1
|
Binte Hanafi Z, Mei Y, Teo HY, Zhu Y, Yong Lionel CC, Chiu JW, Lu J, Liu H. Calpain 2 regulates IL-1α secretion and inhibits tumor development via modulating calpain 1 expression in the tumor microenvironment. Oncoimmunology 2025; 14:2451444. [PMID: 39803956 PMCID: PMC11730618 DOI: 10.1080/2162402x.2025.2451444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/26/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025] Open
Abstract
Tumor-promoting inflammation significantly impacts cancer progression, and targeting inflammatory cytokines has emerged as a promising therapeutic approach in clinical trials. Interleukin (IL)-1α, a member of the IL-1 cytokine family, plays a crucial role in both inflammation and carcinogenesis. How IL-1α is secreted in the tumor microenvironment has been poorly understood, and we previously showed that calpain 1 cleaves pro-IL-1α for mature IL-1α secretion, which exacerbates hepatocellular carcinoma by recruiting myeloid-derived suppressor cells. In this study, we report that calpain 2 also modulates IL-1α secretion. Notably, a deficiency in calpain 2 resulted in enhanced hepatocellular carcinoma development within an IL-1α-enriched tumor microenvironment. Further investigations revealed that calpain 2 deficiency increased calpain 1 expression, implying a compensatory mechanism between the two calpains. Mechanistically, calpain 2 deficiency led to increased expression of FoxO3, which is a forkhead transcription factor that promotes calpain 1 expression. Collectively, these results suggest that calpain 2 modulates calpain 1 expression, and therefore IL-1α secretion through the induction of FoxO3, offering novel potential therapeutic targets for cancer treatment.
Collapse
Affiliation(s)
- Zuhairah Binte Hanafi
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yu Mei
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Huey Yee Teo
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ying Zhu
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chew Chin Yong Lionel
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jing Wen Chiu
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jinhua Lu
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Haiyan Liu
- Immunology Programme, Life Sciences Institute; Centre for Life Sciences, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Programme and Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
2
|
Qi B, Wang Y, Zhu X, Gong Y, Jin J, Wu H, Man X, Liu F, Yao W, Gao J. miR-301a-mediated crosstalk between the Hedgehog and HIPPO/YAP signaling pathways promotes pancreatic cancer. Cancer Biol Ther 2025; 26:2457761. [PMID: 39846248 PMCID: PMC11760222 DOI: 10.1080/15384047.2025.2457761] [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: 06/04/2024] [Revised: 01/06/2025] [Accepted: 01/20/2025] [Indexed: 01/30/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) poses a significant challenge in oncology due to its dismal prognosis and limited therapeutic options. In this study, we investigated the role of miR-301a in facilitating crosstalk between the Hedgehog (Hh) and HIPPO/YAP signaling pathways during the progression of PDAC. Our findings revealed that miR-301a served as a central regulatory node, targeting Gli1 within the Hh pathway and STK4 within the HIPPO/YAP pathway. Immunohistochemical and molecular analyses confirmed dysregulation of pathway components in pancreatic cancer, underscoring the pivotal role of miR-301a. Functional assays demonstrated the impact of miR-301a on cell proliferation and apoptosis, particularly in synergy with TNF-α. Overall, our study elucidated the intricate interplay between the Hh and HIPPO/YAP pathways mediated by miR-301a, providing valuable insights into potential therapeutic strategies for intervening in PDAC.
Collapse
Affiliation(s)
- Bing Qi
- Institute of Oncology, Second Affiliated Hospital, Xi’an Medical University, Xi’an, China
| | - Yuqiong Wang
- Department of Gastroenterology, The Hospital of 92608 People’s Liberation Army of China (PLA) Troops, Shanghai, China
| | - Xian Zhu
- Department of Gastroenterology, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Yanfang Gong
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Jing Jin
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Hongyu Wu
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiaohua Man
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Feng Liu
- Department of Gastroenterology, Tongchuan People’s Hospital, Tongchuan, China
| | - Wenzhu Yao
- Institute of Oncology, Second Affiliated Hospital, Xi’an Medical University, Xi’an, China
| | - Jun Gao
- Institute of Oncology, Second Affiliated Hospital, Xi’an Medical University, Xi’an, China
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University, Shanghai, China
| |
Collapse
|
3
|
Chang HW, Frey G, Wang J, Liu H, Xing C, Chen J, Boyle WJ, Short JM. Preclinical development of ozuriftamab vedotin (BA3021), a novel ROR2-specific conditionally active biologic antibody-drug conjugate. MAbs 2025; 17:2490078. [PMID: 40202784 PMCID: PMC11988251 DOI: 10.1080/19420862.2025.2490078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/01/2025] [Accepted: 04/02/2025] [Indexed: 04/10/2025] Open
Abstract
Receptor tyrosine kinase-like orphan receptor (ROR2) has been identified as a highly relevant tumor-associated antigen in a variety of cancer indications of high unmet medical need, including melanoma, renal cell carcinoma, osteosarcoma, gastrointestinal stromal tumor, colorectal cancer, pancreatic ductal adenocarcinoma, and non-small cell lung cancer. Overexpression of ROR2 often correlates with advanced disease or poor prognosis, making it an attractive target for cancer therapy. We developed a novel, conditionally active biologic (CAB) antibody-drug conjugate (ADC), ozuriftamab vedotin (BA3021), which binds to ROR2 only in the acidic tumor microenvironment. In healthy tissue, binding to ROR2 is greatly reduced by a novel selection mechanism using physiological chemicals as protein-associated chemical switches (PaCS). The CAB anti-ROR2 ADC displays the anticipated binding properties and mediates potent lysis of ROR2-positive cancer cell lines. In vivo, BA3021 has potent and durable antitumor activity in human cancer xenograft mouse models, including patient-derived xenograft models. In non-human primates, BA3021 was well tolerated at doses of up to 10 mg/kg and showed excellent stability in vivo. These preclinical results indicate that CAB anti-ROR2 ADC is efficacious and well tolerated and may be a promising treatment for cancer patients with ROR2-expressing tumors.
Collapse
Affiliation(s)
| | - Gerhard Frey
- Research & Development, BioAtla Inc, San Diego, CA, USA
| | - Jing Wang
- Research & Development, BioAtla Inc, San Diego, CA, USA
| | - Haizhen Liu
- Research & Development, BioAtla Inc, San Diego, CA, USA
| | - Charles Xing
- Research & Development, BioAtla Inc, San Diego, CA, USA
| | - Jian Chen
- Research & Development, BioAtla Inc, San Diego, CA, USA
| | | | - Jay M. Short
- Research & Development, BioAtla Inc, San Diego, CA, USA
| |
Collapse
|
4
|
Liu S, Chen L, Shang Y. CEACAM5 exacerbates asthma by inducing ferroptosis and autophagy in airway epithelial cells through the JAK/STAT6-dependent pathway. Redox Rep 2025; 30:2444755. [PMID: 39844719 PMCID: PMC11758806 DOI: 10.1080/13510002.2024.2444755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
OBJECTIVES Asthma, a prevalent chronic disease, poses significant health threats and burdens healthcare systems. This study focused on the role of bronchial epithelial cells in asthma pathophysiology. METHODS Bioinformatics was used to identify key asthmarelated genes. An ovalbumin-sensitized mouse model and an IL-13-stimulated Beas-2B cell model were established for further investigation. RESULTS Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) was identified as a crucial gene in asthma. CEACAM5 expression was elevated in asthmatic mouse lung tissues and IL-13-stimulated Beas-2B cells, primarily in bronchial epithelial cells. CEACAM5 induced reactive oxygen species (ROS), lipid peroxidation, and ferroptosis. Interfering with CEACAM5 reduced ROS, malondialdehyde levels, and enhanced antioxidant capacity, while inhibiting iron accumulation and autophagy. Overexpression of CEACAM5 in IL-13-stimulated cells activated the JAK/STAT6 pathway, which was necessary for CEACAM5-induced autophagy, ROS accumulation, lipid peroxidation, and ferroptosis. CONCLUSION CEACAM5 promotes ferroptosis and autophagy in airway epithelial cells via the JAK/STAT6 pathway, exacerbating asthma symptoms. It represents a potential target for clinical treatment.
Collapse
Affiliation(s)
- Si Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Li Chen
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Yunxiao Shang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
5
|
Dewhirst MW. A translational review of hyperthermia biology. Int J Hyperthermia 2025; 42:2447952. [PMID: 39799944 DOI: 10.1080/02656736.2024.2447952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 01/15/2025] Open
Abstract
This review was written to be included in the Special Collection 'Therapy Ultrasound: Medicine's Swiss Army Knife?' The purpose of this review is to provide basic presentation and interpretation of the fundamentals of hyperthermia biology, as it pertains to uses of therapeutic ultrasound. The fundamentals are presented but in the setting of a translational interpretation and a view toward the future. Subjects that require future research and development are highlighted. The effects of hyperthermia are time and temperature dependent. Because intra-tumoral temperatures are non-uniform in tumors, one has to account for differential biologic effects in different parts of a tumor that occur simultaneously during and after hyperthermia.
Collapse
Affiliation(s)
- Mark W Dewhirst
- Gustavo S. Montana Distinguished Professor Emeritus of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
6
|
Guo S, Su Y, Li Y, Li C, Pan L. The causal relationship between circulating metabolites and gestational hypertension, pre-eclampsia, eclampsia: A bidirectional two-sample Mendelian randomization study. Clin Exp Hypertens 2025; 47:2508787. [PMID: 40392544 DOI: 10.1080/10641963.2025.2508787] [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: 02/04/2025] [Revised: 05/10/2025] [Accepted: 05/15/2025] [Indexed: 05/22/2025]
Abstract
OBJECTIVE Gestational hypertension, pre-eclampsia, and eclampsia pose significant risks to maternal and fetal health, yet their underlying causes remain unclear. This study investigates the associations between 233 metabolites and these conditions. METHODS We analyzed data from the Genome-Wide Association Studies (GWAS) database for gestational hypertension, pre-eclampsia, and eclampsia. The bidirectional two-sample MR analysis examined causal relationships using inverse variance weighting as the primary method, supplemented by MR-Egger, weighted median, simple mode, and weighted mode. Sensitivity analyses assessed robustness, heterogeneity, and horizontal pleiotropy. RESULTS In the forward Mendelian randomization analysis, a reduction in citrate levels (OR = 0.906, 95% CI = 0.829-0.990, p = .029) is associated with an increased risk of gestational hypertension. The ratio of conjugated linoleic acid to total fatty acids (OR = 1.172, 95% CI = 1.026-1.339, p = .019) is associated with an increased risk of gestational hypertension. The ratio of conjugated linoleic acid to total fatty acids (OR = 1.288, 95% CI = 1.064-1.560, p = .009) is associated with an increased risk of preeclampsia and eclampsia. The phospholipids to total lipids ratio in large HDL (OR = 1.227, 95% CI = 1.120-1.344, p = 9.91 × 10^-6) is associated with an increased risk of preeclampsia and eclampsia. The total cholesterol to total lipids ratio in chylomicrons and extremely large VLDL (OR = 0.884, 95% CI = 0.789-0.990, p = .033) is associated with an increased risk of preeclampsia and eclampsia. In the reverse Mendelian randomization analysis, the occurrence of gestational hypertension is associated with a reduction in Cholesteryl esters to total lipids ratio in very large VLDL (OR = 0.987, 95% CI = 0.975-0.999, p = .044). The occurrence of preeclampsia and eclampsia is associated with a reduction in total choline levels (OR = 0.989, 95% CI = 0.979-0.998, p = .029), and with a reduction in total phosphoglycerides levels (OR = 0.988, 95% CI = 0.978-0.997, p = .012). Sensitivity analysis did not detect significant heterogeneity or pleiotropy. CONCLUSION This research elucidates the causal links between specific metabolites and gestational hypertension, pre-eclampsia, and eclampsia, potentially informing new clinical approaches for diagnosis and treatment.
Collapse
Affiliation(s)
- Shi Guo
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yuting Su
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yajun Li
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Cuiyuan Li
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Lele Pan
- Department of Obstetrics and Gynecology, Guangdong Women and Children Hospital, Guangzhou, People's Republic of China
| |
Collapse
|
7
|
Sang-Ngern M, Fukuchi A, Kondratyuk TP, Park EJ, Simmons CJ, Wall MM, Lorch SE, Pezzuto JM, Chang LC. Biologically active withanolides from Physalis peruviana. PHARMACEUTICAL BIOLOGY 2025; 63:334-343. [PMID: 40285658 PMCID: PMC12035931 DOI: 10.1080/13880209.2025.2488136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/24/2024] [Accepted: 03/24/2025] [Indexed: 04/29/2025]
Abstract
CONTEXT Physalis peruviana L. (Solanaceae), also known as Poha, has been used in traditional medicine since pre-Columbian times, particularly in treating cancer. OBJECTIVE To study the chemical composition and potential medicinal properties of Poha. MATERIALS AND METHODS The fresh fruits and aerial parts of Poha were extracted. The isolation of extract yields a novel withanolide (physaperuvin K; 1) from the edible fruit, and seven withanolides (2-8), including a rare chlorinated withanolide (physalolactone; 2) from the aerial parts. Structure elucidation/determination was performed, some acetate derivatives were prepared (2a-6a), and the compounds were evaluated with in vitro assays indicative of anti-inflammatory activity. RESULTS The structure of 1 was elucidated through NMR spectroscopic analyses. The absolute configuration of compound 2 was determined using single-crystal X-ray diffraction. Compounds 1, 2, and 3 exhibited inhibition of tumor necrosis factor-α-induced nuclear factor-kappa B (NF-κB) activity with IC50 values of 10, 60, and 40 nM, respectively, without causing cytotoxicity at a concentration of 50 μM. Furthermore, compounds 1-3 reduced nitric oxide (NO) production in lipopolysaccharide-activated RAW 264.7 mouse macrophage cells with IC50 values ranging from 0.32 to 13.3 μM without overt cytotoxicity. Overall, acetylation did not significantly impact activity, except for compound 4, wherein the IC50 values in the NF-κB and NO assays were reduced from 11.0 to 0.33 μM, and 1.8 to 0.24 μM, respectively. CONCLUSIONS These findings enhance our understanding of Poha's constituents and potential medicinal properties. One of the most bioactive compounds identified in this study, physaperuvin K, is found in edible fruit.
Collapse
Affiliation(s)
- Mayuramas Sang-Ngern
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo, HI, United States
- School of Cosmetic Science, Mae Fah Luang University, Tasud, Muang District, Chiang Rai, Thailand
| | - Ashley Fukuchi
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo, HI, United States
| | - Tamara P. Kondratyuk
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo, HI, United States
| | - Eun-Jung Park
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo, HI, United States
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, United States
| | - Charles J. Simmons
- X-ray Diffraction Laboratory, Department of Chemistry, University of Hawai’i at Hilo, Hilo, HI, United States
| | - Marisa M. Wall
- Daniel K. Inouye U.S. Pacific Basin Agricultural Research Center, Hilo, HI, United States
| | - Sam E. Lorch
- Lani ko Honua Berry Farm, Hilo, HI, United States
| | - John M. Pezzuto
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo, HI, United States
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, United States
| | - Leng Chee Chang
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai’i at Hilo, Hilo, HI, United States
| |
Collapse
|
8
|
Sun S, Arif Aslam M, Ma EB, Lee G, Muhammad Ahmad Javaid H, Yoon S, Huh JY. Activation of CXCR7 exerts an inhibitory effect on adipogenesis through regulation of β-arrestin2/Wnt and AKT signalling. Adipocyte 2025; 14:2490258. [PMID: 40302245 PMCID: PMC12045560 DOI: 10.1080/21623945.2025.2490258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/22/2025] [Accepted: 03/29/2025] [Indexed: 05/02/2025] Open
Abstract
CXCR7, an alternative receptor for the inflammatory chemokine SDF-1, is involved in cell proliferation and migration. Recent studies have reported that CXCR7 also plays a role in adipose tissue. However, evidence regarding the role of CXCR7 and its ligands in adipocyte differentiation is limited. In this study, we aimed to elucidate changes in CXCR7 expression during adipocyte differentiation and the role of the SDF-1/CXCR7/CXCR4 axis in adipogenesis using recombinant SDF-1, the CXCR7 ligand CCX771, and small interfering RNAs. The results indicated that the levels of SDF-1 and its receptors, CXCR7 and CXCR4, decreased during the early stages of adipogenesis. Treatment with recombinant SDF-1 and CCX771 inhibited adipogenesis and lipid accumulation by inducing β-arrestin2, Wnt expression, and AKT phosphorylation and downregulating C/EBPα, PPARγ, and FABP4 expression. In contrast, knockdown of SDF-1 and CXCR7 in preadipocytes downregulated the β-arrestin2/Wnt and AKT pathway, leading to the induction of adipogenesis. Meanwhile, knockdown of CXCR4 had no significant effect. In mice, basal gene expression levels of SDF-1 and CXCR7 were higher in the stromal vascular fraction compared to mature adipocytes and were significantly upregulated by a high-fat diet. Our results provide new insights into the local role of the SDF-1-CXCR7 axis in adipocytes and offer additional benefits for the prevention of obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Shiyue Sun
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Muhammad Arif Aslam
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Eun Bi Ma
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Gahui Lee
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, Republic of Korea
| | - Hafiz Muhammad Ahmad Javaid
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Somy Yoon
- College of Pharmacy, Chonnam National University, Gwangju, Republic of Korea
| | - Joo Young Huh
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
- Department of Global Innovative Drugs, The Graduate School of Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
9
|
Meza-Torres J, Tinevez JY, Crouzols A, Mary H, Kim M, Hunault L, Chamorro-Rodriguez S, Lejal E, Altamirano-Silva P, Groussard D, Gobaa S, Peltier J, Chassaing B, Dupuy B. Clostridioides difficile binary toxin CDT induces biofilm-like persisting microcolonies. Gut Microbes 2025; 17:2444411. [PMID: 39719371 DOI: 10.1080/19490976.2024.2444411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/07/2024] [Accepted: 12/09/2024] [Indexed: 12/26/2024] Open
Abstract
Clinical symptoms of Clostridioides difficile infection (CDI) range from diarrhea to pseudomembranous colitis. A major challenge in managing CDI is the high rate of relapse. Several studies correlate the production of CDT binary toxin by clinical strains of C. difficile with higher relapse rates. Although the mechanism of action of CDT on host cells is known, its exact contribution to CDI is still unclear. To understand the physiological role of CDT during CDI, we established two hypoxic relevant intestinal models, Transwell and Microfluidic Intestine-on-Chip systems. Both were challenged with the epidemic strain UK1 CDT+ and its isogenic CDT- mutant. We report that CDT induces mucin-associated microcolonies that increase C. difficile colonization and display biofilm-like properties by enhancing C. difficile resistance to vancomycin. Importantly, biofilm-like microcolonies were also observed in the cecum and colon of infected mice. Hence, our study shows that CDT induces biofilm-like microcolonies, increasing C. difficile persistence and risk of relapse.
Collapse
Affiliation(s)
- Jazmin Meza-Torres
- Pathogenesis of Bacterial Anaerobes, Department of Microbiology, Institut Pasteur, Université Paris-Cité, UMR-CNRS 6047, Paris, France
| | - Jean-Yves Tinevez
- Image Analysis Hub, Department of Cell Biology and Infection, Institut Pasteur, Université Paris Cité, Paris, France
| | - Aline Crouzols
- Pathogenesis of Bacterial Anaerobes, Department of Microbiology, Institut Pasteur, Université Paris-Cité, UMR-CNRS 6047, Paris, France
| | - Héloïse Mary
- Biomaterials and Microfluidics Core Facility, Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Minhee Kim
- Biomaterials and Microfluidics Core Facility, Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Lise Hunault
- Antibodies in Therapy and Pathology, Department of Immunology, Institut Pasteur, Paris, France
| | - Susan Chamorro-Rodriguez
- Pathogenesis of Bacterial Anaerobes, Department of Microbiology, Institut Pasteur, Université Paris-Cité, UMR-CNRS 6047, Paris, France
| | - Emilie Lejal
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, Gif-sur-Yvette, France
| | - Pamela Altamirano-Silva
- Centro de Investigación en Enfermedades Tropicales, Universidad de Costa Rica, San José, Costa Rica
| | | | - Samy Gobaa
- Biomaterials and Microfluidics Core Facility, Department of Developmental and Stem Cell Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Johann Peltier
- Institute for Integrative Biology of the Cell (I2BC), Université Paris-Saclay, CEA, CNRS, Gif-sur-Yvette, France
| | - Benoit Chassaing
- Microbiome-Host Interactions, Department of Microbiology, Institut Pasteur, Université Paris Cité, INSERM U1306, Paris, France
- Mucosal Microbiota in Chronic Inflammatory Diseases, INSERM U1016, CNRS UMR 8104, Université Paris Cité, Paris, France
| | - Bruno Dupuy
- Pathogenesis of Bacterial Anaerobes, Department of Microbiology, Institut Pasteur, Université Paris-Cité, UMR-CNRS 6047, Paris, France
| |
Collapse
|
10
|
Mallya S, Gangadhar V, Aldrin SE, Acharya M, Kabekkodu SP, Kolathur KK, Chakrabarty S. Insights into the molecular and genetic role of obesity in breast cancer pathogenesis. Cancer Biol Ther 2025; 26:2501345. [PMID: 40353441 PMCID: PMC12077478 DOI: 10.1080/15384047.2025.2501345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 04/23/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025] Open
Abstract
The epidemic of obesity is a growing concern and is one of the major risk factors for several chronic diseases, including several types of cancers. The correlation of breast cancer with obesity has been extensively studied and involves an interplay of hormonal, metabolic, and genetic factors explored in this review. Inflammation and hormone dysregulation play an important role in promoting a protumorigenic environment through adipose tissue, which is involved in energy storage and functions as an endocrine organ. As a result, various cytokines, primarily proinflammatory in nature, are released, resulting in low-grade inflammation promoting tumor growth. Additionally, obese conditions also induce imbalances in hormones, particularly estrogen and insulin, both of which drive carcinogenesis. Genetic components such as single nucleotide polymorphisms also play critical roles in modulating the correlation between obesity and breast cancer. This review provides a comprehensive overview of various mechanisms underlying obesity and breast cancer incidence and progression.
Collapse
Affiliation(s)
- Sandeep Mallya
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Varsha Gangadhar
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Sophia Evangeline Aldrin
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Meghana Acharya
- Department of Bioinformatics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Kiran Kumar Kolathur
- Department of Pharmaceutical Biotechnology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, India
| | - Sanjiban Chakrabarty
- Department of Public Health and Genomics, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, India
| |
Collapse
|
11
|
Li D, Chu X, Liu W, Ma Y, Tian X, Yang Y. The regulatory roles of RNA-binding proteins in the tumour immune microenvironment of gastrointestinal malignancies. RNA Biol 2025; 22:1-14. [PMID: 39718205 DOI: 10.1080/15476286.2024.2440683] [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] [Revised: 11/03/2024] [Accepted: 12/04/2024] [Indexed: 12/25/2024] Open
Abstract
The crosstalk between the tumour immune microenvironment (TIME) and tumour cells promote immune evasion and resistance to immunotherapy in gastrointestinal (GI) tumours. Post-transcriptional regulation of genes is pivotal to GI tumours progression, and RNA-binding proteins (RBPs) serve as key regulators via their RNA-binding domains. RBPs may exhibit either anti-tumour or pro-tumour functions by influencing the TIME through the modulation of mRNAs and non-coding RNAs expression, as well as post-transcriptional modifications, primarily N6-methyladenosine (m6A). Aberrant regulation of RBPs, such as HuR and YBX1, typically enhances tumour immune escape and impacts prognosis of GI tumour patients. Further, while targeting RBPs offers a promising strategy for improving immunotherapy in GI cancers, the mechanisms by which RBPs regulate the TIME in these tumours remain poorly understood, and the therapeutic application is still in its early stages. This review summarizes current advances in exploring the roles of RBPs in regulating genes expression and their effect on the TIME of GI tumours, then providing theoretical insights for RBP-targeted cancer therapies.
Collapse
Affiliation(s)
- Dongqi Li
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| | - Xiangyu Chu
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Weikang Liu
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| | - Yongsu Ma
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| | - Xiaodong Tian
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| | - Yinmo Yang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| |
Collapse
|
12
|
Zhang Y, Fan W, Su F, Zhang X, Du Y, Li W, Gao Y, Hu W, Zhao J. Discussion on the mechanism of HER2 resistance in esophagogastric junction and gastric cancer in the era of immunotherapy. Hum Vaccin Immunother 2025; 21:2459458. [PMID: 39875210 PMCID: PMC11776468 DOI: 10.1080/21645515.2025.2459458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/11/2025] [Accepted: 01/24/2025] [Indexed: 01/30/2025] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is a critical biomarker and therapeutic target in gastric/gastroesophageal junction (G/GEJ) cancers, despite the initial success of HER2-targeted therapies, such as trastuzumab, resistance to these drugs has emerged as a major impediment to effective long-term treatment. This review examines the mechanisms of drug resistance in HER2-positive G/GEJ cancer, the primary mechanisms of resistance explored include alterations in the HER2 receptor itself, such as mutations and changes in expression levels, as well as downstream signaling pathways, and interactions with the tumor microenvironment (TME). Furthermore, the review discusses the Novel therapeutic approaches, including the use of antibody-drug conjugates (ADCs) and combination therapies are assessed for their potential to enhance outcomes. By integrating recent research findings and clinical trials, this review aims to provide oncologists and researchers with insights into developing more effective treatments for patients with drug-resistant HER2-positive G/GEJ cancer.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Graduate School, Changzhi Medical College, Changzhi, Shanxi, China
| | - Wenxuan Fan
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Graduate School, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Fei Su
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Graduate School, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoling Zhang
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Yunyi Du
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Weiling Li
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Graduate School, Changzhi Medical College, Changzhi, Shanxi, China
| | - Yangjun Gao
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Wenqing Hu
- Department of Gastrointestinal Surgery, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Jun Zhao
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| |
Collapse
|
13
|
Quan Z, Fan S, Zheng H, Ning Y, Yang Y. A pan-cancer analysis of MARCH8: molecular characteristics, clinical relevance, and immuno-oncology features. Cancer Biol Ther 2025; 26:2458773. [PMID: 39881438 PMCID: PMC11784653 DOI: 10.1080/15384047.2025.2458773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 01/31/2025] Open
Abstract
Membrane-associated RING-CH8 (MARCH8) is a member of the recently discovered MARCH family of ubiquitin ligases. MARCH8 has been shown to participate in immune responses. However, the role of MARCH8 in prognosis and immunology in human cancers remains largely unknown. The expression of MARCH8 protein was detected via immunohistochemistry in non-small cell lung cancer (NSCLC) and non-cancerous lung tissues. The study investigated the role of MARCH8 in tumor immunity through pan-cancer analysis of multiple databases. MARCH8 genetic alternations and expression were explored with the cBioPortal, GTEx, and TCGA databases. We investigated the role of MARCH8 expression in clinical relevance, prognosis, tumor immune microenvironment, immune checkpoint (ICP) with a series of bioinformatics tools and methods, such as TISIDB database, ESTIMATE, and CIBERSORT method. MARCH8 expression showed cancer-specific dysregulation and was associated with the prognosis of patients in various cancers. MARCH8 was related to the tumor microenvironment and participated in tumor immune regulation. Furthermore, low expression of MARCH8 was associated with poor prognosis and might serve as an independent prognostic biomarker for NSCLC patients. The comprehensive pan-cancer analysis revealed the potential of MARCH8 in tumor-targeted therapy, and suggested MARCH8 as a promising prognostic and immunological pan-cancer biomarker.
Collapse
Affiliation(s)
- Zihan Quan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hongmei Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yue Ning
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yang Yang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Pathology, The Fourth People’s Hospital of Longgang District, Shenzhen, Guangdong, China
| |
Collapse
|
14
|
Li G, Che X, Wang S, Liu D, Xie D, Jiang B, Zheng Z, Zheng X, Wu G. The role of cisplatin in modulating the tumor immune microenvironment and its combination therapy strategies: a new approach to enhance anti-tumor efficacy. Ann Med 2025; 57:2447403. [PMID: 39757995 PMCID: PMC11705547 DOI: 10.1080/07853890.2024.2447403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 05/27/2024] [Accepted: 11/23/2024] [Indexed: 01/07/2025] Open
Abstract
Cisplatin is a platinum-based drug that is frequently used to treat multiple tumors. The anti-tumor effect of cisplatin is closely related to the tumor immune microenvironment (TIME), which includes several immune cell types, such as the tumor-associated macrophages (TAMs), cytotoxic T-lymphocytes (CTLs), dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs), regulatory T cells (Tregs), and natural killer (NK) cells. The interaction between these immune cells can promote tumor survival and chemoresistance, and decrease the efficacy of cisplatin monotherapy. Therefore, various combination treatment strategies have been devised to enhance patient responsiveness to cisplatin therapy. Cisplatin can augment anti-tumor immune responses in combination with immune checkpoint blockers (such as PD-1/PD-L1 or CTLA4 inhibitors), lipid metabolism disruptors (like FASN inhibitors and SCD inhibitors) and nanoparticles (NPs), resulting in better outcomes. Exploring the interaction between cisplatin and the TIME will help identify potential therapeutic targets for improving the treatment outcomes in cancer patients.
Collapse
Affiliation(s)
- Guandu Li
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xiangyu Che
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Shijin Wang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Dequan Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Deqian Xie
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Bowen Jiang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zunwen Zheng
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xu Zheng
- Department of Cell Biology, College of Basic Medical Science, Dalian Medical University, Dalian, Liaoning, China
| | - Guangzhen Wu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
15
|
Chen P, He L, Chen K, Pan Q, Rong J, Mei Q, Sun P, Zhang C, Li D. Electron-donor planar extension based on donor-acceptor-donor molecules generates charge transfer-mediated J-aggregates for NIR-II photothermal immunotherapy and bacterial elimination. Biomaterials 2025; 323:123435. [PMID: 40450765 DOI: 10.1016/j.biomaterials.2025.123435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 05/06/2025] [Accepted: 05/24/2025] [Indexed: 06/11/2025]
Abstract
Second near-infrared photothermal therapy (NIR-II-PTT) has emerged as a promising therapeutic modality in diverse medical aspects. In particular, J-aggregate is a potential strategy to develop high-performance NIR-II-PTT materials, however, it suffers from specific molecular skeletons and complex control conditions. Herein, we presented a simply electron-donor planar extension strategy to develop donor-acceptor-donor (D-A-D) type small molecules with charge-transfer (CT)-mediated J-aggregation for NIR-II PTT. By integrating ring-fused thiophene units in a benzo[1,2-'c:4,5-c']bis[1,2,5]thiadiazole (BBTDT) scaffold, we enabled tunable electrostatic and π-π interactions, promoting CT-mediated J-aggregate formation. The resulting BDTT nanoparticles showed an impressive light-harvesting capability (ɛ1064) of 2.92 × 104 M-1 cm-1 under 1064 nm excitation, yielding an exceptional photothermal performance (ɛ1064 × PCE = 1.99 × 104), which surpasses those of reported D-A-D type NIR-II-absorbing small molecules. This outstanding NIR-II photothermal property induced sufficient immunogenic cell death and amplified the final ablation of deep-seated tumor in conjunction with programmed cell death protein 1 (PD-1). Meanwhile, the fabricated BDTT nanoparticles also facilitated hyperthermia-triggered bacterial death. Together, this study provides valuable insights into developing NIR-II-absorbing J-aggregates based on D-A-D type small molecules, and offers critical potentials to treat malignant tumors and prevents postsurgical infections.
Collapse
Affiliation(s)
- Pengfei Chen
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Liuliang He
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Kai Chen
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Qiyong Pan
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jie Rong
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Qunbo Mei
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts & Telecommunications, Nanjing, 210023, China
| | - Pengfei Sun
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Jiangsu Key Laboratory for Biosensors, Nanjing University of Posts & Telecommunications, Nanjing, 210023, China.
| | - Chi Zhang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Daifeng Li
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
16
|
Liu C, Wang P, Gao Y, Ma X, Su Y, Wei Y, Qiao R. CD39-Diannexin alleviates the platelet storage lesion by protecting platelets from activation, a new attempt from a traditional perspective. Platelets 2025; 36:2517108. [PMID: 40491155 DOI: 10.1080/09537104.2025.2517108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 05/05/2025] [Accepted: 06/02/2025] [Indexed: 06/11/2025]
Abstract
Due to platelet storage lesions (PSL), transfused platelets are unable to function properly in the prevention and treatment of bleeding in critically ill patients. It is a traditional assumption that PSL is closely related to platelet activation during storage because of the exposure of CD62P, phosphatidylserine (PS), etc. In this standpoint, activated platelets in vitro cannot be reactivated in vivo to exert their hemostatic function and exposed PS accelerates platelet clearance. Therefore, reducing platelet activation is helpful to alleviate PSL. Diannexin is the dimer of annexin that has a higher affinity for PS. CD39 is an ADP hydrolase produced by the vascular endothelium. As a result, we construct CD39-Diannexin (CD39-DA) fusion protein and hypothesize that CD39-DA can reduce platelet activation during storage to alleviate PSL. CD39-DA can bind to the exposed PS on the surface of stored platelets by immunofluorescence. Compared to the control groups, CD39-DA reserves part of stored platelets' aggregation function confirmed by platelet aggregation assay, induced by AA, ADP and collagen. Additionally, CD39-DA reduces lactic dehydrogenase (LDH) levels and CD62P-positive events after three-day storage. Interestingly, we preliminarily discover that CD39-DA may reduce stored platelets' apoptosis and increase aggregatory platelets after activation by thrombin, collagen and calcium, which is marked by GSAO. In conclusion, we confirm that CD39-DA can alleviate PSL by reducing platelet activation.
Collapse
Affiliation(s)
- Cheng Liu
- The Department of Laboratory Medicine, Peking University Third Hospital, Bei Jing, China
| | - Peng Wang
- Department of Transfusion, Peking University First Hospital, Bei Jing, China
| | - Yafei Gao
- The Department of Laboratory Medicine, Peking University Third Hospital, Bei Jing, China
| | - Xiaolong Ma
- The Department of Laboratory Medicine, Peking University Third Hospital, Bei Jing, China
| | - Yang Su
- The Department of Laboratory Medicine, Peking University Third Hospital, Bei Jing, China
| | - Yao Wei
- The Department of Laboratory Medicine, Peking University Third Hospital, Bei Jing, China
| | - Rui Qiao
- The Department of Laboratory Medicine, Peking University Third Hospital, Bei Jing, China
| |
Collapse
|
17
|
Zhou B, Wang W, Yu P, Yang Y, Mi D, Tian Y, Li Y, Zhu F. Expression and clinical significance of histamine receptors in pediatric AML. Hematology 2025; 30:2449319. [PMID: 39918973 DOI: 10.1080/16078454.2024.2449319] [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: 03/23/2024] [Accepted: 12/23/2024] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND This study investigates the expression and clinical significance of the histamine receptor family (HRs) in the bone marrow of children with newly diagnosed acute myeloid leukemia (AML). METHODS RNA sequencing was performed to assess the expression levels of HR family members (HRH1, HRH2, HRH3, and HRH4) in the bone marrow of 140 pediatric AML patients prior to chemotherapy. We compared the expression levels across various risk categories and assessed their relationship with prognosis using ROC curve analysis to evaluate predictive capabilities for outcomes. RESULTS Among the 140 AML patients in our center, those with different FAB subtypes showed varying overall survival (OS), event-free survival (EFS), and relapse-free survival (RFS). Specifically, the M2 and M4 subtypes showed better OS, EFS, and RFS, whereas the M5 subtype had poorer outcomes. Among patients with different fusion genes, those with AML1/ETO had superior OS, EFS, and RFS compared to other subtypes. Additionally, patients with CEBPA mutations demonstrated relatively favorable outcomes, whereas those with FLT3 mutations had poorer survival metrics. HRH1 expression was significantly higher in AML patients than in normal controls (P < 0.05). Patients in the high HRH1 expression group had significantly better EFS and RFS than those in the low expression group (P < 0.05). Furthermore, HRH1 expression was significantly higher in the low-risk (LR) group than in the intermediate and high-risk (IR & HR) groups (P < 0.05). This finding suggests that HRH1 may serve as an early predictor of risk, EFS, and RFS. CONCLUSION The clinical significance of HR family members varies in pediatric AML, with HRH1 identified as a valuable predictor of relapse in children with AML.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/diagnosis
- Child
- Male
- Female
- Child, Preschool
- Adolescent
- Receptors, Histamine/genetics
- Receptors, Histamine/metabolism
- Receptors, Histamine/biosynthesis
- Prognosis
- Infant
- Gene Expression Regulation, Leukemic
- Clinical Relevance
Collapse
Affiliation(s)
- Bi Zhou
- Department of Pediatrics, Suzhou Hospital of Anhui Medical University, Suzhou, People's Republic of China
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - WenPeng Wang
- Department of Pediatrics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Pao Yu
- Department of Pediatrics, Suzhou Hospital of Anhui Medical University, Suzhou, People's Republic of China
| | - Yang Yang
- Department of Oncology, Suzhou Hospital of Anhui Medical University, Suzhou, People's Republic of China
| | - DaWei Mi
- Department of Stomatology, Suzhou Hospital of Anhui Medical University, Suzhou, People's Republic of China
| | - YuanYuan Tian
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ying Li
- Department of Pediatrics, Suzhou Hospital of Anhui Medical University, Suzhou, People's Republic of China
| | - Feng Zhu
- Department of Pediatrics, Suzhou Hospital of Anhui Medical University, Suzhou, People's Republic of China
| |
Collapse
|
18
|
Chriqui LE, Cavin S, Perentes JY. Dual implication of endothelial adhesion molecules in tumor progression and cancer immunity. Cell Adh Migr 2025; 19:2472308. [PMID: 40071851 PMCID: PMC11913389 DOI: 10.1080/19336918.2025.2472308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 10/16/2024] [Accepted: 01/19/2025] [Indexed: 03/19/2025] Open
Abstract
Adhesion molecules are proteins expressed at the surface of various cell types. Their main contribution to immunity is to allow the infiltration of immune cells in an inflamed site. In cancer, adhesion molecules have been shown to promote tumor dissemination favoring the development of metastasis. While adhesion molecule inhibition approaches were unsuccessful for cancer control, their importance for the generation of an immune response alone or in combination with immunotherapies has gained interest over the past years. Currently, the balance of adhesion molecules for tumor promotion/inhibition is unclear. Here we review the role of selectins, intercellular adhesion molecules (ICAM) and vascular cell adhesion molecules (VCAM) from the perspective of the dual contribution of adhesion molecules in tumor progression and immunity.
Collapse
Affiliation(s)
- Louis-Emmanuel Chriqui
- Division of Thoracic Surgery, Department of Surgery, CHUV, Lausanne University Hospital, Lausanne, Switzerland
- Agora Cancer Research Center Lausanne, Lausanne, Switzerland
| | - Sabrina Cavin
- Division of Thoracic Surgery, Department of Surgery, CHUV, Lausanne University Hospital, Lausanne, Switzerland
- Agora Cancer Research Center Lausanne, Lausanne, Switzerland
| | - Jean Yannis Perentes
- Division of Thoracic Surgery, Department of Surgery, CHUV, Lausanne University Hospital, Lausanne, Switzerland
- Agora Cancer Research Center Lausanne, Lausanne, Switzerland
| |
Collapse
|
19
|
Xiang Z, Ma B, Pei X, Wang W, Gong W. Mechanism of action of genistein on breast cancer and differential effects of different age stages. PHARMACEUTICAL BIOLOGY 2025; 63:141-155. [PMID: 39996512 PMCID: PMC11864014 DOI: 10.1080/13880209.2025.2469607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/26/2025]
Abstract
CONTEXT Genistein, a soy-derived isoflavone, exhibits structural similarities with 17β-estradiol and demonstrates antioxidant, anti-inflammatory, and estrogenic properties. Despite its low bioavailability limiting its clinical application, it shows potential for breast cancer prevention and treatment. OBJECTIVE This review aims to summarize the pharmacological effects and molecular mechanisms of genistein in breast cancer, focusing on its therapeutic potential, strategies to overcome bioavailability limitations, and its role in personalized medicine. Differential impacts among population subgroups are also discussed. METHODS A systematic review was conducted using PubMed, ScienceDirect, and Google Scholar databases. Studies were selected based on their focus on genistein's mechanisms of action, strategies to enhance its bioavailability, and interactions with other therapies. RESULTS Genistein exerted anticancer effects by modulating estrogen receptor β (ERβ), inhibiting angiogenesis, arresting the cell cycle, and inducing apoptosis. Its antioxidant properties help mitigate tumor-associated oxidative stress. Bioavailability enhancement strategies, such as nanoparticle and lipid-based formulations, show promise. Age-dependent effects were evident, with distinct responses observed in prepubertal, menopausal, and postmenopausal populations, underscoring its potential for personalized therapies. Furthermore, genistein influences epigenetic modifications, including DNA methylation and miRNA expression, bolstering its anticancer efficacy. CONCLUSION Genistein is a promising candidate for breast cancer therapy, particularly for personalized treatment. Strategies to enhance bioavailability and further clinical research are essential to optimize its therapeutic potential and evaluate its efficacy in combination therapies.
Collapse
Affiliation(s)
- Zhebin Xiang
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Bo Ma
- Zhejiang Hospital, Hangzhou, China
| | - Xiujun Pei
- Shandong Provincial Hospital, Shandong, China
| | - Wenjie Wang
- Zhejiang Chinese Medical University, Hangzhou, China
| | - Weilun Gong
- Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
20
|
Li J, Lin L, Yang S, Mu Y, Zhang L, Ruan H, Xu J. Diagnostic value of CEACAM6 and HE4 in pleural fluid for malignant pleural effusion. Ann Med 2025; 57:2489748. [PMID: 40232301 PMCID: PMC12001857 DOI: 10.1080/07853890.2025.2489748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 01/27/2025] [Accepted: 03/29/2025] [Indexed: 04/16/2025] Open
Abstract
OBJECTIVE This study aimed to assess the diagnostic performance of carcinoembryonic antigen-related adhesion molecule 6 (CEACAM6) and human epididymis protein 4 (HE4) in pleural fluid for the detection of malignant pleural effusion (MPE). MATERIALS AND METHODS In this prospective study, pleural levels of CEACAM6 and HE4 were measured in two independent cohorts. The test cohort included 182 patients with exudative pleural effusions (123 malignant and 59 benign), and the validation cohort comprised 117 patients with exudative pleural effusions (65 malignant and 52 benign). Receiver operating characteristic (ROC) curves were used to assess the diagnostic performance of CEACAM6 and HE4 for MPE. RESULTS Both CEACAM6 and HE4 levels were significantly elevated in MPE compared to benign pleural effusion (BPE) in both cohorts (p < .001). In the test cohort, CEACAM6 and HE4 demonstrated areas under the curve (AUC) values of 0.862 and 0.826, respectively. The combination of CEACAM6 and HE4 yielded a higher AUC of 0.938 compared to either marker alone. In the validation cohort, both CEACAM6 (AUC = 0.811) and HE4 (AUC = 0.721), along with their combination (AUC = 0.834), exhibited strong diagnostic performance for MPE. Notably, in cytology-negative cases, the combination of CEACAM6 and HE4 also demonstrated a favourable diagnostic efficacy, with an AUC of 0.800. The addition of CEA to the CEACAM6/HE4 combination further improved the AUC to 0.819 (p = .04). CONCLUSIONS Pleural CEACAM6 and HE4 are promising biomarkers for distinguishing MPE from BPE. Their combination improves diagnostic accuracy, offering a valuable tool for MPE diagnosis, especially in challenging cases with cytology-negative pleural effusion.
Collapse
Affiliation(s)
- Jie Li
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, China
| | - Liyuan Lin
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, China
| | - Shengrui Yang
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, China
| | - Yuan Mu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, China
| | - Lixia Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, China
| | - Haoyu Ruan
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, China
| | - Jian Xu
- Department of Laboratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Branch of National Clinical Research Center for Laboratory Medicine, Nanjing, China
| |
Collapse
|
21
|
Batur A, Novak R, Salai G, Hrkač S, Ćosić V, Grgurević L. Extracellular vesicles in the pathogenesis and future diagnostics of oral squamous cell carcinoma. Future Sci OA 2025; 11:2461940. [PMID: 39920887 PMCID: PMC11812389 DOI: 10.1080/20565623.2025.2461940] [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: 09/11/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025] Open
Abstract
Extracellular vesicles are a group of heterogeneous particles secreted during both physiological and pathological conditions which serve in intercellular communication and play a role in the development and progression of oral squamous cell carcinoma, the most common malignant tumor of the head and neck with a high mortality rate. Extensive research is being conducted in order to determine the precise role of extracellular vesicles in oncogenic processes and to explore the possible application of extracellular vesicles as early tumor biomarkers. In this review, we aimed to systematize observed roles extracellular vesicles might play in organizing of tumor microenvironment, tumor invasion and metastasis, as well as the impact of extracellular vesicles on immune dysregulation and development of resistance to chemotherapeutics. Additionally, we summarized findings involving the potential use of extracellular vesicles cargo proteins as early disease biomarkers.
Collapse
Affiliation(s)
- Anđela Batur
- School of Dental Medicine, University of Zagreb, Zagreb, Croatia
| | - Ruđer Novak
- Center for Translational and Clinical Research, Department of Proteomics, University of Zagreb, School of Medicine, Zagreb, Croatia
- BIMIS – Biomedical Research Center Šalata, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Grgur Salai
- Department of Pulmonology, University Hospital Dubrava, Zagreb, Croatia
| | - Stela Hrkač
- Department of Clinical Immunology, Allergology and Rheumatology, University Hospital Dubrava, Zagreb, Croatia
| | - Vesna Ćosić
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Lovorka Grgurević
- Center for Translational and Clinical Research, Department of Proteomics, University of Zagreb, School of Medicine, Zagreb, Croatia
- BIMIS – Biomedical Research Center Šalata, University of Zagreb School of Medicine, Zagreb, Croatia
- Department of Anatomy, “Drago Perović”, University of Zagreb, School of Medicine, Zagreb, Croatia
| |
Collapse
|
22
|
Zhang D, Xing Y, Liu L, Zhang X, Ma C, Xu M, Li R, Wei H, Zhao Y, Xu B, Mei S. Prognostic signature based on mitochondria- and angiogenesis-related genes associated with immune microenvironment of multiple myeloma. Hematology 2025; 30:2456649. [PMID: 39873160 DOI: 10.1080/16078454.2025.2456649] [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: 08/12/2024] [Accepted: 01/14/2025] [Indexed: 01/30/2025] Open
Abstract
INTRODUCTION Mitochondria and angiogenesis play key roles in multiple myeloma (MM) development, but their interrelated genes affecting MM prognosis are under-studied. METHODS We analyzed TCGA_MMRF and GSE4581 datasets to identify four genes - CCNB1, CDC25C, HSP90AA1, and PARP1 - that significantly correlate with MM prognosis, with high expression indicating poor outcomes. RESULTS A prognostic signature based on these genes stratified patients into high- and low-risk groups, with the latter showing better survival. The signature was validated as an independent prognostic factor. Biological function analysis revealed differences in cell cycle processes between risk groups, and immune microenvironment analysis showed distinct immune cell infiltration patterns. CONCLUSION This mitochondria- and angiogenesis-related prognostic signature could enhance MM prognosis assessment and offer new therapeutic insights.
Collapse
Affiliation(s)
- Dai Zhang
- Department of Hematology, XuChang Central Hospital, XuChang, People's Republic of China
| | - Yu Xing
- Department of Hematology, XuChang Central Hospital, XuChang, People's Republic of China
| | - Lu Liu
- Department of Hematology, XuChang Central Hospital, XuChang, People's Republic of China
| | - Xiaoqing Zhang
- Department of Hematology, XuChang Central Hospital, XuChang, People's Republic of China
| | - Cong Ma
- Department of Hematology, XuChang Central Hospital, XuChang, People's Republic of China
| | - MengYao Xu
- Department of Hematology, XuChang Central Hospital, XuChang, People's Republic of China
| | - Ruiqi Li
- Department of Hematology, XuChang Central Hospital, XuChang, People's Republic of China
| | - HanJing Wei
- Research Center for Clinical Medical Sciences, XuChang Central Hospital, XuChang, People's Republic of China
- Henan Provincial Health Commission Key Laboratory of Precision Medicine, XuChang, People's Republic of China
| | - Yan Zhao
- Research Center for Clinical Medical Sciences, XuChang Central Hospital, XuChang, People's Republic of China
- Henan Provincial Health Commission Key Laboratory of Precision Medicine, XuChang, People's Republic of China
| | - Bingxin Xu
- Research Center for Clinical Medical Sciences, XuChang Central Hospital, XuChang, People's Republic of China
- Henan Provincial Health Commission Key Laboratory of Precision Medicine, XuChang, People's Republic of China
| | - Shuhao Mei
- Department of Hematology, XuChang Central Hospital, XuChang, People's Republic of China
- Henan Provincial Health Commission Key Laboratory of Precision Medicine, XuChang, People's Republic of China
- XuChang Key Laboratory of Hematology, XuChang, People's Republic of China
| |
Collapse
|
23
|
Li M, Li D, Wang HY, Zhang W, Zhuo Z, Guo H, Liu J, Zhuo Y, Tang J, He J, Miao L. Leptin decreases Th17/Treg ratio to facilitate neuroblastoma via inhibiting long-chain fatty acid catabolism in tumor cells. Oncoimmunology 2025; 14:2460281. [PMID: 39902867 PMCID: PMC11796542 DOI: 10.1080/2162402x.2025.2460281] [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: 11/04/2024] [Revised: 01/05/2025] [Accepted: 01/24/2025] [Indexed: 02/06/2025] Open
Abstract
The exploration of therapeutic targets in neuroblastoma (NB), which needs more attempts, can benefit patients with high-risk NB. Based on metabolomic and transcriptomic data in mediastinal NB tissues, we found that the content of long-chain acylcarnitine (LCAC) was increased and positively associated with leptin expression in advanced NB. Leptin over-expression forced naïve CD4+ T cells to differentiate into Treg cells instead of Th17 cells, which benefited from NB cell proliferation, migration, and drug resistance. Mechanically, leptin in NB cells blunted the activity of carnitine palmitoyltransferase 2 (CPT2), the key enzyme for LCAC catabolism, by inhibiting sirtuin 3-mediated CPT2 deacetylation, which depresses oxidative phosphorylation (OXPHOS) for energy supply and increases lactic acid (LA) production from glycolysis to modulate CD4+ T cell differentiation. These findings highlight that excess leptin contributes to lipid metabolism dysfunction in NB cells and subsequently misdirects CD4+ T cell differentiation in tumor micro-environment (TME), indicating that targeting leptin could be a therapeutic strategy for retarding NB progression.
Collapse
Affiliation(s)
- Meng Li
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Di Li
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Hai-Yun Wang
- Department of Pathology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, National Children’s Medical Center for South Central Region, Guangzhou, Guangdong, China
| | - Weixin Zhang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Zhenjian Zhuo
- Laboratory Animal Center, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, Shenzhen, China
| | - Huiqin Guo
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Jiabin Liu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Yue Zhuo
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jue Tang
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| | - Lei Miao
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, Guangdong, China
| |
Collapse
|
24
|
Kang Z, Zhang L, Yang Z. Role of non-coding RNAs in the pathogenesis of viral myocarditis. Virulence 2025; 16:2466480. [PMID: 39950847 PMCID: PMC11849450 DOI: 10.1080/21505594.2025.2466480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 01/04/2025] [Accepted: 02/09/2025] [Indexed: 02/21/2025] Open
Abstract
Viral myocarditis (VMC) is a common inflammatory disease of the myocardium that is characterized mainly by inflammatory cell infiltration and cardiomyocyte necrosis. Coxsackievirus B3 (CVB3) is a common cause of VMC, although major progress has been made in the treatment of VMC, the long-term prognosis is still not ideal and further research is needed. Non-coding RNAs (ncRNAs) are RNA molecules without coding functions and include microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), which play extensive regulatory roles in gene expression; however, their mechanisms of action in CVB3-induced VMC remain incompletely understood. Here, we review the currently known roles of various ncRNAs in CVB3-induced VMC models, with a focus on cell death, inflammation and viral replication, with the aim of providing a reference for their therapeutic or vaccine development for the treatment of VMC.
Collapse
Affiliation(s)
- Zhijuan Kang
- Department of Nephrology, Rheumatology and Immunology, The Affiliated Children’s Hospital of Xiangya School of Medicine, Central South University (Hunan children’s hospital), Changsha, Hunan, China
- Department of Pediatrics, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Liang Zhang
- Department of Nephrology, Rheumatology and Immunology, The Affiliated Children’s Hospital of Xiangya School of Medicine, Central South University (Hunan children’s hospital), Changsha, Hunan, China
| | - Zuocheng Yang
- Department of Pediatrics, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
25
|
Ulhe A, Raina P, Chaudhary A, Kaul-Ghanekar R. Alpha-linolenic acid-mediated epigenetic reprogramming of cervical cancer cell lines. Epigenetics 2025; 20:2451551. [PMID: 39895102 PMCID: PMC11792827 DOI: 10.1080/15592294.2025.2451551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 11/25/2024] [Accepted: 01/02/2025] [Indexed: 02/04/2025] Open
Abstract
Cervical cancer, the fourth most common cancer globally and the second most prevalent cancer among women in India, is primarily caused by Human Papilloma Virus (HPV). The association of diet with cancer etiology and prevention has been well established and nutrition has been shown to regulate cancer through modulation of epigenetic markers. Dietary fatty acids, especially omega-3, reduce the risk of cancer by preventing or reversing the progression through a variety of cellular targets, including epigenetic regulation. In this work, we have evaluated the potential of ALA (α linolenic acid), an ω-3 fatty acid, to regulate cervical cancer through epigenetic mechanisms. The effect of ALA was evaluated on the regulation of histone deacetylases1, DNA methyltransferases 1, and 3b, and global DNA methylation by ELISA. RT-PCR was utilized to assess the expression of tumor regulatory genes (hTERT, DAPK, RARβ, and CDH1) and their promoter methylation in HeLa (HPV18-positive), SiHa (HPV16-positive) and C33a (HPV-negative) cervical cancer cell lines. ALA increased DNA demethylase, HMTs, and HATs while decreasing global DNA methylation, DNMT, HDMs, and HDACs mRNA expression/activity in all cervical cancer cell lines. ALA downregulated hTERT oncogene while upregulating the mRNA expression of TSGs (Tumor Suppressor Genes) CDH1, RARβ, and DAPK in all the cell lines. ALA reduced methylation in the 5' CpG island of CDH1, RARβ, and DAPK1 promoters and reduced global DNA methylation in cervical cancer cell lines. These results suggest that ALA regulates the growth of cervical cancer cells by targeting epigenetic markers, shedding light on its potential therapeutic role in cervical cancer management.
Collapse
Affiliation(s)
- Amrita Ulhe
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Prerna Raina
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
- Analytical Department (ADT), Lupin Limited, Pune, India
| | - Amol Chaudhary
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Ruchika Kaul-Ghanekar
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
- Symbiosis Centre for Research and Innovation (SCRI); Symbiosis International Deemed University (SIU), Pune, India
- Cancer Research Lab, Symbiosis School of Biological Sciences (SSBS), Symbiosis International Deemed University (SIU), Pune, India
| |
Collapse
|
26
|
Lv Y, Li Y, Zhou J, Liu X, Wang D, Wang D, Tong D, Wang S, An H, Kang X. Exosomal miR-122-5p for regulation of secretory functions of fibroblasts and promotion of breast cancer metastasis by targeting MKP-2: an experimental study. Cancer Biol Ther 2025; 26:2500104. [PMID: 40320567 PMCID: PMC12051585 DOI: 10.1080/15384047.2025.2500104] [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: 08/28/2022] [Revised: 12/14/2024] [Accepted: 04/27/2025] [Indexed: 05/08/2025] Open
Abstract
Tumor metastasis is a major obstacle for the effective treatment of breast cancer. Some studies showed that exosomes could promote tumor distant metastasis by establishing pre-metastasis niches (PMN). MicroRNAs (miRNAs) in exosomes play a critical role in tumor development and invasion. We aimed to investigate the effects of exosomal miRNAs derived from breast cancer cells on metastasis. MiRNA sequencing and RT-PCR approach were used to screen potential exosomal miRNAs. We compared the levels of serum exosomal miRNAs from breast cancer patients and those from MCF10A/MCF7/MDA-MB-231 cells. We found that differential exosomal miRNAs screened from patients with metastasis have higher expression levels in exosomes secreted by MDA-MB-231 cells. Using miRNA mimics or inhibitors, exosomal miR-122-5p was found to enhance the secretion levels of chemokine MCP-1 and SDF-1 from WI-38 lung fibroblast cells. In vitro luciferase assay and western blot confirmed the targeting of 3'-untranslated region of MKP-2 and suppression of MKP-2 expression by miR-122-5p in WI-38 cells. Treatment of xenograft mice with exosomal miR-122-5p increased the levels of MCP-1 and SDF-1 in serum, and promoted lung metastasis of breast cancer. In conclusion, we identified exosomal miR-122-5p from breast cancer cells that could promote the chemokine secretion of lung fibroblasts, which might facilitate the chemotaxis and colonization of breast cancer cells in lung tissue.
Collapse
Affiliation(s)
- Yun Lv
- Department of Medical Oncology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yue Li
- Department of Medical Oncology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jie Zhou
- Department of Medical Oncology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xin Liu
- Department of Medical Oncology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Dandan Wang
- Department of Medical Oncology, Heze Municipal Hospital, Heze, China
| | - Dongmei Wang
- Department of Ultrasonography, Xiang’an Hospital of Xiamen University, Xiamen, China
| | - Dandan Tong
- School of medicine, Huaqiao University, Quanzhou, China
| | - Shuhuai Wang
- Department of Pathology, Cancer Hospital of Harbin Medical University, Harbin, China
| | - Hanxiang An
- Department of Medical Oncology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xinmei Kang
- Department of Medical Oncology, Xiang’an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
27
|
Lou C, Cai X. The emerging roles of platelet-derived extracellular vesicles in disease. Ann Med 2025; 57:2499029. [PMID: 40317251 PMCID: PMC12054590 DOI: 10.1080/07853890.2025.2499029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 04/12/2025] [Accepted: 04/18/2025] [Indexed: 05/07/2025] Open
Abstract
INTRODUCTION Platelet-derived extracellular vesicles (pEVs) are nanoscale, membrane-bound vesicles released by platelets during activation or apoptosis. They contain various bioactive and non-bioactive molecules and play significant roles in numerous physiological and pathological processes through intercellular communication, thus attracting growing attention in biomedical research. METHODS This review comprehensively overviews the biogenesis, clearance, and molecular characteristics of pEVs. It also covers current methodologies for their isolation and characterization. The therapeutic implications of pEVs in key clinical settings like tissue regeneration, hemostasis, immune modulation, and vascular repair, with a focus on cancer progression, wound healing, and hemorrhagic shock management, are explored. Their role in cellular signal transduction is examined, and their functional properties are compared with other platelet-derived products such as platelet-rich plasma. RESULTS pEVs show potential as both therapeutic agents and diagnostic biomarkers. They are involved in modulating inflammatory responses, promoting angiogenesis, and enhancing cellular repair mechanisms. CONCLUSION Future research should concentrate on optimizing their therapeutic efficacy, refining biomarker applications, and exploring targeted delivery strategies to fully utilize their potential in regenerative medicine, oncology, and hemostasis management.
Collapse
Affiliation(s)
- Can Lou
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Blood Transfusion, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaohong Cai
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Blood Transfusion, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
28
|
Lin J, Lin Z, Chen Y, Wang X, Huang Y, Zhang H, Zhu L, Xu Z, Gao X, Zhang Y, Lan B, Chen Y. Differences of clinical features, prognosis and genetic mutations in Chinese patients with malignant melanoma and additional primary tumours. Ann Med 2025; 57:2493769. [PMID: 40317239 PMCID: PMC12051608 DOI: 10.1080/07853890.2025.2493769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/09/2025] [Accepted: 04/08/2025] [Indexed: 05/07/2025] Open
Abstract
BACKGROUND The differences in the clinical features, prognosis and genetic mutations in Chinese patients with malignant melanoma (MM) and additional primary tumours remain unclear. METHODS A retrospective analysis was conducted on patients with malignancies in Fujian Cancer Hospital from January 2007 to September 2022, end follow-up in September 2023. Clinical data were gathered, survival analysis was performed, and genetic mutations were detected. RESULTS There were 58 of 1223 melanoma patients with melanoma and additional primary tumours, an incidence of 4.74%. Acral MM was the most common subtype (26/58), 23 (39.66%) patients had concomitant digestive tumours. Patients who had MM as their first primary tumour (MMFP) had shorter tumour occurrence intervals (9.93 vs. 57.78 months, p = .008) but longer melanoma survival (MM-OS) than the non-MMFP group (100.43 vs. 18.93 months, p = .015). Patients with cancer family histories were more likely to have pathogenic and likely pathogenic (P/LP) mutations (2/5 vs. 4/25). The somatic BRAF gene mutation was frequently observed in MM tissue (8/19, 42.11%). Three patients had whole-genome doubling and microsatellite instability-high (MSI-H). The COSMIC2 signature 3 was significantly higher in the P/LP group. CONCLUSIONS The frequency of MM and additional primary tumours is about 5% in Chinese populations. Patients with melanoma diagnosed first have longer melanoma survival. Digestive system tumours were the most concomitant; a digestive examination is advisable, especially for those with an expected overall survival (OS) greater than 10 months. Meanwhile, patient's family cancer history should be followed up in detail, along with completion of germline P/LP mutation and somatic mutation testing, all of which may provide valuable support for further treatment.
Collapse
Affiliation(s)
- Jing Lin
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Cancer Bio-Immunotherapy Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Zhongqiao Lin
- Phase I Clinical Trial Ward, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yanping Chen
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Xuefeng Wang
- Innovation Center for Cancer Research, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yufang Huang
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Cancer Bio-Immunotherapy Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Huishan Zhang
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Cancer Bio-Immunotherapy Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Li Zhu
- Clinical Oncology School of Fujian Medical University, Fuzhou, China
| | - Zelong Xu
- Geneplus-Beijing Institute, Beijing, China
| | - Xuan Gao
- GenePlus-Shenzhen Clinical Laboratory, Shenzhen, China
| | | | - Bin Lan
- Laboratory of Radiation Oncology and Radiobiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yu Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Cancer Bio-Immunotherapy Center, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| |
Collapse
|
29
|
Xu Y, Li J, Ji X, Chen Q, Liu Z, Ji S. Lymphocyte-to-C-reactive protein ratio predicts prognosis in unresectable locally advanced non-small cell lung cancer patients. Ann Med 2025; 57:2487629. [PMID: 40178370 PMCID: PMC11980205 DOI: 10.1080/07853890.2025.2487629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 03/09/2025] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND The lymphocyte-to-C-reactive protein ratio (LCR) is a promising inflammation-based tool for assessing the status of patients with malignant tumours. This study evaluated the ability of LCR to predict the prognosis of patients with unresectable locally advanced non-small cell lung cancer (LA-NSCLC) after chemoradiotherapy. METHODS We retrospectively investigated 206 consecutive patients with unresectable LA-NSCLC who underwent chemoradiotherapy between January 2016 and November 2019. The LCR was calculated from the differential count by dividing the absolute lymphocyte count by the C-reactive protein level. The optimal cut-off value of LCR was determined using the receiver operating characteristic (ROC) curve, and the enrolled patients were divided into two groups for further analysis according to LCR. Overall survival (OS) and disease-free survival (DFS) were assessed using univariate and multivariate Cox regression analyses. RESULTS In patients with unresectable LA-NSCLC, the level of LCR was significantly associated with pathology (p = 0.042) and TNM stage (p = 0.002). High LCR and low LCR patients had different distinct outcomes (median OS: 36 vs. 34 months, p < 0.0001) and recurrence risk (median DFS: 31 vs. 23 months, p < 0.001). Univariate analysis indicated that Eastern Cooperative Oncology Group (ECOG) performance status, TNM stage, CEA level, response, neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-monocyte ratio (LMR), systemic immune inflammation index (SII), and LCR were predictors of OS and DFS. Multivariate analysis showed that a high LCR was an independent prognostic factor for OS (hazard ratio [HR], 0.526; 95% CI, 0.364-0.762; p = 0.001) and DFS (HR, 0.390; 95% CI, 0.275-0.554; p < 0.001). CONCLUSION LCR is a promising prognostic index in patients with LA-NSCLC undergoing chemoradiotherapy, and an increase in the LCR level contributes to better outcomes.
Collapse
Affiliation(s)
- Yingying Xu
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University Suzhou, Suzhou, China
| | - Jinping Li
- Department of Gastroenterology, Fangzi People’s Hospital, Weifang, China
| | - Xiang Ji
- Department of Gastroenterology, Fangzi People’s Hospital, Weifang, China
| | - Qingqing Chen
- Department of Radiotherapy and Oncology, The affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, China
| | - Zhengcao Liu
- Department of Radiotherapy and Oncology, The affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, China
| | - Shengjun Ji
- Department of Radiotherapy and Oncology, The affiliated Suzhou Hospital of Nanjing Medical University, Gusu School, Nanjing Medical University, Suzhou, China
| |
Collapse
|
30
|
Thulasinathan B, Suvilesh KN, Maram S, Grossmann E, Ghouri Y, Teixeiro EP, Chan J, Kaif JT, Rachagani S. The impact of gut microbial short-chain fatty acids on colorectal cancer development and prevention. Gut Microbes 2025; 17:2483780. [PMID: 40189834 PMCID: PMC11980463 DOI: 10.1080/19490976.2025.2483780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/18/2025] [Accepted: 03/18/2025] [Indexed: 04/11/2025] Open
Abstract
Cancer is a long-term illness that involves an imbalance in cellular and immune functions. It can be caused by a range of factors, including exposure to environmental carcinogens, poor diet, infections, and genetic alterations. Maintaining a healthy gut microbiome is crucial for overall health, and short-chain fatty acids (SCFAs) produced by gut microbiota play a vital role in this process. Recent research has established that alterations in the gut microbiome led to decreased production of SCFA's in lumen of the colon, which associated with changes in the intestinal epithelial barrier function, and immunity, are closely linked to colorectal cancer (CRC) development and its progression. SCFAs influence cancer progression by modifying epigenetic mechanisms such as DNA methylation, histone modifications, and non-coding RNA functions thereby affecting tumor initiation and metastasis. This suggests that restoring SCFA levels in colon through microbiota modulation could serve as an innovative strategy for CRC prevention and treatment. This review highlights the critical relationship between gut microbiota and CRC, emphasizing the potential of targeting SCFAs to enhance gut health and reduce CRC risk.
Collapse
Affiliation(s)
- Boobalan Thulasinathan
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
| | - Kanve N. Suvilesh
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
| | - Sumanas Maram
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
| | - Erik Grossmann
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Department of Medicine, Digestive Centre, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
| | - Yezaz Ghouri
- Department of Medicine, Digestive Centre, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
| | - Emma Pernas Teixeiro
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO, USA
| | - Joshua Chan
- Chemical and Biological Engineering, Colorado State University, Fort Collins, CO, USA
| | - Jussuf T. Kaif
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
- Siteman Cancer Centre, Washington University, St. Louis, MO, USA
| | - Satyanarayana Rachagani
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, USA
- Roy Blunt NextGen Precision Health Institute, University of Missouri, Columbia, MO, USA
- Department of Surgery, Ellis Fischel Cancer Centre, University of Missouri, Columbia, MO, USA
- Harry S. Truman Memorial Veterans’ Hospital, Columbia, MO, USA
- Siteman Cancer Centre, Washington University, St. Louis, MO, USA
| |
Collapse
|
31
|
Wu KP, Li QQ, Luo XQ, Wang XX, Lai YZ, Tian D, Yang HC, Wei XL, Wang LY, Li QM, Zhu D, Chen SJ, Li YS. Chemoimmunotherapy as induction treatment in concurrent chemoradiotherapy for patients with nasopharyngeal carcinoma stage IVa. Ann Med 2025; 57:2453091. [PMID: 39834281 PMCID: PMC11753007 DOI: 10.1080/07853890.2025.2453091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/11/2024] [Accepted: 12/29/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Chemoimmunotherapy is the first-line therapy for patients with recurrent or metastatic nasopharyngeal carcinoma (NPC) and is currently the main induction treatment option for patients with locoregionally advanced NPC. However, it remains unclear whether combining immunotherapy with standard induction chemotherapy enhances its efficacy. This study aimed to evaluate the efficacy, toxicity, and survival outcomes of induction chemoimmunotherapy in patients with locoregionally advanced NPC. METHODS This study analyzed 50 patients with stage IVa NPC between January 2020 and December 2023 in our hospital. Among them, 23 received induction chemoimmunotherapy, and 27 received induction chemotherapy. All patients underwent standard platinum-based concurrent intensity-modulated radiation therapy. We compared tumor response and toxicity during induction treatment and concurrent chemoradiotherapy (CCRT) between the two groups. RESULTS The objective and complete response rates were significantly higher in the induction chemoimmunotherapy group compared to the induction chemotherapy group (95.7% vs 77.8%, and 39.1% vs 22.2%, respectively). All patients completed radical CCRT. Median follow-up was 24 months. Patients who received induction chemoimmunotherapy had longer event-free survival (EFS) compared to those who received induction chemotherapy (p = 0.029, Hazard Ratio and 95%confidence interval [CI]: 0.24 [0.07-0.85]). The 24-month EFS was higher in the induction chemoimmunotherapy group compared with the chemotherapy group (24-month EFS rates and 95%CI: 88.9% [95%CI: 68.3%-100%] vs 62.6% [95%CI: 43.1%-82.1%]). No significant differences in adverse events were observed between the two groups during induction treatment and CCRT. CONCLUSIONS Adding immunotherapy to induction chemotherapy may be an effective and safe choice for treating patients with stage IVa NPC.
Collapse
Affiliation(s)
- Kun-Peng Wu
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
- Heyuan Key Laboratory of Molecular Diagnosis & Disease Prevention and Treatment, Doctors Station of Guangdong Province, Heyuan People’s Hospital, Heyuan, Guangdong, China
| | - Qing-Qing Li
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
- Heyuan Key Laboratory of Molecular Diagnosis & Disease Prevention and Treatment, Doctors Station of Guangdong Province, Heyuan People’s Hospital, Heyuan, Guangdong, China
| | - Xu-Qiang Luo
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
- Heyuan Key Laboratory of Molecular Diagnosis & Disease Prevention and Treatment, Doctors Station of Guangdong Province, Heyuan People’s Hospital, Heyuan, Guangdong, China
| | - Xiao-Xi Wang
- Department of Otolaryngology-Head and Neck Surgery, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yan-Zhen Lai
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
- Heyuan Key Laboratory of Molecular Diagnosis & Disease Prevention and Treatment, Doctors Station of Guangdong Province, Heyuan People’s Hospital, Heyuan, Guangdong, China
| | - Dan Tian
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
- Heyuan Key Laboratory of Molecular Diagnosis & Disease Prevention and Treatment, Doctors Station of Guangdong Province, Heyuan People’s Hospital, Heyuan, Guangdong, China
| | - Hong-cheng Yang
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
- Heyuan Key Laboratory of Molecular Diagnosis & Disease Prevention and Treatment, Doctors Station of Guangdong Province, Heyuan People’s Hospital, Heyuan, Guangdong, China
| | - Xue-Ling Wei
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
| | - Lan-Ying Wang
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
| | - Qiu-Ming Li
- Department of Otolaryngology-Head and Neck Surgery, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
| | - Dao Zhu
- Department of Otolaryngology-Head and Neck Surgery, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
| | - Si-Jie Chen
- Department of Otolaryngology-Head and Neck Surgery, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
| | - Yang-Si Li
- Department of Oncology, Heyuan People’s Hospital, Guangdong Provincial People’s Hospital Heyuan Hospital, Heyuan, Guangdong, China
- Heyuan Key Laboratory of Molecular Diagnosis & Disease Prevention and Treatment, Doctors Station of Guangdong Province, Heyuan People’s Hospital, Heyuan, Guangdong, China
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
32
|
Tang J, Wang M, Li H, Liang Q, Zhang Z, Liu H, Sun J, Ma LJ. Simply structural, reversible and chemical stable ratiometric pH-responsive fluorescence probe of pyrene derivatives and its applications. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 338:126205. [PMID: 40222233 DOI: 10.1016/j.saa.2025.126205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/02/2025] [Accepted: 04/07/2025] [Indexed: 04/15/2025]
Abstract
The pyrene group is commonly used as a fluorophore in ratiometric fluorescence probes. However, these probes almost exclusively utilize the monomer/excimer of the pyrene group to achieve ratiometric fluorescence recognition for analytes. In the study, three structurally simple, mono-substituted pyrene derivatives, namely pyrene-1-ol (Probe1), pyren-1-ylboronic acid (Probe2), pyrene-1-carboxylic acid (Probe3) utilize their protonation or deprotonation effects to influence the electronic excited state of the pyrene group, thereby achieving sensitive ratiometric fluorescence recognition of pH. Among them, Probe1 and Probe2 have been verified to function in alkaline environments with pH values ranging from 8.1 to 12.1 and 9.4 to 13.1, respectively, with pKa values of 8.9 and 11.1. Meanwhile, Probe3 performs optimally in weak acidic conditions with a pH range of 2.3-5.2, having a pKa value of 4.8. Additionally, three fluorescence probes exhibit excellent photo-stability, recyclability, and resistance to ionic interference. Furthermore, the three probes can also achieve ratiometric fluorescence detection of pH in environmental water sources such as tap water and Pearl River water, as well as fluorescence response to pH in living GS cells of epinephelus coioides.
Collapse
Affiliation(s)
- Jiawei Tang
- School of Chemistry, South China Normal University, Guangzhou 510006, PR China; Key Laboratory of Analytical Chemistry for Biomedicine, South China Normal University, Guangzhou 510006, PR China; Key Laboratory of Theoretical Chemistry of Environment Ministry of Education, South China Normal University, Guangzhou 510631, PR China
| | - Muxi Wang
- School of Chemistry, South China Normal University, Guangzhou 510006, PR China
| | - Hongming Li
- School of Chemistry, South China Normal University, Guangzhou 510006, PR China
| | - Qingjian Liang
- School of Fishery, Zhejiang Ocean University, Zhoushan, Zhejiang 316022, PR China
| | - Zhongyan Zhang
- Key Laboratory of Theoretical Chemistry of Environment Ministry of Education, South China Normal University, Guangzhou 510631, PR China
| | - Hong Liu
- Key Laboratory of Theoretical Chemistry of Environment Ministry of Education, South China Normal University, Guangzhou 510631, PR China
| | - Jian Sun
- College of Pharmacy, Xinjiang Key Laboratory of Biopharmaceuticals and Medical Devices, Xinjiang Medical University, Urumqi 830017, China.
| | - Li-Jun Ma
- School of Chemistry, South China Normal University, Guangzhou 510006, PR China; Key Laboratory of Analytical Chemistry for Biomedicine, South China Normal University, Guangzhou 510006, PR China; Key Laboratory of Theoretical Chemistry of Environment Ministry of Education, South China Normal University, Guangzhou 510631, PR China.
| |
Collapse
|
33
|
Liu S, Ren Z, Yan M, Ye W, Hu Y. Strategies to enhance the penetration of nanomedicine in solid tumors. Biomaterials 2025; 321:123315. [PMID: 40185056 DOI: 10.1016/j.biomaterials.2025.123315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 03/16/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Nanomedicine was previously regarded as a promising solution in the battle against cancer. Over the past few decades, extensive research has been conducted to exploit nanomedicine for overcoming tumors. Unfortunately, despite these efforts, nanomedicine has not yet demonstrated its ability to cure tumors, and the research on nanomedicine has reached a bottleneck. For a significant period of time, drug delivery strategies have primarily focused on targeting nanomedicine delivery to tumors while neglecting its redistribution within solid tumors. The uneven distribution of nanomedicine within solid tumors results in limited therapeutic effects on most tumor cells and significantly hampers the efficiency of drug delivery and treatment outcomes. Therefore, this review discusses the challenges faced by nanomedicine in penetrating solid tumors and provides an overview of current nanotechnology strategies (alleviating penetration resistance, size regulation, tumor cell transport, and nanomotors) that facilitate enhanced penetration of nanomedicine into solid tumors. Additionally, we discussed the potential role of nanobionics in promoting effective penetration of nanomedicine.
Collapse
Affiliation(s)
- Sen Liu
- Jiangsu Provincial Engineering Research Center for Biomedical Materials and Advanced Medical Devices, Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Zhendong Ren
- Faculty of Chemical Engineering, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Manqi Yan
- College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Wei Ye
- Faculty of Chemical Engineering, Huaiyin Institute of Technology, Huaian, 223003, China.
| | - Yong Hu
- College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China.
| |
Collapse
|
34
|
Musumeci F, Fasce A, Falesiedi M, Oleari F, Grossi G, Carbone A, Schenone S. Approaching Gallium-68 radiopharmaceuticals for tumor diagnosis: a Medicinal Chemist's perspective. Eur J Med Chem 2025; 294:117760. [PMID: 40393260 DOI: 10.1016/j.ejmech.2025.117760] [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: 04/01/2025] [Revised: 05/09/2025] [Accepted: 05/12/2025] [Indexed: 05/22/2025]
Abstract
Nuclear medicine has revolutionized disease diagnosis and treatment, particularly in oncology, by enabling precise imaging and targeted therapies using radiopharmaceuticals. Recently, Gallium-68 (68Ga) has emerged as a powerful positron emission tomography (PET) imaging agent, with a growing role in theranostics when paired with 177Lu for cancer treatment. The ability to obtain 68Ga from 68Ge/68Ga generators, along with its favorable radiochemical and pharmacokinetic properties, has driven an increasing number of clinical applications, which culminated with the approvals of 68Ga-DOTA-TOC and 68Ga-DOTA-TATE for the treatment of neuroendocrine tumors, and 68Ga-PSMA-11 for prostate cancer over the past decade. This review provides a comprehensive overview of 68Ga radiochemistry, chelators, and key compounds in clinical trials, highlighting the potential of this radionuclide in precision oncology.
Collapse
Affiliation(s)
- Francesca Musumeci
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132, Genoa, Italy.
| | - Alessandro Fasce
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132, Genoa, Italy
| | - Marta Falesiedi
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132, Genoa, Italy
| | - Federica Oleari
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132, Genoa, Italy
| | - Giancarlo Grossi
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132, Genoa, Italy
| | - Anna Carbone
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132, Genoa, Italy
| | - Silvia Schenone
- Department of Pharmacy, University of Genoa, Viale Benedetto XV, 3, 16132, Genoa, Italy
| |
Collapse
|
35
|
Ragab A, Raslan RR, Abusaif MS, Thabet HK, Ammar YA, Gohar NA. Discovery and optimization of 2-pyridones as dual h-DHFR/EGFR TK inhibitors with immunomodulatory potential; design, synthesis, anti-proliferative activity, and apoptosis inducer. Eur J Med Chem 2025; 294:117751. [PMID: 40378576 DOI: 10.1016/j.ejmech.2025.117751] [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: 03/11/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/19/2025]
Abstract
Liver and colorectal cancers present considerable health challenges, underscoring the need to identify innovative targeted therapeutics. Tumor progression can be prevented by targeting EGFR-TK and h-DHFR as essential molecular targets. In this context, we synthesized a new series of 2-pyridones from the reaction of 2-cyanoacrylamide with active methylene or 2-cyanoacetanilide with activated double bonds under basic conditions. The structure of the synthesized 2-pyridones was confirmed through microanalysis and spectroscopic data. In comparison to doxorubicin, the spiro 2-pyridine derivative 9b exhibited the highest anti-proliferative activity, demonstrating IC50 values of 6.89 ± 0.4 μM and 5.68 ± 0.3 μM against HepG-2 and Caco-2 cell lines, respectively, with nearly 2-fold increase in efficacy observed in Caco-2 cells. Additionally, compound 9b demonstrated a significant safety profile concerning normal cells (WI-38), as indicated by selectivity index values of 14.66 and 12.09 against the Caco-2 and HepG-2 cell lines, respectively. Moreover, flow cytometry analysis revealed that compound 9b halted the cell cycle at the G1/S phase in Caco-2 treated cells, demonstrating an increase in the percentage of cells undergoing both early and late apoptosis. The apoptotic potential was corroborated by the up-regulation of BAX and the down-regulation of Bcl-2 levels. Compound 9b exhibited significant inhibitory activity against h-DHFR, with an IC50 value of 0.192 ± 0.011 μM, compared to methotrexate (IC50 = 0.191 ± 0.011 μM). Furthermore, compound 9b demonstrated EGFR inhibitory activity, with IC50 of 0.109 ± 0.005 μM, which is close to the inhibition observed with Lapatinib (IC50 = 0.044 ± 0.002 μM). Compound 9b had better immunomodulatory properties with significant inhibitory efficacy on TNF-α and IL-6, with IC50 values of 0.40 ± 0.03 pg/mL and 0.60 ± 0.02 pg/mL, respectively. These values indicate a greater potency than the positive control drug Lapatinib, which displayed IC50 values of 0.41 ± 0.03 pg/mL and 0.74 ± 0.05 pg/mL for TNF-α and IL-6, respectively. In addition, in silico metabolism prediction using SwissADME and BioTransformer tools revealed that compound 9b is a potential inhibitor of CYP2C9 and CYP3A4, and is predicted to undergo metabolic transformations primarily via aromatic hydroxylation and ketone reduction, while maintaining acceptable stability of its ester moiety. Finally, the molecular docking assessment, together with the direct in vitro enzymatic inhibition results, confirmed that the 2-pyridone derivative 9b can potently bind to and inhibit both EGFR and h-DHFR through favorable binding interactions.
Collapse
Affiliation(s)
- Ahmed Ragab
- Chemistry Department, Faculty of Science (boys), Al-Azhar University, 11884, Nasr City, Cairo, Egypt; Chemistry Department, Faculty of Science, Galala University, Galala City, 43511, Suez, Egypt
| | - Reham R Raslan
- Department of Chemistry, Faculty of Science (Girls), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Moustafa S Abusaif
- Chemistry Department, Faculty of Science (boys), Al-Azhar University, 11884, Nasr City, Cairo, Egypt; Department for Synthesis and Characterization of Polymers, Polymer Institute of the Slovak Academy of Sciences SAS, Dúbravská cesta 9, 845 41, Bratislava, Slovakia
| | - Hamdy Khamees Thabet
- Center for Scientific Research and Entrepreneurship, Northern Border University, Arar, 73213, Saudi Arabia; Department of Chemistry, College of Sciences and Arts, Northern Border University, Rafha, 91911, Saudi Arabia.
| | - Yousry A Ammar
- Chemistry Department, Faculty of Science (boys), Al-Azhar University, 11884, Nasr City, Cairo, Egypt
| | - Nirvana A Gohar
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, 11571, Egypt
| |
Collapse
|
36
|
Wang X, Zeng J, Liu S, Qi F, Xin C, Li X, Gao Y, Fan D, Qu Y, Ren Z, Liu J, Xue R, Chen H, Zhang Y, Ha J, Su X, Wang J. Targeted degradation of DDR1 by proteolytic targeting chimera reverses immune exclusion for tumor immunotherapy. Eur J Med Chem 2025; 294:117750. [PMID: 40382838 DOI: 10.1016/j.ejmech.2025.117750] [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: 02/13/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/20/2025]
Abstract
The formation of immune exclusion microenvironment restricts the infiltration of immune cells into the core of tumors. The extracellular domain of the discoidin domain receptor tyrosine kinase 1 (DDR1) protein plays a pivotal role in this process by aligning collagen fibers to remodel the extracellular matrix (ECM), thereby excluding immune cells. Targeted degradation of DDR1 represents a promising approach to suppress the catalytic functions of the protein and remodel the extracellular matrix of DDR1-related tumors. Here, we report the discovery of a selective DDR1 degrader (DP 1), using the proteolysis targeting chimera (PROTAC) approach. Compound DP 1 exhibited potent DDR1 degradation ability with DC50 values reaching nanomolar range across various cell lines. The degradation of DDR1 effectively blocked the downstream signaling pathways, leading to further inhibition of tumor cell migration and invasion. More importantly, the in vivo studies highlighted the therapeutic potential of DDR1 degradation, indicated by the administration of DP 1 could facilitate the infiltration of immune cells into the tumor core which was associated with enhanced tumor apoptosis. In summary, we report a novel DDR1-targeting degrader with efficacious anti-tumor activity and excellent safety profile. Our studies offer a new perspective for cancer immunotherapy by targeting the immune-exclusion microenvironment.
Collapse
Affiliation(s)
- Xiaoyu Wang
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, China
| | - Jiangmeng Zeng
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, China
| | - Shilin Liu
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, China
| | - Fei Qi
- TPD Pharmaceutical Technology (SJZ) Co., Ltd., Shijiazhuang, 050035, China
| | - Chao Xin
- TPD Pharmaceutical Technology (SJZ) Co., Ltd., Shijiazhuang, 050035, China
| | - Xinyu Li
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, China
| | - Yan Gao
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, China
| | - Dan Fan
- TPD Pharmaceutical Technology (SJZ) Co., Ltd., Shijiazhuang, 050035, China
| | - Yuxuan Qu
- TPD Pharmaceutical Technology (SJZ) Co., Ltd., Shijiazhuang, 050035, China
| | - Zhiyi Ren
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, China
| | - Jinzhi Liu
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, China
| | - Ruoyu Xue
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, China
| | - Hongwei Chen
- The First Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Yong Zhang
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, China
| | - Jing Ha
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, China
| | - Xiangdong Su
- TPD Pharmaceutical Technology (SJZ) Co., Ltd., Shijiazhuang, 050035, China.
| | - Jinxu Wang
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, 050018, China; TPD Pharmaceutical Technology (SJZ) Co., Ltd., Shijiazhuang, 050035, China.
| |
Collapse
|
37
|
Zhao L, Lee ZH, Shah YM. Ammonia as a critical metabolic modulator of anti-tumor immunity. Med Gas Res 2025; 15:446-447. [PMID: 40251027 PMCID: PMC12054676 DOI: 10.4103/mgr.medgasres-d-24-00147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/06/2025] [Accepted: 02/24/2025] [Indexed: 04/20/2025] Open
Affiliation(s)
- Liang Zhao
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
| | - Zheng Hong Lee
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Yatrik M. Shah
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
38
|
Han Z, Wu Q, Rao H, Xu T, Zhou C. An overview of PROTACs targeting KRAS and SOS1 as antitumor agents. Bioorg Med Chem Lett 2025; 125-126:130283. [PMID: 40381703 DOI: 10.1016/j.bmcl.2025.130283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
KRAS is the most frequently mutated oncogene and its mutational activation drives approximately 25 % of human cancers. Son of Sevenless 1 (SOS1) plays a pivotal role in the KRAS signaling pathway through catalyzing the conversion of inactive GDP-bound KRAS to active GTP-bound KRAS, and is thus considered as a promising target for pan-KRAS inhibition. Currently, four KRASG12C-specific inhibitors, namely sotorasib, adagrasib, fulzerasib and garsorasib, have garnered regulatory approval. However, acquired resistance to KRASG12C inhibition rapidly emerges. In addition, the other prevalent KRAS mutations, including G12D and G12V, are still lacking effective therapeutic drugs. PROTAC-mediated KRAS and SOS1 degradation has been emerged as a promising strategy to overcome these issues, and achieved rapid progress in the recent years. This article provides an overview of the chemical structures, design strategies, structure-activity relationship (SAR) studies as well as in vitro and in vivo activities of the PROTACs degrading KRAS and SOS1, and sheds light on future challenges and opportunities to accelerate the development of new chemotherapies for KRAS-driven cancers.
Collapse
Affiliation(s)
- Zhiqiu Han
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Qianping Wu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Hongxin Rao
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China
| | - Tianfeng Xu
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China; Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Beijing 100049, China.
| | - Chuan Zhou
- Department of Medicinal Chemistry, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.
| |
Collapse
|
39
|
Li C, Liang Q, Pan W, Yang Y, Shao Q, Fan B. Design of a ROS-responsive fluorescent probe for the diagnosis and imaging of breast cancer. Bioorg Med Chem Lett 2025; 125-126:130268. [PMID: 40381705 DOI: 10.1016/j.bmcl.2025.130268] [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: 02/19/2025] [Revised: 04/19/2025] [Accepted: 05/11/2025] [Indexed: 05/20/2025]
Abstract
At present, radiotherapy and chemotherapy are a common treatment for breast cancer, but they will attack normal cells and cancer cells indiscriminately, resulting in a series of serious adverse reactions such as immune system damage, bone marrow suppression, and digestive system damage. Here, we have developed a fluorescent probe (Rhod-ben) that can achieve diagnosis and imaging in breast cancer tissue. The mechanism is as follows: (1) In mitochondria overexpressing ROS (reactive oxygen species), HRhod-ben (non-fluorescent) is oxidized to Rhod-ben (fluorescent), thereby achieving mitochondrial localization in breast cancer cells. (2) Under green light (λ ∼ 550 nm), the CO bond in Rhod ben is broken, thus releasing the anticancer drug (bendamostatin) targeted in breast cancer cells. Therefore, Rhod-ben is expected to become a new "diagnosis and imaging" fluorescent probe for breast cancer cells or cancer cells with ROS overexpression.
Collapse
Affiliation(s)
- Chunxiao Li
- School of Pharmacy, Hubei University of Science and Technology, No.88, Xianning avenue, XiananDistrict, Xianning 437000, China
| | - Qiao Liang
- School of Pharmacy, Hubei University of Science and Technology, No.88, Xianning avenue, XiananDistrict, Xianning 437000, China
| | - Wenyun Pan
- School of Pharmacy, Hubei University of Science and Technology, No.88, Xianning avenue, XiananDistrict, Xianning 437000, China
| | - Yuyu Yang
- School of Pharmacy, Hubei University of Science and Technology, No.88, Xianning avenue, XiananDistrict, Xianning 437000, China
| | - Qianshan Shao
- School of Pharmacy, Hubei University of Science and Technology, No.88, Xianning avenue, XiananDistrict, Xianning 437000, China
| | - Baolei Fan
- School of Pharmacy, Hubei University of Science and Technology, No.88, Xianning avenue, XiananDistrict, Xianning 437000, China; Hubei Provincial Key Laboratory of Radiation Chemistry and Functional Materials, Hubei University of Science and Technology, No.88, Xianning Avenue, Xianan District, Xianning 437000, China.
| |
Collapse
|
40
|
Panja S, Kapoor E, Siddhanta K, Jogdeo CM, Sil D, Khan RI, Kumari N, Ding L, Gendelman HE, Singh AB, Oupický D. Bioactive polymers as stimulus-responsive anti-metastatic combination agents to treat pancreatic cancer. Biomaterials 2025; 320:123255. [PMID: 40107179 DOI: 10.1016/j.biomaterials.2025.123255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
The intractable and devastating nature of pancreatic ductal adenocarcinoma (PDAC) necessitates an urgent need for novel therapies. This study presents the development of a novel polymer prodrug system for the combination treatment of PDAC, based on an optimized pharmacologically active anti-metastatic macromolecular carrier, PCQ, conjugated with gemcitabine (GEM). Structure-activity relationship evaluations showed that random PCQ copolymers exhibited superior anti-migratory activity compared to the gradient PCQ analogs. GEM was incorporated into the random PCQ copolymers using disulfide linker to prepare a reduction-responsive prodrug, PCQ(r)6-SS-GEM12. The resultant therapeutic system presents a pharmacologically active delivery strategy that targets both the proliferative and the metastatic phenotype in PDAC. The PCQ(r)6-SS-GEM12 prodrug demonstrated a selective release of GEM under the reductive tumor environment leading to a significant inhibition of tumor growth with pronounced anti-metastatic effect. Collectively, our data show that the combination of anti-metastatic PCQ and cytotoxic GEM-based reduction-responsive prodrug polymer offers an innovative strategy to treat PDAC.
Collapse
Affiliation(s)
- Sudipta Panja
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA; Department of Pharmacology and Experimental Neuroscience, USA
| | - Ekta Kapoor
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Kasturi Siddhanta
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Chinmay M Jogdeo
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Diptesh Sil
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Rubayat I Khan
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Neha Kumari
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | - Ling Ding
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA
| | | | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, College of Pharmacy, USA.
| |
Collapse
|
41
|
Delgado-Cruzata L, Zacarias O, Cheng SY, Tartaglia J, Rosas M, Gonzalez C, Champeil E. Stereoisomeric mitomycins interstrand crosslinks differently impact gene expression in MCF-7 and K562 cancer cells. Chem Biol Interact 2025; 417:111564. [PMID: 40383468 PMCID: PMC12145235 DOI: 10.1016/j.cbi.2025.111564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 04/15/2025] [Accepted: 05/15/2025] [Indexed: 05/20/2025]
Abstract
Mitomycin C (MC) is an anticancer drug used to treat stomach, anal and lung cancers. The main cytotoxicity of MC is due to its ability to form interstrand crosslinks with DNA (ICLs). The stereochemical configuration at C1″ of MC major ICL is R (α-ICL). In contrast, decarbamoylmitomycin C, a synthetic derivative of MC, generates the major S stereoisomeric ICL (β-ICL). Here, we investigated the effect of the stereochemical configuration of the α/β-ICL on the cellular response by focusing on gene expression changes in MCF-7 and K562 cell lines, one with wild type and the other with mutated TP53, upon treatment with both ICLs. We transfected both cell lines with duplex oligonucleotides containing either the α- or β-ICL at a single site and extracted RNA for transcriptome analysis. Results show that the stereochemical configuration of the α/β-ICL is responsible for distinct gene expression changes in MCF-7 and K562 cells. Our data also show that, in MCF-7 cells, α-ICL treatment triggers a strong increase in CDKN1A expression which is also observed at the protein level, contrary to what happens upon β-ICL treatment. In addition, β-ICL treatment led to a strong downregulation of a greater number of genes than the α-ICL in both cell lines, in particular in K562 cells, which harbor a TP53 mutation. This suggests that the β-ICL toxicity relies on a mechanism which leads to an overall downregulation of gene expression and may explain the greater toxicity of DMC toward TP53 mutant cells.
Collapse
Affiliation(s)
- Lissette Delgado-Cruzata
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York, NY, 10019, USA
| | - Owen Zacarias
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York, NY, 10019, USA
| | - Shu-Yuan Cheng
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York, NY, 10019, USA; The Graduate Center of the City University of New York, New York, NY, 10016, USA
| | - Jaxon Tartaglia
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York, NY, 10019, USA
| | - Melissa Rosas
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York, NY, 10019, USA
| | - Christina Gonzalez
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York, NY, 10019, USA
| | - Elise Champeil
- Department of Sciences, John Jay College of Criminal Justice, The City University of New York, New York, NY, 10019, USA; The Graduate Center of the City University of New York, New York, NY, 10016, USA.
| |
Collapse
|
42
|
Załuska-Ogryzek K, Wróblewska-Łuczka P, Góralczyk A, Luszczki JJ. Antiproliferative effects of arvanil, olvanil and LY2183240 on human neuroblastoma and glioblastoma cell lines and their in vitro interactions with cisplatin and temozolomide: an isobolographic analysis. Eur J Pharmacol 2025; 1001:177775. [PMID: 40441593 DOI: 10.1016/j.ejphar.2025.177775] [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: 03/24/2025] [Revised: 05/14/2025] [Accepted: 05/26/2025] [Indexed: 06/02/2025]
Abstract
Glioblastoma is the most aggressive malignant brain tumor characterized by rapid development, poor prognosis and high mortality. The purpose of this in vitro study was to assess cytotoxic and anti-proliferative effects of arvanil, olvanil and LY2183240 (three cannabinoid receptor ligands), when used alone and in combination with cisplatin and temozolomide in various neuroblastoma (CHP-134 and KELLY) and glioblastoma (U87MG, C6, and T98G) cell lines in the MTT assay. Results indicate that arvanil, olvanil and LY2183240 considerably affected the viability of neuroblastoma and glioblastoma cell lines with selectivity index values ranging from 0.50 to 17.48 for arvanil; from 0.93 to 24.06 for olvanil; and from 1.68 to 14.52 for LY2183240. With isobolographic analysis we found that the combination of LY2183240 with cisplatin produced three different types of interactions: a synergy in CHP-134; an antagonism in T98G; and additivity in KELLY, C6 and U87MG cell lines in the MTT assay. The combinations of arvanil and olvanil with cisplatin exerted additive interaction in all the tested cell lines in the MTT assay. Similarly, the combinations of temozolomide with arvanil, olvanil and LY 2183240 produced the additive interaction in all the tested glioblastoma and neuroblastoma cell lines. In conclusion, the combinations of temozolomide with the three cannabinoids, due to their additive interactions, can be considered as a promising therapeutic direction in glioblastoma. Favorable combinations of cisplatin with arvanil, olvanil and LY2183240 can also be recommended, except for a combination of cisplatin with LY2183240 in glioblastoma T98G cell line in the MTT assay.
Collapse
Affiliation(s)
- Katarzyna Załuska-Ogryzek
- Department of Occupational Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland.
| | - Paula Wróblewska-Łuczka
- Department of Occupational Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland.
| | - Agnieszka Góralczyk
- Department of Occupational Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland.
| | - Jarogniew J Luszczki
- Department of Occupational Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090, Lublin, Poland.
| |
Collapse
|
43
|
Zhang Z, Zhang Q, Wang Y. CAF-mediated tumor vascularization: From mechanistic insights to targeted therapies. Cell Signal 2025; 132:111827. [PMID: 40288665 DOI: 10.1016/j.cellsig.2025.111827] [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: 01/21/2025] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025]
Abstract
Cancer-associated fibroblasts (CAFs) are a major component of the tumor microenvironment (TME) and play a crucial role in tumor progression. The biological properties of tumors, such as drug resistance, vascularization, immunosuppression, and metastasis are closely associated with CAFs. During tumor development, CAFs contribute to tumor progression by remodeling the extracellular matrix (ECM), inhibiting immune cell function, promoting angiogenesis, and facilitating tumor cell growth, invasion, and metastasis. Studies have shown that CAFs can promote endothelial cell proliferation by directly secreting cytokines such as vascular endothelial growth factor (VEGF) and fibroblast Growth Factor (FGF), as well as through exosomes. CAFs also secrete the chemokine stromal cell-derived factor 1 (SDF-1) to recruit endothelial progenitor cells (EPCs) into the peripheral blood and guide their migration to the tumor periphery. Additionally, CAFs can induce tumor cells to transform into "endothelial cells" that participate in vascular wall formation. However, the precise mechanisms remain to be further investigated. Due to their widespread presence in various solid tumors and their tumor-promoting function, CAFs are emerging as therapeutic targets. In this review, we summarize the specific mechanisms through which CAFs promote angiogenesis and outline current therapeutic strategies targeting CAF-induced vascularization, ongoing clinical trials targeting CAFs, and discuss potential future treatment approaches. We hope this will contribute to the advancement of CAF-targeted tumor treatment strategies.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China
| | - Qing Zhang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| | - Yang Wang
- Department of Neurosurgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100020, China.
| |
Collapse
|
44
|
Xi Y, Huang Y, Hu J, Wang Y, Qian Q, Tu L, Nie H, Zhu J, Ding C, Gao X, Zheng X, Huang D, Cheng L. EIF2B5 promotes malignant progression of hepatocellular carcinoma by activating the PI3K/AKT signaling pathway through targeting RPL6. Cell Signal 2025; 132:111821. [PMID: 40246131 DOI: 10.1016/j.cellsig.2025.111821] [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: 04/05/2025] [Accepted: 04/15/2025] [Indexed: 04/19/2025]
Abstract
Hepatocellular carcinoma (HCC) is a highly aggressive malignancy with limited treatment options and poor prognosis. In this study, we demonstrated the critical role of EIF2B5 in driving HCC progression. We found EIF2B5 expression is significantly upregulated in HCC tumor tissues in several bioinformatics datasets, including The Cancer Genome Atlas, and that high expression of EIF2B5 predicts poor prognosis for HCC patients. Through a series of in vitro cell biology experiments, we found that EIF2B5 knockdown significantly attenuated Hep3B and HepG2 proliferation, migration, and invasion and increased cell cycle arrest, whereas EIF2B5 overexpression promoted HCC progression. Through mass spectrometry and immunoprecipitation validation, we found that EIF2B5 directly interacted with RPL6 and that when EIF2B5 was overexpressed in HCC cells, it promoted the expression of the downstream protein RPL6, which was able to activate the phosphatidylinositol kinase (PI3K)/serine-threonine kinase (AKT)/mammalian target of rapamycin (mTOR) pathway and thereby increase the proliferation and invasion ability of HCC cell lines, as verified by second-generation sequencing analysis and western blot. We further verified these findings using the mouse ectopic tumor assay, and the results showed that EIF2B5 knockdown significantly inhibited tumor progression in HCC mice. The present study suggests that EIF2B5 promotes malignant progression of HCC by interacting with RPL6 and activating the PI3K/AKT/mTOR signaling pathway and may serve as a potential target for the treatment of HCC.
Collapse
Affiliation(s)
- Yiling Xi
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yue Huang
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiahui Hu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yan Wang
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Qiyi Qian
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Linglan Tu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Huizong Nie
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Jiayao Zhu
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Chenguang Ding
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaotao Gao
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Xiaoliang Zheng
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Dongsheng Huang
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Liyan Cheng
- Zhejiang Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine, School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
45
|
Shaw P, Dey Bhowmik A, Gopinatha Pillai MS, Robbins N, Dwivedi SKD, Rao G. Anoikis resistance in Cancer: Mechanisms, therapeutic strategies, potential targets, and models for enhanced understanding. Cancer Lett 2025; 624:217750. [PMID: 40294841 DOI: 10.1016/j.canlet.2025.217750] [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: 01/21/2025] [Revised: 04/01/2025] [Accepted: 04/26/2025] [Indexed: 04/30/2025]
Abstract
Anoikis, defined as programmed cell death triggered by the loss of cell-extracellular matrix (ECM) and cell-cell interactions, is crucial for maintaining tissue homeostasis and preventing aberrant cell migration. Cancer cells, however, display anoikis resistance (AR) which in turn enables cancer metastasis. AR results from alterations in apoptotic signaling, metabolic reprogramming, autophagy modulation, and epigenetic changes, allowing cancer cells to survive in detached conditions. In this review we describe the mechanisms underlying both anoikis and AR, focusing on intrinsic and extrinsic pathways, disrupted cell-ECM interactions, and autophagy in cancer. Recent findings (i.e., between 2014 and 2024) on epigenetic regulation of AR and its role in metastasis are discussed. Therapeutic strategies targeting AR, including chemical inhibitors, are highlighted alongside a network analysis of 122 proteins reported to be associated with AR which identifies 53 hub proteins as potential targets. We also evaluate in vitro and in vivo models for studying AR, emphasizing their role in advancing metastasis research. Our overall goal is to guide future studies and therapeutic developments to counter cancer metastasis.
Collapse
Affiliation(s)
- Pallab Shaw
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Pathology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA
| | - Arpan Dey Bhowmik
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA
| | - Mohan Shankar Gopinatha Pillai
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA
| | - Nathan Robbins
- James E. Hurley School of Science and Mathematics, Oklahoma Baptist University, Shawnee, OK, USA
| | - Shailendra Kumar Dhar Dwivedi
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA
| | - Geeta Rao
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA; Department of Pathology, The University of Oklahoma Health Sciences, Oklahoma City, 73104, Oklahoma, USA.
| |
Collapse
|
46
|
Wen J, Wu D, Le Y, Yin Z, Chen M, Shen Y, Wu X, Liu K, Luo K, Shu Z, Shu Q, Ouyang D. Engineered nanovesicles targeting SERPINE1 overcome temozolomide resistance in glioblastoma. Cell Signal 2025; 132:111763. [PMID: 40139622 DOI: 10.1016/j.cellsig.2025.111763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/15/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025]
Abstract
Glioblastoma multiforme (GBM) is a highly aggressive brain tumor with limited treatment options due to its resistance to temozolomide (TMZ). This study explores a novel therapeutic approach using engineered cell membrane nanovesicles loaded with SERPINE1 inhibitors to combat TMZ resistance. High-throughput sequencing identified pivotal genes associated with resistance, while the nanovesicles demonstrated excellent stability and the ability to cross the blood-brain barrier. Functional assays revealed significant suppression of GBM cell viability, migration, and invasion, accompanied by reduced expression of SERPINE1 and VEGF, suggesting inhibition of angiogenesis and tumor progression. These findings highlight the potential of SERPINE1-targeted nanovesicles as an innovative and effective strategy for overcoming TMZ resistance in GBM.
Collapse
Affiliation(s)
- Jianping Wen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China.
| | - Dongxu Wu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Yi Le
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Zonghua Yin
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Minglong Chen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Yulong Shen
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Xia Wu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Kebo Liu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Kun Luo
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Zhicheng Shu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Qingxia Shu
- Department of Neurosurgery, Hunan University of Medicine General Hospital, Huaihua 418000, China
| | - Dongsheng Ouyang
- The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
47
|
Zhong R, Qiu C, Chan S, Wang Y, Liu K, Xia Y, Zhang H, Zou B. TDH-11 inhibits the proliferation and colonization of colorectal cancer by reducing the activity of HDAC. Cell Signal 2025; 132:111817. [PMID: 40250693 DOI: 10.1016/j.cellsig.2025.111817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/28/2025] [Accepted: 04/14/2025] [Indexed: 04/20/2025]
Abstract
Histone deacetylase inhibitors (HDACIs) have demonstrated significant efficacy and minimal toxic side effects in certain hematological tumors. Nevertheless, their utilization in the therapy of solid tumors, including colorectal cancer (CRC), is constrained by the occurrence of adverse effects such as myelosuppression and cardiotoxicity. Therefore, the development of more efficient and safer HDACIs is crucial for managing CRC. Here, the effects of TDH-11 (a novel HDAC inhibitor) and the underlying molecular mechanisms that inhibits the deveolpment and progression of CRC cells were investigated using in vitro and in vivo experiments. The results indicated that TDH-11 inhibited CRC tumorigenic behavior while also promoted apoptosis and cell cycle arrest. In vivo, TDH-11 markedly inhibited tumor progression and reduces tumor colonization without causing substantial damage to key organs, such as the kidneys, heart, lungs, spleen, and liver. Results of RNA sequencing and western blot suggested that TDH-11 exerted its antitumor effects through modulation of the p53 signaling pathway and its downstream pathways involved in apoptosis and cell cycle regulation. In summary, TDH-11 exhibited significant potential in suppressing the growth and colonization of CRC, as determined in both cellular and animal models. These results provided novel insights into CRC-associated pathways and suggest promising prospects for managing advanced and metastatic CRC.
Collapse
Affiliation(s)
- Rulei Zhong
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China; Anhui Medical University, Hefei, 230032, Anhui, China
| | - Chenyang Qiu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China; Anhui Medical University, Hefei, 230032, Anhui, China
| | - Shixin Chan
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China; Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yiming Wang
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei 230032, Anhui, China; Anhui Medical University, Hefei, 230032, Anhui, China
| | - Kaige Liu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China; Anhui Medical University, Hefei, 230032, Anhui, China
| | - Yihui Xia
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China; Anhui Medical University, Hefei, 230032, Anhui, China
| | - Huabing Zhang
- Department of Biochemistry and Molecular Biology, Metabolic Disease Research Center, School of Basic Medicine, Anhui Medical University, Hefei 230032, Anhui, China; Anhui Medical University, Hefei, 230032, Anhui, China.
| | - Bingbing Zou
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei 230032, China; Institute of Artificial Intelligence, Hefei Comprehensive National Science Center, Hefei, China; Anhui Medical University, Hefei, 230032, Anhui, China.
| |
Collapse
|
48
|
Li W, Shen Q, Tong T, Tian H, Lian X, Wang H, Yang K, Dai Z, Li Y, Chen X, Wang Q, Yang D, Wang F, Hao F, Wang L. Sequential simulation of regeneration-specific microenvironments using scaffolds loaded with nanoplatelet vesicles enhances bone regeneration. Bioact Mater 2025; 50:475-493. [PMID: 40342486 PMCID: PMC12059598 DOI: 10.1016/j.bioactmat.2025.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 03/27/2025] [Accepted: 04/16/2025] [Indexed: 05/11/2025] Open
Abstract
Bone regeneration is a complex and coordinated physiological process, and the different stages of this process have corresponding microenvironments to support cell development and physiological activities. However, biological scaffolds that provide different three-dimensional environments during different stages of bone regeneration are lacking. In this study, we report a novel composite scaffold (NPE@DCBM) inspired by the stages of bone regeneration; this scaffold was composed of a fibrin hydrogel loaded with nanoplatelet vesicles (NPVs), designated as NPE, and decellularized cancellous bone matrix (DCBM) microparticles. Initially, the NPE rapidly established a temporary microenvironment conducive to cell migration and angiogenesis. Subsequently, the DCBM simulated the molecular structure of bone and promoted new bone formation. In vitro, the NPVs regulated lipid metabolism in bone marrow mesenchymal stem cells (BMSCs), reprogramed the fate of BMSCs by activating the PI3K/AKT and MAPK/ERK positive feedback pathways, and increased BMSC functions, including proliferation, migration and proangiogenic potential. In vivo, NPV@DCBM accelerated bone tissue regeneration and repair. Initially, the NPE rapidly induced angiogenesis between DCBM microparticles, and subsequently, BMSCs differentiated into osteoblasts with DCBM microparticles at their core. In summary, the design of this composite scaffold that sequentially mimics different bone regeneration microenvironments may provide a promising strategy for bone regeneration, with clinical translational potential.
Collapse
Affiliation(s)
- Wenshuai Li
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- The Key Laboratory of Orthopedic Biomechanics of Hebei Province, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- Hangzhou OrigO Biotechnology Co. Ltd., Hangzhou, Zhejiang, 310016, China
| | - Qichen Shen
- Hangzhou OrigO Biotechnology Co. Ltd., Hangzhou, Zhejiang, 310016, China
| | - Tong Tong
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- The Key Laboratory of Orthopedic Biomechanics of Hebei Province, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Hongsen Tian
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Xiaowei Lian
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- The Key Laboratory of Orthopedic Biomechanics of Hebei Province, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Haoli Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Ke Yang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Zhanqiu Dai
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
| | - Yijun Li
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Xianhua Chen
- Zhejiang Institute of Medical Device Testing, Hangzhou, Zhejiang, 310016, China
| | - Qingqing Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Medical College of Zhejiang University, Hangzhou, Zhejiang, 310016, China
- Key Laboratory of Musculoskeletal System Degeneration and Regeneration Translational Research of Zhejiang Province, Hangzhou, Zhejiang, 310016, China
- Cixi Biomedical Research Institute, Wenzhou Medical University, Zhejiang, 315000, China
| | - Dan Yang
- Zhejiang DecellMatrix Biotechnology Co. Ltd., Hangzhou, Zhejiang, 310016, China
| | - Feng Wang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- The Key Laboratory of Orthopedic Biomechanics of Hebei Province, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Feng Hao
- Zhejiang DecellMatrix Biotechnology Co. Ltd., Hangzhou, Zhejiang, 310016, China
| | - Linfeng Wang
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
- The Key Laboratory of Orthopedic Biomechanics of Hebei Province, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| |
Collapse
|
49
|
Wang Q, Chen C, Zhao H, Jiao Y, Chen H, Wang P, Song T. Magnetotactic bacteria-mediated integrated magnetic targeted hyperthermia for in-situ deep-seated tumor. Colloids Surf B Biointerfaces 2025; 252:114658. [PMID: 40168695 DOI: 10.1016/j.colsurfb.2025.114658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/28/2025] [Accepted: 03/24/2025] [Indexed: 04/03/2025]
Abstract
Unlike hyperthermia after intratumoral injection, the method of integrated magnetic targeted hyperthermia (iMTH) guides magnetic medium to the target site and then directly performs in-situ heating, showing great potential for effective treatment of deep-seated tumors in the body. Magnetotactic bacteria (MTB), having chain-like arranged magnetic nanoparticles within its body and active movement along an external magnetic field, are considered as a very fitted material for iMTH. However, the amount of MTB concentrated on the deep-seated tumor posed a significant challenge for the successful implementation of iMTH. Herein, we aim to validate the strategy of integrating magnetic targeting and hyperthermia. An in-situ liver tumor model in mouse was developed as deep-seated tumors. After administering the polar MTB MO-1 intravenously via the tail vein, a focusing magnetic field navigated these bacteria to effectively accumulate at the deep-seated tumor site. Immediately afterwards, this targeted aggregation of MO-1 cells triggered a localized magnetic hyperthermia directly at the cancer site under an applied alternating magnetic field. Our findings demonstrated that this hyperthermia induced by the bacteria led to the death of liver cancer cells, thereby effectively curbing the progression and growth of the cancer. These promising results suggested that an iMTH approach was developed, harnessing the power of MTB. This method stands as an exciting and potential therapeutic strategy for the treatment of deep-seated tumors, offering new hope in the fight against cancer.
Collapse
Affiliation(s)
- Qingmeng Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing, China
| | - Changyou Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing, China.
| | - Haoyu Zhao
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yangkun Jiao
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haitao Chen
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing, China
| | - Pingping Wang
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing, China
| | - Tao Song
- Beijing Key Laboratory of Bioelectromagnetism, Institute of Electrical Engineering, Chinese Academy of Sciences, Beijing 100190, China; International Laboratory of Evolution and Development of Magnetotactic Multicellular Organisms, Beijing, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
50
|
Shi Q, Zhou T, Zhou Y, Wang ZH, Xue YJ, Chen YJ. Discovery of tetracyclic 1,2,4-triazoline-fused dibenzo[b,f][1,4]oxazepine as a potent anti-colorectal cancer agent with good efficacy and low toxicity. Bioorg Med Chem 2025; 125:118203. [PMID: 40267749 DOI: 10.1016/j.bmc.2025.118203] [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: 02/21/2025] [Revised: 04/09/2025] [Accepted: 04/17/2025] [Indexed: 04/25/2025]
Abstract
A series of tetracyclic 1,2,4-triazoline-fused dibenzo[b,f][1,4]oxazepines were evaluated as novel anti-tumor agents. MTT assay conducted in four human cancer cell lines (SW620, A549, MCF-7, HepG2) showed that 1,2,4-triazoline-fused dibenzo[b,f][1,4]oxazepine decorated by a methyl group on the benzene ring of 1,2,4-triazoline moiety exhibited a superior antiproliferative activity against SW620 cells with a IC50 value of 0.86 μM. The above compound was thus chosen for further investigation on its anti-colorectal cancer (CRC) effect, and displayed inhibitory effects on the proliferation of HCT116 and CT26 cells with IC50 values of 0.96 μM and 1.71 μM, respectively. Furthermore, this compound could effectively suppress colony formation and induce cell cycle arrest and apoptosis in SW620 cells. Western blot analysis demonstrated that it exerted the anti-tumor activity through blocking the PI3K-AKT signaling pathway. Next, we examined its in vivo anti-tumor activity by establishing a subcutaneous CT26 xenograft model, and found that it significantly reduced the tumor sizes with limited toxicity. Collectively, these findings suggest that this compound could be utilized as a promising candidate against CRC.
Collapse
Affiliation(s)
- Qiwen Shi
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Tao Zhou
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Yuqi Zhou
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Zhi-Hao Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yao-Jie Xue
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ya-Jing Chen
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|