101
|
Alkhammash A. Pharmacology of epitranscriptomic modifications: Decoding the therapeutic potential of RNA modifications in drug resistance. Eur J Pharmacol 2025; 994:177397. [PMID: 39978710 DOI: 10.1016/j.ejphar.2025.177397] [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: 02/12/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
RNA modifications, collectively known as epitranscriptomic modifications, have emerged as critical regulators of gene expression, cellular adaptation, and therapeutic resistance. This review explores the pharmacological potential of targeting RNA modifications, including N6-methyladenosine (m6A) and 5-methylcytosine (m5C), as strategies to overcome drug resistance in cancer. We examine key regulatory enzymes, writers, erasers, and readers-and their roles in modulating RNA stability, translation, and splicing. Advances in combination therapies, integrating RNA modification modulators with conventional chemotherapies and immune checkpoint inhibitors, have shown promising outcomes in reversing multidrug resistance (MDR). Emerging RNA-targeting technologies, such as CRISPR/Cas13 systems and advanced RNA sequencing platforms, further enable precision manipulation of RNA molecules, opening new therapeutic frontiers. However, several challenges persist, including issues related to pharmacokinetics, acquired resistance, and the complexity of epitranscriptomic networks. This review underscores the need for innovative delivery systems, such as lipid nanoparticles and tissue-specific targeting strategies, and highlights the dynamic nature of RNA modifications in response to environmental and therapeutic stress. Ongoing research into non-coding RNA modifications and the interplay between epitranscriptomics and epigenetics offers exciting possibilities for developing novel RNA-targeting therapies. The continued evolution of RNA-based technologies will be crucial in advancing precision medicine, addressing drug resistance, and improving clinical outcomes across multiple diseases.
Collapse
Affiliation(s)
- Abdullah Alkhammash
- Department of Pharmacology, College of Pharmacy, Shaqra University, Shaqra, 11961, Saudi Arabia.
| |
Collapse
|
102
|
Iskierka-Jażdżewska E, Puła B, Jamroziak K, Robak T. Chronic lymphocytic leukemia: what clinical progress have we seen in the last five years? Expert Opin Investig Drugs 2025:1-19. [PMID: 40314223 DOI: 10.1080/13543784.2025.2500288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 04/15/2025] [Indexed: 05/03/2025]
Abstract
INTRODUCTION Chronic lymphocytic leukemia (CLL) is the most common form of leukemia in adults. Although treatment has shifted from immunochemotherapy to novel targeted drugs over the last 10 years, novel therapies remain under investigation, particularly in relapsed and refractory patients. AREAS COVERED This review describes the use of approved targeted drugs and novel therapies in treatment-naïve and relapsed or refractory CLL. Particular attention is paid to the management of double-refractory patients, and the discovery of novel drugs in the last five years. EXPERT OPINION Targeted drugs are effective and well-tolerated in the treatment of CLL. In the last five years, several novel agents have been investigated in preclinical studies and clinical trials, including combinations of approved drugs, novel BTK and BCL2 inhibitors, BTK degraders, bispecific antibodies and CAR-T cells. It is anticipated that some should be approved in the near future.
Collapse
Affiliation(s)
- Elżbieta Iskierka-Jażdżewska
- Department of Hematology, Medical University of Lodz, Lodz, Poland
- Department of General Hematology, Copernicus Memorial Hospital, Lodz, Poland
| | - Bartosz Puła
- Department of Hematology, Medical University of Lodz, Lodz, Poland
- Department of General Hematology, Copernicus Memorial Hospital, Lodz, Poland
| | - Krzysztof Jamroziak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Tadeusz Robak
- Department of Hematology, Medical University of Lodz, Lodz, Poland
- Department of General Hematology, Copernicus Memorial Hospital, Lodz, Poland
| |
Collapse
|
103
|
Cidre-Aranaz F, Magrin C, Zimmermann M, Li J, Baffa A, Ciccaldo M, Hartmann W, Dirksen U, Sola M, Paganetti P, Grünewald TGP, Papin S. High Tau expression correlates with reduced invasion and prolonged survival in Ewing sarcoma. Cell Death Discov 2025; 11:216. [PMID: 40319030 PMCID: PMC12049433 DOI: 10.1038/s41420-025-02497-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/11/2025] [Accepted: 04/16/2025] [Indexed: 05/07/2025] Open
Abstract
The microtubule-associated protein Tau (encoded by the MAPT gene) is linked to a family of neurodegenerative disorders defined as tauopathies, which are characterized by its brain accumulation in neurofibrillary tangles and neuropil threads. Newly described Tau functions comprise DNA protection, chromatin remodeling, p53 regulation and cell fate modulation, suggesting a role of Tau in oncogenesis. Bioinformatic-supported characterization of Tau in cancer reveals robust expression in bone cancer cells, in particular Ewing sarcoma (EwS) cell lines. EwS is an aggressive cancer caused by a fusion of members of the FET and ETS gene families, primarily EWSR1::FLI1. Here we found that MAPT is a EWSR1::ETS target gene and that higher Tau expression in EwS cells inhibited their migratory and invasive behavior, consistent with a more immobile and proliferative phenotype observed in EwS. Indeed, we report that high Tau expression is associated with improved overall survival of EwS patients. We also show that the sessile but proliferative phenotype of EWSR1::ETS-high cells may result from a modulatory role of Tau on focal adhesion to extracellular matrix proteins. Our data highlight the utility of determining Tau expression as a prognostic factor in EwS as well as the opportunity to target Tau expression as an innovative EwS therapy.
Collapse
Affiliation(s)
- Florencia Cidre-Aranaz
- Hopp-Children's Cancer Center (KiTZ), Heidelberg, Germany
- Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Claudia Magrin
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Malenka Zimmermann
- Hopp-Children's Cancer Center (KiTZ), Heidelberg, Germany
- Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
- Faculty of Medicine, Heidelberg University, Heidelberg, Germany
| | - Jing Li
- Hopp-Children's Cancer Center (KiTZ), Heidelberg, Germany
- Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany
| | - Annalisa Baffa
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Matteo Ciccaldo
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Wolfgang Hartmann
- Gerhard-Domagk-Institute of Pathology, Münster University Hospital, Münster, Germany
| | - Uta Dirksen
- Pediatrics III, University Hospital Essen, West German Cancer Center, German Cancer Consortium (DKTK) site Essen, National Center for Tumor Diseases (NCT) West, Essen, Germany
| | - Martina Sola
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Paolo Paganetti
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland.
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland.
| | - Thomas G P Grünewald
- Hopp-Children's Cancer Center (KiTZ), Heidelberg, Germany.
- Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany.
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Stéphanie Papin
- Laboratory for Aging Disorders, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| |
Collapse
|
104
|
Huang C, Li J, Wu R, Li Y, Zhang C. Targeting pyroptosis for cancer immunotherapy: mechanistic insights and clinical perspectives. Mol Cancer 2025; 24:131. [PMID: 40319304 PMCID: PMC12049004 DOI: 10.1186/s12943-025-02344-4] [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: 03/21/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025] Open
Abstract
Pyroptosis is a distinct form of programmed cell death characterized by the rupture of the cell membrane and robust inflammatory responses. Increasing evidence suggests that pyroptosis significantly affects the tumor microenvironment and antitumor immunity by releasing damage-associated molecular patterns (DAMPs) and pro-inflammatory mediators, thereby establishing it as a pivotal target in cancer immunotherapy. This review thoroughly explores the molecular mechanisms underlying pyroptosis, with a particular focus on inflammasome activation and the gasdermin family of proteins (GSDMs). It examines the role of pyroptotic cell death in reshaping the tumor immune microenvironment (TIME) involving both tumor and immune cells, and discusses recent advancements in targeting pyroptotic pathways through therapeutic strategies such as small molecule modulators, engineered nanocarriers, and combinatory treatments with immune checkpoint inhibitors. We also review recent advances and future directions in targeting pyroptosis to enhance tumor immunotherapy with immune checkpoint inhibitors, adoptive cell therapy, and tumor vaccines. This study suggested that targeting pyroptosis offers a promising avenue to amplify antitumor immune responses and surmount resistance to existing immunotherapies, potentially leading to more efficacious cancer treatments.
Collapse
Affiliation(s)
- Chen Huang
- Department of Biotherapy, Cancer Center, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jiayi Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Ruiyan Wu
- West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yangqian Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Chenliang Zhang
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
105
|
Ito Y, Shimono J, Kawamoto K, Hatanaka KC, Kogure Y, Tabata M, Saito Y, Mizuno K, Horie S, Mizukami Y, Koya J, Murakami K, Teshima T, Hatanaka Y, Chiba K, Okada A, Shiraishi Y, Miyoshi H, Matsuno Y, Ohshima K, Kataoka K, Nakagawa M. TP53 and CDKN2A alterations define a poor prognostic subgroup in patients with nodal T follicular helper cell lymphoma. Leukemia 2025:10.1038/s41375-025-02631-5. [PMID: 40316697 DOI: 10.1038/s41375-025-02631-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 05/04/2025]
Abstract
Nodal T follicular helper cell lymphoma (nTFHL) exhibits unique immunophenotypes and somatic alterations, while the prognostic value of these alterations remains unclear. By analyzing 173 nTFHL cases, we identified 36 driver genes, including 4 novel ones (TET3, HLA-C, NRAS, and KLF2). Then, we classified nTFHL cases into four molecular subgroups by major driver alterations. TR-I (+) and TR-I (-) were characterized by TET2 and/or RHOA mutations with and without IDH2 mutations; AC53 by TP53 and/or CDKN2A alterations and aneuploidy; and NSD with no subgroup-defining alterations (namely without any of the above alterations). AC53 exhibited the worst survival, while NSD, particularly those lacking driver alterations, showed the best prognosis. nTFHL had a better prognosis than peripheral T-cell lymphoma, not otherwise specified, when TP53 and/or CDKN2A alterations were absent. Multivariable analyses showed that AC53, the presence of driver alterations, and international prognostic index high-risk were independently associated with worse survival. Finally, we developed a simple prognostic index (mTFHL-PI), which classified patients into three risk categories with a median OS of 181, 67, and 20 months, respectively. Our study identifies novel prognostic factors, namely TP53 and/or CDKN2A alterations and the presence of driver alterations, demonstrating the clinical relevance of molecular classification in nTFHL.
Collapse
Affiliation(s)
- Yuta Ito
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Joji Shimono
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Keisuke Kawamoto
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
- Institute of Pathology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Kanako C Hatanaka
- Center for Development of Advanced Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Yasunori Kogure
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Mariko Tabata
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuki Saito
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Kota Mizuno
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Sara Horie
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Yosuke Mizukami
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
- Department of Gastroenterology, Keio University School of Medicine, Tokyo, Japan
| | - Junji Koya
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Koichi Murakami
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Takanori Teshima
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan
| | - Yutaka Hatanaka
- Research Division of Genome Companion Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Kenichi Chiba
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Ai Okada
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Yuichi Shiraishi
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Hiroaki Miyoshi
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Yoshihiro Matsuno
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Koichi Ohshima
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan.
| | - Keisuke Kataoka
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan.
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan.
| | - Masao Nakagawa
- Department of Hematology, Hokkaido University Faculty of Medicine, Sapporo, Japan.
| |
Collapse
|
106
|
Fu S, Zhang M, Calzadilla N, Zhao B, Zhang X, Yang B, McKenzie VAA, Zheng A, Ni Q, Zhang F. Smart polymer prodrugs via responsive prodrug-initiated ring-opening polymerization of lactide for improved drug delivery. NANOSCALE 2025; 17:10595-10599. [PMID: 40214986 DOI: 10.1039/d5nr00780a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Smart polymer prodrugs, created via responsive prodrug-initiated controlled polymerization of lactide, demonstrated extremely high drug loading, tuneable stimuli-triggered drug release, and significant tumor growth inhibition and improved survival with minimal toxicity. This adaptable strategy can precisely tailor drugs' physicochemical properties for optimal therapeutic efficacy, demonstrating great promise for cancer treatment.
Collapse
Affiliation(s)
- Shiwei Fu
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Miao Zhang
- Nanomedicine Translational Research Program, NUS Centre for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Nicholas Calzadilla
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Bowen Zhao
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Xiao Zhang
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Bowei Yang
- Nanomedicine Translational Research Program, NUS Centre for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Victoria A A McKenzie
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Ajay Zheng
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
| | - Qianqian Ni
- Nanomedicine Translational Research Program, NUS Centre for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| | - Fuwu Zhang
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, Florida, 33146, USA.
- The Dr John T. Macdonald Foundation Biomedical Nanotechnology Institute, University of Miami, Miami, FL 33136, USA
- Sylvester Comprehensive Cancer Centre, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| |
Collapse
|
107
|
Guo Y, Gong Y, Wu M, Ji M, Xie F, Chen H, Niu H, Tang C. CircRNAs in the tumor microenvironment: new frontiers in cancer progression and therapy. Crit Rev Oncol Hematol 2025; 212:104754. [PMID: 40320223 DOI: 10.1016/j.critrevonc.2025.104754] [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: 04/18/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025] Open
Abstract
The tumor microenvironment (TME), a dynamic ecosystem which including immune cells, cancer-associated fibroblasts (CAFs), endothelial cells, pericytes and acellular components, is orchestrating cancer progression through crosstalk between malignant cells and stromal components and increasingly recognized as a therapeutic frontier. Within this intricate network, circular RNAs (circRNAs) have emerged as pivotal regulators due to their unique covalently closed structures, which confer exceptional stability and multifunctional capabilities. This regulation is mediated through multiple mechanisms, such as acting as microRNA (miRNA) sponges, interacting with proteins, and, in certain instances, encoding functional peptides. The interaction between circRNAs and the TME not only affects cancer growth and metastasis but also influences immune evasion and therapeutic resistance. Elucidating the mechanisms by which circRNAs orchestrate these interactions is essential for identifying novel diagnostic biomarkers and developing effective therapeutic strategies. Such insights are expected to bridge gaps in current cancer biology, offering promising avenues for precision oncology and ultimately improving clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Yipei Guo
- School of Elderly Care Services and Management, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yuanxun Gong
- Guangxi Key Laboratory for Preclinical and Translational Research on Bone and Joint Degenerative Diseases, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China
| | - Man Wu
- School of Clinical Medicine, Wannan Medical College, Wuhu 241002, China
| | - Mengjia Ji
- School of Public Health, Wannan Medical College, Wuhu 241002, China
| | - Fei Xie
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao 266013, China.
| | - Hao Chen
- Department of Pathology, Wannan Medical College, Wuhu 241002, China; Postdoctoral Research Station of Clinical Medicine, Jinan University, Guangzhou 510632, China.
| | - Haitao Niu
- Department of Urology, Affiliated Hospital of Qingdao University, Qingdao 266013, China.
| | - Chao Tang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
108
|
Zhao W, Zhang Z, Xie M, Ding F, Zheng X, Sun S, Du J. Exploring tumor-associated macrophages in glioblastoma: from diversity to therapy. NPJ Precis Oncol 2025; 9:126. [PMID: 40316746 PMCID: PMC12048723 DOI: 10.1038/s41698-025-00920-x] [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: 11/21/2024] [Accepted: 04/22/2025] [Indexed: 05/04/2025] Open
Abstract
Glioblastoma is the most aggressive and lethal cancer of the central nervous system, presenting substantial treatment challenges. The current standard treatment, which includes surgical resection followed by temozolomide and radiation, offers limited success. While immunotherapies, such as immune checkpoint inhibitors, have proven effective in other cancers, they have not demonstrated significant efficacy in GBM. Emerging research highlights the pivotal role of tumor-associated macrophages (TAMs) in supporting tumor growth, fostering treatment resistance, and shaping an immunosuppressive microenvironment. Preclinical studies show promising results for therapies targeting TAMs, suggesting potential in overcoming these barriers. TAMs consist of brain-resident microglia and bone marrow-derived macrophages, both exhibiting diverse phenotypes and functions within the tumor microenvironment. This review delves into the origin, heterogeneity, and functional roles of TAMs in GBM, underscoring their dual roles in tumor promotion and suppression. It also summarizes recent progress in TAM-targeted therapies, which may, in combination with other treatments like immunotherapy, pave the way for more effective and personalized strategies against this aggressive malignancy.
Collapse
Affiliation(s)
- Wenwen Zhao
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhi Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Mingyuan Xie
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Feng Ding
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiangrong Zheng
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shicheng Sun
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jianyang Du
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
109
|
Shan J, Chen Z, Chen M, Wu Z, Zhu H, Jin X, Wang Y, Wu Y, Ding Z, Xiang Z, Wang L, Zhao Y, Lin Z, Wang L. SENP3 induced HADHA deSUMOylation enhances intrahepatic cholangiocarcinoma chemotherapy sensitivity via fatty acid oxidation. Cancer Lett 2025; 625:217770. [PMID: 40320039 DOI: 10.1016/j.canlet.2025.217770] [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: 02/10/2025] [Revised: 04/15/2025] [Accepted: 05/01/2025] [Indexed: 05/09/2025]
Abstract
Chemoresistance contributes to poor outcomes in patients with intrahepatic cholangiocarcinoma (ICC). This study aimed to explore the mechanisms underlying chemotherapy resistance and to develop strategies that can sensitize the chemotherapy. Patient derived organoids (PDOs) drug screening and Lipidomics profiling were performed to investigate the chemoresistance mechanism. Through multi-strategy analysis, we found that SENP3 enhanced chemotherapy sensitivity in a SUMO system dependent manner. Mechanistically, chemotherapy resistance increased METTL3 expression, which regulated SENP3 mRNA stability through YTHDF2-dependent m6A methylation modifications. SENP3 interacted with HADHA and catalyzed its deSUMOylation at two lysine residues. Specifically, SUMOylation and ubiquitination exhibited crosstalk at the same modification sites on HADHA, influencing its protein stability and, consequently, regulating fatty acid oxidation (FAO) levels. The physical interaction of SENP3, HADHA, and USP10 provides a novel molecular mechanism for the abnormal activation of FAO pathway. The lipid metabolism-targeting drug could be a promising therapeutic strategy for sensitizing ICC to chemotherapy.
Collapse
Affiliation(s)
- Jijun Shan
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Zhiwen Chen
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Mo Chen
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Zong Wu
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Hongxu Zhu
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Xin Jin
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Yixiu Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Yibin Wu
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Zhiwen Ding
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Zhen Xiang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China
| | - Longrong Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China.
| | - Yiming Zhao
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China.
| | - Zhenhai Lin
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China.
| | - Lu Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, PR China.
| |
Collapse
|
110
|
Dan L, Kang-Zheng L. Optimizing viral transduction in immune cell therapy manufacturing: key process design considerations. J Transl Med 2025; 23:501. [PMID: 40316943 PMCID: PMC12046913 DOI: 10.1186/s12967-025-06524-0] [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: 12/31/2024] [Accepted: 04/21/2025] [Indexed: 05/04/2025] Open
Abstract
Immune cell therapies have revolutionized the treatment of cancers, autoimmune disorders, and infectious diseases. A critical step in their manufacturing is viral transduction, which enables the delivery of therapeutic genes into immune cells. However, the complexity of this process presents significant challenges for optimization and scalability. This review provides a comprehensive analysis of viral transduction process in immune cell therapy manufacturing, highlighting key design considerations to support the development of safe, effective, and scalable production methods. Additionally, it examines current technological challenges in immune cell transduction and explores future innovations poised to advance the field.
Collapse
Affiliation(s)
- Liu Dan
- Bioprocessing Technology Institute BTI, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Singapore, 138668, Singapore.
| | - Lee Kang-Zheng
- Bioprocessing Technology Institute BTI, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Singapore, 138668, Singapore
| |
Collapse
|
111
|
Han L, Wang K, Jiang Z, Guo X, Yu J. Recent development in bispecific antibody immunotherapy for hematological malignancies. Crit Rev Oncol Hematol 2025; 212:104752. [PMID: 40320222 DOI: 10.1016/j.critrevonc.2025.104752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/24/2025] [Accepted: 04/26/2025] [Indexed: 05/08/2025] Open
Abstract
While monoclonal antibody (mAb)-based therapies have revolutionized cancer treatment, challenges such as resistance mechanisms and tumor progression via alternative pathways underscore the need for novel therapeutic strategies. Bispecific antibodies (BsAbs), which target two distinct antigens simultaneously, represent a promising next-generation solution, improving therapeutic precision, efficacy, and safety. BsAbs also redirect cytotoxic effector cells to tumor sites, providing additional therapeutic mechanisms. Recent advancements in BsAb design, such as enhancements in pharmacokinetics and modular multi-specific formats, are expanding their use in hematological malignancies. Combining BsAbs with immune checkpoint inhibitors and other therapies may overcome resistance and improve clinical outcomes. Leading BsAbs, including mosunetuzumab, glofitamab, and blinatumomab, have demonstrated promising efficacy in clinical trials for leukemia and lymphoma subtypes. Despite remaining challenges, particularly in acute myeloid leukemia (AML), ongoing research into new targets and combination therapies is expected to enhance the efficacy of BsAbs in relapsed or refractory (R/R) disease. This review explores the structural and functional innovations of BsAbs, the challenges in current therapies, and their transformative potential in hematological malignancy immunotherapy.
Collapse
Affiliation(s)
- Lijie Han
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Ke Wang
- Department of Hematology/Oncology, Puyang Oilfield General Hospital Affiliated with Xinxiang Medical College, Puyang, China
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xuejun Guo
- Department of Hematology/Oncology, Puyang Oilfield General Hospital Affiliated with Xinxiang Medical College, Puyang, China.
| | - Jifeng Yu
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China.
| |
Collapse
|
112
|
Lee HB, Park SG, Kim HJ, Jeon JP, Oh S, Lee S, Park SH, Kang S. CD7-Targeted Cytotoxic Potency of Diphtheria Toxin- and Ricin-Based Immunotoxins in Targeted Therapy for T-Cell Acute Lymphoblastic Leukemia. Mol Pharm 2025. [PMID: 40317108 DOI: 10.1021/acs.molpharmaceut.5c00146] [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: 05/07/2025]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive and heterogeneous hematologic malignancy, underscoring the urgent need for innovative treatments such as immunotoxins, which combine specific antigen targeting with potent cytotoxic activity. In this study, we developed two distinct immunotoxins (aCD7Nb/DTA and RTA/aCD7Nb) by combining the CD7-binding nanobody (aCD7Nb) with recombinant diphtheria toxin (DTA) and ricin (RTA), utilizing SpyCatcher/SpyTag (SC/ST) post translational protein ligation system for targeted therapy in T-cell acute lymphoblastic leukemia (T-ALL). Both aCD7Nb/DTA and RTA/aCD7Nb specifically bind to three CD7-expressing T-ALL cell lines, CCRF-CEM, Jurkat, and MOLT-4 cells, based on CD7 expression levels, but not to the CD7-negative Raji B-ALL cells. Both aCD7Nb/DTA and RTA/aCD7Nb demonstrated high cytotoxic against T-ALL cells, with IC50 values inversely correlated to CD7 expression. Notably, RTA/aCD7Nb exhibited approximately 2-fold higher anticancer efficacy compared to aCD7Nb/DTA in both CCRF-CEM and Jurkat cells. In an orthotopic model of CCRF-CEM T-ALL-engrafted NSG mice, systemic administration of RTA/aCD7Nb effectively inhibited T-ALL progression and extended survival, without any adverse effects. These findings underscore the potential of combining a CD7-binding ligand with an appropriate active toxin moiety to significantly enhance the efficacy of immunotoxins against T-ALL.
Collapse
Affiliation(s)
- Hyun Bin Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Seong Guk Park
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Hyo Jeong Kim
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Jun Pyo Jeon
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - SuHyeon Oh
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - SangJoon Lee
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Sung Ho Park
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Sebyung Kang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| |
Collapse
|
113
|
Pan R, Koo C, Su W, You Q, Guo H, Liu B. Circular RNAs modulate cell death in cardiovascular diseases. Cell Death Discov 2025; 11:214. [PMID: 40316538 PMCID: PMC12048724 DOI: 10.1038/s41420-025-02504-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025] Open
Abstract
Cardiovascular diseases (CVDs) remain a global health challenge, with programmed cell death (PCD) mechanisms like apoptosis and necroptosis playing key roles in the progression. Circular RNAs (circRNAs) have recently been recognized as crucial regulators of gene expression, especially in modulating PCD. In current researches, circRNA regulation of apoptosis is the most studied area, followed by autophagy and ferroptosis. Notably, the regulatory role of circRNAs in pyroptosis and necroptosis has also begun to attract attention. From a mechanistic perspective, circRNAs influence cellular processes through several modes of action, including miRNA sponging, protein interactions, and polypeptide translation. Manipulating circRNAs and their downstream targets through inhibition or overexpression offers versatile therapeutic options for CVD treatment. Continued investigation into circRNA-mediated mechanisms may enhance our understanding of CVD pathophysiology and underscore their potential as novel and promising therapeutic targets.
Collapse
Affiliation(s)
- Runfang Pan
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Chinying Koo
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Wenyuan Su
- Sport Medicine & Rehabilitation Center, Shanghai University of Sport, Shanghai, 200438, China
| | - Qianhui You
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Haidong Guo
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Baonian Liu
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
114
|
Mavuluri J, Dhungana Y, Jones LL, Bhatara S, Shi H, Yang X, Lim SE, Reyes N, Chi H, Yu J, Geiger TL. GPR65 Inactivation in Tumor Cells Drives Antigen-Independent CAR T-cell Resistance via Macrophage Remodeling. Cancer Discov 2025; 15:1018-1036. [PMID: 39998425 PMCID: PMC12046320 DOI: 10.1158/2159-8290.cd-24-0841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/28/2024] [Accepted: 02/05/2025] [Indexed: 02/26/2025]
Abstract
SIGNIFICANCE The study identifies GPR65 as an important determinant of B-cell acute lymphoblastic leukemia response to CAR T-cell therapy. Notably, GPR65 absence signals CAR T resistance. By emphasizing the therapeutic potential of targeting VEGFA or host macrophages, our study identifies routes to optimize CAR T-cell therapy outcomes in hematologic malignancies via tumor microenvironment manipulation.
Collapse
Affiliation(s)
- Jayadev Mavuluri
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Yogesh Dhungana
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Lindsay L. Jones
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Sheetal Bhatara
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Hao Shi
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Xu Yang
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Song-Eun Lim
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee
- College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Noemi Reyes
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee
- College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Hongbo Chi
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Jiyang Yu
- Department of Computational Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - Terrence L. Geiger
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee
- Graduate School of Biomedical Sciences, St. Jude Children’s Research Hospital, Memphis, Tennessee
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| |
Collapse
|
115
|
Lyu Z, Niu S, Fang Y, Chen Y, Li YR, Yang L. Addressing graft-versus-host disease in allogeneic cell-based immunotherapy for cancer. Exp Hematol Oncol 2025; 14:66. [PMID: 40317083 PMCID: PMC12046680 DOI: 10.1186/s40164-025-00654-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/13/2025] [Indexed: 05/04/2025] Open
Abstract
Allogeneic cell-based immunotherapies, particularly CAR-T cell therapy, represent a significant advancement in cancer treatment, offering scalable and consistent alternatives to autologous therapies. However, their widespread use is limited by the risk of graft-versus-host disease (GvHD). This review provides a comprehensive overview of GvHD in the context of allogeneic cell-based cancer immunotherapy and evaluates current strategies to mitigate its effects. Key strategies include genetic engineering approaches such as T cell receptor (TCR) knockout (KO) and T cell receptor alpha constant (TRAC) CAR knock-in. Alternative immune cell types like natural killer (NK) cells and natural killer T (NKT) cells offer potential solutions due to their lower alloreactivity. Additionally, stem cell technology, utilizing induced pluripotent stem cells (iPSCs), enables standardized and scalable production of engineered CAR-T cells. Clinical trials evaluating these strategies, such as UCART19 and CTX110, demonstrate promising results in preventing GvHD while maintaining anti-tumor efficacy. The review also addresses manufacturing considerations for allogeneic cell products and the challenges in translating preclinical findings into clinical success. By addressing these challenges, allogeneic cell-based immunotherapy continues to advance, paving the way for more accessible, scalable, and effective cancer treatments.
Collapse
Affiliation(s)
- Zibai Lyu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
| | - Siyue Niu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
| | - Ying Fang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
| | - Yuning Chen
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
| | - Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, 90095, USA.
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA.
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, CA, 90095, USA.
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA, 90095, USA.
- Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, CA, 90095, USA.
- Goodman-Luskin Microbiome Center, University of California, Los Angeles, CA, 90095, USA.
| |
Collapse
|
116
|
Lin GB, Chen WT, Kuo YY, Liu HH, Chen YM, Leu SJ, Chao CY. Thermal cycling‑hyperthermia sensitizes non‑small cell lung cancer A549 cells to EGFR tyrosine kinase inhibitor erlotinib. Oncol Rep 2025; 53:58. [PMID: 40183398 PMCID: PMC11976370 DOI: 10.3892/or.2025.8891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Molecular targeted therapy has emerged as a mainstream treatment for non‑small cell lung cancer (NSCLC), the most common type of lung cancer and the leading cause of cancer‑related death in both men and women. Erlotinib (Erl), a targeted therapy inhibiting EGFR pathways, has shown notable response rate in NSCLC cells. However, limited efficacy of the treatment has been reported due to resistance among a proportion of patients with NSCLC. Therefore, sensitizers are required to potentiate the efficacy of Erl in NSCLC treatment. The present study proposed a novel thermal therapy, thermal cycling‑hyperthermia (TC‑HT), as a supplement to amplify the effects of Erl. It was demonstrated that TC‑HT reduced the half‑maximal inhibitory concentration of Erl to 0.5 µM and TC‑HT sensitized A549 NSCLC cells to Erl via the downstream EGFR signaling cascades. Furthermore, the combination treatment of Erl and TC‑HT induced G2/M cell cycle arrest and inhibition of cell proliferation and migration. In addition, by slightly raising the temperature of TC‑HT, TC‑HT treatment alone produced antineoplastic effects without damaging the normal IMR‑90 lung cells. The method presented in this study may be applicable to other combination therapies and could potentially act as a starter for anticancer treatments, with fewer side effects.
Collapse
Affiliation(s)
- Guan-Bo Lin
- Department of Physics, Laboratory for Medical Physics and Biomedical Engineering, National Taiwan University, Taipei 106319, Taiwan, R.O.C
- Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 100233, Taiwan, R.O.C
| | - Wei-Ting Chen
- Department of Physics, Laboratory for Medical Physics and Biomedical Engineering, National Taiwan University, Taipei 106319, Taiwan, R.O.C
- Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 100233, Taiwan, R.O.C
| | - Yu-Yi Kuo
- Department of Physics, Laboratory for Medical Physics and Biomedical Engineering, National Taiwan University, Taipei 106319, Taiwan, R.O.C
- Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 100233, Taiwan, R.O.C
| | - Hsu-Hsiang Liu
- Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 100233, Taiwan, R.O.C
- Graduate Institute of Applied Physics, Biophysics Division, National Taiwan University, Taipei 106319, Taiwan, R.O.C
| | - You-Ming Chen
- Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 100233, Taiwan, R.O.C
- Graduate Institute of Applied Physics, Biophysics Division, National Taiwan University, Taipei 106319, Taiwan, R.O.C
| | - Shr-Jeng Leu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, R.O.C
| | - Chih-Yu Chao
- Department of Physics, Laboratory for Medical Physics and Biomedical Engineering, National Taiwan University, Taipei 106319, Taiwan, R.O.C
- Molecular Imaging Center, National Taiwan University College of Medicine, Taipei 100233, Taiwan, R.O.C
- Graduate Institute of Applied Physics, Biophysics Division, National Taiwan University, Taipei 106319, Taiwan, R.O.C
| |
Collapse
|
117
|
Lee A, Lam CW. Application of Metabolic Biomarkers in Breast Cancer: A Literature Review. Ann Lab Med 2025; 45:229-246. [PMID: 40091629 PMCID: PMC11996688 DOI: 10.3343/alm.2024.0482] [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: 09/08/2024] [Revised: 11/23/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
Breast cancer is the most common cancer and the second leading cause of cancer death in women worldwide. Novel biomarkers for early diagnosis, treatment, and prognosis in breast cancer are needed and extensively studied. Metabolites, which are small molecules produced during metabolic processes, provide links between genetics, environment, and phenotype, making them useful biomarkers for diagnosis, prognosis, and disease classification. With recent advancements in metabolomics techniques, metabolomics research has expanded, which has led to significant progress in biomarker research. In breast cancer, alterations in metabolic pathways result in distinct metabolomic profiles that can be harnessed for biomarker discovery. Studies using mass spectrometry and nuclear magnetic resonance spectroscopy have helped identify significant changes in metabolites, such as amino acids, lipids, and organic acids, in the tissues, blood, and urine of patients with breast cancer, highlighting their potential as biomarkers. Integrative analysis of these metabolite biomarkers with existing clinical parameters is expected to improve the accuracy of breast cancer diagnosis and to be helpful in predicting prognosis and treatment responses. However, to apply these findings in clinical practice, larger cohorts for validation and standardized analytical methods for QC are necessary. In this review, we provide information on the current state of metabolite biomarker research in breast cancer, highlighting key findings and their clinical implications.
Collapse
Affiliation(s)
- Anbok Lee
- Department of Surgery, Chung-Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, Gyeonggi-do, Korea
| | - Ching-Wan Lam
- Department of Pathology, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
118
|
Alhajahjeh A, Stahl M, Kim TK, Kewan T, Stempel JM, Zeidan AM, Bewersdorf JP. Contemporary understanding of myeloid-derived suppressor cells in the acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) tumor microenvironment. Expert Rev Anticancer Ther 2025; 25:435-456. [PMID: 40122075 DOI: 10.1080/14737140.2025.2483855] [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: 01/03/2025] [Revised: 03/01/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION Myeloid-derived suppressor cells (MDSCs) are a key immunosuppressive component in the tumor microenvironment, contributing to immune evasion and disease progression in acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). AREAS COVERED We searched PubMed for literature that evaluated the effect of MDSCs in myeloid diseases. MDSCs impact outcomes by facilitating leukemic stem cell survival, impairing immune checkpoint efficacy, and modulating the bone marrow niche. While these immunosuppressive properties can mitigate graft-versus-host disease post-transplantation, sustained MDSC-mediated immunosuppression can also increase the risk of leukemia relapse.We review MDSC development and function, including metabolic reprogramming, epigenetic modifications, and cytokine-mediated pathways. Therapeutic strategies targeting MDSCs, such as depletion, functional reprogramming, and inhibition of key metabolic and immune pathways, show promising data in preclinical models. However, clinical translation remains hindered by challenges in MDSC quantification and standardization of functional assays. This review underscores the potential of combining MDSC-targeted therapies with conventional and novel treatments to improve patient outcomes in AML and MDS. EXPERT OPINION Future studies should focus on standardizing MDSC assessment, elucidate their dynamic roles in therapy, and optimize combination approaches for clinical application.
Collapse
Affiliation(s)
- Abdulrahman Alhajahjeh
- School of Medicine, The University of Jordan, Amman, Jordan
- King Hussein Cancer Center (KHCC), Internal Medicine Department, Amman, Jordan
| | - Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Tae K Kim
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Tariq Kewan
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Jessica M Stempel
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Jan Philipp Bewersdorf
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
119
|
Chen Z, Xu L, Yuan Y, Zhang S, Xue R. Metabolic crosstalk between platelets and cancer: Mechanisms, functions, and therapeutic potential. Semin Cancer Biol 2025; 110:65-82. [PMID: 39954752 DOI: 10.1016/j.semcancer.2025.02.001] [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: 12/06/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Platelets, traditionally regarded as passive mediators of hemostasis, are now recognized as pivotal regulators in the tumor microenvironment, establishing metabolic feedback loops with tumor and immune cells. Tumor-derived signals trigger platelet activation, which induces rapid metabolic reprogramming, particularly glycolysis, to support activation-dependent functions such as granule secretion, morphological changes, and aggregation. Beyond self-regulation, platelets influence the metabolic processes of adjacent cells. Through direct mitochondrial transfer, platelets reprogram tumor and immune cells, promoting oxidative phosphorylation. Additionally, platelet-derived cytokines, granules, and extracellular vesicles drive metabolic alterations in immune cells, fostering suppressive phenotypes that facilitate tumor progression. This review examines three critical aspects: (1) the distinctive metabolic features of platelets, particularly under tumor-induced activation; (2) the metabolic crosstalk between activated platelets and other cellular components; and (3) the therapeutic potential of targeting platelet metabolism to disrupt tumor-promoting networks. By elucidating platelet metabolism, this review highlights its essential role in tumor biology and its therapeutic implications.
Collapse
Affiliation(s)
- Zhixue Chen
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Lin Xu
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yejv Yuan
- The First Affiliated Hospital of Anhui University of Science and Technology, Huainan 232001, China
| | - Si Zhang
- NHC Key Laboratory of Glycoconjugate Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Ruyi Xue
- Department of Gastroenterology and Hepatology, Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
120
|
Yan L, Su P, Sun X. Role of multi‑omics in advancing the understanding and treatment of prostate cancer (Review). Mol Med Rep 2025; 31:130. [PMID: 40116118 PMCID: PMC11938414 DOI: 10.3892/mmr.2025.13495] [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: 09/30/2024] [Accepted: 01/27/2025] [Indexed: 03/23/2025] Open
Abstract
The application of multi‑omics methodologies, encompassing genomics, transcriptomics, proteomics, metabolomics and integrative genomics, has markedly enhanced the understanding of prostate cancer (PCa). These methods have facilitated the identification of molecular pathways and biomarkers crucial for the early detection, prognostic evaluation and personalized treatment of PCa. Studies using multi‑omics technologies have elucidated how alterations in gene expression and protein interactions contribute to PCa progression and treatment resistance. Furthermore, the integration of multi‑omics data has been used in the identification of novel therapeutic targets and the development of innovative treatment modalities, such as precision medicine. The evolving landscape of multi‑omics research holds promise for not only deepening the understanding of PCa biology but also for fostering the development of more effective and tailored therapeutic interventions, ultimately improving patient outcomes. The present review aims to synthesize current findings from multi‑omics studies associated with PCa and to assess their implications for the improvement of patient management and therapeutic outcomes. The insights provided may guide future research directions and clinical practices in the fight against PCa.
Collapse
Affiliation(s)
- Li Yan
- Department of Urology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| | - Pengxiao Su
- Department of Urology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| | - Xiaoke Sun
- Department of Urology, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, P.R. China
| |
Collapse
|
121
|
Smallbone P, Mehta RS, Alousi A. Steroid Refractory Acute GVHD: The Hope for a Better Tomorrow. Am J Hematol 2025; 100 Suppl 3:14-29. [PMID: 40123554 DOI: 10.1002/ajh.27592] [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: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 03/25/2025]
Abstract
Steroid-refractory acute graft-versus-host disease (SR-AGVHD) presents a significant barrier to successful outcomes following allogeneic hematopoietic cell transplantation (HCT), despite advancements in GVHD prophylaxis and management. While ruxolitinib therapy has shown improved response rates, survival benefits remain elusive. This review explores the definitions and proposed distinct pathophysiology and treatment landscape of SR-AGVHD. Emerging therapies offer potential, yet further research is critical to better define steroid-refractory populations, improve treatment precision with biomarkers, and overcome resistance, particularly in ruxolitinib-refractory cases.
Collapse
Affiliation(s)
- Portia Smallbone
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rohtesh S Mehta
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Amin Alousi
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
122
|
Cao J, Zheng Y, Li R, Feng D, Ni X, Gao H, Wang M, Cao Y, Zhai W, Zhang R, Yang D, He Y, Feng S, Han M, Zhang H, Pang A, Jiang E. CSF3R Mutations in Hematological Disorders Undergoing Allogeneic Hematopoietic Stem Cell Transplantation. Am J Hematol 2025; 100:925-927. [PMID: 39950584 DOI: 10.1002/ajh.27643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 01/27/2025] [Accepted: 02/03/2025] [Indexed: 04/04/2025]
Affiliation(s)
- Jiaxin Cao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yawei Zheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Ruixin Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Dan Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Xiaofei Ni
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Hongye Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Mingyang Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yigen Cao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Weihua Zhai
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Rongli Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Donglin Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Yi He
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Sizhou Feng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Mingzhe Han
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Haixiao Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Aiming Pang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- Tianjin Institutes of Health Science, Tianjin, China
| |
Collapse
|
123
|
Zhang L, Li MJ, Li XP, Yang B, Xiao T, Wang P, Zhang WD. Respiratory microbiota diversity as a predictive biomarker for the efficacy of PD‑1 blockades in patients with advanced non‑small cell lung cancer: A retrospective exploratory study. Oncol Lett 2025; 29:251. [PMID: 40201032 PMCID: PMC11977453 DOI: 10.3892/ol.2025.14997] [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: 09/07/2024] [Accepted: 03/04/2025] [Indexed: 04/10/2025] Open
Abstract
Despite advancements in immunotherapy, particularly regarding programmed cell death protein 1 (PD-1)/programmed death-ligand 1 blockades, the clinical outcomes in non-small cell lung cancer (NSCLC) remain variable with limited predictive biomarkers currently available. The present study investigated respiratory microbiota diversity as a potential biomarker to predict the efficacy of PD-1 blockades in patients with advanced NSCLC. A retrospective analysis was conducted on 60 patients treated with PD-1 blockades from May 2019 to May 2023. Clinical data were collected and respiratory microbiota from deep induced sputum specimens were analyzed using 16S rRNA gene sequencing. An index of respiratory microbiota α diversity was applied and exploratory analysis was performed accordingly. The objective response rate (ORR) and disease control rate among the 60 patients receiving PD-1 blockades was 23.3% (95% CI, 13.4-36.0%) and 58.3% (95% CI, 44.9-70.9%), respectively. Analysis of prognostic data of patients with advanced NSCLC receiving PD-1 blockades monotherapy demonstrated a median progression-free survival of 3.4 months (95% CI, 2.54-4.26) and a median overall survival (OS) of 12.3 months (95% CI, 6.29-18.31). Patients were stratified into high and low α diversity groups based on the Shannon diversity index of respiratory microbiota. The ORR was increased in the high diversity group (26.7%) compared with that of the low diversity group (20.0%), although the difference was not statistically significant (P=0.542). Notably, the high diversity group demonstrated a longer median PFS (3.9 vs. 2.8 months; P=0.017) and median OS (16.8 vs. 6.8 months; P=0.016) compared with that of the low diversity group. These findings suggested that PD-1 blockades demonstrate promising therapeutic activity for patients with previously treated advanced NSCLC in clinical practice. Respiratory microbiota α diversity might serve as a potential biomarker to predict the efficacy of PD-1 blockades monotherapy in patients with advanced NSCLC in the future. Therefore, further prospective studies are warranted to validate these findings and to explore the underlying mechanisms by which respiratory microbiota might modulate the immune response to cancer therapy.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin 300190, P.R. China
| | - Ming-Jiang Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin 300190, P.R. China
| | - Xiao-Ping Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin 300190, P.R. China
| | - Bo Yang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin 300190, P.R. China
| | - Ting Xiao
- Department of Pharmacy, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300071, P.R. China
| | - Ping Wang
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300202, P.R. China
| | - Wei-Dong Zhang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin 300190, P.R. China
| |
Collapse
|
124
|
Khan MA, Palmer J. SOHO State of the Art Updates and Next Questions | Updates on Myelofibrosis With Cytopenia. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:293-303. [PMID: 39516086 DOI: 10.1016/j.clml.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 11/16/2024]
Abstract
Myelofibrosis (MF) is a rare hematologic malignancy that is characterized by dysregulation of the JAK-STAT pathway resulting in fibrosis of the bone marrow, splenomegaly, and abnormalities in peripheral blood counts including anemia, leukocytosis, and thrombocytopenia. This disease has 2 phenotypic extremes - myeloproliferative and cytopenic. Cytopenic myelofibrosis presents with pronounced cytopenia and a different landscape of genetic mutations which results in worse clinical outcomes and a poor prognosis. Patients with cytopenic MF are at high risk of developing various complications like bleeding, infections, and transfusion dependency. Historically, the only Federal Drug Administration (FDA) approved therapy was ruxolitinib, a JAK1/2 inhibitor, which improved constitutional symptoms and splenomegaly, however, exacerbated anemia and thrombocytopenia.1,2 There were very few options for patients with anemia and thrombocytopenia, and supportive treatments for these problems lack efficacy. Fortunately, there are newer treatment options which may allow for treatment of the symptoms and splenomegaly in the setting of cytopenias and even improve cytopenias. This up-to-date review not only highlights the prevalent options in therapeutic marketplace, but also sheds light on the significant unmet need of addressing anemia and thrombocytopenia in cytopenic MF.
Collapse
Affiliation(s)
| | - Jeanne Palmer
- Department of Medicine, Mayo Clinic Arizona, Phoenix, AZ, USA.
| |
Collapse
|
125
|
Suzuki K, Matsuyama H, Matsubara N, Kazama H, Ueno F, Uemura H. Current Evidence on Cabazitaxel for Prostate Cancer Therapy: A Narrative Review. Int J Urol 2025; 32:475-487. [PMID: 39996439 PMCID: PMC12022742 DOI: 10.1111/iju.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/22/2025] [Accepted: 02/11/2025] [Indexed: 02/26/2025]
Abstract
The incidence of prostate cancer (PC) has recently increased in Japan. Androgen deprivation therapy (ADT) has been a key treatment in patients with castration-sensitive PC (CSPC); however, resistance typically emerges through multiple mechanisms, leading to metastatic castration-resistant PC (mCRPC). Taxane-based therapy (i.e., docetaxel, cabazitaxel) has been standard care in patients with mCRPC. New evidence supporting the addition of androgen receptor signaling inhibitors (ARSIs, e.g., enzalutamide, abiraterone) to docetaxel and ADT for patients with metastatic CSPC (mCSPC) raises questions about the role of taxane-based therapies and their optimal sequencing, as well as how to identify patients who may benefit from taxane-based therapy. Here we review the evidence on taxane-based therapy, including cabazitaxel, in the treatment of PC, with a focus on clinical and real-world evidence from Japan. Cabazitaxel has proven effective for patients with mCRPC who have a history of ARSI and docetaxel use, and it is preferable to a second alternative ARSI, as indicated in the CARD study. The safety profile of cabazitaxel (particularly, the incidence of neutropenia) can be managed through prophylactic use of granulocyte colony-stimulating factor, as well as a lower dosage and possibly variation of the dosage interval. However, a certain dose intensity is required because neutropenia has been identified as a potential prognostic indicator for treatment effectiveness. In the ARSI era for mCSPC, evidence on mCRPC treatment sequencing is limited. A better understanding of PC biology and the collection of real-world data is essential for effective treatment and improved safety-benefit outcomes.
Collapse
Affiliation(s)
- Kazuhiro Suzuki
- Department of Urology, Graduate School of MedicineGunma UniversityMaebashiGunmaJapan
| | | | - Nobuaki Matsubara
- Department of Medical OncologyNational Cancer Center Hospital EastChibaJapan
| | | | - Fumiko Ueno
- Specialty Care, Oncology Medical, Sanofi K.K.TokyoJapan
| | - Hirotsugu Uemura
- Department of Urology Kindai University Faculty of MedicineOsakasayamaJapan
| |
Collapse
|
126
|
Yu J, Ren L, Wu T, Hua L, Wang D, Wang Y, Xie Q, Deng J, Gong Y. Establishment and transcriptomic characteristics of radio-resistant meningioma cell lines. J Neurooncol 2025; 173:105-119. [PMID: 40019713 DOI: 10.1007/s11060-025-04966-6] [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: 01/16/2025] [Accepted: 02/05/2025] [Indexed: 03/01/2025]
Abstract
PURPOSE Radio-resistance poses a significant challenge in meningioma treatment. This study aimed to establish radio-resistant meningioma cell lines and uncover molecular mechanisms driving radio-resistance to identify potential biomarkers and therapeutic targets. METHODS Radio-resistant meningioma cell lines (IOMM-Lee-RR, CH157-RR) were developed using a progressive radiation dose (cumulative 90 Gy). Cell morphology, radiosensitivity, apoptosis, viability, migration, invasion, cell cycle, and DNA damage repair were analyzed via clonogenic assays, flow cytometry, and Western blotting. Transcriptome sequencing was performed to identify differentially expressed genes (DEGs), followed by KEGG and GO enrichment analyses. Protein-protein interaction (PPI) analysis was conducted to identify hub genes. TK1 expression was further validated in a cohort of 350 meningiomas and the GSE189672 dataset. RESULTS Radio-resistant meningioma cell lines exhibited enhanced survival, reduced apoptosis, increased cell viability, and superior migratory and invasive abilities compared to parental cells. Under radiation, these cells showed G0/G1 phase accumulation and reduced G2/M phase arrest, along with enhanced DNA repair capacity, as evidenced by lower γ-H2AX expression and fewer DNA damage foci. Transcriptome analysis revealed significant enrichment in metabolic pathways, DNA repair, and cell cycle regulation. Among 34 hub genes identified, TK1 emerged as a key gene, being highly expressed in recurrent and high-grade meningiomas and positively correlated with Ki67. Analysis of the GSE189672 dataset confirmed TK1 as a poor prognostic factor associated with tumor recurrence. CONCLUSION Radio-resistant meningioma cells exhibit enhanced DNA repair, migration, invasion, and altered cell cycle dynamics. TK1 was identified as a promising biomarker and therapeutic target for overcoming radio-resistance in meningiomas.
Collapse
Affiliation(s)
- Jinxiu Yu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, No. 12 Middle Wulumuqi Road, Jingan District, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Fudan University, Shanghai, China
| | - Leihao Ren
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, No. 12 Middle Wulumuqi Road, Jingan District, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Fudan University, Shanghai, China
| | - Tianqi Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, No. 12 Middle Wulumuqi Road, Jingan District, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Fudan University, Shanghai, China
| | - Lingyang Hua
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, No. 12 Middle Wulumuqi Road, Jingan District, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Fudan University, Shanghai, China
| | - Daijun Wang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, No. 12 Middle Wulumuqi Road, Jingan District, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Fudan University, Shanghai, China
| | - Yang Wang
- Department of Radiotherapy, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qing Xie
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, No. 12 Middle Wulumuqi Road, Jingan District, Shanghai, 200040, China
- Institute of Neurosurgery, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Fudan University, Shanghai, China
| | - Jiaojiao Deng
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, No. 12 Middle Wulumuqi Road, Jingan District, Shanghai, 200040, China.
- Institute of Neurosurgery, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Fudan University, Shanghai, China.
| | - Ye Gong
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, No. 12 Middle Wulumuqi Road, Jingan District, Shanghai, 200040, China.
- Institute of Neurosurgery, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Fudan University, Shanghai, China.
- Department of Critical Care Medicine, Shanghai Medical College, Huashan Hospital, Fudan University, Shanghai, China.
- Department of Neurosurgery, Department of Critical Care Medicine, Shanghai Medical College, Huashan Hospital, Fudan University, No. 12 Middle Wulumuqi Road, Jingan District, Shanghai, 200040, China.
| |
Collapse
|
127
|
Chen SH, Yu BY, Kuo WY, Lin YB, Su SY, Chuang WH, Lu IH, Lin CY. Unveiling the immune microenvironment of complex tissues and tumors in transcriptomics through a deconvolution approach. BMC Cancer 2025; 25:733. [PMID: 40307726 PMCID: PMC12044707 DOI: 10.1186/s12885-025-14089-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Accurately resolving the composition of tumor-infiltrating leukocytes is pivotal for advancing cancer immunotherapy strategies. Despite the success of some clinical trials, applying these strategies remains limited due to the challenges in deciphering the immune microenvironment. In this study, we developed a streamlined, two-step workflow to address the complexity of bioinformatics processes involved in analyzing immune cell composition from transcriptomics data. Our dockerized toolkit, DOCexpress_fastqc, integrates the hisat2-stringtie pipeline with customized scripts within Galaxy/Docker environments, facilitating RNA sequencing (RNA-seq) gene expression profiling. The output from DOCexpress_fastqc is seamlessly formatted with mySORT, a web application that employs a deconvolution algorithm to determine the immune content across 21 cell subclasses. We validated mySORT using synthetic pseudo-bulk data derived from single-cell RNA sequencing (scRNA-seq) datasets. Our predictions exhibit strong concordance with the ground-truth immune cell composition, achieving Pearson's correlation coefficients of 0.871 in melanoma patients and 0.775 in head and neck cancer patients. Additionally, mySORT outperforms existing methods like CIBERSORT in accuracy and provides a wide range of data visualization features, such as hierarchical clustering and cell complexity plots. The toolkit and web application are freely available for the research community, providing enhanced resolution for conventional bulk RNA sequencing data and facilitating the analysis of immune microenvironment responses in immunotherapy. The mySORT demo website and Docker image are free at https://mysort.iis.sinica.edu.tw and https://hub.docker.com/r/lsbnb/mysort_2022 .
Collapse
Affiliation(s)
- Shu-Hwa Chen
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, 250 Wu-Xing Street, Taipei, Taiwan
| | - Bo-Yi Yu
- Research Center for Advanced Science and Technology, the University of Tokyo, 4-6-1 Komaba, Meguro-Ku, Tokyo, 153-8904, Japan
| | - Wen-Yu Kuo
- Institute of Information Science, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan
| | - Ya-Bo Lin
- Institute of Information Science, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan
| | - Sheng-Yao Su
- Department of Smart Computing and Applied Mathematics, Tunghai University, Taichung City, 407224, Taiwan
| | - Wei-Hsuan Chuang
- Institute of Information Science, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan
| | - I-Hsuan Lu
- Institute of Information Science, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan
| | - Chung-Yen Lin
- Institute of Information Science, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 115, Taiwan.
- Institute of Fisheries Science, National Taiwan University, Taipei, Taiwan.
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
128
|
Zou Y, Li S, Li Y, Zhang D, Zheng M, Shi B. Glioblastoma Cell Derived Exosomes as a Potent Vaccine Platform Targeting Primary Brain Cancers and Brain Metastases. ACS NANO 2025. [PMID: 40312770 DOI: 10.1021/acsnano.4c14573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Glioblastoma multiforme (GBM) is the most prevalent brain tumor that remains incurable up to now. The rapid advancement of immunotherapy makes vaccines a promising therapeutic approach for GBM. However, current vaccine platforms, such as peptides, dendritic cells, mRNA, and viral vectors, are subject to limitations such as inadequate antigen loading, insufficient immune system activation, ineffective vector delivery, complicated fabrication process, and complex formulation. Here, we developed a GBM tumor cell derived homologous exosomal nanovaccine that does not need to carry any additional tumor antigens and leads to the activation of antigen-presenting cells (APCs) in lymph nodes, increasing the proportion of immune cells (matured dendritic cells, cytotoxic T cells, and memory T cells) and in turn promoting the expression of cytokines (TNF-α, IL-6, and IFN-γ), which effectively stimulates innate immunity to trigger durable protective immunity against tumor cell insult. Our nanovaccine platform possesses efficient dual-targeting capability to lymph nodes and the brain. More importantly, the developed exosomal nanovaccines protected 100% of treated mice by inducing sustained and strong immunity against GL261-luc GBM tumor cells, resulting in 100% mouse survival (8/8) up to 5 months. Our nanovaccines also induced antitumor immune responses in the immunosuppressed CT2A-luc GBM mouse model with greatly improved survival compared to control mice. Exosomal nanovaccines also demonstrated effectiveness in preventing brain metastasis in the B16F10-luc melanoma malignant brain metastasis mouse model, and the mice showed notably improved survival rates. Our simple and potent exosomes offer a versatile platform for clinical translation as individualized vaccine therapy.
Collapse
Affiliation(s)
- Yan Zou
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Shanshan Li
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yundong Li
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Dongya Zhang
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Meng Zheng
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Bingyang Shi
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| |
Collapse
|
129
|
Cao Z, Wang Z, Yang L, Li T, Tao X, Niu X. Reshaping the immune microenvironment and reversing immunosenescence by natural products: Prospects for immunotherapy in gastric cancer. Semin Cancer Biol 2025; 110:1-16. [PMID: 39923925 DOI: 10.1016/j.semcancer.2025.02.002] [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: 12/10/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025]
Abstract
Gastric cancer (GC) represents a global health-care challenge. Recent progress in immunotherapy has elicited attracted considerable attention as a viable treatment option through modulating the host immune system and unleashing pre-existing immunity, which has profoundly revolutionized oncology, especially GC. Nonetheless, low clinical response and intrinsic and acquired resistance remain persistently challenging. The microenvironment of GC comprising multifarious stromal cell types has remarkable immunosuppressive elements that may impact the efficacy of immunotherapy. Immunosenescence is increasingly regarded as a factor that contributes to cancer development, remodels the tumor microenvironment and affects the efficacy of immunotherapy. Natural products are at the forefront of traditional medicine. Senotherapeutics is a class of drugs and natural products capable of delaying, preventing, or reversing the senescence process (i.e., senolytics) or suppressing senescence-associated secretory phenotype (i.e., senomorphics). Emerging evidence supports that natural products can improve the efficacy of existing immunotherapy and expand their indications in GC mainly based upon remodeling the immunosuppressive microenvironment and reversing immunosenescence. The review provides an integrated review of previously reported and ongoing clinical trials with immunotherapeutic regimens in GC and discusses current challenges. Next, we focus on natural compounds that exert anti-GC functions and possess immunomodulatory properties. More attention is paid to the potential of these natural compounds in modulating the immune microenvironment and immunosenescence. Lastly, we discuss the nanomedicine that can overcome the deficiencies of natural products. Altogether, our review suggests the enormous potential of natural compounds in GC immunotherapy, and provides an important direction for future research.
Collapse
Affiliation(s)
- Zhipeng Cao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, China
| | - Zhilin Wang
- Department of Pain Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Li Yang
- Department of Anesthesiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, Liaoning 110032, China
| | - Tian Li
- Tianjin Key Laboratory of Acute Abdomen Disease-Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin 300100, China.
| | - Xueshu Tao
- Department of Pain Medicine, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | - Xing Niu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
130
|
Tandon R, Kumar S, Handa M, Srivastava N. Exosomes in glioma: mechanistic insights on biological, therapeutic, and diagnostic perspective. Ther Deliv 2025; 16:475-486. [PMID: 39957239 DOI: 10.1080/20415990.2025.2466410] [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: 07/27/2024] [Accepted: 02/10/2025] [Indexed: 02/18/2025] Open
Abstract
Gliomas are prominent and frequent primary malignant brain tumors, with a generally poor prognosis. Current treatment involves radiation, surgery and chemotherapy. Exosomes are nanoscale extracellular vesicles released by cells that enable biological molecule movement and encourage intercellular communication in the tumor microenvironment. This contributes to glioma development, radiation resistance, and overcomes chemotherapy. Exosome functional and structural properties are essential for understanding cancer molecular mechanisms. They can also treat invasive tumors like glioblastomas and serve as diagnostic markers. Recent research depicted exosomes' prominent role in cancer cell maintenance, intercellular signaling, and microenvironment modification. Exosomes hold nucleic acids, proteins, lipids, mRNAs, lncRNAs, miRNAs, and immunological regulatory molecules depending on the origin of the cell. This paper reviews exosomes, their role in glioma etiology, and perspective diagnostic and therapeutic uses.
Collapse
Affiliation(s)
- Reetika Tandon
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, India
| | - Samarth Kumar
- Formulation Research & Development-Non Orals, Sun Pharmaceuticals Industries Limited, Vadodara, India
| | - Mayank Handa
- Formulation Research & Development-Non Orals, Sun Pharmaceuticals Industries Limited, Vadodara, India
| | - Nidhi Srivastava
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow, India
| |
Collapse
|
131
|
Liu X, Lin Y, Zhuang Q, Deng H, Liu A, Sun J. BTK inhibitors resistance in B cell malignancies: Mechanisms and potential therapeutic strategies. Blood Rev 2025; 71:101273. [PMID: 40000280 DOI: 10.1016/j.blre.2025.101273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/02/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025]
Abstract
Bruton tyrosine kinase inhibitors (BTKi) have shown prominent clinical efficacy in patients with B cell malignancies, such as chronic lymphocytic leukemia, mantle cell lymphoma, diffuse large B cell lymphoma, and Waldenström's macroglobulinemia. Nevertheless, numerous factors contribute to BTKi resistance, encompassing genetic mutations, chromosomal aberrations, dysregulation of protein expression, tumor microenvironment, and metabolic reprogramming. Accordingly, potential therapeutic strategies have been explored to surmount BTKi resistance, including noncovalent BTKi, BTK proteolysis-targeting chimeras, and combination therapies. Herein, we summarize the mechanisms responsible for BTKi resistance as well as the current preclinical and clinical strategies to address BTKi resistance in B cell malignancies treatment.
Collapse
Affiliation(s)
- Xin Liu
- Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, Hangzhou, China; Department of Hematology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yufan Lin
- Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, Hangzhou, China; Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qiqi Zhuang
- Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, Hangzhou, China; Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haoren Deng
- Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, Hangzhou, China
| | - Aichun Liu
- Department of Hematology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Jie Sun
- Zhejiang Key Laboratory for Precision Diagnosis and Treatment of Hematological Malignancies, Hangzhou, China; Department of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Zhejiang Provincial Clinical Research Center for Hematological Disorders, Hangzhou, China.
| |
Collapse
|
132
|
Shin H, Hwang S, Jeong JH, Shin SC, Oh Y, Kim J, Hwang I, Kim EE, Choo H, Song EJ. Targeting USP47 enhances the efficacy of KRAS inhibitor in KRAS G12C mutated non-small cell lung cancer by controlling deubiquitination of c-Myc. Pharmacol Res 2025; 215:107722. [PMID: 40180254 DOI: 10.1016/j.phrs.2025.107722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 03/31/2025] [Accepted: 03/31/2025] [Indexed: 04/05/2025]
Abstract
FDA-approved KRASG12C inhibitors, like Sotorasib, target G12C-mutated KRAS in NSCLC. However, issues with insensitivity and drug resistance have emerged, requiring the development of new combination therapies to overcome these limitations. USP47 has been identified as a regulator of cancer-related signaling pathways such as Wnt, Hippo, and p53. However, its role in the KRAS signaling pathway remains largely unexplored and USP47 inhibitors are less developed than those targeting its homolog, USP7. Here, we identify USP47 as a novel therapeutic target in KRASG12C-mutated NSCLC and report K-552, a selective USP47 inhibitor, as a potential treatment strategy. We demonstrate that USP47 stabilizes c-Myc by preventing its proteasomal degradation through deubiquitination, thereby promoting NSCLC cell proliferation. Additionally, the compound K-552, a USP47 inhibitor identified through virtual screening, effectively destabilizes c-Myc and inhibits KRASG12C-mutated NSCLC cell proliferation. Furthermore, USP47 inhibition-either by siRNA knockdown or K-552 treatment-enhances the efficacy of Sotorasib in vitro and in vivo. Together, our findings establish USP47 as a promising therapeutic target in KRASG12C-mutated NSCLC and introduce K-552 as a USP47 inhibitor with potential for combination therapy with KRASG12C inhibitors.
Collapse
Affiliation(s)
- Hyungkyung Shin
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - SuA Hwang
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea; Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, Republic of Korea
| | - Jeong Hyun Jeong
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Sang Chul Shin
- Technological Convergence Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Yeonji Oh
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Jinhyeok Kim
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Inah Hwang
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea; Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, Republic of Korea
| | - Eunice EunKyeong Kim
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea.
| | - Hyunah Choo
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea.
| | - Eun Joo Song
- Graduate School of Pharmaceutical Sciences and College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea; Graduate Program in Innovative Biomaterials Convergence, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
133
|
Xin C, Chen Z, Zhou J, Chen Y, Liu Y, Liu H, Liang C, Zhu X, Qi Y, Wang A, Zhang G. Development and therapeutic assessment of bispecific nanobodies targeting B-cell activating factor and interleukin-17 for the neutralization of inflammatory mediators in autoimmune diseases. Int J Biol Macromol 2025; 305:140057. [PMID: 39870268 DOI: 10.1016/j.ijbiomac.2025.140057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/06/2025] [Accepted: 01/17/2025] [Indexed: 01/29/2025]
Abstract
Autoimmune diseases are characterized by dysregulated immune responses and chronic inflammation. B cell activating factor (BAFF) and interleukin-17 (IL-17) are key mediators in the pathogenesis of several autoimmune diseases, driving B cell hyperactivation, autoantibody production, and tissue damage. Simultaneous targeting of these pathways may provide a synergistic therapeutic approach. In this study, we developed a bispecific nanobody (BiNB) that simultaneously neutralizes BAFF and IL-17 through rational design and screening of a fully synthetic nanobody library. In vitro, BiNB effectively antagonized BAFF-induced B cell proliferation (IC50 = 0.38 nM) and IL-17-driven inflammatory responses (IC50 = 4.98 nM), without interference. In murine models, BiNB suppressed B cell-mediated immune responses and alleviated IL-17-induced systemic inflammation. Pharmacokinetic analysis showed a favorable half-life of ∼6 days and high bioavailability (91 %) after subcutaneous administration. Molecular docking identified BiNB's binding epitopes on BAFF and IL-17, explaining its dual mechanism of action. BiNB demonstrated strong antagonistic activity in vitro and effective target engagement in vivo, highlighting its potential as a therapeutic candidate for autoimmune diseases.
Collapse
Affiliation(s)
- Cheng Xin
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou 450001, China; Longhu Laboratory, Henan, Zhengzhou 450001, China; Henan Key Laboratory of Immunobiology, Henan, Zhengzhou 450001, China
| | - Zhuting Chen
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou 450001, China; Longhu Laboratory, Henan, Zhengzhou 450001, China; Henan Key Laboratory of Immunobiology, Henan, Zhengzhou 450001, China
| | - Jingming Zhou
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou 450001, China; Longhu Laboratory, Henan, Zhengzhou 450001, China; Henan Key Laboratory of Immunobiology, Henan, Zhengzhou 450001, China
| | - Yumei Chen
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou 450001, China; Longhu Laboratory, Henan, Zhengzhou 450001, China; Henan Key Laboratory of Immunobiology, Henan, Zhengzhou 450001, China
| | - Yankai Liu
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou 450001, China; Longhu Laboratory, Henan, Zhengzhou 450001, China; Henan Key Laboratory of Immunobiology, Henan, Zhengzhou 450001, China
| | - Hongliang Liu
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou 450001, China; Longhu Laboratory, Henan, Zhengzhou 450001, China; Henan Key Laboratory of Immunobiology, Henan, Zhengzhou 450001, China
| | - Chao Liang
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou 450001, China; Longhu Laboratory, Henan, Zhengzhou 450001, China; Henan Key Laboratory of Immunobiology, Henan, Zhengzhou 450001, China
| | - Xifang Zhu
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou 450001, China; Longhu Laboratory, Henan, Zhengzhou 450001, China; Henan Key Laboratory of Immunobiology, Henan, Zhengzhou 450001, China
| | - Yanhua Qi
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou 450001, China; Longhu Laboratory, Henan, Zhengzhou 450001, China; Henan Key Laboratory of Immunobiology, Henan, Zhengzhou 450001, China
| | - Aiping Wang
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou 450001, China; Longhu Laboratory, Henan, Zhengzhou 450001, China; Henan Key Laboratory of Immunobiology, Henan, Zhengzhou 450001, China.
| | - Gaiping Zhang
- School of Life Sciences, Zhengzhou University, Henan, Zhengzhou 450001, China; School of Advanced Agricultural Sciences, Peking University, Beijing 100000, China; Longhu Laboratory, Henan, Zhengzhou 450001, China; Henan Key Laboratory of Immunobiology, Henan, Zhengzhou 450001, China; College of Veterinary Medicine, Henan Agricultural University, Henan, Zhengzhou 450001, China.
| |
Collapse
|
134
|
Vasilogianni AM, Achour B, Al-Majdoub ZM, Peters SA, Barber J, Rostami-Hodjegan A. The quest to define cancer-specific systems parameters for personalized dosing in oncology. Expert Opin Drug Metab Toxicol 2025; 21:599-615. [PMID: 40042382 DOI: 10.1080/17425255.2025.2476560] [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: 10/09/2024] [Revised: 02/11/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025]
Abstract
INTRODUCTION Clinical trials in oncology initially recruit heterogeneous populations, without catering for all types of variability. The target cohort is often not representative, leading to variability in pharmacokinetics (PK). To address enrollment challenges in clinical trials, physiologically based pharmacokinetic models (PBPK) models can be used as a guide in the absence of large clinical studies. These models require patient-specific systems data relevant to the handling of drugs in the body for each type of cancer, which are scarce. AREAS COVERED This review explores system parameters affecting PK in cancer and highlights important gaps in data. Changes in drug-metabolizing enzymes (DMEs) and transporters have not been fully investigated in cancer. Their impaired expression can significantly affect capacity for drug elimination. Finally, the use of PBPK modeling for precision dosing in oncology is highlighted. Google Scholar and PubMed were mainly used for literature search, without date restriction. EXPERT OPINION Model-informed precision dosing is useful for dosing in sub-groups of cancer patients, which might not have been included in clinical trials. Systems parameters are not fully characterized in cancer cohorts, which are required in PBPK models. Generation of such data and application of cancer models in clinical practice should be encouraged.
Collapse
Affiliation(s)
- Areti-Maria Vasilogianni
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK
| | - Brahim Achour
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK
| | - Sheila Annie Peters
- Translational Quantitative Pharmacology, BioPharma, R&D Global Early Development, Merck KGaA, Darmstadt, Germany
- Translational Medicine and Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co., Ingelheim am Rhein, Germany
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, University of Manchester, Manchester, UK
- Certara Predictive Technologies (CPT), Simcyp Division, Sheffield, UK
| |
Collapse
|
135
|
Gao Y, He J, Wang J, Xu H, Ma L. Chimeric antigen receptor T cell immunotherapy for gynecological malignancies. Crit Rev Oncol Hematol 2025; 209:104680. [PMID: 40024355 DOI: 10.1016/j.critrevonc.2025.104680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
Gynecologic malignancies pose a serious threat to women's health worldwide. Although immunotherapy has significantly revolutionized cancer treatment strategies, effective therapeutic options for recurrent or advanced gynecologic malignancies are still deficient, posing significant challenges to clinical therapy. Chimeric antigen receptor (CAR) T cell therapy has achieved remarkable efficacy in treating hematologic malignancies, marking a significant change in the oncology treatment paradigm. However, despite the gradual increase in CAR T cell therapy used in treating solid tumors in recent years, its efficacy in treating gynecologic malignancies still needs further validation. This review will thoroughly examine CAR-T cell engineering and its mechanism of action on specific antigens associated with gynecologic malignancies, systematically assess the current application of CAR T cell therapy in gynecologic tumors and the advancements in clinical trials, and discuss the significant challenges and corresponding strategies, thereby offering a scientific foundation and guidance for future research in this area.
Collapse
Affiliation(s)
- Yajuan Gao
- Department of Gynecology and Obstetrics, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang 310008, China.
| | - Jing He
- Department of Emergency, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang 310008, China
| | - Jing Wang
- Department of Gynecology and Obstetrics, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang 310008, China
| | - Haiou Xu
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang 310006, China
| | - Lin Ma
- Department of Gynecology and Obstetrics, Hangzhou Women's Hospital (Hangzhou Maternity and Child Health Care Hospital), Hangzhou, Zhejiang 310008, China.
| |
Collapse
|
136
|
Wang Q, Sun Z, Guo J, Li H, Zhang J, Zhang B, Zhou B, Feng Y. Tumor-derived exosomal LINC01812 induces M2 macrophage polarization to promote perineural invasion in cholangiocarcinoma. Cancer Lett 2025; 617:217596. [PMID: 40081462 DOI: 10.1016/j.canlet.2025.217596] [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: 01/08/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/16/2025]
Abstract
M2 macrophages play a critical role in the tumor microenvironment of invasive solid tumors. They are closely associated with perineural invasion (PNI) and are often linked to poor prognosis. In this context, tumor-derived exosomes serve as important mediators of intercellular communication. However, the relationship between tumor cell-induced M2 macrophages and PNI in cholangiocarcinoma remains unexplored. In this study, we utilized multiplex immunofluorescence and transcriptomic sequencing to demonstrate the upregulation of LINC01812 in cholangiocarcinoma tissues and its positive correlation with M2 macrophage infiltration. Exosomal lncRNA sequencing, exosome uptake experiments, RNA pull-down assays, and mass spectrometry analysis demonstrated that macrophages can internalize exosomal LINC01812 and promote the M2 phenotype in cholangiocarcinoma cells. Additionally, Transwell and in vitro cocultures with the dorsal root ganglia confirmed that the tumor microenvironment significantly enhances the nerve infiltration of cholangiocarcinoma cells via M2 macrophages. The findings of this study indicate that exosomes containing LINC01812 derived from cholangiocarcinoma can induce M2 macrophage polarization and facilitate nerve infiltration, thereby providing new potential therapeutic targets for managing PNI in cholangiocarcinoma.
Collapse
Affiliation(s)
- Qinlei Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province, 266000, China.
| | - Zhaowei Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province, 266000, China.
| | - Jingyun Guo
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province, 266000, China.
| | - Haoran Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province, 266000, China.
| | - Jingru Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province, 266000, China.
| | - Bingyuan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province, 266000, China.
| | - Bin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province, 266000, China.
| | - Yujie Feng
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao City, Shandong Province, 266000, China.
| |
Collapse
|
137
|
Chen M, Zhou Y, Bao K, Chen S, Song G, Wang S. Multispecific Antibodies Targeting PD-1/PD-L1 in Cancer. BioDrugs 2025; 39:427-444. [PMID: 40106158 DOI: 10.1007/s40259-025-00712-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2025] [Indexed: 03/22/2025]
Abstract
The development of immune checkpoint inhibitors has revolutionized the treatment of patients with cancer. Targeting the programmed cell death protein 1 (PD-1)/programmed cell death 1 ligand 1(PD-L1) interaction using monoclonal antibodies has emerged as a prominent focus in tumor therapy with rapid advancements. However, the efficacy of anti-PD-1/PD-L1 treatment is hindered by primary or acquired resistance, limiting the effectiveness of single-drug approaches. Moreover, combining PD-1/PD-L1 with other immune drugs, targeted therapies, or chemotherapy significantly enhances response rates while exacerbating adverse reactions. Multispecific antibodies, capable of binding to different epitopes, offer improved antitumor efficacy while reducing drug-related side effects, serving as a promising therapeutic approach in cancer treatment. Several bispecific antibodies (bsAbs) targeting PD-1/PD-L1 have received regulatory approval, and many more are currently in clinical development. Additionally, tri-specific antibodies (TsAbs) and tetra-specific antibodies (TetraMabs) are under development. This review comprehensively explores the fundamental structure, preclinical principles, clinical trial progress, and challenges associated with bsAbs targeting PD-1/PD-L1.
Collapse
Affiliation(s)
- Miaomiao Chen
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Yuli Zhou
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Kaicheng Bao
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Siyu Chen
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China
| | - Guoqing Song
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China.
| | - Siliang Wang
- Department of Oncology, Shengjing Hospital of China Medical University, 36 Sanhao Road, Shenyang, 110004, China.
| |
Collapse
|
138
|
Jin J, Meng DD, Wen Y, Zhang QL, Lv FF, Chen GL, Ma XJ, Yu BH, Zhang SJ, Liu C, Xia ZG. PBVD regimen (pegylated liposomal doxorubicin, bleomycin, vincristine, dacarbazine) in classical Hodgkin lymphoma patients with cardiovascular risk factors: a retrospective study. Leuk Lymphoma 2025; 66:888-897. [PMID: 39756044 DOI: 10.1080/10428194.2024.2447888] [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: 09/30/2024] [Revised: 11/29/2024] [Accepted: 12/23/2024] [Indexed: 01/07/2025]
Abstract
This retrospective study aimed to evaluate the efficacy and safety of PBVD (pegylated liposomal doxorubicin [PLD], bleomycin, vinblastine, and dacarbazine) in the first-line treatment of classical Hodgkin lymphoma (cHL) patients with cardiovascular risk factors. Overall, 84 patients (53 had stage I-II and 31 had stage III-IV disease) received PBVD. The median PLD treatment duration was 16 weeks (interquartile range [IQR]: 8-24) for stage I-II and 24 weeks (IQR: 12-24) for stage III-IV. Among them, 56 (66.7%) received radiotherapy (45 with stage I-II and 11 with stage III-IV disease). Seventy-four (88.1%) patients achieved complete response. At a median follow-up of 49.7 months, 2- and 5-year progression-free survival were both 83.2%, and overall survival was 98.7% and 94.9%. Adverse events occurred in 73.8% of patients, including 7.1% cardiac events. No treatment-related deaths were observed. This approach showed a favorable benefit-to-risk profile in this population.
Collapse
Affiliation(s)
- Jia Jin
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dan-Dan Meng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Medical Oncology, Kaifeng Central Hospital, Kaifeng, China
| | - Yu Wen
- Department of Biostatistics, School of Public Health, Key Laboratory of Public Health Safety and Collaborative Innovation Center of Social Risks Governance in Health, Fudan University, Shanghai, China
| | - Qun-Ling Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fang-Fang Lv
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Guang-Liang Chen
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xue-Jun Ma
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Bao-Hua Yu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Sheng-Jian Zhang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Chang Liu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zu-Guang Xia
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
139
|
Li F, Zhang Y, Li C, Sun Q, Liu J, Qin T, Xu Z, Li B, Qu S, Pan L, Gao Q, Jiao M, Xiao Z. GATA1 insufficiencies in dysmegakaryopoiesis of myelodysplastic syndromes/neoplasms. Pathol Res Pract 2025; 269:155930. [PMID: 40168773 DOI: 10.1016/j.prp.2025.155930] [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: 09/02/2024] [Revised: 02/25/2025] [Accepted: 03/26/2025] [Indexed: 04/03/2025]
Abstract
GATA1 is one of critical transcription factors for megakaryopoiesis and platelet production. Our study aimed to explore the correlations between GATA1 expression and dysmegakaryopoiesis in myelodysplastic syndromes/neoplasm (MDS). We assessed GATA1 expression level of megakaryocytes by performing immunohistochemical staining on bone marrow biopsy sections from MDS patients. According to GATA1 expression level of megakaryocytes and positive megakaryocyte percentage, we assigned each patient a GATA1 score. Compared with TP53-wildtype patients, GATA1 scores significantly decreased in TP53-mutated patients (P < 0.001). Patients with abnormal karyotypes showed decreased GATA1 scores than those with normal karyotypes (P = 0.024). GATA1 expression levels were significantly downregulated in dysplastic megakaryocytes, especially micromegakaryocytes (P < 0.001). Furthermore, we explored the correlation between GATA1 expression levels and cytogenetic abnormalities of the same megakaryocyte using the morphology antibody chromosome (MAC) technique on fresh bone marrow smears. We found that GATA1-negative megakaryocytes had higher frequencies of cytogenetic abnormalities. Our results indicated that decreased GATA1 expression level of megakaryocytes was significantly associated with TP53 mutations, abnormal karyotypes and dysmegakaryopoiesis in MDS, suggesting that downregulation of GATA1 expression levels of megakaryocytes plays a critical role in the pathogenesis of MDS.
Collapse
Affiliation(s)
- Fuhui Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yudi Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Chengwen Li
- Hematologic Pathology Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Qi Sun
- Hematologic Pathology Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Jinqin Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Tiejun Qin
- MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Zefeng Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Bing Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Shiqiang Qu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Lijuan Pan
- MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Qingyan Gao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Meng Jiao
- MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Zhijian Xiao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; MDS and MPN Centre, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Hematologic Pathology Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China.
| |
Collapse
|
140
|
Sacramento CM, Casati MZ, Casarin RCV, Sallum EA, Silvério KG. Gastrointestinal cholecystokinin signaling pathway drugs modulate osteogenic/cementogenic differentiation of human periodontal ligament stem cells. J Dent 2025; 156:105657. [PMID: 40032153 DOI: 10.1016/j.jdent.2025.105657] [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: 12/16/2024] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025] Open
Abstract
OBJECTIVES Understanding the complexities of periodontal regeneration, particularly the unpredictable osteogenic/cementogenic differentiation of low-potential PDLSCs (LOP-PDLSCs), remains challenging. Identifying new therapeutic targets is crucial for enhancing regeneration. This study investigates the modulation of the Cholecystokinin (CCK) pathway, a key signaling cascade with roles in the gastrointestinal system, as a potential osteogenic/cementogenic pathway in PDLSCs. METHODS Gastrointestinal CCK-related drugs, Lorglumide and Sincalide, were tested for their effects on mineralization in PDLSCs. Lorglumide blocked the CCK pathway in high-potential PDLSCs (HOP-PDLSCs), while Sincalide enhanced mineralization in low-potential PDLSCs (LOP-PDLSCs). Cellular viability was tested under different drug concentrations, followed by a mineralization assay (AR-S) using non-toxic doses. RT-qPCR for osteogenic-related genes (IGF1, OCN, RUNX2) and CCK pathway-related genes (CCK, CCKAR, CCKBR, COX2, FOS, JNK3, RGS2) assessed gene modulation. Alkaline phosphatase (ALP) activity, Ca²⁺ quantification, and IP3 receptor phosphorylation were also evaluated. RESULTS Lorglumide reduced mineralization, ALP activity, and RUNX2, OCN, and IGF1 transcripts in HOP-PDLSCs (p < 0.05). It decreased CCK and CCKAR expression, modulated COX2, FOS, JNK3, and RGS2 genes, reduced IP3 receptor phosphorylation, and lowered calcium levels (p < 0.05). Conversely, Sincalide enhanced mineralization in LOP-PDLSCs, increasing ALP activity and OCN and IGF1 expression (p < 0.05). It upregulated COX2, FOS, JNK3, and RGS2 genes, phosphorylated IP3 receptors in LOP1, and increased calcium levels in all LOP-PDLSCs (p < 0.05). CONCLUSIONS Sincalide and Lorglumide modulate PDLSCs' osteogenesis/cementogenesis, revealing the complex interplay of gastrointestinal drugs in periodontal tissue regeneration and offering insights for innovative therapies. CLINICAL SIGNIFICANCE This study demonstrates the potential of gastrointestinal drugs targeting the CCK signaling pathway as innovative modulators for periodontal regeneration. By regulating osteogenic/cementogenic differentiation in hPDLSCs, these findings may pave the way for the development of novel biomaterials and therapies, promising improved outcomes in periodontal tissue regeneration for clinical applications.
Collapse
Affiliation(s)
- Catharina Marques Sacramento
- Departamento de Prótese e Periodontia, Divisão de Periodontia, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas - UNICAMP, Piracicaba, São Paulo, Brasil.
| | - Márcio Zaffalon Casati
- Departamento de Prótese e Periodontia, Divisão de Periodontia, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas - UNICAMP, Piracicaba, São Paulo, Brasil
| | - Renato Corrêa Viana Casarin
- Departamento de Prótese e Periodontia, Divisão de Periodontia, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas - UNICAMP, Piracicaba, São Paulo, Brasil
| | - Enilson Antonio Sallum
- Departamento de Prótese e Periodontia, Divisão de Periodontia, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas - UNICAMP, Piracicaba, São Paulo, Brasil
| | - Karina Gonzales Silvério
- Departamento de Prótese e Periodontia, Divisão de Periodontia, Faculdade de Odontologia de Piracicaba, Universidade Estadual de Campinas - UNICAMP, Piracicaba, São Paulo, Brasil
| |
Collapse
|
141
|
Saeed A, Tasleem Z, Muhammad SA, Ur Rehman A, Shah S, Jamil QA, Siddiqui H, Karuniawati H, Al-Tamimi SK. Economic Burden of Acute Myeloid Leukemia in European Union: Results from a Systematic Review of Literature. PHARMACOECONOMICS - OPEN 2025; 9:365-378. [PMID: 40169494 PMCID: PMC12037953 DOI: 10.1007/s41669-024-00554-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/22/2024] [Indexed: 04/03/2025]
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a heterogenous malignancy whose management is associated with considerable healthcare resource utilization and high expenditures because of recurrent and extended hospitalizations, multiple outpatient visits, and a wide range of supportive care. Modern therapies with improved safety profiles may assist in reducing healthcare costs; however, they are usually more expensive than standard chemotherapies. Few studies have addressed the expenses and burden of AML. Most of these studies were conducted in the USA. Very little research is available from the European Union (EU). OBJECTIVES The aim of this study was to assess the economic impact of AML and determine the major cost-driving factors for its treatment in the EU. METHODS This systematic review is in accordance with PRISMA guidelines. A systematic search was conducted using PubMed, Embase, ScienceDirect, SCOPUS, and Google Scholar databases to identify relevant studies on the economic impact of AML in various countries of the EU, published before April 15, 2024. Original studies investigating direct costs including expenses for treatment and healthcare services, or resource utilization for AML management were included. The systematic review excluded commentaries, editorials, and pharmacoeconomic modeling studies. Two reviewers independently performed data extraction and quality assessment, and the third reviewer resolved disagreements. We employed the Allison Larg Cost-of-Illness Studies evaluation checklist to assess the risk of bias. The mean cost per patient for induction, consolidation, and transplantation was calculated, and the results were converted into 2024 Euros. RESULTS Twenty-eight studies met our inclusion criteria, with the sample size of AML patients ranging from 12 to 39,568. The calculated per-patient direct costs of induction chemotherapy in Spain, France, Netherlands, Germany, and Italy were €92,378, €77,844, €61,643, €46,113, and €20,254, respectively. The mean per-patient direct cost of consolidation chemotherapy in the Netherlands and Germany was €42,137, and €32,220, respectively. The mean per-patient direct costs of transplantation in Sweden, Austria, France, Netherlands, and Spain were €192,628, €188,453, €132,352, €122,760, and €47,968, respectively. The cost-driving factors associated with AML treatment were inpatient hospitalization and medication costs. CONCLUSION AML seems to incur substantial direct economic expenses. Reducing the days of hospitalization can significantly decrease the economic burden of AML in the European Union. Moreover, there is a necessity for studies that comprehensively evaluate the economic implications, particularly concerning total and indirect costs. REGISTRATION Registered in PROSPERO under the registration number 'CRD42024537725'.
Collapse
Affiliation(s)
- Alisha Saeed
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Zermina Tasleem
- Department of Political Sciences, Bahauddin Zakariya University, Multan, Pakistan
| | - Sohail Ayaz Muhammad
- School of Management Sciences, University Sains Malaysia, Penang, Malaysia
- Operations and Supply Chain Department, Hasan Murad School of Management, University of Management and Technology, Lahore, Pakistan
| | - Anees Ur Rehman
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan.
| | - Shahid Shah
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Qurratul Ain Jamil
- Department of Pharmacy Practice, Faculty of Pharmacy, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Hajra Siddiqui
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Hidayah Karuniawati
- Department of Pharmacology and Clinical Pharmacy, Universitas Muhammadiyah Surakarta, Surakarta, Indonesia
| | | |
Collapse
|
142
|
Martínez-Gamboa DA, Hans R, Moreno-Cortes E, Figueroa-Aguirre J, Garcia-Robledo JE, Vargas-Cely F, Booth N, Castro-Martinez DA, Adams RH, Castro JE. CAR T-cell therapy landscape in pediatric, adolescent and young adult oncology - A comprehensive analysis of clinical trials. Crit Rev Oncol Hematol 2025; 209:104648. [PMID: 39900318 DOI: 10.1016/j.critrevonc.2025.104648] [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: 10/15/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/05/2025] Open
Abstract
Chimeric Antigen Receptor (CAR) T-cell therapy has emerged as a transformative approach in cancer treatment, particularly for hematologic malignancies. This therapy involves the genetic modification of patients' T-cells to target specific tumor antigens, bypassing the traditional MHC-TCR-mediated recognition. This innovation marks a significant step toward personalized medicine and precision oncology. In the pediatric, adolescent, and young adult (P-AYA) populations, Tisagenlecleucel (Kymriah®) exemplifies the success of CAR T-cell therapy, demonstrating significant efficacy in treating relapsed or refractory acute lymphoblastic leukemia (r/r ALL). However, the development of CAR T-cell therapies for P-AYA patients has not progressed as rapidly as for adults, with only one FDA approval for pediatric applications compared to six for adults up to 2024. Several challenges hinder the development of pediatric CAR T-cell therapies, including complex production logistics, limited clinical site access, restrictive patient eligibility criteria, and financial constraints, necessitating more effective incentives for pediatric oncology drug development independent of adult indications. To assess the current landscape of CAR T-cell therapy in P-AYA oncology, we conducted a comprehensive review of clinical trials registered on ClinicalTrials.gov up to May 2024. Our analysis included 77 trials exclusively targeting the P-AYA population from an initial pool of 40,690 studies filtered by age, dates, and specific criteria related to CAR T-cell interventions in cancer therapy. We found that 45 % of these trials originated from the USA and 30 % from China. The data retrieved from these trials provided insights into various aspects, including histological categories, antigenic targets, CAR-T generations, costimulatory domains, manufacturing processes, geographical distribution, and funding sources. This review highlighted a predominant focus on hematologic malignancies, particularly B-cell acute lymphoblastic leukemia (B-ALL), with significant attention to dual antigen targeting (CD19 and CD22) to address resistance mechanisms. Emerging targets such as GD2 for solid tumors and B7-H3 for various cancers also showed promise. Additionally, most trials still utilize second-generation CAR-T constructs with 4-1BB costimulatory domains, reflecting a conservative approach in pediatric populations. Our findings underscore the disparity in CAR T-cell therapy development between pediatric and adult populations, driven by distinct biological, ethical, and economic considerations. Pediatric cancers require specialized treatments tailored to the unique biology and genetic makeup of pediatric oncology. However, research and drug development have historically focused less on pediatric needs. Despite legislative efforts to promote pediatric oncology drug development, significant gaps remain. Clinical trials for P-AYA populations face challenges in patient enrollment, trial design, and funding, often relying on academic and non-profit institutions. Addressing these barriers is critical for advancing CAR T-cell therapy in pediatric oncology, improving outcomes, and ensuring equitable access to innovative treatments for these vulnerable populations. This review aims to inform future research and policy decisions, promoting advancements in CAR T-cell therapy for P-AYA cancer patients.
Collapse
Affiliation(s)
- David A Martínez-Gamboa
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA; Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA.
| | - Rhea Hans
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA; Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA; Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ, USA; Bone Marrow Transplant Fellow at Memorial Sloan Kettering Cancer Center, NY, USA
| | - Eider Moreno-Cortes
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA; Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Juana Figueroa-Aguirre
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA; Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Juan Esteban Garcia-Robledo
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA; Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Fabio Vargas-Cely
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA; Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| | - Natalie Booth
- Blood and Marrow Transplant Physician in the Cancer and Blood Disorders Institute at Johns Hopkins All Children's Hospital, USA
| | | | - Roberta H Adams
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA; Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Januario E Castro
- Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ, USA; Cancer Research and Cellular Therapies Laboratory, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
143
|
Fujioka Y, Abumiya M, Ono T, Takahashi N. Bosutinib for Successful Treatment-Free Remission in Chronic Myeloid Leukemia. Cancer Med 2025; 14:e70822. [PMID: 40296377 PMCID: PMC12037706 DOI: 10.1002/cam4.70822] [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: 12/05/2024] [Revised: 01/18/2025] [Accepted: 03/12/2025] [Indexed: 04/30/2025] Open
Abstract
INTRODUCTION AND OBJECTIVE This study explored the status of patients with chronic myeloid leukemia following the safe discontinuation of frontline or second-line bosutinib treatment. The goal was to assess the long-term outcomes and factors influencing treatment-free remission (TFR) following cessation of bosutinib therapy. METHODS The median duration of bosutinib treatment among 16 patients was 48 months. All patients achieved a deep molecular response before bosutinib discontinuation, which was sustained for a median pre-cessation period of 27 months. Patients were monitored for molecular response and clinical outcomes. RESULTS After bosutinib discontinuation, the major molecular response was lost in six patients: within 6 months in five patients and at 19 months in one patient. All six patients achieved a major molecular response after at least 3 months of bosutinib re-treatment. Ten patients exhibited successful TFR without loss of major molecular response for a median duration of 48 (16-101) months. Kaplan-Meier analysis revealed a 68.8% treatment-free survival at 12 months. After bosutinib cessation, eight patients developed Grade 1-2 withdrawal syndrome. No differences were observed in the clinical characteristics or bosutinib treatment between patients with TFR at 12 months (TFR group) and those without remission (recurrence group), except for the deep molecular response duration before bosutinib cessation (31 vs. 24 months, p = 0.009). T-cell profiling using flow cytometry revealed a higher percentage of effector memory CD8+ T cells at 1 and 3 months after bosutinib discontinuation in the TFR group than in the recurrence group (p = 0.012 and p = 0.005, respectively). CONCLUSION Bosutinib can be safely discontinued under certain conditions, similar to other tyrosine kinase inhibitors. Additionally, T-cell profile analysis before and after bosutinib discontinuation may predict successful TFR.
Collapse
MESH Headings
- Humans
- Aniline Compounds/therapeutic use
- Aniline Compounds/adverse effects
- Aniline Compounds/administration & dosage
- Nitriles/therapeutic use
- Nitriles/adverse effects
- Nitriles/administration & dosage
- Female
- Male
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/mortality
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Quinolines/therapeutic use
- Quinolines/adverse effects
- Quinolines/administration & dosage
- Middle Aged
- Aged
- Remission Induction
- Treatment Outcome
- Protein Kinase Inhibitors/therapeutic use
- Protein Kinase Inhibitors/adverse effects
- Protein Kinase Inhibitors/administration & dosage
- Adult
- Kaplan-Meier Estimate
- Fusion Proteins, bcr-abl/genetics
Collapse
Affiliation(s)
- Yuki Fujioka
- Department of Hematology, Nephrology and RheumatologyAkita University Graduate School of MedicineAkitaJapan
- Division of Central LaboratoryAkita University HospitalAkitaJapan
| | - Maiko Abumiya
- Department of PharmacyAkita University HospitalAkitaJapan
| | - Takaaki Ono
- Division of Transfusion and Cell TherapyHamamatsu University HospitalHamamatsuJapan
| | - Naoto Takahashi
- Department of Hematology, Nephrology and RheumatologyAkita University Graduate School of MedicineAkitaJapan
| |
Collapse
|
144
|
Roerden M, Spranger S. Cancer immune evasion, immunoediting and intratumour heterogeneity. Nat Rev Immunol 2025; 25:353-369. [PMID: 39748116 DOI: 10.1038/s41577-024-01111-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/05/2024] [Indexed: 01/04/2025]
Abstract
Cancers can avoid immune-mediated elimination by acquiring traits that disrupt antitumour immunity. These mechanisms of immune evasion are selected and reinforced during tumour evolution under immune pressure. Some immunogenic subclones are effectively eliminated by antitumour T cell responses (a process known as immunoediting), which results in a clonally selected tumour. Other cancer cells arise to resist immunoediting, which leads to a tumour that includes several distinct cancer cell populations (referred to as intratumour heterogeneity (ITH)). Tumours with high ITH are associated with poor patient outcomes and a lack of responsiveness to immune checkpoint blockade therapy. In this Review, we discuss the different ways that cancer cells evade the immune system and how these mechanisms impact immunoediting and tumour evolution. We also describe how subclonal antigen presentation in tumours with high ITH can result in immune evasion.
Collapse
Affiliation(s)
- Malte Roerden
- Koch Institute for Integrative Cancer Research, Massachusetts Institute for Technology, Cambridge, MA, USA
| | - Stefani Spranger
- Koch Institute for Integrative Cancer Research, Massachusetts Institute for Technology, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute for Technology, Cambridge, MA, USA.
- Ragon Institute of Mass General Hospital, Massachusetts Institute for Technology and Harvard, Cambridge, MA, USA.
| |
Collapse
|
145
|
Liu R, Guo L, Shi D, Sun X, Shang M, Zhao Y, Wang X, Yang Y, Xiao S, Li J. Multilayer cascade-response nanoplatforms as metabolic symbiotic disruptors to reprogram the immunosuppressive microenvironment. J Control Release 2025; 383:113797. [PMID: 40318807 DOI: 10.1016/j.jconrel.2025.113797] [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: 02/05/2025] [Revised: 04/22/2025] [Accepted: 04/28/2025] [Indexed: 05/07/2025]
Abstract
Nanomedicine is extensively utilized in tumor treatment, however, the restricted permeability of nanomaterials within tumor tissues, along with the inherent metabolic complexity of these tissues, have hindered effective control of tumor progression. Hypoxic and normoxic tumor cells utilize monocarboxylic acid transporters (MCTs) for the rapid reutilization of lactate, facilitating accelerated tumor growth. Here, cascade-response nanoplatforms (NPs) with contrast-enhanced ultrasound imaging (CEUI) capability had been established, incorporating basigin siRNA internally and featuring hyaluronidase (HAase) and γ-glutamyltranspeptidase (GGT)-responsive lipid coatings externally (GHB NPs). The GHB NPs took advantage of GGT-responsive HAase release to facilitate deep tumor penetration. Furthermore, ultrasound (US) irradiation decreased the expression of glycolysis-related proteins through the modulation of the β-catenin/c-Myc pathway, and US irradiation induced mitochondrial damage, leading to a low-energy state in tumor cells. On this basis, GHB NPs was paired with US stimulation to provide a combination therapy that disturbed tumor cell metabolic symbiosis and remodeled the immunosuppressive tumor microenvironment. This study formulates an effective therapeutic approach for metabolic-immunotherapy, potentially offering a viable candidate for tumor treatment.
Collapse
Affiliation(s)
- Rui Liu
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Lu Guo
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Dandan Shi
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Xiao Sun
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Mengmeng Shang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yading Zhao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Xiaoxuan Wang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Yuanyuan Yang
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Shan Xiao
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Jie Li
- Department of Ultrasound, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Department of Ultrasound, Qilu Hospital (Qingdao) of Shandong University, Qingdao, Shandong 266035, China.
| |
Collapse
|
146
|
Ayub Ali M, Maalouf MA, Feng D, Rashid M, Gehrke NR, Chhonker YS, Murry DJ, Wiemer DF, Holstein SA. Impact of fixed phosphorus position on activity of triazole bisphosphonates as geranylgeranyl diphosphate synthase inhibitors. Bioorg Med Chem 2025; 122:118140. [PMID: 40043324 DOI: 10.1016/j.bmc.2025.118140] [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: 01/10/2025] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025]
Abstract
Geranylgeranyl diphosphate synthase (GGDPS) produces the 20-carbon isoprenoid species used in protein geranylgeranylation reactions. Inhibition of GGDPS has emerged as a novel means of disrupting the activity of geranylgeranylated proteins in cancers such as myeloma and osteosarcoma. We have focused on developing a series of isoprenoid triazole bisphosphonate-based GGDPS inhibitors, demonstrating a complex structure-activity relationship (SAR), not only at the enzymatic level, but also at the cellular and whole organism levels. To further investigate this SAR, we have prepared a family of novel derivatives that have a fixed phosphorus position by virtue of vinyl, epoxy or cyclopropyl groups that incorporate the α-carbon position. Additional modifications include compounds with homocitronellyl chains instead of homogeranyl or homoneryl chains. All new compounds were evaluated in GGDPS enzyme assays and in cellular assays involving a panel of human myeloma and osteosarcoma cell lines. The homocitronellyl derivatives displayed markedly reduced activity in both enzymatic and cellular assays. While all of the homogeranyl/homoneryl vinyl/epoxy/cyclopropyl compounds had relatively similar activity in the enzyme assay (IC50's 0.37-2.87 μM), the cellular potencies varied more dramatically (ranging from 10 nM to no activity at 100 μM), depending on the olefin stereochemistry, the specific α-carbon modification and the tumor cell type. These findings, coupled with POM-prodrug and membrane permeability studies, support the hypothesis that there are specific membrane transporters mediating cellular uptake of these GGDPS inhibitors. Future studies focused on the identification of the membrane transporters responsible for the cellular uptake will enable further understanding of this complex SAR.
Collapse
Affiliation(s)
- Md Ayub Ali
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, USA; Department of Chemistry, Bangladesh University of Engineering and Technology (BUET), Dhaka 1000, Bangladesh
| | - Mona A Maalouf
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, USA
| | - Dan Feng
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mamunur Rashid
- Department of Pharmacy Practice, University of Nebraska Medical Center, Omaha, NE, USA
| | - Nathaniel R Gehrke
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, USA
| | - Yashpal S Chhonker
- Department of Pharmacy Practice, University of Nebraska Medical Center, Omaha, NE, USA
| | - Daryl J Murry
- Department of Pharmacy Practice, University of Nebraska Medical Center, Omaha, NE, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - David F Wiemer
- Department of Chemistry, University of Iowa, Iowa City, IA 52242-1294, USA
| | - Sarah A Holstein
- Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
147
|
Bushi A, Ma Y, Adu-Amankwaah J, Wang R, Cui F, Xiao R, Zhao J, Yuan J, Tan R. G protein-coupled estrogen receptor biased signaling in health and disease. Pharmacol Ther 2025; 269:108822. [PMID: 39978643 DOI: 10.1016/j.pharmthera.2025.108822] [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: 08/30/2024] [Revised: 02/03/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
G protein-coupled estrogen receptor (GPER) is now recognized for its pivotal role in cellular signaling, influencing diverse physiological processes and disease states. Unlike classical estrogen receptors, GPER exhibits biased signaling, wherein ligand binding triggers selective pathways over others, significantly impacting cellular responses. This review explores the nuanced mechanisms of biased signaling mediated by GPER, underscoring its relevance in cardiovascular health, neurological function, immune modulation, and oncogenic processes. Despite its critical implications, biased signaling through GPER remains underexplored compared to traditional signaling paradigms. We explore recent progress in understanding GPER signaling specificity and its potential therapeutic implications across various diseases. Future research directions aim to uncover the molecular basis of biased signaling, develop selective ligands, and translate these insights into personalized therapeutic approaches. Exploiting the therapeutic potential of GPER biased signaling represents a promising frontier in precision medicine, offering innovative strategies to address unmet medical needs.
Collapse
Affiliation(s)
- Aisha Bushi
- School international education, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Yixuan Ma
- First Clinical Medical School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Joseph Adu-Amankwaah
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Rong Wang
- The second clinical college, China Medical University, Shenyang, Liaoning 110122, China
| | - Fen Cui
- Research Institution of Behavioral Medicine Education, Jining Medical University, Jining 272067, China
| | - Rui Xiao
- Second Clinical Medical School, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Jinming Zhao
- Department of Pathology, College of Basic Medical Sciences, China Medical University, Shenyang 110122, China; Department of Pathology, The First Hospital of China Medical University, Shenyang 110001, China.
| | - Jinxiang Yuan
- Lin He's Academician Workstation of New Medicine and Clinical Translation, Jining Medical University, Jining, Shandong, China.
| | - Rubin Tan
- Department of Physiology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| |
Collapse
|
148
|
Aden D, Zaheer S, Sureka N, Trisal M, Chaurasia JK, Zaheer S. Exploring immune checkpoint inhibitors: Focus on PD-1/PD-L1 axis and beyond. Pathol Res Pract 2025; 269:155864. [PMID: 40068282 DOI: 10.1016/j.prp.2025.155864] [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: 08/31/2024] [Revised: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025]
Abstract
Immunotherapy emerges as a promising approach, marked by recent substantial progress in elucidating how the host immune response impacts tumor development and its sensitivity to various treatments. Immune checkpoint inhibitors have revolutionized cancer therapy by unleashing the power of the immune system to recognize and eradicate tumor cells. Among these, inhibitors targeting the programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have garnered significant attention due to their remarkable clinical efficacy across various malignancies. This review delves into the mechanisms of action, clinical applications, and emerging therapeutic strategies surrounding PD-1/PD-L1 blockade. We explore the intricate interactions between PD-1/PD-L1 and other immune checkpoints, shedding light on combinatorial approaches to enhance treatment outcomes and overcome resistance mechanisms. Furthermore, we discuss the expanding landscape of immune checkpoint inhibitors beyond PD-1/PD-L1, including novel targets such as CTLA-4, LAG-3, TIM-3, and TIGIT. Through a comprehensive analysis of preclinical and clinical studies, we highlight the promise and challenges of immune checkpoint blockade in cancer immunotherapy, paving the way for future advancements in the field.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India.
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Monal Trisal
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| |
Collapse
|
149
|
Bindu S, Bibi R, Pradeep R, Sarkar K. The evolving role of B cells in malignancies. Hum Immunol 2025; 86:111301. [PMID: 40132250 DOI: 10.1016/j.humimm.2025.111301] [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: 08/06/2024] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
B cells play diverse roles in different pathological circumstances, such as neoplastic diseases, autoimmune disorders, and neurological maladies. B cells, which are essential elements of the adaptive immune system, demonstrate exceptional functional variety, including the generation of antibodies, the presentation of antigens, and the secretion of cytokines. Within the field of oncology, B cells display a multifaceted nature in the tumor microenvironment, simultaneously manifesting both tumor-promoting and tumor-suppressing characteristics. Studies have found that the existence of tertiary lymphoid structures, which consist of B cells, is linked to better survival rates in different types of cancers. This article examines the involvement of B cells in different types of malignancies, emphasizing their importance in the development of the diseases and their potential as biomarkers. Additionally, the review also examines the crucial role of B cells in autoimmune illnesses and their potential as targets for therapy. The article also analyses the role of B cells in immunization and exploring their potential uses in cancer immunotherapy. This analysis highlights the intricate and occasionally contradictory roles of B cells, underlining the necessity for additional research to clarify their varied actions in various illness scenarios.
Collapse
Affiliation(s)
- Soham Bindu
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu 603203, India
| | - Roshni Bibi
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu 603203, India
| | - R Pradeep
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, Chennai, Tamil Nadu 603203, India.
| |
Collapse
|
150
|
Terroba-Navajas P, Lu IN, Quast I, Heming M, Keller CW, Ostendorf L, Hauser AE, Mothes R, Radbruch H, Stascheit F, Meisel AGO, Wiendl H, Meyer Zu Hörste G, Willcox N, Lünemann JD. Single-Cell Transcriptomics Identifies a Prominent Role for the MIF-CD74 Axis in Myasthenia Gravis Thymus. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200384. [PMID: 40117520 PMCID: PMC11978437 DOI: 10.1212/nxi.0000000000200384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 01/15/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND AND OBJECTIVES Myasthenia gravis (MG) is an autoimmune disease most frequently caused by autoantibodies (auto-Abs) against the acetylcholine receptor (AChR) located at the neuromuscular junction. Thymic follicular hyperplasia is present in most of the patients with early-onset AChR-Ab+ MG (EOMG), but its cellular and molecular drivers and development remain poorly understood. METHODS We constructed a single cell-based transcriptional profile of lymphoid cell types in thymi from 11 immunotherapy-naïve patients with EOMG. Multiplex histology and ELISA were used to determine migration inhibitory factor (MIF) levels. RESULTS Within EOMG thymi, we consistently observed 6 distinct clusters of B-cell populations maturing toward germinal center (GC)-associated and Ab-secreting cells, featuring prominent GC activity, as indicated by substantial clonal expansions and cycling B-cell subsets. Cell-cell interactome predictions identified strong interactions between T cells and GC-associated and memory B cells, dominated by B-cell prosurvival signaling through the MIF-CD74 axis. Multiplex histology confirmed abundant expression of CD74 in MG thymic B cells. Circulating MIF levels in EOMG correlated with higher disease severity as assessed by Myasthenia Gravis Foundation of America status. DISCUSSION Our data not only illustrate and define hyperplastic thymic niches in MG as favorable environments for pathogenic B-cell proliferation, maturation, and persistence but also suggest that the MIF-CD74 axis should be investigated for potential novel therapeutic targeting in EOMG.
Collapse
Affiliation(s)
- Paula Terroba-Navajas
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Germany
| | - I-Na Lu
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Germany
| | - Isaak Quast
- Department of Immunology, Monash University, Melbourne, VIC, Australia
| | - Michael Heming
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Germany
| | - Christian W Keller
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Germany
- Department of Neurology and Neurophysiology, University Hospital Freiburg, Germany
| | - Lennard Ostendorf
- Department of Nephrology and Medical Intensive Care, Charite Universitatsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
- BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Germany
| | - Anja Erika Hauser
- Department of Rheumatology and Clinical Immunology, Charite Universitatsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Berlin, Germany
| | - Ronja Mothes
- Deutsches Rheuma-Forschungszentrum (DRFZ), an Institute of the Leibniz Association, Berlin, Germany
- Department of Neuropathology, Charite Universitatsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Helena Radbruch
- Department of Neuropathology, Charite Universitatsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany
| | - Frauke Stascheit
- Department of Neurology with Experimental Neurology, Neuroscience Clinical Resarch Center, Charite Universitatsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and
| | - Andreas Georg Otto Meisel
- Department of Neurology with Experimental Neurology, Neuroscience Clinical Resarch Center, Charite Universitatsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Germany; and
| | - Heinz Wiendl
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Germany
- Department of Neurology and Neurophysiology, University Hospital Freiburg, Germany
| | - Gerd Meyer Zu Hörste
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Germany
| | - Nick Willcox
- Departments of Clinical Neurosciences, Royal Free Hospital, London Until 1988, Then Weatherall Institute of Molecular Medicine, University of Oxford, United Kingdom
| | - Jan D Lünemann
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Germany
| |
Collapse
|