1
|
Poirier D. Recent advances in the development of 17beta-hydroxysteroid dehydrogenase inhibitors. Steroids 2025; 213:109529. [PMID: 39532224 DOI: 10.1016/j.steroids.2024.109529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 11/08/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
The family of 17β-hydroxysteroid dehydrogenases (17β-HSDs) occupies a prominent place due to its number of isoforms, which carry out a bidirectional transformation (reduction of a steroid carbonyl to alcohol and oxidation of a steroid alcohol to ketone) depending on the nature of the cofactor present. Involved in the activation or inactivation of key estrogens and androgens, 17β-HSDs are therefore therapeutic targets whose selective inhibition would make it possible to be considered for the treatment of several diseases, such as breast cancer, prostate cancer, endometriosis, Alzheimer's disease and osteoporosis. This review article is a continuation of those having reported the great diversity of inhibitors developed over the last years but focusses on inhibitors recently developed. Work to obtain more effective inhibitors that target the first known isoforms (types 1, 2, 3, 5 and 7) has continued, among others, but new inhibitors that target the isoforms more recently reported in the literature (types 10, 12, 13 and 14) are now being reported. Dual inhibitors of two enzymes (17β-HSD1 and steroid sulfatase) were also reported. These inhibitors were grouped according to the 17β-HSD type inhibited and their backbone (steroidal or non-steroidal) when necessary. They were also reported in chronological order and according to the research group.
Collapse
Affiliation(s)
- Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec Research Center-Université Laval, Québec, QC G1V 4G2, Canada; Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
2
|
Jonnalagadda SK, Duan L, Dow LF, Boligala GP, Kosmacek E, McCoy K, Oberley-Deegan R, Chhonker YS, Murry DJ, Reynolds CP, Maurer BJ, Penning TM, Trippier PC. Coumarin-Based Aldo-Keto Reductase Family 1C (AKR1C) 2 and 3 Inhibitors. ChemMedChem 2024; 19:e202400081. [PMID: 38976686 PMCID: PMC11537819 DOI: 10.1002/cmdc.202400081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/10/2024]
Abstract
A series of 7-substituted coumarin derivatives have been characterized as pan-aldo-keto reductase family 1C (AKR1C) inhibitors. The AKR1C family of enzymes are overexpressed in numerous cancers where they are involved in drug resistance development. 7-hydroxy coumarin ethyl esters and their corresponding amides have high potency for AKR1C3 and AKR1C2 inhibition. Coumarin amide 3 a possessed IC50 values of 50 nM and 90 nM for AKR1C3 and AKR1C2, respectively, and exhibits 'drug-like' metabolic stability and half-life in human and mouse liver microsomes and plasma. Compound 3 a was employed as a chemical tool to determine pan-AKR1C2/3 inhibition effects both as a radiation sensitizer and as a potentiator of chemotherapy cytotoxicity. In contrast to previously reported pan-AKR1C inhibitors, 3 a demonstrated no radiation sensitization effect in a radiation-resistant prostate cancer cell line model. Pan-AKR1C inhibition also did not potentiate the in vitro cytotoxicity of ABT-737, daunorubicin or dexamethasone, in two patient-derived T-cell ALL and pre-B-cell ALL cell lines. In contrast, a highly selective AKR1C3 inhibitor, compound K90, enhanced the cytotoxicity of both ABT-737 and daunorubicin in the T-cell ALL cell line model. Thus, the inhibitory profile required to enhance chemotherapeutic cytotoxicity in leukemia may be AKR1C isoform and drug specific.
Collapse
Affiliation(s)
- Sravan K. Jonnalagadda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Ling Duan
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Louise F. Dow
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Geetha P Boligala
- School of Medicine Cancer Center, Texas Tech University Health Sciences Center, Lubbock, Texas, 79430, United States
| | - Elizabeth Kosmacek
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Kristyn McCoy
- School of Medicine Cancer Center, Texas Tech University Health Sciences Center, Lubbock, Texas, 79430, United States
| | - Rebecca Oberley-Deegan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Yashpal Singh Chhonker
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68106, USA
| | - Darryl J. Murry
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68106, USA
- UNMC Center for Drug Design and Innovation, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - C. Patrick Reynolds
- School of Medicine Cancer Center, Texas Tech University Health Sciences Center, Lubbock, Texas, 79430, United States
| | - Barry J. Maurer
- School of Medicine Cancer Center, Texas Tech University Health Sciences Center, Lubbock, Texas, 79430, United States
| | - Trevor M. Penning
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Paul C. Trippier
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
- UNMC Center for Drug Design and Innovation, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| |
Collapse
|
3
|
Vastrad B, Vastrad C. Screening and identification of key biomarkers associated with endometriosis using bioinformatics and next-generation sequencing data analysis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:116. [DOI: 10.1186/s43042-024-00572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 01/04/2025] Open
Abstract
Abstract
Background
Endometriosis is a common cause of endometrial-type mucosa outside the uterine cavity with symptoms such as painful periods, chronic pelvic pain, pain with intercourse and infertility. However, the early diagnosis of endometriosis is still restricted. The purpose of this investigation is to identify and validate the key biomarkers of endometriosis.
Methods
Next-generation sequencing dataset GSE243039 was obtained from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) between endometriosis and normal control samples were identified. After screening of DEGs, gene ontology (GO) and REACTOME pathway enrichment analyses were performed. Furthermore, a protein–protein interaction (PPI) network was constructed and modules were analyzed using the Human Integrated Protein–Protein Interaction rEference database and Cytoscape software, and hub genes were identified. Subsequently, a network between miRNAs and hub genes, and network between TFs and hub genes were constructed using the miRNet and NetworkAnalyst tool, and possible key miRNAs and TFs were predicted. Finally, receiver operating characteristic curve analysis was used to validate the hub genes.
Results
A total of 958 DEGs, including 479 upregulated genes and 479 downregulated genes, were screened between endometriosis and normal control samples. GO and REACTOME pathway enrichment analyses of the 958 DEGs showed that they were mainly involved in multicellular organismal process, developmental process, signaling by GPCR and muscle contraction. Further analysis of the PPI network and modules identified 10 hub genes, including vcam1, snca, prkcb, adrb2, foxq1, mdfi, actbl2, prkd1, dapk1 and actc1. Possible target miRNAs, including hsa-mir-3143 and hsa-mir-2110, and target TFs, including tcf3 (transcription factor 3) and clock (clock circadian regulator), were predicted by constructing a miRNA-hub gene regulatory network and TF-hub gene regulatory network.
Conclusions
This investigation used bioinformatics techniques to explore the potential and novel biomarkers. These biomarkers might provide new ideas and methods for the early diagnosis, treatment and monitoring of endometriosis.
Collapse
|
4
|
Xiong W, Xie Y, Wang D, Huang X, Hao X, Liu J, Liu X, Gu X, Sun S, Li Y, Li J. Aldo-keto reductase family 1 member C3 mediates radioresistance of esophageal cancer cells through suppressing MAPK and AKT signaling. BMC Cancer 2024; 24:1236. [PMID: 39375680 PMCID: PMC11459825 DOI: 10.1186/s12885-024-13012-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 09/30/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Aldo-keto reductase family 1 member C3 (AKR1C3) is a radioresistance gene in esophageal cancer. This study aimed to investigate the signaling pathways that mediate the regulatory role of AKR1C3 in the radioresistance of esophageal cancer cells. METHODS The protein levels of AKR1C3 in cancer tissue samples were compared between patients with radiosensitive and radioresistant esophageal cancer using immunohistochemical staining. AKR1C3-silenced stable KYSE170R esophageal cancer cells (KY170R-shAKR1C3) were established. Colony formation assay was employed to evaluate the radiosensitivity of cancer cells, while flow cytometry analysis was utilized to quantify reactive oxygen species (ROS) production in these cells. Additionally, Western blotting was conducted to determine protein expression levels. RESULTS AKR1C3 protein exhibited significantly higher expression in radioresistant cancer tissue samples compared to radiosensitive samples. AKR1C3 silencing promoted radiosensitivity and ROS production of KYSE170R cells. At 32 h after X-ray radiation, the levels of total and phosphorylated ERK1/2, JNK, and AKT proteins were significantly elevated in KYSE170R-shAKR1C3 cells compared to untransfected KYSE170R cells. The inhibitor of AKR1C3 remarkably enhanced the radiosensitivity of KYSE170R cells. Conversely, treatment with either a MEK inhibitor or an AKT inhibitor significantly increased the radioresistance of KYSE170R-shAKR1C3 cells. CONCLUSIONS Our results suggest that AKR1C3 mediates radioresistance of KYSE170R cells possibly through MAPK and AKT signaling.
Collapse
Affiliation(s)
- Wei Xiong
- Department of Radiation Oncology, Tangshan People's Hospital, Tangshan, Hebei, 063001, China
- Hebei Key Laboratory of Molecular Oncology, Tangshan People's Hospital, Tangshan, China
| | - Ya Xie
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Dong Wang
- Department of Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Xiaozhi Huang
- Department of Radiation Oncology, Tangshan People's Hospital, Tangshan, Hebei, 063001, China
| | - Xiaohui Hao
- School of Public Health, North China University of Science and Technology, Tangshan, 063210, China
| | - Jianming Liu
- Department of Gastroenterological Surgery, Tangshan People's Hospital, Tangshan, Hebei, 063001, China
| | - Xiaohui Liu
- Department of Gastroenterological Surgery, Tangshan People's Hospital, Tangshan, Hebei, 063001, China
| | - Xiaobin Gu
- Department of Radiation Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shaoqian Sun
- Biochemical Engineering College, Beijing Union University, Beijing, 100023, China
| | - Yufeng Li
- The Cancer Institute, Tangshan People's Hospital, Tangshan, Hebei, 063001, China
- Hebei Key Laboratory of Molecular Oncology, Tangshan People's Hospital, Tangshan, China
| | - Jingwu Li
- Department of Gastroenterological Surgery, Tangshan People's Hospital, Tangshan, Hebei, 063001, China.
- Hebei Key Laboratory of Molecular Oncology, Tangshan People's Hospital, Tangshan, China.
| |
Collapse
|
5
|
Pippione A, Vigato C, Tucciarello C, Hussain S, Salladini E, Truong HH, Henriksen NM, Vanzetti G, Giordano G, Zonari D, Mirza OA, Frydenvang K, Pignochino Y, Oliaro-Bosso S, Boschi D, Lolli ML. AI Based Discovery of a New AKR1C3 Inhibitor for Anticancer Applications. ACS Med Chem Lett 2024; 15:1269-1278. [PMID: 39140045 PMCID: PMC11318022 DOI: 10.1021/acsmedchemlett.4c00150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/23/2024] [Accepted: 06/10/2024] [Indexed: 08/15/2024] Open
Abstract
AKR1C3 is an upregulated enzyme in prostate and other cancers; in addition to regulating hormone synthesis, this enzyme is thought to play a role in the aggressiveness of tumors and in the defense against drugs. We here used an unbiased method to discover new potent AKR1C3 inhibitors: through an AI-based virtual drug screen, compound 4 was identified as a potent and selective enzymatic inhibitor able to translate this activity into a pronounced antiproliferative effect in the 22RV1 prostate cancer cell model. As other known AKR1C3 inhibitors, compound 4 determined a significantly increased activity of abiraterone, a drug approved for advanced prostate cancer. Compound 4 also showed a synergic effect with doxorubicin in osteosarcoma cell lines; specifically, the effect is correlated with AKR1C3 expression. In this research work, therefore, the use of AI allowed the identification of a new structure as an AKR1C3 inhibitor and its potential to enhance the effect of chemotherapeutics.
Collapse
Affiliation(s)
- Agnese
C. Pippione
- Department
of Science and Drug Technology, University
of Turin, via Pietro Giuria 9, 10125 Turin, Italy
| | - Chiara Vigato
- Department
of Science and Drug Technology, University
of Turin, via Pietro Giuria 9, 10125 Turin, Italy
| | - Cristina Tucciarello
- Candiolo
Cancer Institute, FPO-IRCCS, str. Prov 142 km 3.95, 10060 Candiolo, Turin, Italy
- Department
of Clinical and Biological Sciences, University
of Turin, Regione Gonzole 10, 10143 Orbassano, Italy
| | - Samrina Hussain
- Department
of Drug Design and Pharmacology, University
of Copenhagen, Jagtvej 162 DK-2100 Copenhagen, Denmark
| | - Edoardo Salladini
- Department
of Science and Drug Technology, University
of Turin, via Pietro Giuria 9, 10125 Turin, Italy
| | - Ha H. Truong
- Atomwise,
Inc, 250 Sutter St, Suite 650, San Francisco, California 94103, United States
| | - Niel M. Henriksen
- Atomwise,
Inc, 250 Sutter St, Suite 650, San Francisco, California 94103, United States
| | - Gaia Vanzetti
- Department
of Science and Drug Technology, University
of Turin, via Pietro Giuria 9, 10125 Turin, Italy
| | - Giorgia Giordano
- Candiolo
Cancer Institute, FPO-IRCCS, str. Prov 142 km 3.95, 10060 Candiolo, Turin, Italy
- Department
of Oncology, University of Turin, str. Prov 142 km 3.95, 10060 Candiolo, Turin, Italy
| | - Daniele Zonari
- Department
of Science and Drug Technology, University
of Turin, via Pietro Giuria 9, 10125 Turin, Italy
| | - Osman Asghar Mirza
- Department
of Drug Design and Pharmacology, University
of Copenhagen, Jagtvej 162 DK-2100 Copenhagen, Denmark
| | - Karla Frydenvang
- Department
of Drug Design and Pharmacology, University
of Copenhagen, Jagtvej 162 DK-2100 Copenhagen, Denmark
| | - Ymera Pignochino
- Candiolo
Cancer Institute, FPO-IRCCS, str. Prov 142 km 3.95, 10060 Candiolo, Turin, Italy
- Department
of Clinical and Biological Sciences, University
of Turin, Regione Gonzole 10, 10143 Orbassano, Italy
| | - Simonetta Oliaro-Bosso
- Department
of Science and Drug Technology, University
of Turin, via Pietro Giuria 9, 10125 Turin, Italy
| | - Donatella Boschi
- Department
of Science and Drug Technology, University
of Turin, via Pietro Giuria 9, 10125 Turin, Italy
| | - Marco L. Lolli
- Department
of Science and Drug Technology, University
of Turin, via Pietro Giuria 9, 10125 Turin, Italy
| |
Collapse
|
6
|
Huang X, Deng H. Construction of the circRNA-miRNA-mRNA axis based on ferroptosis-related gene AKR1C1 to explore the potential pathogenesis of abdominal aortic aneurysm. Medicine (Baltimore) 2024; 103:e38749. [PMID: 38941402 PMCID: PMC11466172 DOI: 10.1097/md.0000000000038749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 06/07/2024] [Indexed: 06/30/2024] Open
Abstract
Abdominal aortic aneurysm (AAA) is a cardiovascular disease that seriously threatens human health and brings huge economic burden. At present, its pathogenesis remains unclear and its treatment is limited to surgical treatment. With the deepening and analysis of studies on the mechanism of ferroptosis, a new idea has been provided for the clinical management of AAA patients, including diagnosis, treatment and prevention. Therefore, this paper aims to construct a competitive endogenous RNA (ceRNA) regulatory axis based on ferroptosis to preliminarily explore the pathogenesis and potential therapeutic targets of AAA. We obtained upregulated and downregulated ferroptosis-related DEGs (FRGs) from GSE144431 dataset and 60 known ferroptosis-related genes. Pearson correlation analysis was used to find aldoketone reductase 1C (AKR1C1) in AAA samples. Enrichment analysis of these genes was performed via Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). Correlation test between immune cells and AKR1C1 was investigated through single-sample gene set enrichment analysis (ssGSEA). The AKR1C1-miRNA pairs were predicted by the TargetScan database and miRWalk database. Circular RNA (CircRNA)-miRNA pairs were selected by the CircInteractome database. Overlapping miRNA between circRNA-miRNA and AKR1C1-miRNA pairs was visualized by Venn diagram. Finally, the circRNA-miRNA-mRNA axis was constructed by searching for upstream circRNA and downstream mRNA of overlapping miRNA. Only one downregulated AKR1C1 gene was found in GSE144431 and 60 ferroptosis-related genes. Functional Enrichment and Pathway Analysis of AKR1C1-related genes were further explored, and it was observed that they were mainly enriched in "response to oxidative stress," "glutathione biosynthetic process" and "nonribosomal peptide biosynthetic process," "Ferroptosis," "Glutathione metabolism" and "Chemical carcinogenesis-reactive oxygen species." They were also found to be significantly associated with most immune cells, including Activated Dendritic cells, CD56dim Natural killer cells, Gamma Delta T cells, Immature B cells, Plasmacytoid dendritic cell, Type 2 T helper cell, Activated CD4 T cell and Type 1 T helper cell. Has_circ_0005073-miRNA-543 and AKR1C1-miRNA-543 were identified by Online Database analysis. Therefore, we have established the has_circ_0005073/miRNA-543/AKR1C1 axis in AAA. We found AKR1C1 was differentially expressed between normal and AAA groups. Based on AKR1C1, we constructed the has_circ_0005073/miRNA-543/AKR1C1 axis to analyze AAA.
Collapse
Affiliation(s)
- Xuehua Huang
- Department of Neonatology, the First Hospital of China Medical University, Shenyang, China
| | - Huanhuan Deng
- Department of Nephrology, the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
7
|
Cuadrado-Torroglosa I, García-Velasco JA, Alecsandru D. The Impacts of Inflammatory and Autoimmune Conditions on the Endometrium and Reproductive Outcomes. J Clin Med 2024; 13:3724. [PMID: 38999290 PMCID: PMC11242609 DOI: 10.3390/jcm13133724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/17/2024] [Accepted: 06/23/2024] [Indexed: 07/14/2024] Open
Abstract
Background: A healthy pregnancy begins with an adequate endometrial state, even before the arrival of a blastocyst. Proper endometrial priming and the development of a tolerogenic decidua are key steps in creating the perfect environment for implantation and pregnancy. In these processes, the involvement of the maternal immune system seems to be of great relevance, modulating the different decidual immune populations to prepare the endometrium for a potential pregnancy. However, certain local pathologies of an inflammatory and autoimmune nature appear to have a direct impact on these phenomena, thus altering patients' reproductive outcomes. Methods: This literature review analyzes original articles, reviews, systematic reviews, and meta-analyses published between 1990 and 2024, concerning the impact of different inflammatory and autoimmune conditions on endometrial status and fertility. The included papers were obtained from Medline (Pubmed) and the Cochrane library. Results: There is evidence that endometriosis, adenomyosis, and chronic endometritis, through the promotion of a chronic inflammatory environment, are capable of altering endometrial immune populations, and, thus, processes essential for early pregnancy. Among other effects, these conditions have been linked to impaired decidualization, alterations in progesterone responsiveness, and hindered placentation. Similarly, antiphospholipid syndrome (APS), thyroid dysfunction, diabetes, and other pathologies related to glucose and gluten metabolism, due to their autoimmune nature, also appear to have a local impact on the uterine environment, affecting reproductive success through different mechanisms, including altered hormonal response and, again, impaired decidualization. Conclusions: The management of inflammatory and autoimmune diseases in assisted reproduction patients is gaining importance due to their direct impact on the endometrium. It is necessary to follow current expert recommendations and established therapeutic approaches in order to improve patients' prospects, ranging from antibiotic treatment in chronic endometritis to heparin and aspirin in APS, as well as hormonal treatments for endometriosis/adenomyosis or a gluten-free diet in celiac disease. All of them and the rest of the therapeutic perspectives, both current and under investigation, are presented throughout this work, assessing the possible improvements for reproductive outcomes.
Collapse
Affiliation(s)
- Isabel Cuadrado-Torroglosa
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Avenida Fernando Abril Martorell, 106, Torre A, Planta 1a, 46026 Valencia, Spain; (I.C.-T.); (J.A.G.-V.)
| | - Juan A. García-Velasco
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Avenida Fernando Abril Martorell, 106, Torre A, Planta 1a, 46026 Valencia, Spain; (I.C.-T.); (J.A.G.-V.)
- IVIRMA Global Research Alliance, IVIRMA Madrid, Av. del Talgo, 68, 28023 Madrid, Spain
- Department of Obstetrics and Gynaecology, Rey Juan Carlos University, Av. de Atenas, s/n, 28922 Alcorcón, Spain
| | - Diana Alecsandru
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Avenida Fernando Abril Martorell, 106, Torre A, Planta 1a, 46026 Valencia, Spain; (I.C.-T.); (J.A.G.-V.)
- IVIRMA Global Research Alliance, IVIRMA Madrid, Av. del Talgo, 68, 28023 Madrid, Spain
| |
Collapse
|
8
|
Tang W, Zhu X, Bian L, Zhang B. Research progress of dydrogesterone in the treatment of endometriosis. Eur J Obstet Gynecol Reprod Biol 2024; 296:120-125. [PMID: 38430648 DOI: 10.1016/j.ejogrb.2024.02.034] [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: 10/11/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
Endometriosis is a common gynecological disease among women of reproductive age. It is a chronic estrogen and progestin related inflammatory disease. At present, the main treatments for endometriosis are drug therapy and surgery. In drug therapy, progesterone is listed as the first-line recommendation in multinational guidelines. Dydrogesterone, as an oral reversal progesterone, can slow down the metabolism of progesterone, inhibit angiogenesis and extracellular matrix degradation to inhibit the proliferation of the ectopic endometrium, induce the atrophy of the ectopic endometrium through the pro-apoptotic pathway, and treat endometriosis through multiple mechanisms of regulating inflammatory factors to reduce inflammation. Clinically, dydrogesterone treatment of endometriosis can relieve patients' symptoms, promote fertility, be used in combination, and is safe. This article will review the mechanism and clinical application of dydrogesterone in the treatment of endometriosis.
Collapse
Affiliation(s)
- Wenlu Tang
- Department of Gynecology, Fujian Provincial Geriatric Hospital, Fuzhou 350003, Fujian Province, China; The First Clinical College of Fujian Medical University, Fuzhou 350004, Fujian Province, China
| | - Xiaohong Zhu
- The First Clinical College of Fujian Medical University, Fuzhou 350004, Fujian Province, China
| | - Lihong Bian
- The First Clinical College of Fujian Medical University, Fuzhou 350004, Fujian Province, China
| | - Bin Zhang
- The First Clinical College of Fujian Medical University, Fuzhou 350004, Fujian Province, China; Department of Obstetrics and Gynecology, The First Affiliated Hospital of Fujian Medical University, Fuzhou 350005, Fujian Province, China; Department of Gynecology, National Regional Medical Centre, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| |
Collapse
|
9
|
Pippione AC, Kovachka S, Vigato C, Bertarini L, Mannella I, Sainas S, Rolando B, Denasio E, Piercy-Mycock H, Romalho L, Salladini E, Adinolfi S, Zonari D, Peraldo-Neia C, Chiorino G, Passoni A, Mirza OA, Frydenvang K, Pors K, Lolli ML, Spyrakis F, Oliaro-Bosso S, Boschi D. Structure-guided optimization of 3-hydroxybenzoisoxazole derivatives as inhibitors of Aldo-keto reductase 1C3 (AKR1C3) to target prostate cancer. Eur J Med Chem 2024; 268:116193. [PMID: 38364714 DOI: 10.1016/j.ejmech.2024.116193] [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: 12/14/2023] [Revised: 01/16/2024] [Accepted: 01/25/2024] [Indexed: 02/18/2024]
Abstract
AKR1C3 is an enzyme that is overexpressed in several types of radiotherapy- and chemotherapy-resistant cancers. Despite AKR1C3 is a validated target for drug development, no inhibitor has been approved for clinical use. In this manuscript, we describe our study of a new series of potent AKR1C3-targeting 3-hydroxybenzoisoxazole based inhibitors that display high selectivity over the AKR1C2 isoform and low micromolar activity in inhibiting 22Rv1 prostate cancer cell proliferation. In silico studies suggested proper substituents to increase compound potency and provided with a mechanistic explanation that could clarify their different activity, later confirmed by X-ray crystallography. Both the in-silico studies and the crystallographic data highlight the importance of 90° rotation around the single bond of the biphenyl group, in ensuring that the inhibitor can adopt the optimal binding mode within the active pocket. The p-biphenyls that bear the meta-methoxy, and the ortho- and meta-trifluoromethyl substituents (in compounds 6a, 6e and 6f respectively) proved to be the best contributors to cellular potency as they provided the best IC50 values in series (2.3, 2.0 and 2.4 μM respectively) and showed no toxicity towards human MRC-5 cells. Co-treatment with scalar dilutions of either compound 6 or 6e and the clinically used drug abiraterone led to a significant reduction in cell proliferation, and thus confirmed that treatment with both CYP171A1-and AKR1C3-targeting compounds possess the potential to intervene in key steps in the steroidogenic pathway. Taken together, the novel compounds display desirable biochemical potency and cellular target inhibition as well as good in-vitro ADME properties, which highlight their potential for further preclinical studies.
Collapse
Affiliation(s)
- Agnese Chiara Pippione
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Sandra Kovachka
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Chiara Vigato
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Laura Bertarini
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy; Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 103, 41125, Modena, Italy
| | - Iole Mannella
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Stefano Sainas
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Barbara Rolando
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Enrica Denasio
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, West Yorkshire, BD7 1DP, UK
| | - Helen Piercy-Mycock
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, West Yorkshire, BD7 1DP, UK
| | - Linda Romalho
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, DK-2100, Copenhagen, Denmark
| | - Edoardo Salladini
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Salvatore Adinolfi
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Daniele Zonari
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Caterina Peraldo-Neia
- Laboratory of Cancer Genomics, Fondazione Edo ed Elvo Tempia, via Malta 3, 13900, Biella, Italy
| | - Giovanna Chiorino
- Laboratory of Cancer Genomics, Fondazione Edo ed Elvo Tempia, via Malta 3, 13900, Biella, Italy
| | - Alice Passoni
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Osman Asghar Mirza
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, DK-2100, Copenhagen, Denmark
| | - Karla Frydenvang
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, DK-2100, Copenhagen, Denmark
| | - Klaus Pors
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, West Yorkshire, BD7 1DP, UK
| | - Marco Lucio Lolli
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Francesca Spyrakis
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Simonetta Oliaro-Bosso
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy.
| | - Donatella Boschi
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy.
| |
Collapse
|
10
|
Li M, Zhang L, Yu J, Wang X, Cheng L, Ma Z, Chen X, Wang L, Goh BC. AKR1C3 in carcinomas: from multifaceted roles to therapeutic strategies. Front Pharmacol 2024; 15:1378292. [PMID: 38523637 PMCID: PMC10957692 DOI: 10.3389/fphar.2024.1378292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/26/2024] [Indexed: 03/26/2024] Open
Abstract
Aldo-Keto Reductase Family 1 Member C3 (AKR1C3), also known as type 5 17β-hydroxysteroid dehydrogenase (17β-HSD5) or prostaglandin F (PGF) synthase, functions as a pivotal enzyme in androgen biosynthesis. It catalyzes the conversion of weak androgens, estrone (a weak estrogen), and PGD2 into potent androgens (testosterone and 5α-dihydrotestosterone), 17β-estradiol (a potent estrogen), and 11β-PGF2α, respectively. Elevated levels of AKR1C3 activate androgen receptor (AR) signaling pathway, contributing to tumor recurrence and imparting resistance to cancer therapies. The overexpression of AKR1C3 serves as an oncogenic factor, promoting carcinoma cell proliferation, invasion, and metastasis, and is correlated with unfavorable prognosis and overall survival in carcinoma patients. Inhibiting AKR1C3 has demonstrated potent efficacy in suppressing tumor progression and overcoming treatment resistance. As a result, the development and design of AKR1C3 inhibitors have garnered increasing interest among researchers, with significant progress witnessed in recent years. Novel AKR1C3 inhibitors, including natural products and analogues of existing drugs designed based on their structures and frameworks, continue to be discovered and developed in laboratories worldwide. The AKR1C3 enzyme has emerged as a key player in carcinoma progression and therapeutic resistance, posing challenges in cancer treatment. This review aims to provide a comprehensive analysis of AKR1C3's role in carcinoma development, its implications in therapeutic resistance, and recent advancements in the development of AKR1C3 inhibitors for tumor therapies.
Collapse
Affiliation(s)
- Mengnan Li
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Limin Zhang
- Jingzhou Hospital of Traditional Chinese Medicine, Jingzhou, China
- The Third Clinical Medical College of Yangtze University, Jingzhou, China
| | - Jiahui Yu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Xiaoxiao Wang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Le Cheng
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Zhaowu Ma
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Xiaoguang Chen
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Lingzhi Wang
- Department of Haematology–Oncology, National University Cancer Institute, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Boon Cher Goh
- Department of Haematology–Oncology, National University Cancer Institute, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
11
|
Hilpert J, Groettrup-Wolfers E, Kosturski H, Bennett L, Barnes CLK, Gude K, Gashaw I, Reif S, Steger-Hartmann T, Scheerans C, Solms A, Rottmann A, Mao G, Chapron C. Hepatotoxicity of AKR1C3 Inhibitor BAY1128688: Findings from an Early Terminated Phase IIa Trial for the Treatment of Endometriosis. Drugs R D 2023; 23:221-237. [PMID: 37422772 PMCID: PMC10439066 DOI: 10.1007/s40268-023-00427-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2023] [Indexed: 07/11/2023] Open
Abstract
INTRODUCTION BAY1128688 is a selective inhibitor of aldo-keto reductase family 1 member C3 (AKR1C3), an enzyme implicated in the pathology of endometriosis and other disorders. In vivo animal studies suggested a potential therapeutic application of BAY1128688 in treating endometriosis. Early clinical studies in healthy volunteers supported the start of phase IIa. OBJECTIVE This manuscript reports the results of a clinical trial (AKRENDO1) assessing the effects of BAY1128688 in adult premenopausal women with endometriosis-related pain symptoms over a 12-week treatment period. METHODS Participants in this placebo-controlled, multicenter phase IIa clinical trial (NCT03373422) were randomized into one of five BAY1128688 treatment groups: 3 mg once daily (OD), 10 mg OD, 30 mg OD, 30 mg twice daily (BID), 60 mg BID; or a placebo group. The efficacy, safety, and tolerability of BAY1128688 were investigated. RESULTS Dose-/exposure-dependent hepatotoxicity was observed following BAY1128688 treatment, characterized by elevations in serum alanine transferase (ALT) occurring at around 12 weeks of treatment and prompting premature trial termination. The reduced number of valid trial completers precludes conclusions regarding treatment efficacy. The pharmacokinetics and pharmacodynamics of BAY1128688 among participants with endometriosis were comparable with those previously found in healthy volunteers and were not predictive of the subsequent ALT elevations observed. CONCLUSIONS The hepatotoxicity of BAY1128688 observed in AKRENDO1 was not predicted by animal studies nor by studies in healthy volunteers. However, in vitro interactions of BAY1128688 with bile salt transporters indicated a potential risk factor for hepatotoxicity at higher doses. This highlights the importance of in vitro mechanistic and transporter interaction studies in the assessment of hepatoxicity risk and suggests further mechanistic understanding is required. CLINICAL TRIAL REGISTRATION NCT03373422 (date registered: November 23, 2017).
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Charles Chapron
- Department of Gynecology, Obstetrics II, and Reproductive Medicine, Faculté de Santé, Faculté de Médecine Paris Centre, Université de Paris, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Universitaire Paris Centre (HUPC), Centre Hospitalier Universitaire (CHU) Cochin, Paris, France
| |
Collapse
|
12
|
Maddeboina K, Jonnalagadda SK, Morsy A, Duan L, Chhonker YS, Murry DJ, Penning TM, Trippier PC. Aldo-Keto Reductase 1C3 Inhibitor Prodrug Improves Pharmacokinetic Profile and Demonstrates In Vivo Efficacy in a Prostate Cancer Xenograft Model. J Med Chem 2023; 66:9894-9915. [PMID: 37428858 DOI: 10.1021/acs.jmedchem.3c00732] [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] [Indexed: 07/12/2023]
Abstract
Aldo-keto reductase 1C3 (AKR1C3) is overexpressed in castration-resistant prostate cancer where it acts to drive proliferation and aggressiveness by producing androgens. The reductive action of the enzyme leads to chemoresistance development against various clinical antineoplastics across a range of cancers. Herein, we report the continued optimization of selective AKR1C3 inhibitors and the identification of 5r, a potent AKR1C3 inhibitor (IC50 = 51 nM) with >1216-fold selectivity for AKR1C3 over closely related isoforms. Due to the cognizance of the poor pharmacokinetics associated with free carboxylic acids, a methyl ester prodrug strategy was pursued. The prodrug 4r was converted to free acid 5r in vitro in mouse plasma and in vivo. The in vivo pharmacokinetic evaluation revealed an increase in systemic exposure and increased the maximum 5r concentration compared to direct administration of the free acid. The prodrug 4r demonstrated a dose-dependent effect to reduce the tumor volume of 22Rv1 prostate cancer xenografts without observed toxicity.
Collapse
Affiliation(s)
- Krishnaiah Maddeboina
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Sravan K Jonnalagadda
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Ahmed Morsy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Ling Duan
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Yashpal S Chhonker
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Daryl J Murry
- Department of Pharmacy Practice and Science, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| | - Trevor M Penning
- Center of Excellence in Environmental Toxicology, Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
- UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, Nebraska 68106, United States
| |
Collapse
|
13
|
Gashaw I, Reif S, Wiesinger H, Kaiser A, Zollmann FS, Scheerans C, Grevel J, Piraino P, Seidel H, Peters M, Rottmann A, Rohde B, Arlt W, Hilpert J. Novel aldo-keto reductase 1C3 inhibitor affects androgen metabolism but not ovarian function in healthy women: a phase 1 study. Eur J Endocrinol 2023; 188:578-591. [PMID: 37306288 PMCID: PMC10376460 DOI: 10.1093/ejendo/lvad063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/26/2023] [Accepted: 04/11/2023] [Indexed: 06/13/2023]
Abstract
OBJECTIVE Aldo-keto reductase 1C3 (AKR1C3) has been postulated to be involved in androgen, progesterone, and estrogen metabolism. Aldo-keto reductase 1C3 inhibition has been proposed for treatment of endometriosis and polycystic ovary syndrome. Clinical biomarkers of target engagement, which can greatly facilitate drug development, have not yet been described for AKR1C3 inhibitors. Here, we analyzed pharmacodynamic data from a phase 1 study with a new selective AKR1C3 inhibitor, BAY1128688, to identify response biomarkers and assess effects on ovarian function. DESIGN In a multiple-ascending-dose placebo-controlled study, 33 postmenopausal women received BAY1128688 (3, 30, or 90 mg once daily or 60 mg twice daily) or placebo for 14 days. Eighteen premenopausal women received 60 mg BAY1128688 once or twice daily for 28 days. METHODS We measured 17 serum steroids by liquid chromatography-tandem mass spectrometry, alongside analysis of pharmacokinetics, menstrual cyclicity, and safety parameters. RESULTS In both study populations, we observed substantial, dose-dependent increases in circulating concentrations of the inactive androgen metabolite androsterone and minor increases in circulating etiocholanolone and dihydrotestosterone concentrations. In premenopausal women, androsterone concentrations increased 2.95-fold on average (95% confidence interval: 0.35-3.55) during once- or twice-daily treatment. Note, no concomitant changes in serum 17β-estradiol and progesterone were observed, and menstrual cyclicity and ovarian function were not altered by the treatment. CONCLUSIONS Serum androsterone was identified as a robust response biomarker for AKR1C3 inhibitor treatment in women. Aldo-keto reductase 1C3 inhibitor administration for 4 weeks did not affect ovarian function.ClinicalTrials.gov Identifier: NCT02434640; EudraCT Number: 2014-005298-36.
Collapse
Affiliation(s)
- Isabella Gashaw
- Research and Development, Pharmaceuticals, Bayer AG, 13353 Berlin, Germany
| | - Stefanie Reif
- Research and Development, Pharmaceuticals, Bayer AG, 13353 Berlin, Germany
| | - Herbert Wiesinger
- Research and Development, Pharmaceuticals, Bayer AG, 13353 Berlin, Germany
| | - Andreas Kaiser
- Research and Development, Pharmaceuticals, Bayer AG, 13353 Berlin, Germany
| | | | | | - Joachim Grevel
- Clinical Development, Bast GmbH, 69115 Heidelberg, Germany
| | - Paolo Piraino
- Research and Development, Pharmaceuticals, Bayer AG, 13353 Berlin, Germany
| | - Henrik Seidel
- Research and Development, Pharmaceuticals, Bayer AG, 13353 Berlin, Germany
| | - Michaele Peters
- Research and Development, Pharmaceuticals, Bayer AG, 13353 Berlin, Germany
| | - Antje Rottmann
- Research and Development, Pharmaceuticals, Bayer AG, 13353 Berlin, Germany
| | - Beate Rohde
- Research and Development, Pharmaceuticals, Bayer AG, 13353 Berlin, Germany
| | - Wiebke Arlt
- Medical Research Council London Institute of Medical Sciences, W12 0NN London, United Kingdom
- Department of Clinical Sciences, Faculty of Medicine, Imperial College London, W12 0NN London, United Kingdom
| | - Jan Hilpert
- Research and Development, Pharmaceuticals, Bayer AG, 13353 Berlin, Germany
| |
Collapse
|
14
|
Poirier D. Description of Chemical Synthesis, Nuclear Magnetic Resonance Characterization and Biological Activity of Estrane-Based Inhibitors/Activators of Steroidogenesis. Molecules 2023; 28:molecules28083499. [PMID: 37110733 PMCID: PMC10143840 DOI: 10.3390/molecules28083499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/08/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Steroid hormones play a crucial role in several aspects of human life, and steroidogenesis is the process by which hormones are produced from cholesterol using several enzymes that work in concert to obtain the appropriate levels of each hormone at the right time. Unfortunately, many diseases, such as cancer, endometriosis, and osteoporosis as examples, are caused by an increase in the production of certain hormones. For these diseases, the use of an inhibitor to block the activity of an enzyme and, in doing so, the production of a key hormone is a proven therapeutic strategy whose development continues. This account-type article focuses on seven inhibitors (compounds 1-7) and an activator (compound 8) of six enzymes involved in steroidogenesis, namely steroid sulfatase, aldo-keto reductase 1C3, types 1, 2, 3, and 12 of the 17β-hydroxysteroid dehydrogenases. For these steroid derivatives, three topics will be addressed: (1) Their chemical synthesis from the same starting material, estrone, (2) their structural characterization using nuclear magnetic resonance, and (3) their in vitro or in vivo biological activities. These bioactive molecules constitute potential therapeutic or mechanistic tools that could be used to better understand the role of certain hormones in steroidogenesis.
Collapse
Affiliation(s)
- Donald Poirier
- Laboratory of Medicinal Chemistry, Endocrinology and Nephrology Unit, CHU de Québec Research Center-Université Laval, Québec, QC G1V 4G2, Canada
- Department of Molecular Medicine, Faculty of Medicine, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
15
|
Szukiewicz D. Aberrant epigenetic regulation of estrogen and progesterone signaling at the level of endometrial/endometriotic tissue in the pathomechanism of endometriosis. VITAMINS AND HORMONES 2023; 122:193-235. [PMID: 36863794 DOI: 10.1016/bs.vh.2022.11.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Endometriosis is a term referring to a condition whereby the endometrial tissue is found outside the uterine cavity. This progressive and debilitating condition affects up to 15% of women of reproductive age. Due to the fact that endometriosis cells may express estrogen receptors (ERα, Erβ, GPER) and progesterone (P4) receptors (PR-A, PR-B), their growth, cyclic proliferation, and breakdown are similar to the processes occurring in the endometrium. The underlying etiology and pathogenesis of endometriosis are still not fully explained. The retrograde transport of viable menstrual endometrial cells with the retained ability to attach within the pelvic cavity, proliferate, differentiate and invade into the surrounding tissue explains the most widely accepted implantation theory. Endometrial stromal cells (EnSCs) with clonogenic potential constitute the most abundant population of cells within endometrium that resemble the properties of mesenchymal stem cells (MSCs). Accordingly, formation of the endometriotic foci in endometriosis may be due to a kind of EnSCs dysfunction. Increasing evidence indicates the underestimated role of epigenetic mechanisms in the pathogenesis of endometriosis. Hormone-mediated epigenetic modifications of the genome in EnSCs or even MSCs were attributed an important role in the etiopathogenesis of endometriosis. The roles of excess estrogen exposure and P4 resistance were also found to be crucial in the development of epigenetic homeostasis failure. Therefore, the aim of this review was to consolidate the current knowledge regarding the epigenetic background of EnSCs and MSCs and the changed properties due to estrogen/P4 imbalances in the context of the etiopathogenesis of endometriosis.
Collapse
Affiliation(s)
- Dariusz Szukiewicz
- Department of Biophysics, Physiology & Pathophysiology, Faculty of Health Sciences, Medical University of Warsaw, Warsaw, Poland.
| |
Collapse
|
16
|
He S, Liu Y, Chu X, Li Q, Lyu W, Liu Y, Xing S, Feng F, Liu W, Guo Q, Zhao L, Sun H. Discovery of Novel Aldo-Keto Reductase 1C3 Inhibitors as Chemotherapeutic Potentiators for Cancer Drug Resistance. ACS Med Chem Lett 2022; 13:1286-1294. [PMID: 35978698 PMCID: PMC9377021 DOI: 10.1021/acsmedchemlett.2c00175] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/06/2022] [Indexed: 11/29/2022] Open
Abstract
As a crucial target which is overexpressed in a variety of cancers, aldo-keto reductase 1C3 (AKR1C3) confers chemotherapeutic resistance to many clinical agents. However, a limited number of AKR1C3-selective inhibitors are applied clinically, which indicates the importance of identifying active compounds. Herein, we report the discovery, synthesis, and evaluation of novel and potent AKR1C3 inhibitors with structural diversity. Molecular dynamics simulations of these active compounds provide reasonable clarification of the interpreted biological data. Moreover, we demonstrate that AKR1C3 inhibitors have the potential to be superior therapeutic agents for re-sensitizing drug-resistant cell lines to chemotherapy, especially the pan-AKR1C inhibitor S07-2010. Our study identifies new structural classes of AKR1C3 inhibitors and enriches the structural diversity, which facilitates the future rational design of inhibitors and structural optimization. Moreover, these compounds may serve as promising therapeutic adjuvants toward new therapeutics for countering drug resistance.
Collapse
Affiliation(s)
- Siyu He
- School
of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
- State
Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis
and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, People’s Republic of China
| | - Yang Liu
- School
of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Xianglin Chu
- School
of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Qi Li
- Department
of Medical Pharmacy, School of Basic Medicine, Qingdao University, Qingdao 266071, People’s Republic
of China
| | - Weiping Lyu
- Department
of Pharmaceutical Analysis, Key Laboratory of Drug Quality Control
and Pharmacovigilance, China Pharmaceutical
University, Nanjing 211198, People’s Republic of China
| | - Yijun Liu
- School
of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Shuaishuai Xing
- School
of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| | - Feng Feng
- Department
of Natural Medicinal Chemistry, China Pharmaceutical
University, Nanjing 211198, People’s Republic of China
- Jiangsu
Drug Development Engineering Research Center for Central Degenerative
Disease, Jiangsu Food and Pharmaceuticals
Science College, Nanjing 223005, People’s Republic
of China
| | - Wenyuan Liu
- School
of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
- Department
of Pharmaceutical Analysis, Key Laboratory of Drug Quality Control
and Pharmacovigilance, China Pharmaceutical
University, Nanjing 211198, People’s Republic of China
| | - Qinglong Guo
- State
Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis
and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, People’s Republic of China
| | - Li Zhao
- State
Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis
and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, People’s Republic of China
| | - Haopeng Sun
- School
of Pharmacy, China Pharmaceutical University, Nanjing 211198, People’s Republic of China
| |
Collapse
|
17
|
Pan D, Yang W, Zeng Y, Qin H, Xu Y, Gui Y, Fan X, Tian G, Wu Y, Sun H, Ye Y, Yang S, Zhou J, Guo Q, Zhao L. AKR1C3 regulated by NRF2/MAFG complex promotes proliferation via stabilizing PARP1 in hepatocellular carcinoma. Oncogene 2022; 41:3846-3858. [PMID: 35773412 DOI: 10.1038/s41388-022-02379-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 05/18/2022] [Accepted: 06/07/2022] [Indexed: 11/09/2022]
Abstract
Aldo-keto reductase family 1 member C3 (AKR1C3) serves as a contributor to numerous kinds of tumors, and its expression is elevated in patients with hepatocellular carcinoma (HCC). However, the biological function of AKR1C3 in HCC remains unclear. Here we investigated the role of AKR1C3 in liver carcinogenesis using in vitro and in vivo models. We determined that AKR1C3 is frequently increased in HCC tissues with poor prognosis. Genetically manipulated cells with AKR1C3 construction were examined to highlight the pro-tumoral growth of both wild-type AKR1C3 and mutant in vitro and in vivo. We observed promising treatment effects of AKR1C3 shRNA by intratumoral injection in mice. Mechanically, we demonstrated that the transcription factor heterodimer NRF2/MAFG was able to bind directly to AKR1C3 promoter to activate its transcription. Further, AKR1C3 stabilized PARP1 by decreasing its ubiquitination, which resulted in HCC cell proliferation and low sensitivity of Cisplatin. Moreover, we discovered that the tumorigenic role of AKR1C3 was non-catalytic dependent and the NRF2/MAFG-AKR1C3-PARP1 axis might be one of the important proliferation pathways in HCC. In conclusion, blockage of AKR1C3 expression provides potential therapeutic benefits against HCC.
Collapse
Affiliation(s)
- Di Pan
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China.,The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang, 550025, Guizhou, China
| | - Wanwan Yang
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Yao Zeng
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Hongkun Qin
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Yuting Xu
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Yanping Gui
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Xiangshan Fan
- Department of Pathology, Affiliated Nanjing Drum Tower Hospital of Nanjing University School of Medicine, Nanjing, 210000, Jiangsu, China
| | - Geng Tian
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Yujia Wu
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Haopeng Sun
- Department of Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Yuting Ye
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Shihe Yang
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China
| | - Jieying Zhou
- Department of chemistry and biochemistry, Florida International University, Miami, FL, USA
| | - Qinglong Guo
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China. .,Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, 210009, Jiangsu, China.
| | - Li Zhao
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, Jiangsu, China.
| |
Collapse
|
18
|
Möller G, Temml V, Cala Peralta A, Gruet O, Richomme P, Séraphin D, Viault G, Kraus L, Huber-Cantonati P, Schopfhauser E, Pachmayr J, Tokarz J, Schuster D, Helesbeux JJ, Dyar KA. Analogues of Natural Chalcones as Efficient Inhibitors of AKR1C3. Metabolites 2022; 12:99. [PMID: 35208174 PMCID: PMC8876231 DOI: 10.3390/metabo12020099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/27/2022] Open
Abstract
Naturally occurring substances are valuable resources for drug development. In this respect, chalcones are known to be antiproliferative agents against prostate cancer cell lines through various mechanisms or targets. Based on the literature and preliminary results, we aimed to study and optimise the efficiency of a series of chalcones to inhibit androgen-converting AKR1C3, known to promote prostate cancer. A total of 12 chalcones with different substitution patterns were synthesised. Structure-activity relationships associated with these modifications on AKR1C3 inhibition were analysed by performing enzymatic assays and docking simulations. In addition, the selectivity and cytotoxicity of the compounds were assessed. In enzymatic assays, C-6' hydroxylated derivatives were more active than C-6' methoxylated derivatives. In contrast, C-4 methylation increased activity over C-4 hydroxylation. Docking results supported these findings with the most active compounds fitting nicely in the binding site and exhibiting strong interactions with key amino acid residues. The most effective inhibitors were not cytotoxic for HEK293T cells and selective for 17β-hydroxysteroid dehydrogenases not primarily involved in steroid hormone metabolism. Nevertheless, they inhibited several enzymes of the steroid metabolism pathways. Favourable substitutions that enhanced AKR1C3 inhibition of chalcones were identified. This study paves the way to further develop compounds from this series or related flavonoids with improved inhibitory activity against AKR1C3.
Collapse
Affiliation(s)
- Gabriele Möller
- Institute for Diabetes and Cancer, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (J.T.); (K.A.D.)
| | - Veronika Temml
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (V.T.); (E.S.); (D.S.)
| | - Antonio Cala Peralta
- University of Angers, SONAS, SFR QUASAV, F-49000 Angers, France; (A.C.P.); (O.G.); (P.R.); (D.S.); (G.V.); (J.-J.H.)
| | - Océane Gruet
- University of Angers, SONAS, SFR QUASAV, F-49000 Angers, France; (A.C.P.); (O.G.); (P.R.); (D.S.); (G.V.); (J.-J.H.)
| | - Pascal Richomme
- University of Angers, SONAS, SFR QUASAV, F-49000 Angers, France; (A.C.P.); (O.G.); (P.R.); (D.S.); (G.V.); (J.-J.H.)
| | - Denis Séraphin
- University of Angers, SONAS, SFR QUASAV, F-49000 Angers, France; (A.C.P.); (O.G.); (P.R.); (D.S.); (G.V.); (J.-J.H.)
| | - Guillaume Viault
- University of Angers, SONAS, SFR QUASAV, F-49000 Angers, France; (A.C.P.); (O.G.); (P.R.); (D.S.); (G.V.); (J.-J.H.)
| | - Luisa Kraus
- Institute of Pharmacy, Pharmaceutical Biology and Clinical Pharmacy, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (L.K.); (P.H.-C.); (J.P.)
| | - Petra Huber-Cantonati
- Institute of Pharmacy, Pharmaceutical Biology and Clinical Pharmacy, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (L.K.); (P.H.-C.); (J.P.)
| | - Elisabeth Schopfhauser
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (V.T.); (E.S.); (D.S.)
| | - Johanna Pachmayr
- Institute of Pharmacy, Pharmaceutical Biology and Clinical Pharmacy, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (L.K.); (P.H.-C.); (J.P.)
| | - Janina Tokarz
- Institute for Diabetes and Cancer, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (J.T.); (K.A.D.)
| | - Daniela Schuster
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Paracelsus Medical University Salzburg, 5020 Salzburg, Austria; (V.T.); (E.S.); (D.S.)
| | - Jean-Jacques Helesbeux
- University of Angers, SONAS, SFR QUASAV, F-49000 Angers, France; (A.C.P.); (O.G.); (P.R.); (D.S.); (G.V.); (J.-J.H.)
| | - Kenneth Allen Dyar
- Institute for Diabetes and Cancer, Helmholtz Center Munich, German Research Center for Environmental Health, 85764 Neuherberg, Germany; (J.T.); (K.A.D.)
| |
Collapse
|
19
|
Mercorio A, Giampaolino P, Romano A, Dällenbach P, Pluchino N. Is intracrinology of endometriosis relevant in clinical practice? A systematic review on estrogen metabolism. Front Endocrinol (Lausanne) 2022; 13:950866. [PMID: 36204107 PMCID: PMC9531311 DOI: 10.3389/fendo.2022.950866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
UNLABELLED Endometriosis is a chronic, multifactorial, estrogen-dependent disease. The abnormal endocrine microenvironment of endometriosis lesions is considered a main feature and multiple enzymatic pathways leading to local increased synthesis of estrogens have been identified. However, the relevance of intracrinology in clinical practice is still lacking. Medline, Embase, Scopus database were systematically searched for studies reporting on local estrogens metabolism of endometriotic lesions. The main enzymatic pathways involved in the intracrinology of endometriosis such as aromatase (CYP19A1), 17β-hydroxysteroid dehydrogenase (HSD17B) type 1, type 2 and type 5, steroid sulfatase (STS), estrogen sulfotransferase (SULT1E1) were assessed with a critical perspective on their role in disease endocrine phenotyping, drug resistance and as therapeutic targets. Overall, studies heterogeneity and missing clinical data affect the interpretation of the clinical role of these enzymes. Although the use of some drugs such as aromatase inhibitors has been proposed in clinical practice for two decades, their potential clinical value is still under investigation as well as their modality of administration. A closer look at new, more realistic drug targets is provided and discussed. Altered expression of these key enzymes in the lesions have far reaching implication in the development of new drugs aimed at decreasing local estrogenic activity with a minimal effect on gonadal function; however, given the complexity of the evaluation of the expression of the enzymes, multiple aspects still remains to be clarified. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42022311329, identifier CRD42022311329.
Collapse
Affiliation(s)
- Antonio Mercorio
- Department of Pediatrics Gynecology and Obstetrics, Division of Gynecology, Geneva University Hospitals, Geneva, Switzerland
| | - Pierluigi Giampaolino
- Department of Public Health, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Andrea Romano
- Obstetrics and Gynaecology Department, GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, Netherlands
| | - Patrick Dällenbach
- Department of Pediatrics Gynecology and Obstetrics, Division of Gynecology, Geneva University Hospitals, Geneva, Switzerland
| | - Nicola Pluchino
- Department of Pediatrics Gynecology and Obstetrics, Division of Gynecology, Geneva University Hospitals, Geneva, Switzerland
- *Correspondence: Nicola Pluchino,
| |
Collapse
|
20
|
Khalilullah H. Identification of Anti‐Cancer Agents Targeting Aldo‐Keto Reductase (AKR) 1C3 Protein by Pharmacophore Modeling, Virtual Screening and Molecular Docking. ChemistrySelect 2021. [DOI: 10.1002/slct.202103151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Habibullah Khalilullah
- Department of Pharmaceutical Chemistry & Pharmacognosy Unaizah College of Pharmacy, Qassim University Unaizah 51911 Kingdom of Saudi Arabia
| |
Collapse
|
21
|
Sinreih M, Jójárt R, Kele Z, Büdefeld T, Paragi G, Mernyák E, Rižner TL. Synthesis and evaluation of AKR1C inhibitory properties of A-ring halogenated oestrone derivatives. J Enzyme Inhib Med Chem 2021; 36:1500-1508. [PMID: 34227437 PMCID: PMC8266253 DOI: 10.1080/14756366.2021.1937142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Enzymes AKR1C regulate the action of oestrogens, androgens, and progesterone at the pre-receptor level and are also associated with chemo-resistance. The activities of these oestrone halides were investigated on recombinant AKR1C enzymes. The oestrone halides with halogen atoms at both C-2 and C-4 positions (13β-, 13α-methyl-17-keto halogen derivatives) were the most potent inhibitors of AKR1C1. The lowest IC50 values were for the 13α-epimers 2_2I,4Br and 2_2I,4Cl (IC50, 0.7 μM, 0.8 μM, respectively), both of which selectively inhibited the AKR1C1 isoform. The 13α-methyl-17-keto halogen derivatives 2_2Br and 2_4Cl were the most potent inhibitors of AKR1C2 (IC50, 1.5 μM, 1.8 μM, respectively), with high selectivity for the AKR1C2 isoform. Compound 1_2Cl,4Cl showed the best AKR1C3 inhibition, and it also inhibited AKR1C1 (Ki: AKR1C1, 0.69 μM; AKR1C3, 1.43 μM). These data show that halogenated derivatives of oestrone represent a new class of potent and selective AKR1C inhibitors as lead compounds for further optimisations.
Collapse
Affiliation(s)
- Maša Sinreih
- Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, University of Ljubljana, Ljubljana, Slovenia
| | - Rebeka Jójárt
- Department of Organic Chemistry, University of Szeged, Szeged, Hungary
| | - Zoltán Kele
- Department of Medicinal Chemistry, University of Szeged, Szeged, Hungary
| | - Tomaž Büdefeld
- Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, University of Ljubljana, Ljubljana, Slovenia
| | - Gábor Paragi
- MTA-SZTE Biomimetic Systems Research Group, University of Szeged, Szeged, Hungary.,Institute of Physics, University of Pécs, Pécs, Hungary
| | - Erzsébet Mernyák
- Department of Organic Chemistry, University of Szeged, Szeged, Hungary
| | - Tea Lanišnik Rižner
- Faculty of Medicine, Institute of Biochemistry and Molecular Genetics, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
22
|
Saunders PT, Horne AW. Endometriosis: Etiology, pathobiology, and therapeutic prospects. Cell 2021; 184:2807-2824. [DOI: 10.1016/j.cell.2021.04.041] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/31/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023]
|
23
|
The subgroup of 2'-hydroxy-flavonoids: Molecular diversity, mechanism of action, and anticancer properties. Bioorg Med Chem 2021; 32:116001. [PMID: 33444847 DOI: 10.1016/j.bmc.2021.116001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/31/2020] [Indexed: 12/19/2022]
Abstract
Flavonoids are abundant in nature, structurally very diversified and largely investigated. However, the subgroup of 2'-hydroxyflavonoids is much less known and not frequently studied. The present review identifies the major naturally-occurring and synthetic 2'-hydroxyflavonoid derivatives and discusses their structural characteristics and biological properties, with a focus on anticancer activities. The pharmacological properties of 2'-hydroxyflavone (2'-HF) and 2'-hydroxyflavanone (2'-HFa) are detailed. Upon binding to the Ral-interacting protein Rlip implicated in the transport of glutathione conjugates, 2'-HFa inhibits tumor cell proliferation and restrict tumor growth, in particular in breast cancer models. Among the synthetic derivatives, the characteristics of the anticancer product 2D08 (2',3',4'-trihydroxy flavone) are detailed to shed light on the molecular mechanism of action of this compound, as a regulator of protein SUMOylation. Inhibition of protein SUMOylation by 2D08 blocks cancer cell migration and invasion, and the compound greatly enhances the anticancer effects of conventional cytotoxic drugs like etoposide. The structural role of the 2'-hydroxyl group on the phenyl C-ring of the flavonoid is discussed, notably the capacity to engage intramolecular H-bonding interactions with the O1 atom on the B-ring of the chromone unit (or the oxygen of a 3-OH group when it is presents). The 2'-hydroxyl group of flavonoid appears as a regulator of the conformational freedom between the bicyclic A-B unit and the appended phenyl C-ring, favoring the planarity of the molecule. It is an essential group accounting for the biological properties of 2'-HF, 2'-HFa and structurally related compounds. This review shed light on 2'-hydroxyflavonoids to encourage their use and chemical development.
Collapse
|
24
|
Penning TM, Asangani IA, Sprenger C, Plymate S. Intracrine androgen biosynthesis and drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:912-929. [PMID: 35582223 PMCID: PMC8992556 DOI: 10.20517/cdr.2020.60] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/30/2020] [Accepted: 10/10/2020] [Indexed: 06/15/2023]
Abstract
Castration-resistant prostate cancer is the lethal form of prostate cancer and most commonly remains dependent on androgen receptor (AR) signaling. Current therapies use AR signaling inhibitors (ARSI) exemplified by abiraterone acetate, a P450c17 inhibitor, and enzalutamide, a potent AR antagonist. However, drug resistance to these agents occurs within 12-18 months and they only prolong overall survival by 3-4 months. Multiple mechanisms can contribute to ARSI drug resistance. These mechanisms can include but are not limited to germline mutations in the AR, post-transcriptional alterations in AR structure, and adaptive expression of genes involved in the intracrine biosynthesis and metabolism of androgens within the tumor. This review focuses on intracrine androgen biosynthesis, how this can contribute to ARSI drug resistance, and therapeutic strategies that can be used to surmount these resistance mechanisms.
Collapse
Affiliation(s)
- Trevor M. Penning
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Irfan A. Asangani
- Department Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Cynthia Sprenger
- Division of Gerontology & Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Stephen Plymate
- Division of Gerontology & Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA 98109, USA
- Geriatric Research Education and Clinical Center (GRECC), VA Puget Sound Health Care System, Seattle, WA 98108, USA
| |
Collapse
|
25
|
Endo S, Oguri H, Segawa J, Kawai M, Hu D, Xia S, Okada T, Irie K, Fujii S, Gouda H, Iguchi K, Matsukawa T, Fujimoto N, Nakayama T, Toyooka N, Matsunaga T, Ikari A. Development of Novel AKR1C3 Inhibitors as New Potential Treatment for Castration-Resistant Prostate Cancer. J Med Chem 2020; 63:10396-10411. [PMID: 32847363 DOI: 10.1021/acs.jmedchem.0c00939] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Aldo-keto reductase (AKR) 1C3 catalyzes the synthesis of active androgens that promote the progression of prostate cancer. AKR1C3 also contributes to androgen-independent cell proliferation and survival through the metabolism of prostaglandins and reactive aldehydes. Because of its elevation in castration-resistant prostate cancer (CRPC) tissues, AKR1C3 is a promising therapeutic target for CRPC. In this study, we found a novel potent AKR1C3 inhibitor, N-(4-fluorophenyl)-8-hydroxy-2-imino-2H-chromene-3-carboxamide (2d), and synthesized its derivatives with IC50 values of 25-56 nM and >220-fold selectivity over other AKRs (1C1, 1C2, and 1C4). The structural factors for the inhibitory potency were elucidated by crystallographic study of AKR1C3 complexes with 2j and 2l. The inhibitors suppressed proliferation of prostate cancer 22Rv1 and PC3 cells through both androgen-dependent and androgen-independent mechanisms. Additionally, 2j and 2l prevented prostate tumor growth in a xenograft mouse model. Furthermore, the inhibitors significantly augmented apoptotic cell death induced by anti-CRPC drugs (abiraterone or enzalutamide).
Collapse
Affiliation(s)
- Satoshi Endo
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Hiroaki Oguri
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Jin Segawa
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Mina Kawai
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Dawei Hu
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-8555, Japan
| | - Shuang Xia
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-8555, Japan
| | - Takuya Okada
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-8555, Japan
| | - Katsumasa Irie
- Cellular and Structural Physiology Institute, Nagoya University, Furo-cho, Chikusa, Nagoya 464-8601, Japan.,Graduate School of Pharmaceutical Sciences, Nagoya University, Furo-cho, Chikusa, Nagoya 464-8601, Japan
| | - Shinya Fujii
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, Tokyo 101-0062, Japan
| | - Hiroaki Gouda
- School of Pharmacy, Showa University, Tokyo 142-8555, Japan
| | - Kazuhiro Iguchi
- Laboratory of Community Pharmacy, Department of Pharmacy, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| | - Takuo Matsukawa
- Department of Urology, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Naohiro Fujimoto
- Department of Urology, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Toshiyuki Nakayama
- Department of Pathology, University of Occupational and Environmental Health, Kitakyushu 807-8555, Japan
| | - Naoki Toyooka
- Graduate School of Innovative Life Science, University of Toyama, Toyama 930-8555, Japan
| | - Toshiyuki Matsunaga
- Education Center of Green Pharmaceutical Sciences, Gifu Pharmaceutical University, Gifu 502-8585, Japan
| | - Akira Ikari
- Laboratory of Biochemistry, Gifu Pharmaceutical University, Gifu 501-1196, Japan
| |
Collapse
|
26
|
Massarotti C, Mirabelli Badenier I, Paudice M, Scaglione G, Remorgida V, Vellone VG. Steroids receptors immunohistochemical expression in different sites of endometriosis. J Gynecol Obstet Hum Reprod 2020; 50:101861. [PMID: 32652301 DOI: 10.1016/j.jogoh.2020.101861] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/21/2020] [Accepted: 07/01/2020] [Indexed: 01/23/2023]
Abstract
BACKGROUND A better characterization of steroid intracrine pathways in endometriosis lesions may lead to a better understanding of the pathogenesis of the disease and insights on the mechanism of resistance to medical therapy. The study aims to evaluate the expression of steroid receptors in endometriosis lesions, including for the first-time androgen receptors, both in glandular and stromal tissue, and to describe the differences, in any, in receptor expression in the different subtypes. BASIC PROCEDURES This is a retrospective analysis of 76 specimens from 51 women, that underwent laparoscopic surgery for endometriosis at a tertiary hospital between 2015 and 2019. Immunohistochemical detections of estrogen, progesterone and androgen receptors positive cells was performed and the results described in terms of both density and intensity. The density and intensity scores were combined to obtain a final Histological Score (HS). Non-parametric Kruskal-Wallis test or Mann-Whitney U-test were used to compare continuous data, chi square test for categorical data. MAIN FINDINGS Estrogen receptor α expression was moderate/high in almost all specimens, regardless of the site. Samples from endometriomas presented lower progesterone receptor expression in the epithelium, compared to pelvic sites. Androgen receptor density was higher in stromal cells compared to epithelial cells and in pelvic sites compared to ovarian ones. CONCLUSIONS The roles of nuclear receptors in endometriosis, including differences in their expression, could help in defining the pathogenesis of the disease and in explaining different responsivity to therapies. The intracrine regulation of steroids plays a relevant role in the metabolic and inflammatory pathogenetic paths of endometriosis: if better understood, its manipulation could be a relevant therapeutic target for treatment.
Collapse
Affiliation(s)
- Claudia Massarotti
- Physiopathology of Human Reproduction Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | | | - Michele Paudice
- Department of Integrated Surgical and Diagnostic Sciences (DISC), University of Genova, Genova, Italy
| | - Giulia Scaglione
- Department of Integrated Surgical and Diagnostic Sciences (DISC), University of Genova, Genova, Italy
| | - Valentino Remorgida
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal-Child Health (DiNOGMI), University of Genova, Genova, Italy; Academic Unit of Obsterics and Gynecology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Valerio Gaetano Vellone
- Department of Integrated Surgical and Diagnostic Sciences (DISC), University of Genova, Genova, Italy; Academic Unit of Pathology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| |
Collapse
|
27
|
Liu Y, He S, Chen Y, Liu Y, Feng F, Liu W, Guo Q, Zhao L, Sun H. Overview of AKR1C3: Inhibitor Achievements and Disease Insights. J Med Chem 2020; 63:11305-11329. [PMID: 32463235 DOI: 10.1021/acs.jmedchem.9b02138] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Human aldo-keto reductase family 1 member C3 (AKR1C3) is known as a hormone activity regulator and prostaglandin F (PGF) synthase that regulates the occupancy of hormone receptors and cell proliferation. Because of the overexpression in metabolic diseases and various hormone-dependent and -independent carcinomas, as well as the emergence of clinical drug resistance, an increasing number of studies have investigated AKR1C3 inhibitors. Here, we briefly review the physiological and pathological function of AKR1C3 and then summarize the recent development of selective AKR1C3 inhibitors. We propose our viewpoints on the current problems associated with AKR1C3 inhibitors with the aim of providing a reference for future drug discovery and potential therapeutic perspectives on novel, potent, selective AKR1C3 inhibitors.
Collapse
Affiliation(s)
- Yang Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Siyu He
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Ying Chen
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yijun Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Feng Feng
- Jiangsu Food and Pharmaceuticals Science College, Institute of Food and Pharmaceuticals Research, Huaian 223005, People's Republic of China.,Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Wenyuan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Li Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| |
Collapse
|
28
|
Guérit D, Marie P, Morel A, Maurin J, Verollet C, Raynaud-Messina B, Urbach S, Blangy A. Primary myeloid cell proteomics and transcriptomics: importance of β-tubulin isotypes for osteoclast function. J Cell Sci 2020; 133:jcs239772. [PMID: 32265273 DOI: 10.1242/jcs.239772] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 03/19/2020] [Indexed: 08/31/2023] Open
Abstract
Among hematopoietic cells, osteoclasts (OCs) and immature dendritic cells (DCs) are closely related myeloid cells with distinct functions: OCs participate skeleton maintenance while DCs sample the environment for foreign antigens. Such specificities rely on profound modifications of gene and protein expression during OC and DC differentiation. We provide global proteomic and transcriptomic analyses of primary mouse OCs and DCs, based on original stable isotope labeling with amino acids in cell culture (SILAC) and RNAseq data. We established specific signatures for OCs and DCs, including genes and proteins of unknown functions. In particular, we showed that OCs and DCs have the same α- and β-tubulin isotype repertoire but that OCs express much more of the β tubulin isotype Tubb6 (also known as TBB6). In both mouse and human OCs, we demonstrate that elevated expression of Tubb6 in OCs is necessary for correct podosomes organization and thus for the structure of the sealing zone, which sustains the bone resorption apparatus. Hence, lowering Tubb6 expression hinders OC resorption activity. Overall, we highlight here potential new regulators of OC and DC biology, and illustrate the functional importance of the tubulin isotype repertoire in the biology of differentiated cells.
Collapse
Affiliation(s)
- David Guérit
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| | - Pauline Marie
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| | - Anne Morel
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| | - Justine Maurin
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| | - Christel Verollet
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, 31077 Toulouse Cedex 04, France
- International associated laboratory (LIA) CNRS 'IM-TB/HIV' (1167), 31077 Toulouse Cedex 04, France
- International associated laboratory (LIA) CNRS 'IM-TB/HIV' (1167), Buenos Aires C1425AUM, Argentina
| | - Brigitte Raynaud-Messina
- Institut de Pharmacologie et Biologie Structurale, IPBS, Université de Toulouse, CNRS, UPS, 31077 Toulouse Cedex 04, France
- International associated laboratory (LIA) CNRS 'IM-TB/HIV' (1167), 31077 Toulouse Cedex 04, France
- International associated laboratory (LIA) CNRS 'IM-TB/HIV' (1167), Buenos Aires C1425AUM, Argentina
| | - Serge Urbach
- Functional Proteomics Facility, Institute of Functional Genomics, Montpellier Univ., CNRS, 141 rue de la Cardonille, 34000 Montpellier, France
| | - Anne Blangy
- Centre de Recherche de Biologie Cellulaire de Montpellier (CRBM), Montpellier Univ., CNRS, 34000 Montpellier, France
| |
Collapse
|