1
|
Li D, Xie Q, Yang M, Cai Y, Sun K, Jiang S, Yu S, Liu L, Zhang Y, Yu B, Tu W, Li L. Lead Identification of Novel Naphthyridine Derivatives as Potent SOS1 Inhibitors. ACS Med Chem Lett 2024; 15:958-964. [PMID: 38894918 PMCID: PMC11181497 DOI: 10.1021/acsmedchemlett.4c00156] [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: 04/08/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
SOS1, a guanine nucleotide exchange factor (GEF), plays a critical role in catalyzing the conversion of KRAS from its GDP- to GTP-bound form, regardless of KRAS mutation status, and represents a promising new drug target to treat all KRAS-driven tumors. Herein, we employed a scaffold hopping strategy to design, synthesize, and optimize a series of novel binary ring derivatives as SOS1 inhibitors. Among them, compound 10f (HH0043) displayed potent activities in both biochemical and cellular assays and favorable pharmacokinetic profiles. Oral administration of HH0043 resulted in a significant tumor inhibitory effect in a subcutaneous KRAS G12C-mutated NCI-H358 (human lung cancer cell line) xenograft mouse model, and the tumor inhibitory effect of HH0043 was superior to that of BI-3406 at the same dose (total growth inhibition, TGI: 76% vs 49%). On the basis of these results, HH0043, with a novel 1,7-naphthyridine scaffold that is distinct from currently reported SOS1 inhibitors, is nominated as the lead compound for this discovery project.
Collapse
Affiliation(s)
- Dongsheng Li
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865# Zuchongzhi Road Zhangjiang Science City, Shanghai 201203, China
| | - Qing Xie
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865# Zuchongzhi Road Zhangjiang Science City, Shanghai 201203, China
| | - Maozhi Yang
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865# Zuchongzhi Road Zhangjiang Science City, Shanghai 201203, China
| | - Yalei Cai
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865# Zuchongzhi Road Zhangjiang Science City, Shanghai 201203, China
| | - Kang Sun
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865# Zuchongzhi Road Zhangjiang Science City, Shanghai 201203, China
| | - Shujuan Jiang
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865# Zuchongzhi Road Zhangjiang Science City, Shanghai 201203, China
| | - Songda Yu
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865# Zuchongzhi Road Zhangjiang Science City, Shanghai 201203, China
| | - Lei Liu
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865# Zuchongzhi Road Zhangjiang Science City, Shanghai 201203, China
| | - Yixiang Zhang
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865# Zuchongzhi Road Zhangjiang Science City, Shanghai 201203, China
| | - Bing Yu
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865# Zuchongzhi Road Zhangjiang Science City, Shanghai 201203, China
| | - Wangyang Tu
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865# Zuchongzhi Road Zhangjiang Science City, Shanghai 201203, China
| | - Leping Li
- Discovery & Early Development, Haihe Biopharma Co., Ltd., No 865# Zuchongzhi Road Zhangjiang Science City, Shanghai 201203, China
| |
Collapse
|
2
|
Zheng L, Zhang Y, Mei S, Xie T, Zou Y, Wang Y, Jing H, Xu S, Dramou P, Xu Z, Li J, Zhou Y, Niu MM. Discovery of a Potent Dual Son of Sevenless 1 (SOS1) and Epidermal Growth Factor Receptor (EGFR) Inhibitor for the Treatment of Prostate Cancer. J Med Chem 2024; 67:7130-7145. [PMID: 38630077 DOI: 10.1021/acs.jmedchem.3c02433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
Multitarget medications represent an appealing therapy against the disease with multifactorial abnormalities─cancer. Therefore, simultaneously targeting son of sevenless 1 (SOS1) and epidermal growth factor receptor (EGFR), two aberrantly expressed proteins crucial for the oncogenesis and progression of prostate cancer, may achieve active antitumor effects. Here, we discovered dual SOS1/EGFR-targeting compounds via pharmacophore-based docking screening. The most prominent compound SE-9 exhibited nanomolar inhibition activity against both SOS1 and EGFR and efficiently suppressed the phosphorylation of ERK and AKT in prostate cancer cells PC-3. Cellular assays also revealed that SE-9 displayed strong antiproliferative activities through diverse mechanisms, such as induction of cell apoptosis and G1 phase cell cycle arrest, as well as reduction of angiogenesis and migration. Further in vivo findings showed that SE-9 potently inhibited tumor growth in PC-3 xenografts without obvious toxicity. Overall, SE-9 is a novel dual-targeting SOS1/EGFR inhibitor that represents a promising treatment strategy for prostate cancer.
Collapse
Affiliation(s)
- Lufeng Zheng
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 211198, China
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 211198, China
| | - Yuxin Zhang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 211198, China
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 211198, China
| | - Shuang Mei
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 211198, China
| | - Tianyuan Xie
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 211198, China
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 211198, China
| | - Yunting Zou
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 211198, China
| | - Yuting Wang
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 211198, China
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 211198, China
| | - Han Jing
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 211198, China
- School of Life Science and Technology, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing 211198, China
| | - Shengtao Xu
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, China
| | - Pierre Dramou
- Department of Analytical Chemistry, School of Sciences, China Pharmaceutical University, Nanjing 210009, China
| | - Zhen Xu
- Department of Pharmacy, Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
| | - Jindong Li
- Taizhou School of Clinical Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou 225300, China
| | - Yang Zhou
- Department of Pathology, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou 213000, China
| | - Miao-Miao Niu
- Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
3
|
Du SG, Zhang HM, Ji YX, Tian YL, Wang D, Zhu K, Zhang QG, Liu SP. Polyphyllin VII Promotes Apoptosis in Breast Cancer by Inhibiting MAPK/ERK Signaling Pathway through Downregulation of SOS1. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:885-904. [PMID: 38716619 DOI: 10.1142/s0192415x24500368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Polyphyllin VII is a biologically active herbal monomer extracted from the traditional Chinese herbal medicine Chonglou. Many studies have demonstrated the anticancer activity of polyphyllin VII against various types of cancers, such as colon, liver, and lung cancer, but its effect on breast cancer has not been elucidated. In this study, we demonstrate that polyphyllin VII inhibited proliferation, increased production of intracellular reactive oxygen species, and decreased mitochondrial membrane potential in breast cancer cells. Notably, polyphyllin VII also induced apoptosis via the mitochondrial pathway. Transcriptome sequencing was used to analyze the targets of PPVII in regulating breast cancer cells. Mechanistic studies showed that polyphyllin VII downregulated Son of Sevenless1 (SOS1) and inhibited the MAPK/ERK pathway. Furthermore, PPVII exerted strong antitumor effects in vivo in nude mice injected with breast cancer cells. Our results suggest that PPVII may promote apoptosis through regulating the SOS1/MAPK/ERK pathway, making it a possible candidate target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Shu-Guang Du
- Chronic Disease Research Center, Medical College, Dalian University, Xuefu Road 10, Dalian 116622, P. R. China
- Laboratory Department, The First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin 150040, P. R. China
| | - Hua-Min Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Xuefu Road 10, Dalian 116622, P. R. China
- Key Laboratory of Natural Resources of Changbai Mountain & Functional Molecules, Ministry of Education, Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji 133002, P. R. China
| | - Yun-Xia Ji
- Laboratory Department, Zhangjiakou First Hospital, Zhangjiakou 075041, P. R. China
| | - Yu-Lin Tian
- Chronic Disease Research Center, Medical College, Dalian University, Xuefu Road 10, Dalian 116622, P. R. China
| | - Dan Wang
- Chronic Disease Research Center, Medical College, Dalian University, Xuefu Road 10, Dalian 116622, P. R. China
| | - Kun Zhu
- Chronic Disease Research Center, Medical College, Dalian University, Xuefu Road 10, Dalian 116622, P. R. China
| | - Qing-Gao Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Xuefu Road 10, Dalian 116622, P. R. China
| | - Shuang-Ping Liu
- Chronic Disease Research Center, Medical College, Dalian University, Xuefu Road 10, Dalian 116622, P. R. China
| |
Collapse
|
4
|
Chen Y, Liu QP, Xie H, Ding J. From bench to bedside: current development and emerging trend of KRAS-targeted therapy. Acta Pharmacol Sin 2024; 45:686-703. [PMID: 38049578 PMCID: PMC10943119 DOI: 10.1038/s41401-023-01194-4] [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: 07/23/2023] [Accepted: 11/09/2023] [Indexed: 12/06/2023] Open
Abstract
Kirsten rat sarcoma 2 viral oncogene homolog (KRAS) is the most frequently mutated oncogene in human cancers with mutations predominantly occurring in codon 12. These mutations disrupt the normal function of KRAS by interfering with GTP hydrolysis and nucleotide exchange activity, making it prone to the GTP-bound active state, thus leading to sustained activation of downstream pathways. Despite decades of research, there has been no progress in the KRAS drug discovery until the groundbreaking discovery of covalently targeting the KRASG12C mutation in 2013, which led to revolutionary changes in KRAS-targeted therapy. So far, two small molecule inhibitors sotorasib and adagrasib targeting KRASG12C have received accelerated approval for the treatment of non-small cell lung cancer (NSCLC) harboring KRASG12C mutations. In recent years, rapid progress has been achieved in the KRAS-targeted therapy field, especially the exploration of KRASG12C covalent inhibitors in other KRASG12C-positive malignancies, novel KRAS inhibitors beyond KRASG12C mutation or pan-KRAS inhibitors, and approaches to indirectly targeting KRAS. In this review, we provide a comprehensive overview of the molecular and mutational characteristics of KRAS and summarize the development and current status of covalent inhibitors targeting the KRASG12C mutation. We also discuss emerging promising KRAS-targeted therapeutic strategies, with a focus on mutation-specific and direct pan-KRAS inhibitors and indirect KRAS inhibitors through targeting the RAS activation-associated proteins Src homology-2 domain-containing phosphatase 2 (SHP2) and son of sevenless homolog 1 (SOS1), and shed light on current challenges and opportunities for drug discovery in this field.
Collapse
Affiliation(s)
- Yi Chen
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiu-Pei Liu
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Department of Chemical and Environment Engineering, Science and Engineering Building, The University of Nottingham Ningbo China, Ningbo, 315100, China
| | - Hua Xie
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - Jian Ding
- Division of Antitumor Pharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
5
|
Wu J, Li X, Wu C, Wang Y, Zhang J. Current advances and development strategies of targeting son of sevenless 1 (SOS1) in drug discovery. Eur J Med Chem 2024; 268:116282. [PMID: 38430853 DOI: 10.1016/j.ejmech.2024.116282] [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: 01/19/2024] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/05/2024]
Abstract
The Son of Sevenless 1 (SOS1) guanine nucleotide exchange factor, prevalent across eukaryotic species, plays a pivotal role in facilitating the attachment of RAS protein to GTP, thereby regulating the activation of intracellular RAS proteins. This regulation is part of a feedback mechanism involving SOS1, which allows both activators and inhibitors of SOS1 to exert control over downstream signaling pathways, demonstrating potential anti-tumor effects. Predominantly, small molecule modulators that target SOS1 focus on a hydrophobic pocket within the CDC25 protein domain. The effectiveness of these modulators largely depends on their ability to interact with specific amino acids, notably Phe890 and Tyr884. This interaction is crucial for influencing the protein-protein interaction (PPI) between RAS and the catalytic domain of SOS1. Currently, most small molecule modulators targeting SOS1 are in the preclinical research phase, with a few advancing to clinical trials. This progression raises safety concerns, making the assurance of drug safety a primary consideration alongside the enhancement of efficacy in the development of SOS1 modulators. This review encapsulates recent advancements in the chemical categorization of SOS1 inhibitors and activators. It delves into the evolution of small molecule modulation targeting SOS1 and offers perspectives on the design of future generations of selective SOS1 small molecule modulators.
Collapse
Affiliation(s)
- Jialin Wu
- Department of Neurology, Neuro-system and Multimorbidity Laboratory and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiaoxue Li
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Chengyong Wu
- Department of Neurology, Neuro-system and Multimorbidity Laboratory and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China; Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuxi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Jifa Zhang
- Department of Neurology, Neuro-system and Multimorbidity Laboratory and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
6
|
Pang X, Cui D, Lv B, Wang CY. Discovery of Potent SOS1 PROTACs with Effective Antitumor Activities against NCI-H358 Tumor Cells In Vitro/In Vivo. J Med Chem 2024; 67:1563-1579. [PMID: 38206836 DOI: 10.1021/acs.jmedchem.3c02135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2024]
Abstract
Directly targeted KRAS inhibitors are now facing resistance problems, which might be partially solved by the combination of SOS1 inhibitors with KRAS inhibitors. However, this combination may still have some resistance mitigation potential. Comparatively, SOS1 PROTAC may have promising applications in addressing the drug resistance problem by degrading the SOS1 protein. Herein, we report the discovery of novel SOS1 PROTACs and their antitumor activity both in vitro and in vivo. In vitro studies demonstrated that degrader 4 had strong inhibitory effects on the proliferation of NCI-H358 cells with IC50 of 5 nM, together with significant degradation of SOS1 protein with DC50 of 13 nM. In the NCI-H358 xenograft model, degrader 4 exhibited significant antitumor activities with TGITV values of 58.8% at 30 mg/kg bid. The PK and safety profiles also supported degrader 4 for further studies as an effective tool compound.
Collapse
Affiliation(s)
- Xudong Pang
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Zelgen Pharma-Tech Co., Ltd., Building 3, No. 999, Cailun Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Dawei Cui
- Shanghai Zelgen Pharma-Tech Co., Ltd., Building 3, No. 999, Cailun Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Binhua Lv
- Shanghai Zelgen Pharma-Tech Co., Ltd., Building 3, No. 999, Cailun Road, Zhangjiang Hi-Tech Park, Shanghai 201203, China
| | - Cheng-Yun Wang
- Key Laboratory for Advanced Materials and Institute of Fine Chemicals, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
7
|
Chen T, Tang X, Wang Z, Feng F, Xu C, Zhao Q, Wu Y, Sun H, Chen Y. Inhibition of Son of Sevenless Homologue 1 (SOS1): Promising therapeutic treatment for KRAS-mutant cancers. Eur J Med Chem 2023; 261:115828. [PMID: 37778239 DOI: 10.1016/j.ejmech.2023.115828] [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: 07/04/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/03/2023]
Abstract
Kristen rat sarcoma (KRAS) is one of the most common oncogenes in human cancers. As a guanine nucleotide exchange factor, Son of Sevenless Homologue 1 (SOS1) represents a potential therapeutic concept for the treatment of KRAS-mutant cancers because of its activation on KRAS and downstream signaling pathways. In this review, we provide a comprehensive overview of the structure, biological function, and regulation of SOS1. We also focus on the recent advances in SOS1 inhibitors and emphasize their binding modes, structure-activity relationships and pharmacological activities. We hope that this publication can provide a comprehensive compendium on the rational design of SOS1 inhibitors.
Collapse
Affiliation(s)
- Tingkai Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Xu Tang
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China
| | - Zhenqi Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Feng Feng
- School of Pharmacy, Nanjing Medical University, 211166, Nanjing, People's Republic of China
| | - Chunlei Xu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Qun Zhao
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Yulan Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, People's Republic of China.
| | - Yao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
8
|
Baltanás FC, García-Navas R, Rodríguez-Ramos P, Calzada N, Cuesta C, Borrajo J, Fuentes-Mateos R, Olarte-San Juan A, Vidaña N, Castellano E, Santos E. Critical requirement of SOS1 for tumor development and microenvironment modulation in KRAS G12D-driven lung adenocarcinoma. Nat Commun 2023; 14:5856. [PMID: 37730692 PMCID: PMC10511506 DOI: 10.1038/s41467-023-41583-1] [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: 02/21/2023] [Accepted: 09/11/2023] [Indexed: 09/22/2023] Open
Abstract
The impact of genetic ablation of SOS1 or SOS2 is evaluated in a murine model of KRASG12D-driven lung adenocarcinoma (LUAD). SOS2 ablation shows some protection during early stages but only SOS1 ablation causes significant, specific long term increase of survival/lifespan of the KRASG12D mice associated to markedly reduced tumor burden and reduced populations of cancer-associated fibroblasts, macrophages and T-lymphocytes in the lung tumor microenvironment (TME). SOS1 ablation also causes specific shrinkage and regression of LUAD tumoral masses and components of the TME in pre-established KRASG12D LUAD tumors. The critical requirement of SOS1 for KRASG12D-driven LUAD is further confirmed by means of intravenous tail injection of KRASG12D tumor cells into SOS1KO/KRASWT mice, or of SOS1-less, KRASG12D tumor cells into wildtype mice. In silico analyses of human lung cancer databases support also the dominant role of SOS1 regarding tumor development and survival in LUAD patients. Our data indicate that SOS1 is critically required for development of KRASG12D-driven LUAD and confirm the validity of this RAS-GEF activator as an actionable therapeutic target in KRAS mutant LUAD.
Collapse
Affiliation(s)
- Fernando C Baltanás
- Lab 1. Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, CSIC-University of Salamanca and CIBERONC, 37007, Salamanca, Spain.
- Institute of Biomedicine of Seville (IBiS)/"Virgen del Rocío" University Hospital/CSIC/University of Seville and Department of Medical Physiology and Biophysics, University of Seville, Seville, Spain.
| | - Rósula García-Navas
- Lab 1. Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, CSIC-University of Salamanca and CIBERONC, 37007, Salamanca, Spain
| | - Pablo Rodríguez-Ramos
- Lab 1. Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, CSIC-University of Salamanca and CIBERONC, 37007, Salamanca, Spain
| | - Nuria Calzada
- Lab 1. Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, CSIC-University of Salamanca and CIBERONC, 37007, Salamanca, Spain
| | - Cristina Cuesta
- Lab 5. Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, CSIC-University of Salamanca, 37007, Salamanca, Spain
| | - Javier Borrajo
- Departament of Biomedical Sciences and Diagnostic, University of Salamanca, 37007, Salamanca, Spain
| | - Rocío Fuentes-Mateos
- Lab 1. Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, CSIC-University of Salamanca and CIBERONC, 37007, Salamanca, Spain
| | - Andrea Olarte-San Juan
- Lab 1. Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, CSIC-University of Salamanca and CIBERONC, 37007, Salamanca, Spain
| | - Nerea Vidaña
- Lab 1. Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, CSIC-University of Salamanca and CIBERONC, 37007, Salamanca, Spain
| | - Esther Castellano
- Lab 5. Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, CSIC-University of Salamanca, 37007, Salamanca, Spain
| | - Eugenio Santos
- Lab 1. Cancer Research Center, Institute of Cancer Molecular and Cellular Biology, CSIC-University of Salamanca and CIBERONC, 37007, Salamanca, Spain.
| |
Collapse
|
9
|
Pagba CV, Gupta AK, Gorfe AA. Small-Molecule Inhibition of KRAS through Conformational Selection. ACS OMEGA 2023; 8:31419-31426. [PMID: 37663463 PMCID: PMC10468774 DOI: 10.1021/acsomega.3c04013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/09/2023] [Indexed: 09/05/2023]
Abstract
Mutations in KRAS account for about 20% of human cancers. Despite the major progress in recent years toward the development of KRAS inhibitors, including the discovery of covalent inhibitors of the G12C KRAS variant for the treatment of non-small-cell lung cancer, much work remains to be done to discover broad-acting inhibitors to treat many other KRAS-driven cancers. In a previous report, we showed that a 308.4 Da small-molecule ligand [(2R)-2-(N'-(1H-indole-3-carbonyl)hydrazino)-2-phenyl-acetamide] binds to KRAS with low micro-molar affinity [Chem. Biol. Drug Des.2019; 94(2):1441-1456]. Binding of this ligand, which we call ACA22, to the p1 pocket of KRAS and its interactions with residues at beta-strand 1 and the switch loops have been supported by data from nuclear magnetic resonance spectroscopy and microscale thermophoresis experiments. However, the inhibitory potential of the compound was not demonstrated. Here, we show that ACA22 inhibits KRAS-mediated signal transduction in cells expressing wild type (WT) and G12D mutant KRAS and reduces levels of guanosine triphosphate-loaded WT KRAS more effectively than G12D KRAS. We ruled out the direct effect on nucleotide exchange or effector binding as possible mechanisms of inhibition using a variety of biophysical assays. Combining these observations with binding data that show comparable affinities of the compound for the active and inactive forms of the mutant but not the WT, we propose conformational selection as a possible mechanism of action of ACA22.
Collapse
Affiliation(s)
- Cynthia V Pagba
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, Texas 77030, United States
| | - Amit K Gupta
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, Texas 77030, United States
| | - Alemayehu A Gorfe
- Department of Integrative Biology and Pharmacology, McGovern Medical School, University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, Texas 77030, United States
| |
Collapse
|
10
|
Liao Y, Tao S, Wang S, Wu G, Yao W, Yang L, Huang Q, Liu Y, Yang G, Yang P. 5-HT modulates the properties of dendritic cells to interfere with the development of type 1 regulating T cells. Mol Immunol 2023; 160:161-167. [PMID: 37451234 DOI: 10.1016/j.molimm.2023.07.001] [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: 06/23/2022] [Revised: 06/23/2023] [Accepted: 07/02/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND 5-hydroxytryptamine (5-HT, serotonin) is a major mediator in allergic reactions. The number of tolerogenic dendritic cell (tolDC) and regulatory T cell is reduced in allergic disorders. The mechanism is unclear. The objective of this study is to elucidate the role of 5-HT in interfering with tolDC generation and regulatory Type 1 T cell (Tr1 cell). METHODS BALB/c mice were treated with 5-HT-containing nasal instillations. The frequency of tolDC and Tr1 cell was evaluated by flow cytometry. RESULTS Following treatment with 5-HT nasal instillations for one week, the frequency of tolDC and Tr1 cell was significantly reduced in the respiratory tissues. Higher levels of SOS1 were detected in DCs isolated from the airway tissues of mice treated with 5-HT. A complex of SOS1 and c-Maf was detected in DCs in response to 5-HT stimulation. The expression of IL-10 was suppressed by the presence of 5-HT. The induction of Tr1 cell by DC was substantially compromised by 5-HT. CONCLUSIONS 5-HT inhibits the expression of IL-10 in DCs. DCs primed with 5-HT lose the ability to induce Tr1 cells.
Collapse
Affiliation(s)
- Yun Liao
- Departments of Otolaryngology & Allergy. Longgang Central Hospital. Shenzhen. China; Shenzhen Clinical College, Guangzhou Chinese Traditional Medical University, Shenzhen, China; Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China
| | - Shuang Tao
- Shenzhen Clinical College, Guangzhou Chinese Traditional Medical University, Shenzhen, China; Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China
| | - Shiqi Wang
- Shenzhen Clinical College, Guangzhou Chinese Traditional Medical University, Shenzhen, China; Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China
| | - Gaohui Wu
- Departments of General Practice Medicine & Allergy. Third Affiliated Hospital of Shenzhen University. Shenzhen. China
| | - Wenkai Yao
- Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China
| | - Liteng Yang
- Departments of General Practice Medicine & Allergy. Third Affiliated Hospital of Shenzhen University. Shenzhen. China
| | - Qinmiao Huang
- Departments of General Practice Medicine & Allergy. Third Affiliated Hospital of Shenzhen University. Shenzhen. China
| | - Yu Liu
- Departments of General Practice Medicine & Allergy. Third Affiliated Hospital of Shenzhen University. Shenzhen. China.
| | - Gui Yang
- Departments of Otolaryngology & Allergy. Longgang Central Hospital. Shenzhen. China.
| | - Pingchang Yang
- Institute of Allergy & Immunology of Shenzhen University, State Key Laboratory of Respiratory Diseases Allergy Division at Shenzhen University, Shenzhen, China.
| |
Collapse
|
11
|
He H, Chen R, Wang Z, Qing L, Zhang Y, Liu Y, Pan W, Fang H, Zhang S. Design of Orally-bioavailable Tetra-cyclic phthalazine SOS1 inhibitors with high selectivity against EGFR. Bioorg Chem 2023; 136:106536. [PMID: 37054529 DOI: 10.1016/j.bioorg.2023.106536] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/28/2023] [Accepted: 04/07/2023] [Indexed: 04/15/2023]
Abstract
KRAS mutations (G12C, G12D, etc.) are implicated in the oncogenesis and progression of many deadliest cancers. Son of sevenless homolog 1 (SOS1) is a crucial regulator of KRAS to modulate KRAS from inactive to active states. We previously discovered tetra-cyclic quinazolines as an improved scaffold for inhibiting SOS1-KRAS interaction. In this work, we report the design of tetra-cyclic phthalazine derivatives for selectively inhibiting SOS1 against EGFR. The lead compound 6c displayed remarkable activity to inhibit the proliferation of KRAS(G12C)-mutant pancreas cells. 6c showed a favorable pharmacokinetic profile in vivo, with a bioavailability of 65.8% and exhibited potent tumor suppression in pancreas tumor xenograft models. These intriguing results suggested that 6c has the potential to be developed as a drug candidate for KRAS-driven tumors.
Collapse
Affiliation(s)
- Huan He
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China; Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, PR China; Wuhan Yuxiang Pharmaceutical Technology Co., Ltd., Wuhan 430200, PR China
| | - Ruiqi Chen
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, PR China
| | - Ziwei Wang
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, PR China
| | - Luolong Qing
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, PR China
| | - Yu Zhang
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, PR China
| | - Yi Liu
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, PR China; School of Chemical and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, PR China; Hubei Key Laboratory of Radiation Chemistry and Functional Materials, Hubei University of Science and Technology, Xianning 437100, PR China
| | - Weidong Pan
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China.
| | - Huaxiang Fang
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, PR China.
| | - Silong Zhang
- School of Pharmaceutical Sciences, Guizhou University, Guiyang 550025, PR China; Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, College of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, PR China; Wuhan Yuxiang Pharmaceutical Technology Co., Ltd., Wuhan 430200, PR China.
| |
Collapse
|
12
|
Luo G, Wang B, Hou Q, Wu X. Development of Son of Sevenless Homologue 1 (SOS1) Modulators To Treat Cancers by Regulating RAS Signaling. J Med Chem 2023; 66:4324-4341. [PMID: 36987571 DOI: 10.1021/acs.jmedchem.2c01729] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Son of sevenless homologue 1 (SOS1) protein is universally expressed in cells and plays an important role in the RAS signaling pathway. Specifically, this protein interacts with RAS in response to upstream stimuli to promote guanine nucleotide exchange in RAS and activates the downstream signaling pathways. Thus, targeting SOS1 is a new approach for treating RAS-driven cancers. In this Perspective, we briefly summarize the structural and functional aspects of SOS1 and focus on recent advances in the discovery of activators, inhibitors, and PROTACs that target SOS1. This review aims to provide a timely and updated overview on the strategies for targeting SOS1 in cancer therapy.
Collapse
Affiliation(s)
- Guangmei Luo
- Department of Medicinal Chemistry, School of Pharmacy and Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Bingrui Wang
- Department of Medicinal Chemistry, School of Pharmacy and Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Qiangqiang Hou
- Department of Medicinal Chemistry, School of Pharmacy and Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Xiaoxing Wu
- Department of Medicinal Chemistry, School of Pharmacy and Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
13
|
De Vos N, Hofmans M, Lammens T, De Wilde B, Van Roy N, De Moerloose B. Targeted therapy in juvenile myelomonocytic leukemia: Where are we now? Pediatr Blood Cancer 2022; 69:e29930. [PMID: 36094370 DOI: 10.1002/pbc.29930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/07/2022]
Abstract
Juvenile myelomonocytic leukemia (JMML) is a rare and aggressive clonal neoplasm of early childhood, classified as an overlap myeloproliferative/myelodysplastic neoplasm by the World Health Organization. In 90% of the patients with JMML, typical initiating mutations in the canonical Ras pathway genes NF1, PTPN11, NRAS, KRAS, and CBL can be identified. Hematopoietic stem cell transplantation (HSCT) currently is the established standard of care in most patients, although long-term survival is still only 50-60%. Given the limited therapeutic options and the important morbidity and mortality associated with HSCT, new therapeutic approaches are urgently needed. Hyperactivation of the Ras pathway as disease mechanism in JMML lends itself to the use of targeted therapy. Targeted therapy could play an important role in the future treatment of patients with JMML. This review presents a comprehensive overview of targeted therapies already developed and evaluated in vitro and in vivo in patients with JMML.
Collapse
Affiliation(s)
- Nele De Vos
- Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University, Ghent, Belgium
| | - Mattias Hofmans
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium.,Cancer Research Institute Ghent, Ghent, Belgium
| | - Tim Lammens
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Bram De Wilde
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| | - Nadine Van Roy
- Cancer Research Institute Ghent, Ghent, Belgium.,Center for Medical Genetics Ghent, Ghent, Belgium.,Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Barbara De Moerloose
- Cancer Research Institute Ghent, Ghent, Belgium.,Department of Pediatric Hematology-Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, Belgium.,Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
| |
Collapse
|
14
|
Icard P, Simula L, Fournel L, Leroy K, Lupo A, Damotte D, Charpentier MC, Durdux C, Loi M, Schussler O, Chassagnon G, Coquerel A, Lincet H, De Pauw V, Alifano M. The strategic roles of four enzymes in the interconnection between metabolism and oncogene activation in non-small cell lung cancer: Therapeutic implications. Drug Resist Updat 2022; 63:100852. [PMID: 35849943 DOI: 10.1016/j.drup.2022.100852] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
NSCLC is the leading cause of cancer mortality and represents a major challenge in cancer therapy. Intrinsic and acquired anticancer drug resistance are promoted by hypoxia and HIF-1α. Moreover, chemoresistance is sustained by the activation of key signaling pathways (such as RAS and its well-known downstream targets PI3K/AKT and MAPK) and several mutated oncogenes (including KRAS and EGFR among others). In this review, we highlight how these oncogenic factors are interconnected with cell metabolism (aerobic glycolysis, glutaminolysis and lipid synthesis). Also, we stress the key role of four metabolic enzymes (PFK1, dimeric-PKM2, GLS1 and ACLY), which promote the activation of these oncogenic pathways in a positive feedback loop. These four tenors orchestrating the coordination of metabolism and oncogenic pathways could be key druggable targets for specific inhibition. Since PFK1 appears as the first tenor of this orchestra, its inhibition (and/or that of its main activator PFK2/PFKFB3) could be an efficacious strategy against NSCLC. Citrate is a potent physiologic inhibitor of both PFK1 and PFKFB3, and NSCLC cells seem to maintain a low citrate level to sustain aerobic glycolysis and the PFK1/PI3K/EGFR axis. Awaiting the development of specific non-toxic inhibitors of PFK1 and PFK2/PFKFB3, we propose to test strategies increasing citrate levels in NSCLC tumors to disrupt this interconnection. This could be attempted by evaluating inhibitors of the citrate-consuming enzyme ACLY and/or by direct administration of citrate at high doses. In preclinical models, this "citrate strategy" efficiently inhibits PFK1/PFK2, HIF-1α, and IGFR/PI3K/AKT axes. It also blocks tumor growth in RAS-driven lung cancer models, reversing dedifferentiation, promoting T lymphocytes tumor infiltration, and increasing sensitivity to cytotoxic drugs.
Collapse
Affiliation(s)
- Philippe Icard
- Thoracic Surgery Department, Paris Center University Hospitals, AP-HP, Paris, France; Normandie Univ, UNICAEN, CHU de Caen Normandie, Unité de recherche BioTICLA INSERM U1086, 14000 Caen, France.
| | - Luca Simula
- Department of Infection, Immunity and Inflammation, Cochin Institute, INSERM U1016, CNRS UMR8104, Paris University, Paris 75014, France
| | - Ludovic Fournel
- Thoracic Surgery Department, Paris Center University Hospitals, AP-HP, Paris, France; INSERM UMR-S 1124, Cellular Homeostasis and Cancer, University of Paris, Paris, France
| | - Karen Leroy
- Department of Genomic Medicine and Cancers, Georges Pompidou European Hospital, APHP, Paris, France
| | - Audrey Lupo
- Pathology Department, Paris Center University Hospitals, AP-HP, Paris, France; INSERM U1138, Integrative Cancer Immunology, University of Paris, 75006 Paris, France
| | - Diane Damotte
- Pathology Department, Paris Center University Hospitals, AP-HP, Paris, France; INSERM U1138, Integrative Cancer Immunology, University of Paris, 75006 Paris, France
| | | | - Catherine Durdux
- Radiation Oncology Department, Georges Pompidou European Hospital, APHP, Paris, France
| | - Mauro Loi
- Radiotherapy Department, University of Florence, Florence, Italy
| | - Olivier Schussler
- Thoracic Surgery Department, Paris Center University Hospitals, AP-HP, Paris, France
| | | | - Antoine Coquerel
- INSERM U1075, COMETE " Mobilités: Attention, Orientation, Chronobiologie", Université Caen, France
| | - Hubert Lincet
- ISPB, Faculté de Pharmacie, Lyon, France, Université Lyon 1, Lyon, France; INSERM U1052, CNRS UMR5286, Cancer Research Center of Lyon (CRCL), France
| | - Vincent De Pauw
- Thoracic Surgery Department, Paris Center University Hospitals, AP-HP, Paris, France
| | - Marco Alifano
- Thoracic Surgery Department, Paris Center University Hospitals, AP-HP, Paris, France; INSERM U1138, Integrative Cancer Immunology, University of Paris, 75006 Paris, France
| |
Collapse
|
15
|
Abstract
RAS proteins play major roles in many human cancers, but programs to develop direct RAS inhibitors so far have only been successful for the oncogenic KRAS mutant G12C. As an alternative approach, inhibitors for the RAS guanine nucleotide exchange factor SOS1 have been investigated by several academic groups and companies, and major progress has been achieved in recent years in the optimization of small molecule activators and inhibitors of SOS1. Here, we review the discovery and development of small molecule modulators of SOS1 and their molecular binding modes and modes of action. As targeting the RAS pathway is expected to result in the development of resistance mechanisms, SOS1 inhibitors will most likely be best applied in vertical combination approaches where two nodes of the RAS signaling pathway are hit simultaneously. We summarize the current understanding of which combination partners may be most beneficial for patients with RAS driven tumors.
Collapse
Affiliation(s)
| | - Benjamin Bader
- Screening, Lead Discovery, Nuvisan ICB GmbH, Berlin, Germany
| |
Collapse
|