1
|
Tai Y, Kong L, Wang Y, Zhao D, Chen X, Wu Q, Hao J, Wang X, Liu X, Chen D, Li J, Hu Y, Zhang W, Yun CH, Zhan Q. Identification and characterization of Bufalin as a novel EGFR degrader. Cancer Lett 2025; 623:217715. [PMID: 40220852 DOI: 10.1016/j.canlet.2025.217715] [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: 12/27/2024] [Revised: 02/17/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) stands out as a common cancer type worldwide, characterized by its notably high rates of occurrence and mortality. The epidermal growth factor receptor (EGFR) is one of the main targets for cancer treatment as it is one of the genes whose expression is often altered by overexpression, amplification, and mutation in a variety of solid tumors. Substantial efforts have been made to develop EGFR-targeted therapeutic agents, including monoclonal antibodies and tyrosine kinase inhibitors (TKIs). However, these agents exhibited limited efficacy due to the emergence of acquired resistance. Therefore, novel treatment strategies targeting EGFR are urgently needed. Recent studies have identified a few natural compounds that can efficiently inhibit EGFR, indicating that natural products may be potential sources for the development of new EGFR inhibitors. Here, using the Drug Affinity Responsive Target Stability (DARTS) assay combined with liquid chromatography/tandem mass spectrometry analysis, co-crystal method, we discovered that Bufalin directly interacts with EGFR and causes EGFR endocytosis and degradation in the lysosome. Moreover, Bufalin exhibits superior anti-tumor activity compared with another EGFR TKIs. Our study identified Bufalin as the first natural small-molecule EGFR degrader, which suppresses EGFR signaling by inducing the degradation of EGFR via the endosome-lysosome pathway.
Collapse
Affiliation(s)
- Yidi Tai
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Lulu Kong
- Department of Biophysics, Department of Integration of Chinese and Western Medicine, and Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Yan Wang
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Dongyu Zhao
- Soochow University Cancer Institute, Suzhou, 215000, China
| | - Xu Chen
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Qingnan Wu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Jia Hao
- Department of Biophysics, Department of Integration of Chinese and Western Medicine, and Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China
| | - Xi Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Medical Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Xingyang Liu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Dongshao Chen
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Jinting Li
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Yuying Hu
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Weimin Zhang
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China; Department of Oncology, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, China.
| | - Cai-Hong Yun
- Department of Biophysics, Department of Integration of Chinese and Western Medicine, and Beijing Key Laboratory of Tumor Systems Biology, School of Basic Medical Sciences, Peking University, Beijing, 100191, China.
| | - Qimin Zhan
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Research Unit of Molecular Cancer Research, Chinese Academy of Medical Sciences, Beijing, 100021, China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, 518107, China; Department of Oncology, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen Peking University-Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, China; Soochow University Cancer Institute, Suzhou, 215000, China.
| |
Collapse
|
2
|
Wang Z, Pan BS, Manne RK, Chen J, Lv D, Wang M, Tran P, Weldemichael T, Yan W, Zhou H, Martinez GM, Shao J, Hsu CC, Hromas R, Zhou D, Qin Z, Lin HK, Li HY. CD36-mediated endocytosis of proteolysis-targeting chimeras. Cell 2025; 188:3219-3237.e18. [PMID: 40250420 DOI: 10.1016/j.cell.2025.03.036] [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: 05/23/2024] [Revised: 01/25/2025] [Accepted: 03/21/2025] [Indexed: 04/20/2025]
Abstract
Passive diffusion does not explain why many drugs are too large and/or too polar for rule-breaking membrane penetration, such as proteolysis-targeting chimeras (PROTACs, generally of a molecular weight > 800 Da). Here, using biotinylated chemical-probe-based target fishing and genetic knockdown/knockin approaches, we discovered that the membrane cluster of differentiation 36 (CD36) binds to and facilitates the uptake and efficacy of diverse PROTACs (e.g., SIM1-Me, MZ1, and clinical ARV-110) and large and/or polar small-molecule drugs (e.g., rapalink-1, rapamycin, navitoclax, birinapant, tubacin, and doxorubicin) via the CD36-mediated early endosome antigen 1 (EEA1)/Ras-related protein 5A (Rab5) endosomal cascade in vitro and/or in vivo. We then devised a novel chemical endocytic medicinal chemistry strategy to improve binding of PROTACs to CD36 using structural modifications via the prodrug approach, markedly enhancing PROTAC anti-tumor efficacy through spontaneously augmenting permeability and solubility.
Collapse
Affiliation(s)
- Zhengyu Wang
- Department of Pharmacology, The Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Department of Pharmaceutical Science, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Bo-Syong Pan
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27101, USA
| | - Rajesh Kumar Manne
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27101, USA
| | - Jungang Chen
- Department of Pathology and Winthrop P. Rockefeller Cancer Institute, School of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Dongwen Lv
- Department of Biochemistry and Structural Biology, Center for Innovative Drug Discovery, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Minmin Wang
- Department of Pharmacology, The Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Phuc Tran
- Department of Pharmaceutical Science, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Tsigereda Weldemichael
- Department of Pharmaceutical Science, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Wei Yan
- Department of Pharmacology, The Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Department of Pharmaceutical Science, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Hongfei Zhou
- Department of Pharmacology, The Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Gloria M Martinez
- Department of Pharmacology, The Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jingwei Shao
- Department of Pharmaceutical Science, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Che-Chia Hsu
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27101, USA
| | - Robert Hromas
- Department of Medicine and the Mays Cancer, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Daohong Zhou
- Department of Biochemistry and Structural Biology, Center for Innovative Drug Discovery, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Zhiqiang Qin
- Department of Pathology and Winthrop P. Rockefeller Cancer Institute, School of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Hui-Kuan Lin
- Department of Pathology, Duke University Medical Center, Durham, NC 27710, USA; Department of Cancer Biology, Wake Forest University School of Medicine, Winston Salem, NC 27101, USA.
| | - Hong-Yu Li
- Department of Pharmacology, The Long School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Department of Pharmaceutical Science, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
3
|
Yu Y, Hu W, Xu Y, Xu HB, Gao J. Advancements in delivery Systems for Proteolysis-Targeting Chimeras (PROTACs): Overcoming challenges and expanding biomedical applications. J Control Release 2025; 382:113719. [PMID: 40268200 DOI: 10.1016/j.jconrel.2025.113719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 04/25/2025]
Abstract
PROTAC (Proteolysis-Targeting Chimera), an emerging drug development strategy based on small molecule technology, has garnered widespread attention due to its high efficiency, broad applicability, low resistance, and dosage advantages. However, PROTAC molecules still exhibit certain limitations that require urgent resolution. Although significant progress has been made in designing PROTACs that target various disease-related proteins, research on drug delivery systems (DDS) for PROTACs remains relatively limited. This review aims to explore the critical role of delivery system design in addressing the inherent challenges associated with PROTAC molecules from a novel perspective. Beginning with five major challenges-insufficient targeting, poor pharmacokinetic properties, low cell permeability, limited accessibility, and the Hook effect-this article introduces formulation strategies to mitigate these deficiencies. It discusses potential solutions through targeted modifications, nano-delivery systems, intelligent response systems, and membrane biomimetic technologies, among others. Furthermore, it elucidates the mechanisms and principles underlying these approaches and analyzes the advantages of various delivery strategies. The insights provided in this review offer insights for designing delivery systems tailored to PROTACs with diverse characteristics for different disease applications.
Collapse
Affiliation(s)
- Yawei Yu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Weitong Hu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yihua Xu
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hong-Bin Xu
- Department of Pharmacy, The First Affiliated Hospital of Ningbo University, Zhe Jiang 315010, China.
| | - Jianqing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| |
Collapse
|
4
|
Zuo Y, Zhu Y, Luo P, Huang N, Yao K, Zhang K, Wang N, Gou X. Design, synthesis, and biological evaluation of novel PI3Kδ/HDAC6 dual inhibitors for the treatment of non-Hodgkin's lymphoma. Eur J Med Chem 2025; 296:117852. [PMID: 40516393 DOI: 10.1016/j.ejmech.2025.117852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Revised: 05/30/2025] [Accepted: 06/05/2025] [Indexed: 06/16/2025]
Abstract
PI3Kδ and HDAC6 inhibitors play important roles in the treatment of lymphoma. Herein, a novel series of purine-based PI3Kδ/HDAC6 dual inhibitors have been rationally designed and synthesized by incorporating an HDAC pharmacophore into our previously reported PI3Kδ inhibitor scaffold. Structure-activity relationship (SAR) studies led to the discovery of the lead compound 22E, which showed potent inhibitory activity towards PI3Kδ and HDAC6, with IC50 values of 2.4 nM and 6.2 nM, respectively. Compound 22E showed significantly enhanced antiproliferative activities against multiple non-Hodgkin's lymphoma (NHL) cells compared with either selective PI3Kδ inhibitor or HDAC6 inhibitor, with IC50 values of 34 nM and 53 nM against SU-DHL-6 and JEKO-1 cells, respectively. Subsequent mechanistic studies revealed that compound 22E effectively arrested the cell cycle in the G0/G1 phase and induced cell apoptosis in SU-DHL-6 and JEKO-1 cells. Meanwhile, 22E could simultaneously block the PI3K/AKT/mTOR signaling pathway as well as increase the acetylation of α-tubulin and histone H3 levels. In addition, 22E prevented tumor growth in both SU-DHL-6 and JEKO-1 xenograft models, and there was no observable toxicity. These results demonstrated that 22E is a promising lead candidate with novel antitumor mechanism for the treatment of NHL.
Collapse
Affiliation(s)
- Yueqi Zuo
- Institute of Basic and Translational Medicine, Xi'an Medical University, No.1 Xinwang Road, Xi'an 710021, Shaanxi Province, China.
| | - Yongxia Zhu
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Peng Luo
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Nan Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Kun Yao
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Ke Zhang
- Institute of Basic and Translational Medicine, Xi'an Medical University, No.1 Xinwang Road, Xi'an 710021, Shaanxi Province, China
| | - Ningyu Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China.
| | - Xingchun Gou
- Institute of Basic and Translational Medicine, Xi'an Medical University, No.1 Xinwang Road, Xi'an 710021, Shaanxi Province, China.
| |
Collapse
|
5
|
Chen Y, Qin Z, Wang Y, Gu B, Wang J, Zheng Y, Niu Y, Jia L. CD44-targeted virus-mimicking nanomedicine eliminates cancer stem cells and mitigates chemoresistance in head and neck squamous cell carcinoma. Mater Today Bio 2025; 32:101721. [PMID: 40242481 PMCID: PMC12002834 DOI: 10.1016/j.mtbio.2025.101721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/06/2025] [Accepted: 03/29/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer stem cells (CSCs) play critical roles in tumor growth, metastasis, and chemoresistance. Although several small-molecule inhibitors designed to inhibit CSCs have been investigated in clinical trials, their inadequate tumor targeting and potential off-target side effects have led to poor outcomes. A CD44-targeted virus-mimicking nanomedicine encapsulating the BMI1 inhibitor PTC209 (PTC209@VNP-HA) was designed to treat head and neck squamous cell carcinoma (HNSCC). We used a dendritic mesoporous silica nanoparticle (MSN) as the core for virus-mimicking nanoparticle (VNP) formation after adding shell particles to the MSN surface. The VNP surface was then modified with hyaluronic acid (HA), and PTC209 was adsorbed by mesopores to form PTC209@VNP-HA. In this system, HA is used to target CD44+ CSCs. The rough surface of VNP-HA provided better drug delivery efficiency than smooth nanoparticles modified with HA. VNP-HA enhanced the cancer inhibitory effect of PTC209 12-fold compared to the administration of free PTC209, leading to significantly higher bioavailability of PTC209. Both in vitro and in vivo assays showed that PTC209@VNP-HA inhibited cancer stemness, proliferation, and metastasis in HNSCC. Mechanistically, this inhibitory effect is closely associated with DNA damage/apoptosis signaling. Using a series of preclinical models in murine systems, we confirmed that PTC209@VNP-HA eliminated BMI1+ CSCs, and greatly inhibited the proliferation and metastasis of HNSCC when combined with cisplatin. This study investigated PTC209@VNP-HA as a novel and potentially transformative HNSCC treatment option that eliminates CSCs, prevents metastasis, and overcomes cisplatin resistance.
Collapse
Affiliation(s)
- Yiwen Chen
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, PR China
| | - Zhen Qin
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Yujia Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Baoxin Gu
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Jing Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100091, PR China
| | - Yunfei Zheng
- Department of Orthodontics, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Yuting Niu
- Central Laboratory, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
| | - Lingfei Jia
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, Beijing, 100081, PR China
- National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, PR China
- Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Institute of Advanced Clinical Medicine, Peking University, Beijing 100091, PR China
| |
Collapse
|
6
|
Vikal A, Maurya R, Patel BB, Sharma R, Patel P, Patil UK, Das Kurmi B. Protacs in cancer therapy: mechanisms, design, clinical trials, and future directions. Drug Deliv Transl Res 2025; 15:1801-1827. [PMID: 39614036 DOI: 10.1007/s13346-024-01754-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2024] [Indexed: 12/01/2024]
Abstract
Cancer develops as a result of changes in both genetic and epigenetic mechanisms, which lead to the activation of oncogenes and the suppression of tumor suppressor genes. Despite advancements in cancer treatments, the primary approach still involves a combination of chemotherapy, radiotherapy, and surgery, typically providing a median survival of approximately five years for patients. Unfortunately, these therapeutic interventions often bring about substantial side effects and toxicities, significantly impacting the overall quality of life for individuals undergoing treatment. Therefore, urgent need of research required which comes up with effective treatment of cancer. This review explores the transformative role of Proteolysis-Targeting Chimeras (PROTACs) in cancer therapy. PROTACs, an innovative drug development strategy, utilize the cell's protein degradation machinery to selectively eliminate disease-causing proteins. The review covers the historical background, mechanism of action, design, and structure of PROTACs, emphasizing their precision in targeting oncogenic proteins. The discussion extends to the challenges, nanotechnology applications, and ongoing clinical trials, showcasing promising results and clinical progress. The review concludes with insights into patents, future directions, and the potential impact of PROTACs in addressing dysregulated protein expression across various diseases. Overall, it provides a concise yet comprehensive overview for researchers, clinicians, and industry professionals involved in developing targeted therapies.
Collapse
Affiliation(s)
- Akash Vikal
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Rashmi Maurya
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Brij Bihari Patel
- Department of Respiratory Medicine, School of Excellence in Pulmonary Medicines, Netaji Subhash Chandra Bose Medical College, Jabalpur, 482003, Madhya Pradesh, India
| | - Rajeev Sharma
- Department of Pharmacy, Amity University, Gwalior, 474005, Madhya Pradesh, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Umesh K Patil
- Department of Pharmaceutical Sciences, Dr. Hari Singh Gour University, Sagar, 470003, India
| | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College of Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
7
|
Shi X, Hu C, Guo B, Zhang C, Xue Y, Yang Z, Wang F. Programmable Transcriptional-Translation Active Sensors for miRNA-Responsive Gene Imaging and Theranostics in Mammalians. ACS Sens 2025. [PMID: 40434179 DOI: 10.1021/acssensors.5c01016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
Synthetic gene circuits are logical circuits constructed by various biological elements based on engineering principles. However, the majority of current synthetic gene circuits are modulated through exogenous factors or endogenous inhibitory substances, which results in redundant structures and low efficiency, thereby greatly limiting their application scenarios. In this study, we developed a miCU sensor system comprising the Gal4-VP16 gene element, which enables real-time targeted monitoring of miR-9 and miR-124a during neural differentiation through transcriptional-translation two-step active regulation with a relatively low background signal. Additionally, the functional gene was replaced by P21 through the programmability of miCU, thereby achieving miRNA-mediated cell cycle arrest and suppression of cell migration in tumor cells. Furthermore, by strategically substituting the target miRNA with miR-155 and concurrently introducing the therapeutic gene Nrf2 into the miCU system, the integration of disease diagnosis and treatment in lipopolysaccharide (LPS)-induced acute liver injury (ALI) mouse models has been successfully achieved. Our study presented a programmable miRNA-responsive gene regulation platform, which may offer a robust tool for precise diagnosis and treatment in disease settings.
Collapse
Affiliation(s)
- Xiaorui Shi
- Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, China
- Guangzhou Institute of Technology, Xidian University, Guangzhou 510555, China
| | - Chong Hu
- Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, China
| | - Bin Guo
- Institute of Medical Engineering, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an 710061, China
| | - Chuanxian Zhang
- Institute of Medical Engineering, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yaru Xue
- Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, China
| | - Zeping Yang
- Engineering Research Center of Molecular and Neuroimaging, Ministry of Education, School of Life Science and Technology, Xidian University, Xi'an 710071, China
| | - Fu Wang
- Institute of Medical Engineering, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an 710061, China
| |
Collapse
|
8
|
Wu Y, Kawamoto Y, Sun J, Takahashi Y, Higuchi Y, Takakura Y. Improvement of Drug Release from an Aptamer Drug Conjugate Using Reductive-sensitive Linkers for Tumor-targeted Drug Delivery. AAPS J 2025; 27:95. [PMID: 40397061 DOI: 10.1208/s12248-025-01070-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 04/04/2025] [Indexed: 05/22/2025] Open
Abstract
The selective delivery of small molecule compounds such as Gemcitabine to tumor cells is a promising methodology for enhancing therapeutic efficacy and attenuating the side effects of anticancer drugs. Aptamers are useful as target-directed ligands for tumor-selective drug delivery due to their ability to bind specific proteins. However, the drug must be released from the aptamer after the conjugate is taken up by the cell to exert its pharmacological effect. In this study, we designed and synthesized a conjugate in which a linker cleaved by glutathione, which is highly expressed in tumor cells, was inserted between the aptamer (AS1411) and Gemcitabine. Almost all Gemcitabine was released from the conjugate after 30 min in the presence of 6 mM glutathione. AS1411 is known to bind to nucleolin, which is highly expressed on tumor cells. The cytotoxicity of the AS1411 and Gemcitabine conjugate with a disulfide bond on A549 cells was higher than that of the conjugate without a disulfide bond. Furthermore, the cytotoxicity of the disulfide-linked conjugate of AS1411 and Gemcitabine was higher in A549 cells than in MCF10A cells, which were used as the model of normal cells. These results indicate that disulfide conjugation enhanced the tumor cell-selective cytotoxicity of Gemcitabine with AS1411.
Collapse
Affiliation(s)
- You Wu
- Graduate School of Pharmaceutical Science, Kyoto University, 46-29 Yoshida-shimoadachicho, Sakyo-ku, Kyoto, Japan
| | - Yusuke Kawamoto
- Graduate School of Pharmaceutical Science, Kyoto University, 46-29 Yoshida-shimoadachicho, Sakyo-ku, Kyoto, Japan
| | - Jin Sun
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, China.
| | - Yuki Takahashi
- Graduate School of Pharmaceutical Science, Kyoto University, 46-29 Yoshida-shimoadachicho, Sakyo-ku, Kyoto, Japan
| | - Yuriko Higuchi
- Graduate School of Pharmaceutical Science, Kyoto University, 46-29 Yoshida-shimoadachicho, Sakyo-ku, Kyoto, Japan.
| | - Yoshinobu Takakura
- Graduate School of Pharmaceutical Science, Kyoto University, 46-29 Yoshida-shimoadachicho, Sakyo-ku, Kyoto, Japan.
| |
Collapse
|
9
|
Chen J, Wang W, Li M, Pan S, Zhao K. Advances in the diagnosis and treatment of urethral metastasis after urothelial carcinoma surgery: A case series and literature review of 3 total ureterectomy cases. Medicine (Baltimore) 2025; 104:e42329. [PMID: 40388736 PMCID: PMC12091633 DOI: 10.1097/md.0000000000042329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/15/2025] [Indexed: 05/21/2025] Open
Abstract
RATIONALE Urethral metastasis recurrence after surgery for urothelial carcinoma (UC) is a common clinical challenge, yet there is no established optimal treatment strategy to effectively control postoperative metastasis and recurrence. This case report evaluates the efficacy of total urethrectomy combined with adjuvant chemotherapy and PD-1 + antibody-drug conjugate (ADC) immunotherapy in managing UC urethral metastasis. PATIENT CONCERNS Three patients with a history of UC surgery presented with urethral metastasis recurrence, raising concerns about disease progression, treatment options, and long-term survival. DIAGNOSES All 3 patients were definitively diagnosed with UC urethral metastasis recurrence based on clinical, imaging, and histopathological examinations. INTERVENTIONS Each patient underwent total urethrectomy followed by adjuvant chemotherapy and PD-1 inhibitor + ADC combination immunotherapy. OUTCOMES Post-treatment follow-up, including serum tumor marker assessments and imaging studies, showed no significant recurrence. The patients exhibited prolonged overall survival and maintained a satisfactory postoperative quality of life. LESSONS Total urethrectomy combined with chemotherapy and PD-1+ADC immunotherapy may significantly improve outcomes in UC patients with postoperative urethral metastasis recurrence. This multimodal approach warrants further investigation in larger clinical studies.
Collapse
Affiliation(s)
- Jiajun Chen
- Department of Urology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Weihao Wang
- Department of Urology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Mengyao Li
- Department of Pathology, Shaoxing People’s Hospital, Shaoxing, China
| | - Shouhua Pan
- Department of Urology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Keyuan Zhao
- Department of Urology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| |
Collapse
|
10
|
Singh AA, Khan F, Song M. Alleviation of Neurological Disorders by Targeting Neurodegenerative-Associated Enzymes: Natural and Synthetic Molecules. Int J Mol Sci 2025; 26:4707. [PMID: 40429850 PMCID: PMC12112699 DOI: 10.3390/ijms26104707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2025] [Revised: 05/12/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
Neurological disorders, encompassing neurodegenerative and neuroinflammatory conditions, present significant public health and clinical challenges. Recent research has elucidated the pivotal role of various enzymes in the onset and progression of these disorders. This review explores the therapeutic potential of targeting these enzymes with natural and synthetic molecules. Key enzymes, including acetylcholinesterase, monoamine oxidase, beta-secretase, tau kinases, caspases, and cyclooxygenase-2, are implicated in diseases such as Alzheimer's disease, Parkinson's disease, and multiple sclerosis. Modulating these enzymes can alleviate symptoms, slow disease progression, or reverse pathological changes. Natural molecules derived from plants, microbes, seaweeds, and animals have long been noted for their therapeutic potential. Their ability to interact with specific enzymes with high specificity and minimal side effects makes them promising candidates for treatment. These natural agents provide a foundation for developing targeted therapies with improved safety profiles. Simultaneously, the development of synthetic chemistry has resulted in molecules designed to inhibit neurodegenerative enzymes with precision. This review examines the progress in creating small molecules, peptides, and enzyme inhibitors through sophisticated drug design techniques. It evaluates the efficacy, safety, and mechanisms of these synthetic agents, highlighting their potential for clinical application. The review offers a comprehensive overview of recent advancements in enzyme-targeted therapies for neurological disorders, covering both natural and synthetic molecules investigated in preclinical and clinical settings. It discusses the mechanisms through which these molecules exert their effects, the challenges faced in their development, and future research directions. By synthesizing current knowledge, this paper aims to illuminate the potential of enzyme-targeted interventions in managing neurological disorders, showcasing both the promise and limitations of these approaches.
Collapse
Affiliation(s)
- Alka Ashok Singh
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Fazlurrahman Khan
- Ocean and Fisheries Development International Cooperation Institute, Pukyong National University, Busan 48513, Republic of Korea
- International Graduate Program of Fisheries Science, Pukyong National University, Busan 48513, Republic of Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
11
|
Zhu S, Fu K, Li S, Yang C, Pan C, Wang X, Wang F, Yu X, To KKW, Fu L. Cardiotoxicity of small-molecule kinase inhibitors in cancer therapy. Exp Hematol Oncol 2025; 14:68. [PMID: 40346640 PMCID: PMC12063284 DOI: 10.1186/s40164-025-00660-5] [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: 02/03/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025] Open
Abstract
Cancer is one of the leading causes of death worldwide. Recent advances in precision oncology have enabled many specific cancer patient populations to respond well and achieve longer survival with small-molecule kinase inhibitors, which have become a new therapeutic strategy for tumors. Since 2001, the Food and Drug Administration has approved 108 and 63 new anticancer drugs for treating solid tumors and hematological malignancies, respectively, 89 of which belong to the large group of small-molecule kinase inhibitors (SMKIs). Compared to conventional chemotherapeutic agents such as cyclophosphamide, doxorubicin, and 5-FU, SMKIs offer better efficacy with fewer toxic side effects. Nevertheless, with the development of more novel SMKIs and their wider clinical application to a larger population of cancer patients, variable degrees of cardiotoxic adverse events have emerged for some SMKIs during cancer therapy. This review comprehensively summarizes the most updated progress in the cardiotoxicity of SMKIs in cancer therapy and discusses the new findings and mechanisms, which will provide emerging strategies for the prevention of cardiotoxicity caused by small molecule targeted drugs and the design of the next generation of low cardiotoxicity targeted drugs.
Collapse
Affiliation(s)
- Shuangli Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Kai Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Sijia Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Chuan Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Can Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Xueping Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Xiyong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangdong, Guangzhou Medical University, Guangzhou, 511436, China
| | - Kenneth Kin Wah To
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China.
| |
Collapse
|
12
|
Pan M, Fu Z, Hou H, Yang C, Li J. Proteolysis-Targeting Chimera (PROTAC): A Revolutionary Tool for Chemical Biology Research. SMALL METHODS 2025:e2500402. [PMID: 40342226 DOI: 10.1002/smtd.202500402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/24/2025] [Indexed: 05/11/2025]
Abstract
Proteolysis-targeting chimera (PROTAC) technology is a revolutionary tool for drug discovery that simultaneously recruits E3 ligase and the protein of interest to induce ubiquitination and subsequent proteasomal degradation. Since the inaugural PROTAC prototype emerged in 2001, this modality has garnered significant interest across academia and industry, catalyzing transformative applications in drug discovery and chemical biology. The field has evolved from foundational investigations into molecular design, structural optimization, and protein target extension to address more sophisticated challenges, such as structural analysis of ternary complexes, expansion of diversified therapeutic indications, and clinical translation studies. Recent progress across chemical, pharmaceutical, and biochemical sciences has reshaped PROTAC design paradigms, which in turn expanded the chemical biology toolkit. In this review, pivotal milestones are systematically chronicled in PROTAC development, evaluate emerging strategies for diversifying E3 ligase utilization and expanding the scope of degradable targets, and summarize a series of instrumental and biochemical methodologies that propelled sequential breakthroughs. Additionally, forward-looking trajectories are proposed to address current limitations and accelerate the clinical maturation of PROTAC-based therapeutics.
Collapse
Affiliation(s)
- Meichen Pan
- Beijing Life Science Academy, Beijing, 102209, China
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Zhongliang Fu
- Beijing Life Science Academy, Beijing, 102209, China
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Hongwei Hou
- Beijing Life Science Academy, Beijing, 102209, China
| | - Chunrong Yang
- New Cornerstone Science Laboratory, Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084, China
- Beijing Life Science Academy, Beijing, 102209, China
| | - Jinghong Li
- New Cornerstone Science Laboratory, Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing, 100084, China
- Beijing Life Science Academy, Beijing, 102209, China
- Center for Bioanalytical Chemistry, Hefei National Laboratory of Physical Science at Microscale, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
13
|
Qi H, Qiao Q, Sun X, Xing L. Survival Outcomes in EGFR-Mutant Non-Small Cell Lung Cancer With Brain Metastases: Kaplan-Meier and Cox Regression Analyses Across Treatment Stages. THE CLINICAL RESPIRATORY JOURNAL 2025; 19:e70085. [PMID: 40421655 PMCID: PMC12107365 DOI: 10.1111/crj.70085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 02/21/2025] [Accepted: 05/06/2025] [Indexed: 05/28/2025]
Abstract
BACKGROUND Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) have shown significant efficacy in patients with brain metastases (BMs) from EGFR-mutated non-small cell lung cancer (NSCLC). However, acquired resistance is inevitable, and clinical data addressing key questions across treatment stages remain insufficient, limiting the formulation of precise treatment strategies. METHODS This retrospective study analyzed 302 EGFR-mutant NSCLC patients with BMs treated at Shandong Cancer Hospital (2014-2022). Patients were divided into three cohorts: cohort A (first-/second-generation EGFR-TKIs without third-generation use), cohort B (first-/second-generation followed by third-generation EGFR-TKIs), and cohort C (first-line third-generation EGFR-TKIs). Survival outcomes were evaluated using Kaplan-Meier and Cox regression analyses across three treatment stages. Propensity score matching (PSM) adjusted for baseline imbalances. RESULTS Third-generation EGFR-TKIs demonstrated superior progression-free survival (PFS) in first-line therapy compared to earlier-generation agents (median PFS1: 14.2 vs. 11.2 months; p = 0.0021), particularly for intracranial control (median iPFS1: 18.0 vs. 12.2 months; p = 0.0058). Patients with uncommon EGFR mutations had significantly shorter PFS on third-generation EGFR-TKIs than those with common mutations (4.4 vs. 12.9 months; p = 0.012). After resistance, combination therapy with immune checkpoint inhibitors (ICIs), antiangiogenics, and chemotherapy extended overall survival (OS) versus non-ICI regimens (median OS2: 17.3 vs. 10.4 months; p = 0.004). CONCLUSIONS Third-generation EGFR-TKIs are effective first-line options for BMs but show limited efficacy against uncommon mutations. Post-resistance regimens integrating ICIs, antiangiogenics, and chemotherapy may improve survival. Reassessment of genetic and PD-L1 status is critical for guiding sequential therapy.
Collapse
Affiliation(s)
- Haoran Qi
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Qiang Qiao
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| | - Xiaorong Sun
- Department of Nuclear MedicineShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical ScienceJinanShandongChina
| | - Ligang Xing
- Department of Radiation OncologyShandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical SciencesJinanShandongChina
| |
Collapse
|
14
|
Liu L, Zhang S, Ren Y, Wang R, Zhang Y, Weng S, Zhou Z, Luo P, Cheng Q, Xu H, Ba Y, Zuo A, Liu S, Liu Z, Han X. Macrophage-derived exosomes in cancer: a double-edged sword with therapeutic potential. J Nanobiotechnology 2025; 23:319. [PMID: 40287762 PMCID: PMC12034189 DOI: 10.1186/s12951-025-03321-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/11/2025] [Indexed: 04/29/2025] Open
Abstract
Solid cancer contains a complicated communication network between cancer cells and components in the tumor microenvironment (TME), significantly influencing the progression of cancer. Exosomes function as key carriers of signaling molecules in these communications, including the intricate signalings of tumor-associated macrophages (TAMs) on cancer cells and the TME. With their natural lipid bilayer structures and biological activity that relates to their original cell, exosomes have emerged as efficient carriers in studies on cancer therapy. Intrigued by the heterogeneity and plasticity of both macrophages and exosomes, we regard macrophage-derived exosomes in cancer as a double-edged sword. For instance, TAM-derived exosomes, educated by the TME, can promote resistance to cancer therapies, while macrophage-derived exosomes generated in vitro have shown favorable potential in cancer therapy. Here, we depict the reasons for the heterogeneity of TAM-derived exosomes, as well as the manifold roles of TAM-derived exosomes in cancer progression, metastasis, and resistance to cancer therapy. In particular, we emphasize the recent advancements of modified macrophage-derived exosomes in diverse cancer therapies, arguing that these modified exosomes are endowed with unique advantages by their macrophage origin. We outline the challenges in translating these scientific discoveries into clinical cancer therapy, aiming to provide patients with safe and effective treatments.
Collapse
Affiliation(s)
- Long Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Siying Zhang
- Medical School of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Ruizhi Wang
- Medical School of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, 450052, Henan, China.
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
15
|
Herbel N, Postel-Vinay S. Targeted protein degradation in oncology: novel therapeutic opportunity for solid tumours? Mol Oncol 2025. [PMID: 40266852 DOI: 10.1002/1878-0261.70034] [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: 02/27/2025] [Revised: 03/24/2025] [Accepted: 03/16/2025] [Indexed: 04/25/2025] Open
Abstract
Targeted and immune therapies have improved patient outcomes in selected diseases. Still, resistance inevitably occurs, and a significant portion of the proteome remains undruggable due to target localisation, structural or functional constraints. Targeted protein degraders (TPDs) represent a promising strategy to expand druggable targets by redirecting the ubiquitin-proteasome system to selectively degrade proteins of interest (POI). TPDs include proteolysis-targeting chimeras (PROTACs), which are heterobifunctional molecules that create a ternary complex with the POI and the E3 ligase, and molecular glues (MGs), which are monovalent small molecules that create an interface between an E3 ligase and the POI. Here, we provide a viewpoint on novel therapeutic opportunities offered by TPDs, notably through the targeting of previously undruggable proteins or overcoming some resistance mechanisms. We further present challenges that will need to be addressed in order to optimise clinical development, including dose optimisation, patient selection and drug delivery.
Collapse
Affiliation(s)
- Noé Herbel
- ERC Chromatin Remodelling, DNA Repair and Epigenetics Laboratory, INSERM Unit U981, Gustave Roussy Institute, Villejuif, France
- Drug Development Department, Gustave Roussy Institute, Villejuif, France
- Paris-Saclay University, Paris-Sud University XI, Faculty of Medicine, Le Kremlin-Bicêtre, France
| | - Sophie Postel-Vinay
- ERC Chromatin Remodelling, DNA Repair and Epigenetics Laboratory, INSERM Unit U981, Gustave Roussy Institute, Villejuif, France
- Drug Development Department, Gustave Roussy Institute, Villejuif, France
- University College of London Cancer Institute, UK
| |
Collapse
|
16
|
Wang F, Chen Y, Huang R, Lu D, Zhang J, Yang Y, Dang H, Liu M, Chen Z, Sun X, Wang Z. Identification of SURF4 and RALGAPA1 as promising therapeutic targets in glioblastoma and pan-cancer through integrative multi-omics, CRISPR-Cas9 screening and prognostic meta-analysis. Cancer Immunol Immunother 2025; 74:175. [PMID: 40249536 PMCID: PMC12008100 DOI: 10.1007/s00262-025-04034-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/24/2025] [Indexed: 04/19/2025]
Abstract
Glioblastoma (GBM) is the most aggressive and malignant type of primary brain tumor, with a median survival time of less than two years and a uniformly poor prognosis, despite multimodal therapeutic approaches, which highlights an urgent need for novel therapeutic targets. In this study, by integrative multi-omics analysis from CPTAC database, DepMap database and seven independent GBM cohorts, four hub genes (CD44, SURF4, IGSF3 and RALGAPA1) were identified as essential genes regulated by cancer driver genes with robust prognostic value. GBM multi-omics data from public and in-house cohorts validated that CD44 and SURF4 might be synthetic lethal partners of loss-of-function tumor suppressor genes. Analysis for immune-related pathway activity revealed complex regulation relationships of the four hub genes in tumor microenvironment (TME). Further investigation on SURF4 in pathway activity, immune therapy response and drug sensitivity proposed that SURF4 emerged as a promising therapeutic target for GBM, even for pan-cancer. Pan-cancer multi-omics exploration suggested that RALGAPA1 may be a tumor suppressor gene. By screening the first-generation and second-generation DepMap database, four genes (CCDC106, GAL3ST1, GDI2 and HSF1) might be considered as synthetic targets after mutation of RALGAPA1 as a tumor suppressor gene.
Collapse
Affiliation(s)
- Fei Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Yuxuan Chen
- Suzhou Medical College of Soochow University, Suzhou, 215127, Jiangsu Province, China
| | - Run Huang
- Suzhou Medical College of Soochow University, Suzhou, 215127, Jiangsu Province, China
| | - Dengfeng Lu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Juyi Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu Province, China
| | - Yanbo Yang
- Department of Oncology, Division of Pediatric Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Hanhan Dang
- China-Japan Friendship Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100000, China
| | - Meirong Liu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, 215000, Jiangsu Province, China
| | - Zhouqing Chen
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu Province, China.
| | - Xiaoou Sun
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu Province, China.
| | - Zhong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu Province, China.
| |
Collapse
|
17
|
Ma Y, Zhou Y, Xie G, Chen H, Huangfu Y, Shen L, Liu Y, Wang P. HEX-1 reduces colitis-driven colorectal cancer via inactivating the prolyl isomerase PIN1 sensitization and remodeling the gut microbiota. Discov Oncol 2025; 16:565. [PMID: 40251462 PMCID: PMC12008109 DOI: 10.1007/s12672-025-02338-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/08/2025] [Indexed: 04/20/2025] Open
Abstract
Metabolic reprogramming, a pivotal hallmark of cancer, plays a crucial role in both the initiation and progression of colorectal cancer (CRC). Despite the vast unknowns surrounding the identity and biological activities of most natural metabolites in diseases, our study, utilizing native metabolomics results through GC-MS/MS, identified a small molecule, 4,4-Dimethyl-2-cyclohexen-1-one, named HEX-1 in the serum of CRC patients. We have further explored and assessed its biological activities. HEX-1 suppressed the proliferation of cancer cells and tumorigenesis via the inactivation and sensitization of PIN1. Notably, HEX-1 exhibits similar functional effects as all-trans retinoic acid (atRA) but stands out by not inducing the degradation of PIN1 mRNA or protein expression, unlike biological compounds associated with atRA. HEX-1 demonstrated the ability to induce G1/S arrest in vitro and ameliorate the progression of inflammatory CRC in mice by remodeling the gut microbiota. As volatile organic compounds (VOCs), HEX-1 could be detected feasibly. Its unique ability to penetrate whole cell populations positions it as a promising approach for cancer therapy and as an enhancer for chemotherapy and immunotherapy. The findings suggest that HEX-1 holds the potential as a valuable addition to the armamentarium against CRC.
Collapse
Affiliation(s)
- Yanhui Ma
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yunlan Zhou
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Guohua Xie
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Hui Chen
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yuchan Huangfu
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Lisong Shen
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, 200092, China
| | - Yi Liu
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, 200092, China.
| | - Ping Wang
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, 200092, China.
| |
Collapse
|
18
|
Hu J, Arvejeh PM, Bone S, Hett E, Marincola FM, Roh KH. Nanocarriers for cutting-edge cancer immunotherapies. J Transl Med 2025; 23:447. [PMID: 40234928 PMCID: PMC12001629 DOI: 10.1186/s12967-025-06435-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/26/2025] [Indexed: 04/17/2025] Open
Abstract
Cancer immunotherapy aims to harness the body's own immune system for effective and long-lasting elimination of malignant neoplastic tissues. Owing to the advance in understanding of cancer pathology and immunology, many novel strategies for enhancing immunological responses against various cancers have been successfully developed, and some have translated into excellent clinical outcomes. As one promising strategy for the next generation of immunotherapies, activating the multi-cellular network (MCN) within the tumor microenvironment (TME) to deploy multiple mechanisms of action (MOAs) has attracted significant attention. To achieve this effectively and safely, delivering multiple or pleiotropic therapeutic cargoes to the targeted sites of cancerous tissues, cells, and intracellular organelles is critical, for which numerous nanocarriers have been developed and leveraged. In this review, we first introduce therapeutic payloads categorized according to their predicted functions in cancer immunotherapy and their physicochemical structures and forms. Then, various nanocarriers, along with their unique characteristics, properties, advantages, and limitations, are introduced with notable recent applications in cancer immunotherapy. Following discussions on targeting strategies, a summary of each nanocarrier matching with suitable therapeutic cargoes is provided with comprehensive background information for designing cancer immunotherapy regimens.
Collapse
Affiliation(s)
- Joyce Hu
- Translational and Advanced Medicine (TAM) Biosciences, Nashville, TN, 37011, USA
| | - Pooria M Arvejeh
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sydney Bone
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA
| | - Erik Hett
- Translational and Advanced Medicine (TAM) Biosciences, Nashville, TN, 37011, USA
| | | | - Kyung-Ho Roh
- Department of Chemical and Materials Engineering, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA.
- Biotechnology Science and Engineering Program, The University of Alabama in Huntsville, Huntsville, AL, 35899, USA.
| |
Collapse
|
19
|
Chai S, Cui J, Sun Y, Wang X, Cai C. Exploring Novel Therapeutic Targets in Breast Cancer via Comprehensive Omics Profiling and Experimental Verification. BIOLOGY 2025; 14:405. [PMID: 40282270 PMCID: PMC12025194 DOI: 10.3390/biology14040405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Breast cancer is the leading cause of cancer-related deaths among women worldwide. Deciphering the molecular mechanisms of breast cancer is crucial for developing targeted therapeutic approaches. METHODS This study analyzed gene expression profiles from the Gene Expression Omnibus (GEO) database to identify differentially expressed genes (DEGs) in breast cancer. Mendelian randomization (MR) analysis was then employed using publicly available eQTL databases to evaluate potential causal relationships between these DEGs and breast cancer. Enrichment analyses were further conducted to explore their functional significance. Furthermore, external validation of co-expressed genes was conducted using The Cancer Genome Atlas (TCGA) database. In vitro functional assays and drug sensitivity analyses were performed on selected target genes to validate their roles in breast cancer pathogenesis and treatment. RESULTS A total of 1052 upregulated and 1380 downregulated genes were identified in breast cancer. Additionally, MR analysis revealed 12 significant co-expressed genes potentially contributing to breast cancer pathogenesis. These genes were primarily enriched in lipid metabolism and immune responses via regulating microRNA functions and AMPK signaling. Validation through the TCGA database confirmed differential expression of these genes in breast cancer tissues. Strikingly, functional assays of the less-reported genes DNASE2 and ATOH8 demonstrated their involvement in breast cancer pathogenesis through modulating proliferation, migration, and invasion of cancer cells. Notably, several commonly used clinical drugs for breast cancer management, such as 5-Fluorouracil, exhibited dramatically increased sensitivity to DNASE2 and ATOH8 expression. CONCLUSIONS Our study provides novel insights into the molecular basis of breast cancer pathogenesis and identifies promising therapeutic strategies for this condition.
Collapse
Affiliation(s)
- Shengjun Chai
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810008, China; (S.C.); (J.C.); (Y.S.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University Medical College, Xining 810008, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University Medical College, Xining 810008, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University Medical College, Xining 810008, China
- Department of Surgical Oncology, The Affiliated Hospital of Qinghai University, Xining 810001, China
| | - Jiayong Cui
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810008, China; (S.C.); (J.C.); (Y.S.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University Medical College, Xining 810008, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University Medical College, Xining 810008, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University Medical College, Xining 810008, China
| | - Yinuo Sun
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810008, China; (S.C.); (J.C.); (Y.S.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University Medical College, Xining 810008, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University Medical College, Xining 810008, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University Medical College, Xining 810008, China
- Department of Economics, School of Finance and Economics, Qinghai University, Xining 810016, China
| | - Xiaowu Wang
- Department of Surgical Oncology, The Affiliated Hospital of Qinghai University, Xining 810001, China
| | - Chunmei Cai
- Research Center for High Altitude Medicine, Qinghai University Medical College, Xining 810008, China; (S.C.); (J.C.); (Y.S.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University Medical College, Xining 810008, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine (Qinghai-Utah Joint Key Laboratory of Plateau Medicine), Qinghai University Medical College, Xining 810008, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University Medical College, Xining 810008, China
| |
Collapse
|
20
|
Martinengo B, Diamanti E, Uliassi E, Bolognesi ML. Medicinal Chemistry: A Key Driver in Achieving the Global Sustainable Development Goals. J Med Chem 2025; 68:6916-6931. [PMID: 40112026 PMCID: PMC11998007 DOI: 10.1021/acs.jmedchem.4c03016] [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: 12/09/2024] [Revised: 02/13/2025] [Accepted: 03/07/2025] [Indexed: 03/22/2025]
Abstract
In 2015, the United Nations officially launched the Sustainable Development Goals (SDGs) as "the blueprint to achieve a better and more sustainable future for all. They address the global challenges we face, including those related to poverty, inequality, climate change, environmental degradation, peace and justice. The 17 Goals are all interconnected, in order to leave no one behind, it is important that we achieve them all by 2030". Here, we have embedded medicinal chemistry as a key player to achieve SDGs. We firmly believe that medicinal chemistry can and must contribute to a sustainable future and a better world with an improved quality of life for all. We have taken a critical look at each of the SDGs, dividing them into Priority and Foundational, and analyzed how medicinal chemistry has an impact on each of them. Although much has been done, we are determined to make progress in this area.
Collapse
Affiliation(s)
- Bianca Martinengo
- Department of Pharmacy and
Biotechnology, Alma Mater Studiorum - University
of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Eleonora Diamanti
- Department of Pharmacy and
Biotechnology, Alma Mater Studiorum - University
of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Elisa Uliassi
- Department of Pharmacy and
Biotechnology, Alma Mater Studiorum - University
of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| | - Maria Laura Bolognesi
- Department of Pharmacy and
Biotechnology, Alma Mater Studiorum - University
of Bologna, Via Belmeloro 6, 40126 Bologna, Italy
| |
Collapse
|
21
|
Li J, Li X, Chen Y, Wang Y, Wang B, Zhang X, Zhang N. Mesothelin expression prediction in pancreatic cancer based on multimodal stochastic configuration networks. Med Biol Eng Comput 2025; 63:1117-1129. [PMID: 39641869 DOI: 10.1007/s11517-024-03253-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 11/25/2024] [Indexed: 12/07/2024]
Abstract
Predicting tumor biomarkers with high precision is essential for improving the diagnostic accuracy and developing more effective treatment strategies. This paper proposes a machine learning model that utilizes CT images and biopsy whole slide images (WSI) to classify mesothelin expression levels in pancreatic cancer. By combining multimodal learning and stochastic configuration networks, a radiopathomics mesothelin-prediction system named RPMSNet is developed. The system extracts radiomic and pathomic features from CT images and WSI, respectively, and sends them into stochastic configuration networks for the final prediction. Compared to traditional radiomics or pathomics, this system has the capability to capture more comprehensive image features, providing a multidimensional insight into tissue characteristics. The experiments and analyses demonstrate the accuracy and effectiveness of the system, with an area under the curve of 81.03%, an accuracy of 73.67%, a sensitivity of 71.54%, a precision of 76.78%, and a F1-score of 72.61%, surpassing both single-modality and dual-modality models. RPMSNet highlights its potential for early diagnosis and personalized treatment in precision medicine.
Collapse
Affiliation(s)
- Junjie Li
- College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Xuanle Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518071, China
- Department of Radiology, Medical Imaging Research Institute, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Yingge Chen
- College of Sciences, Northeastern University, Shenyang, 110819, China
| | - Yunling Wang
- Department of Radiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Binjie Wang
- Department of Radiology, Medical Imaging Research Institute, Huaihe Hospital of Henan University, Kaifeng, 475000, China.
| | - Xuefeng Zhang
- College of Sciences, Northeastern University, Shenyang, 110819, China.
| | - Na Zhang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518071, China.
| |
Collapse
|
22
|
Russo L, Siena LM, Farina S, Pastorino R, Boccia S, Ioannidis JPA. High-impact trials with genetic and -omics information focus on cancer mutations, are industry-funded, and less transparent. J Clin Epidemiol 2025; 180:111676. [PMID: 39826627 DOI: 10.1016/j.jclinepi.2025.111676] [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: 10/29/2024] [Revised: 12/20/2024] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVES To assess how genetics and -omics information is used in the most cited recent clinical trials and to evaluate industry involvement and transparency patterns. STUDY DESIGN AND SETTING This is a meta-research evaluation using a previously constructed database of the 600 most cited clinical trials published from 2019 to 2022. Trials that utilized genetic or -omics characterization of participants in the trial design, analysis, and results were considered eligible. RESULTS 132 (22%) trials used genetic or -omics information, predominantly for detection of cancer mutations (n = 101). Utilization included eligibility criteria (n = 59), subgroup analysis (n = 82), and stratification factor in randomization (n = 14). Authors addressed the relevance in the conclusions in 82 studies (62%). 102 studies (77%) provided data availability statements and six had data already available. Most studies had industry funding (n = 111 [84.0%]). Oncology trials were more likely to be industry-funded (90.1% vs 64.5%, P = .001), to have industry-affiliated analysts (43.6% vs 22.6%, P = .036), and to favor industry-sponsored interventions (83.2% vs 58.1% P = .004). When compared to other trials, genetic and -omics trials were more likely to be funded by industry (84% vs 63.9%, P < .001) and tended to be less likely to have full protocols (P = .018) and statistical plans (P = .04) available. CONCLUSION Our study highlights the current underutilization of genetic and -omics technologies beyond testing for cancer mutations. Industry involvement in these trials appears to be more substantial and transparency is more limited, raising concerns about potential bias.
Collapse
Affiliation(s)
- Luigi Russo
- Section of Hygiene, Department of Life Sciences and Public Health, Universita Cattolica del Sacro Cuore, Rome, Italy
| | - Leonardo M Siena
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Sara Farina
- Section of Hygiene, Department of Life Sciences and Public Health, Universita Cattolica del Sacro Cuore, Rome, Italy
| | - Roberta Pastorino
- Section of Hygiene, Department of Life Sciences and Public Health, Universita Cattolica del Sacro Cuore, Rome, Italy; Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Stefania Boccia
- Section of Hygiene, Department of Life Sciences and Public Health, Universita Cattolica del Sacro Cuore, Rome, Italy; Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - John P A Ioannidis
- Stanford Prevention Research Center, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA; Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA; Meta-Research Innovation Center at Stanford (METRICS), Stanford University, Stanford, CA, USA.
| |
Collapse
|
23
|
Tandon V, Fistrovich A, Nogales J, Ferro F, Rokey SN, Cabel C, Miller AD, Yagel M, Duncan C, Atmasidha A, Sharma I, Wilms G, Williams K, Elliott R, Chavez T, Shaw Y, McMahon A, Ginn S, Basantes LE, Bedard N, Gokhale V, Ellis N, Prescott AR, Smith SJ, Rahman R, Becker W, Read KD, Chalmers AJ, Carragher N, Masson GR, Montfort W, Thorne C, Hulme C, Banerjee S. Dyr726, a brain-penetrant inhibitor of PI3Kα, Type III receptor tyrosine kinases, and WNT signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645490. [PMID: 40196701 PMCID: PMC11974928 DOI: 10.1101/2025.03.26.645490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
The vast majority of clinical small molecule multi-kinase inhibitors (mKI) report abject failures in targeting cancers with high stem cell contents like high-grade glioma and colorectal cancers. The FDA-approved mKIs to date ablate receptor tyrosine kinase signaling but do not target the paradoxical WNT signaling which is a key survival driver for the self-renewing cancer stem cells. The WNT pathway enhances cancer plasticity and triggers relapse of highly heterogenous tumours. Using de novo synthesis and structure-activity-relationship (SAR) studies with blood-brain-barrier (BBB) penetrant mKI scaffolds, we designed a highly potent and selective small molecule inhibitor of PI3Kα, PDGFR/KIT, and the WNT pathway denoted Dyr726. Dyr726 is superior to clinical mKIs and inhibits PI3K-AKT-mTOR and WNT-pathway signaling at multiple nodes thereby impeding proliferation, invasion, and tumour growth. Phospho-proteomic, structural, and target engagement analyses, combined with in vitro, in vivo efficacy, and pharmacokinetic studies reveal that Dyr726 is a brain-penetrant small molecule which effectively reduces tumour volume and extends survival of murine orthotopic models. Our current work establishes a first-in-class brain penetrant small molecule mKI which simultaneously antagonize the PI3K-AKT-mTOR and WNT pathways in preclinical cancer stem cell cultures, adult and pediatric primary organoids, and orthotopic murine models with positive efficacy in combination with clinical standard of care.
Collapse
Affiliation(s)
- Vasudha Tandon
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Alessandra Fistrovich
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA
- Division of Drug Discovery Development, Department of Pharmacology & Toxicology, The College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Joaquina Nogales
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Febe Ferro
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Samantha N Rokey
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA
- Division of Drug Discovery Development, Department of Pharmacology & Toxicology, The College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Carly Cabel
- University of Arizona Cancer Center, Department of Cellular and Molecular Medicine, College of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Amy Dunne Miller
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Mary Yagel
- University of Arizona Cancer Center, Department of Cellular and Molecular Medicine, College of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Christina Duncan
- Drug Discovery Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Aditi Atmasidha
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Ira Sharma
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - Gerrit Wilms
- Institute of Pharmacology and Toxicology, RWTH Aachen University, 52074 Aachen, Germany
| | - Karin Williams
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | - Richard Elliott
- The Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XR, UK
| | - Timothy Chavez
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA
| | - Yeng Shaw
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA
| | - Aidan McMahon
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA
| | - Sean Ginn
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA
| | - L. Emilia Basantes
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA
| | - Nathan Bedard
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA
| | - Vijay Gokhale
- Division of Drug Discovery Development, Department of Pharmacology & Toxicology, The College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Nathan Ellis
- University of Arizona Cancer Center, Department of Cellular and Molecular Medicine, College of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Alan R Prescott
- Dundee Imaging Facility, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | - Stuart J Smith
- Children’s Brain Tumour Research Centre, School of Medicine, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Ruman Rahman
- Children’s Brain Tumour Research Centre, School of Medicine, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Walter Becker
- Institute of Pharmacology and Toxicology, RWTH Aachen University, 52074 Aachen, Germany
| | - Kevin D Read
- Drug Discovery Unit, School of Life Sciences, University of Dundee, Dundee DD1 5EH, UK
| | | | - Neil Carragher
- The Institute of Genetics and Cancer, University of Edinburgh, Edinburgh EH4 2XR, UK
| | - Glenn R Masson
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| | - William Montfort
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA
| | - Curtis Thorne
- University of Arizona Cancer Center, Department of Cellular and Molecular Medicine, College of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Christopher Hulme
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, USA
- Division of Drug Discovery Development, Department of Pharmacology & Toxicology, The College of Pharmacy, The University of Arizona, Tucson, AZ, 85721, USA
| | - Sourav Banerjee
- Division of Cancer Research, School of Medicine, University of Dundee, Dundee DD1 9SY, UK
| |
Collapse
|
24
|
Senekowitsch S, Freitag T, Dubinski D, Freiman TM, Maletzki C, Hinz B. Validation of an LC-MS/MS Method for the Simultaneous Intracellular Quantification of the CDK4/6 Inhibitor Abemaciclib and the EZH2 Inhibitors GSK126 and Tazemetostat. Pharmaceutics 2025; 17:433. [PMID: 40284428 PMCID: PMC12030624 DOI: 10.3390/pharmaceutics17040433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Background: Inhibitors of cyclin-dependent kinases (CDKs) and epigenetic modifier enhancer of zeste homolog 2 (EZH2) have emerged as promising options in the pharmacotherapy of malignant tumors. Recently, we demonstrated synergistic antitumor effects of the CDK4/6 inhibitor abemaciclib and the EZH2 inhibitors GSK126 or tazemetostat in patient-derived glioblastoma (GBM) models. Importantly, all three drugs are substrates of the two most important plasma membrane multidrug transporters ABCB1 and ABCG2, with abemaciclib and tazemetostat also being inhibitors of these proteins. Methods: To investigate whether increased intracellular accumulation of either of the two drugs used in combination could have contributed to corresponding synergisms, we developed a simple LC-MS/MS method for simultaneous detection of the three substances in cell culture lysates. The method was validated in accordance with the current International Council for Harmonization of Technical Requirements for Pharmaceuticals for Human Use (ICH) guideline M10 on bioanalytical method validation and study sample analysis. Results: All acceptance criteria were met. Subsequent analysis of intracellular drug concentrations confirmed increased cellular uptake of tazemetostat in the presence of abemaciclib in both GBM cell lines studied compared to single agent treatment. A comparable pattern was also observed for GSK126, but in only one of the two cell lines used. Conclusions: In conclusion, the observed synergistic antitumor effect could be partly due to increased intracellular accumulation, although this alone is certainly not sufficient to explain it. Overall, the developed method provides a valuable approach for characterizing interactions at the transport level and for predicting the efficiency of both anticancer substance classes in different cell lines.
Collapse
Affiliation(s)
- Stefan Senekowitsch
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany;
| | - Thomas Freitag
- Department of Medicine, Clinic III—Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, 18057 Rostock, Germany
- Department of Neurosurgery, Rostock University Medical Center, 18057 Rostock, Germany
| | - Daniel Dubinski
- Department of Neurosurgery, Rostock University Medical Center, 18057 Rostock, Germany
| | - Thomas M. Freiman
- Department of Neurosurgery, Rostock University Medical Center, 18057 Rostock, Germany
| | - Claudia Maletzki
- Department of Medicine, Clinic III—Hematology, Oncology, Palliative Medicine, Rostock University Medical Center, 18057 Rostock, Germany
| | - Burkhard Hinz
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, Schillingallee 70, 18057 Rostock, Germany;
| |
Collapse
|
25
|
Bremer HJ, Pflum MKH. Chemoproteomic Profiling of PKA Substrates with Kinase-catalyzed Crosslinking and Immunoprecipitation (K-CLIP). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.23.644825. [PMID: 40166339 PMCID: PMC11957104 DOI: 10.1101/2025.03.23.644825] [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
Phosphorylation is a highly regulated protein post-translational modification catalyzed by kinases. Kinases and phosphorylated proteins are key players in a myriad of cellular events, including cell signaling. When cell signaling networks are improperly regulated by kinases, various pathologies can arise, such as cancers and neurodegenerative disease. With critical roles in normal and disease biology, kinase-substrate interactions must be thoroughly characterized. Previously, the chemoproteomic method, kinase-catalyzed crosslinking and immunoprecipitation (K-CLIP), was developed to identify the kinases of a phosphoprotein substrate of interest. Here, K-CLIP was modified to profile the substrates of a kinase of interest. Specifically, the substrate profile of cAMP-dependent protein kinase (PKA) was studied with K-CLIP using a new ATP analog, ATP-alkyne aryl azide. Kinase-focused K-CLIP discovered SMC3 as a PKA substrate. With versatility for any kinase or phosphoprotein substrate of interest, K-CLIP will expand our understanding of kinase-mediated cell biology in healthy and diseased states.
Collapse
|
26
|
Zhang X, Zhuo J, Wang D, Zhu X. Supramolecular Polymers for Drug Delivery. Chemistry 2025; 31:e202404617. [PMID: 39961052 DOI: 10.1002/chem.202404617] [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: 12/15/2024] [Indexed: 03/21/2025]
Abstract
Supramolecular polymers are constructed through highly reversible and directionally specific non-covalent interactions between monomer units. This unique feature enables supramolecular polymers to undergo controlled structural reconfiguration and functional transformation in response to external stimuli, imparting them with high environmental responsiveness and self-healing properties. In particular, supramolecular polymers exhibit several specific advantages compared to conventional polymers, such as inherent degradability, the ease of preparation and the incorporation of functional units, and smart responsiveness to various biological stimuli. These characters make supramolecular polymers promising candidates for intelligent drug delivery systems in complex biological environments. In this review, we comprehensively summarize the latest developments and representative achievements of supramolecular polymers in drug delivery fields, focusing primarily on the design and synthesis, the properties and functionalities, and the practical applications of supramolecular polymers in small molecule drug delivery, gene therapy, and protein delivery. Finally, we highlight future research directions, focusing on multifunctionality, adaptability, and personalized therapy. We focus on recent studies that address key challenges in the field, providing rational polymer design, important properties, functionality, and understanding delivery strategies. These developments are expected to advance supramolecular polymers as new platforms of intelligent drug delivery systems, offering innovative solutions for the treatment of complex diseases.
Collapse
Affiliation(s)
- Xinyue Zhang
- School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Jiaxin Zhuo
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, 130 Meilong Road, Shanghai, 200237, P. R. China
| | - Dali Wang
- School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering, Zhangjiang Institute for Advanced Study, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, P. R. China
| |
Collapse
|
27
|
Chen Y, Bhattacharya S, Bergmann L, Correy GJ, Tan S, Hou K, Biel J, Lu L, Bakanas I, Polizzi NF, Fraser JS, DeGrado WF. Emergence of specific binding and catalysis from a designed generalist binding protein. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.30.635804. [PMID: 39975260 PMCID: PMC11838529 DOI: 10.1101/2025.01.30.635804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
The evolution of binding and catalysis played a central role in the emergence of life. While natural proteins have finely tuned affinities for their primary ligands, they also bind weakly and promiscuously to other molecules, which serve as starting points for stepwise, incremental evolution of entirely new specificities. Thus, modern proteins emerged from the joint exploration of sequence and structural space. The ability of natural proteins to bind promiscuously to small molecule fragments has been widely evaluated using methods including crystallographic fragment screening. However, this approach had not been applied to de novo proteins. Here, we apply this method to explore the promiscuity of a de novo small molecule-binding protein ABLE. As in Nature, we found ABLE was capable of forming weak complexes, which were found to be excellent starting points for evolving entirely new functions, including a binder of a turn-on fluorophore and a highly efficient and specific Kemp eliminase enzyme. This work shows how Nature and protein designers can take advantage of promiscuous binding interactions to evolve new proteins with specialized functions.
Collapse
Affiliation(s)
- Yuda Chen
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Sagar Bhattacharya
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Lena Bergmann
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - Galen J. Correy
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - Sophia Tan
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Kaipeng Hou
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Justin Biel
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - Lei Lu
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Ian Bakanas
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| | - Nicholas F. Polizzi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - James S. Fraser
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - William F. DeGrado
- Department of Pharmaceutical Chemistry & Cardiovascular Research Institute, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
28
|
Devinat M, Thevenard-Devy J, Ghilane F, Devy J, Chazee L, Terryn C, Duca L, Devarenne-Charpentier E, El Btaouri H. Xanthohumol Sensitizes Melanoma Cells to Vemurafenib by Lowering Membrane Cholesterol and Increasing Membrane Fluidity. Int J Mol Sci 2025; 26:2290. [PMID: 40076912 PMCID: PMC11901044 DOI: 10.3390/ijms26052290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Chemoresistance remains one of the major obstacles to cancer treatment. The search for specific molecules that could improve cancer treatment has become one of the objectives of biomedical research. Identifying new natural molecules to enhance chemotherapy treatment or improve sensitization to conventional therapies has become a key objective. Here, we evaluated the effect of Xanthohumol (XN) extracted from hop on SKMEL-28 melanoma cells and their sensitization to vemurafenib (VEM) treatment. We measured the XN effect on cell viability and apoptosis. We also assessed the effect of XN on membrane fluidity and membrane cholesterol levels. Finally, we studied the impact of XN on cell sensitization to VEM. Here, we showed that XN reduced SKMEL-28 cell viability through an apoptotic mechanism. Our results demonstrated the potential role of XN in sensitizing cancer cells to VEM with a less toxic effect on non-tumor cells. A study of XN's molecular mechanism showed that XN was able to induce cholesterol depletion and increased fluidity in SKMEL-28 cancer cells. This leads to an increase in VEM incorporation. Here, we describe the importance of the strategy to modulate membrane fluidity by XN in order to significantly improve anticancer therapy.
Collapse
Affiliation(s)
- Marine Devinat
- UMR-CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, Université de Reims Champagne Ardenne, Moulin de la Housse, BP 1039, 51687 Reims, CEDEX, France; (M.D.); (J.T.-D.); (J.D.); (L.C.); (L.D.); (E.D.-C.)
| | - Jessica Thevenard-Devy
- UMR-CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, Université de Reims Champagne Ardenne, Moulin de la Housse, BP 1039, 51687 Reims, CEDEX, France; (M.D.); (J.T.-D.); (J.D.); (L.C.); (L.D.); (E.D.-C.)
| | - Fatiha Ghilane
- Laboratoire de Biologie des Pathologies Humaines, Université Mohammed V de Rabat, 4 Avenue Ibn Battouta, Rabat B.P. 1014 RP, Morocco;
| | - Jerome Devy
- UMR-CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, Université de Reims Champagne Ardenne, Moulin de la Housse, BP 1039, 51687 Reims, CEDEX, France; (M.D.); (J.T.-D.); (J.D.); (L.C.); (L.D.); (E.D.-C.)
| | - Lise Chazee
- UMR-CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, Université de Reims Champagne Ardenne, Moulin de la Housse, BP 1039, 51687 Reims, CEDEX, France; (M.D.); (J.T.-D.); (J.D.); (L.C.); (L.D.); (E.D.-C.)
| | - Christine Terryn
- Plateau Technique en Imagerie Cellulaire et Tissulaire (PICT) Pôle Santé, UFR Pharmacie, Université de Reims Champagne Ardenne, 51 Rue Cognacq Jay, 51096 Reims, France;
| | - Laurent Duca
- UMR-CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, Université de Reims Champagne Ardenne, Moulin de la Housse, BP 1039, 51687 Reims, CEDEX, France; (M.D.); (J.T.-D.); (J.D.); (L.C.); (L.D.); (E.D.-C.)
| | - Emmanuelle Devarenne-Charpentier
- UMR-CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, Université de Reims Champagne Ardenne, Moulin de la Housse, BP 1039, 51687 Reims, CEDEX, France; (M.D.); (J.T.-D.); (J.D.); (L.C.); (L.D.); (E.D.-C.)
| | - Hassan El Btaouri
- UMR-CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), UFR Sciences Exactes et Naturelles, Université de Reims Champagne Ardenne, Moulin de la Housse, BP 1039, 51687 Reims, CEDEX, France; (M.D.); (J.T.-D.); (J.D.); (L.C.); (L.D.); (E.D.-C.)
| |
Collapse
|
29
|
Yin J, Zhang H, Sun X, You N, Mou M, Lu M, Pan Z, Li F, Li H, Zeng S, Zhu F. Decoding Drug Response With Structurized Gridding Map-Based Cell Representation. IEEE J Biomed Health Inform 2025; 29:1702-1713. [PMID: 38090819 DOI: 10.1109/jbhi.2023.3342280] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025]
Abstract
A thorough understanding of cell-line drug response mechanisms is crucial for drug development, repurposing, and resistance reversal. While targeted anticancer therapies have shown promise, not all cancers have well-established biomarkers to stratify drug response. Single-gene associations only explain a small fraction of the observed drug sensitivity, so a more comprehensive method is needed. However, while deep learning models have shown promise in predicting drug response in cell lines, they still face significant challenges when it comes to their application in clinical applications. Therefore, this study proposed a new strategy called DD-Response for cell-line drug response prediction. First, a limitation of narrow modeling horizons was overcome to expand the model training domain by integrating multiple datasets through source-specific label binarization. Second, a modified representation based on a two-dimensional structurized gridding map (SGM) was developed for cell lines & drugs, avoiding feature correlation neglect and potential information loss. Third, a dual-branch, multi-channel convolutional neural network-based model for pairwise response prediction was constructed, enabling accurate outcomes and improved exploration of underlying mechanisms. As a result, the DD-Response demonstrated superior performance, captured cell-line characteristic variations, and provided insights into key factors impacting cell-line drug response. In addition, DD-Response exhibited scalability in predicting clinical patient responses to drug therapy. Overall, because of DD-response's excellent ability to predict drug response and capture key molecules behind them, DD-response is expected to greatly facilitate drug discovery, repurposing, resistance reversal, and therapeutic optimization.
Collapse
|
30
|
Tang L, Peng S, Zhuang X, He Y, Song Y, Nie H, Zheng C, Pan Z, Lam AK, He M, Shi X, Li B, Xu WW. Tumor Metastasis: Mechanistic Insights and Therapeutic Intervention. MEDCOMM – ONCOLOGY 2025; 4. [DOI: 10.1002/mog2.70012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/10/2025] [Indexed: 03/04/2025]
Abstract
ABSTRACTMetastasis remains a leading cause of cancer‐related deaths, defined by a complex, multi‐step process in which tumor cells spread and form secondary growths in distant tissues. Despite substantial progress in understanding metastasis, the molecular mechanisms driving this process and the development of effective therapies remain incompletely understood. Elucidating the molecular pathways governing metastasis is essential for the discovery of innovative therapeutic targets. The rapid advancements in sequencing technologies and the expansion of biological databases have significantly deepened our understanding of the molecular drivers of metastasis and associated drug resistance. This review focuses on the molecular drivers of metastasis, particularly the roles of genetic mutations, epigenetic changes, and post‐translational modifications in metastasis progression. We also examine how the tumor microenvironment influences metastatic behavior and explore emerging therapeutic strategies, including targeted therapies and immunotherapies. Finally, we discuss future research directions, stressing the importance of novel treatment approaches and personalized strategies to overcome metastasis and improve patient outcomes. By integrating contemporary insights into the molecular basis of metastasis and therapeutic innovation, this review provides a comprehensive framework to guide future research and clinical advancements in metastatic cancer.
Collapse
Affiliation(s)
- Lin Tang
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Shao‐Cong Peng
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Xiao‐Wan Zhuang
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Yan He
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Yu‐Xiang Song
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| | - Hao Nie
- Department of Radiation Oncology, The Fifth Affiliated Hospital Guangzhou Medical University Guangzhou China
| | - Can‐Can Zheng
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Zhen‐Yu Pan
- Department of Radiation Oncology, The Affiliated Huizhou Hospital Guangzhou Medical University Huizhou China
| | - Alfred King‐Yin Lam
- Cancer Molecular Pathology and Griffith Medical School Griffith University Gold Coast Queensland Australia
| | - Ming‐Liang He
- Department of Biomedical Sciences City University of Hong Kong Hong Kong China
| | - Xing‐Yuan Shi
- Department of Radiation Oncology, The Fifth Affiliated Hospital Guangzhou Medical University Guangzhou China
| | - Bin Li
- State Key Laboratory of Respiratory Disease, Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes The Fifth Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Wen Wen Xu
- State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, The Affiliated Traditional Chinese Medicine Hospital Guangzhou Medical University Guangzhou China
| |
Collapse
|
31
|
Lü X, Wei X, Wang C, Tang M, Jin Y, Fan S, Yang Z. Identification of the therapeutic potential of novel TIGIT/PVR interaction blockers based advanced computational techniques and experimental validation. Biophys Chem 2025; 318:107383. [PMID: 39729827 DOI: 10.1016/j.bpc.2024.107383] [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: 09/02/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024]
Abstract
The inhibition of the TIGIT/PVR interaction demonstrates considerable anticancer properties by enhancing the cytotoxic activity of natural killer (NK) and CD8+ T cells. However, the development of small molecule inhibitors that target TIGIT is currently limited. In this study, small molecules with the capacity to bind TIGIT and block the TIGIT/PVR interaction were screened through an advanced computational process, subsequently confirmed by blocking assays. Combined machine learning model XGBOOST and centroid-based molecular docking were employed to expeditiously exclude negative molecules, thereby reducing the chemical space. Subsequently, a blockade assay targeting the TIGIT/PVR interaction was conducted on 14 candidate molecules along with positive control, wherein compound MCULE-5547257859 exhibited the most potent inhibitory effect. Molecular dynamics simulations and binding free energy analyses revealed that compound MCULE-5547257859 possesses a thermodynamically stable conformation, indicative of a stronger binding affinity to TIGIT. In conclusion, our investigation has delineated that compound MCULE-5547257859 effectively impedes the TIGIT/PVR interaction, thereby offering a novel therapeutic modality for oncology.
Collapse
Affiliation(s)
- Xudong Lü
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Xiyu Wei
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Chenyu Wang
- School of Pharmacy, North China University of Science and Technology, Tangshan 063210, China
| | - Mengjia Tang
- School of Pharmacy, North China University of Science and Technology, Tangshan 063210, China
| | - Yuanyuan Jin
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Shuai Fan
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| | - Zhaoyong Yang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China; NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
32
|
Zhao Y, Yuan C, Shi Y, Liu X, Luo L, Zhang L, Pešić M, Yao H, Li L. Drug screening approaches for small-molecule compounds in cancer-targeted therapy. J Drug Target 2025; 33:368-383. [PMID: 39575843 DOI: 10.1080/1061186x.2024.2427185] [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: 05/30/2024] [Revised: 09/30/2024] [Accepted: 10/27/2024] [Indexed: 02/08/2025]
Abstract
Small-molecule compounds exhibit distinct pharmacological properties and clinical effectiveness. Over the past decade, advances in covalent drug discovery have led to successful small-molecule drugs, such as EGFR, BTK, and KRAS (G12C) inhibitors, for cancer therapy. Researchers are paying more attention to refining drug screening methods aiming for high throughput, fast speed, high specificity, and accuracy. Therefore, the discovery and development of small-molecule drugs has been facilitated by significantly reducing screening time and financial resources, and increasing promising lead compounds compared with traditional methods. This review aims to introduce classical and emerging methods for screening small-molecule compounds in targeted cancer therapy. It includes classification, principles, advantages, disadvantages, and successful applications, serving as valuable references for subsequent researchers.
Collapse
Affiliation(s)
- Yelin Zhao
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chenyu Yuan
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuchen Shi
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaohong Liu
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Xicheng District, Beijing, China
| | - Liaoxin Luo
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Li Zhang
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Milica Pešić
- Department of Neurobiology, Institute for Biological Research, 'Siniša Stanković'- National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Hongjuan Yao
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liang Li
- State Key Laboratory of Respiratory Health and Multimorbidity, NHC Key Laboratory of Biotechnology for Microbial Drugs, Department of Oncology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
33
|
Guo Z, Xiao Y, Wu W, Zhe M, Yu P, Shakya S, Li Z, Xing F. Metal-organic framework-based smart stimuli-responsive drug delivery systems for cancer therapy: advances, challenges, and future perspectives. J Nanobiotechnology 2025; 23:157. [PMID: 40022098 PMCID: PMC11871784 DOI: 10.1186/s12951-025-03252-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 02/18/2025] [Indexed: 03/03/2025] Open
Abstract
Cancer treatment is currently one of the most critical healthcare issues globally. A well-designed drug delivery system can precisely target tumor tissues, improve efficacy, and reduce damage to normal tissues. Stimuli-responsive drug delivery systems (SRDDSs) have shown promising application prospects. Intelligent nano drug delivery systems responsive to endogenous stimuli such as weak acidity, complex redox characteristics, hypoxia, active energy metabolism, as well as exogenous stimuli like high temperature, light, pressure, and magnetic fields are increasingly being applied in chemotherapy, radiotherapy, photothermal therapy, photodynamic therapy, and various other anticancer approaches. Metal-organic frameworks (MOFs) have become promising candidate materials for constructing SRDDSs due to their large surface area, tunable porosity and structure, ease of synthesis and modification, and good biocompatibility. This paper reviews the application of MOF-based SRDDSs in various modes of cancer therapy. It summarizes the key aspects, including the classification, synthesis, modifications, drug loading modes, stimuli-responsive mechanisms, and their roles in different cancer treatment modalities. Furthermore, we address the current challenges and summarize the potential applications of artificial intelligence in MOF synthesis. Finally, we propose strategies to enhance the efficacy and safety of MOF-based SRDDSs, ultimately aiming at facilitating their clinical translation.
Collapse
Affiliation(s)
- Ziliang Guo
- Division of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yuzhen Xiao
- Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking, Union Medical College, Beijing, 100005, China
| | - Wenting Wu
- Department of Pediatric Surgery, Division of Orthopedic Surgery, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Man Zhe
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Peiyun Yu
- Department of Molecular Brain Physiology and Behavior, LIMES Institute, University of Bonn, Carl-Troll-Str. 31, 53115, Bonn, Germany
| | - Sujan Shakya
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhihui Li
- Division of Thyroid and Parathyroid Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Fei Xing
- Department of Pediatric Surgery, Division of Orthopedic Surgery, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
34
|
Huang J, Teixeira AP, Gao T, Xue S, Xie M, Fussenegger M. Aspirin-responsive gene switch regulating therapeutic protein expression. Nat Commun 2025; 16:2028. [PMID: 40016240 PMCID: PMC11868571 DOI: 10.1038/s41467-025-57275-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 02/18/2025] [Indexed: 03/01/2025] Open
Abstract
Current small-molecule-regulated synthetic gene switches face clinical limitations such as cytotoxicity, long-term side-effects and metabolic disturbances. Here, we describe an advanced synthetic platform inducible by risk-free input medication (ASPIRIN), which is activated by acetylsalicylic acid (ASA/aspirin), a multifunctional drug with pain-relieving, anti-inflammatory, and cardiovascular benefits. To construct ASPIRIN, we repurpose plant salicylic acid receptors NPR1 and NPR4. Through domain truncations and high-throughput mutant library screening, we enhance their ASA sensitivity. Optimized NPR1 fused with a membrane-tethering myristoylation signal (Myr-NPR1) forms a complex with NPR4, which is fused with a DNA binding domain (VanR) and a transactivation domain (VP16). ASA induces dissociation of the Myr-NPR1/NPR4-VanR-VP16 complex, allowing nuclear translocation of NPR4-VanR-VP16 to activate VanR-operator-controlled gene expression. In male diabetic mice implanted with microencapsulated ASPIRIN-engineered cells, ASA regulates insulin expression, restores normoglycemia, alleviates pain and reduces biomarkers of diabetic neuropathy and inflammation. We envision this system will pave the way for aspirin-based combination gene therapies.
Collapse
Affiliation(s)
- Jinbo Huang
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Ana Palma Teixeira
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Ting Gao
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Shuai Xue
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Mingqi Xie
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Medicine and School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland.
- Faculty of Science, University of Basel, Basel, Switzerland.
| |
Collapse
|
35
|
Guérin C, Vinchent A, Fernandes M, Damour I, Laratte A, Tellier R, Estevam GO, Meneboo JP, Villenet C, Descarpentries C, Fraser JS, Figeac M, Cortot AB, Rouleau E, Tulasne D. MET variants with activating N-lobe mutations identified in hereditary papillary renal cell carcinomas still require ligand stimulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.11.03.565283. [PMID: 37965202 PMCID: PMC10635098 DOI: 10.1101/2023.11.03.565283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
In hereditary papillary renal cell carcinoma (HPRCC), the hepatocyte growth factor receptor (MET) receptor tyrosine kinase (RTK) mutations recorded to date are located in the kinase domain and lead to constitutive MET activation. This contrasts with MET mutations identified in non-small cell lung cancer (NSCLC), which lead to exon 14 skipping and deletion of a regulatory domain: in this latter case, the mutated receptor still requires ligand stimulation. Sequencing of MET in samples from 158 HPRCC and 2808 NSCLC patients revealed ten uncharacterized mutations. Four of these, all found in HPRCC and leading to amino acid substitutions in the N-lobe of the MET kinase, proved able to induce cell transformation, which was further enhanced by hepatocyte growth factor (HGF) stimulation: His1086Leu, Ile1102Thr, Leu1130Ser and Cis1125Gly. Similar to the variant resulting in MET exon 14 skipping, the two N-lobe MET variants His1086Leu and Ile1102Thr were found to require stimulation by HGF in order to strongly activate downstream signaling pathways and epithelial cell motility. The Ile1102Thr mutation also displayed transforming potential, promoting tumor growth in a xenograft model. In addition, the N-lobe-mutated MET variants were found to trigger a common HGF-stimulation-dependent transcriptional program, consistent with an observed increase in cell motility and invasion. Altogether, this functional characterization revealed that N-lobe variants still require ligand stimulation, in contrast to other RTK variants. This suggests that HGF expression in the tumor microenvironment is important for tumor growth. The sensitivity of these variants to MET inhibitors opens the way for use of targeted therapies for patients harboring the corresponding mutations.
Collapse
|
36
|
Xiao H, Wang A, Shuai W, Qian Y, Wu C, Wang X, Yang P, Sun Q, Wang G, Ouyang L, Sun Q. A first-in-class selective inhibitor of ERK1/2 and ERK5 overcomes drug resistance with a single-molecule strategy. Signal Transduct Target Ther 2025; 10:70. [PMID: 39979271 PMCID: PMC11842588 DOI: 10.1038/s41392-025-02169-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 02/07/2025] [Accepted: 02/07/2025] [Indexed: 02/22/2025] Open
Abstract
Despite significant advancements in kinase-targeted therapy, the emergence of acquired drug resistance to targets such as KRAS and MEK remains a challenge. Extracellular-regulated kinase 1/2 (ERK1/2), positioned at the terminus of this pathway, is highly conserved and less susceptible to mutations, thereby garnering attention as a crucial therapeutical target. However, attempts to use monotherapies that target ERK1/2 have achieved only limited clinical success, mainly due to the issues of limited efficacy and the emergence of drug resistance. Herein, we present a proof of concept that extracellular-regulated kinase 5 (ERK5) acts as a compensatory pathway after ERK1/2 inhibition in triple-negative breast cancer (TNBC). By utilizing the principle of polypharmacology, we computationally designed SKLB-D18, a first-in-class molecule that selectively targets ERK1/2 and ERK5, with nanomolar potency and high specificity for both targets. SKLB-D18 demonstrated excellent tolerability in mice and demonstrated superior in vivo anti-tumor efficacy, not only exceeding the existing clinical ERK1/2 inhibitor BVD-523, but also the combination regimen of BVD-523 and the ERK5 inhibitor XMD8-92. Mechanistically, we showed that SKLB-D18, as an autophagy agonist, played a role in mammalian target of rapamycin (mTOR)/70 ribosomal protein S6 kinase (p70S6K) and nuclear receptor coactivator 4 (NCOA4)-mediated ferroptosis, which may mitigate multidrug resistance.
Collapse
Affiliation(s)
- Huan Xiao
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Aoxue Wang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Wen Shuai
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Yuping Qian
- Department of Pathology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Chengyong Wu
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Xin Wang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Panpan Yang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Qian Sun
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China
| | - Guan Wang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China.
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China.
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer Center, Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
37
|
Guérin C, Vinchent A, Fernandes M, Damour I, Laratte A, Tellier R, Estevam GO, Meneboo JP, Villenet C, Descarpentries C, Fraser JS, Figeac M, Cortot AB, Rouleau E, Tulasne D. MET variants with activating N-lobe mutations identified in hereditary papillary renal cell carcinomas still require ligand stimulation. Mol Oncol 2025. [PMID: 39980226 DOI: 10.1002/1878-0261.13806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 10/16/2024] [Accepted: 01/15/2025] [Indexed: 02/22/2025] Open
Abstract
In hereditary papillary renal cell carcinoma (HPRCC), the hepatocyte growth factor receptor (MET) receptor tyrosine kinase (RTK) mutations recorded to date are located in the kinase domain and lead to constitutive MET activation. This contrasts with MET mutations identified in non-small-cell lung cancer (NSCLC), which lead to exon 14 skipping and deletion of a regulatory domain: In this latter case, the mutated receptor still requires ligand stimulation. Sequencing of MET in samples from 158 HPRCC and 2808 NSCLC patients revealed 10 uncharacterized mutations. Four of these, all found in HPRCC and leading to amino acid substitutions in the N-lobe of the MET kinase, proved able to induce cell transformation, which was further enhanced by hepatocyte growth factor (HGF) stimulation: His1086Leu, Ile1102Thr, Leu1130Ser, and Cis1125Gly. Similar to the variant resulting in MET exon 14 skipping, the two N-lobe MET variants His1086Leu and Ile1102Thr were found to require stimulation by HGF in order to strongly activate downstream signaling pathways and epithelial cell motility. The Ile1102Thr mutation also displayed transforming potential, promoting tumor growth in a xenograft model. In addition, the N-lobe-mutated MET variants were found to trigger a common HGF-stimulation-dependent transcriptional program, consistent with an observed increase in cell motility and invasion. Altogether, this functional characterization revealed that N-lobe variants still require ligand stimulation, in contrast to other RTK variants. This suggests that HGF expression in the tumor microenvironment is important for tumor growth. The sensitivity of these variants to MET inhibitors opens the way for use of targeted therapies for patients harboring the corresponding mutations.
Collapse
Affiliation(s)
- Célia Guérin
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Audrey Vinchent
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Marie Fernandes
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Isabelle Damour
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Agathe Laratte
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Rémi Tellier
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| | - Gabriella O Estevam
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| | - Jean-Pascal Meneboo
- Univ. Lille, Plateau de génomique fonctionnelle et structurale, CHU Lille, France
| | - Céline Villenet
- Univ. Lille, Plateau de génomique fonctionnelle et structurale, CHU Lille, France
| | - Clotilde Descarpentries
- Department of Biochemistry and Molecular Biology, Hormonology Metabolism Nutrition Oncology, CHU Lille, France
| | - James S Fraser
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, USA
| | - Martin Figeac
- Univ. Lille, Plateau de génomique fonctionnelle et structurale, CHU Lille, France
| | - Alexis B Cortot
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
- Thoracic Oncology Department, Univ. Lille, CHU Lille, France
| | - Etienne Rouleau
- Department of Medical Biology and Pathology, Cancer Genetics Laboratory, Gustave Roussy, Villejuif, France
| | - David Tulasne
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, UMR9020 - UMR1277 - Canther - Cancer Heterogeneity, Plasticity and Resistance to Therapies, Lille, France
| |
Collapse
|
38
|
Zhou Y, Wei Y, Tian X, Wei X. Cancer vaccines: current status and future directions. J Hematol Oncol 2025; 18:18. [PMID: 39962549 PMCID: PMC11834487 DOI: 10.1186/s13045-025-01670-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
Cancer continues to be a major global health burden, with high morbidity and mortality. Building on the success of immune checkpoint inhibitors and adoptive cellular therapy, cancer vaccines have garnered significant interest, but their clinical success remains modest. Benefiting from advancements in technology, many meticulously designed cancer vaccines have shown promise, warranting further investigations to reach their full potential. Cancer vaccines hold unique benefits, particularly for patients resistant to other therapies, and they offer the ability to initiate broad and durable T cell responses. In this review, we highlight the antigen selection for cancer vaccines, introduce the immune responses induced by vaccines, and propose strategies to enhance vaccine immunogenicity. Furthermore, we summarize key features and notable clinical advances of various vaccine platforms. Lastly, we delve into the mechanisms of tumor resistance and explore the potential benefits of combining cancer vaccines with standard treatments and other immunomodulatory approaches to improve vaccine efficacy.
Collapse
Affiliation(s)
- Yingqiong Zhou
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xiaohe Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
39
|
An Q, Huang L, Wang C, Wang D, Tu Y. New strategies to enhance the efficiency and precision of drug discovery. Front Pharmacol 2025; 16:1550158. [PMID: 40008135 PMCID: PMC11850385 DOI: 10.3389/fphar.2025.1550158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Drug discovery plays a crucial role in medicinal chemistry, serving as the cornerstone for developing new treatments to address a wide range of diseases. This review emphasizes the significance of advanced strategies, such as Click Chemistry, Targeted Protein Degradation (TPD), DNA-Encoded Libraries (DELs), and Computer-Aided Drug Design (CADD), in boosting the drug discovery process. Click Chemistry streamlines the synthesis of diverse compound libraries, facilitating efficient hit discovery and lead optimization. TPD harnesses natural degradation pathways to target previously undruggable proteins, while DELs enable high-throughput screening of millions of compounds. CADD employs computational methods to refine candidate selection and reduce resource expenditure. To demonstrate the utility of these methodologies, we highlight exemplary small molecules discovered in the past decade, along with a summary of marketed drugs and investigational new drugs that exemplify their clinical impact. These examples illustrate how these techniques directly contribute to advancing medicinal chemistry from the bench to bedside. Looking ahead, Artificial Intelligence (AI) technologies and interdisciplinary collaboration are poised to address the growing complexity of drug discovery. By fostering a deeper understanding of these transformative strategies, this review aims to inspire innovative research directions and further advance the field of medicinal chemistry.
Collapse
Affiliation(s)
| | | | | | - Dongmei Wang
- Scientific Research and Teaching Department, Public Health Clinical Center of Chengdu, Chengdu, Sichuan, China
| | - Yalan Tu
- Scientific Research and Teaching Department, Public Health Clinical Center of Chengdu, Chengdu, Sichuan, China
| |
Collapse
|
40
|
Han Z, Yan G, Jousma J, Nukala SB, Amiri M, Kiniry S, Tabatabaei N, Kwon Y, Zhang S, Rehman J, Pinho S, Ong SB, Baranov PV, Tahmasebi S, Ong SG. Translational regulation of SND1 governs endothelial homeostasis during stress. J Clin Invest 2025; 135:e168730. [PMID: 39895626 PMCID: PMC11785924 DOI: 10.1172/jci168730] [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: 01/17/2023] [Accepted: 11/22/2024] [Indexed: 02/04/2025] Open
Abstract
Translational control shapes the proteome and is particularly important in regulating gene expression under stress. A key source of endothelial stress is treatment with tyrosine kinase inhibitors (TKIs), which lowers cancer mortality but increases cardiovascular mortality. Using a human induced pluripotent stem cell-derived endothelial cell (hiPSC-EC) model of sunitinib-induced vascular dysfunction combined with ribosome profiling, we assessed the role of translational control in hiPSC-ECs in response to stress. We identified staphylococcal nuclease and tudor domain-containing protein 1 (SND1) as a sunitinib-dependent translationally repressed gene. SND1 translational repression was mediated by the mTORC1/4E-BP1 pathway. SND1 inhibition led to endothelial dysfunction, whereas SND1 OE protected against sunitinib-induced endothelial dysfunction. Mechanistically, SND1 transcriptionally regulated UBE2N, an E2-conjugating enzyme that mediates K63-linked ubiquitination. UBE2N along with the E3 ligases RNF8 and RNF168 regulated the DNA damage repair response pathway to mitigate the deleterious effects of sunitinib. In silico analysis of FDA-approved drugs led to the identification of an ACE inhibitor, ramipril, that protected against sunitinib-induced vascular dysfunction in vitro and in vivo, all while preserving the efficacy of cancer therapy. Our study established a central role for translational control of SND1 in sunitinib-induced endothelial dysfunction that could potentially be therapeutically targeted to reduce sunitinib-induced vascular toxicity.
Collapse
Affiliation(s)
- Zhenbo Han
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Gege Yan
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Jordan Jousma
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Sarath Babu Nukala
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Mehdi Amiri
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Stephen Kiniry
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Negar Tabatabaei
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Youjeong Kwon
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Sen Zhang
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA
| | - Jalees Rehman
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA
- University of Illinois Cancer Center, Chicago, Illinois, USA
| | - Sandra Pinho
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA
- University of Illinois Cancer Center, Chicago, Illinois, USA
| | - Sang-Bing Ong
- Department of Medicine and Therapeutics, Faculty of Medicine, Chinese University of Hong Kong (CUHK), Hong Kong SAR, China
- Centre for Cardiovascular Genomics and Medicine (CCGM), Lui Che Woo Institute of Innovative Medicine, CUHK, Hong Kong SAR, China
- Hong Kong Hub of Pediatric Excellence (HK HOPE), Hong Kong Children’s Hospital (HKCH), Hong Kong SAR, China
- Kunming Institute of Zoology — The Chinese University of Hong Kong (KIZ-CUHK) Joint Laboratory of Bioresources and Molecular Research of Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
| | - Pavel V. Baranov
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Soroush Tahmasebi
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA
- University of Illinois Cancer Center, Chicago, Illinois, USA
| | - Sang-Ging Ong
- Department of Pharmacology & Regenerative Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA
- Division of Cardiology, Department of Medicine, University of Illinois College of Medicine, Chicago, Illinois, USA
| |
Collapse
|
41
|
Wang QH, Cheng S, Han CY, Yang S, Gao SR, Yin WZ, Song WZ. Tailoring cell-inspired biomaterials to fuel cancer therapy. Mater Today Bio 2025; 30:101381. [PMID: 39742146 PMCID: PMC11683242 DOI: 10.1016/j.mtbio.2024.101381] [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: 11/04/2024] [Revised: 12/01/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025] Open
Abstract
Cancer stands as a predominant cause of mortality across the globe. Traditional cancer treatments, including surgery, radiotherapy, and chemotherapy, are effective yet face challenges like normal tissue damage, complications, and drug resistance. Biomaterials, with their advantages of high efficacy, targeting, and spatiotemporal controllability, have been widely used in cancer treatment. However, the biocompatibility limitations of traditional synthetic materials have restricted their clinical translation and application. Natural cell-inspired biomaterials inherently possess the targeting abilities of cells, biocompatibility, and immune evasion capabilities. Therefore, cell-inspired biomaterials can be used alone or in combination with other drugs or treatment strategies for cancer therapy. In this review, we first introduce the timeline of key milestones in cell-inspired biomaterials for cancer therapy. Then, we describe the abnormalities in cancer including biophysics, cellular biology, and molecular biology aspects. Afterwards, we summarize the design strategies of cell-inspired antitumor biomaterials. Subsequently, we elaborate on the application of antitumor biomaterials inspired by various cell types. Finally, we explore the current challenges and prospects of cell-inspired antitumor materials. This review aims to provide new opportunities and references for the development of antitumor cell-inspired biomaterials.
Collapse
Affiliation(s)
- Qi-Hui Wang
- Department of Stomatology, China-Japan Union Hospital, Jilin University, 126#Xiantai Street, Jingkai District, Changchun, 130031, PR China
| | - Shi Cheng
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, PR China
| | - Chun-Yu Han
- Department of Stomatology, China-Japan Union Hospital, Jilin University, 126#Xiantai Street, Jingkai District, Changchun, 130031, PR China
| | - Shuang Yang
- Department of Stomatology, China-Japan Union Hospital, Jilin University, 126#Xiantai Street, Jingkai District, Changchun, 130031, PR China
| | - Sheng-Rui Gao
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Hospital of Jilin University, Changchun, 130061, PR China
| | - Wan-Zhong Yin
- Department of Otorhinolaryngology, Head and Neck Surgery, The First Hospital of Jilin University, Changchun, 130061, PR China
| | - Wen-Zhi Song
- Department of Stomatology, China-Japan Union Hospital, Jilin University, 126#Xiantai Street, Jingkai District, Changchun, 130031, PR China
| |
Collapse
|
42
|
Rudalska R, Harbig J, Forster M, Woelffing P, Esposito A, Kudolo M, Botezatu A, Haller V, Janssen N, Holzmayer S, Nahidino P, Trompak O, Pantsar T, Kronenberger T, Yurttas C, Rist E, Weber ANR, Dahlke MH, Ott G, Koenigsrainer A, Rothbauer U, Maerklin M, Muerdter T, Schwab M, Singer S, Zender L, Laufer S, Dauch D. First-in-class ultralong-target-residence-time p38α inhibitors as a mitosis-targeted therapy for colorectal cancer. NATURE CANCER 2025; 6:259-277. [PMID: 39820127 PMCID: PMC11864979 DOI: 10.1038/s43018-024-00899-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 12/12/2024] [Indexed: 01/19/2025]
Abstract
Colorectal cancer (CRC) constitutes the second leading cause of cancer-related death worldwide and advanced CRCs are resistant to targeted therapies, chemotherapies and immunotherapies. p38α (Mapk14) has been suggested as a therapeutic target in CRC; however, available p38α inhibitors only allow for insufficient target inhibition. Here we describe a unique class of p38α inhibitors with ultralong target residence times (designated ULTR-p38i) that robustly inhibit p38α downstream signaling and induce distinct biological phenotypes. ULTR-p38i monotherapy triggers an uncontrolled mitotic entry by activating Cdc25 and simultaneously blocking Wee1. Consequently, CRC cells undergo mitotic catastrophe, resulting in apoptosis or senescence. ULTR-p38i exhibit high selectivity, good pharmaco-kinetic properties and no measurable toxicity with strong therapeutic effects in patient-derived CRC organoids and syngeneic CRC mouse models. Conceptually, our study suggests ultralong-target-residence-time kinase inhibitors as an alternative to covalent inhibitors, which, because of the lack of cysteine residues, cannot be generated for many kinase cancer targets.
Collapse
Affiliation(s)
- Ramona Rudalska
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Jule Harbig
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Michael Forster
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
| | - Pascal Woelffing
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Aylin Esposito
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Mark Kudolo
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
| | - Adelina Botezatu
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Vanessa Haller
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
| | - Nicole Janssen
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Tübingen, Germany
| | - Samuel Holzmayer
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Philipp Nahidino
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
| | - Omelyan Trompak
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Tatu Pantsar
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Thales Kronenberger
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
| | - Can Yurttas
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Elke Rist
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
| | - Alexander N R Weber
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Marc H Dahlke
- Department of General and Visceral Surgery, Robert Bosch Hospital, Stuttgart, Germany
| | - German Ott
- Department of Clinical Pathology, Robert Bosch Hospital, Stuttgart, Germany
| | - Alfred Koenigsrainer
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of General, Visceral and Transplant Surgery, University Hospital Tübingen, Tübingen, Germany
| | - Ulrich Rothbauer
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
- Department of Pharmaceutical Biotechnology, University of Tübingen, Tübingen, Germany
| | - Melanie Maerklin
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Tübingen, Germany
| | - Thomas Muerdter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Tübingen, Germany
| | - Matthias Schwab
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart and University of Tübingen, Tübingen, Germany
- Departments of Clinical Pharmacology, and of Biochemistry and Pharmacy, University of Tübingen, Tübingen, Germany
- German Cancer Research Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephan Singer
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Institute of Pathology and Neuropathology, University Hospital Tübingen, Tübingen, Germany
| | - Lars Zender
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- German Cancer Research Consortium (DKTK), Partner Site Tübingen, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Tübingen Center for Academic Drug Discovery and Development (TüCAD2), Tübingen, Germany
| | - Stefan Laufer
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany
- Department of Pharmaceutical Chemistry, University of Tübingen, Tübingen, Germany
- Tübingen Center for Academic Drug Discovery and Development (TüCAD2), Tübingen, Germany
| | - Daniel Dauch
- Department of Medical Oncology and Pneumology, University Hospital Tübingen, Tübingen, Germany.
- IFIT Cluster of Excellence EXC 2180 'Image-Guided and Functionally Instructed Tumor Therapies', University of Tübingen, Tübingen, Germany.
- Tübingen Center for Academic Drug Discovery and Development (TüCAD2), Tübingen, Germany.
| |
Collapse
|
43
|
Čerina Pavlinović D, Šuto Pavičić J, Njavro A, Librenjak N, Tomaš I, Šeparović R, Pleština S, Bajić Ž, Dedić Plavetić N, Vrdoljak E. Precision Oncology in Clinical Practice: Two Years of Comprehensive Genomic Profiling in Croatia. J Pers Med 2025; 15:59. [PMID: 39997336 PMCID: PMC11856208 DOI: 10.3390/jpm15020059] [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: 01/07/2025] [Revised: 01/17/2025] [Accepted: 01/30/2025] [Indexed: 02/26/2025] Open
Abstract
Background: The widespread adoption of precision medicine in routine cancer care remains a critical challenge, even as advanced technologies expand and personalized therapies demonstrate remarkable success in certain cancer types. While breakthrough innovations in targeted treatments have revolutionized outcomes for specific cancers, translating these scientific advances into standard clinical practice continues to be an evolving and complex endeavor. Croatia has a nationwide project of precision oncology through the comprehensive genomic profiling (CGP) analysis. Since collecting and analyzing real-world data is crucial for clinical research and defining the value of CGP in precision oncology, we aimed to present the data from everyday clinical practice given the opportunities and challenges we faced. Methods: This was a retrospective observational study conducted at the national level in all patients whose tumor samples were subjected to CGP between 1 January 2020 and 31 December 2021. Results: In total, 481 patients with CGP results were included in this study. Gastrointestinal and reproductive malignancies were the most common, accounting for 29.1% and 28.9% of all tested tumors, respectively. Specifically, colorectal tumors made up 19.1% of cases, while uterine tumors represented 11.2%. At least one clinically relevant genomic alteration was found in 76.7% of patients, with the KRAS mutation (27.2%) being the most common. During the two-year study period, 26,709 individuals lost their lives to cancer in Croatia. Combining this with the CGP selection criteria valid at the time, there was an estimated population of approximately 13,350 potentially eligible patients for the CGP analysis, meaning that only 3.6% of potentially eligible patients were tested. Conclusions: The analysis identified clinically actionable genomic alterations in approximately 80% of the evaluated patients, suggesting they could be candidates for targeted therapeutic interventions. The adoption of CGP remains limited, with estimates indicating that under 5% of metastatic cancer patients received testing in the initial two-year implementation period, despite established national insurance coverage guidelines. This low utilization rate suggests a significant gap in access to genomic testing, leaving many eligible cancer patients without the potential benefits of this diagnostic approach.
Collapse
Affiliation(s)
- Dora Čerina Pavlinović
- Department of Oncology, University Hospital Center Split, 21 000 Split, Croatia; (D.Č.P.); (J.Š.P.)
- School of Medicine, University of Split, 21 000 Split, Croatia
| | - Jelena Šuto Pavičić
- Department of Oncology, University Hospital Center Split, 21 000 Split, Croatia; (D.Č.P.); (J.Š.P.)
| | - Antonela Njavro
- Department of Oncology and Nuclear Medicine, Sestre Milosrdnice University Hospital Center, 10 000 Zagreb, Croatia;
| | - Nikša Librenjak
- Department of Oncology, University Hospital Center Zagreb, 10 000 Zagreb, Croatia; (N.L.); (S.P.)
| | - Ilijan Tomaš
- School of Medicine, Josip Juraj Strossmayer University of Osijek, 31 000 Osijek, Croatia;
| | - Robert Šeparović
- Department of Medical Oncology, University Hospital for Tumor, Sestre Milosrdnice, 10 000 Zagreb, Croatia;
| | - Stjepko Pleština
- Department of Oncology, University Hospital Center Zagreb, 10 000 Zagreb, Croatia; (N.L.); (S.P.)
- School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia
| | - Žarko Bajić
- Research Unit “Dr. Mirko Grmek”, Psychiatric Clinic Sveti Ivan, 10 000 Zagreb, Croatia;
| | - Natalija Dedić Plavetić
- Department of Oncology, University Hospital Center Zagreb, 10 000 Zagreb, Croatia; (N.L.); (S.P.)
- School of Medicine, University of Zagreb, 10 000 Zagreb, Croatia
| | - Eduard Vrdoljak
- Department of Oncology, University Hospital Center Split, 21 000 Split, Croatia; (D.Č.P.); (J.Š.P.)
- School of Medicine, University of Split, 21 000 Split, Croatia
| |
Collapse
|
44
|
Bu C, Jiang L, Cui L, Tang M, Song X, Zhao Y, Liang Z, Ye L, Nian J, Gao S, Tao X, Wang Z, Chen W. LC-MS/MS method for quantification of 23 TKIs in Plasma: Assessing the relationship between anlotinib trough concentration and toxicities. Clin Chim Acta 2025; 566:120028. [PMID: 39547553 DOI: 10.1016/j.cca.2024.120028] [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: 03/07/2024] [Revised: 11/02/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
OBJECTIVES To develop a simple, rapid, and sensitive LC-MS/MS method for quantifying 23 tyrosine kinase inhibitors (TKIs) in plasma samples, and evaluate the relationship between the trough concentration of anlotinib(ANL) and its toxicities. METHODS The method was developed in Agilent 1290-6460 UHPLC-MS/MS system. This study prospectively enrolled 55 cancer patients undergoing ANL treatment. Plasma samples were collected at steady-state trough concentration and subsequently analyzed using the method. Patients were recorded for the occurrence of toxicities. Statistical analysis was performed to assess the association of the toxicities with ANL exposure level and patients' characteristics. RESULTS The LC-MS/MS method was developed and validated for all items required by pharmacopoeia. The results revealed a positive association between the trough concentration of ANL and the incidence of toxicities. The exposure level 17.655 ng/mL (AUC 0.82, p = 0.010) was identified as a predictive threshold value for grade ≥ 3 overall toxicities. In addition, lower platelet count (PLT count < 179 × 109 g/L) was significantly associated with higher occurrence of grade ≥ 3 toxicities (AUC 0.75, p = 0.049). A logistic model incorporating these two factors demonstrated improved diagnostic capacity for predicting ≥ 3 overall toxicities (AUC = 0.90, p = 0.001). CONCLUSIONS This study successfully developed and validated a simple, rapid, and sensitive LC-MS/MS method for quantifying 23 TKIs in plasma samples. Besides, this study found that both Ctrough of ANL and PLT count as independent predictors for ANL-induced ≥ 3 overall toxicities. Moreover, a logistic model including these two factors presents better prediction capacity for ≥ 3 overall toxicities.
Collapse
Affiliation(s)
- Chen Bu
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Liansheng Jiang
- Department of Laboratory Medicine, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Lili Cui
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Mao Tang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Xinhua Song
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Yingkui Zhao
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China
| | - Zhengyan Liang
- College of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Liya Ye
- College of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Jiayao Nian
- College of Traditional Chinese Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, China
| | - Shouhong Gao
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China.
| | - Xia Tao
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China.
| | - Zhipeng Wang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China.
| | - Wansheng Chen
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
45
|
Rechberger JS, Toll SA, Biswas S, You HB, Chow WD, Kendall N, Navalkele P, Khatua S. Advances in the Repurposing and Blood-Brain Barrier Penetrance of Drugs in Pediatric Brain Tumors. Cancers (Basel) 2025; 17:439. [PMID: 39941807 PMCID: PMC11816256 DOI: 10.3390/cancers17030439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/22/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Central nervous system (CNS) tumors are the leading cause of cancer-related mortality in children, with prognosis remaining dismal for some of these malignancies. Though the past two decades have seen advancements in surgery, radiation, and targeted therapy, major unresolved hurdles continue to undermine the therapeutic efficacy. These include challenges in suboptimal drug delivery through the blood-brain barrier (BBB), marked intra-tumoral molecular heterogeneity, and the elusive tumor microenvironment. Drug repurposing or re-tasking FDA-approved drugs with evidence of penetration into the CNS, using newer methods of intracranial drug delivery facilitating optimal drug exposure, has been an area of intense research. This could be a valuable tool, as most of these agents have already gone through the lengthy process of drug development and the evaluation of safety risks and the optimal pharmacokinetic profile. They can now be used and tested in clinics with an accelerated and different approach. Conclusions: The next-generation therapeutic strategy should prioritize repurposing oncologic and non-oncologic drugs that have been used for other indication, and have demonstrated robust preclinical activity against pediatric brain tumors. In combination with novel drug delivery techniques, these drugs could hold significant therapeutic promise in pediatric neurooncology.
Collapse
Affiliation(s)
| | - Stephanie A. Toll
- Children’s Hospital of Michigan, Central Michigan University School of Medicine, Saginaw, MI 48602, USA;
| | - Subhasree Biswas
- Bronglais General Hospital, Caradog Road, Aberystwyth SY23 1ER, Wales, UK;
| | - Hyo Bin You
- Mayo Clinic Alix School of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; (H.B.Y.); (W.D.C.)
| | - William D. Chow
- Mayo Clinic Alix School of Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA; (H.B.Y.); (W.D.C.)
| | - Nicholas Kendall
- School of Medicine, University of South Dakota Sanford, Vermillion, SD 57069, USA;
| | - Pournima Navalkele
- Division of Oncology, Children’s Hospital of Orange County, Orange, CA 92868, USA;
| | - Soumen Khatua
- Department of Pediatric Hematology/Oncology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| |
Collapse
|
46
|
Zhao F, Jiang X, Li Y, Huang T, Xiahou Z, Nie W, Li Q. Characterizing tumor biology and immune microenvironment in high-grade serous ovarian cancer via single-cell RNA sequencing: insights for targeted and personalized immunotherapy strategies. Front Immunol 2025; 15:1500153. [PMID: 39896800 PMCID: PMC11782144 DOI: 10.3389/fimmu.2024.1500153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 12/19/2024] [Indexed: 02/04/2025] Open
Abstract
Background High-grade serous ovarian cancer (HGSOC), the predominant subtype of epithelial ovarian cancer, is frequently diagnosed at an advanced stage due to its nonspecific early symptoms. Despite standard treatments, including cytoreductive surgery and platinum-based chemotherapy, significant improvements in survival have been limited. Understanding the molecular mechanisms, immune landscape, and drug sensitivity of HGSOC is crucial for developing more effective and personalized therapies. This study integrates insights from cancer immunology, molecular profiling, and drug sensitivity analysis to identify novel therapeutic targets and improve treatment outcomes. Utilizing single-cell RNA sequencing (scRNA-seq), the study systematically examines tumor heterogeneity and immune microenvironment, focusing on biomarkers influencing drug response and immune activity, aiming to enhance patient outcomes and quality of life. Methods scRNA-seq data was obtained from the GEO database in this study. Differential gene expression was analyzed using gene ontology and gene set enrichment methods. InferCNV identified malignant epithelial cells, while Monocle, Cytotrace, and Slingshot software inferred subtype differentiation trajectories. The CellChat software package predicted cellular communication between malignant cell subtypes and other cells, while pySCENIC analysis was utilized to identify transcription factor regulatory networks within malignant cell subtypes. Finally, the analysis results were validated through functional experiments, and a prognostic model was developed to assess prognosis, immune infiltration, and drug sensitivity across various risk groups. Results This study investigated the cellular heterogeneity of HGSOC using scRNA-seq, focusing on tumor cell subtypes and their interactions within the tumor microenvironment. We confirmed the key role of the C2 IGF2+ tumor cell subtype in HGSOC, which was significantly associated with poor prognosis and high levels of chromosomal copy number variations. This subtype was located at the terminal differentiation of the tumor, displaying a higher degree of malignancy and close association with stage IIIC tissue types. The C2 subtype was also associated with various metabolic pathways, such as glycolysis and riboflavin metabolism, as well as programmed cell death processes. The study highlighted the complex interactions between the C2 subtype and fibroblasts through the MK signaling pathway, which may be closely related to tumor-associated fibroblasts and tumor progression. Elevated expression of PRRX1 was significantly connected to the C2 subtype and may impact disease progression by modulating gene transcription. A prognostic model based on the C2 subtype demonstrated its association with adverse prognosis outcomes, emphasizing the importance of immune infiltration and drug sensitivity analysis in clinical intervention strategies. Conclusion This study integrates molecular oncology, immunotherapy, and drug sensitivity analysis to reveal the mechanisms driving HGSOC progression and treatment resistance. The C2 IGF2+ tumor subtype, linked to poor prognosis, offers a promising target for future therapies. Emphasizing immune infiltration and drug sensitivity, the research highlights personalized strategies to improve survival and quality of life for HGSOC patients.
Collapse
Affiliation(s)
- Fu Zhao
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaojing Jiang
- Affiliated Hospital of Shandong Academy of Traditional Chinese Medicine, Jinan, China
| | - Yumeng Li
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tianjiao Huang
- The First School of Clinical Medicine, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Zhikai Xiahou
- China Institute of Sport and Health Science, Beijing Sport University, Beijing, China
| | - Wenyang Nie
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qian Li
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
47
|
Belmontes B, Slemmons KK, Su C, Liu S, Policheni AN, Moriguchi J, Tan H, Xie F, Aiello DA, Yang Y, Lazaro R, Aeffner F, Rees MG, Ronan MM, Roth JA, Vestergaard M, Cowland S, Andersson J, Sarvary I, Chen Q, Sharma P, Lopez P, Tamayo N, Pettus LH, Ghimire-Rijal S, Mukund S, Allen JR, DeVoss J, Coxon A, Rodon J, Ghiringhelli F, Penel N, Prenen H, Glad S, Chuang CH, Keyvanjah K, Townsley DM, Butler JR, Bourbeau MP, Caenepeel S, Hughes PE. AMG 193, a Clinical Stage MTA-Cooperative PRMT5 Inhibitor, Drives Antitumor Activity Preclinically and in Patients with MTAP-Deleted Cancers. Cancer Discov 2025; 15:139-161. [PMID: 39282709 PMCID: PMC11726016 DOI: 10.1158/2159-8290.cd-24-0887] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 08/20/2024] [Accepted: 09/10/2024] [Indexed: 01/30/2025]
Abstract
One of the most robust synthetic lethal interactions observed in multiple functional genomic screens has been the dependency on protein arginine methyltransferase 5 (PRMT5) in cancer cells with MTAP deletion. We report the discovery of the clinical stage MTA-cooperative PRMT5 inhibitor AMG 193, which preferentially binds PRMT5 in the presence of MTA and has potent biochemical and cellular activity in MTAP-deleted cells across multiple cancer lineages. In vitro, PRMT5 inhibition induces DNA damage, cell cycle arrest, and aberrant alternative mRNA splicing in MTAP-deleted cells. In human cell line and patient-derived xenograft models, AMG 193 induces robust antitumor activity and is well tolerated with no impact on normal hematopoietic cell lineages. AMG 193 synergizes with chemotherapies or the KRAS G12C inhibitor sotorasib in vitro and combination treatment in vivo substantially inhibits tumor growth. AMG 193 is demonstrating promising clinical activity, including confirmed partial responses in patients with MTAP-deleted solid tumors from an ongoing phase 1/2 study. Significance: AMG 193 preferentially inhibits the growth of MTAP-deleted tumor cells by inhibiting PRMT5 when in complex with MTA, thus sparing MTAP wild-type normal cells. AMG 193 shows promise as a targeted therapy in a clinically defined patient population.
Collapse
Affiliation(s)
| | | | - Chun Su
- Amgen Research, South San Francisco, California
| | - Siyuan Liu
- Amgen Research, Thousand Oaks, California
| | | | | | - Hong Tan
- Amgen Research, Thousand Oaks, California
| | - Fang Xie
- Amgen Research, South San Francisco, California
| | | | | | | | | | - Matthew G. Rees
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
| | | | | | | | | | | | | | - Qing Chen
- Amgen Research, Thousand Oaks, California
| | | | | | | | | | | | | | | | | | | | | | | | | | - Hans Prenen
- Universitair Ziekenhuis Antwerpen, Edegem, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Wu D, Sun Q, Tang H, Xiao H, Luo J, Ouyang L, Sun Q. Acquired resistance to tyrosine kinase targeted therapy: mechanism and tackling strategies. Drug Resist Updat 2025; 78:101176. [PMID: 39642660 DOI: 10.1016/j.drup.2024.101176] [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: 09/17/2024] [Revised: 11/21/2024] [Accepted: 11/23/2024] [Indexed: 12/09/2024]
Abstract
Over the past two decades, tyrosine kinase inhibitors (TKIs) have rapidly emerged as pivotal targeted agents, offering promising therapeutic prospects for patients. However, as the cornerstone of targeted therapies, an increasing number of TKIs have been found to develop acquired resistance during treatment, making the challenge of overcoming this resistance a primary focus of current research. This review comprehensively examines the evolution of TKIs from multiple perspectives, with particular emphasis on the mechanisms underlying acquired resistance, innovative drug design strategies, inherent challenges, and future directions.
Collapse
Affiliation(s)
- Defa Wu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Qian Sun
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China; West China Medical Publishers, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Haolin Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Huan Xiao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Jiaxiang Luo
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China
| | - Liang Ouyang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China.
| | - Qiu Sun
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu 610041, China; Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu 610212, China; West China Medical Publishers, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
49
|
Zhang G, Ma Z, Ma Z, Liu P, Zhang L, Lian Z, Guo C. SUZ12-Increased NRF2 Alleviates Cardiac Ischemia/Reperfusion Injury by Regulating Apoptosis, Inflammation, and Ferroptosis. Cardiovasc Toxicol 2025; 25:97-109. [PMID: 39729180 DOI: 10.1007/s12012-024-09950-6] [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: 05/21/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024]
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a redox-sensitive transcriptional factor that enables cells to resist oxidant responses, ferroptosis and inflammation. Here, we set out to probe the effects of NRF2 on cardiomyocyte injury under acute myocardial infarction (AMI) condition and its potential mechanism. Human cardiomyocytes were exposed to hypoxia/reoxygenation (H/R) to induce cell injury. qRT-PCR and western blot assays were used to detect the levels of mRNAs and proteins. Cardiomyocyte injury was determined by detecting the levels of lactate dehydrogenase and creatine Kinase MB (CK-MB). Cell apoptosis was investigated by flow cytometry and related markers. Levels of IL-6, IL-10, and TNF-α were measured by ELISA. Cell ferroptosis was assessed by detecting the production of reactive oxygen species (ROS), malonaldehyde (MDA), reduced glutathione/oxidized glutathione disulfide (GSH/GSSG) ratio, Fe + content, and related regulators. The interaction between NRF2 and the suppressor of zest 12 (SUZ12) was analyzed by using dual-luciferase reporter and RNA immunoprecipitation assays. AMI rat models were established for in vivo analysis. NRF2 was lowly expressed in AMI patients and H/R-induced cardiomyocytes. Forced expression of NRF2 reduced H/R-induced cardiomyocyte injury, apoptosis, inflammation, and ferroptosis. Moreover, NRF2 overexpression improved cardiac function and injury in vivo. Mechanistically, SUZ12 bound to the promoter of NRF2 and promoted its expression. Further functional analyses showed that SUZ12 overexpression reduced H/R-induced cardiomyocyte injury, apoptosis, inflammation, and ferroptosis, which were reversed by NRF2 silencing. SUZ12-increased NRF2 suppressed H/R-induced cardiomyocyte injury, apoptosis, inflammation, and ferroptosis in vitro and improved cardiac functions in rats with I/R injury, suggesting the potential cardioprotective effect of NRF2 in cardiac injury during AMI.
Collapse
Affiliation(s)
- Guoyong Zhang
- Department of Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 3 Chongwenmennei Street, Dongcheng District, Beijing, 100730, China
| | - Zhimin Ma
- Department of Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 3 Chongwenmennei Street, Dongcheng District, Beijing, 100730, China
| | - Zheng Ma
- Department of Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 3 Chongwenmennei Street, Dongcheng District, Beijing, 100730, China
| | - Peilin Liu
- Department of Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 3 Chongwenmennei Street, Dongcheng District, Beijing, 100730, China
| | - Lin Zhang
- Department of Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 3 Chongwenmennei Street, Dongcheng District, Beijing, 100730, China
| | - Zheng Lian
- Department of Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 3 Chongwenmennei Street, Dongcheng District, Beijing, 100730, China
| | - Caixia Guo
- Department of Cardiovascular Center, Beijing Tongren Hospital, Capital Medical University, No. 3 Chongwenmennei Street, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
50
|
Wang J, Wang L, Qiang W, Ge W. The role of DDR1 in cancer and the progress of its selective inhibitors. Bioorg Chem 2025; 154:108018. [PMID: 39642752 DOI: 10.1016/j.bioorg.2024.108018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 11/08/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
Discoidin domain receptor 1 (DDR1) is a member of the receptor tyrosine kinase superfamily, which mainly activates downstream signaling pathways through binding to collagen. The abnormal expression of DDR1 is closely related to the occurrence and development of various tumors, and it is one of the potential targets for molecular targeted therapy. At present, specific antibodies and selective small molecule inhibitors against DDR1 have been approved for Phase I clinical trials. In this review, we summarized the effects of DDR1 on tumor cell proliferation, survival, migration, invasion, energy metabolism and tumor microenvironment, and combed the research progress of selective DDR1 small molecule inhibitors in the field of anti-tumor. It is hoped that more DDR1 inhibitors with excellent performance will be developed to provide more treatment options for tumor patients.
Collapse
Affiliation(s)
- Jianjun Wang
- Department of pharmacy, Nanjing Drum Tower Hospital, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province 210008, China.
| | - Lele Wang
- Department of pharmacy, Nanjing Drum Tower Hospital, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu Province 210008, China.
| | - Weijie Qiang
- Department of pharmacy, Nanjing Drum Tower Hospital, Nanjing, Jiangsu Province 210008, China.
| | - Weihong Ge
- Department of pharmacy, Nanjing Drum Tower Hospital, Nanjing, Jiangsu Province 210008, China.
| |
Collapse
|