1
|
Tian H, Zeng W, Wang Z, Li S, Wei W, Li S, Yin X, Na W, Wang Y, Song K, Zhu P, Liang W. P-Pev: micelle-like complexes transformed from tumor extracellular vesicles by PEG-PE for personalized therapeutic tumor vaccine. Biomaterials 2025; 321:123333. [PMID: 40239595 DOI: 10.1016/j.biomaterials.2025.123333] [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: 11/06/2024] [Revised: 02/18/2025] [Accepted: 04/07/2025] [Indexed: 04/18/2025]
Abstract
The clinical benefits of personalized therapeutic tumor vaccines are mainly challenged by the need to identify immunogenic neoantigens promptly, given the rapid pace of tumor mutations. An increasing body of literature addresses the potential of tumor-derived extracellular vesicles (TEVs) as an anti-tumor "cell-free" vaccine due to their substantial presence of neoantigens. However, their immunosuppression and limited presentation efficiency of dendritic cells (DCs) restrict their further application. Here, we have developed a novel tumor-personalized vaccine, termed P-Pev, based on remodeled TEVs by polymeric surfactant polyethylene glycol-phosphatidyleolamine (PEG-PE) and adjuvant monophosphoryl lipid A (MPLA). Our results show that PEG-PE transforms TEVs into micelle-like complexes by disrupting the original structure, facilitating antigens delivery to the cytoplasm, and cross-presentation by DCs. P-Pev particularly prevents the immunosuppressive impacts of TEVs on the ability of DCs to prime CD8+ T cells and eliminates the potency of TEVs to promote lung metastasis through their membrane-bound PD-L1. Finally, the P-Pev effectively induces neoantigen-specific cytotoxic T lymphocytes (CTLs) responses and exhibits excellent therapeutic effects in various murine tumor models. Also, the P-Pev induces neoantigen-specific antibodies, suggesting the involvement of humoral immunity in its anti-tumor effects. More importantly, it has been shown that P-Pev prepared by mutated tumor cells can retard these mutated tumor cell-established syngeneic tumors better than P-Pev prepared by original tumor cells, indicating the feasibility that leverages TEVs to prepare personalized tumor vaccines, and it is synergistically enhanced by PD-1 mAb combination. Collectively, we present a general strategy that offers a streamlined, cost-effective, and time-consuming approach to preparing personalized therapeutic tumor vaccines.
Collapse
Affiliation(s)
- Hongjian Tian
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Wenfeng Zeng
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Zihao Wang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Siqi Li
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Wenjing Wei
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Shanshan Li
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100864, China
| | - Xiaozhe Yin
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, 100084, China
| | - Wenjing Na
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, TX, 78229, USA
| | - Youwang Wang
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Kai Song
- Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ping Zhu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100864, China; Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Wei Liang
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100864, China.
| |
Collapse
|
2
|
Li L, Liu Q, Shao Y, Wang S, Liu S, Wang X, Wang S, Ren D. Gaudichaudion H inhibits KRAS-mutant pancreatic cancer cell growth through interfering PDEδ-KRAS interaction. Chem Biol Interact 2025; 415:111529. [PMID: 40288433 DOI: 10.1016/j.cbi.2025.111529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 04/03/2025] [Accepted: 04/25/2025] [Indexed: 04/29/2025]
Abstract
KRAS mutation results in higher proliferation rates and miserable prognosis of cancers. Targeting the interaction between KRAS and PDE6D provided an alternative strategy to overcome KRAS-mutant pancreatic cancers. Gaudichaudione H (GH) is a prenylated caged xanthone isolated from Garcinia oligantha. In this work, GH was selected as a potential anti-cancer compound by MTT screening of twelve prenylated xanthonoids from G. oligantha. Further studies demonstrated that GH inhibited proliferation of a panel of cancer cell lines and induced pancreatic cancer cell apoptosis. GH suppressed xenograft tumor growth accompanied with decreased phosphorylation of ERK and AKT. Binding with PDEδ and thus interfering the KRAS-PDEδ interaction was verified as the possible mechanism of GH. These findings implicated GH as a promising candidate for the treatment of pancreatic cancers with KRAS mutation, provided novel insight into the underlying mechanisms of GH-induced anticancer effects.
Collapse
Affiliation(s)
- Lingyu Li
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, PR China
| | - Qingying Liu
- School of Pharmaceutical Sciences, Shandong Xiandai University, PR China
| | - Yuyu Shao
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, PR China
| | - Shuo Wang
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, PR China
| | - Shuangyu Liu
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, PR China
| | - Xiaoning Wang
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, PR China
| | - Shuqi Wang
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, PR China
| | - Dongmei Ren
- Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 44 West Wenhua Road, Jinan, 250012, PR China.
| |
Collapse
|
3
|
Wu J, Jiang S, Shen Q, Gong H. Postoperative metastatic Krukenberg tumors with ARID1A and KRAS mutations in a patient with gastric cancer treated with oxaliplatin and tegafur: A case report. Oncol Lett 2025; 29:262. [PMID: 40230423 PMCID: PMC11995681 DOI: 10.3892/ol.2025.15008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 03/14/2025] [Indexed: 04/16/2025] Open
Abstract
Krukenberg tumors are a notably rare type of metastatic ovarian malignant tumor, often originating from the stomach. Due to their low incidence rate and the short survival time of patients, there is currently a lack of consensus on the diagnosis and treatment of this disease, as well as a deficiency in genomic analyses and research into the pathogenetic molecular mechanisms. In the present study, the case of a patient with gastric cancer who, 2 years after curative surgery and chemotherapy with oxaliplatin and tegafur, developed recurrent metastatic bilateral Krukenberg tumors with distant metastasis in the ovaries. During treatment, a total hysterectomy and bilateral salpingo-oophorectomy were performed, and intraoperative intraperitoneal chemotherapy with cisplatin (70 mg) was administered. Additionally, ureteroscopy and bilateral ureteral stent placement were conducted transurethrally. Post-surgery, assessments of the genomic alterations and microsatellite instability of the tumor revealed an AT-rich interaction domain 1A (ARID1A) exon c.4720delC mutation and a KRAS exon c.35G>C mutation. The potential pathogenic mechanisms and clinical significance of these mutations were then further discussed. Mutations in the ARID1A gene could increase the sensitivity of the patient to immune checkpoint inhibitor therapy. Additionally, the successful application of KRASG12C inhibitors in other cancer types offers a new approach for the targeted therapy of Krukenberg tumors. Therefore, the present study provides further evidence regarding the genomics of Krukenberg tumors, which may aid in the development of targeted treatment strategies.
Collapse
Affiliation(s)
- Jie Wu
- Department of Gynecology, Dongguan Songshan Lake Tungwah Hospital, Dongguan, Guangdong 523000, P.R. China
| | - Suzhen Jiang
- Department of Gynecology, Dongguan Songshan Lake Tungwah Hospital, Dongguan, Guangdong 523000, P.R. China
| | - Qingling Shen
- Department of Gynecology, Dongguan Songshan Lake Tungwah Hospital, Dongguan, Guangdong 523000, P.R. China
| | - Hongxia Gong
- Department of Gynecology, Dongguan Tungwah Hospital, Dongguan, Guangdong 523000, P.R. China
| |
Collapse
|
4
|
Hwang SY, Seo Y, Park S, Kim SA, Moon I, Liu Y, Kim S, Pak ES, Jung S, Kim H, Jeon KH, Seo SH, Sung I, Lee H, Park SY, Na Y, Kim TI, Kwon Y. Targeting the HER2-ELF3-KRAS axis: a novel therapeutic strategy for KRAS G13D colorectal cancer. Mol Cancer 2025; 24:139. [PMID: 40340861 PMCID: PMC12063335 DOI: 10.1186/s12943-025-02343-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 04/25/2025] [Indexed: 05/10/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent cancers worldwide, with KRAS mutations playing a significant role in its tumorigenesis. Among the KRAS variants, the G13D mutation is associated with poor prognosis and distinctive biological behaviors. This study focuses on the role of HER2, a critical prognostic and predictive biomarker, in modulating the unique characteristics of KRASG13D-mutated CRCs. We identified a novel transcriptional regulatory network involving HER2, ELF3, and KRAS, with ELF3 acting as a key transcription factor (TF) that regulates KRAS expression under conditions of HER2 overexpression. Our findings reveal that this HER2-ELF3-KRAS axis is exclusively activated in KRASG13D, driving aggressive oncogenic features and conferring resistance to cetuximab (CTX) therapy. Through comprehensive analysis of gene expression profiles, we demonstrated that HER2 is a crucial therapeutic target specifically for KRASG13D CRCs. To explore this further, we introduced YK1, a small molecule inhibitor designed to disrupt the ELF3-MED23 interaction, leading to the transcriptional downregulation of HER2 and KRAS. This intervention significantly attenuated the HER2-ELF3-KRAS axis, sensitizing KRASG13D CRCs to CTX and reducing their tumorigenic potential by inhibiting the epithelial-to-mesenchymal transition process. Our study underscores the importance of HER2 as a key determinant in the unique biological characteristics of KRASG13D CRCs and highlights the therapeutic potential of targeting the HER2-ELF3-KRAS axis. By presenting YK1 as a novel pharmacological approach, we provide a promising strategy for developing tailored interventions for KRASG13D CRCs, contributing to the ongoing efforts in precision medicine for CRCs.
Collapse
Affiliation(s)
- Soo-Yeon Hwang
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Yoojeong Seo
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, 50 Yonseiro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Seojeong Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seul-Ah Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
- Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Inhye Moon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
- Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Yi Liu
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seojeong Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Eun Seon Pak
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Sehyun Jung
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Hyeyoon Kim
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Kyung-Hwa Jeon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Seung Hee Seo
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Inyoung Sung
- BK21 FOUR Intelligence Computing, Seoul National University, Seoul, Republic of Korea
| | - Heetak Lee
- Center for Genome Engineering, Institute for Basic Science, 55, Expo-Ro, Yuseong-Gu, Daejeon, 34126, Republic of Korea
| | - So-Yeon Park
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Younghwa Na
- College of Pharmacy, CHA University, Pocheon, 11160, Republic of Korea
| | - Tae Il Kim
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, 50 Yonseiro, Seodaemun-Gu, Seoul, 03722, Republic of Korea
| | - Youngjoo Kwon
- College of Pharmacy, Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul, 03760, Republic of Korea.
- Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, 03760, Republic of Korea.
| |
Collapse
|
5
|
Ji X, Li H, Wu G, Zhang Q, He X, Wu Y, Zong B, Xu X, Liang C, Wang B, Zhang Y, Hu Q, Deng C, Shen L, Chen Z, Bai B, Wang L, Ai J, Zhang L, Zhou H, Sun S, Wang Y, Wang Y, Fan Q, Chen D, Zhou T, Kong X, Lu J. Discovery and Characterization of RP03707: A Highly Potent and Selective KRAS G12D PROTAC. J Med Chem 2025. [PMID: 40338735 DOI: 10.1021/acs.jmedchem.5c00428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
KRASG12D, the most prevalent oncogenic mutation in KRAS-associated tumors, represents a highly sought-after drug target for cancer treatment. In this study, we explored a KRASG12D protein degradation approach using the PROTAC technology for the treatment of KRASG12D mutant tumors. Through the rational design of the KRASG12D binder and proper selection of the linker and the E3 ligase ligand, we constructed PROTACs and identified RP03707 as a CRBN-involving, highly potent, and selective KRASG12D degrader. RP03707 effectively inhibits tumor cell growth in multiple KRASG12D cell lines. It also exhibits prolonged PK/PD effects and excellent efficacy in mouse CDX models bearing KRASG12D tumors, highlighting its potential for the treatment of KRASG12D-driven tumors in clinical settings.
Collapse
Affiliation(s)
- Xiang Ji
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Huanping Li
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Gang Wu
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Qiguo Zhang
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Xiaolin He
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Yanpeng Wu
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Bin Zong
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Xiaojin Xu
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Chao Liang
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Beibei Wang
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Yuwei Zhang
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Qingyao Hu
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Chao Deng
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Liqiang Shen
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Zijun Chen
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Bing Bai
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Lin Wang
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Jinchao Ai
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Leduo Zhang
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Honggui Zhou
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Shihao Sun
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Yijie Wang
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Youhong Wang
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Qiming Fan
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Dawei Chen
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Tianlun Zhou
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Xianqi Kong
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
| | - Jiasheng Lu
- Risen (Shanghai) Pharma Tech Co., Ltd., Shanghai 201210, China
- School of Life Sciences, Fudan University, Shanghai 200437, China
| |
Collapse
|
6
|
Sirocchi LS, Scharnweber M, Oberndorfer S, Siszler G, Zak KM, Rumpel K, Neumüller RA, Wilding B. Discovery of Carbodiimide Warheads to Selectively and Covalently Target Aspartic Acid in KRAS G12D. J Am Chem Soc 2025; 147:15787-15795. [PMID: 40267480 DOI: 10.1021/jacs.5c03562] [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: 04/25/2025]
Abstract
Targeted covalent inhibitors are known to be successful therapeutics used in various indications. Covalent drugs typically target cysteine, as cysteine is well suited due to its high nucleophilicity. However, its low abundance in protein binding sites represents a major limitation. As a result, there is a need to covalently target additional nucleophilic amino acids. Recent literature has reported covalent inhibitors labeling aspartic acid in KRASG12D. However, these compounds also covalently bind to KRASG12C, indicating their cross-reactivity to cysteine along with aspartic acid. We report here carbodiimides as a novel reactive group to selectively target aspartic acid. Covalent inhibitors bearing a carbodiimide moiety are shown to covalently label KRASG12D in biochemical and cellular assays. A high-resolution X-ray crystal structure was obtained, which illustrates the mechanism of the covalent bond formation with KRASG12D. Carbodiimide warheads show selectivity toward KRASG12D over other KRAS alleles and represent a new covalent warhead suitable for covalently binding to aspartic acid in a biochemical and cellular context.
Collapse
Affiliation(s)
- Ludovica S Sirocchi
- Boehringer Ingelheim RCV GmbH & Co KG, Dr.-Boehringer-Gasse 5-11, Vienna A-1121, Austria
| | - Maximilian Scharnweber
- Boehringer Ingelheim RCV GmbH & Co KG, Dr.-Boehringer-Gasse 5-11, Vienna A-1121, Austria
| | - Sarah Oberndorfer
- Boehringer Ingelheim RCV GmbH & Co KG, Dr.-Boehringer-Gasse 5-11, Vienna A-1121, Austria
| | - Gabriella Siszler
- Boehringer Ingelheim RCV GmbH & Co KG, Dr.-Boehringer-Gasse 5-11, Vienna A-1121, Austria
| | - Krzysztof M Zak
- Boehringer Ingelheim RCV GmbH & Co KG, Dr.-Boehringer-Gasse 5-11, Vienna A-1121, Austria
| | - Klaus Rumpel
- Boehringer Ingelheim RCV GmbH & Co KG, Dr.-Boehringer-Gasse 5-11, Vienna A-1121, Austria
| | - Ralph A Neumüller
- Boehringer Ingelheim RCV GmbH & Co KG, Dr.-Boehringer-Gasse 5-11, Vienna A-1121, Austria
| | - Birgit Wilding
- Boehringer Ingelheim RCV GmbH & Co KG, Dr.-Boehringer-Gasse 5-11, Vienna A-1121, Austria
| |
Collapse
|
7
|
Krauß D, Moreno-Viedma V, Adachi-Fernandez E, de Sá Fernandes C, Genger JW, Fari O, Blauensteiner B, Kirchhofer D, Bradaric N, Gushchina V, Fotakis G, Mohr T, Abramovich I, Mor I, Holcmann M, Bergthaler A, Haschemi A, Trajanoski Z, Winkler J, Gottlieb E, Sibilia M. EGFR controls transcriptional and metabolic rewiring in KRAS G12D colorectal cancer. EMBO Mol Med 2025:10.1038/s44321-025-00240-4. [PMID: 40329096 DOI: 10.1038/s44321-025-00240-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 05/08/2025] Open
Abstract
Inhibition of the epidermal growth factor receptor (EGFR) shows clinical benefit in metastatic colorectal cancer (CRC) patients, but KRAS-mutations are known to confer resistance. However, recent reports highlight EGFR as a crucial target to be co-inhibited with RAS inhibitors for effective treatment of KRAS mutant CRC. Here, we investigated the tumor cell-intrinsic contribution of EGFR in KRASG12D tumors by establishing murine CRC organoids with key CRC mutations (KRAS, APC, TP53) and inducible EGFR deletion. Metabolomic, transcriptomic, and scRNA-analyses revealed that EGFR deletion in KRAS-mutant organoids reduced their phenotypic heterogeneity and activated a distinct cancer-stem-cell/WNT signature associated with reduced cell size and downregulation of major signaling cascades like MAPK, PI3K, and ErbB. This was accompanied by metabolic rewiring with a decrease in glycolytic routing and increased anaplerotic glutaminolysis. Mechanistically, following EGFR loss, Smoc2 was identified as a key upregulated target mediating these phenotypes that could be rescued upon additional Smoc2 deletion. Validation in patient-datasets revealed that the identified signature is associated with better overall survival of RAS mutant CRC patients possibly allowing to predict therapy responses in patients.
Collapse
Affiliation(s)
- Dana Krauß
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, Austria
| | - Veronica Moreno-Viedma
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, Austria
| | - Emi Adachi-Fernandez
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, Austria
| | - Cristiano de Sá Fernandes
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, 1090, Austria
| | - Jakob-Wendelin Genger
- Institute of Hygiene and Applied Immunology, Department of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, 1090, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, 1090, Austria
| | - Ourania Fari
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, Austria
| | - Bernadette Blauensteiner
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, Austria
| | - Dominik Kirchhofer
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, Austria
| | - Nikolina Bradaric
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Valeriya Gushchina
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, Austria
| | - Georgios Fotakis
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Thomas Mohr
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, Austria
| | - Ifat Abramovich
- Department of Cell Biology and Cancer Science, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Inbal Mor
- Department of Cell Biology and Cancer Science, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Molecular Biology, Ariel University, Ariel, 4070000, Israel
| | - Martin Holcmann
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, Austria
| | - Andreas Bergthaler
- Institute of Hygiene and Applied Immunology, Department of Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, 1090, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, 1090, Austria
| | - Arvand Haschemi
- Department of Laboratory Medicine, Medical University of Vienna, 1090, Vienna, Austria
| | - Zlatko Trajanoski
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Juliane Winkler
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, Austria
| | - Eyal Gottlieb
- Department of Cell Biology and Cancer Science, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maria Sibilia
- Center for Cancer Research, Medical University of Vienna and Comprehensive Cancer Center, Vienna, Austria.
| |
Collapse
|
8
|
Xie L, Luo D, Chen X, Zhang M, Zhang W, Chang J, Zhou H, Zhang X, He J, Chen L, Zhou C. RAS gene polymorphisms confer the risk of neuroblastoma in Chinese children from Jiangsu province. Pediatr Surg Int 2025; 41:130. [PMID: 40323475 DOI: 10.1007/s00383-025-06025-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/11/2025] [Indexed: 05/07/2025]
Abstract
INTRODUCTION To evaluate the potential association between single nucleotide polymorphisms in the RAS gene and neuroblastoma risk, we examined four candidate SNPs within this gene. METHODS Our hospital-based case-control study included 402 cases and 473 controls. Four SNPs (rs12587 G > T, rs7973450 A > G, and rs7312175 G > A in KRAS and rs2273267 A > T in NRAS) were genotyped using the TaqMan assay. The association between RAS gene polymorphisms and neuroblastoma susceptibility was assessed through odds ratios and 95% confidence intervals. RESULTS None of the four candidate SNPs exhibited a significant attribution to neuroblastoma risk. However, the concurrent presence of 2-3 KRAS risk genotypes significantly conferred an increased susceptibility to neuroblastoma (adjusted odds ratio [AOR] = 2.55, 95% confidence interval [CI] 1.33-4.89; P = 0.005). Further stratified analyses indicated that carriers of the KRAS rs12587 TT genotype tended to be more predisposed to neuroblastoma in males and in the subgroup with tumors originating from other sites. Additionally, the co-occurrence of 2-3 KRAS risk genotypes was found to be linked to an increased neuroblastoma risk in subgroups of individuals older than 18 months, males, tumors originating from retroperitoneum, mediastinum, or other sites, and those with tumors at clinical stage III + IV, respectively. CONCLUSIONS In summary, a single KRAS gene polymorphism may be weakly associated with an increased risk of childhood neuroblastoma in Jiangsu province, China, while the presence of more KRAS risk genotypes may increase the contribution to the risk of neuroblastoma.
Collapse
Affiliation(s)
- Lili Xie
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Dongyuan Luo
- Department of Stomatology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Xuemei Chen
- Department of Pharmacy, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, Yunnan, China
| | - Mengzhen Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Wenli Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Jiaming Chang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Haixia Zhou
- Department of Hematology, The Key Laboratory of Pediatric Hematology and Oncology Diseases of Wenzhou, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China
| | - Xinxin Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Liping Chen
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China.
| | - Chunlei Zhou
- Department of Pathology, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
9
|
Mateo-Victoriano B, Samaranayake GJ, Pokharel S, Sahayanathan GJ, Jayaraj C, Troccoli CI, Watson DC, Mohsen MG, Guo Y, Kool ET, Rai P. Oncogenic KRAS addiction states differentially influence MTH1 expression and 8-oxodGTPase activity in lung adenocarcinoma. Redox Biol 2025; 82:103610. [PMID: 40184641 PMCID: PMC11999683 DOI: 10.1016/j.redox.2025.103610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/11/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025] Open
Abstract
The efficacy of strategies targeting oncogenic RAS, prevalent in lung adenocarcinoma (LUAD), is limited by rapid adaptive resistance mechanisms. These include loss of RAS addiction and hyperactivation of downstream signaling pathways, such as PI3K/AKT. We previously reported that oncogenic RAS-driven LUAD cells possess an enhanced reliance on MTH1, the mammalian 8-oxodGTPase, to prevent genomic incorporation of oxidized nucleotides, and that MTH1 depletion compromises tumorigenesis and oncogenic signaling. Here, we show that elevated MTH1 correlates with poor prognosis in LUAD and that its redox-protective 8-oxodGTPase activity is variably regulated in KRAS-addicted vs. non-addicted states. Multiple oncogenic KRAS mutants or overexpression of wildtype (wt) KRAS increased MTH1 expression. Conversely, KRAS depletion or its inhibition by AMG-510 (sotorasib) decreased MTH1 in KRASG12C-addicted LUAD cells. Separation-of-function MEK/ERK1/2-activating mutants recapitulated the elevated MTH1 expression induced by oncogenic RAS in wt KRAS LUAD cells. However, upon inhibition of the MEK/ERK1/2 pathway, compensatory AKT activation maintained MTH1 expression. Indeed, elevated AKT signaling maintained high MTH1 expression even when KRAS oncoprotein was low. We previously reported that cancer cells possess variable MTH1-specific and MTH1-independent 8-oxodGTPase activity levels. Whereas both ERK1/2 and AKT could regulate MTH1 protein levels in KRAS-addicted cells, only AKT signaling was associated with elevated MTH1-specific 8-oxodGTPase activity under KRAS-low or KRAS non-addicted states. Our studies suggest that despite loss of KRAS dependency, LUAD cells retain the requirement for high MTH1 8-oxodGTPase activity due to redox vulnerabilities associated with AKT signaling. Thus, MTH1 may serve as a novel orthogonal vulnerability in LUAD that has lost KRAS addiction.
Collapse
Affiliation(s)
- Beatriz Mateo-Victoriano
- Department of Radiation Oncology, Division of Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Govindi J Samaranayake
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Sheela Pokharel
- Department of Radiation Oncology, Division of Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Gracy Jenifer Sahayanathan
- Department of Radiation Oncology, Division of Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Christina Jayaraj
- College of Arts and Sciences, University of Miami, Coral Gables, FL, 33146, USA
| | - Clara I Troccoli
- Sheila and David Fuente Graduate Program in Cancer Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Dionysios C Watson
- Department of Medicine, Division of Medical Oncology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA
| | - Michael G Mohsen
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Yan Guo
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, 33136, USA; Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA
| | - Eric T Kool
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Priyamvada Rai
- Department of Radiation Oncology, Division of Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Sylvester Comprehensive Cancer Center, Miami, FL, 33136, USA.
| |
Collapse
|
10
|
Ueki Y, Naylor RM, Ghozy SA, Thirupathi K, Rinaldo L, Kallmes DF, Kadirvel R. Advances in sporadic brain arteriovenous malformations: Novel genetic insights, innovative animal models and emerging therapeutic approaches. J Cereb Blood Flow Metab 2025; 45:793-799. [PMID: 39948029 PMCID: PMC11826813 DOI: 10.1177/0271678x251319913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 01/27/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025]
Abstract
Brain arteriovenous malformations (bAVMs) are a notable cause of intracranial hemorrhage, strongly associated with severe morbidity and mortality. Contemporary treatment options include surgery, stereotactic radiosurgery, and endovascular embolization, each of which has limitations. Hence, development of pharmacological interventions is urgently needed. The recent discovery of the presence of activating Kirsten rat sarcoma (KRAS) viral oncogene homologue mutations in most sporadic bAVMs has opened the door for a more comprehensive understanding of the pathogenesis of bAVMs and has pointed to entirely novel possible therapeutic targets. Herein, we review the status quo of genetics, animal models, and therapeutic approaches in bAVMs.
Collapse
Affiliation(s)
- Yasuhito Ueki
- Department of Neurologic Surgery, Mayo Clinic Rochester, MN, USA
- Department of Neurosurgery, Faculty of Medicine, The University of Juntendo, Tokyo, Japan
| | - Ryan M Naylor
- Department of Neurologic Surgery, Mayo Clinic Rochester, MN, USA
| | - Sherief A Ghozy
- Department of Neurologic Surgery, Mayo Clinic Rochester, MN, USA
| | | | - Lorenzo Rinaldo
- Department of Neurologic Surgery, Mayo Clinic Rochester, MN, USA
| | | | - Ramanathan Kadirvel
- Department of Neurologic Surgery, Mayo Clinic Rochester, MN, USA
- Department of Radiology, Mayo Clinic Rochester, MN, USA
| |
Collapse
|
11
|
Song H, Wang G, Gao G, Xia H, Jiao L, Wu K. A Systematic Analysis of Expression and Function of RAS GTPase-Activating Proteins (RASGAPs) in Urological Cancers: A Mini-Review. Cancers (Basel) 2025; 17:1485. [PMID: 40361412 PMCID: PMC12071082 DOI: 10.3390/cancers17091485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/14/2025] [Accepted: 04/25/2025] [Indexed: 05/15/2025] Open
Abstract
The RAS signaling pathway is one of the most commonly dysregulated pathways in urological cancers. This pathway can be regulated by RASGAPs, which catalyze the hydrolysis of RAS-GTP to RAS-GDP. As such, the loss of RASGAPs can promote the activation of the RAS signaling pathway. Dysregulation of RASGAPs significantly contributes to the progression of urological cancers, including prostate cancer, bladder cancer, and renal cell carcinoma. Furthermore, alterations in RASGAP expression may influence sensitivity to chemotherapy, radiotherapy, and targeted therapies, suggesting their potential as therapeutic targets. Despite the challenges involved, a deeper understanding of the complexity of the RAS signaling network, along with the evolution of personalized medicine, holds promise for delivering more precise and effective treatment options targeting RASGAPs in urological cancers.
Collapse
Affiliation(s)
- Hao Song
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (H.S.); (G.W.); (G.G.); (H.X.)
| | - Guojing Wang
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (H.S.); (G.W.); (G.G.); (H.X.)
| | - Guoqiang Gao
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (H.S.); (G.W.); (G.G.); (H.X.)
| | - Huayu Xia
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (H.S.); (G.W.); (G.G.); (H.X.)
| | - Lianying Jiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China;
| | - Kaijie Wu
- Department of Urology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (H.S.); (G.W.); (G.G.); (H.X.)
| |
Collapse
|
12
|
Wang X, Ding L, Sun S, Loo SK, Chen L, Li T, Wang MT, Pennathur A, Huang Y, Gao SJ. CASTOR1 : A Novel Tumor Suppressor Linking mTORC1 and KRAS Pathways in Tumorigenesis and Resistance to KRAS-Targeted Therapies in Non-Small Cell Lung Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.23.650349. [PMID: 40313924 PMCID: PMC12045348 DOI: 10.1101/2025.04.23.650349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Cytosolic arginine sensor for mTORC1 Subunit 1 (CASTOR1) functions as a key regulator of mechanistic target of rapamycin complex 1 (mTORC1) signaling. Despite its frequent dysregulation in cancers via mechanisms such as KSHV microRNA-mediated inhibition or AKT-driven phosphorylation and degradation, the impact of CASTOR1 loss on tumor initiation and progression remains poorly understood. Here, we identify CASTOR1 as a critical tumor suppressor in non-small cell lung cancer (NSCLC) by demonstrating that its genetic ablation amplifies tumorigenesis in a KRAS -driven genetically engineered mouse model (GEMM;LSL- KRAS G12D ). CASTOR1 deficiency markedly enhances lung tumor incidence, accelerates tumor progression, and increases proliferative indices in KRAS G12D -driven tumors ( KRAS G12D ; C1 KO ) compared to CASTOR1 wild type (WT) tumors ( KRAS G12D ; C1 WT ). Advanced-stage tumors exhibit elevated phosphorylated CASTOR1 (pCASTOR1) and reduced total CASTOR1 levels, suggesting active degradation during tumorigenesis. Mechanistically, CASTOR1 loss amplifies mTORC1 signaling, as evidenced by heightened phosphorylation of downstream effectors 4EBP1 and S6, while also augmenting AKT and ERK activation, uncovering a crosstalk between the PI3K/AKT/mTORC1 and KRAS/ERK pathways. Furthermore, CASTOR1 ablation induces genome instability, which may contribute to enhanced tumor incidence and progression. Importantly, CASTOR1 deficiency confers resistance to KRAS G12D -specific inhibitors, while over half of KRAS G12D ; C1 WT tumors also display resistance. Organoids derived from KRAS G12D ; C1 KO and KRAS G12D ; C1 WT tumors reveal a correlation between KRAS inhibitor resistance and hyperactivation of mTORC1, with mTORC1 and PI3K inhibitors sensitizing resistant tumors to KRAS G12D -targeted therapies. These findings position CASTOR1 as a novel tumor suppressor that modulates mTORC1 and KRAS signaling to constrain NSCLC progression. Our study further highlights the therapeutic potential of combining mTORC1 or ERK inhibitors with KRAS-targeted therapies for NSCLC characterized by hyperactive KRAS signaling and impaired CASTOR1 activity. Highlights CASTOR1 functions as a tumor suppressor in NSCLC by limiting KRAS -driven tumor initiation and progression. CASTOR1 is frequently lost or inactivated in wild-type tumors during tumor progression, contributing to advanced-stage malignancies.CASTOR1 deficiency amplifies mTORC1 signaling and enhances PI3K/AKT and KRAS/ERK crosstalk, driving tumorigenesis and resistance to KRAS-specific inhibitors. Combining mTORC1 or PI3K inhibitors with KRAS-targeted therapies effectively overcomes resistance in KRAS -driven NSCLC.
Collapse
|
13
|
Xu H, Yao Q, Hu X, Zheng D, Ren C, Ren Z, Gao Y. On-Membrane Supramolecular Assemblies Serving as Bioorthogonal Gating for Melphalan. Angew Chem Int Ed Engl 2025:e202502922. [PMID: 40272883 DOI: 10.1002/anie.202502922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/24/2025] [Accepted: 04/24/2025] [Indexed: 05/04/2025]
Abstract
Covalent drugs have experienced a revival in recent decades due to their advantageous pharmacodynamic profiles and targeting of "undruggable" proteins. However, balancing selectivity, reactivity, and potency is essential for safe and effective drugs. Here, we employ a cell-selective bioorthogonal prodrug design to enhance the selectivity for covalent inhibitors without compromising the reactivity and potency. The upregulation of phosphatase and integrin facilitates the formation of enzyme-instructed supramolecular assemblies (EISA) on the cancer cell membrane. These assemblies localize bioorthogonal reaction handles tetrazine (Tz), which liberate Melphalan from its bioorthogonal prodrug TCO-Mel. The TCO modification disrupts the LAT1-mediated transportation, reducing cellular permeability of TCO-Mel and the corresponding cytotoxicity to normal cells. Although the cell-selective on-membrane assemblies directed prodrug activation restores Melphalan influx to inhibit cancer cell growth. This prodrug activation strategy further demonstrates potent tumor suppression with satisfactory biocompatibility in vivo. Overall, we extend the scope of bioorthogonal prodrug design for covalent drugs via regulating cellular influx of active pharmaceutical ingredients (APIs).
Collapse
Affiliation(s)
- Hanlin Xu
- State Key Laboratory of Chemical Resource Engineering, MOE Key Lab of Biomedical Materials of Natural Macromolecules, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Qingxin Yao
- State Key Laboratory of Chemical Resource Engineering, MOE Key Lab of Biomedical Materials of Natural Macromolecules, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaoqian Hu
- State Key Laboratory of Chemical Resource Engineering, MOE Key Lab of Biomedical Materials of Natural Macromolecules, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Debin Zheng
- Medicine Medical Innovation Research Division of the Chinese PLA General Hospital, Beijing, 100853, China
| | - Chao Ren
- State Key Laboratory of Chemical Resource Engineering, MOE Key Lab of Biomedical Materials of Natural Macromolecules, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zhibin Ren
- State Key Laboratory of Chemical Resource Engineering, MOE Key Lab of Biomedical Materials of Natural Macromolecules, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yuan Gao
- State Key Laboratory of Chemical Resource Engineering, MOE Key Lab of Biomedical Materials of Natural Macromolecules, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
14
|
El Zaitouni S, Laraqui A, Boustany Y, Benmokhtar S, El Annaz H, Abi R, Tagajdid MR, El Kochri S, Bouaiti EA, Amine IL, Ameziane El Hassani R, Ennibi K. Potency and Safety of KRAS G12C Inhibitors in Solid Tumors: A Systematic Review. Clin Med Insights Oncol 2025; 19:11795549251331759. [PMID: 40297021 PMCID: PMC12035108 DOI: 10.1177/11795549251331759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 03/13/2025] [Indexed: 04/30/2025] Open
Abstract
Background The Kirsten rat sarcoma viral oncogene homolog (KRAS) gene, specifically the cysteine residue mutation KRAS (G12C), has garnered significant attention as a therapeutic target for solid cancer patients with KRAS mutations. Despite this interest, the efficacy and safety profiles of KRAS G12C inhibitors remain incompletely understood. In this study, we comprehensively evaluate the effectiveness and toxicity of relevant KRAS G12C inhibitors (Sotorasib, Adagrasib, Garsorasib, and Divarasib) in patients with colorectal cancer (CRC), non-small-cell lung cancer (NSCLC), and pancreatic ductal adenocarcinomas (PDAC). Methods Our systematic review is guided by Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. We review the available clinical trials data on KRAS G12C inhibitors in KRAS G12C-mutated solid tumors. We searched PubMed, EMBASE, Cochrane Library, and major international conferences for clinical trials from January 2020 until August 2023. Results A total of 17 eligible studies were included. KRAS G12C inhibitions with Sotorasib (41.2%) and Adagrasib (41.2%) each of them were reported in 7 studies. Divarasib was reported in 2 studies (11.8%) and Garsorasib was reported in 1 study (6.7%). Sotorasib showed a significant clinical benefit in terms of objective response rate (ORR) (7.1%-47%), progression-free survival (PFS) (4-6.8 months), and overall survival (OS) (4-24 months); it is more efficient in NSCLC patients with an OS of 2 years, PFS of 6.3 months, and an ORR of 41%. Adagrasib also showed significant clinical activity with an ORR (19%-53%), PFS (3.3-11.1 months), and OS (10.5-23.4 months), with more effectiveness in NSCLC patients with an OS of 23.4 months, PFS of 11.1 months, and an ORR of 53.3%. Adagrasib is more efficient with an ORR of 35.1%, PFS of 7.4 months, and an OS of 14 months in patients with PDAC, than Sotorasib which showed an ORR of 21%, PFS of 4 months, and an OS of 6.9 months. However, Adagrasib and Sotorasib are moderately efficient in CRC clinical trials. Conclusion This study confirms that patients treated with these KRAS G12C inhibitors, exclusively or combined with conventional therapies, achieve better treatment responses and modulate the progressions of these solid tumors.
Collapse
Affiliation(s)
- Sara El Zaitouni
- Laboratory of Biology of Human Pathologies, Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Abdelilah Laraqui
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital in Rabat, Rabat, Morocco
| | - Youssra Boustany
- Microbiology and Molecular Biology Team, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Soukaina Benmokhtar
- Laboratory of Biology of Human Pathologies, Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Hicham El Annaz
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital in Rabat, Rabat, Morocco
| | - Rachid Abi
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital in Rabat, Rabat, Morocco
| | - Mohamed Rida Tagajdid
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital in Rabat, Rabat, Morocco
| | - Safae El Kochri
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital in Rabat, Rabat, Morocco
| | - El Arbi Bouaiti
- Laboratory of Biostatistics, Clinical Research and Epidemiology, Faculty of Medicine and Pharmacy, Mohammed V University in Rabat, Rabat, Morocco
| | - Idriss Lahlou Amine
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital in Rabat, Rabat, Morocco
| | - Rabii Ameziane El Hassani
- Laboratory of Biology of Human Pathologies, Genomic Center of Human Pathologies, Department of Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Khalid Ennibi
- Royal School of Military Health Service, Sequencing Unit, Laboratory of Virology, Center of Virology, Infectious and Tropical Diseases, Mohammed V Military Teaching Hospital in Rabat, Rabat, Morocco
| |
Collapse
|
15
|
Kim HN, Gasmi-Seabrook GMC, Uchida A, Gebregiworgis T, Marshall CB, Ikura M. Switch II Pocket Inhibitor Allosterically Freezes KRAS G12D Nucleotide-binding Site and Arrests the GTPase Cycle. J Mol Biol 2025; 437:169162. [PMID: 40268231 DOI: 10.1016/j.jmb.2025.169162] [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: 02/17/2025] [Revised: 04/14/2025] [Accepted: 04/15/2025] [Indexed: 04/25/2025]
Abstract
KRAS is frequently mutated in multiple cancers, with the most common mutation being G12D. The recently developed KRASG12D inhibitor MRTX1133 binds a cryptic allosteric pocket near switch II (SII-P), similar to covalent G12C inhibitors, with remarkable picoM non-covalent affinity. Despite its advancement to clinical trials, some aspects of the molecular mechanisms-of-action remain unclear, indicating a need to uncover the mechanisms underlying MRTX1133 efficacy and potential acquired resistance, thus we characterized the biochemical and biophysical outcomes of MRTX1133 binding KRAS. Hydrogen/deuterium exchange experiments showed that MRTX1133 binding to the induced SII-P reduces the overall conformational plasticity of KRASG12D. This extends well beyond SII-P, with the nucleotide-binding regions (P-loop and G-3/4/5-box motifs) particularly exhibiting stabilization. This conformational rigidification by MRTX1133 is coupled with complete arrest of the GTPase cycle: When the compound engages KRASG12D-GDP, both intrinsic and GEF-mediated nucleotide exchange are blocked while engagement of KRASG12D-GTP blocks both intrinsic and GAP-mediated hydrolysis. MRTX1133 attenuates the interaction between activated KRASG12D and the RAS-binding domain of the effector BRAF. The binding site in Switch I remains flexible, which enables binding, albeit with ∼10-fold lower affinity, and remarkably, this interaction with BRAF reverses the compound's blockage of intrinsic GTP hydrolysis. Unlike KRASWT, GDP-loaded KRASG12D surprisingly maintains a low-affinity interaction with BRAF-RBD, but MRTX1133 can circumvent this mutant-specific abnormal interaction. Taken together, MRTX1133 allosterically 'freezes' the KRASG12D nucleotide-binding site conformation, arresting the canonical GTPase cycle of this oncogenic mutant. This provides a framework for understanding the mechanisms-of-action of SII-P-directed inhibitors and how tumours may acquire resistance.
Collapse
Affiliation(s)
- Ha-Neul Kim
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | | | - Arisa Uchida
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Teklab Gebregiworgis
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada
| | - Christopher B Marshall
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada.
| | - Mitsuhiko Ikura
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario M5G 1L7, Canada; Department of Medical Biophysics, University of Toronto, 610 University Avenue, Toronto, Ontario M5G 2M9, Canada.
| |
Collapse
|
16
|
Li X, Mendez Q, Chapados C, Acca F, Driscoll H, Oliveira J, Liu J, Jones K, Ferguson M, Wallace RL, Bibikov S, Lionberger T, Harvey KJ, Weiner MP, Mirando G. Site-directed antibodies targeting driver mutations of the KRAS protein. N Biotechnol 2025; 87:112-120. [PMID: 40252917 DOI: 10.1016/j.nbt.2025.04.003] [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: 12/20/2024] [Revised: 03/14/2025] [Accepted: 04/05/2025] [Indexed: 04/21/2025]
Abstract
Kirsten rat sarcoma viral oncogene homolog (KRAS) is the most mutated oncogene in human cancers, found in approximately 30 % of tumors. These mutations primarily consist of single-base missense alterations in codon G12. While extensive efforts have focused on developing allele-specific inhibitors for KRAS mutations, mutation-specific antibodies (Abs) remain largely unexplored, with only a few research-use-only catalog Abs available. In this study, we employed the proprietary Epivolve technology to develop site-directed monoclonal Abs (mAbs) that target KRAS oncogenic driver mutation KRAS G12D. These site-directed mAbs demonstrate high binding affinity, with equilibrium dissociation constants (KD) in the nanomolar range, showing over 1,000-fold greater affinity for KRAS G12D compared to wild-type KRAS. Western blot analyses using both purified KRAS protein variants and tumor cell lines harboring G12D mutations confirmed the high specificity of these mAbs. Furthermore, immunocytochemistry analysis revealed co-localization of the site-directed mAbs with endogenously expressed KRAS in cancer cells bearing G12D mutations. The validated high affinity and specificity of these site-directed mAbs highlight their potential for diagnostic applications and therapeutic development targeting KRAS driver mutations.
Collapse
Affiliation(s)
- Xiaofeng Li
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA.
| | - Qiana Mendez
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | | | - Felicity Acca
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | - Holly Driscoll
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | - Jason Oliveira
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | - Jun Liu
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | - Kezzia Jones
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | - Mary Ferguson
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | - Ryan L Wallace
- Aviva Systems Biology Corporation, 6370 Nancy Ridge Dr., Suite 104, San Diego, CA 92121, USA
| | - Sergei Bibikov
- Aviva Systems Biology Corporation, 6370 Nancy Ridge Dr., Suite 104, San Diego, CA 92121, USA
| | - Troy Lionberger
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | - Kevin J Harvey
- Aviva Systems Biology Corporation, 6370 Nancy Ridge Dr., Suite 104, San Diego, CA 92121, USA
| | - Michael P Weiner
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| | - Greg Mirando
- Abbratech, 25 Business Park Drive, Suite C, Branford, CT 06405, USA
| |
Collapse
|
17
|
Scott E, Yun SD, Moghadamchargari Z, Bahramimoghaddam H, Chang JY, Zhang T, Zhu Y, Lyu J, Laganowsky A. Real time characterization of the MAPK pathway using native mass spectrometry. Commun Biol 2025; 8:617. [PMID: 40240517 PMCID: PMC12003711 DOI: 10.1038/s42003-025-08028-5] [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: 11/04/2024] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
The MAPK pathway is a crucial cell-signaling cascade that is composed of RAS, MEK, BRAF, and ERK, which serves to connect extracellular signals to intracellular responses. Over-activating mutations in the MAPK pathway can lead to uncontrolled cell growth ultimately resulting in various types of cancer. While this pathway has been heavily studied using a battery of techniques, herein we employ native mass spectrometry (MS) to characterize the MAPK pathway, including nucleotide, drug, and protein interactions. We utilize native MS to provide detailed insights into nucleotide and drug binding to BRAF complexes, such as modulation of nucleotide binding in the presence of MEK1. We then demonstrate that different CRAF segments vary in their complex formation with KRAS, with the addition of the cysteine rich domain (CRD) enhancing complex formation compared to Ras binding domain (RBD) alone. We report differences in KRAS GTPase activity in the presence of different RAF segments, with KRAS exhibiting significantly enhanced nucleotide turnover when bound to CRAF fragments. We use ERK2 as a downstream readout to monitor the MAPK phosphorylation cascade. This study demonstrates the utility of native MS to provide detailed characterization of individual MAPK pathway components and monitor the phosphorylation cascade in real time.
Collapse
Affiliation(s)
- Elena Scott
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - Sangho D Yun
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | | | | | - Jing-Yuan Chang
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - Tianqi Zhang
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - Yun Zhu
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - Jixing Lyu
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA
| | - Arthur Laganowsky
- Department of Chemistry, Texas A&M University, College Station, TX, 77843, USA.
| |
Collapse
|
18
|
Yuan T, Liu Y, Wu R, Qian M, Wang W, Li Y, Zhu H, Wang J, Ge F, Zeng C, Dai X, Hu R, Zhou T, He Q, Zhu H, Yang B. Josephin Domain Containing 2 (JOSD2) inhibition as Pan-KRAS-mutation-targeting strategy for colorectal cancer. Nat Commun 2025; 16:3623. [PMID: 40240366 PMCID: PMC12003847 DOI: 10.1038/s41467-025-58923-y] [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: 07/30/2024] [Accepted: 04/07/2025] [Indexed: 04/18/2025] Open
Abstract
KRAS is the most common mutated oncogenes in colorectal cancer (CRC), yet effective therapeutic strategies for targeting multiple KRAS mutations remained challenging. The prolonged protein stability of KRAS mutants contribute to their robust tumor-promoting effects, but the underlying mechanism is elusive. Herein by screening deubiquitinases (DUBs) siRNA library, we identify Josephin domain containing 2 (JOSD2) functions as a potent DUB that regulates the protein stability of KRAS mutants. Mechanistically, JOSD2 directly interacts with and stabilizes KRAS variants across different mutants, by reverting their proteolytic ubiquitination; while KRAS mutants reciprocally inhibit the catalytic activity of CHIP, a bona fide E3 ubiquitin ligase for JOSD2, thus forming a JOSD2/KRAS positive feedback circuit that significantly accelerates KRAS-mutant CRC growth. Inhibition of JOSD2 by RNA interference or its pharmacological inhibitor promotes the polyubiquitination and proteasomal degradation of KRAS mutants, and preferentially impede the growth of KRAS-mutant CRC including patient-derived cells/xenografts/organoids (PDCs/PDXs/PDOs) over that harboring wild-type KRAS. Collectively, this study not only reveals the crucial roles of JOSD2/KRAS positive feedback circuit in KRAS-mutant CRC, but also provides a rationale to target JOSD2 as the promising pan-KRAS-mutation-targeting strategy for the treatment of a broad population of CRC patients with KRAS variant across different mutant types.
Collapse
Affiliation(s)
- Tao Yuan
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yue Liu
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ruilin Wu
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Meijia Qian
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Weihua Wang
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yonghao Li
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Hongdao Zhu
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Jia'er Wang
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Fujing Ge
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Chenming Zeng
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoyang Dai
- Center for Drug Safety Evaluation and Research of Zhejiang University, Hangzhou, China
| | - Ronggui Hu
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tianhua Zhou
- Department of Cell Biology, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, China
| | - Hong Zhu
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Bo Yang
- Institute of Pharmacology & Toxicology, Zhejiang Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
- School of Medicine, Hangzhou City University, Hangzhou, China.
| |
Collapse
|
19
|
Hashimoto Y, Shil S, Tsuruta M, Kawauchi K, Miyoshi D. Three- and four-stranded nucleic acid structures and their ligands. RSC Chem Biol 2025; 6:466-491. [PMID: 40007865 PMCID: PMC11848209 DOI: 10.1039/d4cb00287c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
Nucleic acids have the potential to form not only duplexes, but also various non-canonical secondary structures in living cells. Non-canonical structures play regulatory functions mainly in the central dogma. Therefore, nucleic acid targeting molecules are potential novel therapeutic drugs that can target 'undruggable' proteins in various diseases. One of the concerns of small molecules targeting nucleic acids is selectivity, because nucleic acids have only four different building blocks. Three- and four-stranded non-canonical structures, triplexes and quadruplexes, respectively, are promising targets of small molecules because their three-dimensional structures are significantly different from the canonical duplexes, which are the most abundant in cells. Here, we describe some basic properties of the triplexes and quadruplexes and small molecules targeting the triplexes and tetraplexes.
Collapse
Affiliation(s)
- Yoshiki Hashimoto
- Frontiers of Innovative Research in Science and Technology, Konan University 7-1-20 Minatojima-minamimachi, Chuo-ku, Kobe Hyogo 650-0047 Japan
| | - Sumit Shil
- Frontiers of Innovative Research in Science and Technology, Konan University 7-1-20 Minatojima-minamimachi, Chuo-ku, Kobe Hyogo 650-0047 Japan
| | - Mitsuki Tsuruta
- Frontiers of Innovative Research in Science and Technology, Konan University 7-1-20 Minatojima-minamimachi, Chuo-ku, Kobe Hyogo 650-0047 Japan
| | - Keiko Kawauchi
- Frontiers of Innovative Research in Science and Technology, Konan University 7-1-20 Minatojima-minamimachi, Chuo-ku, Kobe Hyogo 650-0047 Japan
| | - Daisuke Miyoshi
- Frontiers of Innovative Research in Science and Technology, Konan University 7-1-20 Minatojima-minamimachi, Chuo-ku, Kobe Hyogo 650-0047 Japan
| |
Collapse
|
20
|
Ji X, Liu M, Zhang T, Zhang W, Xue F, Wan Q, Liu Y. KRAS/PI3K axis driven GTF3C6 expression and promotes LUAD via FAK pathway. J Adv Res 2025; 70:243-254. [PMID: 38685529 PMCID: PMC11976405 DOI: 10.1016/j.jare.2024.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024] Open
Abstract
INTRODUCTION Effective targeting drugs for KRAS mutation-mediated Lung Adenocarcinoma (LUAD) are currently are limited. OBJECTIVES Investigating and intervening in the downstream key target genes of KRAS is crucial for clinically managing KRAS mutant-driven LUAD. GTF3C6, a newly identified member of the general transcription factor III (GTF3) family, plays a role in the transcription of RNA polymerase III (pol III)-dependent genes. However, its involvement in cancer remains unexplored. METHODS This study examined the expression, roles, and potential molecular mechanisms of GTF3C6 in LUAD tissues, LSL-KrasG12D/+;LSL-p53-/- LUAD mouse models, and LUAD patients-derived organoid using Western blot, qRT-PCR, immunofluorescence, immunohistochemistry, and gene manipulation assays. RESULTS We present the first evidence that GTF3C6 is highly expressed in LUAD tissues, LSL-KrasG12D/+;LSL-p53-/- LUAD mouse models, and LUAD organoids, correlating with poor clinical prognosis. Furthermore, GTF3C6 was found to promote anchorage-independent proliferation, migration, and invasion of LUAD cells. Mechanistically, KRAS mutation drives GTF3C6 expression through the PI3K pathway, and GTF3C6 knockdown reverses the malignant phenotype of KRAS mutation-driven LUAD cells. Additionally, the FAK pathway emerged as a crucial downstream signaling pathway through which GTF3C6 mediates the malignant phenotype of LUAD. Finally, GTF3C6 knockdown suppresses LUAD organoid formation and inhibits tumor growth in vivo. CONCLUSION Our findings demonstrate that GTF3C6, driven by KRAS mutation, promotes LUAD development by regulating FAK phosphorylation, suggesting its potential as a biomarker and therapeutic target in KRAS mutant-driven LUAD.
Collapse
Affiliation(s)
- Xingzhao Ji
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Infections Respiratory Disease, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Mingqiang Liu
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Department of Pharmacy, Pingdu People's Hospital, Qingdao, Shandong 266799, China
| | - Tianyi Zhang
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Weiying Zhang
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Fuyuan Xue
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Qiang Wan
- Key Laboratory of Cell Metabolism in Medical and Health of Shandong Provincial Health Commission, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China.
| | - Yi Liu
- Department of Pulmonary and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Key Laboratory of Infections Respiratory Disease, Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
21
|
Dissanayake UC, Roy A, Maghsoud Y, Polara S, Debnath T, Cisneros GA. Computational studies on the functional and structural impact of pathogenic mutations in enzymes. Protein Sci 2025; 34:e70081. [PMID: 40116283 PMCID: PMC11926659 DOI: 10.1002/pro.70081] [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: 11/08/2024] [Revised: 01/23/2025] [Accepted: 02/12/2025] [Indexed: 03/23/2025]
Abstract
Enzymes are critical biological catalysts involved in maintaining the intricate balance of metabolic processes within living organisms. Mutations in enzymes can result in disruptions to their functionality that may lead to a range of diseases. This review focuses on computational studies that investigate the effects of disease-associated mutations in various enzymes. Through molecular dynamics simulations, multiscale calculations, and machine learning approaches, computational studies provide detailed insights into how mutations impact enzyme structure, dynamics, and catalytic activity. This review emphasizes the increasing impact of computational simulations in understanding molecular mechanisms behind enzyme (dis)function by highlighting the application of key computational methodologies to selected enzyme examples, aiding in the prediction of mutation effects and the development of therapeutic strategies.
Collapse
Affiliation(s)
- Upeksha C. Dissanayake
- Department of Chemistry and BiochemistryThe University of Texas at DallasRichardsonTexasUSA
| | - Arkanil Roy
- Department of Chemistry and BiochemistryThe University of Texas at DallasRichardsonTexasUSA
| | - Yazdan Maghsoud
- Department of Chemistry and BiochemistryThe University of Texas at DallasRichardsonTexasUSA
- Present address:
Department of Biochemistry and Molecular PharmacologyBaylor College of MedicineHoustonTexasUSA
| | - Sarthi Polara
- Department of Chemistry and BiochemistryThe University of Texas at DallasRichardsonTexasUSA
| | - Tanay Debnath
- Department of PhysicsThe University of Texas at DallasRichardsonTexasUSA
- Present address:
Department of Pathology and Molecular MedicineQueen's UniversityKingstonOntarioCanada
| | - G. Andrés Cisneros
- Department of Chemistry and BiochemistryThe University of Texas at DallasRichardsonTexasUSA
- Department of PhysicsThe University of Texas at DallasRichardsonTexasUSA
| |
Collapse
|
22
|
Cregg J, Edwards AV, Chang S, Lee BJ, Knox JE, Tomlinson ACA, Marquez A, Liu Y, Freilich R, Aay N, Wang Y, Jiang L, Jiang J, Wang Z, Flagella M, Wildes D, Smith JAM, Singh M, Wang Z, Gill AL, Koltun ES. Discovery of Daraxonrasib (RMC-6236), a Potent and Orally Bioavailable RAS(ON) Multi-selective, Noncovalent Tri-complex Inhibitor for the Treatment of Patients with Multiple RAS-Addicted Cancers. J Med Chem 2025; 68:6064-6083. [PMID: 40056080 DOI: 10.1021/acs.jmedchem.4c02314] [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: 03/28/2025]
Abstract
Oncogenic RAS mutations are among the most common in human cancers. To target the active, GTP-bound state of RAS(ON) directly, we employed an innovative tri-complex inhibitor (TCI) modality. Formation of a complex with an intracellular chaperone protein CypA, an inhibitor, and a target protein RAS blocks effector binding, inhibiting downstream RAS signaling and tumor cell proliferation. Herein, we describe the structure-guided SAR journey that led to the discovery of daraxonrasib (RMC-6236), a noncovalent, potent tri-complex inhibitor of multiple RAS mutant and wild-type (WT) variants. This orally bioavailable bRo5 macrocyclic molecule occupies a unique composite binding pocket comprising CypA and SWI/SWII regions of RAS(ON). To achieve broad-spectrum RAS isoform activity, we deployed an SAR campaign that focused on interactions with residues conserved between mutants and WT RAS isoforms. Concurrent optimization of potency and drug-like properties led to the discovery of daraxonrasib (RMC-6236), currently in clinical evaluation in RAS mutant advanced solid tumors (NCT05379985; NCT06040541; NCT06162221; NCT06445062; NCT06128551).
Collapse
Affiliation(s)
- James Cregg
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Anne V Edwards
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Stephanie Chang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Bianca J Lee
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - John E Knox
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | | | - Abby Marquez
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Yang Liu
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Rebecca Freilich
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Naing Aay
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Yingyun Wang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Lingyan Jiang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Jingjing Jiang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Zhican Wang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Michael Flagella
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - David Wildes
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | | | - Mallika Singh
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Zhengping Wang
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Adrian L Gill
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| | - Elena S Koltun
- Revolution Medicines, Inc., Redwood City, California 94063, United States
| |
Collapse
|
23
|
Baars B, Orive-Ramos A, Kou Z, Gaire B, Desaunay M, Adamopoulos C, Aaronson SA, Wang S, Gavathiotis E, Poulikakos PI. RAS mutation-specific signaling dynamics in response to paralog- and state- selective RAS inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638317. [PMID: 40166154 PMCID: PMC11956912 DOI: 10.1101/2025.02.14.638317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
A high therapeutic index (TI), balancing potent oncogenic signaling inhibition in tumor cells with minimal effects on normal cells, is critical for effective cancer therapies. Recent advances have introduced diverse RAS-targeting inhibitors, including mutant-specific inhibitors (e.g., KRAS(G12C) and KRAS(G12D)), as well as paralog- and state-selective inhibitors. Non-mutant-specific RAS inhibition can be accomplished by 1) panRAS-GEF(OFF) inhibitors which inactivate RAS indirectly by inhibiting SHP2 or SOS1, thereby blocking the nucleotide exchange step of RAS activation, 2) direct KRAS(OFF)-selective inhibitors sparing NRAS and HRAS, and 3) panRAS(ON) inhibitors that directly target active RAS, by occluding binding of its effector RAF. However, the signaling inhibition index (SII) - the differential inhibition of oncogenic signaling between RAS-mutant (RAS(MUT)) and normal cells - remains poorly defined for these approaches. In this study, we evaluated the SII of state- and paralog-selective RAS inhibitors across diverse RAS-mutant (RAS(MUT)) and RAS-wild-type (RAS(WT)) models. PanRAS-GEF(OFF) inhibitors exhibited neutral or negative SII, with comparable or reduced MAPK suppression in KRAS(G12X) cells relative to RAS(WT) cells. KRAS(G13D) models showed low sensitivity (negative SII) to panRAS-GEF(OFF) inhibitors, particularly in the context of NF1 loss. Combination treatments with SHP2 and MEK inhibitors resulted in low SII, as pathway suppression was similar in RAS(MUT) and RAS(WT) cells. Furthermore, RAS(Q61X) models were resistant to combined SHP2 inhibitor+MEK inhibitor due to dual mechanisms: MEK inhibitor-induced NRAS(Q61X) reactivation and RAS(MUT)-induced SHP2 conformations impairing inhibitor binding. Overall, panRAS-GEF(OFF) inhibitors exhibited the lowest SII. PanKRAS(OFF) inhibitors demonstrated a higher SII, while panRAS(ON) inhibitors displayed broader activity but relatively narrow SII. We observed that tumors that were sensitive to RAS(MUT)-specific inhibitors, were also sensitive to the state-selective RAS inhibitors (OFF, or ON). In fact, all RAS inhibitors (mutant-specific and state- or paralog-selective) were active in the same portion of RAS(MUT) models, while the majority of RAS(MUT) cell lines were insensitive to all of them. These findings reveal significant SII variability among RAS-targeted inhibitors, depending on the specific RAS driver mutation and cell context and underscore the importance of incorporating SII considerations into the design and clinical application of RAS-targeted therapies to improve therapeutic outcomes. Main points PanRAS-GEF(OFF) inhibitors have limited SII and effectiveness: The Signaling Inhibition Index (SII) - i.e. the differential inhibition of oncogenic signaling between tumor and normal cells - was neutral or negative for panRAS-GEF(OFF) inhibitors, with comparable or reduced MAPK suppression in KRAS(G12X) mutant versus RAS(WT) cells. KRAS(G13D) models showed reduced sensitivity, particularly with NF1 loss. SHP2+MEK inhibitor combinations also had low SII, with RAS(Q61X) models demonstrating resistance due to NRAS(Q61X) reactivation and impaired SHP2 inhibitor binding.PanKRAS(OFF) selective inhibitors have higher SII than panRAS-GEF(OFF) inhibitors: panKRAS(OFF)-selective inhibitors have a higher SII compared to panRAS-GEF(OFF) inhibitors, offering better tumor-versus-normal cell selectivity.PanRAS(ON) inhibitors have broad but modest SII: While panRAS(ON) inhibitors displayed a broader activity profile, their ability to selectively inhibit mutant RAS signaling over normal cells remained relatively narrow (low SII).Most KRAS-mutant tumors will be insensitive to any single RAS-targeted inhibitor: State- and paralog-selective inhibitors have enhanced activity in the same RAS-MUT cancer models that are also sensitive to RAS-MUT-specific inhibitors, suggesting that most KRAS-MUT tumors will not respond uniformly to any one RAS-targeting inhibitor.SII varies across RAS inhibitors, necessitating tailored therapeutic strategies: The effectiveness of paralog- and state-selective inhibitors depends on specific RAS mutations and cell context, highlighting the need to integrate SII considerations into the development and clinical application of RAS-targeted therapies.
Collapse
|
24
|
Zhou Y, Tao Q, Luo C, Chen J, Chen G, Sun J. Epacadostat Overcomes Cetuximab Resistance in Colorectal Cancer by Targeting IDO-Mediated Tryptophan Metabolism. Cancer Sci 2025. [PMID: 40103010 DOI: 10.1111/cas.70057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 03/20/2025] Open
Abstract
Primary or acquired mutations in RAS/RAF genes resulting in cetuximab resistance have limited its clinical application in colorectal cancer (CRC) patients. The mechanism of this resistance remains unclear. RNA sequencing from cetuximab-sensitive and -resistant specimens revealed an activation of the tryptophan pathway and elevation of IDO1 and IDO2 in cetuximab-resistant CRC patients. In vitro, in vivo, and clinical specimens confirmed the upregulation of IDO1and IDO2 and the Kyn/Trp after cetuximab treatment. Additionally, the IDO inhibitor, epacadostat, could effectively inhibit the migration and proliferation of cetuximab-resistant CRC cells while promoting apoptosis. Compared to epacadostat monotherapy, the combination of cetuximab and epacadostat showed a stronger synergistic anti-tumor effect. Furthermore, in vivo experiments confirmed that combination therapy effectively suppressed tumor growth. Mechanistically, KEGG pathway analysis revealed the activation of the IFN-γ pathway in cetuximab-resistant CRC tissues. Luciferase reporter assays confirmed the transcriptional activity of IDO1 following cetuximab treatment. Silencing IFN-γ then suppressed the upregulation induced by cetuximab. Moreover, we observed that the combination reduced the concentration of the tryptophan metabolite kynurenine, promoted the infiltration of CD8+ T lymphocytes, and enhanced the polarization of M1 macrophages within the tumor microenvironment, thereby exerting potent anti-tumor immune effects. Overall, our results confirm the remarkable therapeutic efficacy of combining cetuximab with epacadostat in cetuximab-resistant CRC. Our findings may provide a novel target for overcoming cetuximab resistance in CRC.
Collapse
Affiliation(s)
- Yimin Zhou
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiongyan Tao
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chubin Luo
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry of Education, Shanghai, China
| | - Jinsong Chen
- Department of Clinical Medicine, Shaoguan University, Shaoguan, Guangdong, China
| | - Genwen Chen
- Department of Radiation Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianyong Sun
- Department of Gastroenterology, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Schock Vaiani J, Broekgaarden M, Coll JL, Sancey L, Busser B. In vivo vectorization and delivery systems for gene therapies and RNA-based therapeutics in oncology. NANOSCALE 2025; 17:5501-5525. [PMID: 39927415 DOI: 10.1039/d4nr05371k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
Gene and RNA-based therapeutics represent a promising frontier in oncology, enabling targeted modulation of tumor-associated genes and proteins. This review explores the latest advances in payload vectorization and delivery systems developed for in vivo cancer treatments. We discuss viral and non-viral organic particles, including lipid based nanoparticles and polymeric structures, for the effective transport of plasmids, siRNA, and self-amplifying RNA therapeutics. Their physicochemical properties, strategies to overcome intracellular barriers, and innovations in cell-based carriers and engineered extracellular vesicles are highlighted. Moreover, we consider oncolytic viruses, novel viral capsid modifications, and approaches that refine tumor targeting and immunomodulation. Ongoing clinical trials and regulatory frameworks guide future directions and emphasize the need for safe, scalable production. The potential convergence of these systems with combination therapies paves the way toward personalized cancer medicine.
Collapse
Affiliation(s)
- Julie Schock Vaiani
- Univ. Grenoble-Alpes (UGA), INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Allée des Alpes, 38000 Grenoble, France.
| | - Mans Broekgaarden
- Univ. Grenoble-Alpes (UGA), INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Allée des Alpes, 38000 Grenoble, France.
| | - Jean-Luc Coll
- Univ. Grenoble-Alpes (UGA), INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Allée des Alpes, 38000 Grenoble, France.
| | - Lucie Sancey
- Univ. Grenoble-Alpes (UGA), INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Allée des Alpes, 38000 Grenoble, France.
| | - Benoit Busser
- Univ. Grenoble-Alpes (UGA), INSERM U1209, CNRS UMR 5309, Institute for Advanced Biosciences, Allée des Alpes, 38000 Grenoble, France.
- Grenoble Alpes Univ. Hospital (CHUGA), 38043 Grenoble, France
- Institut Universitaire de France (IUF), 75005 Paris, France
| |
Collapse
|
26
|
Chen C, Wei Z. Mechanisms and molecular characterization of relapsed/refractory neuroblastomas. Front Oncol 2025; 15:1555419. [PMID: 40115016 PMCID: PMC11922920 DOI: 10.3389/fonc.2025.1555419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 02/18/2025] [Indexed: 03/22/2025] Open
Abstract
Relapsed/refractory neuroblastoma is a type of malignant solid tumor with a very poor prognosis in children. Its pathogenesis is complex, involving multiple molecular pathways and genetic alterations. Recent studies have shown that MYCN amplification, ALK mutation, TERT promoter mutation, p53 pathway inactivation, and chromosomal instability are the key mechanisms and molecular characteristics of relapsed/refractory neuroblastoma. Precision treatment strategies targeting these molecular mechanisms have shown certain prospects in preclinical studies and clinical practice. This review focuses on the relevant mechanisms and molecular characteristics of relapsed/refractory neuroblastoma, explores its relationship with treatment response and clinical prognosis, and briefly introduces the current treatment strategies to provide a theoretical basis for the development of novel and personalized therapeutic regimens to improve the prognosis of children.
Collapse
Affiliation(s)
- Chong Chen
- Department of Clinical Laboratory, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Zixuan Wei
- Department of Pediatric Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Department of Pediatric Oncology, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Pediatric Oncology, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| |
Collapse
|
27
|
Ge Z, Fan Z, He W, Zhou G, Zhou Y, Zheng M, Zhang S. Recent advances in targeted degradation in the RAS pathway. Future Med Chem 2025; 17:693-708. [PMID: 40065567 PMCID: PMC11938967 DOI: 10.1080/17568919.2025.2476387] [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: 11/26/2024] [Accepted: 02/12/2025] [Indexed: 03/26/2025] Open
Abstract
RAS (rat sarcoma) is one of the most frequently mutated gene families in cancer, encoding proteins classified as small GTPases. Mutations in RAS proteins result in abnormal activation of the RAS signaling pathway, a key driver in the initiation and progression of various malignancies. Consequently, targeting RAS proteins and the RAS signaling pathway has become a critical strategy in anticancer therapy. While RAS was historically considered an "undruggable" target, recent breakthroughs have yielded inhibitors specifically targeting KRASG12C and KRASG12D mutations, which have shown clinical efficacy in patients. However, these inhibitors face limitations due to rapid acquired resistance and the toxic effects of combination therapies in clinical settings. Targeted protein degradation (TPD) strategies, such as PROTACs and molecular glues, provide a novel approach by selectively degrading RAS proteins, or their upstream and downstream regulatory factors, to block aberrant signaling pathways. These degraders offer a promising alternative to traditional inhibitors by potentially circumventing resistance and enhancing therapeutic precision. This review discusses recent advancements in RAS pathway degraders, with an emphasis on targeting RAS mutations as well as their upstream regulators and downstream effectors for potential cancer treatments.
Collapse
Affiliation(s)
- Zhiming Ge
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Zisheng Fan
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Shanghai Institute for Advanced Immunochemical Studies, and School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Lingang Laboratory, Shanghai, China
| | - Wei He
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- School of Pharmacy, Nanchang University, Nanchang, China
| | - Guizhen Zhou
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Shanghai Institute for Advanced Immunochemical Studies, and School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- Lingang Laboratory, Shanghai, China
| | - Yidi Zhou
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Mingyue Zheng
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- University of Chinese Academy of Sciences, Beijing, China
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Shanghai Institute for Advanced Immunochemical Studies, and School of Life Science and Technology, ShanghaiTech University, Shanghai, China
- School of Pharmacy, Nanchang University, Nanchang, China
| | - Sulin Zhang
- University of Chinese Academy of Sciences, Beijing, China
- Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
28
|
Kazi A, Vasiyani H, Ghosh D, Bandyopadhyay D, Shah RD, Vudatha V, Trevino J, Sebti SM. FGTI-2734 Inhibits ERK Reactivation to Overcome Sotorasib Resistance in KRAS G12C Lung Cancer. J Thorac Oncol 2025; 20:331-344. [PMID: 39603412 PMCID: PMC11885004 DOI: 10.1016/j.jtho.2024.11.022] [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: 08/15/2024] [Revised: 10/09/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024]
Abstract
INTRODUCTION KRAS G12C targeted therapies, such as sotorasib, represent a major breakthrough, but overall response rates and progression-free survival for patients with KRAS G12C lung cancer are modest due to the emergence of resistance mechanisms involving adaptive reactivation of ERK, which requires wild-type HRAS and NRAS membrane localization. METHODS AND RESULTS Here, we demonstrate that the dual farnesyltransferase and geranylgeranyltransferase-1 inhibitor FGTI-2734 inhibits wild-type RAS membrane localization and sotorasib-induced ERK feedback reactivation, and overcomes sotorasib adaptive resistance. The combination of FGTI-2734 and sotorasib is synergistic at inhibiting the viability and inducing apoptosis of KRAS G12C lung cancer cells, including those highly resistant to sotorasib. FGTI-2734 enhances sotorasib's anti-tumor activity in vivo leading to significant tumor regression of a patient-derived xenograft (PDX) from a patient with KRAS G12C lung cancer and several xenografts from highly sotorasib-resistant KRAS G12C human lung cancer cells. Importantly, treatment of mice with FGTI-2734 inhibited sotorasib-induced ERK reactivation in KRAS G12C PDX, and treatment of mice with the combination of FGTI-2734 and sotorasib was also significantly more effective at suppressing in vivo the levels of P-ERK in sotorasib-resistant human KRAS G12C lung cancer xenografts and the NSCLC PDX. CONCLUSION Our findings provide a foundation for overcoming sotorasib resistance and potentially improving the treatment outcomes of KRAS G12C lung cancer.
Collapse
Affiliation(s)
- Aslamuzzaman Kazi
- Department of Pharmacology and Toxicology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Hitesh Vasiyani
- Department of Pharmacology and Toxicology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Deblina Ghosh
- Department of Pharmacology and Toxicology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | | | - Rachit D Shah
- Department of Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Vignesh Vudatha
- Department of Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Jose Trevino
- Department of Surgery, Virginia Commonwealth University, Richmond, Virginia
| | - Said M Sebti
- Department of Pharmacology and Toxicology and Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, Virginia.
| |
Collapse
|
29
|
Hu Z, Martí J. Unraveling atomic-scale mechanisms of GDP extraction catalyzed by SOS1 in KRAS-G12 and KRAS-D12 oncogenes. Comput Biol Med 2025; 186:109599. [PMID: 39731920 DOI: 10.1016/j.compbiomed.2024.109599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024]
Abstract
The guanine exchange factor SOS1 plays a pivotal role in the positive feedback regulation of the KRAS signaling pathway. Recently, the regulation of KRAS-SOS1 interactions and KRAS downstream effector proteins has emerged as a key focus in the development of therapies targeting KRAS-driven cancers. However, the detailed dynamic mechanisms underlying SOS1-catalyzed GDP extraction and the impact of KRAS mutations remain largely unexplored. In this study, we unveil and describe in atomic detail the primary mechanisms by which SOS1 facilitates GDP extraction from KRAS oncogenes. For GDP-bound wild-type KRAS (KRAS-G12), four critical amino acids (Lys811, Glu812, Lys939, and Glu942) are identified as essential for the catalytic function of SOS1. Notably, the KRAS-G12D mutation (KRAS-D12) significantly accelerates the rate of GDP extraction. The molecular basis of this enhancement are attributed to hydrogen bonding interactions between the mutant residue Asp12 and a positively charged pocket in the intrinsically disordered region (residues 807-818), comprising Ser807, Trp809, Thr810, and Lys811. These findings provide novel insights into SOS1-KRAS interactions and offer a foundation for developing anti-cancer strategies aimed at disrupting these mechanisms.
Collapse
Affiliation(s)
- Zheyao Hu
- Department of Physics, Polytechnic University of Catalonia-Barcelona Tech, B4-B5 Northern Campus UPC, Barcelona, 08034, Catalonia, Spain
| | - Jordi Martí
- Department of Physics, Polytechnic University of Catalonia-Barcelona Tech, B4-B5 Northern Campus UPC, Barcelona, 08034, Catalonia, Spain.
| |
Collapse
|
30
|
Chen L, Ning J, Linghu L, Tang J, Liu N, Long Y, Sun J, Lv C, Shi Y, Tao T, Xiao D, Cao Y, Wang X, Liu S, Li G, Zhang B, Tao Y. USP13 facilitates a ferroptosis-to-autophagy switch by activation of the NFE2L2/NRF2-SQSTM1/p62-KEAP1 axis dependent on the KRAS signaling pathway. Autophagy 2025; 21:565-582. [PMID: 39360581 PMCID: PMC11849926 DOI: 10.1080/15548627.2024.2410619] [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: 09/01/2023] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024] Open
Abstract
Macroautophagy/autophagyis a lysosomal-regulated degradation process that participates incellular stress and then promotes cell survival or triggers celldeath. Ferroptosis was initially described as anautophagy-independent, iron-regulated, nonapoptotic cell death.However, recent studies have revealed that autophagy is positivelyassociated with sensitivity to ferroptosis. Nonetheless, themolecular mechanisms by which these two types of regulated cell death(RCD) modulate each other remain largely unclear. Here, we screened85 deubiquitinating enzymes (DUBs) and found that overexpression ofUSP13 (ubiquitin specific peptidase 13) could significantlyupregulate NFE2L2/NRF2 (NFE2 like bZIP transcription factor 2)protein levels. In addition, in 39 cases of KRAS-mutated lungadenocarcinoma (LUAD), we found that approximately 76% of USP13overexpression is positively correlated with NFE2L2 overexpression.USP13 interacts with and catalyzes the deubiquitination of thetranscription factor NFE2L2. Additionally, USP13 depletion promotesan autophagy-to-ferroptosis switch invitro andin xenograft tumor mouse models, through the activation of theNFE2L2-SQSTM1/p62 (sequestosome 1)-KEAP1 axis in KRAS mutant cellsand tumor tissues. Hence, targeting USP13 effectively switchedautophagy-to-ferroptosis, thereby inhibiting KRAS (KRASproto-oncogene, GTPase) mutant LUAD, suggesting the therapeuticpromise of combining autophagy and ferroptosis in the KRAS-mutantLUAD.Abbreviation: ACSL4: acyl-CoA synthetase long-chain family member 4; ACTB: actin beta; AL: autolysosomes; AP: autophagosomes; BCL2L1/BCL-xL: BCL2 like 1; CCK8: Cell Counting Kit-8; CQ: chloroquine; CUL3: cullin 3; DMSO: dimethyl sulfoxide; DOX: doxorubicin; DUB: deubiquitinating enzyme; Ferr-1: ferrostatin-1; GPX4: glutathione peroxidase 4; GSEA: gene set enrichment analysis; 4HNE: 4-hydroxynonenal; IKE: imidazole ketone erastin; KEAP1: kelch like ECH associated protein 1; KRAS: KRAS proto-oncogene, GTPase; LCSC: lung squamous cell carcinoma; IF: immunofluorescence; LUAD: lung adenocarcinoma; Lys05: Lys01 trihydrochloride; MAPK1/ERK2/p42: mitogen-activated protein kinase 1; MAPK3/ERK1/p44; MTOR: mechanistic target of rapamycin kinase; NFE2L2/NRF2: NFE2 like bZIP transcription factor, 2; NQO1: NAD(P)H quinone dehydrogenase 1; PG: phagophore; RCD: regulated cell death; RAPA: rapamycin; ROS: reactive oxygen species; SLC7A11/xCT: solute carrier family 7 member 11; SQSTM1/p62: sequestosome 1; TEM: transmission electron microscopy; TUBB/beta-tubulin: tubulin, beta; UPS: ubiquitin-proteasome system; USP13: ubiquitin specific peptidase 13.
Collapse
Affiliation(s)
- Ling Chen
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Jieling Ning
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Department of Histology and Embryology, School of Basic Medicine, Central South University, Changsha, China
| | - Li Linghu
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Jun Tang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Na Liu
- Department of Neurosurgery, Postdoctoral Research Workstation, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yao Long
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Jingyue Sun
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Cairui Lv
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Ying Shi
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Tania Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Desheng Xiao
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
| | - Ya Cao
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Xiang Wang
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guangjian Li
- Department of Thoracic Surgery I, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Kunming, China
| | - Bin Zhang
- Department of Histology and Embryology, School of Basic Medicine, Central South University, Changsha, China
| | - Yongguang Tao
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, NHC Key Laboratory of Carcinogenesis, Cancer Research Institute, Central South University, Changsha, Hunan, China
- Department of Pathology, School of Basic Medicine, Central South University, Changsha, China
- Department of Thoracic Surgery, Hunan Key Laboratory of Early Diagnosis and Precision Therapy in Lung Cancer, Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
31
|
Daya T, Breytenbach A, Gu L, Kaur M. Cholesterol metabolism in pancreatic cancer and associated therapeutic strategies. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159578. [PMID: 39542394 DOI: 10.1016/j.bbalip.2024.159578] [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: 07/24/2024] [Revised: 10/31/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Pancreatic cancer remains one of the most lethal cancers due to late diagnosis and high chemoresistance. Despite recent progression in the development of chemotherapies, immunotherapies, and potential nanoparticles-based approaches, the success rate of therapeutic response is limited which is further compounded by cancer drug resistance. Understanding of emerging biological and molecular pathways causative of pancreatic cancer's aggressive and chemoresistance is vital to improve the effectiveness of existing therapeutics and to develop new therapies. One such under-investigated and relatively less explored area of research is documenting the effect that lipids, specifically cholesterol, and its metabolism, impose on pancreatic cancer. Dysregulated cholesterol metabolism has a profound role in supporting cellular proliferation, survival, and promoting chemoresistance and this has been well established in various other cancers. Thus, we aimed to provide an in-depth review focusing on the significance of cholesterol metabolism in pancreatic cancer and relevant genes at play, molecular processes contributing to cellular cholesterol homeostasis, and current research efforts to develop new cholesterol-targeting therapeutics. We highlight the caveats, weigh in different experimental therapeutic strategies, and provide possible suggestions for future research highlighting cholesterol's importance as a therapeutic target against pancreatic cancer resistance and cancer progression.
Collapse
Affiliation(s)
- Tasvi Daya
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Andrea Breytenbach
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Liang Gu
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa
| | - Mandeep Kaur
- School of Molecular and Cell Biology, University of the Witwatersrand, Private Bag 3, WITS, 2050 Johannesburg, South Africa.
| |
Collapse
|
32
|
Deck SL, Xu M, Milano SK, Cerione RA. Revealing Functional Hotspots: Temperature-Dependent Crystallography of K-RAS Highlights Allosteric and Druggable Sites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.639303. [PMID: 40060414 PMCID: PMC11888411 DOI: 10.1101/2025.02.27.639303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
K-RAS mutations drive oncogenesis in multiple cancers, yet the lack of druggable sites has long hindered therapeutic development. Here, we use multi-temperature X-ray crystallography (MT-XRC) to capture functionally relevant K-RAS conformations across a temperature gradient, spanning cryogenic to physiological and even "fever" conditions, and show how cryogenic conditions may obscure key dynamic states as targets for new drug development. This approach revealed a temperature-dependent conformational landscape of K-RAS, shedding light on the dynamic nature of key regions. We identified significant conformational changes occurring at critical sites, including known allosteric and drug-binding pockets, which were hidden under cryogenic conditions but later discovered to be critically important for drug-protein interactions and inhibitor design. These structural changes align with regions previously highlighted by large-scale mutational studies as functionally significant. However, our MT-XRC analysis provides precise structural snapshots, capturing the exact conformations of these potentially important allosteric sites in unprecedented detail. Our findings underscore the necessity of advancing tools like MT-XRC to visualize conformational transitions that may be important in signal propagation which are missed by standard cryogenic XRC and to address hard-to-drug targets through rational drug design. This approach not only provides unique structural insights into K-RAS signaling events and identifies new potential sites to target with drug candidates but also establishes a powerful framework for discovering therapeutic opportunities against other challenging drug targets.
Collapse
Affiliation(s)
- Samuel L Deck
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| | - Megan Xu
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| | - Shawn K Milano
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| | - Richard A Cerione
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853
| |
Collapse
|
33
|
Huang D, Manoni F, Sun Z, Liu R, Allen JR, Banerjee A, Cee VJ, Eshon J, Frohn MJ, Kaller MR, Lee H, Li C, Li X, Lopez P, Ma V, Medina JM, Mohr C, Mukhina OA, Pickrell AJ, Stellwagen J, Wu W, Zhang W, Zhu K, Dahal UP, Hu LA, Leavitt M, Li W, Li Y, Ma Y, Rex K, Saiki AY, Wang P, Sun Y, Dai D, Tamayo NA, Lanman BA. Identification of Structurally Novel KRAS G12C Inhibitors through Covalent DNA-Encoded Library Screening. J Med Chem 2025; 68:4801-4817. [PMID: 39930787 PMCID: PMC11873997 DOI: 10.1021/acs.jmedchem.4c03071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/28/2025]
Abstract
Covalent inhibition of the KRASG12C oncoprotein has emerged as a promising therapeutic approach for the treatment of nonsmall cell lung cancer (NSCLC). The identification of KRASG12C inhibitors has typically relied on the high-throughput screening (HTS) of libraries of cysteine-reactive small molecules or on the attachment of cysteine-reactive warheads to noncovalent binders of KRAS. Such screening approaches have historically been limited in the size and diversity of molecules that could be effectively screened. DNA-encoded library (DEL) screening has emerged as a promising approach to accelerate the preparation and screening of incredibly large and diverse chemical libraries. Here, we describe the design and synthesis of a covalent DEL to screen ∼16 million compounds against KRASG12C. We additionally describe the hit identification, validation, and structure-based optimization that culminated in the identification of a series of structurally novel, potent, and selective covalent inhibitors of KRASG12C with good pharmacokinetic profiles and promising in vivo pharmacodynamic effects.
Collapse
Affiliation(s)
- David Huang
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Francesco Manoni
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Zhen Sun
- Department
of Therapeutic Discovery, Amgen Asia R&D Center, Amgen Research, 4560
Jinke Road, Pudong, Shanghai 201210, P. R. China
| | - Rongfeng Liu
- Department
of Therapeutic Discovery, Amgen Asia R&D Center, Amgen Research, 4560
Jinke Road, Pudong, Shanghai 201210, P. R. China
| | - Jennifer R. Allen
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Abhisek Banerjee
- Syngene
Amgen Research & Development Center (SARC), Syngene International Ltd., Biocon Park, Jigani Link Road, Bengaluru 560099, India
| | - Victor J. Cee
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Josephine Eshon
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Michael J. Frohn
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Matthew R. Kaller
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Heejun Lee
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Cui Li
- Department
of Therapeutic Discovery, Amgen Asia R&D Center, Amgen Research, 4560
Jinke Road, Pudong, Shanghai 201210, P. R. China
| | - Xun Li
- Department
of Therapeutic Discovery, Amgen Asia R&D Center, Amgen Research, 4560
Jinke Road, Pudong, Shanghai 201210, P. R. China
| | - Patricia Lopez
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Vu Ma
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Jose M. Medina
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Christopher Mohr
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Olga A. Mukhina
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Alexander J. Pickrell
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - John Stellwagen
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Wenting Wu
- Department
of Therapeutic Discovery, Amgen Asia R&D Center, Amgen Research, 4560
Jinke Road, Pudong, Shanghai 201210, P. R. China
| | - Wenhan Zhang
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Kai Zhu
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Upendra P. Dahal
- Department
of Pharmacokinetics and Drug Metabolism, Amgen Research, 750
Gateway Boulevard, Suite 100, South San Francisco, California 94080, United States
| | - Liaoyuan A. Hu
- Department
of Therapeutic Discovery, Amgen Asia R&D Center, Amgen Research, 4560
Jinke Road, Pudong, Shanghai 201210, P. R. China
| | - Monica Leavitt
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Wencui Li
- Department
of Therapeutic Discovery, Amgen Asia R&D Center, Amgen Research, 4560
Jinke Road, Pudong, Shanghai 201210, P. R. China
| | - Yu Li
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Yingli Ma
- Department
of Therapeutic Discovery, Amgen Asia R&D Center, Amgen Research, 4560
Jinke Road, Pudong, Shanghai 201210, P. R. China
| | - Karen Rex
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Anne Y. Saiki
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Paul Wang
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Yaping Sun
- Department
of Therapeutic Discovery, Amgen Asia R&D Center, Amgen Research, 4560
Jinke Road, Pudong, Shanghai 201210, P. R. China
| | - Dongcheng Dai
- Department
of Therapeutic Discovery, Amgen Asia R&D Center, Amgen Research, 4560
Jinke Road, Pudong, Shanghai 201210, P. R. China
| | - Nuria A. Tamayo
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| | - Brian A. Lanman
- Department
of Medicinal Chemistry, Discovery Scale-Up & Development, Oncology Research, Structural Biology, Computational and
Data Sciences, and Lead Discovery & Characterization, Amgen Research, One
Amgen Center Drive, Thousand Oaks, California 91320, United States
| |
Collapse
|
34
|
Nussinov R, Yavuz BR, Jang H. Allostery in Disease: Anticancer Drugs, Pockets, and the Tumor Heterogeneity Challenge. J Mol Biol 2025:169050. [PMID: 40021049 DOI: 10.1016/j.jmb.2025.169050] [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: 01/31/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025]
Abstract
Charting future innovations is challenging. Yet, allosteric and orthosteric anticancer drugs are undergoing a revolution and taxing unresolved dilemmas await. Among the imaginative innovations, here we discuss cereblon and thalidomide derivatives as a means of recruiting neosubstrates and their degradation, allosteric heterogeneous bifunctional drugs like PROTACs, drugging phosphatases, inducers of targeted posttranslational protein modifications, antibody-drug conjugates, exploiting membrane interactions to increase local concentration, stabilizing the folded state, and more. These couple with harnessing allosteric cryptic pockets whose discovery offers more options to modulate the affinity of orthosteric, active site inhibitors. Added to these are strategies to counter drug resistance through drug combinations co-targeting pathways to bypass signaling blockades. Here, we discuss on the molecular and cellular levels, such inspiring advances, provide examples of their applications, their mechanisms and rational. We start with an overview on difficult to target proteins and their properties-rarely, if ever-conceptualized before, discuss emerging innovative drugs, and proceed to the increasingly popular allosteric cryptic pockets-their advantages-and critically, issues to be aware of. We follow with drug resistance and in-depth discussion of tumor heterogeneity. Heterogeneity is a hallmark of highly aggressive cancers, the core of drug resistance unresolved challenge. We discuss potential ways to target heterogeneity by predicting it. The increase in experimental and clinical data, computed (cell-type specific) interactomes, capturing transient cryptic pockets, learned drug resistance, workings of regulatory mechanisms, heterogeneity, and resistance-based cell signaling drug combinations, assisted by AI-driven reasoning and recognition, couple with creative allosteric drug discovery, charting future innovations.
Collapse
Affiliation(s)
- Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, the United States of America; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, the United States of America; Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Bengi Ruken Yavuz
- Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, the United States of America
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, the United States of America; Cancer Innovation Laboratory, National Cancer Institute at Frederick, Frederick, MD 21702, the United States of America
| |
Collapse
|
35
|
Uniyal P, Kashyap VK, Behl T, Parashar D, Rawat R. KRAS Mutations in Cancer: Understanding Signaling Pathways to Immune Regulation and the Potential of Immunotherapy. Cancers (Basel) 2025; 17:785. [PMID: 40075634 PMCID: PMC11899378 DOI: 10.3390/cancers17050785] [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: 12/06/2024] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
The Kirsten rat sarcoma viral oncogene homologue (KRAS) mutation is one of the most prevailing mutations in various tumors and is difficult to cure. Long-term proliferation in carcinogenesis is primarily initiated by oncogenic KRAS-downstream signaling. Recent research suggests that it also activates the autocrine effect and interplays the tumor microenvironment (TME). Here, we discuss the emerging research, including KRAS mutations to immune evasion in TME, which induce immunological modulation that promotes tumor development. This review gives an overview of the existing knowledge of the underlying connection between KRAS mutations and tumor immune modulation. It also addresses the mechanisms to reduce the effect of oncogenes on the immune system and recent advances in clinical trials for immunotherapy in KRAS-mutated cancers.
Collapse
Affiliation(s)
- Priyanka Uniyal
- Department of Pharmaceutical Technology, School of Health Sciences and Technology, UPES, Dehradun 248007, India;
| | - Vivek Kumar Kashyap
- Division of Cancer Immunology and Microbiology, Medicine, and Oncology Integrated Service Unit, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA;
- South Texas Center of Excellence in Cancer Research (ST-CECR), School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Tapan Behl
- Amity School of Pharmaceutical Sciences, Amity University, Mohali 140306, India;
| | - Deepak Parashar
- Division of Hematology & Oncology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Ravi Rawat
- Department of Pharmaceutical Technology, School of Health Sciences and Technology, UPES, Dehradun 248007, India;
| |
Collapse
|
36
|
Liu Z, Li Y, Wang S, Wang Y, Sui M, Liu J, Chen P, Wang J, Zhang Y, Dang C, Hou P. Genome-wide CRISPR screening identifies PHF8 as an effective therapeutic target for KRAS- or BRAF-mutant colorectal cancers. J Exp Clin Cancer Res 2025; 44:70. [PMID: 40001243 PMCID: PMC11853609 DOI: 10.1186/s13046-025-03338-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Mutations in KRAS and BRAF genes are prevalent in colorectal cancer (CRC), which strikingly promote tumorigenesis and lead to poor response to a variety of treatments including immunotherapy by activating the MAPK/ERK pathway. Thus, there is an urgent need to discover effective therapeutic targets and strategies. METHODS CRISPR-Cas9 lentiviral knockout library was used to screen the suppressors of anti-PD1 immunotherapy. Bioinformatic analysis was used to analyze the correlation between PHF8 expression and immune indicators in CRC. In vitro and in vivo experiments were utilized to determine the effects of PHF8 on the immune indexes and malignant phenotypes of CRC cells. qRT-PCR, western blotting, immunohistochemical (IHC) staining, and chromatin immunoprecipitation (ChIP)-qPCR assays were used to determine the regulatory effects of PHF8 on PD-L1, KRAS, BRAF, and c-Myc and the regulatory effect c-Myc/miR-22-3p signaling axis on PHF8 expression in CRC cells. RESULTS This study identified histone lysine demethylase PHF8 as a negative regulator for the efficacy of anti-PD1 therapy and found that it was highly expressed in CRCs and strongly associated with poor patient survival. Functional studies showed that PHF8 played an oncogenic role in KRAS- or BRAF-mutant CRC cells, but not in wild-type ones. Mechanistically, PHF8 up-regulated the expression of PD-L1, KRAS, BRAF, and c-Myc by increasing the levels of transcriptional activation marks H3K4me3 and H3K27ac and decreasing the levels of transcriptional repression mark H3K9me2 within their promoter regions, promoting immune escape and tumor progression. Besides, our data also demonstrated that PHF8 was up-regulated by the c-Myc/miR-22-3p signaling axis to form a positive feedback loop. Targeting PHF8 substantially improved the efficacy of anti-PD1 therapy and inhibited the malignant phenotypes of KRAS- or BRAF-mutant CRC cells. CONCLUSION Our data demonstrate that PHF8 may be an effective therapeutic target for KRAS- or BRAF-mutant CRCs.
Collapse
Affiliation(s)
- Zhao Liu
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Yiqi Li
- Department of General Practice, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, P.R. China
| | - Simeng Wang
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Yubo Wang
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Mengjun Sui
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Jiaxin Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Pu Chen
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Jianling Wang
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Yuchen Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China
| | - Chengxue Dang
- Department of Surgical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China.
| | - Peng Hou
- Department of Endocrinology and International Joint Research Center for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P.R. China.
| |
Collapse
|
37
|
Gupta T, Murtaza M. Advancing Targeted Therapies in Pancreatic Cancer: Leveraging Molecular Aberrations for Therapeutic Success. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025:S0079-6107(25)00016-1. [PMID: 39988056 DOI: 10.1016/j.pbiomolbio.2025.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 02/03/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Pancreatic cancer is one of the most deadly with poor prognosis and overall survival rate due to the dense stroma in the tumors which often is challenging for the delivery of drug to penetrate deep inside the tumor bed and usually results in the progression of cancer. The conventional treatment such as chemotherapy, radiotherapy or surgery shows a minimal benefit in the survival due to the drug resistance, poor penetration, less radiosensitivity or recurrence of tumor. There is an urgent demand to develop molecular- level targeted therapies to achieve therapeutic efficacy in the pancreatic ductal adenocarcinoma (PDAC) patients. The precision oncology focuses on the unique attributes of the patient such as epigenome, proteome, genome, microbiome, lifestyle and diet habits which contributes to promote oncogenesis. The targeted therapy helps to target the mutated proteins responsible for controlling growth, division and metastasis of tumor in the cancer cells. It is very important to consider all the attributes of the patient to provide the suitable personalized treatment to avoid any severe side effects. In this review, we have laid emphasis on the precision medicine; the utmost priority is to improve the survival of cancer patients by targeting molecular mutations through transmembrane proteins, inhibitors, signaling pathways, immunotherapy, gene therapy or the use of nanocarriers for the delivery at the tumor site. It will become beneficial therapeutic window to be considered for the advanced stage pancreatic cancer patients to prolong their survival rate.
Collapse
Affiliation(s)
- Tanvi Gupta
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, 704, Taiwan.
| | - Mohd Murtaza
- Fermentation & Microbial Biotechnology Division, CSIR- Indian Institute of Integrative Medicine, Jammu, 180016, India.
| |
Collapse
|
38
|
Er Özilhan S, Efil SC, Çanakçı D, Ağaçkıran Y, Şener Dede D, Onak Kandemir N, Doğan M, Ünal TDK, Kıran MM, Kayaçetin S, Balta H, Tatlı Doğan H. Correlation of PD-L1 and HIF-1 Alpha Expression with KRAS Mutation and Clinicopathological Parameters in Non-Small Cell Lung Cancer. Curr Issues Mol Biol 2025; 47:121. [PMID: 39996842 PMCID: PMC11854292 DOI: 10.3390/cimb47020121] [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: 12/22/2024] [Revised: 02/11/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025] Open
Abstract
Background: Lung cancer remains the leading cause of cancer-related deaths worldwide, with non-small cell lung carcinomas (NSCLCs) comprising the majority of cases. Among the common driver mutations, KRAS plays a critical role in guiding treatment strategies. This study evaluates the expression of programmed death-ligand 1 (PD-L1) and hypoxia-inducible factor 1-alpha (HIF-1α) in KRAS-mutant NSCLCs and investigates their associations with clinicopathological findings. Methods: A total of 85 cases with KRAS mutations were analyzed. Immunohistochemical staining for HIF-1α and PD-L1 was performed, and their relationships with mutation status and prognostic variables were assessed. Results: A significant correlation was identified between HIF-1α expression and PD-L1 expression in tumor cells. While the KRAS G12C mutation was not significantly associated with HIF-1α expression in tumor cells, it demonstrated a notable relationship with HIF-1α expression in the tumor microenvironment and PD-L1 expression. However, PD-L1 and HIF-1α expression did not significantly influence overall survival outcomes. Conclusions: Expression of PD-L1 was positively correlated with HIF-1α, which may provide evidence for a novel therapy targeting PD-L1 and HIF-1α in NSCLC. Further comprehensive studies are warranted to elucidate the prognostic implications of tumor-microenvironment and mutation interactions.
Collapse
Affiliation(s)
- Seda Er Özilhan
- Department of Pathology, Ankara Bilkent City Hospital, Ankara 06800, Turkey
| | - Safa Can Efil
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara 06800, Turkey
| | - Doğukan Çanakçı
- Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara 06800, Turkey
| | - Yetkin Ağaçkıran
- Department of Pathology, Ankara Atatürk Sanatoryum Hospital, Health Sciences University, Ankara 06290, Turkey
| | - Didem Şener Dede
- Department of Medical Oncology, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara 06800, Turkey
| | | | - Mehmet Doğan
- Department of Pathology, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara 06800, Turkey
| | - Tuba Dilay Kökenek Ünal
- Department of Pathology, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara 06800, Turkey
| | - Merve Meryem Kıran
- Department of Pathology, Ankara Bilkent City Hospital, Ankara 06800, Turkey
| | - Serra Kayaçetin
- Department of Pathology, Ankara Bilkent City Hospital, Health Sciences University, Ankara 06290, Turkey
| | - Hilal Balta
- Department of Pathology, Ankara Bilkent City Hospital, Health Sciences University, Ankara 06290, Turkey
| | - Hayriye Tatlı Doğan
- Department of Pathology, Faculty of Medicine, Ankara Yıldırım Beyazıt University, Ankara 06800, Turkey
| |
Collapse
|
39
|
Li A, Li S, Wang P, Dang C, Fan X, Chen M, Liu D, Li F, Liu H, Zhang W, Wang Y, Wang Y. Design, Structure Optimization, and Preclinical Characterization of JAB-21822, a Covalent Inhibitor of KRAS G12C. J Med Chem 2025; 68:2422-2436. [PMID: 39875337 DOI: 10.1021/acs.jmedchem.4c02939] [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: 01/30/2025]
Abstract
KRAS is the most frequently mutated driver oncogene in human cancer, and KRASG12C mutation is commonly found in non-small-cell lung cancer (NSCLC), colorectal cancer (CRC), and pancreatic ductal adenocarcinoma (PDAC). Inhibitors that covalently modify the mutated codon 12 cysteine have completed proof-of-concept studies in the clinic. Here, we describe structure-based design and cocrystal-aided drug optimization of a series of compounds with the 1,8-naphthyridine-3-carbonitrile scaffold. Biopharmaceutical optimization of the resulting leads to improve the solubility of the compounds and block the possible metabolic hotspots led to the identification of JAB-21822, a covalent KRASG12C inhibitor with high potency and excellent cross-species pharmacokinetic properties. JAB-21822 has finished the pivotal Phase II clinical trials in NSCLC, and a new drug application was submitted to the National Medical Products Administration in 2024.
Collapse
Affiliation(s)
- Amin Li
- Medicinal Chemistry Department, Jacobio Pharmaceuticals Group Co., Ltd., Beijing 100176, P. R. China
| | - Sujing Li
- Medicinal Chemistry Department, Jacobio Pharmaceuticals Group Co., Ltd., Beijing 100176, P. R. China
| | - Peng Wang
- Biology Department, Jacobio Pharmaceuticals Group Co., Ltd., Beijing100176, P. R. China
| | - Chaojie Dang
- Process Development Department, Jacobio Pharmaceuticals Group Co., Ltd., Beijing100176, P. R. China
| | - Xinrui Fan
- Medicinal Chemistry Department, Jacobio Pharmaceuticals Group Co., Ltd., Beijing 100176, P. R. China
| | - Mengran Chen
- Medicinal Chemistry Department, Jacobio Pharmaceuticals Group Co., Ltd., Beijing 100176, P. R. China
| | - Dan Liu
- Biology Department, Jacobio Pharmaceuticals Group Co., Ltd., Beijing100176, P. R. China
| | - Fu Li
- Medicinal Chemistry Department, Jacobio Pharmaceuticals Group Co., Ltd., Beijing 100176, P. R. China
| | - Huan Liu
- Process Development Department, Jacobio Pharmaceuticals Group Co., Ltd., Beijing100176, P. R. China
| | - Wei Zhang
- Hits Discovery Department, Jacobio Pharmaceuticals Group Co., Ltd., Beijing100176, P. R. China
| | - Yanping Wang
- Pharmacology Department, Jacobio Pharmaceuticals Group Co., Ltd., Beijing100176, P. R. China
| | - Yinxiang Wang
- Chief executive officer, Jacobio Pharmaceuticals Group Co., Ltd., Beijing100176, P. R. China
| |
Collapse
|
40
|
Avădănei ER, Căruntu ID, Nucă I, Balan RA, Lozneanu L, Giusca SE, Pricope DL, Dascalu CG, Amalinei C. KRAS Mutation Status in Relation to Clinicopathological Characteristics of Romanian Colorectal Cancer Patients. Curr Issues Mol Biol 2025; 47:120. [PMID: 39996841 PMCID: PMC11854687 DOI: 10.3390/cimb47020120] [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: 11/17/2024] [Revised: 02/06/2025] [Accepted: 02/09/2025] [Indexed: 02/26/2025] Open
Abstract
Our study's aim was to evaluate the clinicopathological profile of colorectal cancer (CRC) patients from North-East Romania in relation to the Kirsten rat sarcoma viral oncogene homolog (KRAS). We designed a retrospective study on 108 CRC patients using the fully automated real-time PCR-based molecular testing system, IdyllaTMKRAS Mutation Test (Biocartis, Mechelen, Belgium). Of the patients, 64 (59.3%) were men and 62 (57.4%) were older than the group average, with left bowel location in 38 cases (35.2%), adenocarcinoma NOS in 102 cases (94.4%), mixed histological pattern in 65 cases (60.2%), T3 in 60 patients (55.6%), N2 in 46 patients (42.6%), and 7-12 tumour buds registered in 58 tumours (53.7%). A total of 54 tumour samples (50%) showed KRAS mutation. Statistical comparative analyses associated KRAS mutations with the histopathological pattern (p = 0.018), tumour grade (p = 0.030), depth of invasion (pT) (p < 0.001), lymph node involvement (pN) (p < 0.001), venous vascular invasion (p = 0.048), and tumour buds' number (p = 0.007). Our results demonstrate the relationship between KRAS mutation and clinicopathological features, with possible impact in clinical tumour stratification and therapeutic management.
Collapse
Affiliation(s)
- Elena-Roxana Avădănei
- Department of Morpho-Functional Sciences I-Histology, Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-D.C.); (R.A.B.); (L.L.); (S.-E.G.); (D.L.P.); (C.A.)
- Praxis Medical Investigation Laboratory, 35 Moara de Vant Street, 700376 Iasi, Romania;
| | - Irina-Draga Căruntu
- Department of Morpho-Functional Sciences I-Histology, Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-D.C.); (R.A.B.); (L.L.); (S.-E.G.); (D.L.P.); (C.A.)
- Romanian Medical Science Academy, 1 I.C. Bratianu Boulevard, 030171 Bucharest, Romania
| | - Irina Nucă
- Praxis Medical Investigation Laboratory, 35 Moara de Vant Street, 700376 Iasi, Romania;
- Department of Mother and Child Medicine-Genetics, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania
| | - Raluca Anca Balan
- Department of Morpho-Functional Sciences I-Histology, Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-D.C.); (R.A.B.); (L.L.); (S.-E.G.); (D.L.P.); (C.A.)
| | - Ludmila Lozneanu
- Department of Morpho-Functional Sciences I-Histology, Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-D.C.); (R.A.B.); (L.L.); (S.-E.G.); (D.L.P.); (C.A.)
- Department of Pathology, “Sf. Spiridon” Clinical Emergency County Hospital, 1 Independentei Street, 700111 Iasi, Romania
| | - Simona-Eliza Giusca
- Department of Morpho-Functional Sciences I-Histology, Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-D.C.); (R.A.B.); (L.L.); (S.-E.G.); (D.L.P.); (C.A.)
| | - Diana Lavinia Pricope
- Department of Morpho-Functional Sciences I-Histology, Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-D.C.); (R.A.B.); (L.L.); (S.-E.G.); (D.L.P.); (C.A.)
| | - Cristina Gena Dascalu
- Department of Preventive Medicine and Interdisciplinarity-Medical Informatics and Biostatistics, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania;
| | - Cornelia Amalinei
- Department of Morpho-Functional Sciences I-Histology, Pathology, “Grigore T. Popa” University of Medicine and Pharmacy, 16 University Street, 700115 Iasi, Romania; (I.-D.C.); (R.A.B.); (L.L.); (S.-E.G.); (D.L.P.); (C.A.)
- Department of Histopathology, Institute of Legal Medicine, 4 Buna Vestire Street, 700455 Iasi, Romania
| |
Collapse
|
41
|
López-Estévez AM, Carrascal-Miniño A, Torres D, Alonso MJ, de Rosales RTM, Pellico J. Biodistribution of 89Zr-Radiolabeled Nanoassemblies for Monoclonal Antibody Delivery Revealed through In Vivo PET Imaging. ACS OMEGA 2025; 10:4763-4773. [PMID: 39959112 PMCID: PMC11822718 DOI: 10.1021/acsomega.4c09823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/08/2025] [Accepted: 01/16/2025] [Indexed: 02/18/2025]
Abstract
Despite the outstanding performance of monoclonal antibodies (mAbs) in the clinic, their full potential has been hindered due to their inability to cross cell membranes and therefore reach intracellular targets. The use of nanotechnology to deliver mAbs to intracellular domains has been highlighted as a strategy with high potential. Working toward this goal, we have recently developed and validated palmitoyl hyaluronate (HAC16)-based nanoassemblies (HANAs), a novel technology for the intracellular delivery of mAbs in Kirsten Rat Sarcoma Virus (KRAS)-mutated tumors, one of the most prevalent and a challenging intracellular oncoprotein. Despite their success, the pharmacokinetics and biodistribution of these delivery vehicles are still unknown due to their chemical complexity, a challenge common to a large proportion of drug delivery nanomedicines. To support further development and clinical translation, we present an efficient radiolabeling approach with the positron emitter zirconium-89 (89Zr) for the in vivo evaluation of HANAs by whole-body PET imaging. Additionally, we assessed the impact of PEGylation and size modulation on the biodistribution profile of mAbs using 89Zr-radiolabeled PEGylated and non-PEGylated HANAs. Our PET imaging results demonstrated that HANAs significantly modify the pharmacokinetics and biodistribution of the 89Zr-mAb. Furthermore, we established that the biodistribution of HANAs can be conveniently modulated by introducing PEG polymers on the surface, facilitating customization for cancer applications. This versatile radiolabeling strategy provides a facile approach for the in vivo evaluation of complex nanoformulations loaded with mAbs, in a quantitative manner with high sensitivity.
Collapse
Affiliation(s)
- Ana M. López-Estévez
- Center
for Research in Molecular Medicine and Chronic Diseases (CiMUS), Health
Research Institute of Santiago de Compostela, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Department
of Pharmacology, Pharmacy and Pharmaceutical Technology, School of
Pharmacy, University of Santiago de Compostela, 15782 Santiago
de Compostela, Spain
| | - Amaia Carrascal-Miniño
- School
of Biomedical Engineering & Imaging Sciences, King’s College
London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Dolores Torres
- Department
of Pharmacology, Pharmacy and Pharmaceutical Technology, School of
Pharmacy, University of Santiago de Compostela, 15782 Santiago
de Compostela, Spain
| | - María José Alonso
- Center
for Research in Molecular Medicine and Chronic Diseases (CiMUS), Health
Research Institute of Santiago de Compostela, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Department
of Pharmacology, Pharmacy and Pharmaceutical Technology, School of
Pharmacy, University of Santiago de Compostela, 15782 Santiago
de Compostela, Spain
| | - Rafael T. M. de Rosales
- School
of Biomedical Engineering & Imaging Sciences, King’s College
London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Juan Pellico
- School
of Biomedical Engineering & Imaging Sciences, King’s College
London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| |
Collapse
|
42
|
Kunimasa K, Aohara D, Nishino K. Driver mutation detected in cerebrospinal fluid despite negative liquid biopsy results. Discov Oncol 2025; 16:145. [PMID: 39928192 PMCID: PMC11811313 DOI: 10.1007/s12672-025-01931-7] [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: 10/27/2024] [Accepted: 02/04/2025] [Indexed: 02/11/2025] Open
Abstract
We report a case where a KRAS G12V driver mutation was identified in cerebrospinal fluid (CSF) but not in peripheral blood cell-free DNA (cfDNA) in a patient with advanced lung adenocarcinoma and significant central nervous system involvement. A 67-year-old man presented with hemoptysis and was diagnosed with stage IVB TTF-1-positive lung adenocarcinoma with brain and bone metastases. Standard chemotherapy was ineffective. While cfDNA analysis detected only a RAD21 mutation, CSF analysis revealed the KRAS G12V mutation. Despite the identification, no effective targeted therapy was available. This case highlights that CSF may be more suitable than peripheral blood cfDNA for detecting driver mutations in patients with predominant CNS lesions.
Collapse
Affiliation(s)
- Kei Kunimasa
- Department of Thoracic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae Chuoku, Osaka, Osaka, 541-8567, Japan.
| | - Daisuke Aohara
- Department of Respiratory Medicine, Osaka General Hospital of West Japan Railway Company, Osaka, Japan
| | - Kazumi Nishino
- Department of Thoracic Oncology, Osaka International Cancer Institute, 3-1-69 Otemae Chuoku, Osaka, Osaka, 541-8567, Japan
| |
Collapse
|
43
|
Qin W, Liu Z, Huang M, Liang L, Gan Y, Huang Z, Huang J, Wei X. Recent Advances in Peptide Inhibitors Targeting Wild-Type Ras Protein Interactions in Cancer Therapy. Int J Mol Sci 2025; 26:1425. [PMID: 40003893 PMCID: PMC11855556 DOI: 10.3390/ijms26041425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/24/2025] [Accepted: 01/26/2025] [Indexed: 02/27/2025] Open
Abstract
Ras proteins are pivotal in the regulation of cell proliferation signals, and their dysregulation is intricately linked to the pathogenesis of various malignancies. Peptide inhibitors hold distinct advantages in targeting Ras proteins, attributable to their extensive binding domains, which result from the smooth surfaces of the proteins. The array of specific strategies includes the employment of full hydrocarbon chains, cyclic peptides, linear peptides, and N-terminal nucleation polypeptides. These methods effectively suppress the Ras signaling pathway through distinct mechanisms, highlighting their potential as anti-neoplastic agents. Moreover, cutting-edge methodologies, including the N-terminal aspartate nucleation strategy and the utilization of hydrocarbon-stapled peptides, are transforming the landscape of therapeutics aimed at Ras proteins. These innovations highlight the promise of peptide libraries and combinatorial chemistry in augmenting binding affinity, specificity, and cellular permeability, which are pivotal for the development of potent anti-cancer agents. The incorporation of dual therapeutic strategies, such as the synergy between peptide inhibitors and conventional chemotherapy or the use of radiotherapy enhancers, emerges as a compelling strategy to bolster the efficacy of cancer treatments targeting the Ras-MAPK pathway. Furthermore, recent studies have demonstrated that Ras-targeting stabilized peptides can amplify the radio-sensitivity of cancer cells, offering an innovative approach to enhance the efficacy of radiation therapy within cancer management.
Collapse
Affiliation(s)
- Weirong Qin
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China; (Z.L.); (M.H.); (L.L.); (Y.G.); (Z.H.); (J.H.)
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, Guangxi Medical University, Nanning 530021, China
- Key Laboratory of Biological Molecular Medicine Research (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Zijian Liu
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China; (Z.L.); (M.H.); (L.L.); (Y.G.); (Z.H.); (J.H.)
| | - Mingyu Huang
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China; (Z.L.); (M.H.); (L.L.); (Y.G.); (Z.H.); (J.H.)
| | - Lin Liang
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China; (Z.L.); (M.H.); (L.L.); (Y.G.); (Z.H.); (J.H.)
| | - Yuxin Gan
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China; (Z.L.); (M.H.); (L.L.); (Y.G.); (Z.H.); (J.H.)
| | - Zubei Huang
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China; (Z.L.); (M.H.); (L.L.); (Y.G.); (Z.H.); (J.H.)
| | - Jin Huang
- Pharmaceutical College, Guangxi Medical University, Nanning 530021, China; (Z.L.); (M.H.); (L.L.); (Y.G.); (Z.H.); (J.H.)
| | - Xiangzan Wei
- Key Laboratory of Biological Molecular Medicine Research (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning 530021, China
- State Key Laboratory of Chemical Oncogenomics, Guangdong Provincial Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen 518055, China
| |
Collapse
|
44
|
Chen R, He A, De Carvalho DD. Viral mimicry evasion: a new role for oncogenic KRAS mutations. Mol Oncol 2025; 19:271-274. [PMID: 39592415 PMCID: PMC11792985 DOI: 10.1002/1878-0261.13771] [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: 11/04/2024] [Revised: 11/11/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
"Viral mimicry" refers to the induction of an innate immune response and interferon signaling by endogenous stimuli such as double-stranded RNA (dsRNA). This response has been shown to have strong cancer therapeutic potential, including by enhancing the effectiveness of immune checkpoint inhibition (ICI) therapies, and may represent a tumor suppression mechanism that needs to be overcome for malignant transformation to proceed. In a recent study, Zhou and colleagues identify KRAS, a frequently mutated oncogene, as a negative regulator of dsRNA and viral mimicry in an ICI-resistant colorectal cancer model. Oncogenic KRASG12D mutations downregulate the RNA-binding protein DDX60 by activating the AKT signaling pathway, which inhibits STAT3, a critical transcription factor regulating DDX60 and other interferon-stimulated genes. Overexpression of DDX60, which competitively binds to dsRNA to prevent RISC-mediated degradation, or targeting of KRASG12D elevated dsRNA levels, resulting in viral mimicry activation and potentiation of ICI treatment. These results establish KRAS as a promising target to sensitize immune "cold" tumors to ICI therapy and demonstrate the potential role of oncogenic mutations in viral mimicry evasion during tumorigenesis.
Collapse
Affiliation(s)
- Raymond Chen
- Department of Medical BiophysicsUniversity of TorontoCanada
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoCanada
| | - Aobo He
- Department of Medical BiophysicsUniversity of TorontoCanada
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoCanada
| | - Daniel D. De Carvalho
- Department of Medical BiophysicsUniversity of TorontoCanada
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoCanada
| |
Collapse
|
45
|
Pandey D, Roy KK. Decoding KRAS dynamics: Exploring the impact of mutations and inhibitor binding. Arch Biochem Biophys 2025; 764:110279. [PMID: 39710177 DOI: 10.1016/j.abb.2024.110279] [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: 06/05/2024] [Revised: 11/25/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
KRAS (Kirsten rat sarcoma viral oncogene homologue), the most common mutated protein in human cancers, is the leading cause of morbidity and mortality. Before Sotorasib (AMG-510) was approved for non-small cell lung cancer treatment in 2020, the oncogenic KRAS mutations were believed to be non-druggable. High-resolution X-ray crystal structures of GDP-bound KRAS mutants with and without inhibitor are resolved and deposited in the Protein Data Bank (PDB). Nevertheless, to develop inhibitors targeting oncogenic KRAS mutants, understanding the dynamics of protein conformations and respective binding sites is crucial. In the present study, multiple molecular dynamics (MD) simulations were conducted on wild-type and mutant KRAS structures to understand how G12C or G12D mutations lead to the stabilization of the active state and how KRAS inhibitors lock the mutated conformations in their inactive state. The study found that the guanosine diphosphate (GDP)-bound KRAS mutants, G12C and G12D, were locked in the inactive state, in terms of stability, when the KRAS inhibitors, AMG-510 and MRTX1133, respectively, bind to the respective Switch-II (S-II) pocket. Covalent inhibitor AMG-510 locked the inactive GDP-bound KRASG12C mutant more efficiently when compared to the non-covalent inhibitor MRTX1133. The Cα atom distance between key highly dynamic amino acids from P-loop, Switch-I, and Switch-II domains, lying within 4 Å of the inhibitor, were stable in the KRAS mutant with bound inhibitors (AMG-510 or MRTX1133), but were varying largely in the absence of any inhibitor throughout the microsecond simulation. According to the per-residue energy decomposition results, S-II amino acids in inhibitor-free KRASG12C and KRASG12D mutants showed larger variations in energy values as compared to AMG-510-bound KRASG12C and MRTX1133-bound KRASG12D, respectively. For example, the inhibitor-free KRASG12C exhibited larger variations in energy values in the S-II residues, namely, Thr58, Gln61, Glu63, and Arg68, as compared to the AMG-510-bound KRASG12C. The study found that the higher stability of AMG-510 in torsion angles was due to its covalent nature of binding to the KRASG12C mutant. The S-II amino acids, namely, Thr58, Glu63, and Arg68 remained stable in AMG-510-bound KRASG12C. The study showed that AMG-510 binding significantly stabilizes the amino acids surrounding it, surpassing that of MRTX1133. The insights gained in the present study is expected to be useful in the design and development of new KRAS-targeted drugs.
Collapse
Affiliation(s)
- Divya Pandey
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, 248007, Uttarakhand, India
| | - Kuldeep K Roy
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, 248007, Uttarakhand, India.
| |
Collapse
|
46
|
Kramer‐Drauberg M, Petrini E, Mira A, Patrucco E, Scardaci R, Savinelli I, Wang H, Qiao K, Carrà G, Nokin M, Zhou Z, Westover KD, Santamaria D, Porporato PE, Ambrogio C. Oncogenic mutant KRAS inhibition through oxidation at cysteine 118. Mol Oncol 2025; 19:311-328. [PMID: 39838816 PMCID: PMC11793020 DOI: 10.1002/1878-0261.13798] [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: 06/09/2024] [Revised: 10/30/2024] [Accepted: 12/24/2024] [Indexed: 01/23/2025] Open
Abstract
Specific reactive oxygen species activate the GTPase Kirsten rat sarcoma virus (KRAS) by reacting with cysteine 118 (C118), leading to an electron transfer between C118 and nucleoside guanosine diphosphate (GDP), which causes the release of GDP. Here, we have mimicked permanent oxidation of human KRAS at C118 by replacing C118 with aspartic acid (C118D) in KRAS to show that oncogenic mutant KRAS is selectively inhibited via oxidation at C118, both in vitro and in vivo. Moreover, the combined treatment of hydrogen-peroxide-producing pro-oxidant paraquat and nitric-oxide-producing inhibitor N(ω)-nitro-l-arginine methyl ester selectively inhibits human mutant KRAS activity by inducing oxidization at C118. Our study shows for the first time the vulnerability of human mutant KRAS to oxidation, thereby paving the way to explore oxidation-based anti-KRAS treatments in humans.
Collapse
Affiliation(s)
- Maximilian Kramer‐Drauberg
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Ettore Petrini
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Alessia Mira
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Enrico Patrucco
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Rossella Scardaci
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Ilenia Savinelli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Haiyun Wang
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Keying Qiao
- School of Life Sciences and TechnologyTongji UniversityShanghaiChina
| | - Giovanna Carrà
- Department of Clinical and Biological SciencesUniversity of TorinoOrbassanoItaly
| | - Marie‐Julie Nokin
- Laboratory of Tumor and Development Biology, GIGA‐CancerUniversity of LiegeBelgium
| | - Zhiwei Zhou
- Department of BiochemistryThe University of Texas Southwestern Medical CenterDallasTXUSA
- Department of Radiation OncologyThe University of Texas Southwestern Medical CenterDallasTXUSA
| | - Kenneth D. Westover
- Department of BiochemistryThe University of Texas Southwestern Medical CenterDallasTXUSA
- Department of Radiation OncologyThe University of Texas Southwestern Medical CenterDallasTXUSA
| | - David Santamaria
- Molecular Mechanisms of Cancer Program, Centro de Investigación del CáncerCSIC‐Universidad de SalamancaSpain
| | - Paolo E. Porporato
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology CenterUniversity of TorinoItaly
| |
Collapse
|
47
|
Isermann T, Sers C, Der CJ, Papke B. KRAS inhibitors: resistance drivers and combinatorial strategies. Trends Cancer 2025; 11:91-116. [PMID: 39732595 DOI: 10.1016/j.trecan.2024.11.009] [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: 09/26/2024] [Revised: 11/20/2024] [Accepted: 11/22/2024] [Indexed: 12/30/2024]
Abstract
In 1982, the RAS genes HRAS and KRAS were discovered as the first human cancer genes, with KRAS later identified as one of the most frequently mutated oncogenes. Yet, it took nearly 40 years to develop clinically effective inhibitors for RAS-mutant cancers. The discovery in 2013 by Shokat and colleagues of a druggable pocket in KRAS paved the way to FDA approval of the first covalently binding KRASG12C inhibitors, sotorasib and adagrasib, in 2021 and 2022, respectively. However, rather than marking the end of a successful assault on the Mount Everest of cancer research, this landmark only revealed new challenges in RAS drug discovery. In this review, we highlight the progress on defining resistance mechanisms and developing combination treatment strategies to improve patient responses to KRAS therapies.
Collapse
Affiliation(s)
- Tamara Isermann
- Charité - Universitätsmedizin Berlin, Institute of Pathology, Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christine Sers
- Charité - Universitätsmedizin Berlin, Institute of Pathology, Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Channing J Der
- Charité - Universitätsmedizin Berlin, Institute of Pathology, Berlin, Germany; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bjoern Papke
- Charité - Universitätsmedizin Berlin, Institute of Pathology, Berlin, Germany; German Cancer Consortium (DKTK), Partner Site Berlin, German Cancer Research Center (DKFZ), Heidelberg, Germany; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
48
|
Fujiki K, Kakisawa Y, Mahmoud EM, Ueno Y. Synthesis and Application of 4'- C-[( N-alkyl)aminoethyl]thymidine Analogs for Optimizing Oligonucleotide Properties. Molecules 2025; 30:581. [PMID: 39942684 PMCID: PMC11820600 DOI: 10.3390/molecules30030581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/20/2025] [Accepted: 01/24/2025] [Indexed: 02/16/2025] Open
Abstract
Gapmer-type antisense oligonucleotides (ASOs) are an emerging class of therapeutic agents that directly inhibit pathogenic mRNA. In this study, three new 4'-C-substituted thymidine analogs were generated using a synthetic strategy recently established by our group, namely, 4'-C-(N-ethyl) aminoethyl (4'-EAE-T), 4'-C-(N-butyl) aminoethyl (4'-BAE-T), and 4'-C-(N-octyl) aminoethyl (4'-OAE-T). Their properties were evaluated and compared with those of previously reported analogs, including 4'-C-aminoethyl (4'-AE-T) and 4'-C-(N-methyl) aminoethyl (4'-MAE-T). The novel nucleoside analogs were subsequently incorporated into gapmer-type ASOs featuring phosphorothioate (PS) linkages and locked nucleic acids (LNAs) in the wing regions. The incorporation of 4'-EAE-T and 4'-BAE-T analogs resulted in RNA binding affinities similar to that of the previously reported 4'-MAE-T analog, whereas a marked decrease in RNA affinity was noted for 4'-OAE-T, however, this reduction was mitigated when combined with other chemical modifications. Furthermore, the structural modifications conferred enhanced nuclease resistance under bovine serum conditions, with 4'-EAE-T resulting in the highest stability, followed by 4'-BAE-T and 4'-OAE-T. Additionally, oligonucleotides modified with the developed analogs preserved their RNase H cleavage susceptibility, albeit inducing minor alterations in the cleavage pattern. Finally, the oligonucleotides were applied in a gene silencing experiment targeting the KRAS gene, conducted without the use of transfection agents, displaying gene silencing activities comparable to that of the control, with the exception of the 4'-OAE-modified nucleotide, which exhibited low activity.
Collapse
Affiliation(s)
- Kota Fujiki
- Department of Life Science and Chemistry, The Graduate School of Natural Science and Technology, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan;
| | - Yuri Kakisawa
- Course of Applied Life Science, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan (E.M.M.)
| | - Elsayed M. Mahmoud
- Course of Applied Life Science, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan (E.M.M.)
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Yoshihito Ueno
- Department of Life Science and Chemistry, The Graduate School of Natural Science and Technology, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan;
- Course of Applied Life Science, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan (E.M.M.)
- United Graduate School of Agricultural Science, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Tokai National Higher Education and Research System, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| |
Collapse
|
49
|
Takeda M, Yoshida S, Inoue T, Sekido Y, Hata T, Hamabe A, Ogino T, Miyoshi N, Uemura M, Yamamoto H, Doki Y, Eguchi H. The Role of KRAS Mutations in Colorectal Cancer: Biological Insights, Clinical Implications, and Future Therapeutic Perspectives. Cancers (Basel) 2025; 17:428. [PMID: 39941797 PMCID: PMC11816235 DOI: 10.3390/cancers17030428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/17/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Colorectal cancer (CRC) remains a leading cause of cancer mortality globally, with KRAS mutations occurring in 30-40% of cases, contributing to poor prognosis and resistance to anti-EGFR therapy. This review explores the biological significance, clinical implications, and therapeutic targeting of KRAS mutations in CRC. Methods: A comprehensive analysis of the existing literature and clinical trials was performed, highlighting the role of KRAS mutations in CRC pathogenesis, their impact on prognosis, and recent advancements in targeted therapies. Specific attention was given to emerging therapeutic strategies and resistance mechanisms. Results: KRAS mutations drive tumor progression through persistent activation of MAPK/ERK and PI3K/AKT signaling pathways. These mutations influence the tumor microenvironment, cancer stem cell formation, macropinocytosis, and cell competition. KRAS-mutant CRC exhibits poor responsiveness to anti-EGFR monoclonal antibodies and demonstrates primary and acquired resistance to KRAS inhibitors. Recent breakthroughs include the development of KRAS G12C inhibitors (sotorasib and adagrasib) and promising agents targeting G12D mutations. However, response rates in CRC remain suboptimal compared to other cancers, necessitating combination therapies and novel approaches, such as vaccines, nucleic acid-based therapeutics, and macropinocytosis inhibitors. Conclusions: KRAS mutations are central to CRC pathogenesis and present a significant therapeutic challenge. Advances in KRAS-targeted therapies offer hope for improved outcomes, but resistance mechanisms and organ-specific differences limit efficacy. Continued efforts in personalized treatment strategies and translational research are critical for overcoming these challenges and improving patient survival.
Collapse
Affiliation(s)
- Mitsunobu Takeda
- Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Rolim TDS, Sampaio ALF, Mazzei JL, Moreira DL, Siani AC. Synthesis, Bioproduction and Bioactivity of Perillic Acid-A Review. Molecules 2025; 30:528. [PMID: 39942631 PMCID: PMC11820084 DOI: 10.3390/molecules30030528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Perillic acid (PA) is a limonene derivative in which the exocyclic methyl is oxidized to a carboxyl group. Although endowed with potential anticancer activity, PA has been much less explored regarding its biological properties than analogous compounds such as perillyl alcohol, perillaldehyde, or limonene itself. PA is usually described in mixture with alcohols and ketones produced in the oxidation of monoterpenes, with relatively few existing reports focusing on the PA molecule. This study provides a comprehensive review of PA, addressing its origin, the processes of obtaining it through organic synthesis and biotransformation, and the pharmacological tests in which it is either the lead compound or reference for in vitro efficacy in experimental models. Although feasible and generally poorly yielded, the synthesis of PA from limonene requires multiple steps and the use of unusual catalysts. The most economical process involves using (-)-β-pinene epoxide as the starting material, ending up with (-)-PA. On the other hand, some bacteria and yeasts are successful in producing, exclusively or at satisfactory purity level, PA from limonene or a few other monoterpenes, through environmentally friendly approaches. The compiled data revealed that, with few exceptions, most reports on PA bioactivity are related to its ability to interfere with the prenylation process of oncogenic proteins, an essential step for the growth and dissemination of cancer cells. The present survey reveals that there is still a vast field to disclose regarding the obtaining and scaling of PA via the fermentative route, as well as extending prospective studies on its properties and possible pharmacological applications, especially in the preclinical oncology field.
Collapse
Affiliation(s)
- Thaís de Souza Rolim
- Institute of Drug Technology, Fiocruz, Sizenando Nabuco St. 100, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (T.d.S.R.); (A.L.F.S.); (J.L.M.)
| | - André Luiz Franco Sampaio
- Institute of Drug Technology, Fiocruz, Sizenando Nabuco St. 100, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (T.d.S.R.); (A.L.F.S.); (J.L.M.)
| | - José Luiz Mazzei
- Institute of Drug Technology, Fiocruz, Sizenando Nabuco St. 100, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (T.d.S.R.); (A.L.F.S.); (J.L.M.)
| | - Davyson Lima Moreira
- Institute of Drug Technology, Fiocruz, Sizenando Nabuco St. 100, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (T.d.S.R.); (A.L.F.S.); (J.L.M.)
- Rio de Janeiro Botanical Garden Research Institute, Pacheco Leão St. 915, Jardim Botânico, Rio de Janeiro 22460-030, RJ, Brazil
| | - Antonio Carlos Siani
- Institute of Drug Technology, Fiocruz, Sizenando Nabuco St. 100, Manguinhos, Rio de Janeiro 21041-250, RJ, Brazil; (T.d.S.R.); (A.L.F.S.); (J.L.M.)
| |
Collapse
|