101
|
Gagliardi PA, Pertz O. Gossiping about death: Apoptosis-induced ERK waves as coordinators of multicellular fate decisions. Semin Cell Dev Biol 2025; 171:103615. [PMID: 40279729 DOI: 10.1016/j.semcdb.2025.103615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/03/2025] [Accepted: 04/06/2025] [Indexed: 04/29/2025]
Abstract
Apoptosis is now recognized as a highly dynamic process that involves the release of a large set of signaling molecules that convey information to cells neighboring an apoptotic site. Recent studies in epithelial systems have discovered that apoptotic cells trigger waves of pulses of mitogen-activated protein kinase (MAPK) / extracellular signal-regulated kinase (ERK) pathway activity in their neighbors. At the single-cell level, the ERK pulses emerge from the MAPK pathway's excitable network properties, such as ultrasensitivity and adaptation. At the cell population level, apoptosis-induced ERK waves (AiEWs) emerge from propagation of ERK pulses across cells via a mechanism that involves mechanical inputs and paracrine signaling. AiEWs enable cell populations to dynamically coordinate fate decision signaling during tissue homeostasis and development. This spatio-temporal signaling mechanism can be hijacked by cancer cells to induce drug-tolerant persister states when apoptosis is triggered by cytotoxic or targeted therapies, undermining treatment efficacy. In this review, we summarize our current understanding of AiEWs, including their initiation, propagation, and coordination of fate decision signaling within a population. We discuss how the relatively simple properties of single cells, and their interactions within a collective coordinate these dynamic signaling patterns. We highlight their implication in resistance to cancer therapy and explore potential strategies to target these waves to re-sensitize cancer cells. Finally, we discuss emerging technologies and future directions to expand the study of this biological phenomenon.
Collapse
Affiliation(s)
| | - Olivier Pertz
- Institute of Cell Biology, University of Bern, Bern, Switzerland.
| |
Collapse
|
102
|
Dhar SS, Brown C, Rizvi A, Reed L, Kotla S, Zod C, Abraham J, Abe JI, Rajaram V, Chen K, Lee MG. Heterozygous Kmt2d loss diminishes enhancers to render medulloblastoma cells vulnerable to combinatory inhibition of LSD1 and OXPHOS. Cell Rep 2025; 44:115619. [PMID: 40286267 DOI: 10.1016/j.celrep.2025.115619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 02/17/2025] [Accepted: 04/04/2025] [Indexed: 04/29/2025] Open
Abstract
The histone H3 lysine 4 (H3K4) methyltransferase KMT2D (also called MLL4) is one of the most frequently mutated epigenetic modifiers in many cancers, including medulloblastoma (MB). Notably, heterozygous KMT2D loss frequently occurs in MB and other cancers. However, its oncogenic role remains largely uncharacterized. Here, we show that heterozygous Kmt2d loss in murine cerebellar regions promotes MB genesis driven by heterozygous loss of the MB-suppressor gene Ptch via the upregulation of tumor-promoting programs (e.g., oxidative phosphorylation [OXPHOS]). Downregulation of the transcription-repressive tumor suppressor NCOR2 by heterozygous Kmt2d loss, along with Ptch+/--increased MYCN, upregulated tumor-promoting genes. Heterozygous Kmt2d loss substantially diminished enhancer marks (H3K4me1 and H3K27ac) and the H3K4me3 signature, including those for Ncor2. Combinatory pharmacological inhibition of the enhancer-decommissioning H3K4 demethylase LSD1 and OXPHOS significantly reduced the tumorigenicity of MB cells bearing heterozygous Kmt2d loss. Our findings suggest the molecular and epigenetic pathogenesis underlying the MB-promoting effect of heterozygous KMT2D loss.
Collapse
Affiliation(s)
- Shilpa S Dhar
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| | - Calena Brown
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Ali Rizvi
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Lauren Reed
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Constantin Zod
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Janak Abraham
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Jun-Ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Veena Rajaram
- Department of Pathology, The University of Texas Southwestern Medical Center, Dallas, TX 75235, USA
| | - Kaifu Chen
- Basic and Translational Research Division, Department of Cardiology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
| | - Min Gyu Lee
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
103
|
Ma Q, Zhu Y, Zhang D, Su X, Jiang C, Zhang Y, Zhang X, Han N, Shu G, Yin G, Wang M. Reprogramming and targeting of cholesterol metabolism in tumor-associated macrophages. J Mater Chem B 2025. [PMID: 40266660 DOI: 10.1039/d5tb00236b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Cholesterol, as a major component of cell membranes, is closely related to the metabolic regulation of cells and organisms; tumor-associated macrophages play an important push role in tumor progression. We know that tumor-associated macrophages are polarized from macrophages, and the abnormalities of cholesterol metabolism that may be induced during their polarization are worth discussing. This manuscript focuses on metabolic abnormalities in tumor-associated macrophages, and first provides a basic summary of the regulatory mechanisms of abnormal macrophage polarization. Subsequently, it comprehensively describes the features of abnormal glucose, lipid and cholesterol metabolism in TAMs as well as the different regulatory pathways. Then, the paper also discusses the link between abnormal cholesterol metabolism in TAMs and tumors, chronic diseases and aging. Finally, the paper summarizes cancer therapeutic strategies targeting cholesterol metabolism that are already in clinical trials, as well as nanomaterials capable of targeting cholesterol metabolism that are in the research stage, in the hope of providing value for the design of targeting materials. Overall, elucidating metabolic abnormalities in tumor-associated macrophages, particularly cholesterol metabolism, could provide assistance in tumor therapy and the design of targeted drugs.
Collapse
Affiliation(s)
- Qiaoluo Ma
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Ying Zhu
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Dongya Zhang
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Xiaohan Su
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Can Jiang
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Yuzhu Zhang
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Xingting Zhang
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Na Han
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Guang Shu
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Gang Yin
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| | - Maonan Wang
- Department of Pathology, Xiangya Hospital, Xiangya School of Basic Medical Sciences, Central South University, Changsha, China.
| |
Collapse
|
104
|
Schaub-Clerigué A, Hermanova I, Pintor-Rial A, Sydorenko M, Valcarcel-Jimenez L, Macchia A, Lectez B, Garcia-Longarte S, Fagoaga-Eugui M, Astobiza I, Martín-Martín N, Zabala-Letona A, Pujana-Vaquerizo M, Royo F, Azkargorta M, Berra E, Sutherland JD, Peinado H, Falcón-Perez JM, Elortza F, Carracedo A, Torrano V. Secreted spermidine synthase reveals a paracrine role for PGC1α-induced growth suppression in prostate cancer. Cell Death Dis 2025; 16:330. [PMID: 40268923 PMCID: PMC12019391 DOI: 10.1038/s41419-025-07639-4] [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: 10/08/2024] [Revised: 04/01/2025] [Accepted: 04/04/2025] [Indexed: 04/25/2025]
Abstract
Prostate cancer is the fifth cause of death by cancer worldwide, second in incidence in the male population. The definition of the molecular basis of its development and the oncogenic signals driving lethality continue to be important objectives in prostate cancer research. Prior work from others and us has demonstrated that loss of PGC1α expression results in a metabolic, signaling and transcriptional reprogramming that supports the development of metastatic disease. However, we do not fully understand the spectrum of tumor suppressive effects regulated by this co-regulator. Here we show that PGC1α governs non-cell autonomous paracrine tumor suppression in prostate cancer. A systematic analysis of the transcriptional landscapes associated to PGC1α loss of expression revealed that PGC1α alters the expression of genes encoding for secreted proteins. Cell secretome studies corroborated that PGC1α-dependent ERRα regulation in prostate cancer cells suppresses the growth of tumor cells exposed to their conditioned media, independently of androgen receptor status. The integration of in vitro and in vivo secretomics data and genetic perturbation assays revealed spermidine synthase as a transcriptional target of PGC1α and mediator of the paracrine metabolic growth suppressive effect. Moreover, the activity of the regulatory axis PGC1α-ERRα-SRM was reflected in patients and had prognostic value. Altogether, this work provides unprecedented evidence of the non-cell autonomous suppressive role of PGC1α, which broadens the view of this co-regulator as a multifactorial tumor suppressor in prostate cancer.
Collapse
Affiliation(s)
- Ariane Schaub-Clerigué
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Leioa, Spain
- Cancer Cell Signaling and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Ivana Hermanova
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Ainara Pintor-Rial
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Mariia Sydorenko
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Lorea Valcarcel-Jimenez
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Leioa, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Alice Macchia
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Leioa, Spain
- Cancer Cell Signaling and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Benoit Lectez
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Saioa Garcia-Longarte
- Cancer Cell Signaling and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Maider Fagoaga-Eugui
- Cancer Cell Signaling and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Ianire Astobiza
- Cancer Cell Signaling and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Natalia Martín-Martín
- Cancer Cell Signaling and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Amaia Zabala-Letona
- Cancer Cell Signaling and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Traslational prostate cancer Research lab, CIC bioGUNE-Basurto, Biobizkaia Health Research Institute, Bilbao, Spain
| | - Mikel Pujana-Vaquerizo
- Cancer Cell Signaling and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Félix Royo
- Exosome Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Mikel Azkargorta
- Proteomic Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Edurne Berra
- Cancer Cell Signaling and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - James D Sutherland
- Ubiquitin-likes And Development Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Héctor Peinado
- Microenvironment and Metastasis Laboratory, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Juan Manuel Falcón-Perez
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Exosome Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Félix Elortza
- Proteomic Platform, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
| | - Arkaitz Carracedo
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Leioa, Spain
- Cancer Cell Signaling and Metabolism Laboratory, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
- Traslational prostate cancer Research lab, CIC bioGUNE-Basurto, Biobizkaia Health Research Institute, Bilbao, Spain
| | - Verónica Torrano
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Leioa, Spain.
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
105
|
Gujar V, Li H, Paull TT, Neumann CA, Weyemi U. Unraveling the nexus: Genomic instability and metabolism in cancer. Cell Rep 2025; 44:115540. [PMID: 40208791 PMCID: PMC12043202 DOI: 10.1016/j.celrep.2025.115540] [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: 11/25/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/12/2025] Open
Abstract
The DNA-damage response (DDR) is a signaling network that enables cells to detect and repair genomic damage. Over the past three decades, inhibiting DDR has proven to be an effective cancer therapeutic strategy. Although cancer drugs targeting DDR have received approval for treating various cancers, tumor cells often develop resistance to these therapies, owing to their ability to undergo energetic metabolic reprogramming. Metabolic intermediates also influence tumor cells' ability to sense oxidative stress, leading to impaired redox metabolism, thus creating redox vulnerabilities. In this review, we summarize recent advances in understanding the crosstalk between DDR and metabolism. We discuss combination therapies that target DDR, metabolism, and redox vulnerabilities in cancer. We also outline potential obstacles in targeting metabolism and propose strategies to overcome these challenges.
Collapse
Affiliation(s)
- Vaibhavi Gujar
- NCI Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Haojian Li
- NCI Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tanya T Paull
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, USA
| | - Carola A Neumann
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Urbain Weyemi
- NCI Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
106
|
Tajpara P, Sobkowiak MJ, Healy K, Naud S, Gündel B, Halimi A, Khan ZA, Gabarrini G, Le Guyader S, Imreh G, Reisz JA, Del Chiaro M, D’Alessandro A, Heuchel R, Löhr JM, Özenci V, Sällberg Chen M. Patient-derived pancreatic tumor bacteria exhibit oncogenic properties and are recognized by MAIT cells in tumor spheroids. Front Immunol 2025; 16:1553034. [PMID: 40330456 PMCID: PMC12053177 DOI: 10.3389/fimmu.2025.1553034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 03/19/2025] [Indexed: 05/08/2025] Open
Abstract
Introduction Tumor-residing microbiota poses a new challenge in cancer progression and therapy; however, the functional behavior of patient tumor-derived microbes remains poorly understood. We previously reported the presence of tumor microbiota in intraductal papillary mucinous neoplasms (IPMNs), which are precursors of pancreatic cancer. Methods We examined the metabolic and pathogenic potential of clinical microbiota strains obtained from IPMN tumors using various pancreatic cell lines and 3D spheroid models. Results Our findings revealed that several strains from IPMNs with invasive cancer or high-grade dysplasia, such as E. cloacae, E. faecalis, and K. pneumoniae, induced a cancer metabolite signature in human pancreatic cells when infected ex vivo. Bacterial invasiveness was significantly correlated with DNA damage in spheroids derived from normal and tumor-derived pancreatic cells, particularly in strains derived from advanced neoplasia IPMN and under hypoxic conditions. Additionally, microbial metabolites activate human mucosal-associated invariant T (MAIT) cells and restrict the infection, both extra- and intracellularly, in hypoxic tumor conditions and in synergy with antibiotics. Discussion Immune sensing of tumor microbiota metabolites may have clinical implications in cancer management.
Collapse
Affiliation(s)
- Poojabahen Tajpara
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Michał Jacek Sobkowiak
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
- Division of Oral Diagnostics and Surgery, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Katie Healy
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Sabrina Naud
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Beate Gündel
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Sweden
| | - Asif Halimi
- Division of Surgery, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
- Department of Surgical and Perioperative Sciences, Surgery, Umeå University, Umea, Sweden
| | - Zara Ahmad Khan
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Giorgio Gabarrini
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Sylvie Le Guyader
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Gabriela Imreh
- Department of Medicine Huddinge, Karolinska Institutet, Huddinge, Sweden
| | - Julie A. Reisz
- Department of Biochemistry and Molecular Genetics, University of Colorado, Aurora, CO, United States
| | - Marco Del Chiaro
- Division of Surgical Oncology, Department of Surgery, University of Colorado of Medicine, Aurora, CO, United States
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado, Aurora, CO, United States
| | - Rainer Heuchel
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Sweden
| | - J Matthias Löhr
- Pancreas Cancer Research Lab, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Huddinge, Sweden
| | - Volkan Özenci
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Margaret Sällberg Chen
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
- Division of Oral Diagnostics and Surgery, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer Center, Stockholm, Sweden
| |
Collapse
|
107
|
Rao K, Zhang X, Luo Y, Xia Q, Jin Y, He J. Lactylation orchestrates ubiquitin-independent degradation of cGAS and promotes tumor growth. Cell Rep 2025; 44:115441. [PMID: 40106438 DOI: 10.1016/j.celrep.2025.115441] [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: 07/12/2024] [Revised: 01/06/2025] [Accepted: 02/26/2025] [Indexed: 03/22/2025] Open
Abstract
Lactate extensively associates with metabolic reprogramming, signal transduction, and immune modulation. Nevertheless, the regulatory role of lactate in immune sensing of cytosolic DNA remains uncertain. Here, we report that lactate serves as an initiator to facilitate proteasomal degradation of cyclic GMP-AMP synthase (cGAS) independent of ubiquitin, thus repressing the production of interferon and contributing to tumor growth. Mechanistically, lactylation of K21 stimulates cGAS translocation from the nucleus to the proteasome for degradation, which is compromised by phosphorylation of PSMA4 S188 via disrupting its association with cGAS. Concurrently, lactylation of K415 rewires PIK3CB activity and impairs ULK1-driven phosphorylation of PSMA4 S188. Physiologically, lactylation of cGAS sustains tumor growth. Expression of cGAS correlates with the antitumor effect of the LDHA inhibitor FX11. Finally, the lactate-cGAS axis indicates a prognostic outcome of lung adenocarcinoma. Collectively, these findings not only put forth a mechanism of cGAS degradation but also unravel the clinical relevance of cGAS lactylation.
Collapse
Affiliation(s)
- Keqiang Rao
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China; Shanghai Institute of Transplantation, Shanghai 200120, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai 200120, China
| | - Xinchao Zhang
- Department of Pathology, College of Basic Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yi Luo
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China; Shanghai Institute of Transplantation, Shanghai 200120, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai 200120, China
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China; Shanghai Institute of Transplantation, Shanghai 200120, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai 200120, China.
| | - Yuting Jin
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China; Shanghai Institute of Transplantation, Shanghai 200120, China; Shanghai Engineering Research Center of Transplantation and Immunology, Shanghai 200120, China.
| | - Jing He
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200120, China.
| |
Collapse
|
108
|
Dai J, Rozenblit M, Li X, Shan NL, Wang Y, Mane S, Marczyk M, Pusztai L. Genomic alterations in normal breast tissues preceding breast cancer diagnosis. Breast Cancer Res 2025; 27:60. [PMID: 40264151 PMCID: PMC12013151 DOI: 10.1186/s13058-025-02018-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Accepted: 04/07/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Normal breast tissues adjacent to cancer often harbor many of the same genomic alterations as the cancer itself. However, it remains unclear whether histologically normal breast tissues carry genomic changes related to cancer development years before a cancer diagnosis. METHODS Whole exome sequencing was performed to examine germline and somatic alterations in histologically normal breast tissues from women who subsequently developed breast cancer (n = 79, pre-diagnosis tissues) and compared these with results from breast tissues of women who did not (n = 81). No patient had germline mutations in cancer predisposition genes. RESULTS The pre-diagnosis tissues had significantly more high functional impact germline variants per sample than the healthy controls (P = 0.034), 36.5% of affected genes were cancer hallmark genes, among these 62.4% were involved with evading growth suppressors and 5.7% with genome instability. The average number of somatic mutations were similar between the two cohorts. Mutation signature analysis revealed COSMIC signatures 3 (associated with impaired homologous recombination) as a dominant signature more frequent in pre-diagnosis tissues. At gene and variant level, nine common germline polymorphisms in two immune regulatory genes, FCGBP and TPSBP2, and along with three somatic mutations in F13A1, FRY and TMLHE, were significantly more frequently mutated in the pre-diagnosis samples. CONCLUSIONS Individuals who develop breast cancer have a higher germline variant burden in normal breast tissues leading to subtle deficiencies in DNA repair that in the context of other germline and somatic mutations could facilitate malignant transformation.
Collapse
Affiliation(s)
- Jiawei Dai
- Yale Cancer Center, Yale School of Medicine, Suite 120, Rm 133, 300 George Street, New Haven, CT, 06511, USA
| | - Mariya Rozenblit
- Yale Cancer Center, Yale School of Medicine, Suite 120, Rm 133, 300 George Street, New Haven, CT, 06511, USA
| | - Xiaoyue Li
- Yale Cancer Center, Yale School of Medicine, Suite 120, Rm 133, 300 George Street, New Haven, CT, 06511, USA
| | - Naing Lin Shan
- Yale Cancer Center, Yale School of Medicine, Suite 120, Rm 133, 300 George Street, New Haven, CT, 06511, USA
| | - Yueyue Wang
- Yale Cancer Center, Yale School of Medicine, Suite 120, Rm 133, 300 George Street, New Haven, CT, 06511, USA
| | - Shrikant Mane
- Yale Center for Genome Analysis, West Haven, CT, USA
| | - Michal Marczyk
- Department of Data Mining and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Lajos Pusztai
- Yale Cancer Center, Yale School of Medicine, Suite 120, Rm 133, 300 George Street, New Haven, CT, 06511, USA.
| |
Collapse
|
109
|
Barbosa JMG, de Oliveira CG, Soares MFG, Vieira LFM, Filho OC, Cardoso DMM, Beato PMM, Moro CATM, de Oliveira AE, Antoniosi Filho NR. Cerumenogram as an assay for the metabolic diagnosis of precancer, cancer, and cancer remission. Sci Rep 2025; 15:13929. [PMID: 40263376 PMCID: PMC12015372 DOI: 10.1038/s41598-025-97440-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/04/2025] [Indexed: 04/24/2025] Open
Abstract
Early diagnosis is crucial for successful cancer treatment. As a mitochondrial metabolic disease, cancer produces volatile organic metabolites that are present in earwax, allowing differentiation between healthy individuals and those with cancer through an assay called cerumenogram. In this case series study, we demonstrated that this assay also enables the diagnosis of precancerous stages, such as hypermetabolic inflammation and dysplasia, which can aid in treatments to prevent cancer progression. Additionally, this assay reveals that oncological metabolism differs from that observed in metaplasias, cysts, and benign tumors, helping to avoid unnecessary oncological procedures due to suspected malignancy. Cerumenogram can also be used to assess cancer remission. Thus, the cerumenogram emerges as an assay that might enable the diagnosis of cancer, monitor remission, and identify precancerous stages, covering key steps of tumorigenesis.
Collapse
Affiliation(s)
- João Marcos Gonçalves Barbosa
- Laboratório de Métodos de Extração e Separação (LAMES), Instituto de Química (IQ), Universidade Federal de Goiás (UFG), Campus II - Samambaia, Goiânia, GO, 74690-900, Brazil
| | - Camilla Gabriela de Oliveira
- Laboratório de Métodos de Extração e Separação (LAMES), Instituto de Química (IQ), Universidade Federal de Goiás (UFG), Campus II - Samambaia, Goiânia, GO, 74690-900, Brazil
| | - Marina Ferraz Gontijo Soares
- Laboratório de Métodos de Extração e Separação (LAMES), Instituto de Química (IQ), Universidade Federal de Goiás (UFG), Campus II - Samambaia, Goiânia, GO, 74690-900, Brazil
| | | | - Omar Carneiro Filho
- Insituto de Medicina Nuclear (IMEN), Alameda dos Buritis, 600, St. Central, Goiânia, GO, 74015-080, Brazil
| | | | | | | | - Anselmo Elcana de Oliveira
- Laboratório de Química Teórica e Computacional (LQTC), Instituto de Química (IQ), Universidade Federal de Goiás (UFG), Campus II - Samambaia, Goiânia, GO, 74690-970, Brazil
| | - Nelson Roberto Antoniosi Filho
- Laboratório de Métodos de Extração e Separação (LAMES), Instituto de Química (IQ), Universidade Federal de Goiás (UFG), Campus II - Samambaia, Goiânia, GO, 74690-900, Brazil.
| |
Collapse
|
110
|
Choppavarapu L, Fang K, Liu T, Ohihoin AG, Jin VX. Hi-C profiling in tissues reveals 3D chromatin-regulated breast tumor heterogeneity informing a looping-mediated therapeutic avenue. Cell Rep 2025; 44:115450. [PMID: 40112000 DOI: 10.1016/j.celrep.2025.115450] [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: 08/23/2024] [Revised: 01/12/2025] [Accepted: 02/28/2025] [Indexed: 03/22/2025] Open
Abstract
The limitations of Hi-C (high-throughput chromosome conformation capture) profiling in in vitro cell culture include failing to recapitulate disease-specific physiological properties and lacking a clinically relevant disease microenvironment. In this study, we conduct Hi-C profiling in a pilot cohort of 12 breast tissues comprising two normal tissues, five ER+ breast primary tumors, and five tamoxifen-treated recurrent tumors. We demonstrate 3D chromatin-regulated breast tumor heterogeneity and identify a looping-mediated target gene, CA2, which might play a role in driving tamoxifen resistance. The inhibition of CA2 impedes tumor growth both in vitro and in vivo and reverses chromatin looping. The disruption of CA2 looping reduces tamoxifen-resistant cancer cell proliferation, decreases CA2 mRNA and protein expression, and weakens the looping interaction. Our study thus provides mechanistic and functional insights into the role of 3D chromatin architecture in regulating breast tumor heterogeneity and informs a new looping-mediated therapeutic avenue for treating breast cancer.
Collapse
Affiliation(s)
- Lavanya Choppavarapu
- Divison of Biostatistics, Data Science Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Kun Fang
- Divison of Biostatistics, Data Science Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Tianxiang Liu
- Divison of Biostatistics, Data Science Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Aigbe G Ohihoin
- Cell and Developmental Biology PhD program, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Victor X Jin
- Divison of Biostatistics, Data Science Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA; MCW Cancer Center, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
111
|
Wu MJ, Yang SM, Fang WK, Chen TJ, Wu CY, Hsu YJ, Shen CE, Cheng YC, Hsieh WC, Yuh CH, Yang MH, Kung HJ, Wang WC. KDM4C works in concert with GATA1 to regulate heme metabolism in head and neck squamous cell carcinoma. Cell Mol Life Sci 2025; 82:170. [PMID: 40259045 PMCID: PMC12011672 DOI: 10.1007/s00018-025-05693-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 03/19/2025] [Accepted: 04/01/2025] [Indexed: 04/23/2025]
Abstract
Head and neck squamous cell carcinoma (HNSCC), the sixth most common cancer worldwide, presents significant public health challenges due to its genetic instability and late-stage diagnosis. Despite advancements in treatment, the median overall survival remains below one year, emphasizing the need for improved detection, prognosis, and therapeutic strategies. This study investigates the role of KDM4C and its interaction with GATA1 in regulating heme metabolism and tumor progression in HNSCC. KDM4C knockdown (KDM4C-KD) hindered HNSCC cell migration using in vitro assays, inhibited metastasis through zebrafish xenotransplantation, and suppressed tumor growth in mouse xenograft models. RNA-seq and CUT&Tag-seq analyses on KDM4C-KD SAS cells identified KDM4C-regulated genes, including ferrochelatase (FECH), in heme metabolism. Immunoprecipitation and docking analyses confirmed the KDM4C-GATA1 interaction. Notably, FECH overexpression in KDM4C or GATA1 knockdown cells restored cell migration, invasion, and proliferation, highlighting FECH as a crucial downstream target. KDM4 inhibitors myricetin and BPRKD022S0 (22S0) increased H3K9me3 levels, downregulated heme metabolism genes, and reduced cell survival in HNSCC cells. Zebrafish and mouse models demonstrated that these inhibitors effectively suppressed tumor growth and metastasis. Immunohistochemical analysis of HNSCC patient samples revealed high KDM4C and GATA1 expression correlated with advanced clinical stages and poor survival outcomes. Our findings elucidate the critical role of the KDM4C/GATA1-FECH axis in HNSCC progression and suggest that targeting this pathway with KDM4 inhibitors shows promising therapeutic potential for HNSCC treatment.
Collapse
Affiliation(s)
- Meng-Jen Wu
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Shan-Min Yang
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Wei-Kai Fang
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Tsan-Jan Chen
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Chun-Yi Wu
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Yen-Jung Hsu
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Cheng-En Shen
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Yu-Chia Cheng
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Wan-Chen Hsieh
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC
| | - Chiou-Hwa Yuh
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, 35053, Taiwan, ROC
| | - Muh-Hwa Yang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, 11221, Taiwan, ROC
| | - Hsing-Jien Kung
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, 11031, Taiwan, ROC
- Department of Biochemistry and Molecular Medicine, University of California Davis School of Medicine, University of California Davis Cancer Centre, Sacramento, CA, 95817, USA
| | - Wen-Ching Wang
- Institute of Molecular and Cellular Biology and Department of Life Science, National Tsing-Hua University, Hsinchu, 30013, Taiwan, ROC.
| |
Collapse
|
112
|
Yamada Y, Sankoda N, Yamada Y. In Vivo Reprogramming Highlights Epigenetic Regulation That Shapes Cancer Hallmarks. Cancer Sci 2025. [PMID: 40259515 DOI: 10.1111/cas.70067] [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: 12/29/2024] [Revised: 03/07/2025] [Accepted: 03/19/2025] [Indexed: 04/23/2025] Open
Abstract
Douglas Hanahan added "non-mutational epigenetic reprogramming" and "unlocking phenotypic plasticity" as new hallmarks of cancer, proposing that cancer cells possess fundamental features that are not directly linked to their genetic abnormalities. In vivo reprogramming studies have demonstrated that non-mutational epigenetic regulation can cause cellular reprogramming, leading to cancer development at the organismal level. Given that epigenetic regulation functions as an interface between the cellular environment and gene expression, these results suggest that intercellular communications in the tumor microenvironment play a critical role in cancer development. This review first introduces genetic aberrations that cause cancer development. Then, it illustrates the impact of epigenetic abnormalities in cancer, especially with reference to studies that use in vivo reprogramming technologies. Finally, it discusses the importance of histological evaluations of tumor tissue to understand non-cell-autonomous epigenetic regulation that establishes cancer hallmarks.
Collapse
Affiliation(s)
- Yosuke Yamada
- Department of Molecular Pathology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Nao Sankoda
- Department of Molecular Pathology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yasuhiro Yamada
- Department of Molecular Pathology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
113
|
Zhang Y, Ye L, Qin Y, Qiu C, Sun Q, Fan T, Chen Y, Jiang Y. Serum metabolomics to identify molecular subtypes and predict XELOX efficacy in colorectal cancer. Sci Rep 2025; 15:13671. [PMID: 40258977 PMCID: PMC12012017 DOI: 10.1038/s41598-025-97463-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/04/2025] [Indexed: 04/23/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers; however, accurately predicting prognosis based on existing molecular subtypes remains challenging. The XELOX regimen, which combines oxaliplatin and capecitabine, is the cornerstone of chemotherapy for CRC treatment. However, there is a notable lack of reliable predictive models for determining the sensitivity of this treatment. This study aimed to establish a novel classification system for CRC and develop a predictive model for XELOX chemotherapeutic sensitivity using serum metabolomics. We recruited 89 patients with CRC and 89 age- and sex-matched healthy controls for untargeted metabolomic studies to identify tumor-specific serum metabolites. The patients were grouped into distinct metabolic subtypes using unsupervised clustering. A serum metabolite combination predictive of the efficacy of XELOX was established using Cox regression analysis in 34 patients with stage III CRC. Using unsupervised clustering based on the serum metabolites, three distinct clusters were identified. Notably, Cluster 3, which was characterized by uniform lipid and amino acid levels, demonstrated the best prognosis. Our analysis revealed that D-glucose 6-phosphate, presqualene diphosphate, and leukotriene B4 levels were negatively correlated with XELOX sensitivity, whereas 15-HETE and N-acetyl-l-methionine levels were positively correlated. Based on these findings, we constructed a predictive model validated in an independent cohort of 34 patients with stage III CRC. In summary, this study identified a novel classification of CRC based on serum metabolites and developed a potential prognostic model for XELOX chemotherapeutic efficacy, which may have direct effects on the treatment and prognosis of CRC.
Collapse
Affiliation(s)
- Yijie Zhang
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Lizhen Ye
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, China
| | - Ying Qin
- Department of Gastrointestinal Surgery, Shenzhen Second People'S Hospital, Shenzhen, China
| | - Cheng Qiu
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, China
| | - Qinsheng Sun
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
| | - Tingting Fan
- Institute of Biomedical Health Technology and Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yan Chen
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China.
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, China.
| | - Yuyang Jiang
- Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen, China
- State Key Laboratory of Chemical Oncogenomics, Tsinghua Shenzhen International Graduate School, Shenzhen, China
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| |
Collapse
|
114
|
Valladares-Ayerbes M, Toledano-Fonseca M, Graña B, Jimenez-Fonseca P, Pulido-Cortijo G, Gil S, Sastre J, Salud A, Rivera F, Salgado M, García-Alfonso P, López López R, Guillén-Ponce C, Rodríguez-Ariza A, Vieitez JM, Díaz-Rubio E, Aranda E. Associations of blood RNA biomarkers and circulating tumour cells in patients with previously untreated metastatic colorectal cancer. BMC Cancer 2025; 25:743. [PMID: 40259317 PMCID: PMC12013160 DOI: 10.1186/s12885-025-14098-9] [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: 07/05/2024] [Accepted: 04/07/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND In patients with metastatic colorectal cancer, analysis of the number of basal circulating tumour cells (bCTCs) has been shown to be a strong prognostic indicator. In this study, we aim to explore the potential associations between whole blood mRNA and microRNA expression profiles and bCTC counts, tumour mutations and prognosis in untreated metastatic colorectal cancer patients. METHODS A total of 151 patients previously screened for inclusion in two clinical trials (VISNÚ1 and VISNÚ2) were enrolled in this study. Real-time quantitative PCR (qPCR) analyses were performed to determine the whole blood expression of selected RNAs (mRNAs and microRNAs) involved in the metastatic process. The CellSearch system was used to enumerate circulating tumour cells. The primary objective was to correlate RNA expression with the number of bCTCs, while the secondary objectives were to investigate the relationship between the levels of circulating RNA biomarkers in whole blood and the clinical, pathological, and molecular characteristics and prognosis of patients with metastatic colorectal cancer. RESULTS bCTC count was significantly associated with AGR2 mRNA in the entire cohort of 151 patients. AGR2, ADAR1 and LGR5 were associated with the number of bCTC, both in the subgroup with bCTC ≥ 3 and in the subgroup with native RAS/BRAF/PIK3 CA tumours. In patients with RAS/BRAF/PIK3 CA mutations no correlations with bCTC were detected, but an upregulation of miR-224-5p and the stemness marker LGR5 and a downregulation of immune regulatory CD274 were found. Lower levels of miR-106a-5p/miR-26a-5p were associated with shorter overall survival, with independent statistical significance in the multivariate analysis. CONCLUSIONS A correlation was identified between the levels of a subset of whole blood RNAs, including AGR2, ADAR1, and LGR5, and the number of bCTC and RAS/BRAF/PIK3 CA mutational status. Furthermore, another set of whole blood RNAs, specifically miR-106a-5p and miR-26a-5p, was found to be associated with poor prognosis. This may be helpful for risk stratification. TRIAL REGISTRATION Clinical Trials Gov. NCT01640405 and NCT01640444. Registered on 13 June 2012. https://clinicaltrials.gov/ .
Collapse
Affiliation(s)
- Manuel Valladares-Ayerbes
- Department of Medical Oncology, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina (IBIS), Seville, Spain.
| | - Marta Toledano-Fonseca
- Department of Medical Oncology, IMIBIC, Universidad de Córdoba, CIBERONC, Instituto de Salud Carlos III, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Begoña Graña
- Department of Medical Oncology, Instituto de Investigación Biomédica (INIBIC), Hospital Universitario de A Coruña, A Coruña, Spain
| | - Paula Jimenez-Fonseca
- Department of Medical Oncology, Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - Gema Pulido-Cortijo
- Department of Medical Oncology, IMIBIC, Universidad de Córdoba, CIBERONC, Instituto de Salud Carlos III, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Silvia Gil
- Department of Medical Oncology, Hospital Regional Universitario de Málaga, Málaga, Spain
| | - Javier Sastre
- Department of Medical Oncology, Hospital Clínico San Carlos, Instituto de Investigación (IdISSC), Universidad Complutense, Madrid, Spain
| | - Antonieta Salud
- Department of Medical Oncology, Hospital Universitario Arnau de Vilanova, Lleida, Spain
| | - Fernando Rivera
- Department of Medical Oncology, Hospital Universitario Marqués de Valdecilla, IDIVAL, Santander, Spain
| | - Mercedes Salgado
- Department of Medical Oncology, Complejo Hospitalario Universitario de Ourense, Ourense, Spain
| | - Pilar García-Alfonso
- Department of Medical Oncology, Hospital Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Universidad Complutense, Madrid, Spain
| | - Rafael López López
- Department of Medical Oncology and Translational Medical Oncology Group, Hospital Clínico Universitario, Instituto de Investigación Sanitaria de Santiago (IDIS), CIBERONC, Universidad de Santiago de Compostela, Santiago de Compostela, Spain
| | - Carmen Guillén-Ponce
- Department of Medical Oncology, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Antonio Rodríguez-Ariza
- Department of Medical Oncology, IMIBIC, Universidad de Córdoba, CIBERONC, Instituto de Salud Carlos III, Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Jose Mª Vieitez
- Department of Medical Oncology, Hospital Universitario Central de Asturias, ISPA, Oviedo, Spain
| | - Eduardo Díaz-Rubio
- Department of Medical Oncology, Hospital Clínico San Carlos, Instituto de Investigación (IdISSC), Universidad Complutense, Madrid, Spain
| | - Enrique Aranda
- Department of Medical Oncology, IMIBIC, Universidad de Córdoba, CIBERONC, Instituto de Salud Carlos III, Hospital Universitario Reina Sofía, Córdoba, Spain
| |
Collapse
|
115
|
Zhao CY, Liu F, Dong JM, Du CP, Zhang CL, Wang CY, Zhang XY, Zhou Q, Liu W, Yang AJ, Zhou YN, Dang Y, Shang LN, Wang M, Li M. SDCBP Orchestrated Gastric Cancer Aggression Through Epithelial- Mesenchymal Transition and Macrophages M2 Polarization. Mol Carcinog 2025. [PMID: 40256939 DOI: 10.1002/mc.23923] [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: 12/02/2024] [Revised: 03/13/2025] [Accepted: 03/24/2025] [Indexed: 04/22/2025]
Abstract
Gastric cancer remains a significant global health burden with limited treatment options and high mortality. Syndecan-binding protein (SDCBP), a scaffolding protein involved in tumor differentiation, has attracted attention as a potential therapeutic target in cancers. However, its precise role in gastric cancer progression is not fully understood. In this study, through bioinformatics analysis and gastric cancer samples detection, we discovered that SDCBP was highly expressed in gastric cancer tissues, which was correlated with clinicopathological features such as tumor invasion depth and distant metastasis, and exhibited heterogeneity across histological or molecular subtypes. Elevated SDCBP expression promoted the proliferation, invasion and migration of gastric cancer cells, and modulated epithelial-mesenchymal transition (EMT) via the ERK signaling pathway. Xenograft experiments in mice confirmed that inhibiting SDCBP or ERK signaling could delay cancer progression. We also found that gastric cancer cells with SDCBP knockdown were able to inhibit the M2 polarization of cocultured macrophages, reduce chemotaxis and enhance phagocytosis of macrophages. Therefore, SDCBP plays a crucial role in driving gastric cancer progression. Targeting SDCBP in gastric cancer can partially reverse the malignant phenotype, and SDCBP is expected to be a promising therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Chan-Yuan Zhao
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Feng Liu
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
| | - Jia-Ming Dong
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Cun-Pu Du
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Chen-Li Zhang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
| | - Chen-Yu Wang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
| | - Xiao-Yu Zhang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Quan Zhou
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Wei Liu
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
| | - Ai-Jun Yang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
| | - Yong-Ning Zhou
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yun Dang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Gansu Provincial Maternity and Child-care Hospital/Gansu Provincial Central Hospital, Lanzhou, China
| | - Li-Na Shang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Medical College of Northwest Minzu University, Lanzhou, China
| | - Min Wang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
- Experimental Teaching Center of Basic Medicine, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Min Li
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, China
| |
Collapse
|
116
|
Ghogare SS, Pathan EK. Intratumor fungi specific mechanisms to influence cell death pathways and trigger tumor cell apoptosis. Cell Death Discov 2025; 11:188. [PMID: 40258837 PMCID: PMC12012188 DOI: 10.1038/s41420-025-02483-z] [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: 12/26/2024] [Revised: 03/29/2025] [Accepted: 04/07/2025] [Indexed: 04/23/2025] Open
Abstract
Cancer, uncontrolled cell growth due to the loss of cell cycle regulation, is often found to be associated with viral infections and, as recent studies show, with bacterial infections as well. Emerging reports also suggest a strong link between fungi and cancer. The crucial virulence trait of fungi, the switch from yeast (Y) to hyphal (H) form, is found to be associated with carcinogenesis. The physicochemical properties and signal transduction pathways involved in the switch to the hyphal form overlap with those of tumor cell formation. Inhibiting differentiation causes apoptosis in fungi, whereas preventing apoptosis leads to cancer in multicellular organisms. Literature on the fungi-cancer linkage, though limited, is increasing rapidly. This review examines cancer-specific fungal communities, the impact of fungal microbiome on cancer cell progression, similarities between fungal differentiation and cells turning cancerous at biochemical and molecular levels, including the overlaps in signal transduction pathways between fungi and cancer. Based on the available evidence, we suggest that molecules inhibiting the yeast-hyphal transition in fungi can be combined with those targeting tumor cell apoptosis for effective cancer treatment. The review points out fertile research areas where mycologists and cancer researchers can collaborate to unravel common molecular mechanisms. Moreover, antibodies targeting fungal-specific chitin and glucan can be used for the selective neutralization of tumor cells. These new combinations of potential therapies are expected to facilitate the development of target-specific, less harmful and commercially feasible anticancer therapies. We bring together available evidence to argue that fungal infections could either trigger cancer or have a significant role in the development and progression of cancer. Hence, cancer-associated fungal populations could be utilized as a target for a combination therapy involving the integration of anticancer and antifungal drugs as well as inhibitors of fungal morphogenesis to develop more effective anticancer therapies.
Collapse
Affiliation(s)
- Simran S Ghogare
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) Lavale, Pune, 412115, Maharashtra, India
| | - Ejaj K Pathan
- Symbiosis School of Biological Sciences, Symbiosis International (Deemed University) Lavale, Pune, 412115, Maharashtra, India.
| |
Collapse
|
117
|
Cai S, Mi S, Chen J, Shao L, Yang X, Xue M. METTL1-mediated m7G modification promotes colorectal cancer metastasis via stabilization of ICAM-1. Mol Cell Biochem 2025:10.1007/s11010-025-05293-0. [PMID: 40252158 DOI: 10.1007/s11010-025-05293-0] [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: 12/04/2024] [Accepted: 04/13/2025] [Indexed: 04/21/2025]
Abstract
Colorectal cancer (CRC) is one of the most common tumors worldwide, and metastatic CRC is likely to have a poor prognosis. N7-methylguanosine (m7G) is a common methylation modification that is catalyzed primarily by methyltransferase 1 (METTL1). However, the role of m7G in metastatic CRC remains unclear. The role of METTL1 in progressive CRC was initially explored using bioinformatics analysis. Subsequently, its relationship with CRC was further validated through in vitro and in vivo experiments. Potential downstream targets were identified through RNA-seq and quantitative real-time PCR (RT‒qPCR), and the underlying mechanisms were investigated using methylated RNA immunoprecipitation (MeRIP) and RNA degradation assays. Our results revealed that METTL1 is differentially expressed and significantly upregulated in metastatic CRC. This correlation was further confirmed by in vivo and in vitro experiments. RNA sequencing of CRC cells with METTL1 knockdown revealed that intercellular adhesion molecule-1 (ICAM-1) was a significant downstream target and could be stabilized by m7G modification. We revealed that METTL1 is significantly upregulated in metastatic CRC and plays a critical role in CRC progression by stabilizing ICAM-1 through m7G modification.
Collapse
Affiliation(s)
- Shangwen Cai
- Department of Gastroenterology, the Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, 310009, Zhejiang, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Shuyi Mi
- Department of Gastroenterology, the Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, 310009, Zhejiang, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Jingyu Chen
- Department of Gastroenterology, the Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, 310009, Zhejiang, China
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China
| | - Liming Shao
- Department of Gastroenterology, the Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, 310009, Zhejiang, China
| | - Xiaoyun Yang
- Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Meng Xue
- Department of Gastroenterology, the Second Affiliated Hospital of Zhejiang University School of Medicine, No.88 Jiefang Road, Shangcheng District, Hangzhou, 310009, Zhejiang, China.
- Institute of Gastroenterology, Zhejiang University, Hangzhou, China.
| |
Collapse
|
118
|
Bao Z, Yi B. Analysis of preoperative serum cytokine levels in patients with oral squamous cell carcinoma. Sci Rep 2025; 15:13537. [PMID: 40253468 PMCID: PMC12009433 DOI: 10.1038/s41598-025-89816-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 02/07/2025] [Indexed: 04/21/2025] Open
Abstract
This study investigates preoperative serum cytokine levels in patients with oral squamous cell carcinoma (OSCC). The study included 51 patients with OSCC and 42 healthy controls (HCs). Serum samples of 12 cytokines were analyzed using a multiplex bead-based flow cytometry immunoassay. Mann-Whitney U test and binary logistic regression analysis were performed to identify significant indicators of OSCC. Receiver operating characteristic (ROC) curves evaluated the diagnostic performance.Spearman analysis was assessed the correlation between cytokines and tumor-node-metastasis staging of OSCC. Serum levels of interleukin (IL)-2, IL-5, IL-6, IL-8, IL-12P70, IL-17, and interferon gamma (IFN-γ) were significantly higher (P < 0.05) in patients with OSCC than in HCs. IL-5, IL-6, IL-8, IL-12P70, IL-17, IFN-γ, combination 1 (IL-6 and IL-8), and combination 2 (IL-6 and IL-12P70) had area under the curve (AUC) values > 0.7, with combination 2 exhibited the highest AUC of 0.995. Serum cytokine profiles were significantly different (P < 0.05) between the patients with OSCC and HCs. IL-5, IL-6, IL-8, IL-12P70, IL-17, IFN-γ, combination 1, and combination 2 effectively distinguished between HCs and patients with OSCC. Cytokine combinations enhanced OSCC diagnostic accuracy, with significantly elevated IL-6 levels (P < 0.05) in advanced-stage compared to early-stage OSCC, indicating its potential impact on disease progression and prognosis.
Collapse
Affiliation(s)
- Zhenying Bao
- Department of Clinical Laboratory Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Author's, No.22 ,Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China
| | - Biao Yi
- Department of Clinical Laboratory Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices & Beijing Key Laboratory of Digital Stomatology & NHC Key Laboratory of Digital Stomatology & NMPA Key Laboratory for Dental Materials Author's, No.22 ,Zhongguancun South Avenue, Haidian District, Beijing, 100081, PR China.
| |
Collapse
|
119
|
Han Z, Wen L. G-quadruplex in cancer energy metabolism: A potential therapeutic target. Biochim Biophys Acta Gen Subj 2025; 1869:130810. [PMID: 40254103 DOI: 10.1016/j.bbagen.2025.130810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 04/07/2025] [Accepted: 04/16/2025] [Indexed: 04/22/2025]
Abstract
In recent years, energy metabolism in cancer has received increasing attention as an important component of tumor biology, and the functions of transcription factors, mitochondria, reactive oxygen species (ROS) and the autophagy-lysosome system in which have been elucidated. G-quadruplex (G4) is a molecular switch that regulates gene transcription or translation. As an anticancer target, the effect of G4 on cancer cell proliferation, apoptosis, cycle and autophagy has been recognized. The energy metabolism system is a unified whole composed of transcription factors, metabolic regulators, metabolites and signaling pathways that run through the entire cancer process. However, the role of G4 in this complex metabolic network has not been systematically elucidated. In this review, we analyze the close correlation between G4 and transcription factors, mitochondria, ROS and the autophagy-lysosome system and suggest that G4 can exert a marked effect on cancer energy metabolism by regulating the above mentioned key regulatory elements. The anticancer effects of some G4 ligands through regulation of energy metabolism have also been summarized, confirming the clear involvement of G4 in energy metabolism. Although much more research is needed, we propose that G4 may play a critical role in the complex energy metabolism system of cancer, which is a promising target for anticancer strategies focusing on energy metabolism.
Collapse
Affiliation(s)
- Zongqiang Han
- Department of Laboratory Medicine, Beijing Xiaotangshan Hospital, Beijing 102211, China
| | - Lina Wen
- Department of Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China.
| |
Collapse
|
120
|
Ma Y, Zhou Y, Xie G, Chen H, Huangfu Y, Shen L, Liu Y, Wang P. HEX-1 reduces colitis-driven colorectal cancer via inactivating the prolyl isomerase PIN1 sensitization and remodeling the gut microbiota. Discov Oncol 2025; 16:565. [PMID: 40251462 PMCID: PMC12008109 DOI: 10.1007/s12672-025-02338-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/08/2025] [Indexed: 04/20/2025] Open
Abstract
Metabolic reprogramming, a pivotal hallmark of cancer, plays a crucial role in both the initiation and progression of colorectal cancer (CRC). Despite the vast unknowns surrounding the identity and biological activities of most natural metabolites in diseases, our study, utilizing native metabolomics results through GC-MS/MS, identified a small molecule, 4,4-Dimethyl-2-cyclohexen-1-one, named HEX-1 in the serum of CRC patients. We have further explored and assessed its biological activities. HEX-1 suppressed the proliferation of cancer cells and tumorigenesis via the inactivation and sensitization of PIN1. Notably, HEX-1 exhibits similar functional effects as all-trans retinoic acid (atRA) but stands out by not inducing the degradation of PIN1 mRNA or protein expression, unlike biological compounds associated with atRA. HEX-1 demonstrated the ability to induce G1/S arrest in vitro and ameliorate the progression of inflammatory CRC in mice by remodeling the gut microbiota. As volatile organic compounds (VOCs), HEX-1 could be detected feasibly. Its unique ability to penetrate whole cell populations positions it as a promising approach for cancer therapy and as an enhancer for chemotherapy and immunotherapy. The findings suggest that HEX-1 holds the potential as a valuable addition to the armamentarium against CRC.
Collapse
Affiliation(s)
- Yanhui Ma
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yunlan Zhou
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Guohua Xie
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Hui Chen
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Yuchan Huangfu
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
| | - Lisong Shen
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, 200092, China
| | - Yi Liu
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Faculty of Medical Laboratory Science, College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, 200092, China.
| | - Ping Wang
- Department of Laboratory Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, China.
- Institute of Artificial Intelligence Medicine, Shanghai Academy of Experimental Medicine, Shanghai, 200092, China.
| |
Collapse
|
121
|
Singh M, Arora HL, Naik R, Joshi S, Sonawane K, Sharma NK, Sinha BK. Ferroptosis in Cancer: Mechanism and Therapeutic Potential. Int J Mol Sci 2025; 26:3852. [PMID: 40332483 PMCID: PMC12028135 DOI: 10.3390/ijms26083852] [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: 02/11/2025] [Revised: 04/11/2025] [Accepted: 04/15/2025] [Indexed: 05/08/2025] Open
Abstract
Cancer drug resistance occurs when cancer cells evade cell death following treatment with chemotherapy, radiation therapy, and targeted therapies. This resistance is often linked to the reprogramming of programmed cell death (PCD) pathways, allowing cancer cells to survive drug-induced stress. However, certain anticancer therapies, when combined with specific agents or inhibitors, can induce ferroptosis-a form of cell death driven by iron-dependent lipid peroxidation. Currently, extensive preclinical and clinical research is underway to investigate the molecular, cellular, and tissue-specific mechanisms underlying ferroptosis, with the goal of identifying strategies to overcome drug resistance in cancers unresponsive to conventional PCD pathways. By harnessing ferroptosis, cancer cells can be compelled to undergo lipid peroxidation-induced death, potentially improving therapeutic outcomes in patients with cancer. This short review aims to enhance the understanding of ferroptosis inducers in cancer therapy and stimulate further research into ferroptosis-based approaches for more effective clinical cancer treatment.
Collapse
Affiliation(s)
- Mansaa Singh
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Hasmiq L. Arora
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Rutuja Naik
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Shravani Joshi
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Kaveri Sonawane
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Nilesh Kumar Sharma
- Cancer and Translational Research Lab, Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, India; (M.S.); (H.L.A.); (R.N.); (S.J.); (K.S.)
| | - Birandra K. Sinha
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
| |
Collapse
|
122
|
Pang B, Wu Y, An S, Chang Y, Yan H, Lin H, Zhao Z, Wu F, Chang Q, Jia W, Jiang T, Wang Y, Chai R. Ara-C suppresses H3 K27-altered spinal cord diffuse midline glioma growth and enhances immune checkpoint blockade sensitivity. SCIENCE ADVANCES 2025; 11:eadu3956. [PMID: 40238864 PMCID: PMC12002095 DOI: 10.1126/sciadv.adu3956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/10/2025] [Indexed: 04/18/2025]
Abstract
H3 K27-altered spinal cord diffuse midline glioma (H3-SCDMG) poses therapeutic challenges. Analysis of 73 clinical samples revealed heightened proliferation in H3-SCDMG versus wild-type tumors, suggesting therapeutic vulnerabilities. Drug screening identified cytarabine (Ara-C) as highly effective in inhibiting proliferation in H3 K27M cell models, recently established patient-derived cells, and patient-derived xenograft models. Mechanistically, Ara-C can suppress tumor growth through DNA damage, cell-cycle arrest, and apoptosis. An investigator-initiated clinical trial involving four patients showed benefits in three cases. In addition, a subset of cells exhibited senescence and senescence-associated secretory phenotype post-Ara-C treatment, accompanied by several immune checkpoint ligands' up-regulation and more immune cell infiltration. Combining Ara-C with dual Programmed cell death protein 1 (PD-1) and TIGIT blockade emerged as a promising strategy to disrupt immune evasion by senescent cells, enhancing antitumor responses. These findings highlight Ara-C's potential as a monotherapy and in synergy with immunotherapy for H3-SCDMG, offering potential strategies for clinical management.
Collapse
Affiliation(s)
- Bo Pang
- Department of Molecular Neuropathology, Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yilin Wu
- Department of Molecular Neuropathology, Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - SongYuan An
- Department of Molecular Neuropathology, Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yuzhou Chang
- Department of Molecular Neuropathology, Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hao Yan
- Department of Molecular Neuropathology, Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Han Lin
- Department of Molecular Neuropathology, Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zheng Zhao
- Department of Molecular Neuropathology, Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Research Unit of Accurate Diagnosis, Treatment, and Translational Medicine of Brain Tumors, Chinese Academy of Medical Sciences, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Fan Wu
- Department of Molecular Neuropathology, Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Research Unit of Accurate Diagnosis, Treatment, and Translational Medicine of Brain Tumors, Chinese Academy of Medical Sciences, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Qing Chang
- Department of Molecular Neuropathology, Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Wenqing Jia
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tao Jiang
- Department of Molecular Neuropathology, Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Research Unit of Accurate Diagnosis, Treatment, and Translational Medicine of Brain Tumors, Chinese Academy of Medical Sciences, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Yongzhi Wang
- Department of Molecular Neuropathology, Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Research Unit of Accurate Diagnosis, Treatment, and Translational Medicine of Brain Tumors, Chinese Academy of Medical Sciences, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| | - Ruichao Chai
- Department of Molecular Neuropathology, Department of Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
- Research Unit of Accurate Diagnosis, Treatment, and Translational Medicine of Brain Tumors, Chinese Academy of Medical Sciences, Beijing, China
- Chinese Glioma Genome Atlas Network (CGGA) and Asian Glioma Genome Atlas Network (AGGA), Beijing, China
| |
Collapse
|
123
|
Luo Z, Wang Y, Zeng S, Yu L, Zhao Y, Wang H, Fan Y, Zhang Y, Wang L, Li Y, Niu Z, Zhang X, Zhang Y. Harnessing lysosomal genetics: development of a risk stratification panel and unveiling of DPP7 as a biomarker for colon adenocarcinoma. J Genet Genomics 2025:S1673-8527(25)00118-3. [PMID: 40254156 DOI: 10.1016/j.jgg.2025.04.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 04/08/2025] [Accepted: 04/12/2025] [Indexed: 04/22/2025]
Abstract
Lysosomal dysfunction has been implicated in the progression of colon adenocarcinoma (COAD), yet the prognostic significance and therapeutic potential of lysosome-related genes (LRGs) remain underexplored. In this study, we construct a 6-LRG-based prognostic risk stratification model (DPP7, ADAM8, CD1B, LRP2, ATP6V1C2, and PLAAT3) by integrating LASSO and Cox regression analyses. Stratifying patients based on median risk scores, we demonstrate that high-risk patients exhibit significantly worse clinical outcomes across the TCGA cohort and five independent GEO datasets. Furthermore, this panel outperforms 136 previously published models in terms of predictive accuracy for 1-, 3-, and 5-year survival rates. Validation multiplex immunofluorescence using an in-house tissue microarray cohort confirms the 6-LRG signature serves as an independent prognostic factor. Additionally, high-risk patients exhibit distinct immunosuppressive tumor microenvironment and aggressive malignancy characteristics. Functional depletion of DPP7 significantly inhibits tumor cell proliferation, migration, and metastasis in both in vitro and in vivo settings. Moreover, DPP7 silencing attenuates epithelial-mesenchymal transition, as evidenced by the upregulation of E-cadherin and downregulation of N-cadherin, Vimentin, and Snail. In conclusion, this study establishes an LRG-based model for COAD prognostic prediction and nominates DPP7 as a promising therapeutic target for COAD treatment.
Collapse
Affiliation(s)
- Zhengdong Luo
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China
| | - Yanlei Wang
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China
| | - Shunjie Zeng
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China
| | - Longchen Yu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China
| | - Yuxiao Zhao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China
| | - Hong Wang
- Department of Anesthesiology, Yidu Central Hospital, Weifang Medical University, Qingzhou, Shandong 262500, China
| | - Yingjing Fan
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China
| | - Yanli Zhang
- Department of Clinical Laboratory, Shandong Provincial Third Hospital, Jinan, Shandong 250000, China
| | - Lili Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China
| | - Yaping Li
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China
| | - Zhongfang Niu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China
| | - Xin Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China.
| | - Yi Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, Shandong 250012, China; Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan, Shandong 250012, China.
| |
Collapse
|
124
|
Chen M, Bie L. Intratumoral microbiota for hepatocellular carcinoma: from preclinical mechanisms to clinical cancer treatment. Cancer Cell Int 2025; 25:152. [PMID: 40247312 PMCID: PMC12007317 DOI: 10.1186/s12935-025-03745-7] [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: 12/30/2024] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
Intratumoral microbiota has been found to be a crucial component of hepatocellular carcinoma (HCC). Due to insufficient recognition, technical limitations, and low biomass of intratumoral microbiota, it is poorly understood. Intratumoral microbiota exhibit significant diversity in HCC tissues. It is involved in the development of HCC through several mechanisms, such as remodeling the immunosuppressive microenvironment, metabolic reprogramming, and genetic alterations. Moreover, intratumoral microbiota is associated with the metastasis of HCC cells. Herein, we reviewed the history of intratumoral microbiota, applied biotechnology to depict the signatures of intratumoral microbiota, investigated the potential sources of intratumoral microbiota, and assessed their functions, mechanisms, and heterogeneity. Furthermore, in this review, we summarized the development of therapeutics that can be used in the treatment of HCC and proposed future perspectives for research in this field.
Collapse
Affiliation(s)
- Muhua Chen
- Department of Hepato-Pancreato-Biliary & Gastric Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| | - Lei Bie
- Department of Thoracic Surgery, Wuhan No.1 Hospital, Wuhan, 430030, Hubei, China
| |
Collapse
|
125
|
Göbel C, Niccolai R, de Groot MHP, Jayachandran J, Traets J, Kloosterman DJ, Gregoricchio S, Morris B, Kreft M, Song JY, Azarang L, Kasa E, Oskam N, de Groot D, Hoekman L, Bleijerveld OB, Kersten MJ, Aslam MA, van Leeuwen F, Jacobs H. Targeting DOT1L and EZH2 synergizes in breaking the germinal center identity of diffuse large B-cell lymphoma. Blood 2025; 145:1802-1813. [PMID: 39792929 DOI: 10.1182/blood.2024025500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/27/2024] [Accepted: 10/13/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT Differentiation of antigen-activated B cells into proproliferative germinal center (GC) B cells depends on the activity of the transcription factors myelocytoma (MYC) and B-cell lymphoma 6 (BCL6), and the epigenetic writers disruptor of telomeric silencing 1-like (DOT1L) and enhancer of zeste homolog 2 (EZH2). GCB-like diffuse large B-cell lymphomas (GCB-DLBCLs) arise from GCB cells and closely resemble their cell of origin. Given the dependency of GCB cells on DOT1L and EZH2, we investigated the role of these epigenetic regulators in GCB-DLBCLs and observed that GCB-DLBCLs synergistically depend on the combined activity of DOT1L and EZH2. Mechanistically, inhibiting both enzymes led to enhanced derepression of polycomb repressive complex 2 target genes compared with EZH2 single treatment, along with the upregulation of BCL6 target genes and suppression of MYC target genes. The sum of all these alterations results in a "cell identity crisis," wherein GCB-DLBCLs lose their proproliferative GC identity and partially undergo plasma cell differentiation, a state associated with poor survival. In support of this model, combined epidrugging of DOT1L and EZH2 prohibited the outgrowth of human GCB-DLBCL xenografts in vivo. We conclude that the malignant behavior of GCB-DLBCLs strongly depends on DOT1L and EZH2 and that combined targeting of both epigenetic writers may provide an alternative differentiation-based treatment modality for GCB-DLBCL.
Collapse
MESH Headings
- Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors
- Enhancer of Zeste Homolog 2 Protein/genetics
- Enhancer of Zeste Homolog 2 Protein/metabolism
- Humans
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Germinal Center/pathology
- Germinal Center/metabolism
- Germinal Center/drug effects
- Animals
- Mice
- Histone-Lysine N-Methyltransferase/antagonists & inhibitors
- Cell Line, Tumor
- Gene Expression Regulation, Neoplastic/drug effects
- Cell Differentiation
- Epigenesis, Genetic
Collapse
Affiliation(s)
- Camiel Göbel
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Rachele Niccolai
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marnix H P de Groot
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jayashree Jayachandran
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Joleen Traets
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Daan J Kloosterman
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Ben Morris
- Robotics and Screening Center, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Maaike Kreft
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Ji-Ying Song
- Division of Experimental Animal Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Leyla Azarang
- Biostatistics Center, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Eirini Kasa
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Nienke Oskam
- Immunopathology, Sanquin Research, Amsterdam, The Netherlands
| | - Daniel de Groot
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Liesbeth Hoekman
- Mass Spectrometry/Proteomics Facility, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Onno B Bleijerveld
- Mass Spectrometry/Proteomics Facility, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Marie José Kersten
- Department of Hematology, Amsterdam University Medical Center (location University of Amsterdam), Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Muhammad A Aslam
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan, Pakistan
| | - Fred van Leeuwen
- Division of Gene Regulation, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Department of Medical Biology, Amsterdam University Medical Center (location University of Amsterdam), Amsterdam, The Netherlands
| | - Heinz Jacobs
- Division of Tumor Biology and Immunology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
126
|
Pfeiffer A, Di Leo L, Bechmann MB, Nawabi M, Ambjørner S, Ardeshir-Larijani D, Colstrup LT, Borchert SV, Saaby L, Brodin B, Gajhede M, Lund XL, Čečková M, Brünner N, Stenvang J. Inhibition of ABCG2 by SCO-101 Enhances Chemotherapy Efficacy in Cancer. Int J Mol Sci 2025; 26:3790. [PMID: 40332396 PMCID: PMC12027554 DOI: 10.3390/ijms26083790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/03/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
Chemotherapy resistance, particularly multidrug resistance (MDR), remains a significant barrier to effective cancer treatment, leading to high mortality rates. The development of novel therapeutic strategies targeting key molecular mechanisms to counteract drug resistance is thus an urgent clinical need. In this study, we evaluated the potential of the small molecule SCO-101 to restore chemotherapy sensitivity in drug-resistant cancer cells. Using in silico and in vitro models such as molecular docking, cell viability, colony formation, dye efflux, transporter assays and chemotherapy retention, we assessed the impact of SCO-101 on drug retention and response in several drug-resistant cancer cells. SCO-101 was found to inhibit the activity of breast cancer resistance protein (BCRP/ABCG2) and UDP Glucuronosyltransferase Family 1 Member A1 (UGT1A1), two key proteins involved in drug resistance by cellular drug excretion and drug metabolism. Our results demonstrate that inhibition of these proteins by SCO-101 leads to increased intracellular drug accumulation, enhancing the cytotoxic effects of chemotherapy agents. Additionally, we identified a strong correlation between high ABCG2 expression and MDR in non-drug-resistant models, where cells exhibiting elevated ABCG2 levels displayed chemotherapy resistance, which was effectively reversed by SCO-101 co-treatment. These findings highlight the therapeutic potential of SCO-101 in overcoming MDR by inhibiting drug efflux mechanisms and metabolism, thereby enhancing chemotherapy efficacy. SCO-101 is currently undergoing clinical trials as an orally administered drug and is considered a promising strategy for improving cancer treatment outcomes in patients with drug-resistant tumors.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily G, Member 2/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/chemistry
- Humans
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/metabolism
- Neoplasm Proteins/chemistry
- Neoplasm Proteins/genetics
- Drug Resistance, Neoplasm/drug effects
- Antineoplastic Agents/pharmacology
- Molecular Docking Simulation
- Cell Line, Tumor
- Neoplasms/drug therapy
- Neoplasms/metabolism
- Drug Resistance, Multiple/drug effects
- Cell Survival/drug effects
- Heterocyclic Compounds, 4 or More Rings
- Diketopiperazines
Collapse
Affiliation(s)
- Anamarija Pfeiffer
- Biognosys AG, 8952 Schlieren, Switzerland;
- Scandion Oncology A/S, Symbion, 2100 Copenhagen, Denmark; (L.D.L.); (M.B.B.); (D.A.-L.); (L.T.C.); (N.B.)
| | - Luca Di Leo
- Scandion Oncology A/S, Symbion, 2100 Copenhagen, Denmark; (L.D.L.); (M.B.B.); (D.A.-L.); (L.T.C.); (N.B.)
| | - Marc Baker Bechmann
- Scandion Oncology A/S, Symbion, 2100 Copenhagen, Denmark; (L.D.L.); (M.B.B.); (D.A.-L.); (L.T.C.); (N.B.)
| | - Mubeen Nawabi
- Genmab A/S, Carl Jacobsens Vej 30, 2500 Valby, Denmark;
| | - Sophie Ambjørner
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark (M.G.); (X.L.L.)
| | - Diba Ardeshir-Larijani
- Scandion Oncology A/S, Symbion, 2100 Copenhagen, Denmark; (L.D.L.); (M.B.B.); (D.A.-L.); (L.T.C.); (N.B.)
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark (M.G.); (X.L.L.)
| | - Louise Thybo Colstrup
- Scandion Oncology A/S, Symbion, 2100 Copenhagen, Denmark; (L.D.L.); (M.B.B.); (D.A.-L.); (L.T.C.); (N.B.)
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark (M.G.); (X.L.L.)
| | - Signe Vedel Borchert
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark;
| | - Lasse Saaby
- Bioneer A/S, Kogle Alle 2, 2970 Hørsholm, Denmark;
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark;
| | - Birger Brodin
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark;
| | - Michael Gajhede
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark (M.G.); (X.L.L.)
| | - Xamuel Loft Lund
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark (M.G.); (X.L.L.)
- Institut Laue–Langevin, 71 Avenue de Martyrs, 38042 Grenoble, France
| | - Martina Čečková
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Akademika Heyrovskeho 1203, 500 05 Hradec Kralove, Czech Republic;
| | - Nils Brünner
- Scandion Oncology A/S, Symbion, 2100 Copenhagen, Denmark; (L.D.L.); (M.B.B.); (D.A.-L.); (L.T.C.); (N.B.)
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark (M.G.); (X.L.L.)
| | - Jan Stenvang
- Scandion Oncology A/S, Symbion, 2100 Copenhagen, Denmark; (L.D.L.); (M.B.B.); (D.A.-L.); (L.T.C.); (N.B.)
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark (M.G.); (X.L.L.)
| |
Collapse
|
127
|
Leoni I, Galvani G, Monti E, Vianello C, Valenti F, Pincigher L, Grolla AA, Moro M, Coada CA, Perrone A, Righi V, Marinelli S, Ravegnini G, Giovannini C, Baldassarre M, Pariali M, Ravaioli M, Cescon M, Vasuri F, Domenicali M, Negrini M, Piscaglia F, Fato R, Stefanelli C, Gramantieri L, Bergamini C, Fornari F. MiR-22/GLUT1 Axis Induces Metabolic Reprogramming and Sorafenib Resistance in Hepatocellular Carcinoma. Int J Mol Sci 2025; 26:3808. [PMID: 40332478 PMCID: PMC12027541 DOI: 10.3390/ijms26083808] [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: 03/08/2025] [Revised: 04/01/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
The approval of immunotherapy has revolutionized the management of hepatocellular carcinoma (HCC) patients. However, sorafenib remains a first-line therapeutic option for advanced patients and, in particular, for patients not eligible for immune checkpoint inhibitors, but its efficacy is limited by the onset of acquired resistance, highlighting the urgent need for predictive biomarkers. This study investigates the role of miR-22 in metabolic reprogramming and its potential as a biomarker in HCC. The analysis of miR-22 expression was performed in HCC patients and preclinical models by qPCR. Functional analyses in HCC cells evaluated GLUT1 as a direct miR-22 target. Cellular and metabolic assays evaluated the miR-22/GLUT1 axis's role in metabolic changes, tumor aggressiveness, and sorafenib response. Circulating miR-22 was analyzed in sorafenib-treated HCC patients and rats. MiR-22 was downregulated in HCCs and associated with aggressive tumor features. Functionally, miR-22 modulated the HIF1A pathway, enhanced survival in stressful conditions, promoted a glycolytic shift, and enhanced cancer cell plasticity and sorafenib resistance via GLUT1 targeting. In addition, high serum miR-22 levels were associated with sorafenib resistance in HCC patients and rats. GLUT1 inhibition sensitized low miR-22-expressing HCC cells to sorafenib in preclinical models. These findings suggest that circulating miR-22 deserves attention as a predictive biomarker of sorafenib response. GLUT1 inhibition may represent a therapeutic strategy to combine with sorafenib, particularly in patients exhibiting high circulating miR-22 levels.
Collapse
Affiliation(s)
- Ilaria Leoni
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy; (I.L.); (G.G.); (E.M.); (C.V.); (V.R.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Giuseppe Galvani
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy; (I.L.); (G.G.); (E.M.); (C.V.); (V.R.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Elisa Monti
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy; (I.L.); (G.G.); (E.M.); (C.V.); (V.R.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Clara Vianello
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy; (I.L.); (G.G.); (E.M.); (C.V.); (V.R.); (C.S.)
- Centre for Applied Biomedical Research—CRBA, University of Bologna, 40138 Bologna, Italy
| | - Francesca Valenti
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.V.); (L.P.); (G.R.); (R.F.)
| | - Luca Pincigher
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.V.); (L.P.); (G.R.); (R.F.)
| | - Ambra A. Grolla
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (A.A.G.); (M.M.)
| | - Marianna Moro
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale, 28100 Novara, Italy; (A.A.G.); (M.M.)
| | - Camelia A. Coada
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (C.A.C.); (A.P.); (C.G.); (M.R.); (M.C.); (F.V.); (M.D.); (F.P.)
| | - Alessandro Perrone
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (C.A.C.); (A.P.); (C.G.); (M.R.); (M.C.); (F.V.); (M.D.); (F.P.)
| | - Valeria Righi
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy; (I.L.); (G.G.); (E.M.); (C.V.); (V.R.); (C.S.)
| | - Sara Marinelli
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.M.); (L.G.)
| | - Gloria Ravegnini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.V.); (L.P.); (G.R.); (R.F.)
| | - Catia Giovannini
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (C.A.C.); (A.P.); (C.G.); (M.R.); (M.C.); (F.V.); (M.D.); (F.P.)
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.M.); (L.G.)
| | - Maurizio Baldassarre
- Unit of Semiotics, Liver and Alcohol-Related Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Milena Pariali
- U.O. Genetica Medica, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Matteo Ravaioli
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (C.A.C.); (A.P.); (C.G.); (M.R.); (M.C.); (F.V.); (M.D.); (F.P.)
- Hepato-Biliary Surgery and Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Matteo Cescon
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (C.A.C.); (A.P.); (C.G.); (M.R.); (M.C.); (F.V.); (M.D.); (F.P.)
- Hepato-Biliary Surgery and Transplant Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesco Vasuri
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (C.A.C.); (A.P.); (C.G.); (M.R.); (M.C.); (F.V.); (M.D.); (F.P.)
- Pathology Unit, Santa Maria delle Croci Hospital, 40121 Ravenna, Italy
| | - Marco Domenicali
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (C.A.C.); (A.P.); (C.G.); (M.R.); (M.C.); (F.V.); (M.D.); (F.P.)
| | - Massimo Negrini
- Department of Translational Medicine, University of Ferrara, 44100 Ferrara, Italy;
| | - Fabio Piscaglia
- Department of Medical and Surgical Sciences, Bologna University, 40138 Bologna, Italy; (C.A.C.); (A.P.); (C.G.); (M.R.); (M.C.); (F.V.); (M.D.); (F.P.)
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.M.); (L.G.)
| | - Romana Fato
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.V.); (L.P.); (G.R.); (R.F.)
| | - Claudio Stefanelli
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy; (I.L.); (G.G.); (E.M.); (C.V.); (V.R.); (C.S.)
| | - Laura Gramantieri
- Division of Internal Medicine, Hepatobiliary and Immunoallergic Diseases, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (S.M.); (L.G.)
| | - Christian Bergamini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy; (F.V.); (L.P.); (G.R.); (R.F.)
| | - Francesca Fornari
- Department for Life Quality Studies, University of Bologna, 47921 Rimini, Italy; (I.L.); (G.G.); (E.M.); (C.V.); (V.R.); (C.S.)
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| |
Collapse
|
128
|
Cheikh IA, Hayar B, Ghanem N, Saleh LA, El-Baba C, Al-Hadeethi S, El-Khoury R, Usta J, Darwiche N. Therapeutic Targeting of the Pentose Phosphate Pathway in Colorectal Cancer Using 6-Aminonicotinamide and 5-Fluorouracil. Mol Carcinog 2025. [PMID: 40235275 DOI: 10.1002/mc.23920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 03/25/2025] [Indexed: 04/17/2025]
Abstract
Colorectal cancer (CRC) is a significant global health concern with rising incidence and mortality rates. 5-Fluorouracil (5-FU) is the standard chemotherapy for CRC but is often constrained by resistance and toxicity, highlighting the need for more efficient treatments. The pentose phosphate pathway (PPP), a glucose metabolic shunt, is significantly upregulated in CRC to support nucleotide synthesis and redox balance. Therefore, we hypothesized that targeting the PPP decreases CRC cell growth, reduces tumor progression, and improves 5-FU therapy. Consequently, we investigated the anti-tumor activities, cell death mechanism, and mode of action of the PPP inhibitor, 6-aminonicotinamide (6-AN), and 5-FU alone or in combination against CRC. We used human CRC cell lines with different p53 and 5-FU resistance statuses and a CRC xenograft model. Our findings show that 6-AN reduced the viability of human CRC cells independently of their p53 and 5-FU resistance profile, with its effect further enhanced in combination with 5-FU. The 6-AN/5-FU combination treatment synergized by reducing the total dehydrogenase activity of the PPP, inducing oxidative stress, and promoting senescence in CRC cells. Furthermore, 6-AN treatment significantly decreased tumor growth in a CRC xenograft mouse model. However, combining 6-AN with 5-FU did not reduce tumor volume significantly, highlighting the complexities of translating in vitro findings to animal models. These results suggest that interfering with the PPP activity suppresses CRC cell growth and may reduce 5-FU resistance. This study underscores targeting cancer metabolism as a novel therapeutic strategy to minimize drug resistance and to improve CRC therapeutic outcomes.
Collapse
Affiliation(s)
- Israa Ahmad Cheikh
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Berthe Hayar
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Noorhan Ghanem
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Lara Al Saleh
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Chirine El-Baba
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Sadaf Al-Hadeethi
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Riyad El-Khoury
- Department of Pathology and Laboratory Medicine, American University of Beirut, Beirut, Lebanon
| | - Julnar Usta
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| | - Nadine Darwiche
- Department of Biochemistry and Molecular Genetics, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
129
|
Jung LA, Demir S, Hotes A, Hiyama E, Hishiki T, Indersie E, Branchereau S, Cairo S, Kappler R. Targeting HSP90 with Ganetespib to Induce CDK1 Degradation and Promote Cell Death in Hepatoblastoma. Cancers (Basel) 2025; 17:1341. [PMID: 40282517 PMCID: PMC12026307 DOI: 10.3390/cancers17081341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Hepatoblastoma, the most common malignant liver tumor in pediatric patients, is characterized by a remarkably low mutation rate, thereby impeding targeted therapies. Current treatment regimens rely on conventional cytotoxic agents that often cause severe adverse effects, necessitating the search for novel, less toxic therapeutic approaches. METHODS In this study, we explored the anti-tumor potential of heat shock protein 90 (HSP90) inhibitors using a unique collection of hepatoblastoma in vitro models. RESULTS Among the five tested inhibitors, we identified ganetespib as the most effective, significantly suppressing tumor cell growth while sparing healthy, non-tumor cells. Ganetespib treatment at low nanomolar concentrations markedly reduced cell proliferation, impaired long-term survival, and inhibited three-dimensional spheroid growth, ultimately leading to the induction of apoptosis. Mechanistically, ganetespib downregulated the expression of the HSP90 client protein cyclin-dependent kinase 1, a key cell cycle regulator controlling G2/M phase transition, which is heavily upregulated in hepatoblastoma. This disruption consequently resulted in cell cycle arrest, further contributing to its anti-tumor effects. CONCLUSIONS HSP90 inhibition by ganetespib demonstrates significant potential as a novel therapeutic strategy for hepatoblastoma, offering a potential alternative to current cytotoxic treatments with fewer adverse effects.
Collapse
Affiliation(s)
- Lea Amelie Jung
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, LMU University Hospital, 80337 Munich, Germany; (L.A.J.); (S.D.); (A.H.)
| | - Salih Demir
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, LMU University Hospital, 80337 Munich, Germany; (L.A.J.); (S.D.); (A.H.)
| | - Alina Hotes
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, LMU University Hospital, 80337 Munich, Germany; (L.A.J.); (S.D.); (A.H.)
| | - Eiso Hiyama
- Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima 739-0046, Japan;
| | - Tomoro Hishiki
- Department of Pediatric Surgery, Graduate School of Medicine, Chiba University, Chiba 260-8677, Japan;
| | | | | | - Stefano Cairo
- Champions Oncology, Inc., Rockville, MD 20850, USA;
- Pediatric Research Institute (IRP), 35127 Padova, Italy
| | - Roland Kappler
- Department of Pediatric Surgery, Dr. von Hauner Children’s Hospital, LMU University Hospital, 80337 Munich, Germany; (L.A.J.); (S.D.); (A.H.)
| |
Collapse
|
130
|
Zhou ZY, Bai N, Zheng WJ, Ni SJ. MultiOmics analysis of metabolic dysregulation and immune features in breast cancer. Int Immunopharmacol 2025; 152:114376. [PMID: 40054322 DOI: 10.1016/j.intimp.2025.114376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 02/09/2025] [Accepted: 02/24/2025] [Indexed: 03/24/2025]
Abstract
Metabolic disorders and diminished immune response are hallmark characteristics of tumors. However, limited studies have comprehensively integrated metabolic and immunological factors to evaluate or predict the prognosis of cancer patients. In this study, we utilized 72 metabolic pathway gene sets from the MsigDB database to conduct GSVA, univariate regression, and prognostic analyses on 247 breast cancer samples sourced from the TCGA and GEO databases. Consequently, five metabolic pathways with significant research value were identified. Based on these findings, unsupervised clustering was performed on the breast cancer samples to compare differences in gene expression, clinicopathological features, immune infiltration levels, and prognosis across different clusters. This process led to the identification of nine metabolism-related characteristic genes. Additionally, single-cell sequencing analysis was employed to assess the spatial expression patterns of these characteristic genes, revealing significantly higher expression indices in tumor cells compared to non-tumor cells. Subsequently, machine learning algorithms were applied to reconstruct metabolic risk models for evaluating the prognosis of breast cancer patients. The results indicated that the high metabolic risk group exhibited higher gene mutation scores, a greater proportion of unfavorable clinicopathological parameters, and lower chemokine and immune scores compared to the low-risk group. In conclusion, the metabolic risk model constructed using metabolism-related characteristic genes can accurately distinguish and predict the survival prognosis and immunotherapy outcomes of breast cancer patients, offering novel targets and insights for personalized treatment strategies.
Collapse
Affiliation(s)
- Zuo-Yuan Zhou
- Department of Oncology, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China
| | - Nan Bai
- Medical school of Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China; Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China
| | - Wen-Jie Zheng
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China.
| | - Su-Jie Ni
- Department of Oncology, Affiliated Hospital of Nantong University, #20 Xisi Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
131
|
Shan W, Zhang SL, Assaraf YG, Tam KY. Combined inhibition of hexokinase 2 and pyruvate dehydrogenase surmounts SHP2 inhibitor resistance in non-small cell lung cancer with hybrid metabolic state harboring KRAS Q61H mutation. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167859. [PMID: 40250775 DOI: 10.1016/j.bbadis.2025.167859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 03/02/2025] [Accepted: 04/15/2025] [Indexed: 04/20/2025]
Abstract
KRAS Q61H is an aggressive oncogenic driver mutation rendering cancer cells drug resistant to SHP2 inhibitors (SHP2i). Some metastatic and chemoresistant non-small cell lung cancer (NSCLC) cells, exhibit a hybrid metabolic state in which both glycolysis and oxidative phosphorylation (OXPHOS) coexist. Hence, we evaluated the in vitro and in vivo efficacy of a combination of hexokinase 2 (HK2) and pyruvate dehydrogenase (PDH) inhibitors, benserazide (Benz) and CPI-613, respectively, against NSCLC NCI-H460 cells harboring the driver KRAS Q61H mutation. This combination synergistically disrupted the hybrid metabolic state, inhibited NCI-H460 cell proliferation in vitro, and markedly suppressed tumor growth in NCI-H460 cell xenograft model in mice. The molecular basis underlying this antitumor activity was apparently due to suppression of SHP2/SOS1/RAS/MAPK signaling pathways, leading to enhanced apoptosis. Moreover, this drug combination restored the sensitivity to SHP2i. Consistently, SHP2 overexpression in NCI-H460 cells abrogated the antitumor activity of this drug combination. These findings reveal that the combination of Benz and CPI-613 targets the metabolic vulnerability of KRAS Q61H mutant-bearing NSCLC tumors. These results offer a combination therapeutic strategy for the possible treatment of cancer cells displaying a hybrid metabolic state, thereby surmounting chemoresistance.
Collapse
Affiliation(s)
- Wenying Shan
- Faculty of Health Sciences, University of Macau, Taipa, Macau
| | - Shao-Lin Zhang
- School of Pharmaceutical Sciences, Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Chongqing University, Chongqing 401331, PR China
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Faculty of Biology, Technion-Israel Institute of Technology, Haifa 3200003, Israel.
| | - Kin Yip Tam
- Faculty of Health Sciences, University of Macau, Taipa, Macau.
| |
Collapse
|
132
|
Li SS, Zhang B, Huang C, Fu Y, Zhao Y, Gong L, Tan Y, Wang H, Chen W, Luo J, Zhang Y, Ma S, Fu L, Liu C, Huang J, Ju HQ, Lee AWM, Guan XY. FAO-fueled OXPHOS and NRF2-mediated stress resilience in MICs drive lymph node metastasis. Proc Natl Acad Sci U S A 2025; 122:e2411241122. [PMID: 40215279 PMCID: PMC12012528 DOI: 10.1073/pnas.2411241122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 02/25/2025] [Indexed: 04/24/2025] Open
Abstract
Metastasis is an inefficient process requiring cancer cells to adapt metabolically for survival and colonization in new environments. The contributions of tumor metabolic reprogramming to lymph node (LN) metastasis and its underlying mechanisms remain elusive. Through single-cell RNA sequencing, we identified rare metastasis-initiating cells (MICs) with stem-like properties that drive early LN metastasis. Integrated transcriptome, lipidomic, metabolomic, and functional analyses demonstrated that MICs depend on oxidative phosphorylation (OXPHOS) fueled by fatty acid oxidation (FAO) in the lipid-rich LN microenvironment. Mechanistically, the NRF2-SLC7A11 axis promotes glutathione synthesis to mitigate oxidative stress, thereby enhancing stress resistance and metastatic potential of MICs. Inhibition of NRF2-SLC7A11 reduced LN metastasis and sensitized tumors to cisplatin. Clinically, elevated NRF2-SLC7A11 expression was observed in tumors, with high expression correlating with LN metastasis, chemoresistance, and poor prognosis in esophageal squamous cell carcinoma (ESCC). These findings highlight the pivotal roles of FAO-fueled OXPHOS and NRF2 in LN metastasis and suggest targeting these pathways as a promising therapeutic strategy for metastatic ESCC.
Collapse
Affiliation(s)
- Shan-Shan Li
- Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518000, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou516007, China
| | - Baifeng Zhang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong999077, China
| | - Cuicui Huang
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong999077, China
| | - Yuying Fu
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou516007, China
| | - Yuying Zhao
- Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518000, China
| | - Lanqi Gong
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong999077, China
| | - Yanan Tan
- Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518000, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou516007, China
| | - Huali Wang
- Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518000, China
| | - Wenqi Chen
- Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518000, China
| | - Jie Luo
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong999077, China
| | - Yu Zhang
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510060, China
| | - Stephanie Ma
- Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518000, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong999077, China
| | - Li Fu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology and International Cancer Center, Shenzhen University Health Science Center, Shenzhen518055, China
| | - Chenli Liu
- State Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen518055, China
| | - Jiandong Huang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong999077, China
- State Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen518055, China
| | - Huai-Qiang Ju
- Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518000, China
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510060, China
| | - Anne Wing-Mui Lee
- Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518000, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong999077, China
| | - Xin-Yuan Guan
- Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen518000, China
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou516007, China
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong999077, China
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou510060, China
| |
Collapse
|
133
|
Zhang BW, Huang T, Yang YF, Li MY, Shao GB. Lysine methyltransferase SETD7 in cancer: functions, molecular mechanisms and therapeutic implications. Mol Biol Rep 2025; 52:389. [PMID: 40232640 DOI: 10.1007/s11033-025-10494-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
Since its discovery as a histone methyltransferase, SETD7 has been implicated in many signaling pathways and carcinogenesis. SETD7 catalyzes the methylation of histone H3 and non-histone proteins, regulating their translation, stability and activity. SETD7 is frequently abnormally expressed and has a significant influence on cell proliferation, invasion, autophagy and immune response. As cancer is a complex disease, an outstanding concept in cancer biology is the "hallmarks of cancer". In this review, we focus on the involvement of SETD7 in the hallmarks of cancer, describing its functions and underlying mechanisms in detail. Additionally, we discuss non-coding RNAs and chemical inhibitors targeting SETD7, highlighting the potential and importance of SETD7 in cancer therapy.
Collapse
Affiliation(s)
- Bo-Wen Zhang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Ting Huang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Yi-Fan Yang
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Ming-Yang Li
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China
| | - Gen-Bao Shao
- Department of Basic Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China.
- Present Address: Jiangsu University, No.301 Xuefu Road, Jingkou District, Zhenjiang, Jiangsu province, China.
| |
Collapse
|
134
|
Hu L, Lin Y, Zheng J, Wan L, Zhao R, Ma Y, Li J. Transcriptome sequencing revealed that lymph node metastasis of papillary thyroid microcarcinoma is associated with high THBS4 expression and PDGFRA+ cancer-associated fibroblasts. Front Oncol 2025; 15:1536063. [PMID: 40303998 PMCID: PMC12037473 DOI: 10.3389/fonc.2025.1536063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
Background Cervical lymph node metastasis is a major factor influencing recurrence after surgery for papillary thyroid cancer. Molecular markers that can predict the presence of lymph node metastasis and assess the aggressiveness of papillary thyroid microcarcinoma (PTMC) remain poorly understood. The research question addressed whether specific genes, such as thrombospondin-4 (THBS4), could serve as predictive biomarkers for guiding surgical strategies, particularly in cases where current imaging modalities fail to detect LNM in the central region, and the decision for prophylactic central neck dissection remains controversial. Methods Transcriptome sequencing was employed to screen for differentially expressed genes and perform enrichment analysis. The study defined two groups of PTMC patients: LNM(n=50) and NLNM(n=50). 10 samples from each group were used for transcriptome sequencing. The expression of THBS4 was evaluated in both groups. Additionally, the correlation between THBS4 expression and cancer-associated fibroblasts (CAFs), specifically the PDGFRA+ inflammatory CAFs, was investigated to understand the stromal regulatory protein's role in PTMC aggressiveness. Results The analysis of sequencing data revealed that THBS4 expression was significantly higher in LNM PTMC compared to the NLNM group (Fold Change > 1.6 and P < 0.05). LNM PTMCs were also associated with a higher presence of PDGFRA+ inflammatory CAFs (P < 0.05), while no significant difference in the quantity of SMA+ myofibroblastic CAFs was observed between the two groups(P>0.05). Immunohistochemical analysis demonstrated increased THBS4(P < 0.01) and PDGFRA(P < 0.001) expression in LNM groups, while SMA staining showed no significant intergroup differences(P>0.05). Conclusion This study's findings indicate that THBS4 could be a potential biomarker for predicting the risk of lymph node metastasis in papillary thyroid microcarcinoma, thus potentially guiding more personalized surgical interventions. Further validation in larger patient cohorts and the interactions between THBS4 and CAFs are necessary.
Collapse
Affiliation(s)
- LeYin Hu
- Department of Pathology, Wenzhou Medical University First Affiliated Hospital, Wenzhou, Zhejiang, China
| | - Yi Lin
- Department of Pathology, Sanmen People’s Hospital, Taizhou, Zhejiang, China
| | - JingYu Zheng
- Department of Pathology, Wenzhou Medical University First Affiliated Hospital, Wenzhou, Zhejiang, China
| | - Li Wan
- Department of Pathology, Wenzhou Medical University First Affiliated Hospital, Wenzhou, Zhejiang, China
| | - Rui Zhao
- Department of Gastroenterology, Wenzhou Medical University First Affiliated Hospital, Wenzhou, Zhejiang, China
| | - Yi Ma
- Department of Pathology, Sanmen People’s Hospital, Taizhou, Zhejiang, China
| | - JianMin Li
- Department of Pathology, Wenzhou Medical University First Affiliated Hospital, Wenzhou, Zhejiang, China
| |
Collapse
|
135
|
Tang R, Luo S, Liu H, Sun Y, Liu M, Li L, Ren H, Angele MK, Börner N, Yu K, Guo Z, Yin G, Luo H. Circulating Tumor Microenvironment in Metastasis. Cancer Res 2025; 85:1354-1367. [PMID: 39992721 PMCID: PMC11997552 DOI: 10.1158/0008-5472.can-24-1241] [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: 04/15/2024] [Revised: 11/12/2024] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Activation of invasion and metastasis is a central hallmark of cancer, contributing to the primary cause of death for patients with cancer. In the multistep metastatic process, cancer cells must infiltrate the circulation, survive, arrest at capillary beds, extravasate, and form metastatic clones in distant organs. However, only a small proportion of circulating tumor cells (CTC) successfully form metastases, with transit of CTCs in the circulation being the rate-limiting step. The fate of CTCs is influenced by the circulating tumor microenvironment (cTME), which encompasses factors affecting their biological behaviors in the circulation. This liquid and flowing microenvironment differs significantly from the primary TME or the premetastatic niche. This review summarizes the latest advancements in identifying the biophysical cues, key components, and biological roles of the cTME, highlighting the network among biophysical attributes, blood cells, and nonblood factors in cancer metastasis. In addition to the potential of the cTME as a therapeutic target for inhibiting metastasis, the cTME could also represent as a biomarker for predicting patient outcomes and developing strategies for treating cancer.
Collapse
Affiliation(s)
- Rui Tang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Shujuan Luo
- Department of Obstetrics, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Liu
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yan Sun
- Department of Cell Biology and Medical Genetics, Basic Medical School, Chongqing Medical University, Chongqing, China
| | - Manran Liu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Lu Li
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Haoyu Ren
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Martin K. Angele
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich Munich, Germany
| | - Nikolaus Börner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich Munich, Germany
| | - Keda Yu
- Department of Breast Surgery, Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Zufeng Guo
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Guobing Yin
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Haojun Luo
- Department of Thyroid and Breast Surgery, Renji Hospital, School of Medicine, Chongqing University, Chongqing, China
| |
Collapse
|
136
|
George S, Saju H, Jaikumar T, Raj R, Nisarga R, Sontakke S, Sangshetti J, Paul MK, Arote RB. Deciphering a crosstalk between biological cues and multifunctional nanocarriers in lung cancer therapy. Int J Pharm 2025; 674:125395. [PMID: 40064384 DOI: 10.1016/j.ijpharm.2025.125395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/08/2025] [Accepted: 02/21/2025] [Indexed: 03/17/2025]
Abstract
In recent years, the utilization of nanocarriers has significantly broadened across a diverse spectrum of biomedical applications. However, the clinical translation of these tiny carriers is limited and encounters hurdles, particularly in the intricate landscape of the tumor microenvironment. Lung cancer poses unique hurdles for nanocarrier design. Multiple physiological barriers hinder the efficient drug delivery to the lungs, such as the complex anatomy of the lung, the presence of mucus, immune responses, and rapid clearance mechanisms. Overcoming these obstacles necessitates a targeted approach that minimizes off-target effects while effectively penetrating nanoparticles/cargo into specific lung tissues or cells. Furthermore, understanding the cellular uptake mechanisms of these nano carriers is also essential. This knowledge aids in developing nanocarriers that efficiently enter cells and transfer their payload for the most effective therapeutic outcome. Hence, a thorough understanding of biological cues becomes crucial in designing multifunctional nanocarriers tailored for treating lung cancer. This review explores the essential biological cues critical for developing a flexible nanocarrier specifically intended to treat lung cancer. Additionally, it discusses advancements in nanotheranostics in lung cancer.
Collapse
Affiliation(s)
- Sharon George
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - Hendry Saju
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - Tharun Jaikumar
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - Reshma Raj
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - R Nisarga
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - Samruddhi Sontakke
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India
| | - Jaiprakash Sangshetti
- Y. B. Chavan College of Pharmacy, Dr. Rafiq Zakaria Campus, Rauza Baugh, Aurangabad 431001, India
| | - Manash K Paul
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India; Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles (UCLA), 90095 CA, USA.
| | - Rohidas B Arote
- Centre for Nano and Material Sciences, Jain (Deemed to be) University, Jain Global Campus, Bangalore, Karnataka 562112, India; Dental Research Institute, School of Dentistry, Seoul National University, Gwanak-ku, Seoul 08826, Republic of Korea.
| |
Collapse
|
137
|
Kołodziejczak-Guglas I, Simões RLS, de Souza Santos E, Demicco EG, Lazcano Segura RN, Ma W, Wang P, Geffen Y, Storrs E, Petralia F, Colaprico A, da Veiga Leprevost F, Pugliese P, Ceccarelli M, Noushmehr H, Nesvizhskii AI, Kamińska B, Priebe W, Lubiński J, Zhang B, Lazar AJ, Kurzawa P, Mesri M, Robles AI, Ding L, Malta TM, Wiznerowicz M. Proteomic-based stemness score measures oncogenic dedifferentiation and enables the identification of druggable targets. CELL GENOMICS 2025:100851. [PMID: 40250426 DOI: 10.1016/j.xgen.2025.100851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 10/13/2024] [Accepted: 03/21/2025] [Indexed: 04/20/2025]
Abstract
Cancer progression and therapeutic resistance are closely linked to a stemness phenotype. Here, we introduce a protein-expression-based stemness index (PROTsi) to evaluate oncogenic dedifferentiation in relation to histopathology, molecular features, and clinical outcomes. Utilizing datasets from the Clinical Proteomic Tumor Analysis Consortium across 11 tumor types, we validate PROTsi's effectiveness in accurately quantifying stem-like features. Through integration of PROTsi with multi-omics, including protein post-translational modifications, we identify molecular features associated with stemness and proteins that act as active nodes within transcriptional networks, driving tumor aggressiveness. Proteins highly correlated with stemness were identified as potential drug targets, both shared and tumor specific. These stemness-associated proteins demonstrate predictive value for clinical outcomes, as confirmed by immunohistochemistry in multiple samples. The findings emphasize PROTsi's efficacy as a valuable tool for selecting predictive protein targets, a crucial step in customizing anti-cancer therapy and advancing the clinical development of cures for cancer patients.
Collapse
Affiliation(s)
- Iga Kołodziejczak-Guglas
- International Institute for Molecular Oncology, 60-203 Poznań, Poland; Postgraduate School of Molecular Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Renan L S Simões
- School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil
| | - Emerson de Souza Santos
- School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto 14040-900, Brazil
| | - Elizabeth G Demicco
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital and Laboratory Medicine and Pathobiology, University of Toronto, Toronto ON M5G 1X5, Canada
| | - Rossana N Lazcano Segura
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Weiping Ma
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Pei Wang
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yifat Geffen
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA; Cancer Center and Department of Pathology, Massachusetts General Hospital, Boston, MA 02115, USA
| | - Erik Storrs
- Department of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA; McDonnell Genome Institute, Washington University in St. Louis, St. Louis, MO 63108, USA
| | - Francesca Petralia
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Antonio Colaprico
- Sylvester Comprehensive Cancer Center and Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | | | - Pietro Pugliese
- Department of Science and Technology, University of Sannio, 82100 Benevento, Italy
| | - Michele Ceccarelli
- Sylvester Comprehensive Cancer Center and Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Houtan Noushmehr
- Hermelin Brain Tumor Center, Henry Ford Health System, Detroit, MI 48202, USA
| | - Alexey I Nesvizhskii
- Departments of Pathology and Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Bożena Kamińska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology, 02-093 Warsaw, Poland
| | - Waldemar Priebe
- Department of Experimental Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Jan Lubiński
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University in Szczecin, 70-204 Szczecin, Poland
| | - Bing Zhang
- Lester and Sue Smith Breast Center and Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexander J Lazar
- Departments of Pathology & Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Paweł Kurzawa
- Department of Oncological Pathology, University Clinical Hospital in Poznan, Poznan University of Medical Sciences, 60-514 Poznań, Poland
| | - Mehdi Mesri
- Office of Cancer Clinical Proteomics Research, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD 20850, USA
| | - Ana I Robles
- Office of Cancer Clinical Proteomics Research, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Rockville, MD 20850, USA
| | - Li Ding
- Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Tathiane M Malta
- School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo, Ribeirão Preto 14040-903, Brazil; Ribeirao Preto Medical School, University of São Paulo, Ribeirão Preto 14040-900, Brazil.
| | - Maciej Wiznerowicz
- International Institute for Molecular Oncology, 60-203 Poznań, Poland; Department of Oncology, Institute of Oncology, University Clinical Hospital in Poznan, Poznan University of Medical Sciences, 60-659 Poznań, Poland.
| |
Collapse
|
138
|
Zhang Y, Katkhada K, Meng LZ, Zhao B, Tong S, Chaabane W, Kallai A, Tobin NP, Östman A, Mega A, Ehnman M. Myogenic IGFBP5 levels in rhabdomyosarcoma are nourished by mesenchymal stromal cells and regulate growth arrest and apoptosis. Cell Commun Signal 2025; 23:184. [PMID: 40234830 PMCID: PMC12001570 DOI: 10.1186/s12964-025-02171-6] [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: 01/01/2025] [Accepted: 03/24/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Mesenchymal stromal cells belong to a diverse collection of cells in different states that are poorly characterized in soft-tissue sarcomas. In this study, we explored tumor growth-regulatory signaling between differentially educated non-malignant mesenchymal stromal cells and malignant cells in pediatric rhabdomyosarcoma (RMS). METHODS Xenograft experiments demonstrated that non-malignant stromal cells influence tumor behavior. Gene expression analysis identified deregulated genes, which were further studied using cell culture assays and patient data. Clinicopathological correlations were made in a discovery cohort (N = 147) and a validation cohort (N = 101). RESULTS The results revealed transiently suppressive paracrine effects of orthotopic stromal cells derived from skeletal muscle. These effects were lost when the stromal cells were exposed to RMS cells, either short-term in vitro, or long-term in hindlimb muscle in vivo. High resolution microarray-based Clariom D gene expression analysis identified insulin-like growth factor binding protein 5 (IGFBP5) as the top upregulated gene in RMS cells exposed to naïve stromal cells, and effects on growth arrest, caspase 3/7 activation, and myogenic cell identity were demonstrated in functional assays. Furthermore, IGFBP5 associated with the caspase 3 substrate growth arrest specific protein 2 (GAS2), lower disease stage and favorable survival in patient cohorts. CONCLUSIONS This study uses functional modeling and omics approaches to identify IGFBP5 as a candidate mediator of anti-tumor growth mechanisms originating from tumor-neighboring mesenchymal stromal cells. Tumors of mesenchymal origin, such as RMS, are known for their heterogeneity, and this could potentially pose a limitation to the study. However, a clinical relevance is emphasized by consistent findings across patient cohorts. These insights pave the way for novel therapeutic strategies modulating activities of stromal cell subsets at primary and metastatic sites in RMS.
Collapse
Affiliation(s)
- Yue Zhang
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Karim Katkhada
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Liu Zhen Meng
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Binbin Zhao
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Shanlin Tong
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Wiem Chaabane
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Aditi Kallai
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Nicholas P Tobin
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Breast Center, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Stockholm, Sweden
| | - Arne Östman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
- Centre for Cancer Biomarkers CCBIO, University of Bergen, 5021, Bergen, Norway
| | - Alessandro Mega
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Monika Ehnman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden.
- PO Bröst- och endokrina tumörer och sarkom, Tema Cancer, Karolinska University Hospital, Visionsgatan 4, SE-171 76, Stockholm, Sweden.
| |
Collapse
|
139
|
Lan JX, Huang LJ, Kang SS, Hao-Huang, Liu SL, Dai W, Xu XL, Wang JY, Shu GZ, Hou W. Design, synthesis, biological evaluation, and mechanism of action of new pyrazines as anticancer agents in vitro and in vivo. Bioorg Med Chem 2025; 121:118108. [PMID: 39955800 DOI: 10.1016/j.bmc.2025.118108] [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: 11/24/2024] [Revised: 02/02/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
Cancer is the second leading cause of mortality worldwide. The development of innovative antitumor pharmaceuticals is urgently needed to alter this circumstance. N-heterocycles, pyrazines for example are prevalent pharmacophores in the architecture of anticancer medicines. This research involved the design and synthesis of seventy-seven new pyrazine derivatives, followed by an evaluation of their anticancer activity in vitro and in vivo. Several new pyrazines exhibiting remarkable antiproliferative activity and selectivity were identified. The links between structure and function were analyzed, and the mechanisms of action were examined. Our mechanistic investigations indicated that these chemicals triggered mitochondria-associated apoptosis in cancer cells. Moreover, they suppressed the phosphorylation of STAT3, concomitant with the down-regulation of BcL-2, BcL-XL, c-Myc, XIAP, GLI1, TAZ, MCL1, JAK1, JAK2 and up-regulation of Bax, p21. Furthermore, the lead compounds B-11 and C-27 demonstrated significant anticancer activity in vivo in the SKOV3 xenograft nude mouse model. Our research establishes a basis for the identification of pyrazines as JAK/STAT3 inhibition based anticancer lead compounds.
Collapse
Affiliation(s)
- Jin-Xia Lan
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China; School of Public Health and Health Management, Gannan Medical University, Ganzhou 341000 PR China
| | - Le-Jun Huang
- School of Rehabilitation, Gannan Medical University, Ganzhou 341000 PR China
| | - Si-Shuang Kang
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Hao-Huang
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Sheng-Lan Liu
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Wei Dai
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Xin-Liang Xu
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Jin-Yang Wang
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Guang-Zhao Shu
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China
| | - Wen Hou
- Jiangxi Province Key Laboratory of Pharmacology of Traditional Chinese Medicine, National Engineering Research Center for Modernization of Traditional Chinese Medicine-Hakka Medical Resources Branch, School of Pharmacy, Gannan Medical University, Ganzhou 341000 PR China.
| |
Collapse
|
140
|
Jessen SB, Vogelsang RP, Dolin TG, Jørgensen J, Olsson JB, Kirkegaard T, Gögenur I, Troelsen JT. Surgery-related change in cancer cell adhesion associates with recurrence in patients undergoing colorectal cancer surgery. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:110055. [PMID: 40253751 DOI: 10.1016/j.ejso.2025.110055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 04/12/2025] [Indexed: 04/22/2025]
Abstract
OBJECTIVES This study aimed to investigate the effect of pre- and postoperative serum on the adhesion of cultured colon cancer cells and their relationship with colorectal cancer recurrence. BACKGROUND Colorectal cancer is common, and surgery is the primary treatment choice. However, surgical procedures may be associated with an increased risk of recurrence. METHOD 434 patients undergoing curatively intended colorectal cancer surgery at Copenhagen University Hospital, Herlev, Denmark, between July 15, 2014, and March 31, 2019, were included in the study. Pre- and postoperative serum samples were collected, and the effect on cellular adhesion was analyzed using a novel high-throughput approach based on CRISPR/Cas9 modified Caco-2 cells and secreted luciferase, named the AdhesionScore assay. The relative risk of postoperative recurrence was estimated using Cox proportional regression analysis. RESULTS The difference in adhesion between modified Caco-2 cells seeded in the pre- and postoperative serum showed a significant increase in postoperative adhesion in patients with a recurrence event (p=0.0293). Modeling the adhesion data using multiple logistic regression and Cox proportional regression analyses showed a statistically significant association between increased postoperative adhesion and recurrence (p=0.0155 and p=0.0126, respectively). Patients with the highest AdhesionScore showed the greatest risk of recurrence (HR=7, 95% CI 1.6-37.8, p=0.0130). CONCLUSIONS The study found that a difference in the adhesion of Caco-2 cells seeded in pre- and postoperative serum was associated with cancer recurrence following intended curative surgery. This suggests that increased postoperative adhesion may serve as a novel biological marker of recurrence in patients undergoing surgery for colorectal cancer.
Collapse
Affiliation(s)
- Stine Bull Jessen
- Department of Science and Environment, Enhanced Perioperative Oncology (EPeOnc) Consortium, Roskilde University, Universitetsvej 1, 4000, Roskilde, Denmark; Center for Surgical Science, Enhanced Perioperative Oncology (EPeOnc) Consortium, Department of Surgery, Zealand University Hospital, Lykkebækvej 1, Køge, 4600, Denmark
| | - Rasmus Peuliche Vogelsang
- Center for Surgical Science, Enhanced Perioperative Oncology (EPeOnc) Consortium, Department of Surgery, Zealand University Hospital, Lykkebækvej 1, Køge, 4600, Denmark
| | - Troels Gammeltoft Dolin
- Department of Medicine, Copenhagen University Hospital - Herlev and Gentofte, Borgmester Ib Juuls Vej 1, Herlev, 2730, Denmark; Department of Surgery, Copenhagen University Hospital - Herlev and Gentofte, Borgmester Ib Juuls Vej 1, Herlev, 2730, Denmark
| | - Jannie Jørgensen
- Department of Science and Environment, Enhanced Perioperative Oncology (EPeOnc) Consortium, Roskilde University, Universitetsvej 1, 4000, Roskilde, Denmark; Department of Clinical Immunology, Naestved Hospital, Ringstedgade 77B, Naestved, 4700, Denmark
| | - Josephine Bjergbæk Olsson
- Department of Science and Environment, Enhanced Perioperative Oncology (EPeOnc) Consortium, Roskilde University, Universitetsvej 1, 4000, Roskilde, Denmark; Department of Clinical Immunology, Naestved Hospital, Ringstedgade 77B, Naestved, 4700, Denmark
| | - Tove Kirkegaard
- Center for Surgical Science, Enhanced Perioperative Oncology (EPeOnc) Consortium, Department of Surgery, Zealand University Hospital, Lykkebækvej 1, Køge, 4600, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Enhanced Perioperative Oncology (EPeOnc) Consortium, Department of Surgery, Zealand University Hospital, Lykkebækvej 1, Køge, 4600, Denmark
| | - Jesper Thorvald Troelsen
- Department of Science and Environment, Enhanced Perioperative Oncology (EPeOnc) Consortium, Roskilde University, Universitetsvej 1, 4000, Roskilde, Denmark.
| |
Collapse
|
141
|
Huang C, Wang L, Zhuo C, Chen W, Fan H, Hong Y, Zhang Y, Zhou D, Lin W, Zhang L, Zhao J, Chen S, Yu C, Ye Y. ID3 enhances PD-L1 expression by restructuring MYC to promote colorectal cancer immune evasion. Proc Natl Acad Sci U S A 2025; 122:e2423490122. [PMID: 40208940 PMCID: PMC12012548 DOI: 10.1073/pnas.2423490122] [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: 11/13/2024] [Accepted: 03/12/2025] [Indexed: 04/12/2025] Open
Abstract
The inhibitor of DNA binding protein ID3 has been associated with the progression of colorectal cancer (CRC). Despite its significance, its specific role in the immune evasion strategies utilized by CRC remains unclear. RNA-seq analysis revealed that ID3 was positively associated with the PD-L1 immune checkpoint. We further demonstrated that tumor cell-expressed ID3 enhanced PD-L1 expression, suppressed the infiltration and activation of CD8+ T cells, and facilitated the immune evasion of CRC cells. Additionally, we found that knockdown of ID3 significantly enhanced the effectiveness of PD-L1 antibody blockade treatment in combating CRC, reduced the upregulation of PD-L1 induced by the antibody, and altered the immune microenvironment within CRC. Mechanistically, ID3 interacted with the transcription factor MYC and reconstructed the four-dimensional structure of MYC, thereby enhancing its binding affinity to the PD-L1 promoter and augmenting PD-L1 transcriptional activity. By integrating analysis of ChIP-seq, RNA-seq, and ImmPort gene sets, we found that ID3's DNA-assisted binding function was widespread and could either enhance or suppress gene transcription, not only affecting tumor immune escape through immune checkpoints but also regulating various cytokines and immune cells involved in tumor immunity. In conclusion, our study uncovers a mechanism by which ID3 promotes immune evasion in CRC and implicates that targeting ID3 may improve the efficacy of anti-PD-1/PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Chuanzhong Huang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou350014, People’s Republic of China
| | - Ling Wang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
| | - Changhua Zhuo
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
| | - Wenxin Chen
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
| | - Hongmei Fan
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
| | - Yilin Hong
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen361102, People’s Republic of China
| | - Yu Zhang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
| | - Dongmei Zhou
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
| | - Wansong Lin
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
| | - Lingyu Zhang
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
| | - Jingjing Zhao
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
| | - Shuping Chen
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou350122, China
| | - Chundong Yu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen361102, People’s Republic of China
| | - Yunbin Ye
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian Medical University, Fuzhou350014, People’s Republic of China
| |
Collapse
|
142
|
Yasukawa K, Shimada S, Akiyama Y, Taniai T, Igarashi Y, Tsukihara S, Tanji Y, Umemura K, Kamachi A, Nara A, Yamane M, Akahoshi K, Shimizu A, Soejima Y, Tanabe M, Tanaka S. ACVR2A attenuation impacts lactate production and hyperglycolytic conditions attracting regulatory T cells in hepatocellular carcinoma. Cell Rep Med 2025; 6:102038. [PMID: 40139191 PMCID: PMC12047472 DOI: 10.1016/j.xcrm.2025.102038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 10/01/2024] [Accepted: 03/03/2025] [Indexed: 03/29/2025]
Abstract
Although ACVR2A mutations are prevalent in non-viral hepatocellular carcinomas (HCCs), the underlying mechanism remains unelucidated. Our molecular investigation reveals that ACVR2A impairment induces hyperglycolysis through the inactivation of the SMAD signaling pathway. Using syngeneic transplantation models and human clinical samples, we clarify that ACVR2A-deficient HCC cells produce and secrete lactate via the upregulation of lactate dehydrogenase A (LDHA) and monocarboxylate transporter 4 (MCT4) expression levels, which promotes regulatory T (Treg) cell accumulation and then acquires resistance to immune checkpoint inhibitors. Remarkably, genetic knockdown and pharmacological inhibition of MCT4 ameliorate the high-lactate milieu in ACVR2A-deficient HCC, resulting in the suppression of intratumoral Treg cell recruitment and the restoration of the sensitivity to PD-1 blockade. These findings furnish compelling evidence that lactate attenuates anti-tumor immunity and that therapeutics targeting this pathway present a promising strategy for mitigating immunotherapy resistance in ACVR2A-deficient HCC.
Collapse
Affiliation(s)
- Koya Yasukawa
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; Division of Gastroenterological, Hepato-Biliary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Shu Shimada
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan.
| | - Yoshimitsu Akiyama
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Tomohiko Taniai
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo 105-8471, Japan
| | - Yosuke Igarashi
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo 105-8471, Japan
| | - Shu Tsukihara
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; Department of Surgery, The Jikei University School of Medicine, Tokyo 105-8471, Japan
| | - Yoshiaki Tanji
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; Division of Hepatobiliary and Pancreas Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo 105-8471, Japan
| | - Kentaro Umemura
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; Division of Gastroenterological, Hepato-Biliary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Atsushi Kamachi
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; Division of Gastroenterological, Hepato-Biliary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Atsushi Nara
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; Department of Hepato-Biliary-Pancreatic Surgery, Tokyo Medical and Dental University, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Masahiro Yamane
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; Department of Hepato-Biliary-Pancreatic Surgery, Tokyo Medical and Dental University, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Keiichi Akahoshi
- Department of Hepato-Biliary-Pancreatic Surgery, Tokyo Medical and Dental University, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Akira Shimizu
- Division of Gastroenterological, Hepato-Biliary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Yuji Soejima
- Division of Gastroenterological, Hepato-Biliary-Pancreatic, Transplantation and Pediatric Surgery, Department of Surgery, Shinshu University School of Medicine, Matsumoto 390-8621, Japan
| | - Minoru Tanabe
- Department of Hepato-Biliary-Pancreatic Surgery, Tokyo Medical and Dental University, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan
| | - Shinji Tanaka
- Department of Molecular Oncology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan; Department of Hepato-Biliary-Pancreatic Surgery, Tokyo Medical and Dental University, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo 113-8519, Japan.
| |
Collapse
|
143
|
Chen Q, Zhang Y, Gao J, Zhang J. CPPCGM: A Highly Efficient Sequence-Based Tool for Simultaneously Identifying and Generating Cell-Penetrating Peptides. J Chem Inf Model 2025; 65:3357-3369. [PMID: 40105337 DOI: 10.1021/acs.jcim.5c00199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Cell-penetrating peptides (CPPs) are usually short oligopeptides with 5-30 amino acid residues. CPPs have been proven as important drug delivery vehicles into cells through different mechanisms, demonstrating their potential as therapeutic candidates. However, experimental screening and synthesis of CPPs could be time-consuming and expensive. Recently, numerous attempts have been made to develop computational methods as a cost-effective way for screening a number of potential CPP candidates. Despite significant advancements, current methods exhibit limited feature representation capabilities, thereby constraining the potential for further performance enhancements. In this study, we developed a deep learning framework called CPPCGM, which uses protein language models (PLMs) to identify and generate novel CPPs. There are two separate blocks in this framework: CPPClassifier and CPPGenerator. The former utilizes three pretrained models for simple voting, thereby accurately categorizing CPPs and non-CPPs. The latter, similar to a generative adversarial network, including a discriminator and a generator, generates peptides that are not present in the training data set. Our proposed CPPCGM has achieved remarkably high Matthews correlation coefficient scores of 0.876, 0.923, and 0.664 on three data sets based on the classification results. Compared with the state-of-the-art methods, the performance of our method is significantly improved. The results also demonstrated the generating potential of CPPCGM through qualitative and quantitative evaluation of the generated samples. Significantly, using PLM-based methods can optimize peptides for biochemical functions, benefiting drug delivery and biomedical applications. Materials related are publicly available at https://github.com/QiufenChen/CPPCGM.
Collapse
Affiliation(s)
- Qiufen Chen
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Yuewei Zhang
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Jiali Gao
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518055, China
- School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen 518055, China
- Department of Chemistry and Supercomputing Institute, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Jun Zhang
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen 518055, China
| |
Collapse
|
144
|
Jaber S, Eldawra E, Rakotopare J, Simeonova I, Lejour V, Gabriel M, Cañeque T, Volochtchouk V, Licaj M, Fajac A, Rodriguez R, Morillon A, Bardot B, Toledo F. Oncogenic and teratogenic effects of Trp53Y217C, an inflammation-prone mouse model of the human hotspot mutant TP53Y220C. eLife 2025; 13:RP102434. [PMID: 40223808 PMCID: PMC11996178 DOI: 10.7554/elife.102434] [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] [Indexed: 04/15/2025] Open
Abstract
Missense 'hotspot' mutations localized in six p53 codons account for 20% of TP53 mutations in human cancers. Hotspot p53 mutants have lost the tumor suppressive functions of the wildtype protein, but whether and how they may gain additional functions promoting tumorigenesis remain controversial. Here, we generated Trp53Y217C, a mouse model of the human hotspot mutant TP53Y220C. DNA damage responses were lost in Trp53Y217C/Y217C (Trp53YC/YC) cells, and Trp53YC/YC fibroblasts exhibited increased chromosome instability compared to Trp53-/- cells. Furthermore, Trp53YC/YC male mice died earlier than Trp53-/- males, with more aggressive thymic lymphomas. This correlated with an increased expression of inflammation-related genes in Trp53YC/YC thymic cells compared to Trp53-/- cells. Surprisingly, we recovered only one Trp53YC/YC female for 22 Trp53YC/YC males at weaning, a skewed distribution explained by a high frequency of Trp53YC/YC female embryos with exencephaly and the death of most Trp53YC/YC female neonates. Strikingly, however, when we treated pregnant females with the anti-inflammatory drug supformin (LCC-12), we observed a fivefold increase in the proportion of viable Trp53YC/YC weaned females in their progeny. Together, these data suggest that the p53Y217C mutation not only abrogates wildtype p53 functions but also promotes inflammation, with oncogenic effects in males and teratogenic effects in females.
Collapse
Affiliation(s)
- Sara Jaber
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Eliana Eldawra
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Jeanne Rakotopare
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Iva Simeonova
- Chromatin Dynamics, Institut Curie, CNRS UMR3664, Sorbonne University, PSL UniversityParisFrance
| | - Vincent Lejour
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Marc Gabriel
- Non Coding RNA, Epigenetic and Genome Fluidity, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Tatiana Cañeque
- Chemical Biology, Institut Curie, CNRS UMR3666, INSERM U1143, PSL UniversityParisFrance
| | - Vitalina Volochtchouk
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Monika Licaj
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Anne Fajac
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Raphaël Rodriguez
- Chemical Biology, Institut Curie, CNRS UMR3666, INSERM U1143, PSL UniversityParisFrance
| | - Antonin Morillon
- Non Coding RNA, Epigenetic and Genome Fluidity, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
| | - Boris Bardot
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
- Signaling and Neural Crest Development, Institut Curie, CNRS UMR3347, INSERM U1021, Université Paris-Saclay, PSL UniversityOrsayFrance
| | - Franck Toledo
- Genetics of Tumor Suppression, Institut Curie, CNRS UMR3244, Sorbonne University, PSL UniversityParisFrance
- Hematopoietic and Leukemic Development, Centre de Recherche Saint-Antoine, INSERM UMRS938, Sorbonne UniversityParisFrance
| |
Collapse
|
145
|
Espinoza-Ferrao S, Echeverría-Garcés G, Rivera-Orellana S, Bueno-Miño J, Castellanos-Molina E, Benítez-Núñez M, López-Cortés A. Global analysis of actionable genomic alterations in thyroid cancer and precision-based pharmacogenomic strategies. Front Pharmacol 2025; 16:1524623. [PMID: 40297138 PMCID: PMC12034932 DOI: 10.3389/fphar.2025.1524623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 04/01/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Thyroid cancer, a prevalent endocrine malignancy, has an age-standardized incidence rate of 9.1 per 100,000 people and a mortality rate of 0.44 per 100,000 as of 2024. Despite significant advances in precision oncology driven by large-scale international consortia, gaps persist in understanding the genomic landscape of thyroid cancer and its impact on therapeutic efficacy across diverse populations. Methods To address this gap, we performed comprehensive data mining and in silico analyses to identify pathogenic variants in thyroid cancer driver genes, calculate allele frequencies, and assess deleteriousness scores across global populations, including African, Amish, Ashkenazi Jewish, East and South Asian, Finnish and non-Finnish European, Latino, and Middle Eastern groups. Additionally, pharmacogenomic profiling, in silico drug prescription, and clinical trial data were analyzed to prioritize targeted therapeutic strategies. Results Our analysis examined 56,622 variants in 40 thyroid cancer-driver genes across 76,156 human genomes, identifying 5,001 known and predicted oncogenic variants. Enrichment analysis revealed critical pathways such as MAPK, PI3K-AKT-mTOR, and p53 signaling, underscoring their roles in thyroid cancer pathogenesis. High-throughput validation strategies confirmed actionable genomic alterations in RET, BRAF, NRAS, KRAS, and EPHA7. Ligandability assessments identified these proteins as promising therapeutic targets. Furthermore, our findings highlight the clinical potential of targeted drug inhibitors, including vandetanib, dabrafenib, and selumetinib, for improving treatment outcomes. Discussion This study underscores the significance of integrating genomic insights with pharmacogenomic strategies to address disparities in thyroid cancer treatment. The identification of population-specific oncogenic variants and actionable therapeutic targets provides a foundation for advancing precision oncology. Future efforts should focus on including underrepresented populations, developing population-specific prevention strategies, and fostering global collaboration to ensure equitable access to pharmacogenomic testing and innovative therapies. These initiatives have the potential to transform thyroid cancer care and align with the broader goals of personalized medicine.
Collapse
Affiliation(s)
| | - Gabriela Echeverría-Garcés
- Centro de Referencia Nacional de Genómica, Secuenciación y Bioinformática, Instituto Nacional de Investigación en Salud Pública “Leopoldo Izquieta Pérez”, Quito, Ecuador
- Latin American Network for the Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Santiago, Chile
| | | | - José Bueno-Miño
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | | | - Melanie Benítez-Núñez
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| | - Andrés López-Cortés
- Cancer Research Group (CRG), Faculty of Medicine, Universidad de Las Américas, Quito, Ecuador
| |
Collapse
|
146
|
Xavierselvan M, Shethia RT, Bednarke B, Yang V, Moses L, Yalamarty SSK, Cook J, Mallidi S. Oxygen-Releasing Nanodroplets Relieve Intratumoral Hypoxia and Potentiate Photodynamic Therapy in 3D Head and Neck Cancer Spheroids. ACS Biomater Sci Eng 2025; 11:2378-2395. [PMID: 40041949 PMCID: PMC12002064 DOI: 10.1021/acsbiomaterials.4c02031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 02/01/2025] [Accepted: 02/20/2025] [Indexed: 03/12/2025]
Abstract
Hypoxia in solid tumors, including head and neck cancer (HNC), contributes to treatment resistance, aggressive tumor phenotypes, and poorer clinical outcomes. Perfluorocarbon nanodroplets have emerged as promising drugs to alleviate tumor hypoxia. These versatile nanocarriers can also encapsulate and deliver various therapeutic agents, offering a multifunctional approach to cancer treatment. However, a detailed characterization of hypoxia alleviation, particularly the duration of hypoxia treatment drug residence, has not been thoroughly investigated. In this study, we developed and characterized perfluoropentane nanodroplets (PFP NDs) for the codelivery of oxygen and the photoactivatable drug benzoporphyrin derivative (BPD) to hypoxic HNC spheroids. The PFP NDs exhibited excellent stability, efficient oxygen loading/release, and biocompatibility. Using 3D multicellular tumor spheroids of FaDu and SCC9 HNC cells, we investigated the spatiotemporal dynamics of hypoxia within these spheroids and the ability of oxygenated PFP NDs to alleviate hypoxia. Our results showed that oxygen-loaded PFP NDs effectively penetrated the core of tumor spheroids, significantly reducing hypoxia, as evidenced by the downregulation of hypoxia-inducible factors HIF-1α and HIF-2α. Importantly, we demonstrated sustained hypoxia alleviation for up to 3 h post-treatment with PFP NDs. BPD-loaded PFP NDs successfully delivered the photosensitizer into the spheroid core in a time-dependent manner. Furthermore, we evaluated the efficacy of oxygen-dependent treatment modality, namely, photodynamic therapy (PDT) with BPD and oxygen-loaded PFP NDs compared to free BPD. The NDs formulation exhibited superior PDT outcomes, which were attributed to improved oxygen availability during the treatment. This study provides comprehensive evidence for the potential of PFP NDs as a codelivery platform to overcome hypoxia-mediated treatment resistance and enhance PDT efficacy in HNC. Our findings pave the way for further investigation of this promising approach in more complex in vivo models, potentially leading to improved therapeutic strategies for hypoxic solid tumors.
Collapse
Affiliation(s)
- Marvin Xavierselvan
- Department
of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155-5801, United
States
| | - Ronak Tarun Shethia
- Department
of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155-5801, United
States
| | - Brooke Bednarke
- Department
of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155-5801, United
States
| | - Vicky Yang
- Department
of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155-5801, United
States
| | - Leah Moses
- Department
of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155-5801, United
States
| | | | - Jason Cook
- NanoHybrids,
Inc., Acton, Massachusetts 01720, United States
| | - Srivalleesha Mallidi
- Department
of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155-5801, United
States
- Wellman Center
for Photomedicine, Massachusetts General
Hospital, Boston, Massachusetts 02114, United States
| |
Collapse
|
147
|
Nam SB, Choi JH, Lee GE, Kim JY, Lee MH, Yang G, Cho YY, Jeong HG, Bang G, Lee CJ. Extracts from Allium pseudojaponicum Makino Target STAT3 Signaling Pathway to Overcome Cisplatin Resistance in Lung Cancer. Mar Drugs 2025; 23:167. [PMID: 40278288 PMCID: PMC12028371 DOI: 10.3390/md23040167] [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: 03/20/2025] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Lung cancer, particularly non-small-cell lung cancer (NSCLC), remains a leading cause of cancer-related mortality, with cisplatin-based chemotherapy being a standard treatment. However, the development of chemoresistance significantly limits its efficacy, necessitating alternative therapeutic approaches. Here, we demonstrate the anticancer effects of the extracts of Allium pseudojaponicum Makino (APE), a salt-tolerant plant, in cisplatin-resistant NSCLC. Metabolite profiling using UPLC-Q-TOF-MSE identified 13 major compounds, predominantly alkaloids (71.65%) and flavonoids (8.81%), with key bioactive constituents such as lycorine (29.81%), tazettine (17.22%), and tricetin (8.19%). APE significantly inhibited cell viability in A549 and H460 cells, reducing viability to 38.6% (A549-Ctr), 37.2% (A549-CR), 28.4% (H460-Ctr), and 30.4% (H460-CR) at 40 µg/mL after 48 h. APE also suppressed colony formation by over 90% in both 2D and soft agar assays, while showing no cytotoxicity in normal human keratinocytes up to 80 µg/mL. Flow cytometry analysis revealed APE-induced G1 phase arrest, with the G1 population increasing from 50.4% to 56.6% (A549-Ctr) and 47.5% to 58.4% (A549-CR), accompanied by reduced S phase populations. This effect was associated with the downregulation of G1/S transition regulators, including cyclin D1, CDK4, cyclin E, and CDK2. Furthermore, proteomic analysis identified STAT3 signaling as a major target of APE; APE decreased phosphorylated STAT3 and c-Myc expression, and STAT3 knockdown phenocopied the effects of APE. These findings highlight the potential of APE as a natural product-based therapeutic strategy for overcoming cisplatin resistance in NSCLC.
Collapse
Affiliation(s)
- Soo-Bin Nam
- Biopharmaceutical Research Center, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea; (S.-B.N.); (G.-E.L.)
- College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea;
| | - Jung Hoon Choi
- Digital Omics Research Center, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea; (J.H.C.); (J.Y.K.)
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea;
| | - Ga-Eun Lee
- Biopharmaceutical Research Center, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea; (S.-B.N.); (G.-E.L.)
| | - Jin Young Kim
- Digital Omics Research Center, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea; (J.H.C.); (J.Y.K.)
| | - Mee-Hyun Lee
- College of Korean Medicine, Dongshin University, Naju 58245, Republic of Korea;
| | - Gabsik Yang
- Department of Korean Medicine, College of Korean Medicine, Woosuk University, Jeonju 55338, Republic of Korea;
| | - Yong-Yeon Cho
- College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea;
| | - Hye Gwang Jeong
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea;
| | - Geul Bang
- Digital Omics Research Center, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea; (J.H.C.); (J.Y.K.)
| | - Cheol-Jung Lee
- Biopharmaceutical Research Center, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea; (S.-B.N.); (G.-E.L.)
- Department of Bio-Analytical Science, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| |
Collapse
|
148
|
Wang X, Wang Z, Liu Z, Huang F, Pan Z, Zhang Z, Liu T. Nutritional strategies in oncology: The role of dietary patterns in modulating tumor progression and treatment response. Biochim Biophys Acta Rev Cancer 2025; 1880:189322. [PMID: 40228747 DOI: 10.1016/j.bbcan.2025.189322] [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: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/16/2025]
Abstract
Dietary interventions can influence tumor growth by restricting tumor-specific nutritional requirements, altering the nutrient availability in the tumor microenvironment, or enhancing the cytotoxicity of anticancer drugs. Metabolic reprogramming of tumor cells, as a significant hallmark of tumor progression, has a profound impact on immune regulation, severely hindering tumor eradication. Dietary interventions can modify tumor metabolic processes to some extent, thereby further improving the efficacy of tumor treatment. In this review, we emphasize the impact of dietary patterns on tumor progression. By exploring the metabolic differences of nutrients in normal cells versus cancer cells, we further clarify how dietary patterns influence cancer treatment. We also discuss the effects of dietary patterns on traditional treatments such as immunotherapy, chemotherapy, radiotherapy, and the gut microbiome, thereby underscoring the importance of precision nutrition.
Collapse
Affiliation(s)
- Xueying Wang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zeyao Wang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zihan Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Fanxuan Huang
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China
| | - Zhaoyu Pan
- Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital, Central South University, Hunan, China
| | - Zhiren Zhang
- Departments of Cardiology and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China; Departments of Cardiology and Pharmacy and Breast Cancer surgery, Harbin Medical University Cancer Hospital, Institute of Metabolic Disease, Heilongjiang Academy of Medical Science, Heilongjiang Key Laboratory for Metabolic Disorder and Cancer Related Cardiovascular Diseases, Harbin, China.
| | - Tong Liu
- Department of Oncological Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang Province 150000, China; Departments of Cardiology and Critical Care Medicine, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, China.
| |
Collapse
|
149
|
Murtazina A, Jimenez-Martinez Y, Ruiz Alcala G, Marchal JA, Tarabayeva A, Bitanova E, Rakhimbayev I, McDougall GJ, Bishimbayeva N, Boulaiz H. In Vitro Inhibition of Colon Cancer Stem Cells by Natural Polysaccharides Obtained from Wheat Cell Culture. Polymers (Basel) 2025; 17:1048. [PMID: 40284312 PMCID: PMC12030112 DOI: 10.3390/polym17081048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/05/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
Natural polysaccharides (PSs) have shown inhibitory effects on differentiated cancer cells (DCCs), but their activity against cancer stem cells (CSCs) remains poorly understood. Here, we report that PSs from wheat cell cultures (WCCPSs) inhibit the proliferation of both DCCs and CSCs derived from HCT-116 colorectal cancer cells. Among them, NA and DC fractions showed the strongest anti-CSC activity. NA, rich in xylose, was effective at lower concentrations, while DC, enriched in xylose and galacturonic acid (GalUA), exhibited higher potency, with a lower IC50 and preferential activity against CSCs at higher doses. WCCPSs reduced β-catenin levels, and some fractions also downregulated Ep-CAM, CD44, and c-Myc. Notably, DC increased caspase-3 without inducing cytochrome C and caspase-8 overexpression, suggesting a mechanism promoting CSC differentiation rather than apoptosis. Correlation analysis linked xylose content to reduced c-Myc expression, and GalUA levels to increased caspase-3. These results suggest that WCCPS bioactivity may be related to their monosaccharide composition. Overall, our findings support the potential of wheat-derived PSs as CSC-targeting agents that suppress self-renewal and promote differentiation, offering a promising approach to reduce tumor aggressiveness and recurrence.
Collapse
Affiliation(s)
- Alima Murtazina
- Department of General Immunology, Faculty of Medicine, Asfendyarov Kazakh National Medical University, Almaty 050012, Kazakhstan; (A.M.); (A.T.); (E.B.)
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18016 Granada, Spain; (Y.J.-M.); (G.R.A.); (J.A.M.)
- Research Center “Bioscience Technologies”, Almaty 050057, Kazakhstan
| | - Yaiza Jimenez-Martinez
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18016 Granada, Spain; (Y.J.-M.); (G.R.A.); (J.A.M.)
- Instituto de Investigación Biosanitaria ibs. GRANADA, University Hospitals of Granada-University of Granada, 18014 Granada, Spain
| | - Gloria Ruiz Alcala
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18016 Granada, Spain; (Y.J.-M.); (G.R.A.); (J.A.M.)
- Instituto de Investigación Biosanitaria ibs. GRANADA, University Hospitals of Granada-University of Granada, 18014 Granada, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18016 Granada, Spain; (Y.J.-M.); (G.R.A.); (J.A.M.)
- Instituto de Investigación Biosanitaria ibs. GRANADA, University Hospitals of Granada-University of Granada, 18014 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
- Research Unit “Modeling Nature” (MNat), University of Granada, 18016 Granada, Spain
| | - Anel Tarabayeva
- Department of General Immunology, Faculty of Medicine, Asfendyarov Kazakh National Medical University, Almaty 050012, Kazakhstan; (A.M.); (A.T.); (E.B.)
| | - Elmira Bitanova
- Department of General Immunology, Faculty of Medicine, Asfendyarov Kazakh National Medical University, Almaty 050012, Kazakhstan; (A.M.); (A.T.); (E.B.)
| | | | - Gordon J. McDougall
- Plant Biochemistry and Food Quality Group, Environmental and Biochemical Sciences Department, The James Hutton Institute, Invergowrie, Dundee DD2 5DA, UK;
| | - Nazira Bishimbayeva
- Research Center “Bioscience Technologies”, Almaty 050057, Kazakhstan
- Research Institute for Problems of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan
| | - Houria Boulaiz
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, University of Granada, 18016 Granada, Spain; (Y.J.-M.); (G.R.A.); (J.A.M.)
- Research Center “Bioscience Technologies”, Almaty 050057, Kazakhstan
- Instituto de Investigación Biosanitaria ibs. GRANADA, University Hospitals of Granada-University of Granada, 18014 Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| |
Collapse
|
150
|
Liu Z, Mao Y, Wang S, Zheng H, Yang K, Yang L, Huang P. A bibliometric and visual analysis of the impact of senescence on tumor immunotherapy. Front Immunol 2025; 16:1566227. [PMID: 40292294 PMCID: PMC12021824 DOI: 10.3389/fimmu.2025.1566227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Background Recently, many studies have focused on the relationship between senescence and immunotherapy in cancer treatment. However, relatively few studies have examined the intrinsic links between the three. Whether these studies can act synergistically in the fight against cancer and the specific links between them are still unclear. Methods We extracted, quantified, and visualized data from the literature (n = 2396) for the period 2004-2023 after rigorous quality control using citespace, GraphPad Prism, the R software package, and VOSviewer. Results Linear fit analyses were generated to predict the number of annual publications and citations as a function of the top-performing authors, journals, countries, and affiliations academically over the past two decades such as Weiwei, Aging-us, China, and the UT MD Anderson Cancer Center. Vosviewer-based hierarchical clustering further categorized study characteristics into six clusters, including two major clusters of immunotherapy research, immunosenescence-related research factors, and timeline distributions suggesting that cellular senescence and tumor progression is a relatively new research cluster that warrants further exploration and development. Study characterization bursts and linear regression analyses further confirmed these findings and revealed other important results, such as aging (a = 1.964, R² = 0.6803) and immunotherapy (a = 16.38, R² = 0.8812). Furthermore, gene frequency analysis in this study revealed the most abundant gene, APOE, and SIRT1-7 proteins. Conclusion The combination of aging therapies with tumor immunotherapies is currently in its preliminary stages. Although senescence has the greatest impact on ICB therapies, mechanistic investigations, and drug development for APOE and sirt1-7 (Sirtuins family) targets may be the key to combining senescence therapies with immunotherapies in the treatment of tumors.
Collapse
Affiliation(s)
- Zixu Liu
- Center for Evidence-Based Medicine, School of Public Health, Jiangxi Medical College. Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
- First Clinical Medicine School, Nanchang University, Nanchang, China
| | - Yuchen Mao
- First Clinical Medicine School, Nanchang University, Nanchang, China
| | - Shukai Wang
- First Clinical Medicine School, Nanchang University, Nanchang, China
| | - Haoyu Zheng
- School of Stomatology, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Kangping Yang
- Department of Gastroenterological Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Liang Yang
- Department of Gastroenterological Surgery, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Peng Huang
- Center for Evidence-Based Medicine, School of Public Health, Jiangxi Medical College. Nanchang University, Nanchang, China
- Jiangxi Province Key Laboratory of Preventive Medicine, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|