1
|
Ciebiera M, Kociuba J, Ali M, Madueke-Laveaux OS, Yang Q, Bączkowska M, Włodarczyk M, Żeber-Lubecka N, Zarychta E, Corachán A, Alkhrait S, Somayeh V, Malasevskaia I, Łoziński T, Laudański P, Spaczynski R, Jakiel G, Al-Hendy A. Uterine fibroids: current research on novel drug targets and innovative therapeutic strategies. Expert Opin Ther Targets 2024; 28:669-687. [PMID: 39136530 DOI: 10.1080/14728222.2024.2390094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024]
Abstract
INTRODUCTION Uterine fibroids, the most common nonmalignant tumors affecting the female genital tract, are a significant medical challenge. This article focuses on the most recent studies that attempted to identify novel non-hormonal therapeutic targets and strategies in UF therapy. AREAS COVERED This review covers the analysis of the pharmacological and biological mechanisms of the action of natural substances and the role of the microbiome in reference to UFs. This study aimed to determine the potential role of these compounds in UF prevention and therapy. EXPERT OPINION While there are numerous approaches for treating UFs, available drug therapies for disease control have not been optimized yet. This review highlights the biological potential of vitamin D, EGCG and other natural compounds, as well as the microbiome, as promising alternatives in UF management and prevention. Although these substances have been quite well analyzed in this area, we still recommend conducting further studies, particularly randomized ones, in the field of therapy with these compounds or probiotics. Alternatively, as the quality of data continues to improve, we propose the consideration of their integration into clinical practice, in alignment with the patient's preferences and consent.
Collapse
Affiliation(s)
- Michal Ciebiera
- Second Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, Warsaw, Poland
- Warsaw Institute of Women's Health, Warsaw, Poland
- Development and Research Center of Non-Invasive Therapies, Pro-Familia Hospital, Rzeszow, Poland
| | - Jakub Kociuba
- Second Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, Warsaw, Poland
- Warsaw Institute of Women's Health, Warsaw, Poland
| | - Mohamed Ali
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
| | | | - Qiwei Yang
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
| | - Monika Bączkowska
- Second Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, Warsaw, Poland
| | - Marta Włodarczyk
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
- Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Natalia Żeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Center of Postgraduate Medical Education, Warsaw, Poland
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Elżbieta Zarychta
- Second Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, Warsaw, Poland
| | - Ana Corachán
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain
| | - Samar Alkhrait
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
| | - Vafaei Somayeh
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
| | | | - Tomasz Łoziński
- Development and Research Center of Non-Invasive Therapies, Pro-Familia Hospital, Rzeszow, Poland
- Department of Obstetrics and Gynecology, Pro-Familia Hospital, Rzeszow, Poland
- Department of Gynecology and Obstetrics, Institute of Medical Sciences, Medical College of Rzeszow University, Rzeszow, Poland
| | - Piotr Laudański
- Department of Obstetrics, Gynecology and Gynecological Oncology, Medical University of Warsaw, Warsaw, Poland
- Women's Health Research Institute, Calisia University, Kalisz, Poland
- OVIklinika Infertility Center, Warsaw, Poland
| | - Robert Spaczynski
- Center for Gynecology, Obstetrics and Infertility Treatment, Poznan, Poland
- Collegium Medicum, University of Zielona Gora, Zielona Gora, Poland
| | - Grzegorz Jakiel
- First Department of Obstetrics and Gynecology, Center of Postgraduate Medical Education, Warsaw, Poland
| | - Ayman Al-Hendy
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL, USA
| |
Collapse
|
2
|
Wang B, Wang J, Yang W, Zhao L, Wei B, Chen J. Unveiling Allosteric Regulation and Binding Mechanism of BRD9 through Molecular Dynamics Simulations and Markov Modeling. Molecules 2024; 29:3496. [PMID: 39124901 PMCID: PMC11314499 DOI: 10.3390/molecules29153496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Bromodomain-containing protein 9 (BRD9) is a key player in chromatin remodeling and gene expression regulation, and it is closely associated with the development of various diseases, including cancers. Recent studies have indicated that inhibition of BRD9 may have potential value in the treatment of certain cancers. Molecular dynamics (MD) simulations, Markov modeling and principal component analysis were performed to investigate the binding mechanisms of allosteric inhibitor POJ and orthosteric inhibitor 82I to BRD9 and its allosteric regulation. Our results indicate that binding of these two types of inhibitors induces significant structural changes in the protein, particularly in the formation and dissolution of α-helical regions. Markov flux analysis reveals notable changes occurring in the α-helicity near the ZA loop during the inhibitor binding process. Calculations of binding free energies reveal that the cooperation of orthosteric and allosteric inhibitors affects binding ability of inhibitors to BRD9 and modifies the active sites of orthosteric and allosteric positions. This research is expected to provide new insights into the inhibitory mechanism of 82I and POJ on BRD9 and offers a theoretical foundation for development of cancer treatment strategies targeting BRD9.
Collapse
Affiliation(s)
- Bin Wang
- Center for Medical Artificial Intelligence, Shandong University of Traditional Chinese Medicine, Qingdao 266112, China;
| | - Jian Wang
- School of Science, Shandong Jiaotong University, Jinan 250357, China; (J.W.); (W.Y.); (L.Z.)
| | - Wanchun Yang
- School of Science, Shandong Jiaotong University, Jinan 250357, China; (J.W.); (W.Y.); (L.Z.)
| | - Lu Zhao
- School of Science, Shandong Jiaotong University, Jinan 250357, China; (J.W.); (W.Y.); (L.Z.)
| | - Benzheng Wei
- Center for Medical Artificial Intelligence, Shandong University of Traditional Chinese Medicine, Qingdao 266112, China;
| | - Jianzhong Chen
- School of Science, Shandong Jiaotong University, Jinan 250357, China; (J.W.); (W.Y.); (L.Z.)
| |
Collapse
|
3
|
Mohan N, Dashwood RH, Rajendran P. A-Z of Epigenetic Readers: Targeting Alternative Splicing and Histone Modification Variants in Cancer. Cancers (Basel) 2024; 16:1104. [PMID: 38539439 PMCID: PMC10968829 DOI: 10.3390/cancers16061104] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/03/2024] [Accepted: 03/06/2024] [Indexed: 11/03/2024] Open
Abstract
Epigenetic 'reader' proteins, which have evolved to interact with specific chromatin modifications, play pivotal roles in gene regulation. There is growing interest in the alternative splicing mechanisms that affect the functionality of such epigenetic readers in cancer etiology. The current review considers how deregulation of epigenetic processes and alternative splicing events contribute to pathophysiology. An A-Z guide of epigenetic readers is provided, delineating the antagonistic 'yin-yang' roles of full-length versus spliced isoforms, where this is known from the literature. The examples discussed underscore the key contributions of epigenetic readers in transcriptional regulation, early development, and cancer. Clinical implications are considered, offering insights into precision oncology and targeted therapies focused on epigenetic readers that have undergone alternative splicing events during disease pathogenesis. This review underscores the fundamental importance of alternative splicing events in the context of epigenetic readers while emphasizing the critical need for improved understanding of functional diversity, regulatory mechanisms, and future therapeutic potential.
Collapse
Affiliation(s)
- Nivedhitha Mohan
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX 77030, USA
- Department of Translational Medical Sciences, Antibody & Biopharmaceuticals Core, Texas A&M School of Medicine, Houston, TX 77030, USA
| | - Roderick H. Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX 77030, USA
- Department of Translational Medical Sciences, Antibody & Biopharmaceuticals Core, Texas A&M School of Medicine, Houston, TX 77030, USA
| | - Praveen Rajendran
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX 77030, USA
- Department of Translational Medical Sciences, Antibody & Biopharmaceuticals Core, Texas A&M School of Medicine, Houston, TX 77030, USA
| |
Collapse
|
4
|
Ross SA, Emenaker NJ, Kumar A, Riscuta G, Biswas K, Gupta S, Mohammed A, Shoemaker RH. Green Cancer Prevention and Beyond. Cancer Prev Res (Phila) 2024; 17:107-118. [PMID: 38251904 PMCID: PMC10911807 DOI: 10.1158/1940-6207.capr-23-0308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/13/2023] [Accepted: 01/18/2024] [Indexed: 01/23/2024]
Abstract
The concept of green chemoprevention was introduced in 2012 by Drs. Jed Fahey and Thomas Kensler as whole-plant foods and/or extract-based interventions demonstrating cancer prevention activity. Refining concepts and research demonstrating proof-of-principle approaches are highlighted within this review. Early approaches included extensively investigated whole foods, including broccoli sprouts and black raspberries showing dose-responsive effects across a range of activities in both animals and humans with minimal or no apparent toxicity. A recent randomized crossover trial evaluating the detoxification of tobacco carcinogens by a broccoli seed and sprout extract in the high-risk cohort of current smokers highlights the use of a dietary supplement as a potential next-generation green chemoprevention or green cancer prevention approach. Challenges are addressed, including the selection of dose, duration and mode of delivery, choice of control group, and standardization of the plant food or extract. Identification and characterization of molecular targets and careful selection of high-risk cohorts for study are additional important considerations when designing studies. Goals for precision green cancer prevention include acquiring robust evidence from carefully controlled human studies linking plant foods, extracts, and compounds to modulation of targets for cancer risk reduction in individual cancer types.
Collapse
Affiliation(s)
- Sharon A. Ross
- Division of Cancer Prevention, Nutritional Sciences Research Group, National Cancer Institute, Rockville, Maryland
| | - Nancy J. Emenaker
- Division of Cancer Prevention, Nutritional Sciences Research Group, National Cancer Institute, Rockville, Maryland
| | - Amit Kumar
- Division of Cancer Prevention, Nutritional Sciences Research Group, National Cancer Institute, Rockville, Maryland
| | - Gabriela Riscuta
- Division of Cancer Prevention, Nutritional Sciences Research Group, National Cancer Institute, Rockville, Maryland
| | - Kajal Biswas
- Division of Cancer Prevention, Chemopreventive Agent Development Research Group, National Cancer Institute, Rockville, Maryland
| | - Shanker Gupta
- Division of Cancer Prevention, Chemopreventive Agent Development Research Group, National Cancer Institute, Rockville, Maryland
| | - Altaf Mohammed
- Division of Cancer Prevention, Chemopreventive Agent Development Research Group, National Cancer Institute, Rockville, Maryland
| | - Robert H. Shoemaker
- Division of Cancer Prevention, Chemopreventive Agent Development Research Group, National Cancer Institute, Rockville, Maryland
| |
Collapse
|
5
|
Kehrberg RJ, Bhyravbhatla N, Batra SK, Kumar S. Epigenetic regulation of cancer-associated fibroblast heterogeneity. Biochim Biophys Acta Rev Cancer 2023; 1878:188901. [PMID: 37120098 PMCID: PMC10375465 DOI: 10.1016/j.bbcan.2023.188901] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 04/13/2023] [Accepted: 04/22/2023] [Indexed: 05/01/2023]
Abstract
Cancer-associated fibroblasts (CAFs), a significant component of the tumor microenvironment (TME), contribute to cancer progression through the secretion of extracellular matrix (ECM), growth factors, and metabolites. It is now well recognized that CAFs are a heterogenous population with ablation experiments leading to reduced tumor growth and single-cell RNA sequencing demonstrating CAF subgroups. CAFs lack genetic mutations yet substantially differ from their normal stromal precursors. Here, we review epigenetic changes in CAF maturation, focusing on DNA methylation and histone modifications. DNA methylation changes in CAFs have been demonstrated globally, while roles of methylation at specific genes affect tumor growth. Further, loss of CAF histone methylation and gain of histone acetylation has been shown to promote CAF activation and tumor promotion. Many CAF activating factors, such as transforming growth factor β (TGFβ), lead to these epigenetic changes. MicroRNAs (miRNAs) serve as targets and orchestrators of epigenetic modifications that influence gene expression. Bromodomain and extra-terminal domain (BET), an epigenetic reader, recognizes histone acetylation and activates the transcription of genes leading to the pro-tumor phenotype of CAFs.
Collapse
Affiliation(s)
- Rachel J Kehrberg
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Namita Bhyravbhatla
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
6
|
Kapoor S, Damiani E, Wang S, Dharmanand R, Tripathi C, Tovar Perez JE, Dashwood WM, Rajendran P, Dashwood RH. BRD9 Inhibition by Natural Polyphenols Targets DNA Damage/Repair and Apoptosis in Human Colon Cancer Cells. Nutrients 2022; 14:nu14204317. [PMID: 36297001 PMCID: PMC9610492 DOI: 10.3390/nu14204317] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 11/07/2022] Open
Abstract
Epigenetic mechanisms play an important role in the etiology of colorectal cancer (CRC) and other malignancies due, in part, to deregulated bromodomain (BRD) functions. Inhibitors of the bromodomain and extraterminal (BET) family have entered into clinical trials as anticancer agents, and interest has grown in other acetyl 'reader' proteins as therapeutic targets, including non-BET member bromodomain-containing protein 9 (BRD9). We report here that overexpression of BRD9 is associated with poor prognosis in CRC patients, and that siRNA-mediated knockdown of BRD9 decreased cell viability and activated apoptosis in human colon cancer cells, coincident with increased DNA damage. Seeking natural compounds as BRD9 antagonists, molecular docking in silico identified several polyphenols such as Epigallocatechin-3-gallate (EGCG), Equol, Quercetin, and Aspalathin, with favorable binding energies, supported by BROMOscan® (DiscoverX) and isothermal titration calorimetry experiments. Polyphenols mimicked BRD9 knockdown and iBRD9 treatment in reducing colon cancer cell viability, inhibiting colony formation, and enhancing DNA damage and apoptosis. Normal colonic epithelial cells were unaffected, signifying cancer-specific effects. These findings suggest that natural polyphenols recognize and target BRD9 for inhibition, and might serve as useful lead compounds for bromodomain therapeutics in the clinical setting.
Collapse
Affiliation(s)
- Sabeeta Kapoor
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX 77030, USA
| | - Elisabetta Damiani
- Department of Life and Environmental Sciences, Università Politecnica delle Marche, 60121 Ancona, Italy
| | - Shan Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Hangzhou 310030, China
| | - Ravirajan Dharmanand
- Center for Infectious & Inflammatory Diseases, Texas A&M Health, Houston, TX 77030, USA
| | - Chakrapani Tripathi
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX 77030, USA
| | | | - Wan Mohaiza Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX 77030, USA
| | - Praveen Rajendran
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX 77030, USA
- Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA
- Correspondence: (P.R.); (R.H.D.); Tel.: +1-713-677-7803 (P.R.); +1-713-677-7806 (R.H.D.)
| | - Roderick Hugh Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Houston, TX 77030, USA
- Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA
- Correspondence: (P.R.); (R.H.D.); Tel.: +1-713-677-7803 (P.R.); +1-713-677-7806 (R.H.D.)
| |
Collapse
|
7
|
Ruzic D, Djoković N, Srdić-Rajić T, Echeverria C, Nikolic K, Santibanez JF. Targeting Histone Deacetylases: Opportunities for Cancer Treatment and Chemoprevention. Pharmaceutics 2022; 14:pharmaceutics14010209. [PMID: 35057104 PMCID: PMC8778744 DOI: 10.3390/pharmaceutics14010209] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/06/2022] [Accepted: 01/12/2022] [Indexed: 02/06/2023] Open
Abstract
The dysregulation of gene expression is a critical event involved in all steps of tumorigenesis. Aberrant histone and non-histone acetylation modifications of gene expression due to the abnormal activation of histone deacetylases (HDAC) have been reported in hematologic and solid types of cancer. In this sense, the cancer-associated epigenetic alterations are promising targets for anticancer therapy and chemoprevention. HDAC inhibitors (HDACi) induce histone hyperacetylation within target proteins, altering cell cycle and proliferation, cell differentiation, and the regulation of cell death programs. Over the last three decades, an increasing number of synthetic and naturally derived compounds, such as dietary-derived products, have been demonstrated to act as HDACi and have provided biological and molecular insights with regard to the role of HDAC in cancer. The first part of this review is focused on the biological roles of the Zinc-dependent HDAC family in malignant diseases. Accordingly, the small-molecules and natural products such as HDACi are described in terms of cancer therapy and chemoprevention. Furthermore, structural considerations are included to improve the HDACi selectivity and combinatory potential with other specific targeting agents in bifunctional inhibitors and proteolysis targeting chimeras. Additionally, clinical trials that combine HDACi with current therapies are discussed, which may open new avenues in terms of the feasibility of HDACi’s future clinical applications in precision cancer therapies.
Collapse
Affiliation(s)
- Dusan Ruzic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.R.); (N.D.); (K.N.)
| | - Nemanja Djoković
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.R.); (N.D.); (K.N.)
| | - Tatjana Srdić-Rajić
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia;
| | - Cesar Echeverria
- Facultad de Medicina, Universidad de Atacama, Copayapu 485, Copiapo 1531772, Chile;
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe 450, 11221 Belgrade, Serbia; (D.R.); (N.D.); (K.N.)
| | - Juan F. Santibanez
- Group for Molecular Oncology, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Dr. Subotica 4, POB 102, 11129 Belgrade, Serbia
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, Santiago 8370854, Chile
- Correspondence: ; Tel.: +381-11-2685-788; Fax: +381-11-2643-691
| |
Collapse
|
8
|
Chun KS, Raut PK, Kim DH, Surh YJ. Role of chemopreventive phytochemicals in NRF2-mediated redox homeostasis in humans. Free Radic Biol Med 2021; 172:699-715. [PMID: 34214633 DOI: 10.1016/j.freeradbiomed.2021.06.031] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/14/2021] [Accepted: 06/24/2021] [Indexed: 12/17/2022]
Abstract
While functioning as a second messenger in the intracellular signaling, ROS can cause oxidative stress when produced in excess or not neutralized/eliminated properly. Excessive ROS production is implicated in multi-stage carcinogenesis. Our body is equipped with a defense system to cope with constant oxidative stress caused by the external insults, including redox-cycling chemicals, radiation, and microbial infection as well as endogenously generated ROS. The transcription factor, nuclear transcription factor erythroid 2-related factor 2 (NRF2) is a master switch in the cellular antioxidant signaling and plays a vital role in adaptive survival response to ROS-induced oxidative stress. Although NRF2 is transiently activated when cellular redox balance is challenged, this can be overwhelmed by massive oxidative stress. Therefore, it is necessary to maintain the NRF2-mediated antioxidant defense capacity at an optimal level. This review summarizes the natural NRF2 inducers/activators, especially those present in the plant-based diet, in relation to their cancer chemopreventive potential in humans. The molecular mechanisms underlying their stabilization or activation of NRF2 are also discussed.
Collapse
Affiliation(s)
- Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 42691, South Korea
| | - Pawan Kumar Raut
- College of Pharmacy, Keimyung University, Daegu 42691, South Korea
| | - Do-Hee Kim
- Department of Chemistry, College of Convergence and Integrated Science, Kyonggi University, Suwon, Gyeonggi-do 16227, South Korea
| | - Young-Joon Surh
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, South Korea; Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, South Korea; Cancer Research Institute, Seoul National University, Seoul 03080, South Korea.
| |
Collapse
|
9
|
Miller MS, Allen PJ, Brown PH, Chan AT, Clapper ML, Dashwood RH, Demehri S, Disis ML, DuBois RN, Glynn RJ, Kensler TW, Khan SA, Johnson BD, Liby KT, Lipkin SM, Mallery SR, Meuillet EJ, Roden RB, Schoen RE, Sharp ZD, Shirwan H, Siegfried JM, Rao CV, You M, Vilar E, Szabo E, Mohammed A. Meeting Report: Translational Advances in Cancer Prevention Agent Development Meeting. J Cancer Prev 2021; 26:71-82. [PMID: 33842408 PMCID: PMC8020174 DOI: 10.15430/jcp.2021.26.1.71] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 12/11/2022] Open
Abstract
The Division of Cancer Prevention of the National Cancer Institute (NCI) and the Office of Disease Prevention of the National Institutes of Health co-sponsored the Translational Advances in Cancer Prevention Agent Development Meeting on August 27 to 28, 2020. The goals of this meeting were to foster the exchange of ideas and stimulate new collaborative interactions among leading cancer prevention researchers from basic and clinical research; highlight new and emerging trends in immunoprevention and chemoprevention as well as new information from clinical trials; and provide information to the extramural research community on the significant resources available from the NCI to promote prevention agent development and rapid translation to clinical trials. The meeting included two plenary talks and five sessions covering the range from pre-clinical studies with chemo/immunopreventive agents to ongoing cancer prevention clinical trials. In addition, two NCI informational sessions describing contract resources for the preclinical agent development and cooperative grants for the Cancer Prevention Clinical Trials Network were also presented.
Collapse
Affiliation(s)
- Mark Steven Miller
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland, USA
| | - Peter J. Allen
- Division of Surgical Oncology, Duke Cancer Institute, Durham, NC, USA
| | - Powel H. Brown
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, MD Anderson Cancer Center, Houston, TX, USA
| | - Andrew T. Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Margie L. Clapper
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Roderick H. Dashwood
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, Texas A&M Health Science Center, Houston, TX, USA
| | - Shadmehr Demehri
- Department of Dermatology, Massachusetts General Hospital, Boston, MA, USA
| | - Mary L. Disis
- Cancer Vaccine Institute, University of Washington School of Medicine, Seattle, WA, USA
| | - Raymond N. DuBois
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Robert J. Glynn
- Division of Preventive Medicine, Brigham & Women’s Hospital, Boston, MA, USA
| | - Thomas W. Kensler
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Seema A. Khan
- Department of Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Bryon D. Johnson
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Karen T. Liby
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, Michigan, NY, USA
| | - Steven M. Lipkin
- Division of Gastroenterology and Hepatology, Weill Cornell University, New York, NY, USA
| | - Susan R. Mallery
- Department of Oral and Maxillofacial Pathology, College of Dentistry, Ohio State University, Columbus, OH, USA
| | | | - Richard B.S. Roden
- Department of Pathology, Cancer Prevention and Control Program, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Robert E. Schoen
- Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zelton D. Sharp
- Department of Molecular Medicine, University of Texas Science Center at San Antonio, San Antonio, TX, USA
| | - Haval Shirwan
- Department of Child Health and Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - Jill M. Siegfried
- Department of Pharmacology, Masonic Cancer Center, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Chinthalapally V. Rao
- Medical Oncology Center for Cancer Prevention & Drug Development, Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Ming You
- Department of Pharmacology and Toxicology, Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, Division of Cancer Prevention and Population Sciences, MD Anderson Cancer Center, Houston, TX, USA
| | - Eva Szabo
- Lung and Upper Aerodigestive Cancer Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, MD, USA
| | - Altaf Mohammed
- Chemopreventive Agent Development Research Group, Division of Cancer Prevention, National Cancer Institute, Rockville, Maryland, USA
| |
Collapse
|
10
|
Kapoor S, Gustafson T, Zhang M, Chen YS, Li J, Nguyen N, Perez JET, Dashwood WM, Rajendran P, Dashwood RH. Deacetylase Plus Bromodomain Inhibition Downregulates ERCC2 and Suppresses the Growth of Metastatic Colon Cancer Cells. Cancers (Basel) 2021; 13:cancers13061438. [PMID: 33809839 PMCID: PMC8004213 DOI: 10.3390/cancers13061438] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/19/2021] [Indexed: 01/14/2023] Open
Abstract
There is growing evidence that DNA repair factors have clinical value for cancer treatment. Nucleotide excision repair (NER) proteins, including excision repair cross-complementation group 2 (ERCC2), play a critical role in maintaining genome integrity. Here, we examined ERCC2 expression following epigenetic combination drug treatment. Attention was drawn to ERCC2 for three reasons. First, from online databases, colorectal cancer (CRC) patients exhibited significantly reduced survival when ERCC2 was overexpressed in colon tumors. Second, ERCC2 was the most highly downregulated RNA transcript in human colon cancer cells, plus Ercc2 in rat tumors, after treatment with the histone deacetylase 3 (HDAC3) inhibitor sulforaphane (SFN) plus JQ1, which is an inhibitor of the bromodomain and extraterminal domain (BET) family. Third, as reported here, RNA-sequencing of polyposis in rat colon (Pirc) polyps following treatment of rats with JQ1 plus 6-methylsulfinylhexyl isothiocyanate (6-SFN) identified Ercc2 as the most highly downregulated gene. The current work also defined promising second-generation epigenetic drug combinations with enhanced synergy and efficacy, especially in metastasis-lineage colon cancer cells cultured as 3D spheroids and xenografts. This investigation adds to the growing interest in combination approaches that target epigenetic 'readers', 'writers', and 'erasers' that are deregulated in cancer and other pathologies, providing new avenues for precision oncology and cancer interception.
Collapse
Affiliation(s)
- Sabeeta Kapoor
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Trace Gustafson
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Mutian Zhang
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Ying-Shiuan Chen
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Jia Li
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Nhung Nguyen
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Jorge Enrique Tovar Perez
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Wan Mohaiza Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Praveen Rajendran
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
- Antibody & Biopharmaceuticals Core, Texas A&M Health, Houston, TX 77030, USA
- Correspondence: (P.R.); (R.H.D.)
| | - Roderick H. Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
- Correspondence: (P.R.); (R.H.D.)
| |
Collapse
|