1
|
He Q, Liu X, Jiang L, Liu P, Xuan W, Wang Y, Meng R, Feng H, Lv S, Miao Q, Zheng D, Xu Y, Wang M. First-line treatments for KRAS-mutant non-small cell lung cancer: current state and future perspectives. Cancer Biol Ther 2025; 26:2441499. [PMID: 39681355 PMCID: PMC11651285 DOI: 10.1080/15384047.2024.2441499] [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: 10/16/2024] [Revised: 11/20/2024] [Accepted: 12/09/2024] [Indexed: 12/18/2024] Open
Abstract
KRAS mutations are common in non-small cell lung cancer (NSCLC) and are associated with patient prognosis; however, targeting KRAS has faced various difficulties. Currently, immunotherapy, chemotherapy, and chemoimmunotherapy play pivotal roles in the first-line treatment of KRAS-mutated NSCLC. Here, we summarize the current evidence on first-line therapies and compare the treatment outcomes and biomarkers for different regimens. KRAS inhibitors and other emerging alternative treatments are also discussed, as combining these drugs with immunotherapy may serve as a promising first-line treatment for KRAS-mutated NSCLC in the future. We hope that this review will assist in first-line treatment choices and shed light on the development of novel agents for KRAS-mutated NSCLC.
Collapse
Affiliation(s)
- Qi He
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyan Liu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liyan Jiang
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Ping Liu
- Department of Respiratory Medicine, Changsha Hospital Affiliated to Xiangya Medical College, Central South University (The First Hospital of Changsha), Changsha, China
| | - Weixia Xuan
- Department of Respiratory and Critical Care Medicine, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Yudong Wang
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Rui Meng
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huijing Feng
- Department of Thoracic Oncology, Cancer Center, Shanxi Bethune Hospital, Taiyuan, Shanxi, China
| | - Shuang Lv
- Department of Internal Medicine-Oncology, Inner Mongolia People’s Hospital, Huhehot, Inner Mongolia, P.R. China
| | - Qian Miao
- Department of Thoracic Oncology, Clinical Oncology School of Fujian Medical University, Fuzhou, China
| | - Di Zheng
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| | - Yan Xu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengzhao Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
2
|
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
|
3
|
Kale R, Samant C, Nandakumar K, Ranganath Pai KS, Bhonde M. Drugging the Undruggable and beyond: Emerging precision oncology approaches to target acquired resistance to KRAS G12C and KRAS G12D inhibitors. Biochem Biophys Res Commun 2025; 760:151688. [PMID: 40174369 DOI: 10.1016/j.bbrc.2025.151688] [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: 11/22/2024] [Revised: 02/21/2025] [Accepted: 03/22/2025] [Indexed: 04/04/2025]
Abstract
Development of mutant specific KRAS inhibitors validated KRAS as a 'druggable' target. However, excellent initial efficacy was eventually overshadowed by failure to exhibit sustained clinical response, primarily due to acquired resistance. Some targeted therapies like SOS1, SHP2, and MEK inhibitors, in combination with mutant KRAS G12C inhibitors (G12Ci), are currently under clinical investigation with evidences of improving efficacy. However, a deep understanding of the underlying molecular pathways behind the acquired resistance is still at a nascent stage. Recent preclinical studies have uncovered a role of novel proteins and pathways responsible for resistance and their inhibition demonstrated a robust anticancer efficacy in combination. Plethora of combination therapy approaches are now being proposed with emergence of AXL, ULK1, Tissue factor, farnesyltransferase, etc. as targets to counter G12Ci resistance. This review summarizes in a comprehensive manner, some of the novel combination modalities to overcome G12Ci resistance, based on current understanding and with great potential to hit clinical success. Along with G12C, KRAS G12D (G12D) was also considered a formidable foe, until the discovery of selective inhibitors. However, eventual clinical resistance can eclipse the early success and requires an in-depth understanding of resistance mechanisms. Evidences of G12Ci resistance can be exploited as probable combination strategies to tackle ensuing resistance to G12D inhibitors (G12Di), and can translate in superior clinical efficacy. Early preclinical studies of G12Di in combination with ERBB, SOS1, AKT and immune-checkpoints inhibitors indicate encouraging response. This review further describes some of the early affirmations on combination strategies with G12Di. We postulate to go beyond 'Drugging the Undruggable' with advanced combination approaches mitigating G12C and G12D inhibitor resistance.
Collapse
Affiliation(s)
- Ramesh Kale
- Research Scholar, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India; Department of Pharmacology, Novel Drug Discovery and Development (NDDD), Lupin Limited, Survey No. 46A/47A, Village Nande, Taluka Mulshi, Pune, 412115, Maharashtra, India
| | - Charudatt Samant
- Research Scholar, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India; Department of Pharmacology, Novel Drug Discovery and Development (NDDD), Lupin Limited, Survey No. 46A/47A, Village Nande, Taluka Mulshi, Pune, 412115, Maharashtra, India.
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - K Sreedhara Ranganath Pai
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Mandar Bhonde
- Department of Pharmacology, Novel Drug Discovery and Development (NDDD), Lupin Limited, Survey No. 46A/47A, Village Nande, Taluka Mulshi, Pune, 412115, Maharashtra, India
| |
Collapse
|
4
|
Guo H, Hong J, Zhu Y, Gui H, Liu H, Ren R, Li Y, Shan S, Guan Z, Liu M, Yang Z. A Mannosylated peptidyl lipid CManDA doped into cytidinyl/cationic lipids efficiently delivers siG12Ss to lung cancer in vivo. J Control Release 2025; 381:113624. [PMID: 40073943 DOI: 10.1016/j.jconrel.2025.113624] [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: 12/31/2024] [Revised: 03/02/2025] [Accepted: 03/08/2025] [Indexed: 03/14/2025]
Abstract
Gene therapy has attracted widespread attention in recent years, and one of the important delivery systems is the LNP. However, many LNPs have potential toxicity and accumulate in the liver. Here, we designed and synthesized a Gemini-type mannosylated peptidyl lipid called CManDA(M), which, in combination with the cytidinyl lipid DNCA(D) and the peptidyl lipid CLD(C) (D/50C/50M), could transfect siRNA (siG12S) into A549 cells to target and silence the KRASG12S gene. The fluorescence intensity in the tumor area of the D/50C/50M/Cy5.5-siG12S group increased by approximately 2.5 times. Furthermore, full 2'-F/2'-OMe-modified siG12Ss could also be transfected by D/50C/50M into cells, resulting in target gene silencing. The tumor weight in the D/50C/50M/M3 group (1.5 mg/kg, i.v.) was reduced by 50 % after administration in a mouse axillary tumor (A549) model, whereas the tumor bioluminescence intensity was only approximately 30 % of that in the blank group in a mouse orthotopic lung cancer model and showed no significant toxicity. Further studies revealed that the mannose groups of CManDA can be exposed on the nanoparticle surface to bind lectins, and CManDA can also shield the formation of a protein corona and alter the composition of the protein corona, which aids in the enhancement of its active targeting function. CManDA is expected to be a safe and effective helper lipid for tumor-targeted delivery of siRNA in vivo.
Collapse
Affiliation(s)
- Hua Guo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Jiamei Hong
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yuejie Zhu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Genable (Beijing) Biotechnology Co., Ltd, (#)38 Yongda Road, Beijing 102609, China
| | - Hongzhe Gui
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Hongyi Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Runan Ren
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yi Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Saijun Shan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Zhu Guan
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Mingzhe Liu
- Key Laboratory of Structure-Based Drug Design and Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, China
| | - Zhenjun Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China; Genable (Beijing) Biotechnology Co., Ltd, (#)38 Yongda Road, Beijing 102609, China.
| |
Collapse
|
5
|
Shang Y, Pang M, Fu S, Fei W, Chen B, Zhang Y, Wang J, Shen T. Design, synthesis and biological evaluation of pyrrolopyrimidine urea derivatives as novel KRAS G12C inhibitors for the treatment of cancer. Eur J Med Chem 2025; 289:117391. [PMID: 40024167 DOI: 10.1016/j.ejmech.2025.117391] [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: 12/21/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 03/04/2025]
Abstract
The KRASG12C mutation, which occurs in approximately 14 % of lung adenocarcinomas, has recently become a crucial target for therapy via small molecules that covalently bind to the mutated cysteine. In this study, a novel series of pyrrolopyrimidine derivatives was rationally designed and synthesized, employing a structure-based drug design strategy. Through structure-activity relationship (SAR) analysis, compound SK-17 emerged as a direct and highly potent inhibitor of KRASG12C. Cellular assays illustrated that SK-17 exhibits potent antiproliferative effects, induces apoptosis, possesses anti-tumor metastasis properties, and effectively inhibits the downstream KRAS pathway in a dose-dependent manner. Moreover, the synergistic enhancement observed when SK-17 is combined with SHP2 inhibitors in vitro underscores its innovative potential in combinatorial therapies. In the xenograft mouse model, SK-17 demonstrated outstanding tumor growth suppression with good safety. Importantly, the in vivo test results show that compound SK-17 has a superior PK profile and lower toxicity in zebrafish test. These results demonstrated the potential of SK-17 with novel scaffold as a promising lead compound targeting KRASG12C to guide in-depth structural optimization.
Collapse
Affiliation(s)
- Yanguo Shang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Miaomiao Pang
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Shengnan Fu
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wenjuan Fei
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Boxuan Chen
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China
| | - Yaoyao Zhang
- Cerebrovascular Disease Center, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing, 210024, Jiangsu, China.
| | - Jinxin Wang
- Jiangsu Key Laboratory of Drug Design and Optimization, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Tao Shen
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, China.
| |
Collapse
|
6
|
Wang W, Zhai Y, Yang X, Ye L, Lu G, Shi X, Zhai G. Effective design of therapeutic nanovaccines based on tumor neoantigens. J Control Release 2025; 380:17-35. [PMID: 39892648 DOI: 10.1016/j.jconrel.2025.01.078] [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: 09/05/2024] [Revised: 01/17/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Neoantigen vaccines are among the most potent immunotherapies for personalized cancer treatment. Therapeutic vaccines containing tumor-specific neoantigens that elicit specific T cell responses offer the potential for long-term clinical benefits to cancer patients. Unlike immune-checkpoint inhibitors (ICIs), which rely on pre-existing specific T cell responses, personalized neoantigen vaccines not only promote existing specific T cell responses but importantly stimulate the generation of neoantigen-specific T cells, leading to the establishment of a persistent specific memory T cell pool. The review discusses the current state of clinical research on neoantigen nanovaccines, focusing on the application of vectors, adjuvants, and combinational strategies to address a range of challenges and optimize therapeutic outcomes.
Collapse
Affiliation(s)
- Weilin Wang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT 84124, United States of America
| | - Xiaoye Yang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Lei Ye
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guoliang Lu
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland 1142, New Zealand; Maurice Wilkins Centre, University of Auckland, Private Bag 92019, Auckland 1142, New Zealand
| | - Xiaoqun Shi
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| | - Guangxi Zhai
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China.
| |
Collapse
|
7
|
Ling H, Li L, Duan L, Huang W, Zheng J, Zhang S, Li X, Qiu X, Zhou Y, Ma N, Ren X, Zhang J, Wang Z, Zhao Y, Tian R, Zhang ZM, Ding K. O-Cyanobenzaldehydes Irreversibly Modify Both Buried and Exposed Lysine Residues in Live Cells. J Am Chem Soc 2025; 147:11955-11963. [PMID: 40150802 DOI: 10.1021/jacs.4c18006] [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: 03/29/2025]
Abstract
Lysine residue represents an attractive site for covalent drug development due to its high abundance (5.6%) and critical functions. However, very few lysines have been characterized to be accessible to covalent ligands and perturb the protein functions, owing to their protonation state and adjacent steric hindrance. Herein, we report a new lysine bioconjugation chemistry, O-cyanobenzaldehyde (CNBA), that enables selective modification of the lysine ε-amine to form iso-indolinones under physiological conditions. Activity-based proteome profiling enabled the mapping of 3451 lysine residues and 85 endogenous kinases in live cells, highlighting its potential for modifying hyper-reactive lysines within the proteome or buried catalytic lysines within the kinome. Further protein crystallography and mass spectrometry confirmed that K271_ABL1 and K162_AURKA are covalently targetable sites in kinases. Leveraging a structure-based drug design, we incorporated CNBA into the core structure of Nutlin-3 to irreversibly inhibit the MDM2-p53 interaction by targeting an exposed lysine K94 on the surface of murine double minute 2. Importantly, we have demonstrated the potential application of CNBA as a lysine-recognized bioconjugation agent for developing new antibody-drug conjugates. The results collectively validate CNBA as a new selective and efficient modifying agent with broad applications for both buried and exposed lysine residues in live cells.
Collapse
Affiliation(s)
- Huan Ling
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
- University of Chinese Academy of Sciences, No. 1 Yanxihu Road, Huairou District, Beijing 101408, China
| | - Lin Li
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
| | - Liping Duan
- State Key Laboratory of Bioactive Molecules and Draggability Assessment, School of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Weixue Huang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
| | - Jiangnan Zheng
- Department of Chemistry, School of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Shijie Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Xinling Li
- Department of Chemistry, School of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiaorong Qiu
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
| | - Yang Zhou
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Discovery of Chinese Ministry of Education (MOE), Guangzhou City Key Laboratory of Precision Chemical Drug Development, College of Pharmacy, Jinan University, 855 Xingye Avenue East, Guangzhou 511400, China
| | - Nan Ma
- State Key Laboratory of Bioactive Molecules and Draggability Assessment, School of Pharmacy, Jinan University, Guangzhou 511436, China
- Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Pharmacy, Jinan University, Guangzhou 510632, China
- Guangdong-Hong Kong-Macau Joint Laboratory for Pharmacodynamic Constituents of TCM and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Xiaomei Ren
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
| | - Jinwei Zhang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
| | - Zhen Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
| | - Yujun Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
- University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Ruijun Tian
- Department of Chemistry, School of Science, Southern University of Science and Technology, Shenzhen 518055, China
| | - Zhi-Min Zhang
- State Key Laboratory of Bioactive Molecules and Draggability Assessment, School of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Ke Ding
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Road, Shanghai 200032, China
| |
Collapse
|
8
|
He Y, Yang K, Li S, Zeller M, McAlister GC, Stewart HI, Hock C, Damoc E, Zabrouskov V, Gygi SP, Paulo JA, Yu Q. TMT-based Multiplexed (Chemo)proteomics on the Orbitrap Astral Mass Spectrometer. Mol Cell Proteomics 2025:100968. [PMID: 40210101 DOI: 10.1016/j.mcpro.2025.100968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 04/01/2025] [Accepted: 04/06/2025] [Indexed: 04/12/2025] Open
Abstract
Ongoing advancements in instrumentation has established mass spectrometry (MS) as an essential tool in proteomics research and drug discovery. The newly released Asymmetric Track Lossless (Astral) analyzer represents a major step forward in MS instrumentation. Here, we evaluate the Orbitrap Astral mass spectrometer in the context of tandem mass tag-based multiplexed proteomics and activity-based proteome profiling, highlighting its sensitivity boost relative to the Orbitrap Tribrid platform-50% at the peptide and 20% at the protein level. We compare TMT DDA and label-free DIA on the same instrument, both of which quantify over 10,000 human proteins per sample within one hour. TMT offers higher quantitative precision and data completeness, while DIA is free of ratio compression and is thereby more accurate. Our results suggest that ratio compression is prevalent with the high-resolution MS2-based quantification on the Astral, while real-time search-based MS3 quantification on the Orbitrap Tribrid platform effectively restores accuracy. Additionally, we benchmark TMT-based activity-based proteome profiling by interrogating cysteine ligandability. The Astral measures over 30,000 cysteines in a single-shot experiment, a 54% increase relative to the Orbitrap Eclipse. We further leverage this remarkable sensitivity to profile the target engagement landscape of FDA-approved covalent drugs, including Sotorasib and Adagrasib. We herein provide a reference for the optimal use of the advanced MS platform.
Collapse
Affiliation(s)
- Yuchen He
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States
| | - Ka Yang
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States
| | - Shaoxian Li
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States
| | - Martin Zeller
- Thermo Fisher Scientific, Hanna-Kunath-Straße 11, 28199 Bremen, Germany
| | | | - Hamish I Stewart
- Thermo Fisher Scientific, Hanna-Kunath-Straße 11, 28199 Bremen, Germany
| | - Christian Hock
- Thermo Fisher Scientific, Hanna-Kunath-Straße 11, 28199 Bremen, Germany
| | - Eugen Damoc
- Thermo Fisher Scientific, Hanna-Kunath-Straße 11, 28199 Bremen, Germany
| | | | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, United States.
| | - Qing Yu
- Department of Biochemistry and Molecular Biotechnology, University of Massachusetts Chan Medical School, Worcester, MA 01605, United States.
| |
Collapse
|
9
|
Pulido I, Luan Q, Pastor-Puente S, Gunder L, Wang Y, Ying C, Li J, Sun Y, Dai Y, Ascoli C, Abdelhady K, Massad M, Prince T, Wang G, Foley K, Ying W, Papautsky I, Carretero J, Shimamura T. Chaperone directed heterobifunctional molecules circumvent KRAS G12C inhibitor resistance. Cancer Lett 2025:217691. [PMID: 40204148 DOI: 10.1016/j.canlet.2025.217691] [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: 01/16/2025] [Revised: 03/28/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
While KRASG12C inhibitors have shown promising results in clinical activity, acquired resistance remains a significant barrier to durable responses. Combination therapies have been explored to improve the efficacy of KRASG12C inhibitors; however, their use is often restricted due to toxicity and limitations in clinically amendable dosing schedules. Transcriptomic profiling and functional assays on acquired resistant models to adagrasib identified an enrichment of HSP90 client proteins in resistant phenotypes, suggesting a therapeutic vulnerability. To address the finding, RNK07421, a novel heterobifunctional molecule, was developed to simultaneously target KRASG12C and HSP90-client oncoproteins. Structural and biochemical analyses demonstrated that RNK07421 disrupts KRASG12C interactions by inducing a non-natural interface with HSP90, thereby impairing oncogenic signaling. In vitro, RNK07421 effectively suppressed ERK reactivation and reduced viability in KRASG12C-mutant cell lines exhibiting either intrinsic or acquired resistance. In vivo, RNK07421 significantly reduced tumor burden in xenograft models, outperforming both monotherapies and combination therapies. These findings highlight dual KRASG12C and HSP90 inhibition as a promising strategy to overcome resistance in KRASG12C-driven cancers.
Collapse
Affiliation(s)
- Ines Pulido
- Department of Surgery, Division of Cardiothoracic Surgery; University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Qiyue Luan
- Department of Bioengineering, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Sara Pastor-Puente
- Department of Ophthalmology and Visual Science, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Laura Gunder
- Department of Surgery, Division of Cardiothoracic Surgery; University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Yaya Wang
- Ranok Therapeutics, Waltham, Massachusetts, 02451, Hangzhou, 310020, China
| | - Chenghao Ying
- Ranok Therapeutics, Waltham, Massachusetts, 02451, Hangzhou, 310020, China
| | - Jinhua Li
- Ranok Therapeutics, Waltham, Massachusetts, 02451, Hangzhou, 310020, China
| | - Yuetong Sun
- Ranok Therapeutics, Waltham, Massachusetts, 02451, Hangzhou, 310020, China
| | - Yan Dai
- Ranok Therapeutics, Waltham, Massachusetts, 02451, Hangzhou, 310020, China
| | - Christian Ascoli
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - K Abdelhady
- Department of Surgery, Division of Cardiothoracic Surgery
| | - Malek Massad
- Department of Surgery, Division of Cardiothoracic Surgery
| | - Thomas Prince
- Ranok Therapeutics, Waltham, Massachusetts, 02451, Hangzhou, 310020, China
| | - Guoqiang Wang
- Ranok Therapeutics, Waltham, Massachusetts, 02451, Hangzhou, 310020, China
| | - Kevin Foley
- Ranok Therapeutics, Waltham, Massachusetts, 02451, Hangzhou, 310020, China
| | - Weiwen Ying
- Ranok Therapeutics, Waltham, Massachusetts, 02451, Hangzhou, 310020, China
| | - Ian Papautsky
- Department of Bioengineering, University of Illinois Chicago, Chicago, IL, 60612, USA
| | - Julian Carretero
- Department of Physiology, Universitat de Valencia, Valencia, 46100, Spain
| | - Takeshi Shimamura
- Department of Surgery, Division of Cardiothoracic Surgery; University of Illinois Hospital & Health Sciences System Cancer Center, University of Illinois Chicago, Chicago, IL, 60612, USA.
| |
Collapse
|
10
|
Mehmood A, Hakami MA, Ogaly HA, Subramaniyan V, Khalid A, Wadood A. Evolution of computational techniques against various KRAS mutants in search for therapeutic drugs: a review article. Cancer Chemother Pharmacol 2025; 95:52. [PMID: 40195161 DOI: 10.1007/s00280-025-04767-8] [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: 10/24/2024] [Accepted: 02/23/2025] [Indexed: 04/09/2025]
Abstract
KRAS was (Kirsten rat sarcoma viral oncogene homolog) revealed as an important target in current therapeutic cancer research because alteration of RAS (rat sarcoma viral oncogene homolog) protein has a critical role in malignant modification, tumor angiogenesis, and metastasis. For cancer treatment, designing competitive inhibitors for this attractive target was difficult. Nevertheless, computational investigations of the protein's dynamic behavior displayed the existence of temporary pockets that could be used to design allosteric inhibitors. The last decade witnessed intensive efforts to discover KRAS inhibitors. In 2021, the first KRAS G12C covalent inhibitor, AMG 510, received FDA (Food and drug administration) approval as an anticancer medication that paved the path for future treatment strategies against this target. Computer-aided drug designing discovery has long been used in drug development research targeting different KRAS mutants. In this review, the major breakthroughs in computational methods adapted to discover novel compounds for different mutations have been discussed. Undoubtedly, virtual screening and molecular dynamic (MD) simulation and molecular docking are the most considered approach, producing hits that can be employed in subsequent refinements. After comprehensive analysis, Afatinib and Quercetin were computationally identified as hits in different publications. Several authors conducted covalent docking studies with acryl amide warheads groups containing inhibitors. Future studies are needed to demonstrate their true potential. In-depth studies focusing on various allosteric pockets demonstrate that the switch I/II pocket is a suitable site for drug designing. In addition, machine learning and deep learning based approaches provide new insights for developing anti-KRAS drugs. We believe that this review provides extensive information to researchers globally and encourages further development in this particular area of research.
Collapse
Affiliation(s)
- Ayesha Mehmood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Mohammed Ageeli Hakami
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Shaqra University, Al- Quwayiyah, Riyadh, Saudi Arabia
| | - Hanan A Ogaly
- Chemistry Department, College of Science, King Khalid University, Abha, 61421, Saudi Arabia
| | - Vetriselvan Subramaniyan
- Division of Pharmacology, School of Medical and Life Sciences, Sunway University No. 5, Jalan Universiti, Bandar Sunway, Selangor Darul Ehsan, 47500, Malaysia
| | - Asaad Khalid
- Health Research Center, Jazan University, 114, Jazan, 45142, Saudi Arabia
| | - Abdul Wadood
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan.
| |
Collapse
|
11
|
Su L, Wang D, Purwin TJ, Ran S, Yang Q, Zhang Q, Cai W. Selective USP7 inhibition synergizes with MEK1/2 inhibitor to enhance immune responses and potentiate anti-PD-1 therapy in NRAS mutant melanoma. J Invest Dermatol 2025:S0022-202X(25)00384-7. [PMID: 40204067 DOI: 10.1016/j.jid.2025.03.021] [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: 07/17/2024] [Revised: 02/28/2025] [Accepted: 03/21/2025] [Indexed: 04/11/2025]
Abstract
Targeted therapy for NRAS mutant melanoma remains an unmet clinical need. We found that inhibiting Ubiquitin Specific Peptidase 7 (USP7) with the selective USP7 inhibitor (USP7i) FT671 inhibited cell proliferation in NRAS mutant melanoma cell lines. In addition, we identified and validated the knockout of TP53BP1, TP53 or CDKN1A conferred resistance to FT671, suggesting the activation of a functional p53 signaling pathway is essential for the efficacy of USP7i. In Nras mutant melanoma isograft models, FT671 treatment delayed tumor growth. Moreover, the combinatorial treatment with FT671 and MEK1/2 inhibitor (MEKi) was synergistic and induced pyroptosis in vitro. In immunocompetent mice, the combined treatment profoundly suppressed tumor growth, prolonged survival and enhanced intratumoral immune cell infiltration, particularly increasing the ratios of CD8+ T cells and mature dendritic cells, indicative of activated antitumor immunity. Notably, the triple combination of USP7i, MEKi, and anti-PD-1 antibody resulted in durable tumor regression, with effects persisting beyond 80 days after treatment cessation. These findings establish USP7i+MEKi as a promising strategy for targeting NRAS, an 'undruggable' mutation in melanoma, and provide a strong rationale for the clinical development of USP7i plus MEKi as an adjuvant therapy to enhance anti-PD-1 immunotherapy in NRAS mutant melanoma patients.
Collapse
Affiliation(s)
- Liya Su
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Dinghao Wang
- Department of Mathematics and Statistics, University of Calgary, Calgary, Alberta, Canada
| | - Timothy J Purwin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Sophia Ran
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois, USA; Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - Qi Yang
- Department of Pediatrics,; Child Health Institute of New Jersey; Rutgers Institute for Translational Medicine and Science, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Qingrun Zhang
- Department of Mathematics and Statistics, University of Calgary, Calgary, Alberta, Canada; Department of Biochemistry and Molecular Biology,; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Weijia Cai
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, Illinois, USA; Simmons Cancer Institute, Southern Illinois University School of Medicine, Springfield, Illinois, USA; Department of Surgery, Southern Illinois University School of Medicine, Springfield, Illinois, USA.
| |
Collapse
|
12
|
Geng C, Zeng J, Deng X, Xia F, Xu X. Molecular Dynamics Investigation into the Stability of KRas and CRaf Multimeric Complexes. J Phys Chem B 2025; 129:3306-3316. [PMID: 40126127 DOI: 10.1021/acs.jpcb.4c08767] [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: 03/25/2025]
Abstract
In the Ras/Raf/MAPK signaling pathway, Ras and Raf proteins interact synergistically to form a tetrameric complex. NMR experiments have demonstrated that Ras dimerizes in solution and binds stably to Raf, forming Ras·Raf complexes. In this study, we constructed the ternary and quaternary complexes of KRas and CRaf based on crystal structures, denoted as (KRas)2·CRaf and (KRas)2·(CRaf)2, respectively. Molecular dynamics (MD) simulations were performed to investigate the stability of these complexes, while hydrogen bonds as well as salt bridges formed at the protein-protein interaction interfaces were analyzed based on simulation trajectories. The results revealed that the KRas·CRaf complex is more stable in explicit solvent compared with the KRas dimer. Formation of the stable quaternary complex (KRas)2·(CRaf)2 might be attributed to the association of two binary KRas·CRaf complexes. Additionally, MD simulations of the KRasG12D·CRaf complex revealed a stable and extended binding site at the KRas-CRaf interaction interface. This binding site was identified as a potential therapeutic target to block abnormal signal transmission in the pathway.
Collapse
Affiliation(s)
- Chongli Geng
- School of Chemistry and Molecular Engineering, NYU-ECNU Center for Computational Chemistry at NYU Shanghai, East China Normal University, Shanghai 200062, China
| | - Juan Zeng
- School of Biomedical Engineering, Guangdong Medical University, Dongguan 523808, China
| | - Xianming Deng
- State-province Joint Engineering Laboratory of Targeted Drugs from Natural Products, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian 361102, China
- Department of Hematology, The First Affiliated Hospital of Xiamen University, Xiamen University, 361003 Xiamen, China
| | - Fei Xia
- School of Chemistry and Molecular Engineering, NYU-ECNU Center for Computational Chemistry at NYU Shanghai, East China Normal University, Shanghai 200062, China
| | - Xin Xu
- Collaborative Innovation Center of Chemistry for Energy Materials, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, MOE Key Laboratory of Computational Physical Sciences, Department of Chemistry, Fudan University, Shanghai 200433, China
| |
Collapse
|
13
|
Tedeschi A, Schischlik F, Rocchetti F, Popow J, Ebner F, Gerlach D, Geyer A, Santoro V, Boghossian AS, Rees MG, Ronan MM, Roth JA, Lipp J, Samwer M, Gmachl M, Kraut N, Pearson M, Rudolph D. Pan-KRAS Inhibitors BI-2493 and BI-2865 Display Potent Antitumor Activity in Tumors with KRAS Wild-type Allele Amplification. Mol Cancer Ther 2025; 24:550-562. [PMID: 39711431 PMCID: PMC11962398 DOI: 10.1158/1535-7163.mct-24-0386] [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: 05/24/2024] [Revised: 10/11/2024] [Accepted: 12/12/2024] [Indexed: 12/24/2024]
Abstract
KRASG12C selective inhibitors, such as sotorasib and adagrasib, have raised hopes of targeting other KRAS-mutant alleles in patients with cancer. We report that KRAS wild-type (WT)-amplified tumor models are sensitive to treatment with the small-molecule KRAS inhibitors BI-2493 and BI-2865. These pan-KRAS inhibitors directly target the "OFF" state of KRAS and result in potent antitumor activity in preclinical models of cancers driven by KRAS-mutant proteins. In this study, we used the high-throughput cellular viability Profiling Relative Inhibition Simultaneously in Mixtures assay to assess the antiproliferative activity of BI-2493 in a 900+ cancer cell line panel, expanding on our previous work. KRAS WT-amplified cancer cell lines, with a copy number >7, were identified as the most sensitive, across cell lines with any KRAS alterations, to our pan-KRAS inhibitors. Importantly, our data suggest that a KRAS "OFF" inhibitor is better suited to treat KRAS WT-amplified tumors than a KRAS "ON" inhibitor. KRAS WT amplification is common in patients with gastroesophageal cancers in which it has been shown to act as a unique cancer driver with little overlap to other actionable mutations. The pan-KRAS inhibitors BI-2493 and BI-2865 show potent antitumor activity in vitro and in vivo in KRAS WT-amplified cell lines from this and other tumor types. In conclusion, this is the first study to demonstrate that direct pharmacologic inhibition of KRAS shows antitumor activity in preclinical models of cancer with KRAS WT amplification, suggesting a novel therapeutic concept for patients with cancers bearing this KRAS alteration.
Collapse
Affiliation(s)
| | | | | | | | - Florian Ebner
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | | | - Antonia Geyer
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | | | | | - Matthew G. Rees
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | | | | | - Jesse Lipp
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | | | | | - Norbert Kraut
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | - Mark Pearson
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | | |
Collapse
|
14
|
Shin H, Hwang S, Jeong JH, Shin SC, Oh Y, Kim J, Hwang I, Kim EE, Choo H, Song EJ. Targeting USP47 enhances the efficacy of KRAS inhibitor in KRAS G12C mutated non-small cell lung cancer by controlling deubiquitination of c-Myc. Pharmacol Res 2025; 215:107722. [PMID: 40180254 DOI: 10.1016/j.phrs.2025.107722] [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: 11/13/2024] [Revised: 03/31/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
FDA-approved KRASG12C inhibitors, like Sotorasib, target G12C-mutated KRAS in NSCLC. However, issues with insensitivity and drug resistance have emerged, requiring the development of new combination therapies to overcome these limitations. USP47 has been identified as a regulator of cancer-related signaling pathways such as Wnt, Hippo, and p53. However, its role in the KRAS signaling pathway remains largely unexplored and USP47 inhibitors are less developed than those targeting its homolog, USP7. Here, we identify USP47 as a novel therapeutic target in KRASG12C-mutated NSCLC and report K-552, a selective USP47 inhibitor, as a potential treatment strategy. We demonstrate that USP47 stabilizes c-Myc by preventing its proteasomal degradation through deubiquitination, thereby promoting NSCLC cell proliferation. Additionally, the compound K-552, a USP47 inhibitor identified through virtual screening, effectively destabilizes c-Myc and inhibits KRASG12C-mutated NSCLC cell proliferation. Furthermore, USP47 inhibition-either by siRNA knockdown or K-552 treatment-enhances the efficacy of Sotorasib in vitro and in vivo. Together, our findings establish USP47 as a promising therapeutic target in KRASG12C-mutated NSCLC and introduce K-552 as a USP47 inhibitor with potential for combination therapy with KRASG12C inhibitors.
Collapse
Affiliation(s)
- Hyungkyung Shin
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - SuA Hwang
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea; Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, Republic of Korea
| | - Jeong Hyun Jeong
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Sang Chul Shin
- Technological Convergence Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Yeonji Oh
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Jinhyeok Kim
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Inah Hwang
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea; Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, Republic of Korea
| | - Eunice EunKyeong Kim
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.
| | - Hyunah Choo
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea.
| | - Eun Joo Song
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea; Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Tachibana M, Ito Y, Fujikawa R, Tsukamoto K, Uehara M, Kobayashi J, Hayashi T. Lung Adenocarcinoma Exhibiting Thanatosomes (Hyaline Bodies), Cytoplasmic Clearing, and Nuclear Pleomorphism, with a KRAS Mutation. Diagnostics (Basel) 2025; 15:894. [PMID: 40218244 PMCID: PMC11988772 DOI: 10.3390/diagnostics15070894] [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: 02/13/2025] [Revised: 03/09/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
Since epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors were introduced in 2004, various driver gene mutations have been identified in non-small cell lung cancer, particularly adenocarcinoma, where mutations are typically mutually exclusive. EGFR and Kirsten rat sarcoma viral oncogene (KRAS) mutations are most prevalent in Japan, with routine testing now standard. However, hematoxylin and eosin staining often fails to detect mutations, except in cases such as ALK fusion lung cancer. We report a 76-year-old non-smoking Japanese woman diagnosed with adenocarcinoma confirmed as KRAS G12D/S-positive. Histological features, including thanatosomes (hyaline globules), nuclear pleomorphism, and cytoplasmic clearing, may aid in identifying mutations. Numerous thanatosomes were identified, some containing nuclear dust. Thanatosomes revealed periodic acid-Schiff reactivity with diastase resistance, fuchsinophilia with Masson's trichrome stain, and dark blue-black color with Mallory's PTAH stain. This is the first report linking thanatosomes in KRAS-mutant pulmonary adenocarcinoma to apoptosis via cleaved caspase-3 staining.
Collapse
Affiliation(s)
- Mitsuhiro Tachibana
- Department of Diagnostic Pathology, Shimada General Medical Center, Shimada 427-8502, Japan
| | - Yutaro Ito
- Department of Respiratory Medicine, Shimada General Medical Center, Shimada 427-8502, Japan
| | - Ryo Fujikawa
- Department of Respiratory Surgery, Shimada General Medical Center, Shimada 427-8502, Japan
| | - Kei Tsukamoto
- Department of Diagnostic Radiology, Shimada General Medical Center, Shimada 427-8502, Japan
| | - Masahiro Uehara
- Department of Respiratory Medicine, Shimada General Medical Center, Shimada 427-8502, Japan
| | - Jun Kobayashi
- Department of Respiratory Surgery, Shimada General Medical Center, Shimada 427-8502, Japan
| | - Takuo Hayashi
- Department of Human Pathology, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo 113-8431, Japan;
| |
Collapse
|
16
|
Cardona P, Houk B. Impact of Sotorasib, a KRAS G12C Inhibitor, on the Pharmacokinetics and Therapeutic Window of Digoxin, a P-Glycoprotein Substrate. Clin Pharmacol Drug Dev 2025; 14:298-303. [PMID: 39777405 DOI: 10.1002/cpdd.1501] [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: 10/25/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025]
Abstract
Sotorasib is a small-molecule Kirsten rat sarcoma viral oncogene homolog (KRAS) G12C inhibitor indicated for the treatment of KRAS G12C-driven cancers. KRAS G12C is a common mutation in solid tumors, including non-small cell lung cancer. In vitro studies suggested that sotorasib is a weak inhibitor of P-glycoprotein transporter. Digoxin is a known substrate for P-glycoprotein. The primary objective of this study was to assess the impact of sotorasib on digoxin pharmacokinetics in healthy subjects. This Phase 1, open-label, fixed-sequence study enrolled 14 healthy subjects. Each subject received 0.5 mg of digoxin on Day 1 and 960 mg of sotorasib followed by 0.5 mg of digoxin on Day 7. Blood samples for digoxin pharmacokinetics were collected before dosing and up to 144 hours after the digoxin dose. Digoxin median time to maximum observed plasma concentration and mean terminal half-life were similar following coadministration of digoxin with sotorasib compared with those of digoxin alone. Geometric mean digoxin area under the concentration-time curve from time 0 extrapolated to infinity following coadministration of digoxin with sotorasib (40.3 h•ng/mL) was similar to that of digoxin alone (33.2 h•ng/mL). Geometric mean digoxin maximum observed plasma concentration following coadministration of digoxin with sotorasib (3.64 ng/mL) was higher compared with that of digoxin alone (1.90 ng/mL). Coadministration of digoxin and sotorasib did not impact sotorasib exposure. Single doses of 0.5 mg of digoxin were safe and well tolerated when administered alone or coadministered with 960 mg of sotorasib. Coadministration of digoxin with a single dose of sotorasib increased digoxin area under the concentration-time curve from time 0 extrapolated to infinity and maximum observed plasma concentration by factors of 1.21 and 1.91, respectively, compared with digoxin alone.
Collapse
Affiliation(s)
- Panli Cardona
- Clinical Pharmacology Modeling and Simulation, Amgen Inc., Thousand Oaks, CA, USA
| | - Brett Houk
- Clinical Pharmacology Modeling and Simulation, Amgen Inc., Thousand Oaks, CA, USA
| |
Collapse
|
17
|
Liu Y, Gao X, Li Y, He X, Shi Z, Zhang L, Wang Y, Shi A. Effect of Food on the Pharmacokinetic Characteristics of a Single Oral Dose of D-1553, a Selective Inhibitor of KRAS G12C, in Healthy Chinese Subjects. Clin Drug Investig 2025; 45:201-206. [PMID: 40102370 DOI: 10.1007/s40261-025-01430-1] [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] [Accepted: 02/24/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND AND OBJECTIVE D-1553 (garsorasib) is a novel and selective oral KRASG12C inhibitor. This study aims to evaluate the effect of food on the single-dose pharmacokinetics (PK) of D-1553 tablet in healthy Chinese subjects. Also the safety and tolerability of single-dose D-1553 in subjects are also evaluated. METHODS A randomized, open-label, single-dose, two-intervention (fed vs fasting), two-period, two-sequence crossover study was performed on 14 healthy Chinese subjects. Plasma concentrations of D-1553 were determined by the liquid chromatography-tandem mass spectrometry method. Safety evaluations were carried out during the study period. The main PK parameters of the two formulations of D-1553 were calculated by non-compartmental analysis using Phoenix WinNonlin (Version 8.3) software. RESULTS The geometric mean ratios (90% confidence interval [CI]) of AUC0-t and AUC0-∞ in the high-fat meal condition versus the fasting condition were 86.19% (78.30%, 94.87%) and 83.30% (75.77%, 91.58%), respectively. The geometric mean ratio (90% CI) of Cmax values in high-fat meal condition to that observed in fasting condition were 109.74% (100.22%,120.15%). The p value of Tmax was 0.1484 (fed vs fasting). Two subjects (14.3%) reported 4 treatment-emergent adverse events (TEAEs) in the fasting condition, and no subjects reported TEAEs in the fed condition. All adverse reactions were mild and had recovered by the end of the study. CONCLUSION The study indicated that a high-calorie and high-fat meal has no clinically relevant impact on the PK and bioavailability of D-1553 in healthy Chinese subjects. D-1553 was generally safe and well-tolerated under both fasting and fed conditions. The findings suggest that D-1553 could be administered orally with or without food. CLINICAL TRIALS ClinicalTrials.gov Identifer CTR20212761; registered on 4 Nov 2021.
Collapse
Affiliation(s)
- Yue Liu
- Clinical Trial Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Key Laboratory for Artificial Intelligence and Biotechnology Collaboratories in Gerontology Research, Beijing, People's Republic of China
| | - Xin Gao
- Clinical Trial Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Key Laboratory for Artificial Intelligence and Biotechnology Collaboratories in Gerontology Research, Beijing, People's Republic of China
| | - Yang Li
- Clinical Trial Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Key Laboratory for Artificial Intelligence and Biotechnology Collaboratories in Gerontology Research, Beijing, People's Republic of China
| | - Xuemei He
- Clinical Trial Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Key Laboratory for Artificial Intelligence and Biotechnology Collaboratories in Gerontology Research, Beijing, People's Republic of China
| | - Zhe Shi
- R&D, InventisBio Co., Ltd, Shanghai, People's Republic of China
| | - Ling Zhang
- R&D, InventisBio Co., Ltd, Shanghai, People's Republic of China
| | - Yaolin Wang
- R&D, InventisBio Co., Ltd, Shanghai, People's Republic of China
| | - Aixin Shi
- Clinical Trial Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Key Laboratory for Artificial Intelligence and Biotechnology Collaboratories in Gerontology Research, Beijing, People's Republic of China.
- Clinical Trial Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Key Laboratory of Drug Clinical Risk and Personalized Medication Evaluation, No. 1 Da Hua Road, Dong Dan, Beijing, 100730, People's Republic of China.
| |
Collapse
|
18
|
Zhang Y, Ma Y, Zhang K, Wang Y, Sun X, Kan C, Han F. KRAS Mutations in Cancer: From Molecular Insights to Therapeutic Strategies. Am J Clin Oncol 2025:00000421-990000000-00275. [PMID: 40167108 DOI: 10.1097/coc.0000000000001192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
The global burden of cancer remains a major public health challenge, with Kirsten rat sarcoma viral oncogene homolog (KRAS) emerging as the most common mutated oncogene across diverse malignancies. Once considered "undruggable" due to its unique structure, KRAS has garnered intense research focus, resulting in significant advancements. This paper aims to review recent developments in our understanding of KRAS biology, including its structural and functional aspects, and to explore the latest insights into its mutations across various cancer types. Emphasis is placed on prognosis, predictive roles, and emerging therapeutic strategies targeting KRAS. This review aspires to deepen our comprehension of KRAS and potentially enhance treatment outcomes for cancer patients harboring KRAS mutations in the future.
Collapse
Affiliation(s)
- Yuanzhu Zhang
- Department of Endocrinology and Metabolism, Shandong Provincial Key Medical and Health Discipline of Endocrinology and Laboratory of Endocrinology and Metabolic Diseases, Clinical Research Center
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| | - Yujie Ma
- Department of Endocrinology and Metabolism, Shandong Provincial Key Medical and Health Discipline of Endocrinology and Laboratory of Endocrinology and Metabolic Diseases, Clinical Research Center
| | - Kexin Zhang
- Department of Endocrinology and Metabolism, Shandong Provincial Key Medical and Health Discipline of Endocrinology and Laboratory of Endocrinology and Metabolic Diseases, Clinical Research Center
| | - Yuqun Wang
- Department of Endocrinology and Metabolism, Shandong Provincial Key Medical and Health Discipline of Endocrinology and Laboratory of Endocrinology and Metabolic Diseases, Clinical Research Center
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Shandong Provincial Key Medical and Health Discipline of Endocrinology and Laboratory of Endocrinology and Metabolic Diseases, Clinical Research Center
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Shandong Provincial Key Medical and Health Discipline of Endocrinology and Laboratory of Endocrinology and Metabolic Diseases, Clinical Research Center
| | - Fang Han
- Department of Endocrinology and Metabolism, Shandong Provincial Key Medical and Health Discipline of Endocrinology and Laboratory of Endocrinology and Metabolic Diseases, Clinical Research Center
- Department of Pathology, Affiliated Hospital of Shandong Second Medical University, Weifang, China
| |
Collapse
|
19
|
Itogawa K, Saito T, Nakata Y, Amari H, Tahara H, Yoshino N, Maeda Y, Nomura M, Shiihara J, Nagai Y, Ohta H, Yamaguchi Y. First Reported Case of Pure Red Cell Aplasia Related to Sotorasib. Intern Med 2025; 64:1089-1092. [PMID: 39231674 DOI: 10.2169/internalmedicine.3961-24] [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] [Indexed: 09/06/2024] Open
Abstract
We herein report a 64-year-old man with KRASG12C-mutated advanced lung adenocarcinoma previously treated with immune checkpoint inhibitors (ICIs). One month after starting second-line sotorasib treatment, the patient experienced a progressive decline in serum hemoglobin levels. Anemia was accompanied by markedly elevated serum erythropoietin levels and decreased reticulocyte levels. Bone marrow aspiration revealed pure red cell aplasia. No secondary causes other than medication use were identified. Suspected causative drugs were sotorasib and ICIs. Discontinuation of sotorasib for one week improved his anemia; therefore, the causative drug was identified as sotorasib.
Collapse
Affiliation(s)
- Katsuhiro Itogawa
- Division of Pulmonary Medicine, Jichi Medical University Saitama Medical Center, Japan
| | - Tatsuya Saito
- Division of Pulmonary Medicine, Jichi Medical University Saitama Medical Center, Japan
| | - Yuya Nakata
- Division of Hematology, Jichi Medical University Saitama Medical Center, Japan
| | - Hikari Amari
- Division of Pulmonary Medicine, Jichi Medical University Saitama Medical Center, Japan
| | - Hiroki Tahara
- Division of Pulmonary Medicine, Jichi Medical University Saitama Medical Center, Japan
| | - Nozomu Yoshino
- Division of Hematology, Jichi Medical University Saitama Medical Center, Japan
| | - Yuki Maeda
- Division of Pulmonary Medicine, Jichi Medical University Saitama Medical Center, Japan
| | - Motoko Nomura
- Division of Pulmonary Medicine, Jichi Medical University Saitama Medical Center, Japan
| | - Jun Shiihara
- Division of Pulmonary Medicine, Jichi Medical University Saitama Medical Center, Japan
| | - Yoshiaki Nagai
- Division of Pulmonary Medicine, Jichi Medical University Saitama Medical Center, Japan
| | - Hiromitsu Ohta
- Division of Pulmonary Medicine, Jichi Medical University Saitama Medical Center, Japan
| | - Yasuhiro Yamaguchi
- Division of Pulmonary Medicine, Jichi Medical University Saitama Medical Center, Japan
| |
Collapse
|
20
|
Maki H, Ayabe RI, Haddad A, Nishioka Y, Newhook TE, Tran Cao HS, Chun YS, Tzeng CWD, Vauthey JN. Associations of KRAS Point Mutations with Survival of Patients Who Underwent Curative-Intent Resection of Colorectal Liver Metastases. Ann Surg Oncol 2025; 32:2425-2434. [PMID: 39821490 DOI: 10.1245/s10434-024-16822-4] [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: 10/20/2023] [Accepted: 12/19/2024] [Indexed: 01/19/2025]
Abstract
BACKGROUND The oncologic significance of specific KRAS point mutations for patients with colorectal liver metastases (CLM) is uncertain. This study aimed to assess the prognostic impact of KRAS point mutations on patients who underwent surgery for CLM. METHODS Patients who underwent curative-intent surgery for CLM from 2001 to 2020 were selected for the study. In the study, KRAS point mutations and other clinicopathologic variables were examined for association with survival. RESULTS The study classified 798 patients into five groups by KRAS mutation status as follows: wild-type (n = 412, 51.6%), G12D (n = 123, 15.4%), G12V (n = 88, 11.0%), G13D (n = 61, 7.6%), and "Other" mutations (n = 114, 14.3%). For the patients with G12V substitutions, TP53 mutation was associated with worse overall survival (OS) (hazard ratio [HR], 2.64; 95% confidence interval [CI], 1.04-6.66; P = 0.041), but was not associated with a survival difference for the other four groups. The patients with co-occurring KRAS G12V and TP53 had a median OS of 4.4 years and a 5-year OS rate of 39.8%. In contrast, the patients with KRAS G12V mutation and wild-type TP53 had a median OS of 7.3 years and a 5-year OS rate of 75.9%, similar to the corresponding values for the patients with wild-type KRAS. Co-occurring KRAS G12V and TP53 mutations were independently associated with worse OS in the entire cohort (HR, 2.08; 95% CI, 1.15-3.76; P = 0.015). CONCLUSIONS This study showed that KRAS G12V mutation is associated with worse OS for patients undergoing curative-intent CLM resection, but only those with co-occurring TP53 mutation. Prognosis after surgery for CLM should not be stratified by KRAS mutation site alone.
Collapse
Affiliation(s)
- Harufumi Maki
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Reed I Ayabe
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Antony Haddad
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yujiro Nishioka
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Timothy E Newhook
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hop S Tran Cao
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yun Shin Chun
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ching-Wei D Tzeng
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jean-Nicolas Vauthey
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
21
|
Seo YD, Katz MHG, Snyder RA. The Landmark Series: The Future of Pancreatic Cancer Clinical Trials. Ann Surg Oncol 2025; 32:2777-2785. [PMID: 39815074 DOI: 10.1245/s10434-024-16840-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/25/2024] [Indexed: 01/18/2025]
Abstract
Pancreatic cancer has a poor prognosis despite ongoing advances in systemic and multimodal therapies. This review analyzes recent progress and future directions in pancreatic cancer clinical trials, emphasizing the evolution from traditional approaches to a more personalized and biologically-driven treatment paradigm. While improvements in overall survival have been achieved through perioperative therapies, gaps remain in our understanding of optimal treatment strategies. Key questions include selection of specific chemotherapeutic agents, duration of preoperative therapy, the role of radiotherapy, and accurate and real-time assessment of response to therapy. Historically, pancreatic cancer clinical trials have been designed based on anatomic criteria, failing to account for the inherent biologic heterogeneity of this disease. The field is now moving towards a precision oncology approach, leveraging genomic and transcriptomic data to identify predictive biomarkers and personalize treatment selection. Novel clinical trial designs, such as platform and basket trials, are accelerating the evaluation of new therapeutic strategies and facilitating efficient patient selection, particularly in the context of new emerging targeted therapies such as KRAS inhibitors. Furthermore, implementation of dynamic response assessment techniques, such as circulating tumor DNA and radiomics, may inform treatment decision-making and improve prediction of long-term outcomes. By integrating these evolving strategies, the emerging clinical trial landscape has the potential to transform the treatment of pancreatic cancer and yield meaningful improvements in patient outcomes.
Collapse
Affiliation(s)
- Yongwoo David Seo
- Division of Surgery, Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matthew H G Katz
- Division of Surgery, Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rebecca A Snyder
- Division of Surgery, Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
22
|
Parry CW, Pellicano F, Schüttelkopf AW, Beyer KS, Bower J, Bryson A, Cameron K, Cerutti NM, Clark JP, Davidson SC, Davies K, Drysdale MJ, Engelman J, Estevan-Barber A, Gohlke A, Gray CH, Guthy DA, Hong M, Hopkins A, Hutchinson LD, Konczal J, Maira M, McArthur D, Mezna M, McKinnon H, Nepravishta R, Ostermann N, Pasquali CC, Pollock K, Pugliese A, Rooney N, Schmiedeberg N, Shaw P, Velez-Vega C, West C, West R, Zecri F, Taylor JB. Reversible Small Molecule Multivariant Ras Inhibitors Display Tunable Affinity for the Active and Inactive Forms of Ras. J Med Chem 2025. [PMID: 40162713 DOI: 10.1021/acs.jmedchem.4c02929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Activating mutations of Ras are one of the most prevalent drivers of cancer and are often associated with poor clinical outcomes. Despite FDA approval for two irreversible inhibitors that target the inactive state of KRasG12C, significant unmet clinical need still exists, and the susceptibility of non-G12C mutants to inactive-state inhibition remains unclear. Here we report the discovery of a novel series of reversible inhibitors that bind in an enlarged version of the switch I-II pocket with nanomolar affinities. Dependent on chemotype these can either preferentially bind to the inactive or active state or bind both with similar affinity. The active-state binders inhibit the Raf interaction for wild-type Ras, and a broad range of oncogenic KRas mutants with nanomolar potency. A subseries of these molecules displays cellular inhibition of Ras-Raf binding, as well as decreased phosphorylation of the downstream protein ERK, demonstrating that potent multivariant Ras inhibitors can be accessed from this novel pocket.
Collapse
Affiliation(s)
- Charles W Parry
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Francesca Pellicano
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Alexander W Schüttelkopf
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Kim S Beyer
- Novartis Institute for BioMedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Justin Bower
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Amy Bryson
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Kenneth Cameron
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Nichole M Cerutti
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Jonathan P Clark
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Stuart C Davidson
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Keneth Davies
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Martin J Drysdale
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Jeffrey Engelman
- Novartis Institutes for BioMedical Research, Inc., 181 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Anna Estevan-Barber
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Andrea Gohlke
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Christopher H Gray
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Daniel A Guthy
- Novartis Institute for BioMedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Min Hong
- NCI RAS Initiative, Frederick National Laboratory for Cancer Research, P.O. Box B, Frederick, Maryland 21702, United States
| | - Alana Hopkins
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Luke D Hutchinson
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Jennifer Konczal
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Michel Maira
- Novartis Institute for BioMedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Duncan McArthur
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Mokdad Mezna
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Heather McKinnon
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Ridvan Nepravishta
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Nils Ostermann
- Novartis Institute for BioMedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Camila C Pasquali
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Katie Pollock
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Angelo Pugliese
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Nicholas Rooney
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Niko Schmiedeberg
- Novartis Institute for BioMedical Research, Fabrikstrasse 2, Novartis Campus, CH-4056 Basel, Switzerland
| | - Paul Shaw
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Camilo Velez-Vega
- Novartis Institutes for BioMedical Research, Inc., 181 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - Christopher West
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Ryan West
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| | - Frederic Zecri
- Novartis Institutes for BioMedical Research, Inc., 181 Massachusetts Avenue, Cambridge, Massachusetts 02139, United States
| | - John B Taylor
- Cancer Research Horizons, CRUK Scotland Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, U.K
| |
Collapse
|
23
|
Hao MW, Zhang TX, Dong D, Zhou X, Gao H. Enhancing KRAS G12D inhibitor sensitivity in pancreatic cancer through SHP2/PI3K pathway. Med Oncol 2025; 42:139. [PMID: 40146324 DOI: 10.1007/s12032-025-02683-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025]
Abstract
Pancreatic cancer with the KRAS G12D mutation, found in 40% of cases, is challenging to treat. MRTX1133, a non-covalent KRAS G12D inhibitor, shows therapeutic promise but faces resistance issues. Our study combines MRTX1133 with the SHP2 inhibitor SHP099 or PI3K inhibitor Buparlisib, showing synergistic inhibition of pancreatic cancer cell growth and enhanced apoptosis. These combination therapies could improve clinical outcomes for patients with KRAS G12D mutation in pancreatic cancer.
Collapse
Affiliation(s)
- Man-Wei Hao
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Tian-Xing Zhang
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Dan Dong
- Gynaecology and Obstetrics Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xin Zhou
- Cancer Research Institute of Jilin University, The First Hospital of Jilin University, Changchun, 130021, China
| | - Haicheng Gao
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China.
| |
Collapse
|
24
|
Schaaf RE, Quirke JCK, Ghavami M, Tonogai EJ, Lee HY, Barlock SL, Trzupek TR, Abo KR, Rees MG, Ronan MM, Roth JA, Hergenrother PJ. Identification of a Selective Anticancer Agent from a Collection of Complex-And-Diverse Compounds Synthesized from Stevioside. J Am Chem Soc 2025; 147:10647-10661. [PMID: 40070033 DOI: 10.1021/jacs.5c00919] [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: 03/27/2025]
Abstract
Compounds constructed by distorting the ring systems of natural products serve as a ready source of complex and diverse molecules, useful for a variety of applications. Herein is presented the use of the diterpenoids steviol and isosteviol as starting points for the construction of >50 new compounds through this complexity-to-diversity approach, featuring novel ring system distortions and a noteworthy thallium(III) nitrate (TTN)-mediated ring fusion. Evaluation of this collection identified SteviX4 as a potent and selective anticancer compound, inducing cell death at low nanomolar concentrations against some cancer cell lines in culture, compared to micromolar activity against others. SteviX4 induces ferroptotic cell death in susceptible cell lines, and target identification experiments reveal SteviX4 acts as an inhibitor of glutathione peroxidase 4 (GPX4), a critical protein that protects cancer cells against ferroptosis. In its induction of cell death, SteviX4 displays enhanced cell line selectivity relative to most known GPX4 inhibitors. SteviX4 was used to reveal dependency on GPX4 as a vulnerability of certain cancer cell lines, not tied to any one type of cancer, suggesting GPX4 inhibition as a cancer type-agnostic anticancer strategy. With its high fraction of sp3-hybridized carbons and considerable cell line selectivity and potency, SteviX4 is unique among GPX4 inhibitors, serving as an outstanding probe compound and basis for further translational development.
Collapse
Affiliation(s)
- Rachel E Schaaf
- Department of Chemistry, Cancer Center at Illinois, Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Jonathan C K Quirke
- Department of Chemistry, Cancer Center at Illinois, Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Maryam Ghavami
- Department of Chemistry, Cancer Center at Illinois, Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Emily J Tonogai
- Department of Chemistry, Cancer Center at Illinois, Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Hyang Yeon Lee
- Department of Chemistry, Cancer Center at Illinois, Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Samantha L Barlock
- Department of Chemistry, Cancer Center at Illinois, Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Thomas R Trzupek
- Department of Chemistry, Cancer Center at Illinois, Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Kyle R Abo
- Department of Chemistry, Cancer Center at Illinois, Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Matthew G Rees
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, Massachusetts 02142, United States
| | - Melissa M Ronan
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, Massachusetts 02142, United States
| | - Jennifer A Roth
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, Massachusetts 02142, United States
| | - Paul J Hergenrother
- Department of Chemistry, Cancer Center at Illinois, Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
25
|
Wu J, Xie K, Zhang Y, Zhang W, Cheng R, Zhang Y, Xia Y, Liu T, Yin R, Qiu Y, Xu T, Li R, Sun Q, Yan C. RASON promotes KRAS G12C-driven tumor progression and immune evasion in non-small cell lung cancer. J Exp Clin Cancer Res 2025; 44:106. [PMID: 40128846 PMCID: PMC11934775 DOI: 10.1186/s13046-025-03369-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/17/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND KRAS is the most frequently mutated oncogene in human cancers, with KRASG12C being a prevalent driver mutation in 12-13% non-small cell lung cancer (NSCLC) cases. Despite breakthroughs in KRASG12C inhibitors such as sotorasib (AMG-510) and adagrasib (MRTX-849), clinical resistance remains a challenging issue, highlighting the need for deeper understanding of the molecular mechanisms underlying KRASG12C-driven oncogenic signaling in NSCLC. Previously, we identified RASON as a novel regulator of KRASG12D/V signaling in pancreatic cancer. Herein, we aim to explore the role of RASON in KRASG12C-driven NSCLC and its therapeutic potential. METHODS Immunohistochemistry analysis of NSCLC patient cohorts was performed to demonstrate the correlation between RASON expression and NSCLC progression. Immunoblotting was performed to evaluate the effects of RASON on KRASG12C downstream signaling. In vitro and in vivo assays including cell proliferation, sphere formation, tumor implantation and genetic mouse models were performed to determine the oncogenic role of RASON. RNA-seq analysis was utilized to identify the key signaling pathway regulated by RASON. Immunofluorescence, immunoprecipitation, nuclear magnetic resonance and biochemistry assays were used to validate the interaction between KRASG12C and RASON. Phagocytosis assay and flow cytometry were conducted to explore the effects of RASON on the tumor immune microenvironment. Pharmacological inhibition in subcutaneous xenograft model was used to determine the therapeutical potential of RASON. RESULTS RASON is overexpressed in NSCLC with KRASG12C mutation and correlates with poor patient prognosis. Genetic knockout of RASON significantly reduced lung tumor burden in LSL-KRASG12D; Trp53R172H/+ mice. In KRASG12C-mutant lung cancer cell lines, RASON overexpression enhanced, while CRISPR-mediated knockout suppressed, both in vitro proliferation and in vivo tumor growth. Mechanistically, RASON directly binds KRASG12C, stabilizes it in the GTP-bound hyperactive state and promotes downstream signaling. RASON knockout significantly reduced CD47 expression, enhancing macrophage-mediated phagocytosis and anti-tumor immunity. Therapeutically, antisense oligonucleotides targeting RASON not only exhibited tumor-suppressive effects, but also synergized with the KRASG12C inhibitor AMG-510 to significantly enhance anti-tumor efficacy. CONCLUSION This study reveals RASON as a key oncogenic regulator of KRASG12C signaling, driving lung tumorigenesis and progression, and identifies RASON as a promising therapeutic target for KRASG12C mutant non-small cell lung cancer.
Collapse
Affiliation(s)
- Jianzhuang Wu
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Kexin Xie
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yixuan Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Weiyi Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Rongjie Cheng
- Cancer Stem Cell Laboratory, The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
| | - Yaliang Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yugui Xia
- Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, China
| | - Tongyan Liu
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Jiangsu Cancer Hospital and Nanjing Medical University Affiliated Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
| | - Rong Yin
- Department of Thoracic Surgery, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Jiangsu Cancer Hospital and Nanjing Medical University Affiliated Cancer Hospital and Jiangsu Institute of Cancer Research, Nanjing, China
- Collaborative Innovation Centre for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, China
| | - Yudong Qiu
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tao Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Rutian Li
- The Comprehensive Cancer Center, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qi Sun
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - Chao Yan
- Department of Pancreatic and Metabolic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
- Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, China.
| |
Collapse
|
26
|
Decker M, Huang BJ, Ware T, Boone C, Tang M, Ybarra J, Ballapuram AC, Taran KA, Chen PY, Amendáriz M, Leung CJ, Harris M, Tjoa K, Hongo H, Abelson S, Rivera J, Ngo N, Herbst DM, Suciu RM, Guijas C, Sedighi K, Andalis T, Roche E, Xie B, Liu Y, Smith CC, Stieglitz E, Niphakis MJ, Cravatt BF, Shannon K. In vivo validation of the palmitoylation cycle as a therapeutic target in NRAS -mutant cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.20.644389. [PMID: 40166265 PMCID: PMC11957127 DOI: 10.1101/2025.03.20.644389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Normal and oncogenic Ras proteins are functionally dependent on one or more lipid modifications 1,2 . Whereas K-Ras4b farnesylation is sufficient for stable association with the plasma membrane, farnesylated H-Ras, K-Ras4a, and N-Ras traffic to the Golgi where they must undergo palmitoylation before regulated translocation to cell membranes. N-Ras palmitoylation by the DHHC family of palmitoyl acyl transferases (PATs) and depalmitoylation by ABHD17 serine hydrolases is a dynamic process that is essential for the growth of acute myeloid leukemias (AMLs) harboring oncogenic NRAS mutations 3-6 . Here, we have tested whether co-targeting ABHD17 enzymes and Ras signal output would cooperatively inhibit the proliferation and survival of NRAS -mutant AMLs while sparing normal tissues that retain K-Ras4b function. We show that ABD778, a potent and selective ABHD17 inhibitor with in vivo activity, selectively reduces the growth of NRAS -mutant AML cells in vitro and is synergistic with the allosteric MEK inhibitor PD0325901 (PD901) 7,8 . Similarly, ABD778 and PD901 significantly extended the survival of recipient mice transplanted with three independent primary mouse AMLs harboring an oncogenic Nras G12D driver mutation. Resistant leukemias that emerged during continuous drug treatment acquired by-pass mutations that confer adaptive drug resistance and increase mitogen activated protein kinase (MAPK) signal output. ABD778 augmented the anti-leukemia activity of the pan-PI3 kinase inhibitor pictilisib 9 , the K/N-Ras G12C inhibitor sotorasib 10 , and the FLT3 inhibitor gilteritinib 11 . Co-treatment with ABD778 and gilteritinib restored drug sensitivity in a patient-derived xenograft model of adaptive resistance to FLT3 inhibition. These data validate the palmitoylation cycle as a promising therapeutic target in AML and support exploring it in other NRAS -mutant cancers.
Collapse
Affiliation(s)
- Matthew Decker
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Benjamin J. Huang
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| | - Timothy Ware
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Christopher Boone
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Michelle Tang
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Julia Ybarra
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | | | - Katrine A. Taran
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Pan-Yu Chen
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Marcos Amendáriz
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Camille J. Leung
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Max Harris
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Karensa Tjoa
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Henry Hongo
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Sydney Abelson
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Jose Rivera
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - Nhi Ngo
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | | | - Radu M. Suciu
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Carlos Guijas
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Kimia Sedighi
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Taylor Andalis
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Elysia Roche
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Boer Xie
- Lundbeck La Jolla Research Center, Inc., San Diego, CA, USA
| | - Yunlong Liu
- Center for Center for Medical Genomics, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Catherine C. Smith
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Elliot Stieglitz
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | | | | | - Kevin Shannon
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
27
|
Jin Y, Jana S, Abbasov ME, Lin H. Antibiotic target discovery by integrated phenotypic and activity-based profiling of electrophilic fragments. Cell Chem Biol 2025; 32:434-448.e9. [PMID: 40020665 PMCID: PMC11995724 DOI: 10.1016/j.chembiol.2025.02.001] [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: 04/27/2024] [Revised: 10/05/2024] [Accepted: 02/06/2025] [Indexed: 03/03/2025]
Abstract
The emergence of antibiotic resistance necessitates the discovery of novel bacterial targets and antimicrobial agents. Here, we present a bacterial target discovery framework that integrates phenotypic screening of cysteine-reactive fragments with competitive activity-based protein profiling to map and functionally characterize the targets of screening hits. Using this approach, we identify β-ketoacyl-acyl carrier protein synthase III (FabH) and MiaA tRNA prenyltransferase as primary targets of a hit fragment, 10-F05, that confer bacterial stress resistance and virulence in Shigella flexneri. Mechanistic investigations elucidate that covalent C112 modification in FabH, an enzyme involved in bacterial fatty acid synthesis, results in its inactivation and consequent growth inhibition. We further demonstrate that irreversible C273 modification at the MiaA RNA-protein interaction interface abrogates substrate tRNA binding, attenuating resistance and virulence through decreased translational accuracy. Our findings underscore the efficacy of integrating phenotypic and activity-based profiling of electrophilic fragments to accelerate the identification and pharmacologic validation of new therapeutic targets.
Collapse
Affiliation(s)
- Yizhen Jin
- Graduate Program of Biochemistry, Molecular and Cell Biology, Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA; Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA; Department of Medicine and Department of Chemistry, The University of Chicago, 900 E. 57(th) Street, Chicago, IL 60637, USA
| | - Sadhan Jana
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA; Department of Medicine and Department of Chemistry, The University of Chicago, 900 E. 57(th) Street, Chicago, IL 60637, USA
| | - Mikail E Abbasov
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA.
| | - Hening Lin
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA; Howard Hughes Medical Institute, Department of Medicine and Department of Chemistry, The University of Chicago, 900 E. 57(th) Street, Chicago, IL 60637, USA.
| |
Collapse
|
28
|
Cao L, Li T, Xu X, Sun M, Teng W, Zhu M. Texasin, A main product from Caragana Jubata (Pall.) Poir, induces proliferation arrest and protective autophagy in lung adenocarcinoma. BMC Cancer 2025; 25:513. [PMID: 40114111 PMCID: PMC11924753 DOI: 10.1186/s12885-025-13933-3] [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: 11/18/2024] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Lung cancer, a leading cause of mortality worldwide, necessitates effective therapeutic strategies. Caragana jubata, a traditional Chinese medicinal plant, harbors Texasin, a potential anti-tumor agent. This study aimed to evaluate the anti-cancer effects of Texasin on lung cancer cells, while assessing its impact on normal lung cells. METHODS The study utilized cell lines H1299 and A549, alongside normal lung embryonic cells, to investigate Texasin's effects through Cell Counting Kit-8, Transwell, and wound healing assays. Transcriptome sequencing and analysis were performed to identify potential mechanisms. β-galactosidase activity and Retinoblastoma(RB) protein expression were assessed, and autophagy and apoptosis were explored through chloroquine co-treatment. Mice bearing H1299 cell-derived tumors were treated with Texasin. Tumor changes were assessed through in vivo imaging, and autophagy levels within the tumors were analyzed. RESULTS Texasin inhibited lung cancer cell proliferation, migration, and invasion without harming normal cells. It promoted cell senescence, arrested the cell cycle in G1 phase, and upregulated β-galactosidase and RB protein expression. Texasin induced protective autophagy, which could be converted to apoptosis by chloroquine co-treatment. Texasin inhibits the proliferation of lung adenocarcinoma cells in vivo, as evidenced by immunohistochemistry showing an increase in autophagy levels within the tumors. CONCLUSIONS Texasin emerges as a promising non-cytotoxic anti-lung adenocarcinoma cancer compound, significantly inhibiting malignant phenotypes, highlighting its potential for lung adenocarcinoma cancer therapy.
Collapse
Affiliation(s)
- Liuzhao Cao
- Department of pulmonary and critical care medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu, People's Republic of China
- Northern Jiangsu People's Hospital, Jiangsu, People's Republic of China
| | - Tiantian Li
- Department of Ultrasound, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu, People's Republic of China
- Northern Jiangsu People's Hospital, Jiangsu, People's Republic of China
| | - Xingxiang Xu
- Department of pulmonary and critical care medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu, People's Republic of China
- Northern Jiangsu People's Hospital, Jiangsu, People's Republic of China
| | - Mei Sun
- Department of Hematology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu, People's Republic of China
- Northern Jiangsu People's Hospital, Jiangsu, People's Republic of China
| | - Weiyun Teng
- Department of pulmonary and critical care medicine, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu, People's Republic of China.
- Northern Jiangsu People's Hospital, Jiangsu, People's Republic of China.
| | - Miao Zhu
- Department of Hematology, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Jiangsu, People's Republic of China.
- Northern Jiangsu People's Hospital, Jiangsu, People's Republic of China.
| |
Collapse
|
29
|
Cecchini M, LoRusso P. Challenges and opportunities of emerging mechanisms of resistance to KRAS G12C inhibitors. Ann Oncol 2025:S0923-7534(25)00115-2. [PMID: 40118149 DOI: 10.1016/j.annonc.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/23/2025] Open
Affiliation(s)
- M Cecchini
- Yale University School of Medicine, New Haven, USA
| | - P LoRusso
- Yale University School of Medicine, New Haven, USA.
| |
Collapse
|
30
|
Negrao MV, Paula AG, Molkentine D, Hover L, Nilsson M, Vokes N, Engstrom L, Calinisan A, Briere DM, Waters L, Hallin J, Diao L, Altan M, Blumenschein GR, Skoulidis F, Wang J, Kopetz SE, Hong DS, Gibbons DL, Olson P, Christensen JG, Heymach JV. Impact of Co-mutations and Transcriptional Signatures in Non-Small Cell Lung Cancer Patients Treated with Adagrasib in the KRYSTAL-1 Trial. Clin Cancer Res 2025; 31:1069-1081. [PMID: 39804166 PMCID: PMC11911804 DOI: 10.1158/1078-0432.ccr-24-2310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/01/2024] [Accepted: 01/09/2025] [Indexed: 03/18/2025]
Abstract
PURPOSE KRAS inhibitors are revolutionizing the treatment of non-small cell lung cancer (NSCLC), but clinico-genomic determinants of treatment efficacy warrant continued exploration. EXPERIMENTAL DESIGN Patients with advanced KRASG12C-mutant NSCLC treated with adagrasib [KRYSTAL-1 (NCT03785249)] were included in the analysis. Pretreatment next-generation sequencing data were collected per protocol. HTG EdgeSeq Transcriptome Panel was used for gene expression profiling. Clinical endpoints included objective response, progression-free survival (PFS), and overall survival (OS). KRASG12C-mutant NSCLC cell lines and xenograft models were used for sensitivity analyses and combination drug screens. RESULTS KEAP1 MUT and STK11MUT were associated with shorter survival to adagrasib [KEAP1: PFS 4.1 vs. 9.9 months, HR 2.7, P < 0.01; OS 5.4 vs. 19.0 months, HR 3.6, P < 0.01; STK11: PFS 4.2 vs. 11.0 months, HR 2.2, P < 0.01; OS 9.8 months vs. not reached (NR), HR 2.6, P < 0.01]. KEAP1WT/STK11WT status identified adagrasib-treated patients with significantly longer PFS (16.9 months) and OS (NR). Preclinical analyses further validate the association between KEAP1 loss of function and adagrasib resistance. Adagrasib and mTOR inhibitor combinations produced higher treatment efficacy in NSCLC models harboring STK11 and KEAP1 co-mutations. NRF2HIGH signaling was associated with shorter survival to adagrasib (PFS: 4.2 vs. 8.4 months, HR 2.0, P = 0.02; OS: 6.5 vs. 19.0 months, HR 2.8, P < 0.01) even in patients with KEAP1WT NSCLC. KEAP1WT/STK11WT/NRF2LOW status identified patients-32%-with longer survival to adagrasib (PFS 12.0 vs. 4.2 months, HR 0.2, P < 0.01; OS NR vs. 8.0 months, HR 0.1, P < 0.01). CONCLUSIONS KEAP1, STK11, and NRF2 status define patients with KRASG12C-mutant NSCLC with markedly distinct outcomes to adagrasib. These results further support the use of genomic features-mutational and nonmutational-for the treatment selection of patients with KRASG12C-mutant NSCLC.
Collapse
Affiliation(s)
- Marcelo V. Negrao
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Alvaro G. Paula
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - David Molkentine
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | | | - Monique Nilsson
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Natalie Vokes
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Lars Engstrom
- Mirati Therapeutics, Inc., a Bristol Myers Squibb Company, San Diego, California
| | - Andrew Calinisan
- Mirati Therapeutics, Inc., a Bristol Myers Squibb Company, San Diego, California
| | - David M. Briere
- Mirati Therapeutics, Inc., a Bristol Myers Squibb Company, San Diego, California
| | - Laura Waters
- Mirati Therapeutics, Inc., a Bristol Myers Squibb Company, San Diego, California
| | - Jill Hallin
- Mirati Therapeutics, Inc., a Bristol Myers Squibb Company, San Diego, California
| | - Lixia Diao
- Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mehmet Altan
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - George R. Blumenschein
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Ferdinandos Skoulidis
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Jing Wang
- Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Scott E. Kopetz
- Department of Gastro-Intestinal Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - David S. Hong
- Department of Investigational Cancer Therapeutics, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Don L. Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Peter Olson
- Mirati Therapeutics, Inc., a Bristol Myers Squibb Company, San Diego, California
| | - James G. Christensen
- Mirati Therapeutics, Inc., a Bristol Myers Squibb Company, San Diego, California
| | - John V. Heymach
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, University of Texas, Houston, Texas
| |
Collapse
|
31
|
Jiao B, Yan L, Zhang R, Huang W, Wang X, Liu C, Wang P, Xu P, Wang J, Fang Z, Li D, Xia Z, Li J, Ji S, Zhang Q, Wu M, Wang S, Liu P, Ren R. Loss of Golga7 Suppresses Oncogenic Nras-Driven Leukemogenesis without Detectable Toxicity in Adult Mice. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2412208. [PMID: 40091521 DOI: 10.1002/advs.202412208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/24/2025] [Indexed: 03/19/2025]
Abstract
NRAS mutations are prevalent in human hematological malignancies and are also common in certain solid tumors, including melanoma and colon cancer. Despite their crucial role in oncogenesis, no effective therapies targeting NRAS have been developed. Inhibiting NRAS localization to the plasma membrane (PM) represents a promising strategy for cancer therapy, as its oncogenic signaling relies on PM localization. Knocking out Golgin subfamily A member 7 (Golga7), an accessory protein of RAS palmitoyltransferases, through a conditional gene editing approach drastically suppresses the development of myeloid leukemia induced by the activation of NrasG12D/G12D knock-in alleles in mice. The loss of Golga7 disrupts NRASG12D PM localization in bone marrow cells without altering the level of NRASG12D palmitoylation. Notably, Golga7 is dispensable for normal hematopoiesis in adult mice. While constitutive Golga7 knockout leads to embryonic lethality, the ubiquitous knockout of Golga7 induced in adult mice does not manifest any measurable toxic effects. These findings indicate that GOLGA7 is an effective and safe therapeutic target for NRAS-driven leukemias.
Collapse
Affiliation(s)
- Bo Jiao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lei Yan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Rui Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wei Huang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xinru Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chenxuan Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Peihong Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Pengfei Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jinzeng Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhou Fang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Donghe Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhizhou Xia
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jiaoyang Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shiyu Ji
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qianqian Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Min Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shengyue Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ping Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ruibao Ren
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- International Center for Aging and Cancer, Department of Hematology of The First Affiliated Hospital, Hainan Medical University, Haikou, 571199, China
| |
Collapse
|
32
|
Tanaka E, Oda N, Ogawa T, Takata I, Terao M, Ueki T. Cancer of Unknown Primary Presenting with Chylothorax. Intern Med 2025; 64:887-891. [PMID: 39111886 PMCID: PMC11986303 DOI: 10.2169/internalmedicine.3988-24] [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: 04/15/2024] [Accepted: 06/16/2024] [Indexed: 03/18/2025] Open
Abstract
A 56-year-old man presented to our hospital with dyspnea on exertion for two months. Bilateral pleural effusions were found, and a close examination revealed a chylothorax, including adenocarcinoma. The primary tumor could not be identified by systemic examination. Therefore, the patient was diagnosed with cancer of unknown primary (CUP) presenting with chylothorax. Chemotherapy was administered for CUP, and thoracentesis, pleurodesis, ascites puncture, and nutritional therapy were performed for chylothorax and chylous ascites. Although drainage frequency and tumor marker levels (CA19-9, DUPAN-2, and Span-1) temporarily decreased, disease control deteriorated, and the patient died 12 months after the initial diagnosis.
Collapse
Affiliation(s)
- Emi Tanaka
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Japan
- Department of Internal Medicine, Fukuyama City Hospital, Japan
| | - Naohiro Oda
- Department of Internal Medicine, Fukuyama City Hospital, Japan
| | | | - Ichiro Takata
- Department of Internal Medicine, Fukuyama City Hospital, Japan
| | - Masako Terao
- Department of Internal Medicine, Fukuyama City Hospital, Japan
| | - Toru Ueki
- Department of Internal Medicine, Fukuyama City Hospital, Japan
| |
Collapse
|
33
|
Ghazali N, Garassino MC, Leighl NB, Bestvina CM. Immunotherapy in advanced, KRAS G12C-mutant non-small-cell lung cancer: current strategies and future directions. Ther Adv Med Oncol 2025; 17:17588359251323985. [PMID: 40093982 PMCID: PMC11907553 DOI: 10.1177/17588359251323985] [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: 11/15/2024] [Accepted: 02/05/2025] [Indexed: 03/19/2025] Open
Abstract
Kirsten rat sarcoma (KRAS) mutations are present in up to 25% of non-small-cell lung cancer (NSCLC). KRAS G12C is the most common type of mutation, representing approximately half of the cases in KRAS-mutant NSCLC. Mutations in KRAS activate the RAF-MEK-ERK pathway, leading to increased cell proliferation and survival. Recent advances in drug development have led to the approval of KRAS G12C inhibitors sotorasib and adagrasib. This review explores the emerging therapeutic strategies in KRAS G12C-mutant NSCLC, including dual checkpoint blockade and combinations with checkpoint inhibitors, with a focus on the setting of advanced disease.
Collapse
Affiliation(s)
- Nadia Ghazali
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | | | - Natasha B Leighl
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Christine M Bestvina
- Department of Medicine, The University of Chicago Medicine, 5841 S. Maryland Avenue, MC 2115, Chicago, IL 60637, USA
| |
Collapse
|
34
|
Alshahrani M, Parikh V, Foley B, Hu G, Verkhivker G. Atomistic Profiling of KRAS Interactions with Monobodies and Affimer Proteins Through Ensemble-Based Mutational Scanning Unveils Conserved Residue Networks Linking Cryptic Pockets and Regulating Mechanisms of Binding, Specificity and Allostery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.11.642708. [PMID: 40161650 PMCID: PMC11952430 DOI: 10.1101/2025.03.11.642708] [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
KRAS, a historically "undruggable" oncogenic driver, has eluded targeted therapies due to its lack of accessible binding pockets in its active state. This study investigates the conformational dynamics, binding mechanisms, and allosteric communication networks of KRAS in complexes with monobodies (12D1, 12D5) and affimer proteins (K6, K3, K69) to characterize the binding and allosteric mechanisms and hotspots of KRAS binding. Through molecular dynamics simulations, mutational scanning, binding free energy analysis and network-based analyses, we identified conserved allosteric hotspots that serve as critical nodes for long-range communication in KRAS. Key residues in β-strand 4 (F78, L80, F82), α-helix 3 (I93, H95, Y96), β-strand 5 (V114, N116), and α-helix 5 (Y157, L159, R164) consistently emerged as hotspots across diverse binding partners, forming contiguous networks linking functional regions of KRAS. Notably, β-strand 4 acts as a central hub for propagating conformational changes, while the cryptic allosteric pocket centered around H95/Y96 positions targeted by clinically approved inhibitors was identified as a universal hotspot for both binding and allostery. The study also reveals the interplay between structural rigidity and functional flexibility, where stabilization of one region induces compensatory flexibility in others, reflecting KRAS's adaptability to perturbations. We found that monobodies stabilize the switch II region of KRAS, disrupting coupling between switch I and II regions and leading to enhanced mobility in switch I of KRAS. Similarly, affimer K3 leverages the α3-helix as a hinge point to amplify its effects on KRAS dynamics. Mutational scanning and binding free energy analysis highlighted the energetic drivers of KRAS interactions. revealing key hotspot residues, including H95 and Y96 in the α3 helix, as major contributors to binding affinity and selectivity. Network analysis identified β-strand 4 as a central hub for propagating conformational changes, linking distant functional sites. The predicted allosteric hotspots strongly aligned with experimental data, validating the robustness of the computational approach. Despite distinct binding interfaces, shared hotspots highlight a conserved allosteric infrastructure, reinforcing their universal importance in KRAS signaling. The results of this study can inform rational design of small-molecule inhibitors that mimic the effects of monobodies and affimer proteins, challenging the "undruggable" reputation of KRAS.
Collapse
|
35
|
Bandi DSR, Nagaraju GP, Sarvesh S, Carstens JL, Foote JB, Graff EC, Fang YHD, Keeton AB, Chen X, Valiyaveettil J, Berry KL, Bae S, Akce M, Gorman G, Yoon KJ, Manne U, Boyd MR, Buchsbaum DJ, Azmi AS, Maxuitenko YY, Piazza GA, El-Rayes BF. ADT-1004: a first-in-class, oral pan-RAS inhibitor with robust antitumor activity in preclinical models of pancreatic ductal adenocarcinoma. Mol Cancer 2025; 24:76. [PMID: 40082968 PMCID: PMC11905721 DOI: 10.1186/s12943-025-02288-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 02/28/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND Oncogenic KRAS mutations occur in nearly, 90% of patients with pancreatic ductal adenocarcinoma (PDAC). Targeting KRAS has been complicated by mutational heterogeneity and rapid resistance. We developed a novel pan-RAS inhibitor, ADT-1004 (an oral prodrug of ADT-007) and evaluated antitumor activity in murine and human PDAC models. METHODOLOGY Murine PDAC cells with KRASG12D mutation (KPC-luc or 2838c3-luc) were orthotopically implanted into the pancreas of C57BL/6J mice, and four PDX PDAC tumors with KRAS mutations were implanted subcutaneously in NSG mice. To assess potential to overcome RAS inhibitor resistance, parental and resistant MIA PaCa-2 PDAC cells (KRASG12C mutation) were implanted subcutaneously. Subcutaneously implanted RASWT BxPC-3 cells were used to assess the selectivity of ADT-1004. RESULTS ADT-1004 potently blocked tumor growth and RAS activation in mouse PDAC models without discernable toxicity with target engagement and reduced activated RAS and ERK phosphorylation. In addition, ADT-1004 suppressed tumor growth in PDX PDAC models with KRASG12D, KRASG12V, KRASG12C, or KRASG13Q mutations and increased CD4+ and CD8+ T cells in the TME consistent with exhaustion and increased MHCII+ M1 macrophage and dendritic cells. ADT-1004 demonstrated superior efficacy over sotorasib and adagrasib in tumor models resistant to these KRASG12C inhibitors and MRTX1133 resistant KRASG12D mutant cells. As evidence of selectivity for tumors with mutant KRAS, ADT-1004 did not impact the growth of tumors from RASWT PDAC cells. CONCLUSION/SIGNIFICANCE ADT-1004 has strong antitumor activity in aggressive and clinically relevant PDAC models with unique selectivity to block RAS-mediated signaling in RAS mutant cells. As a pan-RAS inhibitor, ADT-1004 has broad activity and potential efficacy advantages over allele-specific KRAS inhibitors. These findings support clinical trials of ADT-1004 for KRAS mutant PDAC.
Collapse
Affiliation(s)
- Dhana Sekhar Reddy Bandi
- Department of Hematology and Oncology, O'Neal Comprehensive Cancer Center, Heersink School of Medicine, University of Alabama, Birmingham, AL, 35233, USA
| | - Ganji Purnachandra Nagaraju
- Department of Hematology and Oncology, O'Neal Comprehensive Cancer Center, Heersink School of Medicine, University of Alabama, Birmingham, AL, 35233, USA
| | - Sujith Sarvesh
- Department of Hematology and Oncology, O'Neal Comprehensive Cancer Center, Heersink School of Medicine, University of Alabama, Birmingham, AL, 35233, USA
| | - Julienne L Carstens
- Department of Hematology and Oncology, O'Neal Comprehensive Cancer Center, Heersink School of Medicine, University of Alabama, Birmingham, AL, 35233, USA
| | - Jeremy B Foote
- Department of Microbiology, University of Alabama, Birmingham, AL, 35294, USA
| | - Emily C Graff
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, 36849, USA
| | - Yu-Hua D Fang
- Radiology and Neurology, University of Alabama, Birmingham, AL, 35233, USA
| | - Adam B Keeton
- Drug Discovery and Development Department, Harrison College of Pharmacy, Auburn University, Auburn, AL, 36849, USA
- ADT Pharmaceuticals, LLC, Orange Beach, AL, 31691, USA
| | - Xi Chen
- Drug Discovery and Development Department, Harrison College of Pharmacy, Auburn University, Auburn, AL, 36849, USA
- ADT Pharmaceuticals, LLC, Orange Beach, AL, 31691, USA
| | - Jacob Valiyaveettil
- Drug Discovery and Development Department, Harrison College of Pharmacy, Auburn University, Auburn, AL, 36849, USA
- ADT Pharmaceuticals, LLC, Orange Beach, AL, 31691, USA
| | - Kristy L Berry
- Drug Discovery and Development Department, Harrison College of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Sejong Bae
- Division of General Internal Medicine and Population Science, University of Alabama School of Medicine, Birmingham, AL, 35233, USA
| | - Mehmet Akce
- Department of Hematology and Oncology, O'Neal Comprehensive Cancer Center, Heersink School of Medicine, University of Alabama, Birmingham, AL, 35233, USA
| | - Greg Gorman
- Department of Pharmaceutical, Social and Administrative Sciences, Samford University, Birmingham, AL, 35229, USA
| | - Karina J Yoon
- Department of Pharmacology and Toxicology, University of Alabama, Birmingham, AL, 35233, USA
- Department of Cell, Developmental and Integrative Biology, University of Alabama, Birmingham, AL, 35294, USA
| | - Upender Manne
- Department of Pathology, University of Alabama, Birmingham, AL, 35233, USA
| | | | - Donald J Buchsbaum
- Department of Obstetrics and Gynecology, University of Alabama, Birmingham, AL, 35233, USA
| | - Asfar S Azmi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Yulia Y Maxuitenko
- Drug Discovery and Development Department, Harrison College of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Gary A Piazza
- Drug Discovery and Development Department, Harrison College of Pharmacy, Auburn University, Auburn, AL, 36849, USA
- ADT Pharmaceuticals, LLC, Orange Beach, AL, 31691, USA
| | - Bassel F El-Rayes
- Department of Hematology and Oncology, O'Neal Comprehensive Cancer Center, Heersink School of Medicine, University of Alabama, Birmingham, AL, 35233, USA.
| |
Collapse
|
36
|
Paolino M, Tassone G, Governa P, Saletti M, Lami M, Carletti R, Sacchetta F, Pozzi C, Orlandini M, Manetti F, Olivucci M, Cappelli A. Morita-Baylis-Hillman Adduct Chemistry as a Tool for the Design of Lysine-Targeted Covalent Ligands. ACS Med Chem Lett 2025; 16:397-405. [PMID: 40104796 PMCID: PMC11912265 DOI: 10.1021/acsmedchemlett.4c00479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 02/21/2025] [Accepted: 02/24/2025] [Indexed: 03/20/2025] Open
Abstract
The use of Targeted Covalent Inhibitors (TCIs) is an expanding strategy for the development of innovative drugs. It is driven by two fundamental steps: (1) recognition of the target site by the molecule and (2) establishment of the covalent interaction by its reactive group. The development of new TCIs depends on the development of new warheads. Here, we propose the use of Morita-Baylis-Hillman adducts (MBHAs) to covalently bind Lys strategically placed inside a lipophilic pocket. A human cellular retinoic acid binding protein II mutant (M2) was selected as a test bench for a library of 19 MBHAs. The noncovalent interaction step was investigated by molecular docking studies, while experimentally the entire library was incubated with M2 and crystallized to confirm covalent binding with the target lysine. The results, rationalized through covalent docking analysis, support our hypothesis of MBHAs as reactive scaffolds for the design of lysine-TCIs.
Collapse
Affiliation(s)
- Marco Paolino
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Giusy Tassone
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Paolo Governa
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Mario Saletti
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Matteo Lami
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Riccardo Carletti
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Filippo Sacchetta
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Cecilia Pozzi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Maurizio Orlandini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Fabrizio Manetti
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Massimo Olivucci
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
- Chemistry Department, Bowling Green State University, Overman Hall, Bowling Green, Ohio 43403, United States
| | - Andrea Cappelli
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via A. Moro 2, 53100 Siena, Italy
| |
Collapse
|
37
|
Wei JR, Lu MY, Wei TH, Fleishman JS, Yu H, Chen XL, Kong XT, Sun SL, Li NG, Yang Y, Ni HW. Overcoming cancer therapy resistance: From drug innovation to therapeutics. Drug Resist Updat 2025; 81:101229. [PMID: 40081221 DOI: 10.1016/j.drup.2025.101229] [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: 12/26/2024] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
One of the major limitations of cancer therapy is the emergence of drug resistance. This review amis to provide a focused analysis of the multifactorial mechanisms underlying therapy resistance,with an emphasis on actionable insights for developing novel therapeutic strategies. It concisely outlines key factors contributing to therapy resistance, including drug delivery barriers, cancer stem cells (CSCs), epithelial-mesenchymal transition (EMT), cancer heterogeneity, tumor microenvironment (TME), genetic mutations, and alterlations in gene expression. Additionally, we explore how tumors evade targeted therapies through pathway-specific mechanisms that restore disrupted signaling pathways. The review critically evaluates innovative strategies designed to sensitize resistant tumor cells, such as targeted protein dedgradation, antibody-drug conjugates, structure-based drug design, allosteric drugs, multitarget drugs, nanomedicine and others We also highlight the importance of understanding the pharmacological actions of these agents and their integration into treatment regimens. By synthesizing current knowledge and identifying gaps in our understanding, this review aims to guide future research and improve patient outcomes in cancer therapy.
Collapse
Affiliation(s)
- Jin-Rui Wei
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China; The First Clinical College of Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Meng-Yi Lu
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Tian-Hua Wei
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Joshua S Fleishman
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Hui Yu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Xiao-Li Chen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Xiang-Tu Kong
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China
| | - Shan-Liang Sun
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China; State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 211198, China.
| | - Nian-Guang Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Ye Yang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Hai-Wen Ni
- Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Provincial Hospital of Traditional Chinese Medicine, Nanjing 210029, China.
| |
Collapse
|
38
|
Mahadevan KK, Maldonado AS, Li B, Bickert AA, Perdyan A, Kumbhar SV, Piya S, Sockwell A, Morse SJ, Arian K, Sugimoto H, Shalapour S, Hong DS, Heffernan TP, Maitra A, Kalluri R. Inhibitors of oncogenic Kras specifically prime CTLA4 blockade to transcriptionally reprogram Tregs and overcome resistance to suppress pancreas cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640711. [PMID: 40093186 PMCID: PMC11908235 DOI: 10.1101/2025.02.28.640711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Lack of sustained response to oncogenic Kras (Kras*) inhibition in preclinical models and patients with pancreatic ductal adenocarcinoma (PDAC) emphasizes the need to identify impactful synergistic combination therapies to achieve robust clinical benefit. Kras* targeting results in an influx of global T cell infiltrates including Tregs, effector CD8 + T cells and exhausted CD8 + T cells expressing several immune checkpoint molecules in PDAC. Here, we probe whether the T cell influx induced by diverse Kras* inhibitors open a therapeutic window to target the adaptive immune response in PDAC. We show a specific synergy of anti-CTLA4 immune checkpoint blockade with Kras* targeting primed by Kras G12D allele specific inhibitor, MRTX1133 and multi-selective pan-RAS inhibitor, RMC-6236, both currently in clinical testing phase. In contrast, attempted therapeutic combination following Kras* targeting with multiple checkpoint inhibitors, including anti-PD1, anti-Tim3, anti-Lag3, anti-Vista and anti-4-1BB agonist antibody failed due to compensatory mechanisms mediated by other checkpoints on exhausted CD8 + T cells. Anti-CTLA4 therapy in Kras* targeted PDAC transcriptionally reprograms effector T regs to a naïve phenotype, reverses CD8 + T cell exhaustion and is associated with recruitment of tertiary lymphoid structures (TLS) containing interferon (IFN)-stimulated/ activated B cells and germinal center B cells to enable immunotherapy efficacy and overcome resistance with long-term survival. Single cell ATAC sequencing analysis revealed that transcriptional reprogramming of Tregs is epigenetically regulated by downregulation of AP-1 family of transcription factors including Fos, Fos-b, Jun-b, Jun-d in the IL-35 promoter region. This study reveals an actionable vulnerability in the adaptive immune response in Kras* targeted PDAC with important clinical implications. Graphical abstract
Collapse
|
39
|
Godinez-Macias KP, Chen D, Wallis JL, Siegel MG, Adam A, Bopp S, Carolino K, Coulson LB, Durst G, Thathy V, Esherick L, Farringer MA, Flannery EL, Forte B, Liu T, Godoy Magalhaes L, Gupta AK, Istvan ES, Jiang T, Kumpornsin K, Lobb K, McLean KJ, Moura IMR, Okombo J, Payne NC, Plater A, Rao SPS, Siqueira-Neto JL, Somsen BA, Summers RL, Zhang R, Gilson MK, Gamo FJ, Campo B, Baragaña B, Duffy J, Gilbert IH, Lukens AK, Dechering KJ, Niles JC, McNamara CW, Cheng X, Birkholtz LM, Bronkhorst AW, Fidock DA, Wirth DF, Goldberg DE, Lee MCS, Winzeler EA. Revisiting the Plasmodium falciparum druggable genome using predicted structures and data mining. NPJ DRUG DISCOVERY 2025; 2:3. [PMID: 40066064 PMCID: PMC11892419 DOI: 10.1038/s44386-025-00006-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/22/2025] [Indexed: 03/19/2025]
Abstract
Identification of novel drug targets is a key component of modern drug discovery. While antimalarial targets are often identified through the mechanism of action studies on phenotypically derived inhibitors, this method tends to be time- and resource-consuming. The discoverable target space is also constrained by existing compound libraries and phenotypic assay conditions. Leveraging recent advances in protein structure prediction, we systematically assessed the Plasmodium falciparum genome and identified 867 candidate protein targets with evidence of small-molecule binding and blood-stage essentiality. Of these, 540 proteins showed strong essentiality evidence and lack inhibitors that have progressed to clinical trials. Expert review and rubric-based scoring of this subset based on additional criteria such as selectivity, structural information, and assay developability yielded 27 high-priority antimalarial target candidates. This study also provides a genome-wide data resource for P. falciparum and implements a generalizable framework for systematically evaluating and prioritizing novel pathogenic disease targets.
Collapse
Affiliation(s)
| | - Daisy Chen
- Department of Pediatrics, University of California, San Diego, La Jolla, CA USA
| | | | | | - Anna Adam
- MMV Medicines for Malaria Venture, 1215, Geneva, Switzerland
| | - Selina Bopp
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Krypton Carolino
- Department of Pediatrics, University of California, San Diego, La Jolla, CA USA
| | - Lauren B. Coulson
- Holistic Drug Discovery and Development (H3D) Centre, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Greg Durst
- Lgenia, Inc., 412 S Maple St, Fortville, IN USA
| | - Vandana Thathy
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY USA
| | - Lisl Esherick
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Madeline A. Farringer
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA USA
- Division of Infectious Diseases, Boston Children’s Hospital, Boston, MA USA
| | | | - Barbara Forte
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Science, University of Dundee, Dundee, UK
| | - Tiqing Liu
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA USA
| | - Luma Godoy Magalhaes
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Science, University of Dundee, Dundee, UK
| | - Anil K. Gupta
- Calibr-Skaggs Institute for Innovative Medicines, a division of The Scripps Research Institute, La Jolla, CA USA
| | - Eva S. Istvan
- Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, MO USA
| | - Tiantian Jiang
- Department of Pediatrics, University of California, San Diego, La Jolla, CA USA
| | - Krittikorn Kumpornsin
- Calibr-Skaggs Institute for Innovative Medicines, a division of The Scripps Research Institute, La Jolla, CA USA
| | - Karen Lobb
- Lgenia, Inc., 412 S Maple St, Fortville, IN USA
| | - Kyle J. McLean
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Igor M. R. Moura
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY USA
- São Carlos Institute of Physics, University of São Paulo, São Carlos, São Paulo, Brazil
| | - John Okombo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY USA
| | - N. Connor Payne
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA USA
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA USA
| | - Andrew Plater
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Science, University of Dundee, Dundee, UK
| | | | - Jair L. Siqueira-Neto
- Department of Pediatrics, University of California, San Diego, La Jolla, CA USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA USA
| | | | - Robert L. Summers
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA USA
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA USA
| | - Rumin Zhang
- Global Health Drug Discovery Institute, Beijing, China
| | - Michael K. Gilson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA USA
| | | | - Brice Campo
- MMV Medicines for Malaria Venture, 1215, Geneva, Switzerland
| | - Beatriz Baragaña
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Science, University of Dundee, Dundee, UK
| | - James Duffy
- MMV Medicines for Malaria Venture, 1215, Geneva, Switzerland
| | - Ian H. Gilbert
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, School of Life Science, University of Dundee, Dundee, UK
| | - Amanda K. Lukens
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA USA
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA USA
| | | | - Jacquin C. Niles
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA USA
| | - Case W. McNamara
- Calibr-Skaggs Institute for Innovative Medicines, a division of The Scripps Research Institute, La Jolla, CA USA
| | - Xiu Cheng
- Global Health Drug Discovery Institute, Beijing, China
| | - Lyn-Marie Birkholtz
- Department of Biochemistry, Genetics & Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Private Bag X20, Hatfield, Pretoria, South Africa
| | | | - David A. Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY USA
| | - Dyann F. Wirth
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA USA
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA USA
| | - Daniel E. Goldberg
- Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, MO USA
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, MO USA
| | - Marcus C. S. Lee
- Division of Biological Chemistry and Drug Discovery, Wellcome Centre for Anti-Infectives Research, University of Dundee, Dundee, UK
| | - Elizabeth A. Winzeler
- Department of Pediatrics, University of California, San Diego, La Jolla, CA USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA USA
| |
Collapse
|
40
|
Foote JB, Mattox TE, Keeton AB, Chen X, Smith FT, Berry K, Holmes TW, Wang J, Huang CH, Ward A, Mitra AK, Ramirez-Alcantara V, Hardy C, Fleten KG, Flatmark K, Yoon KJ, Sarvesh S, Nagaraju GP, Bandi DSR, Maxuitenko YY, Valiyaveettil J, Carstens JL, Buchsbaum DJ, Yang J, Zhou G, Nurmemmedov E, Babic I, Gaponenko V, Abdelkarim H, Boyd MR, Gorman G, Manne U, Bae S, El-Rayes BF, Piazza GA. A Pan-RAS Inhibitor with a Unique Mechanism of Action Blocks Tumor Growth and Induces Antitumor Immunity in Gastrointestinal Cancer. Cancer Res 2025; 85:956-972. [PMID: 39700396 PMCID: PMC11875992 DOI: 10.1158/0008-5472.can-24-0323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 06/04/2024] [Accepted: 12/11/2024] [Indexed: 12/21/2024]
Abstract
Activated RAS is a common driver of cancer that was considered undruggable for decades. Recent advances have enabled the development of RAS inhibitors, but the efficacy of these inhibitors remains limited by resistance. In this study, we developed a pan-RAS inhibitor, ADT-007, (Z)-2-(5-fluoro-1-(4-hydroxy-3,5-dimethoxybenzylidene)-2-methyl-1H-inden-3-yl)-N-(furan-2-ylmethyl)acetamide, that binds nucleotide-free RAS to block GTP activation of effector interactions and MAPK/AKT signaling, resulting in mitotic arrest and apoptosis. ADT-007 potently inhibited the growth of RAS-mutant cancer cells irrespective of the RAS mutation or isozyme. Wild-type RAS (RASWT) cancer cells with GTP-activated RAS from upstream mutations were equally sensitive. Conversely, RASWT cancer cells harboring downstream BRAF mutations and normal cells were essentially insensitive to ADT-007. Sensitivity of cancer cells to ADT-007 required activated RAS and dependence on RAS for proliferation, whereas insensitivity was attributed to metabolic deactivation by UDP-glucuronosyltransferases that were expressed in RASWT and normal cells but repressed in RAS-mutant cancer cells. ADT-007 displayed unique advantages over KRAS mutant-specific, pan-KRAS, and pan-RAS inhibitors that could impact in vivo antitumor efficacy by escaping compensatory mechanisms that lead to resistance. Local administration of ADT-007 showed robust antitumor activity in syngeneic immunocompetent and xenogeneic immune-deficient mouse models of colorectal and pancreatic cancers. The antitumor activity of ADT-007 was associated with the suppression of MAPK signaling and activation of innate and adaptive immunity in the tumor immune microenvironment. Oral administration of ADT-007 prodrug also inhibited tumor growth. Thus, ADT-007 has the potential to address the complex RAS mutational landscape of many human cancers and to improve treatment of RAS-driven tumors. Significance: ADT-007, a first-in-class pan-RAS inhibitor, has unique selectivity for cancer cells with mutant RAS or activated RAS protein and the capability to circumvent resistance to suppress tumor growth, supporting further development of ADT-007 analogs.
Collapse
Affiliation(s)
- Jeremy B. Foote
- Department of Microbiology, University of Alabama at Birmingham, Birmingham AL
| | | | - Adam B. Keeton
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL
- ADT Pharmaceuticals LLC, Orange Beach, AL
| | - Xi Chen
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL
- ADT Pharmaceuticals LLC, Orange Beach, AL
| | - Forrest T. Smith
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL
| | - Kristy Berry
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL
| | - Thomas W. Holmes
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL
| | - Junwei Wang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL
| | - Chung-hui Huang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL
| | | | - Amit K. Mitra
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL
| | | | - Cherlene Hardy
- Department of Microbiology, University of Alabama at Birmingham, Birmingham AL
| | - Karrianne G. Fleten
- Department of Gastroenterological Surgery, Oslo University Hospital, The Radium Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Kjersti Flatmark
- Department of Gastroenterological Surgery, Oslo University Hospital, The Radium Hospital, Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Karina J. Yoon
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL
| | - Sujith Sarvesh
- Department of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL
| | - Ganji P. Nagaraju
- Department of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL
| | | | - Yulia Y. Maxuitenko
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL
| | - Jacob Valiyaveettil
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL
| | - Julienne L. Carstens
- Department of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL
| | - Donald J. Buchsbaum
- Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL
| | | | - Gang Zhou
- Georgia Cancer Center, University of Augusta, Augusta, GA
| | | | | | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL
| | - Hazem Abdelkarim
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL
| | | | - Greg Gorman
- Department of Pharmaceutical, Social and Administrative Sciences, McWhorter School of Pharmacy, Samford University; Birmingham, AL
| | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - Sejong Bae
- Division of Preventive Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Bassel F. El-Rayes
- Department of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL
| | - Gary A. Piazza
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL
- ADT Pharmaceuticals LLC, Orange Beach, AL
| |
Collapse
|
41
|
Maciag AE, Stice JP, Wang B, Sharma AK, Chan AH, Lin K, Singh D, Dyba M, Yang Y, Setoodeh S, Smith BP, Ju JH, Jeknic S, Rabara D, Zhang Z, Larsen EK, Esposito D, Denson JP, Ranieri M, Meynardie M, Mehdizadeh S, Alexander PA, Abreu Blanco M, Turner DM, Xu R, Lightstone FC, Wong KK, Stephen AG, Wang K, Simanshu DK, Sinkevicius KW, Nissley DV, Wallace E, McCormick F, Beltran PJ. Discovery of BBO-8520, a First-In-Class Direct and Covalent Dual Inhibitor of GTP-Bound (ON) and GDP-Bound (OFF) KRASG12C. Cancer Discov 2025; 15:578-594. [PMID: 39642212 PMCID: PMC11873722 DOI: 10.1158/2159-8290.cd-24-0840] [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: 06/12/2024] [Revised: 10/02/2024] [Accepted: 11/26/2024] [Indexed: 12/08/2024]
Abstract
Approved inhibitors of KRASG12C prevent oncogenic activation by sequestering the inactive, GDP-bound (OFF) form rather than directly binding and inhibiting the active, GTP-bound (ON) form. This approach provides no direct target coverage of the active protein. Expectedly, adaptive resistance to KRASG12C (OFF)-only inhibitors is observed in association with increased expression and activity of KRASG12C(ON). To provide optimal KRASG12C target coverage, we have developed BBO-8520, a first-in-class, direct dual inhibitor of KRASG12C(ON) and (OFF) forms. BBO-8520 binds in the Switch-II/Helix3 pocket, covalently modifies the target cysteine, and disables effector binding to KRASG12C(ON). BBO-8520 exhibits potent signaling inhibition in growth factor-activated states, in which current (OFF)-only inhibitors demonstrate little measurable activity. In vivo, BBO-8520 demonstrates rapid target engagement and inhibition of signaling, resulting in durable tumor regression in multiple models, including those resistant to KRASG12C(OFF)-only inhibitors. BBO-8520 is in phase 1 clinical trials in patients with KRASG12C non-small cell lung cancer. Significance: BBO-8520 is a first-in-class direct, small molecule covalent dual inhibitor that engages KRASG12C in the active (ON) and inactive (OFF) conformations. BBO-8520 represents a novel mechanism of action that allows for optimal target coverage and delays the emergence of adaptive resistance seen with (OFF)-only inhibitors in the clinic. See related commentary by Zhou and Westover, p. 455.
Collapse
Affiliation(s)
- Anna E. Maciag
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - James P. Stice
- BridgeBio Oncology Therapeutics, South San Francisco, California
| | - Bin Wang
- BridgeBio Oncology Therapeutics, South San Francisco, California
| | - Alok K. Sharma
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Albert H. Chan
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Ken Lin
- BridgeBio Oncology Therapeutics, South San Francisco, California
| | - Devansh Singh
- BridgeBio Oncology Therapeutics, South San Francisco, California
| | - Marcin Dyba
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Yue Yang
- Physical and Life Sciences (PLS) Directorate, Lawrence Livermore National Laboratory, Livermore, California
| | - Saman Setoodeh
- BridgeBio Oncology Therapeutics, South San Francisco, California
| | - Brian P. Smith
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Jin Hyun Ju
- BridgeBio Oncology Therapeutics, South San Francisco, California
| | - Stevan Jeknic
- BridgeBio Oncology Therapeutics, South San Francisco, California
| | - Dana Rabara
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Zuhui Zhang
- BridgeBio Oncology Therapeutics, South San Francisco, California
| | - Erik K. Larsen
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Dominic Esposito
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - John-Paul Denson
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Michela Ranieri
- Perlmutter Cancer Center, New York University, New York, New York
| | - Mary Meynardie
- Perlmutter Cancer Center, New York University, New York, New York
| | - Sadaf Mehdizadeh
- BridgeBio Oncology Therapeutics, South San Francisco, California
| | - Patrick A. Alexander
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Maria Abreu Blanco
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - David M. Turner
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Rui Xu
- BridgeBio Oncology Therapeutics, South San Francisco, California
| | - Felice C. Lightstone
- Physical and Life Sciences (PLS) Directorate, Lawrence Livermore National Laboratory, Livermore, California
| | - Kwok-Kin Wong
- Perlmutter Cancer Center, New York University, New York, New York
| | - Andrew G. Stephen
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Keshi Wang
- BridgeBio Oncology Therapeutics, South San Francisco, California
| | - Dhirendra K. Simanshu
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | | | - Dwight V. Nissley
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
| | - Eli Wallace
- BridgeBio Oncology Therapeutics, South San Francisco, California
| | - Frank McCormick
- NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, Maryland
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Pedro J. Beltran
- BridgeBio Oncology Therapeutics, South San Francisco, California
| |
Collapse
|
42
|
Zhang Y, Liu Z, Hirschi M, Brodsky O, Johnson E, Won SJ, Nagata A, Bezwada D, Petroski MD, Majmudar JD, Niessen S, VanArsdale T, Gilbert AM, Hayward MM, Stewart AE, Nager AR, Melillo B, Cravatt BF. An allosteric cyclin E-CDK2 site mapped by paralog hopping with covalent probes. Nat Chem Biol 2025; 21:420-431. [PMID: 39294320 DOI: 10.1038/s41589-024-01738-7] [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: 01/21/2024] [Accepted: 08/20/2024] [Indexed: 09/20/2024]
Abstract
More than half of the ~20,000 protein-encoding human genes have paralogs. Chemical proteomics has uncovered many electrophile-sensitive cysteines that are exclusive to subsets of paralogous proteins. Here we explore whether such covalent compound-cysteine interactions can be used to discover ligandable pockets in paralogs lacking the cysteine. Leveraging the covalent ligandability of C109 in the cyclin CCNE2, we substituted the corresponding residue in paralog CCNE1 to cysteine (N112C) and found through activity-based protein profiling that this mutant reacts stereoselectively and site-specifically with tryptoline acrylamides. We then converted the tryptoline acrylamide-CCNE1-N112C interaction into in vitro NanoBRET (bioluminescence resonance energy transfer) and in cellulo activity-based protein profiling assays capable of identifying compounds that reversibly inhibit both the N112C mutant and wild-type CCNE1:CDK2 (cyclin-dependent kinase 2) complexes. X-ray crystallography revealed a cryptic allosteric pocket at the CCNE1:CDK2 interface adjacent to N112 that binds the reversible inhibitors. Our findings, thus, show how electrophile-cysteine interactions mapped by chemical proteomics can extend the understanding of protein ligandability beyond covalent chemistry.
Collapse
Affiliation(s)
- Yuanjin Zhang
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Zhonglin Liu
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Marscha Hirschi
- Medicine Design, Pfizer Research and Development, Pfizer, Inc., La Jolla, CA, USA
| | - Oleg Brodsky
- Medicine Design, Pfizer Research and Development, Pfizer, Inc., La Jolla, CA, USA
| | - Eric Johnson
- Medicine Design, Pfizer Research and Development, Pfizer, Inc., La Jolla, CA, USA
| | - Sang Joon Won
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Asako Nagata
- Medicine Design, Pfizer Research and Development, Pfizer, Inc., La Jolla, CA, USA
| | - Divya Bezwada
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Jaimeen D Majmudar
- Discovery Sciences, Pfizer Research and Development, Pfizer, Inc., Cambridge, MA, USA
| | - Sherry Niessen
- Oncology Research and Development, Pfizer, Inc., La Jolla, CA, USA
- Belharra Therapeutics, San Diego, CA, USA
| | - Todd VanArsdale
- Oncology Research and Development, Pfizer, Inc., La Jolla, CA, USA
| | - Adam M Gilbert
- Discovery Sciences, Pfizer Research and Development, Pfizer, Inc., Groton, CT, USA
| | - Matthew M Hayward
- Discovery Sciences, Pfizer Research and Development, Pfizer, Inc., Groton, CT, USA
- Magnet Biomedicine, Boston, MA, USA
| | - Al E Stewart
- Medicine Design, Pfizer Research and Development, Pfizer, Inc., La Jolla, CA, USA
| | - Andrew R Nager
- Oncology Research and Development, Pfizer, Inc., La Jolla, CA, USA
| | - Bruno Melillo
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Benjamin F Cravatt
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
43
|
Khedkar NR, Sindkhedkar M, Joseph A. Fragment-Based Drug Discovery: Small Fragments, Big Impact - Success Stories of Approved Oncology Therapeutics. Bioorg Chem 2025; 156:108197. [PMID: 39879825 DOI: 10.1016/j.bioorg.2025.108197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 12/01/2024] [Accepted: 01/18/2025] [Indexed: 01/31/2025]
Abstract
Fragment-Based Drug Discovery (FBDD) has revolutionized drug discovery by overcoming the challenges of traditional methods like combinatorial chemistry and high-throughput screening (HTS). Leveraging small, low-molecular-weight fragments, FBDD achieves higher hit rates, reduced screening costs, and faster development timelines for clinically relevant drug candidates. This review explores FBDD's core principles, innovative methodologies, and its success in targeting diverse protein classes, including previously "undruggable" targets. Key advancements in fragment libraries, screening techniques, and computational tools are discussed, along with the efficient progression from fragment hits to clinical drugs. Notably, we highlight FDA-approved fragment-derived drugs, including capivasertib, which has increased the total number of fragment-based oncology drugs to seven. As FBDD continues to evolve, its potential to address unmet therapeutic needs and drive the discovery of groundbreaking treatments across various disease areas becomes increasingly evident.
Collapse
Affiliation(s)
- Nilesh Raghunath Khedkar
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal, Academy of Higher Education, Manipal, Karnataka 576104, India; Novel Drug Discovery & Development, Lupin Research Park, Lupin Ltd., Pune 412115, India; Research Scholar, Manipal Academy of Higher Education, India
| | - Milind Sindkhedkar
- Novel Drug Discovery & Development, Lupin Research Park, Lupin Ltd., Pune 412115, India.
| | - Alex Joseph
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal, Academy of Higher Education, Manipal, Karnataka 576104, India
| |
Collapse
|
44
|
Waliany S, Lin JJ, Gainor JF. Evolution of first versus next-line targeted therapies for metastatic non-small cell lung cancer. Trends Cancer 2025; 11:245-257. [PMID: 39890507 DOI: 10.1016/j.trecan.2025.01.005] [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: 09/25/2024] [Revised: 12/21/2024] [Accepted: 01/10/2025] [Indexed: 02/03/2025]
Abstract
The expanding armamentarium of targeted therapies has revolutionized treatment for metastatic oncogene-addicted lung cancers. For multiple subsets, such as those harboring EGFR mutations and fusions in ALK or ROS1, successive generation of increasingly potent, selective, and brain-penetrating targeted therapies have shifted the treatment paradigm towards preferential first-line use of next-generation drugs. This evolution in clinical practice provides a lens through which to review the lessons learned from drug development in oncogene-addicted lung cancers, guided by translational insights into tumor biology and mechanisms of therapeutic resistance. For oncogenic drivers that are less sensitive to single-agent targeted therapies, rationally designed combination strategies will be needed to enable first-line use of targeted agents.
Collapse
Affiliation(s)
- Sarah Waliany
- Cancer Center and Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jessica J Lin
- Cancer Center and Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Justin F Gainor
- Cancer Center and Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
45
|
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
|
46
|
Xia RX, Zou PC, Xie JT, Tang YB, Gong MM, Fan F, Aihemaiti A, Liu YQ, Shen Y, Zhou BBS, Zhu L, Lei HM. Dependence of NPPS creates a targetable vulnerability in RAS-mutant cancers. Acta Pharmacol Sin 2025; 46:728-739. [PMID: 39506063 PMCID: PMC11845791 DOI: 10.1038/s41401-024-01409-2] [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: 07/11/2024] [Accepted: 10/09/2024] [Indexed: 11/08/2024]
Abstract
RAS is the most frequently mutated oncoprotein for cancer driving. Understanding of RAS biology and discovery of druggable lynchpins in RAS pathway is a prerequisite for targeted therapy of RAS-mutant cancers. The recent identification of KRASG12C inhibitor breaks the "undruggable" curse on RAS and has changed the therapy paradigm of KRAS-mutant cancers. However, KRAS mutations, let alone KRASG12C mutation, account for only part of RAS-mutated cancers. Targeted therapies for cancers harboring other RAS mutations remain the urgent need. In this study we explored the pivotal regulatory molecules that allow for broad inhibition of RAS mutants. By comparing the expression levels of nucleotide pyrophosphatase (NPPS) in a panel of cell lines and the functional consequence of increased NPPS expression in RAS-mutant cells, we demonstrated that cancer cells with various kinds of RAS mutations depended on NPPS for growth and survival, and that this dependence conferred a vulnerability of RAS-mutant cancer to treatment of NPPS inhibition. RAS-mutant cells, compared with RAS-wildtype cells, bored and required an upregulation of NPPS. Transcriptomics and metabolomics analyses revealed a NPPS-dependent hyperglycolysis in RAS-mutant cells. We demonstrated that NPPS promoted glucose-derived glycolytic intermediates in RAS-mutant cells by enhancing its interaction with hexokinase 1 (HK1), the enzyme catalyzing the first committed step of glycolysis. Pharmacological inhibition of NPPS-HK1 axis using NPPS inhibitor Enpp-1-IN-1 or HK1 inhibitor 2-deoxyglucose (2-DG), or genetic interfere with NPPS suppressed RAS-mutant cancers in vitro and in vivo. In conclusion, this study reveals an unrecognized mechanism and druggable lynchpin for modulation of pan-mutant-RAS pathway, proposing a new potential therapeutic approach for treating RAS-mutant cancers.
Collapse
Affiliation(s)
- Rui-Xue Xia
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Pei-Chen Zou
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jun-Ting Xie
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ya-Bin Tang
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Miao-Miao Gong
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Fu Fan
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Ayinazhaer Aihemaiti
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yu-Qing Liu
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ying Shen
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Bin-Bing S Zhou
- Key Laboratory of Pediatric Hematology and Oncology Ministry of Health, Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Liang Zhu
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hui-Min Lei
- Department of Pharmacology and Chemical Biology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
47
|
Tao H, Yang B, Farhangian A, Xu K, Li T, Zhang ZY, Li J. Covalent-Allosteric Inhibitors: Do We Get the Best of Both Worlds? J Med Chem 2025; 68:4040-4052. [PMID: 39937154 DOI: 10.1021/acs.jmedchem.4c02760] [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: 02/13/2025]
Abstract
Covalent-allosteric inhibitors (CAIs) may achieve the best of both worlds: increased potency, long-lasting effects, and reduced drug resistance typical of covalent ligands, along with enhanced specificity and decreased toxicity inherent in allosteric modulators. Therefore, CAIs can be an effective strategy to transform many undruggable targets into druggable ones. However, CAIs are challenging to design. In this perspective, we analyze the discovery of known CAIs targeting three protein families: protein phosphatases, protein kinases, and GTPases. We also discuss how computational methods and tools can play a role in addressing the practical challenges of rational CAI design.
Collapse
Affiliation(s)
- Hui Tao
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Bo Yang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Atena Farhangian
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Ke Xu
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Tongtong Li
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Zhong-Yin Zhang
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Jianing Li
- Borch Department of Medicinal Chemistry and Molecular Pharmacology, Purdue Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
48
|
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
|
49
|
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
|
50
|
Kyoya T, Ishida H, Saitoh T, Itoh T. Preparation of ethynylsulfonamides and study of their reactivity with nucleophilic amino acids. Org Biomol Chem 2025; 23:1901-1908. [PMID: 39815739 DOI: 10.1039/d4ob01873g] [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: 01/18/2025]
Abstract
The development of covalent drugs, particularly those utilizing Michael acceptors, has garnered significant attention in recent pharmaceutical research due to the ability of such molecules to irreversibly inhibit protein function. This study focusses on the synthesis and evaluation of ethynylsulfonamides, which are predicted to have superior covalent binding ability, metabolic stability, and water solubility compared to traditional amides. We developed a straightforward synthesis method for ethynylsulfonamides and comprehensively evaluated the covalent binding abilities of these compounds using NMR with various nucleophilic amino acids in different solvents. Our results revealed that ethynylsulfonamides exhibit rapid and selective reactivity with cysteine residues, particularly in phosphate-buffered saline (PBS), where the reaction progressed quantitatively within five minutes. Notably, propynylsulfonamide demonstrated high reactivity and selectivity toward cysteine, suggestive of the significant potential of this molecule for applications in antibody-drug conjugates (ADCs) and other therapeutic areas where metabolic stability, water solubility, and selective reactivity are crucial.
Collapse
Affiliation(s)
- Tatsuhiro Kyoya
- Laboratory of Drug Design and Medicinal Chemistry, Showa Pharmaceutical University, 3-2-1 Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan.
| | - Hiroaki Ishida
- Laboratory of Drug Design and Medicinal Chemistry, Showa Pharmaceutical University, 3-2-1 Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan.
| | - Toshiaki Saitoh
- Division of Medicinal Informatics, Nihon Pharmaceutical University, 10281 Komuro, lna-machi, Kitaadachi-gun, Saitama 362-0806, Japan
| | - Toshimasa Itoh
- Laboratory of Drug Design and Medicinal Chemistry, Showa Pharmaceutical University, 3-2-1 Higashi-Tamagawagakuen, Machida, Tokyo 194-8543, Japan.
| |
Collapse
|