1
|
Acharyya RK, Rej RK, Hu B, Chen Z, Wu D, Lu J, Metwally H, McEachern D, Wang Y, Jiang W, Bai L, Tošović J, Gersch CL, Xu G, Zhang W, Wu W, Priestley ES, Sui Z, Sarkari F, Wen B, Sun D, Rae JM, Wang S. Discovery of ERD-1233 as a Potent and Orally Efficacious Estrogen Receptor PROTAC Degrader for the Treatment of ER+ Human Breast Cancer. J Med Chem 2024. [PMID: 39485242 DOI: 10.1021/acs.jmedchem.4c01521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Despite the development of highly effective therapies for the treatment of estrogen receptor α (ERα)-positive human breast cancer, clinical resistance to current therapies requires the development of novel therapeutic strategies. Herein, we report the discovery of ERD-1233 as a potent and orally efficacious ERα degrader designed using the PROTAC technology. ERD-1233 was developed based on Lasofoxifene as the ER binding moiety and a novel cereblon ligand through extensive optimization of the linker. ERD-1233 potently and effectively reduces the ERα protein in vitro and achieves excellent oral bioavailability in mice and rats. Oral administration of ERD-1233 effectively reduces ER protein in ER+ tumors and achieves tumor regression in the ER wild-type MCF-7 xenograft tumor model and strong tumor growth inhibition in the ESR1Y537S mutated model in mice. Our data demonstrate that ERD-1233 is a promising ER PROTAC degrader for extensive evaluation as a new therapy for the treatment of ER+ human breast cancer.
Collapse
Affiliation(s)
- Ranjan Kumar Acharyya
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Rohan Kalyan Rej
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Biao Hu
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Zhixiang Chen
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Dimin Wu
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jianfeng Lu
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hoda Metwally
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Donna McEachern
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yu Wang
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Wei Jiang
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Longchuan Bai
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jelena Tošović
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Christina L Gersch
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Guozhang Xu
- SK Life Science Laboratories, 2500 Renaissance Blvd, King of Prussia, Pennsylvania 19406, United States
| | - Weihong Zhang
- SK Life Science Laboratories, 2500 Renaissance Blvd, King of Prussia, Pennsylvania 19406, United States
| | - WenXue Wu
- SK Life Science Laboratories, 2500 Renaissance Blvd, King of Prussia, Pennsylvania 19406, United States
| | - E Scott Priestley
- SK Life Science Laboratories, 2500 Renaissance Blvd, King of Prussia, Pennsylvania 19406, United States
| | - Zhihua Sui
- SK Life Science Laboratories, 2500 Renaissance Blvd, King of Prussia, Pennsylvania 19406, United States
| | - Farzad Sarkari
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - James M Rae
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Shaomeng Wang
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
2
|
Saini H, Gupta PK, Mahapatra AK, Rajagopala S, Tripathi R, Nesari T. Deciphering the multi-scale mechanism of herbal phytoconstituents in targeting breast cancer: a computational pharmacological perspective. Sci Rep 2024; 14:23795. [PMID: 39394443 PMCID: PMC11479599 DOI: 10.1038/s41598-024-75059-z] [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: 06/29/2024] [Accepted: 10/01/2024] [Indexed: 10/13/2024] Open
Abstract
Breast Cancer (BC) is the most common cause of cancer-associated deaths in females worldwide. Despite advancements in BC treatment driven by extensive characterization of its molecular hallmarks, challenges such as drug resistance, tumor relapse, and metastasis persist. Therefore, there is an urgent need for alternative treatment approaches with multi-modal efficacy to overcome these hurdles. In this context, natural bioactives are increasingly recognized for their pivotal role as anti-cancer compounds. This study focuses on predicting molecular targets for key herbal phytoconstituents-gallic acid, piperine, quercetin, resveratrol, and beta-sitosterol-present in the polyherbal formulation, Krishnadi Churna. Using an in-silico network pharmacology model, key genes were identified and docked against these marker compounds and controls. Mammary carcinoma emerged as the most significant phenotype of the putative targets. Analysis of an online database revealed that out of 135 predicted targets, 134 were mutated in breast cancer patients. Notably, ESR1, CYP19A1, and EGFR were identified as key genes which are known to regulate the BC progression. Docking studies demonstrated that the herbal phytoconstituents had similar or better docking scores than positive controls for these key genes, with convincing protein-ligand interactions confirmed by molecular dynamics simulations, MM/GBSA and free energy landscape (FEL) analysis. Overall, this study highlights the predictive potential of herbal phytoconstituents in targeting BC genes, suggesting their promise as a basis for developing new therapeutic formulations for BC.
Collapse
Affiliation(s)
- Heena Saini
- Integrated Translational Molecular Biology Unit (ITMBU), Department of Rog Nidan Evam Vikriti Vigyan (Pathology), All India Institute of Ayurveda, New Delhi, 110076, India.
| | - Prashant Kumar Gupta
- Ayurinformatics Laboratory, Department of Kaumarabhritya (Pediatrics), All India Institute of Ayurveda, New Delhi, 110076, India
| | - Arun Kumar Mahapatra
- Ayurinformatics Laboratory, Department of Kaumarabhritya (Pediatrics), All India Institute of Ayurveda, New Delhi, 110076, India
| | - Shrikrishna Rajagopala
- Ayurinformatics Laboratory, Department of Kaumarabhritya (Pediatrics), All India Institute of Ayurveda, New Delhi, 110076, India
| | - Richa Tripathi
- Integrated Translational Molecular Biology Unit (ITMBU), Department of Rog Nidan Evam Vikriti Vigyan (Pathology), All India Institute of Ayurveda, New Delhi, 110076, India
| | - Tanuja Nesari
- Department of DravyaGuna (Materia Medica & Pharmacology), All India Institute of Ayurveda, New Delhi, 110076, India.
| |
Collapse
|
3
|
Chen Z, Hu B, Rej RK, Wu D, Acharyya RK, Wang M, Xu T, Lu J, Metwally H, Wang Y, McEachern D, Bai L, Gersch CL, Wang M, Zhang W, Li Q, Wen B, Sun D, Rae JM, Wang S. Discovery of ERD-3111 as a Potent and Orally Efficacious Estrogen Receptor PROTAC Degrader with Strong Antitumor Activity. J Med Chem 2023; 66:12559-12585. [PMID: 37647546 DOI: 10.1021/acs.jmedchem.3c01186] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Estrogen receptor α (ERα) is a prime target for the treatment of ER-positive (ER+) breast cancer. Despite the development of several effective therapies targeting ERα signaling, clinical resistance remains a major challenge. In this study, we report the discovery of a new class of potent and orally bioavailable ERα degraders using the PROTAC technology, with ERD-3111 being the most promising compound. ERD-3111 exhibits potent in vitro degradation activity against ERα and demonstrates high oral bioavailability in mice, rats, and dogs. Oral administration of ERD-3111 effectively reduces the levels of wild-type and mutated ERα proteins in tumor tissues. ERD-3111 achieves tumor regression or complete tumor growth inhibition in the parental MCF-7 xenograft model with wild-type ER and two clinically relevant ESR1 mutated models in mice. ERD-3111 is a promising ERα degrader for further extensive evaluations for the treatment of ER+ breast cancer.
Collapse
Affiliation(s)
- Zhixiang Chen
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Biao Hu
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Rohan Kalyan Rej
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Dimin Wu
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ranjan Kumar Acharyya
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Mingliang Wang
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Tianfeng Xu
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jianfeng Lu
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hoda Metwally
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yu Wang
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Donna McEachern
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Longchuan Bai
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Christina L Gersch
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Meilin Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Wenjing Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Qiuxia Li
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Bo Wen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Duxin Sun
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - James M Rae
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Shaomeng Wang
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Pharmacology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan 48109, United States
- The Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan 48109, United States
| |
Collapse
|
4
|
Xin L, Min J, Hu H, Li Y, Du C, Xie B, Cheng Y, Deng X, Deng X, Shen K, Huang J, Chen CC, Guo RT, Dong C, Zhou HB. Structure-guided identification of novel dual-targeting estrogen receptor α degraders with aromatase inhibitory activity for the treatment of endocrine-resistant breast cancer. Eur J Med Chem 2023; 253:115328. [PMID: 37037140 DOI: 10.1016/j.ejmech.2023.115328] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/28/2023] [Accepted: 03/28/2023] [Indexed: 04/12/2023]
Abstract
Drug resistance is a major challenge in conventional endocrine therapy for estrogen receptor (ER) positive breast cancer (BC). BC is a multifactorial disease, in which simultaneous aromatase (ARO) inhibition and ERα degradation may effectively inhibit the signal transduction of both proteins, thus potentially overcoming drug resistance caused by overexpression or mutation of target proteins. In this study, guided by the X-ray structure of a hit compound 30a in complex with ER-Y537S, a structure-based optimization was performed to get a series of multiacting inhibitors targeting both ERα and ARO, and finally a novel class of potent selective estrogen receptor degraders (SERDs) based on a three-dimensional oxabicycloheptene sulfonamide (OBHSA) scaffold equipped with aromatase inhibitor (AI) activity were identified. Of these dual-targeting SERD-AI hybrids, compound 31q incorporating a 1H-1,2,4-triazole moiety showed excellent ERα degradation activity, ARO inhibitory activity and remarkable antiproliferative activity against BC resistant cells. Furthermore, 31q manifested efficient tumor suppression in MCF-7 tumor xenograft models. Taken together, our study reported for the first time the highly efficient dual-targeting SERD-AI hybrid compounds, which may lay the foundation of translational research for improved treatment of endocrine-resistant BC.
Collapse
Affiliation(s)
- Lilan Xin
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Jian Min
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Hebing Hu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Yuanyuan Li
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Chuanqian Du
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Baohua Xie
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yan Cheng
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xiaofei Deng
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xiangping Deng
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Kang Shen
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Jian Huang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Chun-Chi Chen
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China
| | - Rey-Ting Guo
- State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei Hongshan Laboratory, Hubei Collaborative Innovation Center for Green Transformation of Bio-Resources, Hubei Key Laboratory of Industrial Biotechnology, School of Life Sciences, Hubei University, Wuhan, 430062, China.
| | - Chune Dong
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China.
| | - Hai-Bing Zhou
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; Frontier Science Center for Immunology and Metabolism, State Key Laboratory of Virology, Provincial Key Laboratory of Developmentally Originated Disease, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
5
|
Bhatia N, Hazra S, Thareja S. Selective Estrogen receptor degraders (SERDs) for the treatment of breast cancer: An overview. Eur J Med Chem 2023; 256:115422. [PMID: 37163948 DOI: 10.1016/j.ejmech.2023.115422] [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: 03/14/2023] [Revised: 04/17/2023] [Accepted: 04/26/2023] [Indexed: 05/12/2023]
Abstract
Discovery of SERDs has changed the direction of anticancer research, as more than 70% of breast cancer cases are estrogen receptor positive (ER+). Therapies such as selective estrogen receptor modulators (SERM) and aromatase inhibitors (AI's) have been effective, but due to endocrine resistance, SERDs are now considered essential therapeutics for the treatment of ER+ breast cancer. The present review deliberates the pathophysiology of SERDs from the literature covering various molecules in clinical trials. Estrogen receptors active sites distinguishing characteristics and interactions with currently available FDA-approved drugs have also been discussed. Designing strategy of previously reported SERDs, their SAR analysis, in silico, and the biological efficacy have also been summarized along with appropriate examples.
Collapse
Affiliation(s)
- Neha Bhatia
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Shreejita Hazra
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India.
| |
Collapse
|
6
|
Ehtezazi T, Rahman K, Davies R, Leach AG. The Pathological Effects of Circulating Hydrophobic Bile Acids in Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:173-211. [PMID: 36994114 PMCID: PMC10041467 DOI: 10.3233/adr-220071] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/19/2023] Open
Abstract
Recent clinical studies have revealed that the serum levels of toxic hydrophobic bile acids (deoxy cholic acid, lithocholic acid [LCA], and glycoursodeoxycholic acid) are significantly higher in patients with Alzheimer's disease (AD) and amnestic mild cognitive impairment (aMCI) when compared to control subjects. The elevated serum bile acids may be the result of hepatic peroxisomal dysfunction. Circulating hydrophobic bile acids are able to disrupt the blood-brain barrier and promote the formation of amyloid-β plaques through enhancing the oxidation of docosahexaenoic acid. Hydrophobic bile acid may find their ways into the neurons via the apical sodium-dependent bile acid transporter. It has been shown that hydrophobic bile acids impose their pathological effects by activating farnesoid X receptor and suppressing bile acid synthesis in the brain, blocking NMDA receptors, lowering brain oxysterol levels, and interfering with 17β-estradiol actions such as LCA by binding to E2 receptors (molecular modelling data exclusive to this paper). Hydrophobic bile acids may interfere with the sonic hedgehog signaling through alteration of cell membrane rafts and reducing brain 24(S)-hydroxycholesterol. This article will 1) analyze the pathological roles of circulating hydrophobic bile acids in the brain, 2) propose therapeutic approaches, and 3) conclude that consideration be given to reducing/monitoring toxic bile acid levels in patients with AD or aMCI, prior/in combination with other treatments.
Collapse
Affiliation(s)
- Touraj Ehtezazi
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Khalid Rahman
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Rhys Davies
- The Walton Centre, NHS Foundation Trust, Liverpool, UK
| | - Andrew G Leach
- School of Pharmacy, University of Manchester, Manchester, UK
| |
Collapse
|
7
|
Elavarasan S, Preety J, Abinaya R, Saravanan T, Balasunramaniam KK, Venkatramaiah N, Baskar B. Visible Light Driven Metal-Free Photoredox Catalyzed α-benzylation and α-oxygenation of N-substituted tetrahydroisoquinolines: Applications to Synthesis of Natural Products. Chem Asian J 2022; 17:e202200878. [PMID: 36073541 DOI: 10.1002/asia.202200878] [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: 08/25/2022] [Revised: 09/06/2022] [Indexed: 11/07/2022]
Abstract
Herein, visible light mediated organophoto redox catalysed simple and convenient method for the a-benzylation and a-oxygenation of tertiary amines is demonstrated. Synthesis of novel thiophenol based donor acceptor organophotoredox catalysts 4a - 4d were investigated along with commercial catalyst 4-CzIPN ( 4e ). A diverse biologically active a-benzylated tetrahydroisoquinolines and natural products such as (±)-Norlaudanosine, (±)-laudanosine and (±)-xylopinine have been synthesized under the optimized conditions in yields ranging from from 62-91%. Exploitation of synthesized a-benzylated compound using present phtoredox catalyzed conditions gave rise to dehydyrogenative benzylic oxidation product under oxygen atmosphere which is known to display biologically and structurally important properties. Also, various N-protected tertiary amines were found to be suitable for the a-oxygenation reactions using catalyst 4e and resulted in good yields (61-85%).
Collapse
Affiliation(s)
- S Elavarasan
- SRM Institute of Science and Technology, Chemistry, INDIA
| | - J Preety
- SRM Institute of Science and Technology, Chemistry, INDIA
| | - R Abinaya
- SRM Institute of Science and Technology, Chemistry, INDIA
| | - T Saravanan
- University of Hyderabad, School of Chemistry, INDIA
| | | | | | - Baburaj Baskar
- SRM University, Chemistry, Kattankulathur, 603203, India, 603203, Chennai, INDIA
| |
Collapse
|
8
|
Zhang T, Wang Y, Wang B, Jin W, Xia Y, Liu C, Zhang Y. Visible‐Light‐Induced Oxidation of Diazenyl‐Protected Tetrahydroisoquinolines and Isoindolines for the Synthesis of Functionalized Lactams. Adv Synth Catal 2022. [DOI: 10.1002/adsc.202101506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Tao Zhang
- Urumqi Key Laboratory of Green Catalysis and Synthesis Technology Key Laboratory of Oil and Gas Fine Chemicals, Ministry of Education & Xinjiang Uygur Autonomous Region State Key Laboratory of Chemistry and Utilization of Carbon Based Energy Resources College of Chemistry Xinjiang University Urumqi 830046 P. R. China
| | - Yanhong Wang
- Urumqi Key Laboratory of Green Catalysis and Synthesis Technology Key Laboratory of Oil and Gas Fine Chemicals, Ministry of Education & Xinjiang Uygur Autonomous Region State Key Laboratory of Chemistry and Utilization of Carbon Based Energy Resources College of Chemistry Xinjiang University Urumqi 830046 P. R. China
| | - Bin Wang
- Urumqi Key Laboratory of Green Catalysis and Synthesis Technology Key Laboratory of Oil and Gas Fine Chemicals, Ministry of Education & Xinjiang Uygur Autonomous Region State Key Laboratory of Chemistry and Utilization of Carbon Based Energy Resources College of Chemistry Xinjiang University Urumqi 830046 P. R. China
| | - Weiwei Jin
- Urumqi Key Laboratory of Green Catalysis and Synthesis Technology Key Laboratory of Oil and Gas Fine Chemicals, Ministry of Education & Xinjiang Uygur Autonomous Region State Key Laboratory of Chemistry and Utilization of Carbon Based Energy Resources College of Chemistry Xinjiang University Urumqi 830046 P. R. China
| | - Yu Xia
- Urumqi Key Laboratory of Green Catalysis and Synthesis Technology Key Laboratory of Oil and Gas Fine Chemicals, Ministry of Education & Xinjiang Uygur Autonomous Region State Key Laboratory of Chemistry and Utilization of Carbon Based Energy Resources College of Chemistry Xinjiang University Urumqi 830046 P. R. China
| | - Chenjiang Liu
- Urumqi Key Laboratory of Green Catalysis and Synthesis Technology Key Laboratory of Oil and Gas Fine Chemicals, Ministry of Education & Xinjiang Uygur Autonomous Region State Key Laboratory of Chemistry and Utilization of Carbon Based Energy Resources College of Chemistry Xinjiang University Urumqi 830046 P. R. China
- College of Future Technology Xinjiang University Urumqi 830046 P. R. China
| | - Yonghong Zhang
- Urumqi Key Laboratory of Green Catalysis and Synthesis Technology Key Laboratory of Oil and Gas Fine Chemicals, Ministry of Education & Xinjiang Uygur Autonomous Region State Key Laboratory of Chemistry and Utilization of Carbon Based Energy Resources College of Chemistry Xinjiang University Urumqi 830046 P. R. China
| |
Collapse
|
9
|
Shyamsivappan S, Vivek R, Suresh T, Naveen P, Kaviyarasu A, Amsaveni S, Athimoolam S, Mohan PS. New N-(3′-acetyl-8-nitro-2,3-dihydro-1 H,3′ H-spiro[quinoline-4,2′-[1,3,4]thiadiazol]-5′-yl) acetamides induced cell death in MCF-7 cells via G2/M phase cell cycle arrest. NEW J CHEM 2022. [DOI: 10.1039/d1nj02550c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A series of new N-(3′-acetyl-8-nitro-2,3-dihydro-1H,3′H-spiro[quinoline-4,2′-[1,3,4]thiadiazol]-5′-yl) acetamide derivatives were synthesized from potent 8-nitroquinoline-thiosemicarbazones.
Collapse
Affiliation(s)
- Selvaraj Shyamsivappan
- School of Chemical Sciences, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
- Department of Chemistry, Dr N.G.P. Arts and Science College, Coimbatore 641048, Tamil Nadu, India
| | - Raju Vivek
- Cancer Research Program (CRP), Bio-Nano Therapeutics Research Laboratory, School of Life Sciences, Department of Zoology, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Thangaraj Suresh
- School of Chemical Sciences, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Palanivel Naveen
- Department of Chemistry, Dr N.G.P. Arts and Science College, Coimbatore 641048, Tamil Nadu, India
| | - Adhigaman Kaviyarasu
- School of Chemical Sciences, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | - Sundarasamy Amsaveni
- School of Chemical Sciences, Bharathiar University, Coimbatore 641046, Tamil Nadu, India
| | | | | |
Collapse
|
10
|
Luo G, Lin X, Ren S, Wu S, Wang X, Ma L, Xiang H. Development of novel tetrahydroisoquinoline-hydroxamate conjugates as potent dual SERDs/HDAC inhibitors for the treatment of breast cancer. Eur J Med Chem 2021; 226:113870. [PMID: 34610548 DOI: 10.1016/j.ejmech.2021.113870] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 09/23/2021] [Accepted: 09/23/2021] [Indexed: 12/25/2022]
Abstract
Concomitant inhibition of estrogen receptor alpha (ERα) and histone deacetylase (HDAC) signaling has been proven effective in endocrine-resistant ER+ breast cancers. Herein, a series of tetrahydroisoquinoline (THIQ)-hydroxamate conjugates were rationally designed and synthesized as dual SERDs/HDAC inhibitors by incorporating the hydroxamate, a known HDAC pharmacophore, into a privileged THIQ scaffold of selective ERα degraders (SERDs). Some of these THIQ-hydroxamate conjugates displayed remarkable HDAC6 inhibition and improved antiproliferative activity against MCF-7 cells. Particularly, the most potent HDAC inhibitor 19k also exhibits potent ERα binding affinity, good ERα degradation efficacy and the best antiproliferative activity. Besides, 19k displayed superior antitumor efficacy than the drug combination (Fulvestrant + SAHA) through promoting ERα degradation and histone acetylation in an MCF-7 xenograft model, without causing observable toxicity. Collectively, this study validates the therapeutic potential of a dual-acting compound with potent ERα degradation efficacy and HDAC6 inhibition in breast cancer.
Collapse
Affiliation(s)
- Guoshun Luo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Xin Lin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Shengnan Ren
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Shuangjie Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Xin Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Luyu Ma
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hua Xiang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
11
|
Scott JS, Barlaam B. Selective estrogen receptor degraders (SERDs) and covalent antagonists (SERCAs): a patent review (2015-present). Expert Opin Ther Pat 2021; 32:131-151. [PMID: 34763600 DOI: 10.1080/13543776.2022.2006185] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION The estrogen receptor (ER) is a clinically validated oncology target with a pivotal role in hormonally driven breast cancer, the most prevalent form of female cancer. Current treatments which directly modulate ER include antagonists (SERMs), such as tamoxifen, and degraders (SERDs), such as fulvestrant which is administered by intramuscular injection. AREAS COVERED This review covers patent applications that claim estrogen receptor degraders (SERDs) and covalent antagonists (SERCAs) between the period January 2015 to June 2021. A total of 114 patent applications from 23 different applicants are evaluated with stratification into acidic SERDs, basic SERDs and SERCAs. EXPERT OPINION The clinical success of fulvestrant in the treatment of ER+ breast cancer has spurred research over the last decade into the discovery and development of novel SERDs, with a particular focus on the discovery of orally bioavailable drugs. This has resulted in a diverse range of candidates entering clinical trials. Although some have faltered in development, a cohort of oral SERDs has generated encouraging efficacy and safety data that has allowed advancement into late stage clinical trials. Data from these trials is eagerly awaited, with these molecules having the potential to offer significant benefits in the treatment of ER+ breast cancer.
Collapse
Affiliation(s)
- James S Scott
- Oncology R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | - Bernard Barlaam
- Oncology R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| |
Collapse
|
12
|
Discovery of GNE-502 as an orally bioavailable and potent degrader for estrogen receptor positive breast cancer. Bioorg Med Chem Lett 2021; 50:128335. [PMID: 34425201 DOI: 10.1016/j.bmcl.2021.128335] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 08/09/2021] [Accepted: 08/16/2021] [Indexed: 11/20/2022]
Abstract
Fulvestrant is an FDA-approved drug with a dual mechanism of action (MOA), acting as a full antagonist and degrader of the estrogen receptor protein. A significant limitation of fulvestrant is the dosing regimen required for efficacy. Due to its high lipophilicity and poor pharmacokinetic profile, fulvestrant needs to be administered through intramuscular injections which leads to injection site soreness. This route of administration also limits the dose and target occupancy in patients. We envisioned a best-in-class molecule that would function with the same dual MOA as fulvestrant, but with improved physicochemical properties and would be orally bioavailable. Herein we report our progress toward that goal, resulting in a new lead GNE-502 which addressed some of the liabilities of our previously reported lead molecule GNE-149.
Collapse
|
13
|
Faheem, Karan Kumar B, Venkata Gowri Chandra Sekhar K, Chander S, Kunjiappan S, Murugesan S. 1,2,3,4-Tetrahydroisoquinoline (THIQ) as privileged scaffold for anticancer de novo drug design. Expert Opin Drug Discov 2021; 16:1119-1147. [PMID: 33908322 DOI: 10.1080/17460441.2021.1916464] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Cancer is a dreadful disorder that is emerging as one of the leading causes of mortality across the globe. The complex tumor environment, supplemented with drawbacks of the existing drugs, has made it a global health concern. The Tetrahydroisoquinoline (THIQ) ring holds an important position in medicinal chemistry due to its wide range of pharmacological properties. Several THIQ based natural products have been previously explored for their antitumor properties, making it a vital scaffold for anticancer drug design.Areas covered: This review article addresses the potential of THIQ as anticancer agents. Various medicinal chemistry strategies employed for the design and development of THIQ analogs as inhibitors or modulators of relevant anticancer targets have been discussed in detail. Moreover, the common strategies employed for the synthesis of the core scaffold are also highlighted.Expert opinion: Evidently, THIQs have tremendous potential in anticancer drug design. Some of these analogs exhibited potent activity against various cancer molecular targets. However, there are some drawbacks, such as selectivity that need addressing. The synthetic ease for constructing the core scaffold complimented with its reactivity makes it ideal for further structure-activity relationship studies. For these reasons, THIQ is a privileged scaffold for the design and development of novel anticancer agents.
Collapse
Affiliation(s)
- Faheem
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani, India
| | - Banoth Karan Kumar
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani, India
| | | | - Subhash Chander
- Amity Institute of Phytomedicine and Phytochemistry, Amity University Uttar Pradesh, Noida, India
| | - Selvaraj Kunjiappan
- Department of Biotechnology, Kalasalingam Academy of Research and Education, Krishnankoil, India
| | - Sankaranarayanan Murugesan
- Medicinal Chemistry Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science-Pilani, Pilani, India
| |
Collapse
|
14
|
Terashima K, Kawasaki-Takasuka T, Yamazaki T. Construction of fully substituted carbon centers containing a heteroatom and a CF 3 group via in situ generated p-quinone methides. Org Biomol Chem 2021; 19:1305-1314. [PMID: 33503080 DOI: 10.1039/d0ob02469d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
1,6-Conjugate additions of in situ generated δ-CF3-δ-substituted p-quinone methides have been achieved with a variety of heteronucleophiles under mild conditions, which led to facile and practical construction of fully substituted carbon centers including a heteroatom and a CF3 group. In particular, it was revealed that some amines themselves worked for efficient cleavage of the TBS protective group, and addition of a catalytic amount of an appropriate Brønsted acid was found to sometimes improve the progress of the desired process.
Collapse
Affiliation(s)
- Kyu Terashima
- Division of Applied Chemistry, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei 184-8588, Japan.
| | - Tomoko Kawasaki-Takasuka
- Division of Applied Chemistry, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei 184-8588, Japan.
| | - Takashi Yamazaki
- Division of Applied Chemistry, Institute of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Nakamachi, Koganei 184-8588, Japan.
| |
Collapse
|
15
|
Matsumoto S, Marumoto H, Akazome M, Otani Y, Kaiho T. Chemoselective Reduction of α, β-Unsaturated Carbonyl and Carboxylic Compounds by Hydrogen Iodide. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2021. [DOI: 10.1246/bcsj.20200341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Shoji Matsumoto
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
- Soft Molecular Activation Research Center (SMARC), Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Hayato Marumoto
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Motohiro Akazome
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| | - Yasuhiko Otani
- GODO SHIGEN CO., LTD., 1365 Nanaido, Chosei-mura, Chosei-gun, Chiba 299-4333, Japan
| | - Tatsuo Kaiho
- GODO SHIGEN CO., LTD., 1365 Nanaido, Chosei-mura, Chosei-gun, Chiba 299-4333, Japan
- Chiba Iodine Resource Innovation Center (CIRIC), Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba 263-8522, Japan
| |
Collapse
|
16
|
Cirillo D, Angelucci F, Bjørsvik H. Functionalization of the Imidazole Backbone by Means of a Tailored and Optimized Oxidative Heck Cross‐Coupling. Adv Synth Catal 2020. [DOI: 10.1002/adsc.202000909] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Davide Cirillo
- Department of Chemistry University of Bergen Allégaten 41 N-5007 Bergen Norway
| | - Francesco Angelucci
- Department of Chemistry University of Bergen Allégaten 41 N-5007 Bergen Norway
| | - Hans‐René Bjørsvik
- Department of Chemistry University of Bergen Allégaten 41 N-5007 Bergen Norway
| |
Collapse
|
17
|
Lu Y, Liu W. Selective Estrogen Receptor Degraders (SERDs): A Promising Strategy for Estrogen Receptor Positive Endocrine-Resistant Breast Cancer. J Med Chem 2020; 63:15094-15114. [PMID: 33138369 DOI: 10.1021/acs.jmedchem.0c00913] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Estrogen receptor (ER) plays important roles in gene transcription and the proliferation of ER positive breast cancers. Selective modulation of ER has been a therapeutic target for this specific type of breast cancer for more than 30 years. Selective estrogen receptor modulators (SERMs) and aromatase inhibitors (AIs) have been demonstrated to be effective therapeutic approaches for ER positive breast cancers. Unfortunately, 30-50% of ER positive tumors become resistant to SERM/AI treatment after 3-5 years. Fulvestrant, the only approved selective estrogen receptor degrader (SERD), is currently an important therapeutic approach for the treatment of endocrine-resistant breast cancers. The poor pharmacokinetic properties of fulvestrant have inspired the development of a new generation of oral SERDs to overcome drug resistance. In this review, we describe recent advances in ERα structure, functions, and mechanisms of endocrine resistance and summarize the development of oral SERDs in both academic and industrial areas.
Collapse
Affiliation(s)
- Yunlong Lu
- School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.,Department of Pharmaceutical Sciences, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois 60612, United States
| | - Wukun Liu
- School of Medicine & Holistic Integrative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China.,State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, P. R. China.,State Key Laboratory of Coordination Chemistry, Nanjing University, Nanjing 210023, P. R. China
| |
Collapse
|
18
|
Wang L, Sharma A. The Quest for Orally Available Selective Estrogen Receptor Degraders (SERDs). ChemMedChem 2020; 15:2072-2097. [PMID: 32916035 DOI: 10.1002/cmdc.202000473] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/31/2020] [Indexed: 01/10/2023]
Abstract
Estrogen receptor-alpha (ERα) is the target of endocrine therapies for the treatment of more than 70 % of ERα-positive breast cancers. Selective estrogen receptor degraders (SERDs) antagonize estrogen binding and target the receptor for degradation, representing the last line of treatment for resistant metastatic breast cancer patients. However, the clinical efficacy of the lone clinically approved SERD (Fulvestrant) is limited by its poor oral bioavailability. Recently, several analogues of GW5638, an acrylic acid-based ERα ligand developed by Glaxo Research Institute in 1994, have been reported as promising orally bioavailable SERDs. Some of these compounds are currently in clinical trials, while various other structurally novel SERDs have also been reported by pharma as well as academic research groups. This review provides a critical analysis of the recent developments in orally available SERDs, with a focus on the structure-activity relationships, binding interactions and pharmacokinetic properties of these compounds.
Collapse
Affiliation(s)
- Lucia Wang
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey, 07030, USA
| | - Abhishek Sharma
- Department of Chemistry and Chemical Biology, Stevens Institute of Technology, Hoboken, New Jersey, 07030, USA
| |
Collapse
|
19
|
Meng Q, Ma X, Xie B, Deng X, Huang J, Zhou HB, Dong C. Establishment of evaluation criteria for the development of high quality ERα-targeted fluorescent probes. Analyst 2020; 145:5989-5995. [PMID: 32856648 DOI: 10.1039/d0an01172j] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
ERα-targeted fluorescent probes are important tools for ERα study. In order to develop high quality ERα-targeted probes, a sound and complete evaluation system is essential but has not been established yet. Herein, we set up a series of evaluation criteria for ERα-targeted fluorescent probes including ERα binding affinity, fluorescence quantum yield, cytotoxicity, ERα tracking capacity, ERα selectivity and ERα labeling ability. To verify the practicability of the evaluation criteria, we designed and synthesized two ERα-targeted fluorescent probes and fully characterized their properties based on the proposed evaluation criteria. It showed that the probes exhibited better performance. Moreover, we applied the probes in MCF-7 cells to study the ERα motion characteristics for the first time. We hope that our evaluation criteria could be helpful for the establishment of a complete evaluation system for ERα-targeted fluorescent probes.
Collapse
Affiliation(s)
- Qiuyu Meng
- Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Hubei Provincial Key Laboratory of Developmentally Originated Disease, State Key Laboratory of Virology, Wuhan University School of Pharmaceutical Sciences, Wuhan 430071, China.
| | | | | | | | | | | | | |
Collapse
|
20
|
Talele TT. Opportunities for Tapping into Three-Dimensional Chemical Space through a Quaternary Carbon. J Med Chem 2020; 63:13291-13315. [PMID: 32805118 DOI: 10.1021/acs.jmedchem.0c00829] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
A quaternary carbon bears four other carbon substituents or combination of four non-hydrogen substituents at four vertices of a tetrahedron. The spirocyclic quaternary carbon positioned at the center of a bioactive molecule offers conformational rigidity, which in turn reduces the penalty for conformational entropy. The quaternary carbon is a predominant feature of natural product structures and has been associated with more effective and selective binding to target proteins compared to planar compounds with a high sp2 count. The presence of a quaternary carbon stereocenter allows the exploration of novel chemical space to obtain new molecules with enhanced three-dimensionality. These characteristics, coupled to an increasing awareness to develop sp3-rich molecules, boosted utility of quaternary carbon stereocenters in bioactive compounds. It is hoped that this Perspective will inspire the chemist to utilize quaternary carbon stereocenters to enhance potency, selectivity, and other drug-like properties.
Collapse
Affiliation(s)
- Tanaji T Talele
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York 11439, United States
| |
Collapse
|
21
|
Liang J, Blake R, Chang J, Friedman LS, Goodacre S, Hartman S, Ingalla ER, Kiefer JR, Kleinheinz T, Labadie S, Li J, Lai KW, Liao J, Mody V, McLean N, Metcalfe C, Nannini M, Otwine D, Ran Y, Ray N, Roussel F, Sambrone A, Sampath D, Vinogradova M, Wai J, Wang T, Yeap K, Young A, Zbieg J, Zhang B, Zheng X, Zhong Y, Wang X. Discovery of GNE-149 as a Full Antagonist and Efficient Degrader of Estrogen Receptor alpha for ER+ Breast Cancer. ACS Med Chem Lett 2020; 11:1342-1347. [PMID: 32551022 PMCID: PMC7294714 DOI: 10.1021/acsmedchemlett.0c00224] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 05/26/2020] [Indexed: 12/19/2022] Open
Abstract
Estrogen receptor alpha (ERα) is a well-validated drug target for ER-positive (ER+) breast cancer. Fulvestrant is FDA-approved to treat ER+ breast cancer and works through two mechanisms-as a full antagonist and selective estrogen receptor degrader (SERD)-but lacks oral bioavailability. Thus, we envisioned a "best-in-class" molecule with the same dual mechanisms as fulvestrant, but with significant oral exposure. Through lead optimization, we discovered a tool molecule 12 (GNE-149) with improved degradation and antiproliferative activity in both MCF7 and T47D cells. To illustrate the binding mode and key interactions of this scaffold with ERα, we obtained a cocrystal structure of 6 that showed ionic interaction of azetidine with Asp351 residue. Importantly, 12 showed favorable metabolic stability and good oral exposure. 12 exhibited antagonist effect in the uterus and demonstrated robust dose-dependent efficacy in xenograft models.
Collapse
Affiliation(s)
- Jun Liang
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Robert Blake
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jae Chang
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Lori S. Friedman
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Simon Goodacre
- Charles
River Discovery Research Services UK Limited, 7-9 Spire Green Center, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Steven Hartman
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Ellen Rei Ingalla
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - James R. Kiefer
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Tracy Kleinheinz
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Sharada Labadie
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jun Li
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Kwong Wah Lai
- WuXi
AppTec Co., Ltd, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, P. R.
China
| | - Jiangpeng Liao
- WuXi
AppTec Co., Ltd, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, P. R.
China
| | - Vidhi Mody
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Neville McLean
- Charles
River Discovery Research Services UK Limited, 7-9 Spire Green Center, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Ciara Metcalfe
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Michelle Nannini
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Daniel Otwine
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Yingqing Ran
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Nick Ray
- Charles
River Discovery Research Services UK Limited, 7-9 Spire Green Center, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Fabien Roussel
- Charles
River Discovery Research Services UK Limited, 7-9 Spire Green Center, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Amy Sambrone
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Deepak Sampath
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Maia Vinogradova
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - John Wai
- WuXi
AppTec Co., Ltd, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, P. R.
China
| | - Tao Wang
- WuXi
AppTec Co., Ltd, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, P. R.
China
| | - Kuen Yeap
- Charles
River Discovery Research Services UK Limited, 7-9 Spire Green Center, Flex Meadow, Harlow, Essex CM19 5TR, United Kingdom
| | - Amy Young
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Jason Zbieg
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Birong Zhang
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Xiaoping Zheng
- WuXi
AppTec Co., Ltd, 288 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, P. R.
China
| | - Yu Zhong
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| | - Xiaojing Wang
- Genentech,
Inc., 1 DNA Way, South San Francisco, California 94080, United States
| |
Collapse
|
22
|
Hanan EJ, Liang J, Wang X, Blake RA, Blaquiere N, Staben ST. Monomeric Targeted Protein Degraders. J Med Chem 2020; 63:11330-11361. [DOI: 10.1021/acs.jmedchem.0c00093] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
23
|
Zhang C, Li Q, Ren Y, Liu F. Molecular modeling studies of benzothiophene-containing derivatives as promising selective estrogen receptor downregulators: a combination of 3D-QSAR, molecular docking and molecular dynamics simulations. J Biomol Struct Dyn 2020; 39:2702-2723. [PMID: 32249694 DOI: 10.1080/07391102.2020.1751717] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Selective estrogen receptor downregulators (SERDs) for the treatment of positive breast cancer can act both as estrogen alpha receptor (ERα) antagonists and degraders. In this study, the optimal antagonist models (CoMFA-A, q2 = 0.660, r2 = 0.996; CoMSIA-A, q2 = 0.728, r2 = 0.992) and degrader models (CoMFA-D, q2 = 0.850, r2 = 0.996; CoMSIA-D, q2 = 0.719, r2 = 0.995) of a series of potent benzothiophene-containing SERDs were constructed to explore the three-dimensional quantitative structure-activity relationship. Internal and external validation indicated that all models exhibited good applicability, high predictive ability and robustness. Contour maps revealed the relationships between the essential structural features and antagonistic and degradation activities. Additionally, molecular docking, molecular dynamics and free energy calculation studies were further performed to investigate the detailed binding mode. Results indicated that several key residues, ARG394, GLU353, PHE404 and ILE424, were crucial for the stability of the ligand binding domain. The hydrophobic, electrostatic and Van der Waals interactions played significant effect on the binding affinity. Finally, ten novel compounds were designed based on above findings, where the predicted activity of compound D8 was equivalent to that of the compound LSZ102. 3D-QSAR, ADMET and bioavailability predictions indicated that all designed compounds with good predicted activity, good physicochemical and bioavailability could be potential candidates for SERDs. These results and combinations of computational methods provided guidance for the rational drug design of novel potential SERDs.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Cuihua Zhang
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, China
| | - Qunlin Li
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, China
| | - Yujie Ren
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, China
| | - Fei Liu
- School of Chemical and Environmental Engineering, Shanghai Institute of Technology, Shanghai, China
| |
Collapse
|
24
|
Shagufta, Ahmad I, Mathew S, Rahman S. Recent progress in selective estrogen receptor downregulators (SERDs) for the treatment of breast cancer. RSC Med Chem 2020; 11:438-454. [PMID: 33479648 PMCID: PMC7580774 DOI: 10.1039/c9md00570f] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 02/11/2020] [Indexed: 12/11/2022] Open
Abstract
Selective estrogen receptor downregulators (SERDs) are a novel class of compounds capable of reducing the ERα protein level and blocking ER activity. Therefore, SERDs are considered as a significant therapeutic approach to treat ER+ breast cancer in both early stage and more advanced drug-resistant cases. After the FDA approval of a steroidal drug, fulvestrant, as a SERD for the treatment of breast cancer in patients who have progressed on antihormonal agents, several molecules with diverse chemical structures have been rapidly developed, studied and evaluated for selective estrogen receptor downregulation activity. Here we compile the promising SERDs reported in recent years and discuss the chemical structure and pharmacological profile of the most potent compound of the considered series. Because of the availability of only a limited number of effective drugs for the treatment of breast cancer, the quest for a potent SERD with respectable activity and bioavailability is still ongoing. The goal of this article is to make available to the reader an overview of the current progress in SERDs and provide clues for the future discovery and development of novel pharmacological potent SERDs for the treatment of breast cancer.
Collapse
Affiliation(s)
- Shagufta
- Department of Mathematics and Natural Sciences , School of Arts and Sciences , American University of Ras Al Khaimah , P. O. Box 10021 , Ras Al Khaimah , United Arab Emirates . ;
| | - Irshad Ahmad
- Department of Mathematics and Natural Sciences , School of Arts and Sciences , American University of Ras Al Khaimah , P. O. Box 10021 , Ras Al Khaimah , United Arab Emirates . ;
| | - Shimy Mathew
- Department of Biotechnology , School of Arts and Sciences , American University of Ras Al Khaimah , P. O. Box 10021 , Ras Al Khaimah , United Arab Emirates
| | - Sofia Rahman
- Department of Biotechnology , School of Arts and Sciences , American University of Ras Al Khaimah , P. O. Box 10021 , Ras Al Khaimah , United Arab Emirates
| |
Collapse
|
25
|
Gao Z, Xia Z, Dai L, Ye S. N‐Heterocyclic Carbene Catalyzed Photooxidation: Intramolecular Cross Dehydrogenative Coupling of Tetrahydroisoquinoline‐Tethered Aldehydes. Adv Synth Catal 2020. [DOI: 10.1002/adsc.202000164] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Zhong‐Hua Gao
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Molecular Recognition and Function, Institute of ChemistryChinese Academy of Sciences Beijing 100190 People's Republic of China
| | - Zi‐Hao Xia
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Molecular Recognition and Function, Institute of ChemistryChinese Academy of Sciences Beijing 100190 People's Republic of China
- University of Chinese Academy of Sciences Beijing 100049 People's Republic of China
| | - Lei Dai
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Molecular Recognition and Function, Institute of ChemistryChinese Academy of Sciences Beijing 100190 People's Republic of China
- University of Chinese Academy of Sciences Beijing 100049 People's Republic of China
| | - Song Ye
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Molecular Recognition and Function, Institute of ChemistryChinese Academy of Sciences Beijing 100190 People's Republic of China
- University of Chinese Academy of Sciences Beijing 100049 People's Republic of China
| |
Collapse
|
26
|
In silico toxicological analyzes of selected toxic compounds from dumpsite or contaminated soils on human health. NOVA BIOTECHNOLOGICA ET CHIMICA 2019. [DOI: 10.2478/nbec-2019-0017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
The soil is a key component of natural ecosystems because environmental sustainability depends largely on a sustainable soil ecosystem. The objective of this study was to predict the impact of selected toxic compounds from dumpsite or contaminated soils on human health at the molecular level of biological processes. The in silico methods that were used include toxicokinetics and target gene prediction, molecular docking, and gene expressing network analysis. The result showed bisphenol A (BPA), 2,20-bis(p-chlorophenyl)-1,1-dichloroethane (DDD), 2,20-bis(p-chlorophenyl)-1,1-trichloroethane (DDT), diethylhexyl phthalate (DEHP), nonylphenol (NP) and tetrachlorodibenzodioxin (TCDD) as the active toxic compounds that can modulate biological system and are considered as potential cause of several diseases including cancer. The principal target genes include substance-P receptor (also known as Neurokinin 1 receptor), 5-hydroxytryptamine receptor, human serotonin transporter; estrogen receptor alpha; and aryl hydrocarbon receptor. These genes implicated SUZ12, STAT3, and TRIM28 as the major transcription factors while mitogen-activated protein kinases and cyclin-dependent kinases were the major kinases from the protein-protein interaction. All the six toxicants investigated showed good free binding energies (ΔG) which were below - 5.0 kcal.mol−1. These toxic compounds showed ligand efficiency greater than 0.25 kcal.mol−1. HA and would possibly cause fatal damage on human health. The order of in silico predicted toxicity of these compounds were BPA > DDD = DDT > TCDD > NP > DEHP. Our results identified potential threats, which the selected toxicants can pose to public health. More importantly, it provides basis for investigation of super bugs (microorganisms) that can remediate these toxicants in our environment. Environmental monitoring and modern wastes management system should be implemented and enforced in the affected countries in order to safeguard the health of the citizenry.
Collapse
|
27
|
Scott JS, Breed J, Carbajo RJ, Davey PR, Greenwood R, Huynh HK, Klinowska T, Morrow CJ, Moss TA, Polanski R, Nissink JWM, Varnes J, Yang B. Building Bridges in a Series of Estrogen Receptor Degraders: An Application of Metathesis in Medicinal Chemistry. ACS Med Chem Lett 2019; 10:1492-1497. [PMID: 31620239 DOI: 10.1021/acsmedchemlett.9b00370] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 09/23/2019] [Indexed: 11/30/2022] Open
Abstract
Herein we report the use of metathesis to construct a novel tetracyclic core in a series of estrogen receptor degraders. This improved the chemical stability, as assessed using an NMR-MS based assay, and gave a molecule with excellent physicochemical properties and pharmacokinetics in rat. X-ray crystallography established minimal perturbation of the bridged compounds relative to the unbridged analogues in the receptor binding pocket. Unfortunately, despite retaining excellent binding to ERα, this adversely affected the ability of the compounds to degrade the receptor.
Collapse
Affiliation(s)
- James S. Scott
- Oncology R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | - Jason Breed
- Discovery Sciences, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | | | - Paul R. Davey
- Oncology R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | - Ryan Greenwood
- Oncology R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | - Hoan K. Huynh
- Oncology R&D, AstraZeneca, R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | | | | | - Thomas A. Moss
- Oncology R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | | | | | - Jeffrey Varnes
- Oncology R&D, AstraZeneca, R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Bin Yang
- Oncology R&D, AstraZeneca, R&D Boston, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| |
Collapse
|
28
|
Wardell SE, Yllanes AP, Chao CA, Bae Y, Andreano KJ, Desautels TK, Heetderks KA, Blitzer JT, Norris JD, McDonnell DP. Pharmacokinetic and pharmacodynamic analysis of fulvestrant in preclinical models of breast cancer to assess the importance of its estrogen receptor-α degrader activity in antitumor efficacy. Breast Cancer Res Treat 2019; 179:67-77. [PMID: 31562570 PMCID: PMC6985185 DOI: 10.1007/s10549-019-05454-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 09/19/2019] [Indexed: 12/11/2022]
Abstract
Purpose Fulvestrant is a selective estrogen receptor downregulator (SERD) that is approved for first- or second-line use as a single agent or in combination with cyclin dependent kinase or phosphatidylinositol 3-kinase inhibitors for the treatment of metastatic breast cancer. Fulvestrant exhibits exceptionally effective antitumor activity in preclinical models of breast cancer, a success that has been attributed to its robust SERD activity despite modest receptor downregulation in patient tumors. By modeling human exposures in animal models we probe the absolute need for SERD activity. Methods Three xenograft models of endocrine therapy-resistant breast cancer were used to evaluate the efficacy of fulvestrant administered in doses historically used in preclinical studies in the field or by using a dose regimen intended to model clinical exposure levels. Pharmacokinetic and pharmacodynamic analyses were conducted to evaluate plasma exposure and intratumoral ER downregulation. Results A clinically relevant 25 mg/kg dose of fulvestrant exhibited antitumor efficacy comparable to the historically used 200 mg/kg dose, but at this lower dose it did not result in robust ER downregulation. Further, the antitumor efficacy of the lower dose of fulvestrant was comparable to that observed for other oral SERDs currently in development. Conclusion The use of clinically unachievable exposure levels of fulvestrant as a benchmark in preclinical development of SERDs may negatively impact the selection of those molecules that are advanced for clinical development. Further, these studies suggest that antagonist efficacy, as opposed to SERD activity, is likely to be the primary driver of clinical response. Electronic supplementary material The online version of this article (10.1007/s10549-019-05454-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Suzanne E Wardell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Box 3813, Durham, NC, 27710, USA
| | - Alexander P Yllanes
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Box 3813, Durham, NC, 27710, USA
| | - Christina A Chao
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Box 3813, Durham, NC, 27710, USA
| | - Yeeun Bae
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Box 3813, Durham, NC, 27710, USA
| | - Kaitlyn J Andreano
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Box 3813, Durham, NC, 27710, USA
| | - Taylor K Desautels
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Box 3813, Durham, NC, 27710, USA
| | - Kendall A Heetderks
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Box 3813, Durham, NC, 27710, USA
| | | | - John D Norris
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Box 3813, Durham, NC, 27710, USA
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Box 3813, Durham, NC, 27710, USA.
| |
Collapse
|
29
|
Li Y, Zhang S, Zhang J, Hu Z, Xiao Y, Huang J, Dong C, Huang S, Zhou HB. Exploring the PROTAC degron candidates: OBHSA with different side chains as novel selective estrogen receptor degraders (SERDs). Eur J Med Chem 2019; 172:48-61. [DOI: 10.1016/j.ejmech.2019.03.058] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/23/2019] [Accepted: 03/24/2019] [Indexed: 12/23/2022]
|
30
|
Scott JS, Bailey A, Buttar D, Carbajo RJ, Curwen J, Davey PRJ, Davies RDM, Degorce SL, Donald C, Gangl E, Greenwood R, Groombridge SD, Johnson T, Lamont S, Lawson M, Lister A, Morrow CJ, Moss TA, Pink JH, Polanski R. Tricyclic Indazoles-A Novel Class of Selective Estrogen Receptor Degrader Antagonists. J Med Chem 2019; 62:1593-1608. [PMID: 30640465 DOI: 10.1021/acs.jmedchem.8b01837] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Herein, we report the identification and synthesis of a series of tricyclic indazoles as a novel class of selective estrogen receptor degrader antagonists. Replacement of a phenol, present in our previously reported tetrahydroisoquinoline scaffold, with an indazole group led to the removal of a reactive metabolite signal in an in vitro glutathione trapping assay. Further optimization, guided by X-ray crystal structures and NMR conformational work, varied the alkyl side chain and pendant aryl group and resulted in compounds with low turnover in human hepatocytes and enhanced chemical stability. Compound 9 was profiled as a representative of the series in terms of pharmacology and demonstrated the desired estrogen receptor α degrader-antagonist profile and demonstrated activity in a xenograft model of breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Eric Gangl
- Oncology IMED Biotech Unit , AstraZeneca, R&D Boston , 35 Gatehouse Drive , Waltham , Massachusetts 02451 , United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Tria GS, Abrams T, Baird J, Burks HE, Firestone B, Gaither LA, Hamann LG, He G, Kirby CA, Kim S, Lombardo F, Macchi KJ, McDonnell DP, Mishina Y, Norris JD, Nunez J, Springer C, Sun Y, Thomsen NM, Wang C, Wang J, Yu B, Tiong-Yip CL, Peukert S. Discovery of LSZ102, a Potent, Orally Bioavailable Selective Estrogen Receptor Degrader (SERD) for the Treatment of Estrogen Receptor Positive Breast Cancer. J Med Chem 2018; 61:2837-2864. [PMID: 29562737 DOI: 10.1021/acs.jmedchem.7b01682] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In breast cancer, estrogen receptor alpha (ERα) positive cancer accounts for approximately 74% of all diagnoses, and in these settings, it is a primary driver of cell proliferation. Treatment of ERα positive breast cancer has long relied on endocrine therapies such as selective estrogen receptor modulators, aromatase inhibitors, and selective estrogen receptor degraders (SERDs). The steroid-based anti-estrogen fulvestrant (5), the only approved SERD, is effective in patients who have not previously been treated with endocrine therapy as well as in patients who have progressed after receiving other endocrine therapies. Its efficacy, however, may be limited due to its poor physicochemical properties. We describe the design and synthesis of a series of potent benzothiophene-containing compounds that exhibit oral bioavailability and preclinical activity as SERDs. This article culminates in the identification of LSZ102 (10), a compound in clinical development for the treatment of ERα positive breast cancer.
Collapse
Affiliation(s)
- George S Tria
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Tinya Abrams
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Jason Baird
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Heather E Burks
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Brant Firestone
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - L Alex Gaither
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Lawrence G Hamann
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Guo He
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Christina A Kirby
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Sunkyu Kim
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Franco Lombardo
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Kaitlin J Macchi
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Donald P McDonnell
- Department of Pharmacology and Cancer Biology , Duke University School of Medicine , Durham , North Carolina 27710 , United States
| | - Yuji Mishina
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - John D Norris
- Department of Pharmacology and Cancer Biology , Duke University School of Medicine , Durham , North Carolina 27710 , United States
| | - Jill Nunez
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Clayton Springer
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Yingchuan Sun
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Noel M Thomsen
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Chunrong Wang
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Jianling Wang
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Bing Yu
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Choi-Lai Tiong-Yip
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| | - Stefan Peukert
- Novartis Institutes for BioMedical Research, Inc. , 250 Massachusetts Avenue , Cambridge , Massachusetts 02139 , United States
| |
Collapse
|
32
|
Jayaram V, Sridhar T, Sharma GVM, Berrée F, Carboni B. Synthesis of Polysubstituted Isoquinolines and Related Fused Pyridines from Alkenyl Boronic Esters via a Copper-Catalyzed Azidation/Aza-Wittig Condensation Sequence. J Org Chem 2018; 83:843-853. [DOI: 10.1021/acs.joc.7b02831] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Vankudoth Jayaram
- Organic
and Biomolecular Chemistry Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad-500 007, India
| | - Tailor Sridhar
- Organic
and Biomolecular Chemistry Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad-500 007, India
| | - Gangavaram V. M. Sharma
- Organic
and Biomolecular Chemistry Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad-500 007, India
| | - Fabienne Berrée
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000 Rennes, France
| | - Bertrand Carboni
- Univ Rennes, CNRS, ISCR (Institut des Sciences Chimiques de Rennes) - UMR 6226, F-35000 Rennes, France
| |
Collapse
|