma (ALCL), 842 Zhang et al. Chi-CHOEP for previously untreated PTCL and other rare entities. PTCLs account for 10%–15% of according to the 2016 WHO classification10, and ALCL all non-Hodgkin lymphomas (NHLs) worldwide, being (ALK+), NK/T-cell lymphoma, and primary cutaneous lym- significantly higher in China (23.3%–30.2%)1,2. No stand- phoma were excluded. ard treatments have been established and commonly used (cyclophosphamide, doxorubicin, vincristine, and prednisone Treatment protocol (CHOP) or etoposide with CHOP (CHOEP) regimens result in poor outcomes, except for anaplastic lymphoma kinase The treatment protocol involved induction therapy with up (ALK)-positive ALCL3. Upfront autologous stem cell trans- to 6 cycles of the Chi-CHOEP regimen as follows: CHOEP plantation (ASCT) has been recommended to improve the [intravenous cyclophosphamide (750 mg/m²), doxorubicin prognosis of PTCL4. Frequent epigenetic dysregulation has (50 mg/m²), or epirubicin (70 mg/m²), and vincristine been documented in patients with PTCL, which makes his- (1.4 mg/m²) or vindesine (4 mg on day 1), etoposide (100 mg tone deacetylase (HDAC) a potential target5. Several HDAC on days 1–3), and oral prednisone (60 mg/m² on days 1–5), inhibitors have been approved for PTCL in a relapsed/refrac- repeated every 21 days] in combination with oral escalating tory setting, such as belinostat6, romidepsin7, and chidamide8. doses of chidamide twice a week. Patients were restaged by Chidamide, a novel benzamide class of HDAC inhibitor, has computed tomography (CT) or positron emission tomogra- demonstrated broad spectrum activity against various neo- phy-CT after the first 3 cycles according to the revised response plasms9. In a pivotal phase 2 trial with refractory or relapsed criteria for NHL. Patients with stable disease (SD) or progres- PTCL, chidamide produced an overall response rate (ORR) sive disease (PD) discontinued study treatment and went on and complete response (CR) rate of 28% and 14%, respec- to receive salvage therapy at the discretion of the treating phy- tively, leading to approval by the National Medical Products sician. Patients with a complete response or partial response Administration (China) in 2014 for treatment of refractory or received 3 additional cycles to complete the planned 6 cycles. relapsed PTCL8. Whether first-line use of HADC inhibitors in The selection of subsequent therapy was left to the discretion combination with the CHOEP regimen will further improve the of the treating physician (either ASCT consolidation or chid- prognosis in PTCL is unknown. Thus, we conducted a prospec- amide maintenance). tive, multicenter, phase 1b/2 study to evaluate the efficacy and safety of combination chidamide and CHOEP (Chi-CHOEP) Study endpoints in previously untreated patients with PTCL, hoping to establish a more effective regimen for this aggressive lymphoma. This study consisted of dose-escalation (phase 1b) and expan- sion (phase 2) phases. In phase 1b, 3 dose levels of chidamide Materials and methods (15, 20, and 25 mg twice a week) were evaluated at a tradi- tional 3 + 3 dose-escalation design. Dose-limited toxicity Study design and participants (DLT) was defined in the first 2 cycles and predefined DLTs included non-hematological toxicity of any grade ≥ 3, grade A prospective, multicenter, single arm, phase 1b/2 study was 4 agranulocytosis with granulocyte colony-stimulating factor conducted at 17 sites across China to evaluate the safety and support, or grade 4 thrombocytopenia lasting > 7 days. The efficacy of Chi-CHOEP in previously untreated patients with primary endpoint of the phase 2 study was 2-year progres- PTCL (ClinicalTrials.gov Identifier: NCT02987244). This sion-free survival (PFS), and secondary endpoints included study was carried out in accordance with the principles of overall response rate (ORR), CR, 2-year overall survival (OS), good clinical practice and the Declaration of Helsinki. The and adverse events (AEs). PFS was defined as the time from protocol, informed consent forms, and other relevant study treatment initiation until disease progression, death from any documentation were approved by the Ethics Committee of cause, or last follow-up, whichever came first. each center. All patients provided written informed consent before study entry. Statistical analysis A total of 142 patients (18–70 years of age) were screened and 128 were enrolled between March 1, 2016 and November All statistical analyses were performed with SPSS statistical 30, 2019 (Figure 1). The diagnosis of PTCL was confirmed software for Windows, version 24.0 (SPSS, Chicago, IL, USA). Cancer Biol Med Vol 18, No 3 Month 2021 843 Screened (n = 142) Exclusion (n = 14): -Histological diagnosis not met (n = 7) - Patients withdrew (n = 4) - Investigator decision (n = 3) Enrolled (n = 128) Phase 1b chidamide dose: 15 mg (n = 6), 20 mg (n = 6), 25 mg (n = 3) Phase 2 chidamide dose: 20 mg (n = 113) In phase 2 study Completed 6 cycles Completed 3–5 cycles Completed 1–2 cycles (n = 49) (n = 37) (n = 27) Reason for early discontinuation Investigator decision Disease progression Adverse
Recommended publications
  • An Overview of the Role of Hdacs in Cancer Immunotherapy
    International Journal of Molecular Sciences Review Immunoepigenetics Combination Therapies: An Overview of the Role of HDACs in Cancer Immunotherapy Debarati Banik, Sara Moufarrij and Alejandro Villagra * Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, 800 22nd St NW, Suite 8880, Washington, DC 20052, USA; [email protected] (D.B.); [email protected] (S.M.) * Correspondence: [email protected]; Tel.: +(202)-994-9547 Received: 22 March 2019; Accepted: 28 April 2019; Published: 7 May 2019 Abstract: Long-standing efforts to identify the multifaceted roles of histone deacetylase inhibitors (HDACis) have positioned these agents as promising drug candidates in combatting cancer, autoimmune, neurodegenerative, and infectious diseases. The same has also encouraged the evaluation of multiple HDACi candidates in preclinical studies in cancer and other diseases as well as the FDA-approval towards clinical use for specific agents. In this review, we have discussed how the efficacy of immunotherapy can be leveraged by combining it with HDACis. We have also included a brief overview of the classification of HDACis as well as their various roles in physiological and pathophysiological scenarios to target key cellular processes promoting the initiation, establishment, and progression of cancer. Given the critical role of the tumor microenvironment (TME) towards the outcome of anticancer therapies, we have also discussed the effect of HDACis on different components of the TME. We then have gradually progressed into examples of specific pan-HDACis, class I HDACi, and selective HDACis that either have been incorporated into clinical trials or show promising preclinical effects for future consideration.
    [Show full text]
  • Chidamide, a Histone Deacetylase Inhibitor, Induces Growth Arrest and Apoptosis in Multiple Myeloma Cells in a Caspase‑Dependent Manner
    ONCOLOGY LETTERS 18: 411-419, 2019 Chidamide, a histone deacetylase inhibitor, induces growth arrest and apoptosis in multiple myeloma cells in a caspase‑dependent manner XIANG-GUI YUAN*, YU-RONG HUANG*, TENG YU, HUA-WEI JIANG, YANG XU and XIAO-YING ZHAO Department of Hematology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China Received August 1, 2018; Accepted March 29, 2019 DOI: 10.3892/ol.2019.10301 Abstract. Chidamide, a novel histone deacetylase (HDAC) Introduction inhibitor, induces antitumor effects in various types of cancer. The present study aimed to evaluate the cytotoxic effect of Multiple myeloma (MM) is the second most frequent hemato- chidamide on multiple myeloma and the underlying mecha- logical neoplasm in the USA in 2018 (1) and is characterized nisms involved. Viability of multiple myeloma cells upon by the infiltration of clonal plasma cells in the bone marrow, chidamide treatment was determined by the Cell Counting secretion of monoclonal immunoglobulins and end organ Kit-8 assay. Apoptosis induction and cell cycle alteration were damage (2). Over the last decade, the introduction of protea- detected by flow cytometry. Specific apoptosis-associated some inhibitors [bortezomib (BTZ) and carfilzomib] and proteins and cell cycle proteins were evaluated by western immunomodulatory drugs (thalidomide and lenalidomide), blot analysis. Chidamide suppressed cell viability in a time- combined with autologous stem cell transplantation, have and dose-dependent manner. Chidamide treatment markedly significantly improved the prognosis of patients with MM. suppressed the expression of type I HDACs and further The 5-year overall survival (OS) rate of patients diagnosed induced the acetylation of histones H3 and H4.
    [Show full text]
  • Histone Deacetylase Inhibitors: a Prospect in Drug Discovery Histon Deasetilaz İnhibitörleri: İlaç Keşfinde Bir Aday
    Turk J Pharm Sci 2019;16(1):101-114 DOI: 10.4274/tjps.75047 REVIEW Histone Deacetylase Inhibitors: A Prospect in Drug Discovery Histon Deasetilaz İnhibitörleri: İlaç Keşfinde Bir Aday Rakesh YADAV*, Pooja MISHRA, Divya YADAV Banasthali University, Faculty of Pharmacy, Department of Pharmacy, Banasthali, India ABSTRACT Cancer is a provocative issue across the globe and treatment of uncontrolled cell growth follows a deep investigation in the field of drug discovery. Therefore, there is a crucial requirement for discovering an ingenious medicinally active agent that can amend idle drug targets. Increasing pragmatic evidence implies that histone deacetylases (HDACs) are trapped during cancer progression, which increases deacetylation and triggers changes in malignancy. They provide a ground-breaking scaffold and an attainable key for investigating chemical entity pertinent to HDAC biology as a therapeutic target in the drug discovery context. Due to gene expression, an impending requirement to prudently transfer cytotoxicity to cancerous cells, HDAC inhibitors may be developed as anticancer agents. The present review focuses on the basics of HDAC enzymes, their inhibitors, and therapeutic outcomes. Key words: Histone deacetylase inhibitors, apoptosis, multitherapeutic approach, cancer ÖZ Kanser tedavisi tüm toplum için büyük bir kışkırtıcıdır ve ilaç keşfi alanında bir araştırma hattını izlemektedir. Bu nedenle, işlemeyen ilaç hedeflerini iyileştirme yeterliliğine sahip, tıbbi aktif bir ajan keşfetmek için hayati bir gereklilik vardır. Artan pragmatik kanıtlar, histon deasetilazların (HDAC) kanserin ilerleme aşamasında deasetilasyonu arttırarak ve malignite değişikliklerini tetikleyerek kapana kısıldığını ifade etmektedir. HDAC inhibitörleri, ilaç keşfi bağlamında terapötik bir hedef olarak HDAC biyolojisiyle ilgili kimyasal varlığı araştırmak için, çığır açıcı iskele ve ulaşılabilir bir anahtar sağlarlar.
    [Show full text]
  • Small Molecule HDAC Inhibitors Promising Agents for Breast Cancer
    Bioorganic Chemistry 91 (2019) 103184 Contents lists available at ScienceDirect Bioorganic Chemistry journal homepage: www.elsevier.com/locate/bioorg Small molecule HDAC inhibitors: Promising agents for breast cancer T treatment ⁎ ⁎ Meiling Huanga,1, Jian Zhangb,1, Changjiao Yana, Xiaohui Lic, Juliang Zhanga, , Rui Linga, a Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi’an 710032, PR China b Department of Burns and Cutaneous Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, PR China c School of Life Science and Biotechnology, Dalian University of Technology, Dalian 116024, PR China ARTICLE INFO ABSTRACT Keywords: Breast cancer, a heterogeneous disease, is the most frequently diagnosed cancer and the second leading cause of HDACs cancer-related death among women worldwide. Recently, epigenetic abnormalities have emerged as an im- HDAC inhibitors portant hallmark of cancer development and progression. Given that histone deacetylases (HDACs) are crucial to Anticancer drug chromatin remodeling and epigenetics, their inhibitors have become promising potential anticancer drugs for Breast cancer research. Here we reviewed the mechanism and classification of histone deacetylases (HDACs), association Clinical trials between HDACs and breast cancer, classification and structure–activity relationship (SAR) of HDACIs, phar- macokinetic and toxicological properties of the HDACIs, and registered clinical studies for breast cancer treat- ment. In conclusion, HDACIs have shown desirable effects on breast cancer, especially when they are usedin combination with other anticancer agents. In the coming future, more multicenter and randomized Phase III studies are expected to be conducted pushing promising new therapies closer to the market. In addition, the design and synthesis of novel HDACIs are also needed.
    [Show full text]
  • Chidamide Increases the Sensitivity of Non-Small Cell Lung Cancer to Crizotinib by Decreasing C-MET Mrna Methylation
    bioRxiv preprint doi: https://doi.org/10.1101/2020.03.28.012971; this version posted March 30, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Chidamide increases the sensitivity of non-small cell lung cancer to crizotinib by decreasing c-MET mRNA methylation Nan Ding, Abin You, Wei Tian, Liankun Gu, Dajun Deng* Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital & Institute, Fu-Cheng-Lu #52, Haidian District, Beijing, 100142, China Corresponding author: Professor Dajun Deng, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Division of Etiology, Peking University Cancer Hospital & Institute, Fu-Cheng-Lu #52, Haidian District, Beijing, 100142, China. Phone: +8610-8816-96752; Fax: +8610-88122437; E-mail: [email protected] Running title: Chidamide increases crizotinib sensitivity of cancer Total number of figures and tables: 6 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.03.28.012971; this version posted March 30, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. ABSTRACT Introduction: Crizotinib is a kinase inhibitor targeting c-MET/ALK/ROS1 used as the first-line chemical for the treatment of non-small cell lung cancer (NSCLC) with ALK mutations.
    [Show full text]
  • Preclinical Evaluation of a Regimen Combining Chidamide and ABT-199
    Chen et al. Cell Death and Disease (2020) 11:778 https://doi.org/10.1038/s41419-020-02972-2 Cell Death & Disease ARTICLE Open Access Preclinical evaluation of a regimen combining chidamide and ABT-199 in acute myeloid leukemia Kai Chen 1,2,3, Qianying Yang4, Jie Zha2,ManmanDeng2, Yong Zhou2,GuofengFu4, Silei Bi2, Liying Feng2, Zijun Y. Xu-Monette5,XiaoLeiChen 4,GuoFu 4,YunDai6, Ken H. Young5 and Bing Xu1,2 Abstract Acute myeloid leukemia (AML) is a heterogeneous myeloid neoplasm with poor clinical outcome, despite the great progress in treatment in recent years. The selective Bcl-2 inhibitor venetoclax (ABT-199) in combination therapy has been approved for the treatment of newly diagnosed AML patients who are ineligible for intensive chemotherapy, but resistance can be acquired through the upregulation of alternative antiapoptotic proteins. Here, we reported that a newly emerged histone deacetylase inhibitor, chidamide (CS055), at low-cytotoxicity dose enhanced the anti-AML activity of ABT-199, while sparing normal hematopoietic progenitor cells. Moreover, we also found that chidamide showed a superior resensitization effect than romidepsin in potentiation of ABT-199 lethality. Inhibition of multiple HDACs rather than some single component might be required. The combination therapy was also effective in primary AML blasts and stem/progenitor cells regardless of disease status and genetic aberrance, as well as in a patient-derived xenograft model carrying FLT3-ITD mutation. Mechanistically, CS055 promoted leukemia suppression through DNA double-strand break and altered unbalance of anti- and pro-apoptotic proteins (e.g., Mcl-1 and Bcl-xL downregulation, and Bim upregulation).
    [Show full text]
  • (Hdacs) Guided Novel Therapies for T-Cell Lymphomas Qing Zhang1, Shaobin Wang2, Junhui Chen1, Zhendong Yu3
    Int. J. Med. Sci. 2019, Vol. 16 424 Ivyspring International Publisher International Journal of Medical Sciences 2019; 16(3): 424-442. doi: 10.7150/ijms.30154 Review Histone Deacetylases (HDACs) Guided Novel Therapies for T-cell lymphomas Qing Zhang1, Shaobin Wang2, Junhui Chen1, Zhendong Yu3 1. Department of Minimally Invasive Intervention, Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China. 2. Health Management Center of Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China. 3. China Central Laboratory of Peking University Shenzhen Hospital, Shenzhen, Guangdong, 518036, China. Corresponding authors: Dr. Qing Zhang, email: [email protected]; Dr. Zhendong Yu, email: [email protected] © Ivyspring International Publisher. This is an open access article distributed under the terms of the Creative Commons Attribution (CC BY-NC) license (https://creativecommons.org/licenses/by-nc/4.0/). See http://ivyspring.com/terms for full terms and conditions. Received: 2018.09.24; Accepted: 2018.12.19; Published: 2019.01.29 Abstract T-cell lymphomas are a heterogeneous group of cancers with different pathogenesis and poor prognosis. Histone deacetylases (HDACs) are epigenetic modifiers that modulate many key biological processes. In recent years, HDACs have been fully investigated for their roles and potential as drug targets in T-cell lymphomas. In this review, we have deciphered the modes of action of HDACs, HDAC inhibitors as single agents, and HDACs guided combination therapies in T-cell lymphomas. The overview of HDACs on the stage of T-cell lymphomas, and HDACs guided therapies both as single agents and combination regimens endow great opportunities for the cure of T-cell lymphomas. Key words: Histone deacetylases (HDACs), T-cell lymphomas, cutaneous T-cell lymphoma, peripheral T-cell lymphoma, combination therapy 1.
    [Show full text]
  • Why ACE—Overview of the Development of the Subtype-Selective Histone Deacetylase Inhibitor Chidamide in Hormone Receptor Positive Advanced Breast Cancer
    11 Review Article Page 1 of 11 Why ACE—overview of the development of the subtype-selective histone deacetylase inhibitor chidamide in hormone receptor positive advanced breast cancer Zhiqiang Ning Shenzhen Chipscreen Biosciences Ltd., Shenzhen 518057, China Correspondence to: Zhiqiang Ning. Shenzhen Chipscreen Biosciences Ltd., Bio-Incubator 2-601, 1st Ave. of Gaoxin Road, Hi-Tech Industrial Park, Shenzhen 518057, China. Email: [email protected]. Abstract: ACE stands for “A Phase III Trial of Chidamide in Combination with Exemestane in Patients with Hormone Receptor-Positive Advanced Breast Cancer”. Chidamide, or by its International Nonproprietary Name (INN), tucidinostat, is an oral subtype-selective histone deacetylase (HDAC) inhibitor with unique epigenetic modulating mechanisms for anti-cancer treatment. Chidamide has been approved in China for the indications of relapsed or refractory peripheral T-cell lymphoma, and hormone receptor-positive (HR+) advanced breast cancer. In the current review, epigenetic regulations and HDACs, the potential roles of epigenetic aberrations in disease progression and resistance to endocrine therapy in HR+ breast cancer, and the development of HDAC inhibitors as a promising therapeutic strategy in the field are overlooked. Then the review focuses on the ACE study in its rationale, trial design and results that led to the first-in-disease approval of chidamide in the epigenetic modulating drug class in breast cancer. Keywords: Chidamide/Tucidinostat; selective histone deacetylase inhibitor (selective HDAC inhibitor); breast cancer; endocrine therapy resistance Received: 03 March 2020; Accepted: 24 March 2020; Published: 10 April 2020. doi: 10.21037/tbcr.2020.03.06 View this article at: http://dx.doi.org/10.21037/tbcr.2020.03.06 Introduction therapies.
    [Show full text]
  • Improving Drug Discovery Using Image-Based Multiparametric Analysis of Epigenetic Landscape
    bioRxiv preprint doi: https://doi.org/10.1101/541151; this version posted February 5, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Improving drug discovery using image-based multiparametric analysis of epigenetic landscape. Chen Farhy1, Luis Orozco1, Fu-Yue Zeng1, Ian Pass1, Jarkko Ylanko2, Santosh Hariharan2, Chun-Teng Huang1, David Andrews2,3, and Alexey V. Terskikh1*. 1Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA 2Biological Sciences Platform, Sunnybrook Research Institute. 3Departments of Biochemistry and Medical Biophysics, University of Toronto, Ontario, Canada. *Correspondence should be addressed to A.V.T. ([email protected]). Abstract With the advent of automatic cell imaging and machine learning, high-content phenotypic screening has become the approach of choice for drug discovery due to its ability to extract drug specific multi- layered data and compare it to known profiles. In the field of epigenetics such screening approaches has suffered from the lack of tools sensitive to selective epigenetic perturbations. Here we describe a novel approach Microscopic Imaging of Epigenetic Landscapes (MIEL) that captures patterns of nuclear staining of epigenetic marks (e.g. acetylated and methylated histones) and employs machine learning to accurately distinguish between such patterns (1). We demonstrated that MIEL has superior resolution compared to conventional intensity thresholding techniques and enables efficient detection of epigenetically active compounds, function-based classification, flagging possible off-target effects and even predict novel drug function. We validated MIEL platform across multiple cells lines and using dose-response curves to insure the robustness of this approach for the high content high throughput drug discovery.
    [Show full text]
  • WO 2016/070089 A2 6 May 2016 (06.05.2016) W P O P C T
    (12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization International Bureau (10) International Publication Number (43) International Publication Date WO 2016/070089 A2 6 May 2016 (06.05.2016) W P O P C T (51) International Patent Classification: (81) Designated States (unless otherwise indicated, for every C07K 16/28 (2006.01) A61K 31/00 (2006.01) kind of national protection available): AE, AG, AL, AM, A61K 47/48 (2006.01) A61P 35/00 (2006.01) AO, AT, AU, AZ, BA, BB, BG, BH, BN, BR, BW, BY, BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, (21) International Application Number: DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, PCT/US2015/058389 HN, HR, HU, ID, IL, IN, IR, IS, JP, KE, KG, KN, KP, KR, (22) International Filing Date: KZ, LA, LC, LK, LR, LS, LU, LY, MA, MD, ME, MG, 30 October 2015 (30.10.201 5) MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SA, SC, (25) Filing Language: English SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ, TM, TN, (26) Publication Language: English TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA, ZM, ZW. (30) Priority Data: (84) Designated States (unless otherwise indicated, for every 62/073,824 31 October 2014 (3 1. 10.2014) US kind of regional protection available): ARIPO (BW, GH, GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, ST, SZ, (71) Applicant: ABBVIE BIOTHERAPEUTICS INC.
    [Show full text]
  • The Timeline of Epigenetic Drug Discovery: from Reality to Dreams A
    Ganesan et al. Clinical Epigenetics (2019) 11:174 https://doi.org/10.1186/s13148-019-0776-0 REVIEW Open Access The timeline of epigenetic drug discovery: from reality to dreams A. Ganesan1, Paola B. Arimondo2, Marianne G. Rots3, Carmen Jeronimo4,5 and María Berdasco6,7* Abstract The flexibility of the epigenome has generated an enticing argument to explore its reversion through pharmacological treatments as a strategy to ameliorate disease phenotypes. All three families of epigenetic proteins—readers, writers, and erasers—are druggable targets that can be addressed through small-molecule inhibitors. At present, a few drugs targeting epigenetic enzymes as well as analogues of epigenetic modifications have been introduced into the clinic use (e.g. to treat haematological malignancies), and a wide range of epigenetic-based drugs are undergoing clinical trials. Here, we describe the timeline of epigenetic drug discovery and development beginning with the early design based solely on phenotypic observations to the state-of-the-art rational epigenetic drug discovery using validated targets. Finally, we will highlight some of the major aspects that need further research and discuss the challenges that need to be overcome to implement epigenetic drug discovery into clinical management of human disorders. To turn into reality, researchers from various disciplines (chemists, biologists, clinicians) need to work together to optimise the drug engineering, read-out assays, and clinical trial design. Keywords: Epigenetics, DNA methylation, Histone modifications, Epidrugs, Therapy Background regions so as to register, signal, or perpetuate altered ac- Seventy-five years ago, the British biologist Conrad tivity states. Waddington coined the term epigenetics to describe the At the molecular level, epigenetics involves a highly com- mechanisms by which an organism stably adapts its plex and dynamically reversible set of structural modifica- phenotype to the environment [1].
    [Show full text]
  • Chidamide Increases the Sensitivity of Refractory Or Relapsed Acute Myeloid
    Wang et al. Journal of Experimental & Clinical Cancer Research (2020) 39:278 https://doi.org/10.1186/s13046-020-01792-8 RESEARCH Open Access Chidamide increases the sensitivity of refractory or relapsed acute myeloid leukemia cells to anthracyclines via regulation of the HDAC3 -AKT-P21-CDK2 signaling pathway Hao Wang1†, Yu-chen Liu1†, Cheng-ying Zhu1†, Fei Yan2, Meng-zhen Wang1, Xiao-su Chen3, Xiao-kai Wang4, Bao-xu Pang5, Yong-hui Li1, Dai-hong Liu1*, Chun-ji Gao1*, Shu-jun Liu6* and Li-ping Dou1* Abstract Background: Induction therapy for acute myeloid leukemia (AML) is an anthracycline-based chemotherapy regimen. However, many patients experience a relapse or exhibit refractory disease (R/R). There is an urgent need for more effective regimens to reverse anthracycline resistance in these patients. Methods: In this paper, Twenty-seven R/R AML patients with anthracycline resistance consecutively received chidamide in combination with anthracycline-based regimen as salvage therapy at the Chinese PLA General Hospital. Results: Of the 27 patients who had received one course of salvage therapy, 13 achieved a complete response and 1 achieved a partial response. We found that the HDAC3-AKT-P21-CDK2 signaling pathway was significantly upregulated in anthracycline-resistant AML cells compared to non-resistant cells. AML patients with higher levels of HDAC3 had lower event-free survival (EFS) and overall survival (OS) rates. Moreover, anthracycline-resistant AML cells are susceptible to chidamide, a histone deacetylase inhibitor which can inhibit cell proliferation, increase cell apoptosis and induce cell-cycle arrest in a time- and dose-dependent manner. Chidamide increases the sensitivity of anthracycline-resistant cells to anthracycline drugs, and these effects are associated with the inhibition of the HDAC3-AKT-P21-CDK2 signaling pathway.
    [Show full text]