1
|
Castagnino PA, Haas DA, Musante L, Tancler NA, Tran BV, Kean R, Steck AR, Martinez LA, Mostaghel EA, Hooper DC, Kim FJ. Sigma1 inhibitor suppression of adaptive immune resistance mechanisms mediated by cancer cell derived extracellular vesicles. Cancer Biol Ther 2025; 26:2455722. [PMID: 39863992 PMCID: PMC11776462 DOI: 10.1080/15384047.2025.2455722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/28/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025] Open
Abstract
Adaptive immune resistance in cancer describes the various mechanisms by which tumors adapt to evade anti-tumor immune responses. IFN-γ induction of programmed death-ligand 1 (PD-L1) was the first defined and validated adaptive immune resistance mechanism. The endoplasmic reticulum (ER) is central to adaptive immune resistance as immune modulatory secreted and integral membrane proteins are dependent on ER. Sigma1 is a unique ligand-regulated integral membrane scaffolding protein enriched in the ER of cancer cells. PD-L1 is an integral membrane glycoprotein that is translated into the ER and processed through the cellular secretory pathway. At the cell surface, PD-L1 is an immune checkpoint molecule that binds PD-1 on activated T-cells and blocks anti-tumor immunity. PD-L1 can also be incorporated into cancer cell-derived extracellular vesicles (EVs), and EV-associated PD-L1 can inactivate T-cells within the tumor microenvironment. Here, we demonstrate that a selective small molecule inhibitor of Sigma1 can block IFN-γ mediated adaptive immune resistance in part by altering the incorporation of PD-L1 into cancer cell-derived EVs. Sigma1 inhibition blocked post-translational maturation of PD-L1 downstream of IFN-γ/STAT1 signaling. Subsequently, EVs released in response to IFN-γ stimulation were significantly less potent suppressors of T-cell activation. These results suggest that by reducing tumor derived immune suppressive EVs, Sigma1 inhibition may promote antitumor immunity. Sigma1 modulation presents a novel approach to regulating the tumor immune microenvironment by altering the content and production of EVs. Altogether, these data support the notion that Sigma1 may play a role in adaptive immune resistance in the tumor microenvironment.
Collapse
Affiliation(s)
- Paola A. Castagnino
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Derick A. Haas
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Luca Musante
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Nathalia A. Tancler
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Bach V. Tran
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Rhonda Kean
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Alexandra R. Steck
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Luis A. Martinez
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Elahe A. Mostaghel
- Geriatric Research, Education and Clinical Center, U.S. Department of Veterans Affairs Puget Sound Health Care System, Seattle, WA, USA
| | - D. Craig Hooper
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| | - Felix J. Kim
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
- Sidney Kimmel Comprehensive Cancer Center at Jefferson, Philadelphia, PA, USA
| |
Collapse
|
2
|
Zhang F, Ramar S, Wang Y, Xu H, Zhang K, Awadasseid A, Rao G, Zhang W. Advances in cancer immunotherapy using small-molecular inhibitors targeting the PD-1/PD-L1 interaction. Bioorg Med Chem 2025; 127:118238. [PMID: 40367914 DOI: 10.1016/j.bmc.2025.118238] [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: 01/20/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/16/2025]
Abstract
Cancer cells evade immune responses by interacting with PD-1 and its ligand, PD-L1. Although monoclonal antibodies targeting this pathway have revolutionized oncology, their high production costs, poor oral bioavailability, and limited tumor penetration remain significant challenges. Small-molecule inhibitors provide a promising alternative, offering advantages such as improved tumor penetration and cost-effectiveness. This review highlights advancements in small-molecule PD-1/PD-L1 inhibitors, focusing on their mechanisms, structural designs, and therapeutic potential. Key innovations, including biphenyl scaffolds, heterocyclic frameworks, enhance binding efficiency and immune activation. The article effectively integrates fundamental principles of drug chemistry with real-world clinical needs, offering a comprehensive approach to the design of PD-1/PD-L1 small-molecule inhibitors. It systematically classifies various molecular structures, analyzes relevant industrial cases, and incorporates the most recent research findings. By examining these aspects, it uncovers the underlying logic driving the design process and provides a fresh, innovative perspective on advancing the field of immune small-molecule inhibitors for cancer therapy.
Collapse
Affiliation(s)
- Feng Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China
| | - Sivaramakarthikeyan Ramar
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China; Moganshan Institute ZJUT, Deqing 313202, China
| | - Yu Wang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China
| | - Haoran Xu
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China
| | - Koutian Zhang
- Zhejiang Qingzhenghong Technology Co., Ltd, Hangzhou 311121, China
| | - Annoor Awadasseid
- Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China; Zhejiang Qingzhenghong Technology Co., Ltd, Hangzhou 311121, China.
| | - Guowu Rao
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China.
| | - Wen Zhang
- Lab of Chemical Biology and Molecular Drug Design, College of Pharmaceutical Science, Zhejiang University of Technology, Deqing 313299, China; Institute of Drug Development & Chemical Biology, Zhejiang University of Technology, Deqing 313299, China; Zhejiang Jieyuan Med-Tech Co., Ltd., Hangzhou 311113, China.
| |
Collapse
|
3
|
Ton Nu QC, Deka G, Park PH. CD8 + T cell-based immunotherapy: Promising frontier in human diseases. Biochem Pharmacol 2025; 237:116909. [PMID: 40179991 DOI: 10.1016/j.bcp.2025.116909] [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: 02/28/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
The abundant cell components of the adaptive immune system called T lymphocytes (T cells) play important roles in mediating immune responses to eliminate the invaders and create the memory of the germs to form a new immunity for the next encounter. Among them, cytotoxic T cells expressing cell-surface CD8 are the most critical effector cells that directly eradicate the target infected cells by recognizing antigens presented by major histocompatibility complex class I molecules to protect our body from pathological threats. In the continuous evolution of immunotherapy, various CD8+ T cell-based therapeutic strategies have been developed based on the role and molecular concept of CD8+ T cells. The emergence of such remarkable therapies provides promising hope for multiple human disease treatments such as autoimmunity, infectious disease, cancer, and other non-infectious diseases. In this review, we aim to discuss the current knowledge on the utilization of CD8+ T cell-based immunotherapy for the treatment of various diseases, the molecular basis involved, and its limitations. Additionally, we summarize the recent advances in the use of CD8+ T cell-based immunotherapy and provide a comprehensive overview of CD8+ T cells, including their structure, underlying mechanism of function, and markers associated with CD8+ T cell exhaustion. Building upon these foundations, we delineate the advancement of CD8+ T cell-based immunotherapies with fundamental operating principles followed by research studies, and challenges, as well as illustrate human diseases involved in this development.
Collapse
Affiliation(s)
- Quynh Chau Ton Nu
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Gitima Deka
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea; Research institute of cell culture, Yeungnam University, Gyeongsan, Republic of Korea.
| |
Collapse
|
4
|
Jalaguier S, Kuehn A, Petitpas C, Dulom A, Jacquemont R, Assi C, Sixou S, Jeschke U, Colinge J, Cavaillès V. The transcription factor RIP140 regulates interferon γ signaling in breast cancer. Int J Cancer 2025; 157:170-182. [PMID: 40065499 PMCID: PMC12062925 DOI: 10.1002/ijc.35405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 02/18/2025] [Accepted: 02/25/2025] [Indexed: 05/11/2025]
Abstract
RIP140 (receptor interacting protein of 140 kDa) is an important player in breast cancer (BC) by regulating key cellular pathways such as nuclear hormone receptor signaling. In order to identify additional genes specifically regulated by RIP140 in BC, we performed a transcriptomic analysis after silencing its expression in MCF-7 cells. We identified the interferon γ (IFNγ) signaling as being substantially repressed by RIP140 knockdown. Using the GBP1 (guanylate binding protein 1) gene as a reporter of IFNγ signaling, we demonstrated its robust induction by RIP140 through an ISRE motif, leading to a significant reduction of its induction upon IFNγ treatment. Furthermore, we showed that low levels of RIP140 amplified the IFNγ-dependent inhibition of BC cell proliferation. In line with these data, reanalysis of transcriptomic data obtained in human BC samples revealed that IFNγ levels were associated with good prognosis only for BC patients exhibiting tumors expressing low levels of RIP140, thus confirming its effect on the anti-tumor activity of IFNγ provided by our experimental data. Altogether, this study identifies RIP140 as a new regulator of IFNγ signaling in breast tumorigenesis.
Collapse
Affiliation(s)
- Stéphan Jalaguier
- IRCM, Institut de Recherche en Cancérologie de MontpellierMontpellierFrance
- INSERMMontpellierFrance
- Université de MontpellierMontpellierFrance
- Institut régional du Cancer de MontpellierMontpellierFrance
| | - Axel Kuehn
- IRCM, Institut de Recherche en Cancérologie de MontpellierMontpellierFrance
- INSERMMontpellierFrance
- Université de MontpellierMontpellierFrance
- Institut régional du Cancer de MontpellierMontpellierFrance
| | - Chloé Petitpas
- IRCM, Institut de Recherche en Cancérologie de MontpellierMontpellierFrance
- INSERMMontpellierFrance
- Université de MontpellierMontpellierFrance
- Institut régional du Cancer de MontpellierMontpellierFrance
| | - Arnaud Dulom
- IRCM, Institut de Recherche en Cancérologie de MontpellierMontpellierFrance
- INSERMMontpellierFrance
- Université de MontpellierMontpellierFrance
- Institut régional du Cancer de MontpellierMontpellierFrance
| | - Rémy Jacquemont
- IRCM, Institut de Recherche en Cancérologie de MontpellierMontpellierFrance
- INSERMMontpellierFrance
- Université de MontpellierMontpellierFrance
- Institut régional du Cancer de MontpellierMontpellierFrance
| | - Cindy Assi
- IRCM, Institut de Recherche en Cancérologie de MontpellierMontpellierFrance
- INSERMMontpellierFrance
- Université de MontpellierMontpellierFrance
- Institut régional du Cancer de MontpellierMontpellierFrance
| | - Sophie Sixou
- Faculté des Sciences PharmaceutiquesUniversité Toulouse III—Paul SabatierToulouseFrance
| | - Udo Jeschke
- Department of Obstetrics and GynecologyUniversity Hospital AugsburgAugsburgGermany
| | - Jacques Colinge
- IRCM, Institut de Recherche en Cancérologie de MontpellierMontpellierFrance
- INSERMMontpellierFrance
- Université de MontpellierMontpellierFrance
- Institut régional du Cancer de MontpellierMontpellierFrance
| | - Vincent Cavaillès
- IRCM, Institut de Recherche en Cancérologie de MontpellierMontpellierFrance
- INSERMMontpellierFrance
- Université de MontpellierMontpellierFrance
- Institut régional du Cancer de MontpellierMontpellierFrance
- CNRSMontpellierFrance
| |
Collapse
|
5
|
Sabit H, Adel A, Abdelfattah MM, Ramadan RM, Nazih M, Abdel-Ghany S, El-Hashash A, Arneth B. The role of tumor microenvironment and immune cell crosstalk in triple-negative breast cancer (TNBC): Emerging therapeutic opportunities. Cancer Lett 2025; 628:217865. [PMID: 40516902 DOI: 10.1016/j.canlet.2025.217865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 06/03/2025] [Accepted: 06/07/2025] [Indexed: 06/16/2025]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype characterized by its lack of estrogen, progesterone, and HER2 receptors, leading to limited treatment options and poor prognosis. This review synthesizes current research on the tumor microenvironment (TME) and immune cell crosstalk in TNBC to identify emerging therapeutic opportunities. The TME in TNBC is a complex ecosystem comprising immune cells, fibroblasts, and extracellular matrix components, which significantly influence tumor growth and metastasis. Single-cell RNA sequencing reveals T-cell heterogeneity and identifies prognostic genes. Regulatory T cells (Tregs) play a key role in immunosuppression, with thymidine kinase-1 (TK1) identified as a potential therapeutic target. MUC1-C and CXCL9 modulate the TME, impacting T-cell depletion and macrophage differentiation. Spatial analysis highlights the importance of cell-to-cell interactions in predicting recurrence. Epithelial-mesenchymal transition (EMT) and thermogenesis also influence the TME, while epigenetic modifications, such as HDAC inhibition, can induce pyroptosis and enhance immune cell recruitment. Integrating genomic information with TME analysis is crucial for developing personalized treatments, considering racial disparities in immune infiltration. Emerging therapies targeting immune checkpoints, modulating Treg activity, and inducing pyroptosis hold promise for improving TNBC patient outcomes. Future research should focus on multi-omics data, spatial transcriptomics, and patient-derived models to refine therapeutic interventions.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Medical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P.O. Box 77, Giza, 3237101, Egypt.
| | - Amro Adel
- Department of Pharmaceutical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P.O. Box 77, Giza, 3237101, Egypt
| | - Mariam M Abdelfattah
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, P.O. Box 77, Giza, 3237101, Egypt
| | - Rehab M Ramadan
- Department of Pharmaceutical Biotechnology, College of Biotechnology, Misr University for Science and Technology, P.O. Box 77, Giza, 3237101, Egypt
| | - Mahmoud Nazih
- Al Ryada University for Science and Technology (RST), ElMehwar ElMarkazy-2, Cairo - Alex desert RD K92, Sadat City, 16504, Egypt; Scientific Office, Egyptian Society of Pharmacogenomics and Personalized Medicine (ESPM), Cairo, Egypt
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, P.O. Box 77, Giza, 3237101, Egypt
| | - Ahmed El-Hashash
- Elizabeth City State campus of the University of North Carolina (UNC), NC, 27909, USA
| | - Borros Arneth
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Philipps University Marburg, Baldingerstr. 1, Marburg, 35043, Germany; Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Hospital of the Universities of Giessen and Marburg (UKGM), Justus Liebig University Giessen, Feulgenstr 12, Giessen, 35392, Germany.
| |
Collapse
|
6
|
Griffiths A, Udomjarumanee P, Georgescu AS, Barri M, Zinovkin DA, Pranjol MZI. The Immunomodulatory Role of Galectin-1 in the Tumour Microenvironment and Strategies for Therapeutic Applications. Cancers (Basel) 2025; 17:1888. [PMID: 40507367 PMCID: PMC12153884 DOI: 10.3390/cancers17111888] [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: 04/21/2025] [Revised: 05/30/2025] [Accepted: 06/04/2025] [Indexed: 06/16/2025] Open
Abstract
With the morbidity of cancer currently on a perpetual rise, there is a critical need for new treatment options. Current therapeutic options, such as chemotherapy and radiotherapy, are frequently employed; however, the high rate of recurrence underscores the incomplete understanding of tumour growth, progression, and the intricacies of their microenvironments. In this study, we review the roles that galectin-1 (Gal1) plays in suppressing immune surveillance in the tumour microenvironment. Studies have shown that Gal1 changes the immune system parameters: suppressing T cell function, sensitising resting T lymphocytes to Fas/FasL, decreasing cell proliferation, reducing adhesion to extracellular matrix, inhibiting Th1 cytokines, increasing M2 phenotype macrophages, and promoting angiogenesis. Gal1 has garnered increasing attention as a potential therapeutic target due to its involvement in tumour progression and immune evasion. Given the limitations and toxic side effects associated with current treatment options, alternative strategies targeting Gal1 have been explored for their therapeutic potential. Approaches such as OTX008, anti-Gal1 monoclonal antibodies, and Gal1-targeted vaccines have demonstrated the ability to downregulate tumour progression by inhibiting Gal1 activity. These findings highlight the therapeutic promise of Gal1 not only as a novel target for cancer therapy but also as a potential prognostic biomarker, offering opportunities for the development of more effective and less toxic treatment strategies.
Collapse
Affiliation(s)
- Alice Griffiths
- Department of Biochemistry, School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | - Palita Udomjarumanee
- School of Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Andrei-Stefan Georgescu
- Department of Biochemistry, School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | - Muruj Barri
- Department of Biochemistry, School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| | - Dmitry A. Zinovkin
- Department of Pathology, Gomel State Medical University, 246000 Gomel, Belarus
| | - Md Zahidul I. Pranjol
- Department of Biochemistry, School of Life Sciences, University of Sussex, Brighton BN1 9QG, UK
| |
Collapse
|
7
|
Bhattacharya P, Linnenbach A, South AP, Martinez-Outschoorn U, Curry JM, Johnson JM, Harshyne LA, Mahoney MG, Luginbuhl AJ, Vadigepalli R. Tumor microenvironment governs the prognostic landscape of immunotherapy for head and neck squamous cell carcinoma: A computational model-guided analysis. PLoS Comput Biol 2025; 21:e1013127. [PMID: 40460357 PMCID: PMC12162103 DOI: 10.1371/journal.pcbi.1013127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 06/12/2025] [Accepted: 05/08/2025] [Indexed: 06/11/2025] Open
Abstract
Immune checkpoint inhibition (ICI) has emerged as a critical treatment strategy for squamous cell carcinoma of the head and neck (HNSCC) that halts the immune escape of the tumor cells. Increasing evidence suggests that the onset, progression, and lack of/no response of HNSCC to ICI are emergent properties arising from the interactions within the tumor microenvironment (TME). Deciphering how the diversity of cellular and molecular interactions leads to distinct HNSCC TME subtypes subsequently governing the ICI response remains largely unexplored. We developed a cellular-molecular model of the HNSCC TME that incorporates multiple cell types, cellular states, and transitions, and molecularly mediated paracrine interactions. Simulation across the selected parameter space of the HNSCC TME network shows that distinct mechanistic balances within the TME give rise to the five clinically observed TME subtypes such as immune/non-fibrotic, immune/fibrotic, fibrotic only and immune/fibrotic desert. We predict that the cancer-associated fibroblast, beyond a critical proliferation rate, drastically worsens the ICI response by hampering the accessibility of the CD8 + killer T cells to the tumor cells. Our analysis reveals that while an Interleukin-2 (IL-2) + ICI combination therapy may improve response in the immune desert scenario, Osteopontin (OPN) and Leukemia Inhibition Factor (LIF) knockout with ICI yields the best response in a fibro-dominated scenario. Further, we predict Interleukin-8 (IL-8), and lactate can serve as crucial biomarkers for ICI-resistant HNSCC phenotypes. Overall, we provide an integrated quantitative framework that explains a wide range of TME-mediated resistance mechanisms for HNSCC and predicts TME subtype-specific targets that can lead to an improved ICI outcome.
Collapse
Affiliation(s)
- Priyan Bhattacharya
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Alban Linnenbach
- Department of Otolaryngology, Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Andrew P. South
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Ubaldo Martinez-Outschoorn
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Joseph M. Curry
- Department of Otolaryngology, Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Jennifer M. Johnson
- Department of Otolaryngology, Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Larry A. Harshyne
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Mỹ G. Mahoney
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Adam J. Luginbuhl
- Department of Otolaryngology, Head and Neck Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Rajanikanth Vadigepalli
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
8
|
Zhao S, Zhang H, Shang G. Research progress of B7-H3 in malignant tumors. Front Immunol 2025; 16:1586759. [PMID: 40519928 PMCID: PMC12162284 DOI: 10.3389/fimmu.2025.1586759] [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: 03/03/2025] [Accepted: 05/09/2025] [Indexed: 06/18/2025] Open
Abstract
B7 homolog 3 (B7-H3, also known as CD276) is a novel member of the B7 immune protein family. There is a marked difference in the expression and distribution of B7-H3 protein and mRNA between normal and tumor tissues, with widespread expression in tumor tissues and a close relationship with tumor progression. B7-H3 activates or inhibits tumor immune responses by binding to receptors on the surface of immune cells. Apart from participating in tumor immune activities, it has regulatory effects on non-immunological functions, such as tumor migration and invasion, angiogenesis, glycometabolism, and drug resistance. Thus, it has important biological functions in regulating the progression of malignant tumors. Current research on the structure, function, and therapeutic methods of B7-H3 is continuously breaking new ground, deepening our understanding of B7-H3, and promoting the development of therapeutic drugs targeting this new protein. This review briefly discusses the structure and distribution of B7-H3, as well as its immune and non-immune functions in the progression of cancer. It also summarizes the research progress on drugs targeting B7-H3 and the latest developments in clinical trials, highlighting their significant potential for the treatment of malignant tumors.
Collapse
Affiliation(s)
- Shuaixiang Zhao
- The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
- Department of Bone and Soft Tissue Oncology, Department of Orthopaedic, Shengjing Hospital, China Medical University, Shenyang, Liaoning, China
| | - He Zhang
- Department of Bone and Soft Tissue Oncology, Department of Orthopaedic, Shengjing Hospital, China Medical University, Shenyang, Liaoning, China
| | - Guanning Shang
- The First Clinical College, Liaoning University of Traditional Chinese Medicine, Shenyang, Liaoning, China
- Department of Bone and Soft Tissue Oncology, Department of Orthopaedic, Shengjing Hospital, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
9
|
Chen E, Zhou W. Immunotherapy in microsatellite-stable colorectal cancer: Strategies to overcome resistance. Crit Rev Oncol Hematol 2025; 212:104775. [PMID: 40409481 DOI: 10.1016/j.critrevonc.2025.104775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 05/04/2025] [Accepted: 05/19/2025] [Indexed: 05/25/2025] Open
Abstract
Colorectal cancer (CRC) is among the foremost causes of cancer-related mortality worldwide; however, individuals with microsatellite-stable (MSS) disease-who constitute most CRC diagnoses-derive limited benefit from existing immunotherapeutic approaches. Here, we outline emerging methods designed to address the inherent resistance of MSS CRC to immune checkpoint inhibitors (ICIs). Recent findings emphasize how the immunosuppressive tumor microenvironment (TME) in MSS CRC, marked by diminished immunogenicity and high levels of regulatory T cells and myeloid-derived suppressor cells, restricts effective antitumor immune activity. Combination regimens that merge ICIs with chemotherapy, anti-angiogenic agents, or targeted blockade of pathways such as TGF-β and VEGF have shown encouraging early outcomes, including enhanced antigen presentation and T-cell penetration. Novel immunomodulatory platforms-such as epigenetic modifiers, oncolytic viruses, and engineered probiotic vaccines-are under assessment to further reprogram the TME and boost therapeutic efficacy. Concurrently, progress in adoptive cell therapies (for example, chimeric antigen receptor (CAR) T cells) and the development of cancer vaccines targeting tumor-associated and neoantigens promise to extend immune control over MSS CRC. In parallel, improving patient selection through predictive biomarkers-from circulating tumor DNA (ctDNA) to gene expression signatures and specific molecular subtypes-could refine individualized treatment strategies. Finally, interventions that alter the gut microbiome, including probiotics and fecal transplantation, serve as complementary tools to strengthen ICI responses. Taken together, these insights and combined treatment strategies lay the foundation for more successful immunotherapeutic interventions in MSS CRC, ultimately aiming to provide sustained clinical benefits to a broader spectrum of patients.
Collapse
Affiliation(s)
- Engeng Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou 310016, China
| | - Wei Zhou
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital of Zhejiang University, Hangzhou 310016, China.
| |
Collapse
|
10
|
Bauer M, Santos P, Wilfer A, van den Berg E, Zietsman A, Vetter M, Kaufhold S, Wickenhauser C, Dos-Santos-Silva I, Chen WC, Cubasch H, Murugan N, McCormack V, Joffe M, Seliger B, Kantelhardt E. HIV status alters immune cell infiltration and activation profile in women with breast cancer. Nat Commun 2025; 16:4699. [PMID: 40393975 DOI: 10.1038/s41467-025-59408-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 04/23/2025] [Indexed: 05/22/2025] Open
Abstract
The breast cancer (BC)-related mortality is higher and the immunity is altered in women living with HIV (WLWH) compared to HIV-negative women. Therefore, tumor samples of 296 black BC patients from South Africa and Namibia with known age, HIV status, tumor stage, hormone receptor and HER2 status and overall survival (OS) are analyzed for components of the tumor microenvironment (TME). WLWH (n = 117), either with suppressed viral activity (HR = 1.25) or with immune suppression (HR = 2.04), have a shorter OS. HIV status is associated with increased numbers of CD8+ T cells in the TME compared to HIV-negative patients; no correlation is found with CD4+ T cell numbers in the blood. Moreover, an increased expression of CD276/B7-H3 and a more pronounced IFN-γ signaling in the tumors are found in WLWH, independent of age, stage, and BC subtypes. In conclusion, altered T cell composition and CD276 expression in WLWH may contribute to inferior survival and can be used for targeted treatment.
Collapse
Affiliation(s)
- Marcus Bauer
- Institute of Pathology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
- Global and Planetary Health Working Group, Institute of Medical Epidemiology, Biometrics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
| | - Pablo Santos
- Global and Planetary Health Working Group, Institute of Medical Epidemiology, Biometrics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Andreas Wilfer
- Institute of Pathology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Krukenberg Cancer Center, University Hospital Halle, Halle (Saale), Germany
| | - Eunice van den Berg
- Department of Anatomical Pathology, University of the Witwatersrand, National Health Laboratory Service, Johannesburg, South Africa
| | - Annelle Zietsman
- AB May Cancer Centre, Windhoek Central Hospital, Windhoek, Namibia
| | - Martina Vetter
- Department of Gynecology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Sandy Kaufhold
- Department of Gynecology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Claudia Wickenhauser
- Institute of Pathology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Isabel Dos-Santos-Silva
- Department of Non-Communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine (LSHTM), London, UK
| | - Wenlong Carl Chen
- Global and Planetary Health Working Group, Institute of Medical Epidemiology, Biometrics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- National Cancer Registry, National Health Laboratory Service, Johannesburg, South Africa
| | - Herbert Cubasch
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nivashini Murugan
- Department of Surgery, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Valerie McCormack
- International Agency for Research on Cancer (IARC/WHO), Environment and Lifestyle Epidemiology Branch, Lyon, France
| | - Maureen Joffe
- Global and Planetary Health Working Group, Institute of Medical Epidemiology, Biometrics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Strengthening Oncology Services Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Noncommunicable Diseases Research Division, Wits Health Consortium (PTY) Ltd, University Witwatersrand, Johannesburg, South Africa
- Strengthening Oncology Services Research Unit,Faculty of Health Sciences, University Witwatersrand, Johannesburg, South Africa
| | - Barbara Seliger
- Medical Faculty, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany.
- Institute of Translational Immunology, Medical School Theodor Fontane, Brandenburg an der Havel, Germany.
| | - Eva Kantelhardt
- Global and Planetary Health Working Group, Institute of Medical Epidemiology, Biometrics and Informatics, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Department of Gynecology, University Hospital Halle, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
11
|
Lin YH, Chen CW, Chen MY, Xu L, Tian X, Cheung SH, Wu YL, Siriwon N, Wu SH, Mou KY. The Bacterial Outer Membrane Vesicle-Cloaked Immunostimulatory Nanoplatform Reinvigorates T Cell Function and Reprograms Tumor Immunity. ACS NANO 2025. [PMID: 40392526 DOI: 10.1021/acsnano.5c02541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
Bacterial outer membrane vesicles (OMVs) represent powerful immunoadjuvant nanocarriers with the capacity to reprogram the tumor microenvironment (TME) and activate immune responses. Here, we investigate a nanotherapeutic platform integrating immunostimulatory cytosine-phosphate-guanine oligodeoxynucleotides (CpG-ODNs, hereafter termed CpG) into mesoporous silica nanoparticles cloaked with OMVs (CpG@MSN-PEG/PEI@OMVs) for cancer immunotherapy. Systemic administration of these nanohybrids facilitates precise tumor targeting, induces antitumor cytokines such as IFNγ, and suppresses immunosuppressive cytokine TGF-β, reshaping the TME. Additionally, CpG@MSN-PEG/PEI@OMVs promote M1 macrophage polarization, dendritic cell maturation, and the generation of durable tumor-specific immune memory, resulting in pronounced tumor regression with minimal systemic toxicity. The platform demonstrates efficacy against metastatic and solid tumor models including 4T1 breast and MC38 colorectal cancers. Transcriptomic analyses reveal that CpG@MSN-PEG/PEI@OMVs enhance mitochondrial oxidative phosphorylation in T cells within tumor-draining lymph nodes, mitigating T cell exhaustion and restoring metabolic fitness. These results support the potential of CpG@MSN-PEG/PEI@OMVs as a modular nanoplatform to modulate innate and adaptive immunity in cancer immunotherapy.
Collapse
Affiliation(s)
- Yu-Han Lin
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 11529, Taiwan
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Chia-Wei Chen
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Mei-Yi Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| | - Li Xu
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Xuejiao Tian
- Research Center for Applied Sciences, Academia Sinica, Taipei 11529, Taiwan
- Nano Science and Technology Program, Taiwan International Graduate Program, Academia Sinica, Taipei, 11529, Taiwan
- Department of Engineering and System Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Siu-Hung Cheung
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Yen-Ling Wu
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Natnaree Siriwon
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samutprakarn 10540, Thailand
| | - Si-Han Wu
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program in Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Kurt Yun Mou
- Taiwan International Graduate Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei 11529, Taiwan
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
| |
Collapse
|
12
|
Krug A, Ernst LM, Mhaidly R, Ramis J, Gusta MF, Bastus NG, Martinez-Turtos A, Tosolini M, Di Mascio L, Tari G, Boyer L, Gaulard P, Lemonnier F, Ricci JE, Verhoeyen E, Puntes V. Scavenging Reactive Oxygen Species by Cerium Oxide Nanoparticles Prevents Death in a Peripheral T Cell Lymphoma Preclinical Mouse Model. ACS NANO 2025; 19:18644-18660. [PMID: 40346022 DOI: 10.1021/acsnano.5c02860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2025]
Abstract
Cancer cell survival and proliferation are correlated with increased metabolic activity and consequent oxidative stress, driving metabolic shifts that interfere with the immune response to malignant cells. This is the case of high-energy-demanding angioimmunoblastic T cell lymphoma (AITL), a highly aggressive cancer with poor survival rates, where malignant CD4+ PD-1high T cells show increased mitochondrial activity and Reactive oxygen species (ROS) accumulation. Here, we report that administration of ROS scavenging cerium oxide (CeO2) nanoparticles in an AITL preclinical mouse model leads to their preferential accumulation in the spleen, where the CD4+ PD-1high T cells driving malignancy were significantly reduced. This was accompanied by activation of previously exhausted cytotoxic CD8+ T cells, restoring their potent antitumor function. As a result, survival rates dramatically increase with no observed toxicity to healthy cells or tissues. Overall, it highlights the correlation between increased energy demand, increased mitochondrial mass, increased PD-1 expression, increased ROS production, and immune suppression and how this vicious loop can be stopped by scavenging ROS.
Collapse
Affiliation(s)
- Adrien Krug
- Université Côte d'Azur, INSERM, C3M, Nice 06204, France
- Equipe labellisée Ligue Contre le Cancer, Nice 06204, France
| | - Lena M Ernst
- Vall d'Hebron Research Institute (VHIR), Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, Barcelona 08193, Spain
- Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona 08010, Spain
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Rana Mhaidly
- Université Côte d'Azur, INSERM, C3M, Nice 06204, France
- Equipe labellisée Ligue Contre le Cancer, Nice 06204, France
| | - Joana Ramis
- Vall d'Hebron Research Institute (VHIR), Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, Barcelona 08193, Spain
- Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona 08010, Spain
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Muriel F Gusta
- Vall d'Hebron Research Institute (VHIR), Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, Barcelona 08193, Spain
- Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona 08010, Spain
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Neus G Bastus
- Vall d'Hebron Research Institute (VHIR), Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, Barcelona 08193, Spain
- Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona 08010, Spain
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| | | | - Marie Tosolini
- CRCT, Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse 31100, France
| | - Léa Di Mascio
- Université Côte d'Azur, INSERM, C3M, Nice 06204, France
- Equipe labellisée Ligue Contre le Cancer, Nice 06204, France
| | - Gamze Tari
- INSERMU955; Unité hémopathies lymphoïdes, Hôpitaux Universitaires Henri Mondor, Assistance publique des Hôpitaux de Paris, Université Paris-Est Créteil; Institut Mondor de Recherche Biomédicale, Créteil F-94010, France
| | - Laurent Boyer
- Université Côte d'Azur, INSERM, C3M, Nice 06204, France
| | - Philippe Gaulard
- département de pathologie, AP-HP, Groupe hospitalo-universitaire Chenevier Mondor, Créteil F-94010, France
- Service Unité Hémopathies Lymphoides, AP-HP, Groupe hospitalo-universitaire Chenevier Mondor, Créteil F-94010, France
| | - François Lemonnier
- INSERMU955; Unité hémopathies lymphoïdes, Hôpitaux Universitaires Henri Mondor, Assistance publique des Hôpitaux de Paris, Université Paris-Est Créteil; Institut Mondor de Recherche Biomédicale, Créteil F-94010, France
| | - Jean-Ehrland Ricci
- Université Côte d'Azur, INSERM, C3M, Nice 06204, France
- Equipe labellisée Ligue Contre le Cancer, Nice 06204, France
| | - Els Verhoeyen
- Université Côte d'Azur, INSERM, C3M, Nice 06204, France
- Equipe labellisée Ligue Contre le Cancer, Nice 06204, France
- CIRI, Université de Lyon; INSERM U1111; ENS de Lyon; University Lyon1; CNRS UMR5308, Lyon 69007, France
| | - Victor Puntes
- Vall d'Hebron Research Institute (VHIR), Passeig Vall d'Hebron 119-129, Barcelona 08035, Spain
- Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and BIST, Campus UAB, Bellaterra, Barcelona 08193, Spain
- Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona 08010, Spain
- Networking Research Centre for Bioengineering, Biomaterials, and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
| |
Collapse
|
13
|
Yodweerapong T, Ueno Y, Yamaguchi R, Yarangsee P, Kimura KI, Kataoka T. Kujigamberol Inhibits IFN-γ and IL-2 mRNA Expression and NFATc2 Binding to Their Promoters in Response to a Phorbol Ester and Ionomycin Stimulation. Molecules 2025; 30:2214. [PMID: 40430387 PMCID: PMC12114272 DOI: 10.3390/molecules30102214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Revised: 05/12/2025] [Accepted: 05/15/2025] [Indexed: 05/29/2025] Open
Abstract
Kujigamberol, a dinorlabdane compound isolated from Kuji amber, exerts multiple biological effects, including anti-allergic and anti-inflammatory activities. The present study demonstrated that kujigamberol inhibited cytokine production by T cells. In response to a phorbol 12-myristate 13-acetate (PMA) and ionomycin (IM) stimulation, kujigamberol suppressed interferon-γ (IFN-γ) and interleukin-2 (IL-2) mRNA expression in murine T-cell lymphoma BW5147 cells stably transfected with the T-box transcription factor eomesodermin. IL-4 and Fas ligand mRNA expression was also inhibited by kujigamberol. In the murine cytotoxic T-cell line CTLL-2, kujigamberol more strongly decreased IFN-γ mRNA expression induced by IM alone than that induced by the combination of PMA and IM. A luciferase reporter assay showed that kujigamberol preferentially reduced nuclear factor of activated T cell (NFAT)-dependent transcription in human embryonic kidney 293T cells. Unlike the calcineurin inhibitor FK506, kujigamberol did not markedly affect NFATc2 protein levels in BW5147 cells but interfered with the binding of NFATc2 to the IFN-γ and IL-2 promoters. These results indicate that kujigamberol inhibited IFN-γ and IL-2 mRNA expression by preventing the binding of NFATc2 to their promoters; therefore, it has potential as an immunosuppressive agent.
Collapse
Affiliation(s)
- Tanpitcha Yodweerapong
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Yuto Ueno
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Rikako Yamaguchi
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Piimwara Yarangsee
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| | - Ken-ichi Kimura
- The United Graduate School of Agricultural Sciences, Iwate University, 3-18-8 Ueda, Morioka 020-8550, Japan
| | - Takao Kataoka
- Department of Applied Biology, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
- Center for Social and Biomedical Engineering, Kyoto Institute of Technology, Matsugasaki, Sakyo-ku, Kyoto 606-8585, Japan
| |
Collapse
|
14
|
Xu L, Xiao T, Chao T, Xiong H, Yao W. From genes to therapy: a lipid Metabolism-Related genetic risk model predicts HCC outcomes and enhances immunotherapy. BMC Cancer 2025; 25:895. [PMID: 40389832 PMCID: PMC12090435 DOI: 10.1186/s12885-025-14306-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 05/09/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Hepatocellular Carcinoma (HCC) is related to dysregulated lipid metabolism and immunosuppressive microenvironment. This study developed a genetic risk model using lipid metabolism-related genes to predict survival and immune patterns in HCC patients. METHODS Differentially expressed genes (DEGs) related to lipid metabolism were identified in HCC via the TCGA-LIHC dataset. A risk model for survival prediction was constructed via DEGs related to survival. The immune signature associated with the risk model was also evaluated by the CIBERSORT algorithm, tumor immune dysfunction and exclusion algorithm, and single sample gene set enrichment analysis. RESULTS This study identified six lipid metabolism-related genes, ADH4, LCAT, CYP2C9, CYP17A1, LPCAT1, and ACACA, to construct a lipid metabolism-related gene risk model that can divide HCC patients into low- and high-risk groups. Internal and external validation verified that the risk model could be a signature that could effectively predict HCC patient prognosis. High-risk patients showed disrupted immune cell profiles, reduced tumor-killing capacity, and increased expression of immune checkpoint genes. However, they responded more favorably to immune checkpoint inhibitor (ICB) therapy. The top ten hub genes related to the risk model were associated with tumor progression and deteriorating prognosis. In vitro experiments verified that the downregulation of the top 1 hub gene CDK1 was correlated to the HCC cell proliferation. CONCLUSION The risk model constructed using lipid metabolism-related genes could effectively predict prognosis and was related to the immunosuppressive microenvironment and ICB immunotherapy. The hub genes related to the risk model were potential therapeutic targets.
Collapse
Affiliation(s)
- Lei Xu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Ting Xiao
- Department of Ultrasonography, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Tengfei Chao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Wei Yao
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
15
|
Zhang ZC, Shen Y, Lin YS, Yang B, Cao J, Li J, Zhao WB. Peptide-MHC I regulatory mechanisms and intervention strategies in anti-tumor T cell immunity. Acta Pharmacol Sin 2025:10.1038/s41401-025-01574-y. [PMID: 40379886 DOI: 10.1038/s41401-025-01574-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 04/22/2025] [Indexed: 05/19/2025]
Abstract
T cell immune responses are triggered by antigenic peptides presented through major histocompatibility complex class Is (pMHC-Is), which play an important role in the genesis, development, and therapy of tumors. The capacity of a specific pMHC-I to elicit T cell responses is deeply influenced by its expression level (quantity) and its immunogenicity (quality). Tumor cells can evade T cell immunity by down-regulating the quantity of pMHC-Is or selectively eliminating highly immunogenic antigenic peptides. Augmenting the quantity or quality of pMHC-Is is essential for tumor immunotherapy. However, the complexity of pMHC-I regulation and tumor heterogeneity pose challenges to clinical strategies. Consequently, developing approaches grounded in comprehensive analyses of pMHC-I regulatory mechanisms remains a focal point in the research of T cell immunity. In this review, we discuss how tumors modulate their surface pMHC-Is through genetic, epigenetic, and proteomic mechanisms and summarize potential therapeutic strategies targeting these mechanisms, which may provide a valuable reference for the development of novel tumor immunotherapies based on pMHC-I modulation. Tumor cells can achieve immune escape by interfering with the quantity and quality of pMHC-Is, and corresponding immunotherapy can also be achieved by the regulation of pMHC-Is.
Collapse
Affiliation(s)
- Zhi-Chao Zhang
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, China
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ying Shen
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, China
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310018, China
| | - Yu-Shen Lin
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, China
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Bo Yang
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, China
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310018, China
- School of Medicine, Hangzhou City University, Hangzhou, 310015, China
| | - Ji Cao
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, China.
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310018, China.
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
| | - Jun Li
- Cancer Center, Zhejiang University, Hangzhou, 310058, China.
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences, Hangzhou, 310009, China.
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, 310000, China.
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| | - Wen-Bin Zhao
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, 310018, China.
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China.
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, 310018, China.
| |
Collapse
|
16
|
Gill GS, Kharb S, Goyal G, Das P, Kurdia KC, Dhar R, Karmakar S. Immune Checkpoint Inhibitors and Immunosuppressive Tumor Microenvironment: Current Challenges and Strategies to Overcome Resistance. Immunopharmacol Immunotoxicol 2025:1-45. [PMID: 40376861 DOI: 10.1080/08923973.2025.2504906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 05/06/2025] [Indexed: 05/18/2025]
Abstract
Immune checkpoint inhibitors (ICIs) are shown to improve cancer treatment effectiveness by boosting the immune system of the patient. Nevertheless, the unique and highly suppressive TME poses a significant challenge, causing heterogeneity of response or resistance in a considerable number of patients. This review focuses on the evasive attributes of the TME. Immune evasion mechanism in TME include immunosuppressive cells, cytokine and chemokine signaling, metabolic alterations and overexpression of immune checkpoint molecules such as PD-1, CTLA-4, LAG-3, TIM-3, TIGIT, BTLA and their interactions within the TME. In addition, this review focuses on the overcoming resistance by targeting immunosuppressive cells, normalizing tumor blood vessels, blocking two or three checkpoints simultaneously, combining vaccines, oncolytic viruses and metabolic inhibitors with ICIs or other therapies. This review also focuses on the necessity of finding predictive markers for the stratification of patients and to check response of ICIs treatment. It remains to be made certain by new research and intelligent innovations how these discoveries of the TME and its interplay facilitate ICI treatment and change the face of cancer treatment.
Collapse
Affiliation(s)
- Gurpreet Singh Gill
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Simmi Kharb
- Department of Biochemistry, Pt. B.D. Sharma Postgraduate Institute of Medical Sciences, Rohtak, India
| | - Gitanjali Goyal
- Department of Biochemistry, All India Institute of Medical Sciences, Bathinda, India
| | - Prasenjit Das
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Kailash Chand Kurdia
- Department of GI Surgery & Liver Transplantation, All India Institute of Medical Sciences, New Delhi, India
| | - Ruby Dhar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
17
|
Aindelis G, Spyridopoulou K, Kyriakou S, Tiptiri-Kourpeti A, Panayiotidis MI, Pappa A, Chlichlia K. Evaluating the Chemical Composition and Antitumor Activity of Origanum vulgare ssp. hirtum Essential Oil in a Preclinical Colon Cancer Model. Int J Mol Sci 2025; 26:4737. [PMID: 40429884 PMCID: PMC12111866 DOI: 10.3390/ijms26104737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 05/05/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
Origanum vulgare ssp. hirtum is an aromatic plant native to various Mediterranean regions and has been traditionally used in folk medicine. This study investigates the chemical composition and the potential antitumor activity of its essential oil in a preclinical model of CT26 colorectal cancer in BALB/c mice. Mice received prophylactic oral administration of the essential oil, and tumor progression, immune modulation, and apoptosis were evaluated. Even treatment with low doses (350 parts per million, ppm in 100 μL final volume) of the essential oil significantly suppressed tumor growth by approximately 44%. This effect correlated with the enhanced expression of antitumorigenic cytokines, including a 2.7-fold increase in type I interferons (IFN), IFN-γ (from 46.5 to 111.9 pg/μL per mg of protein) and tumor necrosis factor alpha (TNF-α) (from 34.5 to 103 pg/μL per mg of protein). Furthermore, the production of granzyme B, a key mediator of cytotoxic immune cell function, was notably increased from 96.1 to 319.6 pg/μL per mg of protein. An elevated activation of caspase 3, a central effector caspase of all apoptotic cascades, was also observed in tumors from oregano-treated mice. These findings suggest that O. vulgare ssp. hirtum essential oil exhibits promising antitumor properties through immune modulation and immunity-mediated apoptosis induction, supporting its potential development as a bioactive compound for cancer prevention or therapy.
Collapse
Affiliation(s)
- Georgios Aindelis
- Department of Molecular Biology and Genetics, School of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (G.A.); (K.S.); (A.T.-K.); (A.P.)
| | - Katerina Spyridopoulou
- Department of Molecular Biology and Genetics, School of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (G.A.); (K.S.); (A.T.-K.); (A.P.)
| | - Sotiris Kyriakou
- Department of Cancer Genetics, Therapeutics & Ultrastructural Pathology, The Cyprus Institute of Neurology & Genetics, Nicosia 2371, Cyprus;
| | - Angeliki Tiptiri-Kourpeti
- Department of Molecular Biology and Genetics, School of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (G.A.); (K.S.); (A.T.-K.); (A.P.)
| | - Mihalis I. Panayiotidis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA;
| | - Aglaia Pappa
- Department of Molecular Biology and Genetics, School of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (G.A.); (K.S.); (A.T.-K.); (A.P.)
| | - Katerina Chlichlia
- Department of Molecular Biology and Genetics, School of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (G.A.); (K.S.); (A.T.-K.); (A.P.)
| |
Collapse
|
18
|
Kim SY, Yi JM, Chun J, Park M, Yeo H, Park SM, Jeong MK. Deciphering the immunomodulatory mechanisms of Bojungikki-tang via systematic transcriptomic and immune cell interaction network analysis. Biomed Pharmacother 2025; 188:118129. [PMID: 40378772 DOI: 10.1016/j.biopha.2025.118129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/30/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025] Open
Abstract
Bojungikki-tang (BJIKT), a traditional herbal formula with immunomodulatory properties, has synergistic effects with immune checkpoint inhibitors. However, the detailed molecular mechanisms underlying its effects on various immune cell types remain largely unexplored. Therefore, in this study, we aimed to propose a framework for understanding how herbal medicine modulates immunity through a systematic analysis of the drug-induced transcriptome and immune cell interaction networks. We obtained large-scale RNA-seq data for distinct five immune cell types (T cells, natural killer cells, B cells, macrophages, and dendritic cells) treated with BJIKT and its four major constituent herbs, totaling 180 sequenced samples. Transcriptomic analysis indicated that BJIKT significantly upregulated interferon-γ, TNF-α, and inflammatory pathways in B cells, macrophages, and dendritic cells. Although BJIKT did not directly activate T cells, it indirectly modulated their function through immune cell-cell interactions. The reconstructed network and cytokine assays identified IL-1β, IL-6, IL-8, MIP-1β, CXCL9, CXCL10, and TNF-α as critical cytokines influenced by BJIKT, playing pivotal roles in activating and recruiting various immune cells. Our findings provide insights into the immunomodulatory mechanisms of BJIKT and the unique actions of its constituent herbs. We highlight the potential for combining traditional herbal medicine with omics technologies to explore the therapeutic mechanisms of natural products as complementary therapies for advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Sang-Yun Kim
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jin-Mu Yi
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Jaemoo Chun
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Musun Park
- KM Data Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Heerim Yeo
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Sang-Min Park
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea.
| | - Mi-Kyung Jeong
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea.
| |
Collapse
|
19
|
Enduru N, Manuel AM, Zhao Z. Genetic, Transcriptomic, and Epigenomic Insights into Sjögren's Disease: An Integrative Network Investigation and Immune Diseases Comparison. Int J Mol Sci 2025; 26:4637. [PMID: 40429780 PMCID: PMC12111751 DOI: 10.3390/ijms26104637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/25/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Sjögren's disease (SjD) is a systemic autoimmune disorder primarily causing dry eyes and mouth. It frequently overlaps with other autoimmune diseases (AIDs), including rheumatoid arthritis (RA) and systemic lupus erythematosus (SLE). However, the genetic basis of SjD remains underexplored, limiting our understanding of its connections to other immune-mediated conditions. In this study, we aimed to identify gene networks associated with SjD through the integration of genetic, transcriptomic, and epigenomic data. We further compared the genetic factors of SjD with other immune-mediated diseases. We analyzed genome-wide association studies (GWAS) summary statistics, DNA methylation, and transcriptomic data using our in-house network-based methods, dmGWAS and EW_dmGWAS, to identify key gene modules associated with SjD. In dmGWAS analysis, discovery and evaluation datasets were used to identify consensus results. We conducted gene-set, cell-type, and disease-enrichment analyses on significant gene modules and explored potential drug targets. Genetic correlations and Mendelian randomization were applied to assess SjD's link with 17 other AIDs and 16 cancer types. dmGWAS identified 207 and 211 gene modules in the discovery and evaluation phases, respectively, while EW_dmGWAS detected 886 modules. Key modules highlighted 55 genes (discovery), 52 genes (evaluation), and 59 genes (EW_dmGWAS), with at least 50 genes from each analysis linked to AIDs and cancer. Enrichment analyses confirmed their relevance to immune and oncogenic pathways. We pinpointed four candidate drug targets associated with AIDs. We developed a novel integrative omics approach to identify potential genetic markers of SjD and compared them with AIDs and cancers. Our approach can be similarly applied to other disease studies.
Collapse
Affiliation(s)
- Nitesh Enduru
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (N.E.); (A.M.M.)
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Astrid M. Manuel
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (N.E.); (A.M.M.)
- Molecular & Human Genetics Department, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zhongming Zhao
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA; (N.E.); (A.M.M.)
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| |
Collapse
|
20
|
Murray NP. Immunomodulation and Immunotherapy for Patients with Prostate Cancer: An Up-to-Date Review. Biomedicines 2025; 13:1179. [PMID: 40427006 PMCID: PMC12109314 DOI: 10.3390/biomedicines13051179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Immunotherapy alone or in combination with chemotherapy or radiotherapy is the frontline treatment for melanoma and lung cancer. However, its role in prostate cancer is usually as a fourth-line treatment. It is usually employed in patients with metastasis, after androgen blockade and chemotherapy. This article reviews the immunosuppressive effects of prostate cancer and possible uses of various types of immunotherapies. It also considers when would be the optimal time to employ this type of therapy.
Collapse
Affiliation(s)
- Nigel P. Murray
- Faculty of Medicine, Universidad Finis Terrae, Santiago 7501015, Chile;
- Department of Medicine, Hospital de Carabineros de Chile, Santiago 7770199, Chile
| |
Collapse
|
21
|
Coelho KBCA, Wosniaki DK, Pereira JL, Luz M, Albrecht L, Nardin JM, Aoki MN, Reis LO, dos Reis RB, Zanette DL. Comparative Analysis of Cytokine Expression Profiles in Prostate Cancer Patients. BIOLOGY 2025; 14:505. [PMID: 40427694 PMCID: PMC12109277 DOI: 10.3390/biology14050505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/24/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025]
Abstract
This study aimed to identify the cytokine expression profile in prostate cancer (PCa) patients compared to healthy individuals. Plasma samples from 75 PCa patients and 14 healthy controls were analyzed using Multiplex ELISA (Luminex) to measure the expression levels of 12 cytokines: IL-4, IL-5, IL-6, IL-10, IL-1β, IL-17A, IL-12p70, MCP-1/CCL2, MIP-1α/CCL3, MIP-1β/CCL4, TNF-α, and IFN-γ. Differences in cytokine expression levels were analyzed using the Mann-Whitney test, Wilcoxon's rank-sum test, Spearman's rank correlation, and K-means Clustering unsupervised machine learning to validate cytokine expression patterns. In PCa patients, MIP-1α/CCL3, MIP-1β/CCL4, IFN-γ, and interleukins exhibited significantly higher expression levels; conversely, TNF-α and MCP-1/CCL2 both had decreased expression compared to healthy individuals. The clustering analysis confirmed that PCa patients exclusively exhibit the highest associations with MIP-1α/CCL3, IFN- γ, IL-12p70, IL-4, and IL-5. Furthermore, specific correlations between IL-4 and MIP-1 beta, IL-4 and IFN-gamma, IL-5 and IL-12p70, and IL-5 and IFN-gamma in PCa patients did not occur in healthy individuals. Such results will guide forthcoming in vitro and in vivo human prostate cancer-drug treatment models, paving the way for exploration of future drug targets and candidates with potential to predict FDA-approved prostate cancer treatment responses by targeting cytokine levels and the oncogenesis pathways.
Collapse
Affiliation(s)
- Karoline Brito Caetano Andrade Coelho
- Uro-Oncology Laboratory, Surgery and Anatomy Department, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirão Preto 14090-000, SP, Brazil; (K.B.C.A.C.); (R.B.d.R.)
| | - Denise Kusma Wosniaki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81350-010, PR, Brazil; (D.K.W.); (L.A.); (M.N.A.)
| | | | - Murilo Luz
- Erasto Gaertner Hospital, Curitiba 81520-060, PR, Brazil; (J.L.P.); (M.L.)
| | - Letusa Albrecht
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81350-010, PR, Brazil; (D.K.W.); (L.A.); (M.N.A.)
| | - Jeanine Marie Nardin
- School of Medicine and Life Sciences, Pontifícia Universidade Católica do Paraná, Curitiba 87013-250, PR, Brazil;
| | - Mateus Nobrega Aoki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81350-010, PR, Brazil; (D.K.W.); (L.A.); (M.N.A.)
| | - Leonardo O. Reis
- UroScience, State University of Campinas, Unicamp, Campinas 13083-872, SP, Brazil
- ImmunOncology, Pontifical Catholic University of Campinas, PUC-Campinas, Campinas 13087-571, SP, Brazil
- UroGen, National Institute of Science, Technology and Innovation in Genitourinary Cancer (INCT), Campinas 13087-571, SP, Brazil
| | - Rodolfo Borges dos Reis
- Uro-Oncology Laboratory, Surgery and Anatomy Department, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirão Preto 14090-000, SP, Brazil; (K.B.C.A.C.); (R.B.d.R.)
- UroGen, National Institute of Science, Technology and Innovation in Genitourinary Cancer (INCT), Campinas 13087-571, SP, Brazil
| | - Dalila Lucíola Zanette
- Uro-Oncology Laboratory, Surgery and Anatomy Department, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirão Preto 14090-000, SP, Brazil; (K.B.C.A.C.); (R.B.d.R.)
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba 81350-010, PR, Brazil; (D.K.W.); (L.A.); (M.N.A.)
- UroGen, National Institute of Science, Technology and Innovation in Genitourinary Cancer (INCT), Campinas 13087-571, SP, Brazil
| |
Collapse
|
22
|
Aquino A, Franzese O. Reciprocal Modulation of Tumour and Immune Cell Motility: Uncovering Dynamic Interplays and Therapeutic Approaches. Cancers (Basel) 2025; 17:1547. [PMID: 40361472 PMCID: PMC12072109 DOI: 10.3390/cancers17091547] [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: 03/31/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Dysregulated cell movement is a hallmark of cancer progression and metastasis, the leading cause of cancer-related mortality. The metastatic cascade involves tumour cell migration, invasion, intravasation, dissemination, and colonisation of distant organs. These processes are influenced by reciprocal interactions between cancer cells and the tumour microenvironment (TME), including immune cells, stromal components, and extracellular matrix proteins. The epithelial-mesenchymal transition (EMT) plays a crucial role in providing cancer cells with invasive and stem-like properties, promoting dissemination and resistance to apoptosis. Conversely, the mesenchymal-epithelial transition (MET) facilitates metastatic colonisation and tumour re-initiation. Immune cells within the TME contribute to either anti-tumour response or immune evasion. These cells secrete cytokines, chemokines, and growth factors that shape the immune landscape and influence responses to immunotherapy. Notably, immune checkpoint blockade (ICB) has transformed cancer treatment, yet its efficacy is often dictated by the immune composition of the tumour site. Elucidating the molecular cross-talk between immune and cancer cells, identifying predictive biomarkers for ICB response, and developing strategies to convert cold tumours into immune-active environments is critical to overcoming resistance to immunotherapy and improving patient survival.
Collapse
Affiliation(s)
| | - Ornella Franzese
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
| |
Collapse
|
23
|
Yabuki Y, Mitsuhashi A, Ogino H, Yoshida A, Nguyen NT, Yoneda H, Ozaki R, Tsukazaki Y, Morita Y, Nokihara H, Sato S, Shinohara T, Hanibuchi M, Nishioka Y. Hypoxia-inducible factor-targeting therapy augmented the sensitivity to programmed death ligand-1 blockade by enhancing interferon-γ-induced chemokines in tumor cells. Int J Cancer 2025; 156:1814-1825. [PMID: 39686841 DOI: 10.1002/ijc.35301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 10/06/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024]
Abstract
Immune checkpoint inhibitors (ICIs) targeting programmed death ligand-1 (PD-L1) provide clinical benefits for various advanced malignancies. However, the predictive factors that determine sensitivity to ICIs have not been fully elucidated. We focused on tumor-derived CXCL10/11 as a pivotal factor that determines the response to PD-L1 blockade by regulating T cell accumulation and tumor angiogenesis. We previously reported that CXCL10/11 was upregulated by interferon (IFN)-γ in ICI-sensitive tumor cells but not in ICI-resistant cells, including mouse Lewis lung carcinoma (LLC). In the present study, gene silencing of tumor-derived CXCL10/11 induced resistance to PD-L1 blockade in AB1-HA mesothelioma cell-bearing mice. To identify the mechanisms underlying ICI resistance, we performed a microarray analysis to compare the IFN-γ-inducible genes between ICI-sensitive AB1-HA and ICI-resistant LLC in vitro. A pathway analysis based on microarray data indicated that hypoxia-inducible factor (HIF) 1A is the key signal that inhibits CXCL10/11 expression. We revealed that the HIF1A inhibitors echinomycin (EC) and YC-1 upregulated CXCL10/11 genes induced by IFN-γ in tumor cells in vitro. In addition, combination therapy with PD-L1 blockade and EC demonstrated synergistic antitumor effects in LLC-bearing mice. Combination therapy enhanced tumor infiltration of CD8 T cells and suppressed tumor angiogenesis. The present study suggests that HIF1A signaling in tumor cells dominates ICI resistance via the downregulation of tumor-derived CXCL10/11.
Collapse
Affiliation(s)
- Yohei Yabuki
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Atsushi Mitsuhashi
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hirokazu Ogino
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Aito Yoshida
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Na Thi Nguyen
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hiroto Yoneda
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Ryohiko Ozaki
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yuki Tsukazaki
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yutaka Morita
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hiroshi Nokihara
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Seidai Sato
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Tsutomu Shinohara
- Department of Community Medicine for Respirology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Masaki Hanibuchi
- Department of Community Medicine for Respirology, Hematology and Metabolism, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Yasuhiko Nishioka
- Department of Respiratory Medicine and Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
- Department of Community Medicine for Rheumatology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
24
|
Safaei S, Yari A, Pourbagherian O, Maleki LA. The role of cytokines in shaping the future of Cancer immunotherapy. Cytokine 2025; 189:156888. [PMID: 40010034 DOI: 10.1016/j.cyto.2025.156888] [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: 10/28/2024] [Revised: 01/13/2025] [Accepted: 02/05/2025] [Indexed: 02/28/2025]
Abstract
As essential immune system regulators, cytokines are essential for modulating both innate and adaptive immunological responses. They have become important tools in cancer immunotherapy, improving the immune system's capacity to identify and destroy tumor cells. This article examines the background, workings, and therapeutic uses of cytokines, such as interleukins, interferons, and granulocyte-macropHage colony-stimulating factors, in the management of cancer. It examines the many ways that cytokines affect immune cell activation, signaling pathways, tumor development, metastasis, and prognosis by modifying the tumor microenvironment. Despite the limited effectiveness of cytokine-based monotherapy, recent developments have concentrated on new fusion molecules such as immunocytokines, cytokine delivery improvements, and combination techniques to maximize treatment efficacy while reducing adverse effects. Current FDA-approved cytokine therapeutics and clinical trial results are also included in this study, which offers insights into how cytokines might be used with other therapies including checkpoint inhibitors, chemotherapy, and radiation therapy to address cancer treatment obstacles. This study addresses the intricacies of cytokine interactions in the tumor microenvironment, highlighting the possibility for innovative treatment methods and suggesting fresh techniques for enhancing cytokine-based immunotherapies. PEGylation, viral vector-mediated cytokine gene transfer, antibody-cytokine fusion proteins (immunocytokines), and other innovative cytokine delivery techniques are among the novelties of this work, which focuses on the most recent developments in cytokine-based immunotherapy. Additionally, the study offers a thorough examination of the little-reviewed topic of cytokine usage in conjunction with other treatment techniques. It also discusses the most recent clinical studies and FDA-approved therapies, providing a modern perspective on the developing field of cancer immunotherapy and suggesting creative ways to improve treatment effectiveness while lowering toxicity. BACKGROUND: Cytokines are crucial in cancer immunotherapy for regulating immune responses and modifying the tumor microenvironment (TME). However, challenges with efficacy and safety have driven research into advanced delivery methods and combination therapies to enhance their therapeutic potential.
Collapse
Affiliation(s)
- Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - AmirHossein Yari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Omid Pourbagherian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
25
|
Gopalakrishnan R, Wang Y, Kapczinski F, Frey BN, Wollenhaupt-Aguiar B. Peripheral protein inflammatory biomarkers in bipolar disorder and major depressive disorder: A systematic review and meta-analysis. J Affect Disord 2025; 376:149-168. [PMID: 39894226 DOI: 10.1016/j.jad.2025.01.150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 01/18/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
OBJECTIVES Bipolar disorder (BD) and major depressive disorder (MDD) are mood disorders. The most frequent clinical presentation of BD and MDD is depression, which contributes to high rates of misdiagnosis between disorders. To support diagnostic discrimination and therapeutic stratification, we aim to perform a systematic review and meta-analysis evaluating peripheral protein inflammatory biomarkers between BD and MDD, with a focus on the depressive state. METHODS We conducted a literature search on PubMed, PsycInfo and Embase with no year/language restrictions. Original studies including human participants with a BD or MDD diagnosis which directly compared levels of peripheral protein inflammatory biomarkers between groups were included. A random effects meta-analysis was performed. RESULTS 35 studies were included in the systematic review. 9 studies were included in the meta-analysis. The meta-analysis showed IL-7 (p < 0.01) levels were significantly decreased in BD, and IL-9 (p < 0.01), CCL3 (p = 0.03), CCL4 (p = 0.01), CCL5 (p = 0.02) and CCL11 (p = 0.04) levels were significantly increased in BD. LIMITATIONS High heterogeneity and limited dataset size restricted our meta-analysis to a small subset of biomarkers and limited our exploration of the effects of moderator variables. CONCLUSION This study found differences in IL-7, IL-9, CCL3, CCL4, CCL5 and CCL11 between BD and MDD in a depressive state. These findings support the notion that inflammation is associated with mood disorder pathophysiology, particularly with respect to T-cell network dysregulation. Further studies can assist in better understanding differences between disorders and work towards clinical applications.
Collapse
Affiliation(s)
- Ridhi Gopalakrishnan
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Centre for Clinical Neurosciences, McMaster University, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Yifan Wang
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Centre for Clinical Neurosciences, McMaster University, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Flavio Kapczinski
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Department of Psychiatry, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Benicio N Frey
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada; Centre for Clinical Neurosciences, McMaster University, St. Joseph's Healthcare, Hamilton, ON, Canada; Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Bianca Wollenhaupt-Aguiar
- Centre for Clinical Neurosciences, McMaster University, St. Joseph's Healthcare, Hamilton, ON, Canada; Mood Disorders Program and Women's Health Concerns Clinic, St. Joseph's Healthcare, Hamilton, ON, Canada.
| |
Collapse
|
26
|
Lee SW, Yun JS, Kim YJ, Jeong S, Noh JE, Kim HO, Cho HJ, Park CK, Oh IJ, Cho JH. Progressive accumulation of circulating CD27 -CD28 - effector/memory CD8 + T cells in patients with lung cancer blunts responses to immune checkpoint inhibitor therapy. Exp Mol Med 2025:10.1038/s12276-025-01448-7. [PMID: 40307573 DOI: 10.1038/s12276-025-01448-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 12/30/2024] [Accepted: 03/03/2025] [Indexed: 05/02/2025] Open
Abstract
Suppression of tumor-reactive CD8+ T cells is common within the tumor microenvironment. However, little is known about how tumors systemically affect the overall CD8+ T cell compartment. Here we demonstrate that peripheral blood CD8+ T cells from patients with lung cancer showed altered compositions particularly within CD45RA-CCR7- effector memory subpopulation. Specifically, patients with lung cancer exhibited increased frequency of more differentiated effector memory cells, which are less susceptible to T cell-receptor-induced proliferation. Further analysis using single-cell RNA sequencing revealed that these alterations were correlated with reduced quiescence and increased spontaneous activation at a systemic level, indicative of homeostatic dysregulation of the entire CD8+ T cell population. This phenomenon was found to be correlated with a poor clinical response to immune checkpoint inhibitor therapy across four independent cohorts, consisting of a total of 224 patients with lung cancer. These findings suggest that lung cancers continue to counteract potentially tumor-reactive CD8+ T cells by inducing homeostatic dysregulation of the entire CD8+ T cell compartment systematically.
Collapse
Affiliation(s)
- Sung-Woo Lee
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Ju Sik Yun
- Department of Thoracic and Cardiovascular Surgery, Chonnam National University Medical School, Hwasun Hospital, Gwangju, Republic of Korea
| | - Young Ju Kim
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Gwangju, Republic of Korea
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Gwangju, Republic of Korea
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Saei Jeong
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Gwangju, Republic of Korea
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Gwangju, Republic of Korea
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Jeong Eun Noh
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Gwangju, Republic of Korea
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Gwangju, Republic of Korea
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hee-Ok Kim
- Selecxine Inc., Seoul, Republic of Korea
| | - Hyun-Ju Cho
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun Hospital, Gwangju, Republic of Korea
| | - Cheol-Kyu Park
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun Hospital, Gwangju, Republic of Korea
| | - In-Jae Oh
- Department of Internal Medicine, Chonnam National University Medical School, Hwasun Hospital, Gwangju, Republic of Korea.
| | - Jae-Ho Cho
- Department of Microbiology and Immunology, Chonnam National University Medical School, Gwangju, Republic of Korea.
- Medical Research Center for Combinatorial Tumor Immunotherapy, Chonnam National University Medical School, Gwangju, Republic of Korea.
- National Immunotherapy Innovation Center, Chonnam National University Medical School, Gwangju, Republic of Korea.
- BioMedical Sciences Graduate Program, Chonnam National University Medical School, Gwangju, Republic of Korea.
| |
Collapse
|
27
|
Cai Z, Meng K, Yu T, Xi Y, Yuan Z, Wang X, Wang C, Li L, Fu X. IFN-γ-mediated suppression of ANGPT2-Tie2 in endothelial cells facilitates tumor vascular normalization during immunotherapy. Front Immunol 2025; 16:1551322. [PMID: 40370455 PMCID: PMC12075545 DOI: 10.3389/fimmu.2025.1551322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
Introduction Tumor angiogenesis is a critical biological hallmark of cancer, which involves multiple molecularly regulated signaling pathways, including the angiopoietin (ANGPT)-Tie2 and the vascular endothelial growth factor (VEGF) signaling pathways. Despite initial optimism, targeting tumor angiogenesis in the treatment of lung adenocarcinoma (LUAD) has been unsatisfactory. Currently, monotherapy with PD-1/PD-L1 inhibitors, or their combination with bevacizumab, is considered the standard therapeutic approach for LUAD. Recent studies have shown that immunotherapy suppresses tumor angiogenesis and facilitates vascular normalization. However, whether and how anti-PD-L1 therapy influences tumor vasculature remains unclear. Methods To investigate the impact of immunotherapy on the vasculature of LUAD, a mouse model of lung adenocarcinoma was established by subcutaneous implantation of Lewis lung carcinoma cells in vivo. The effects of different treatments on microvessel density and pericyte coverage were explored, and the expression of angiogenesis-related factors was analyzed. Furthermore, to explore the molecular mechanisms through which IFN-γ regulates tumor blood vessels during immunotherapy, we elucidated the specific mechanisms in vitro by means of techniques such as siRNA, ChIP, RT-qPCR, Western blot, and immunofluorescence. Finally, the effects of IFN-γ on the proliferation, migration, and angiogenic function of endothelial cells (ECs) were evaluated through CCK-8, Transwell, and HUVEC tube formation assays. Results Employing a mouse model of LUAD, we demonstrated that PD-L1 blockade therapy inhibits tumor angiogenesis and normalizes vasculature in an IFN-γ-signaling-dependent manner. Notably, anti-PD-L1 therapy reduced Tie2 and ANGPT2 expression, and these effects were reversed by the JAK1/2 inhibitor. Mechanistically, we demonstrated that IFN-γ inhibited Tie2 and ANGPT2 expression in ECs, and suppressed ANGPT2 gene transcription through the AKT-FOXO1 signaling pathway. Interestingly, IFN-γ-mediated activation of STAT1 exerts negative regulation by directly binding to the promoter regions of the ANGPT2 and TEK genes. Functionally, IFN-γ limits the migration, proliferation, and tube formation of ECs. Discussion In conclusion, our results revealed a novel mechanism wherein IFN-γ-mediated inhibition of ANGPT2-Tie2 facilitates vascular normalization during immunotherapy in LUAD, which performs an essential function in the antitumor efficacy of immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lequn Li
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, China
| | - Xiangning Fu
- Thoracic Surgery Laboratory, Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, Wuhan, China
| |
Collapse
|
28
|
Sato J, Motai Y, Yamagami S, Win SY, Horio F, Saeki H, Maekawa N, Okagawa T, Konnai S, Ohashi K, Murata S. Programmed Cell Death-1 Expression in T-Cell Subsets in Chickens Infected with Marek's Disease Virus. Pathogens 2025; 14:431. [PMID: 40430752 PMCID: PMC12114408 DOI: 10.3390/pathogens14050431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/21/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Marek's disease virus (MDV) causes Marek's disease (MD) in chickens, characterized by malignant lymphomas and immunosuppression. Sporadic MD outbreaks continue to occur even among vaccinated flocks in certain regions due to the increased virulence of the field strains. However, the mechanisms of tumorigenesis and immunosuppression caused by MDV remain to be fully elucidated. We previously reported that the mRNA expression of programmed cell death 1 (PD-1), an immune checkpoint molecule, was increased in tumor lesions caused by MDV, and its expression was positively correlated with the mRNA expression of Meq, an MDV-specific oncogene. In this study, we characterized PD-1-expressing T-cell subsets in the spleen and tissues of chickens that developed tumors to investigate the association between PD-1 expression and immunosuppression. Flow cytometric analysis revealed that the proportion of PD-1-expressing CD4+ T-cells, which are targets of MDV tumorigenesis, increased in the spleen and tumor tissues of chickens with MD. The proportion of PD-1+ CD4+ T-cells was higher in Meq-expressing cells than in those that were not. In the spleens of chickens with MD, the proportions of PD-1-expressing cells were increased in CD8+ and γδ T-cells, which play pivotal roles in defense against MD pathogenesis, relative to those of spleens from uninfected chickens. Moreover, the proportion of PD-1+ CD8+ T-cells expressing interferon (IFN)-γ did not increase in the spleen of chickens with MD. Additionally, no difference in the proportion of IFN-γ+ γδ T-cells expressing and not expressing PD-1 was observed in the spleens of chickens with MD, although the proportion of IFN-γ+ γδ T-cells expressing PD-1 in the spleens of uninfected chickens was higher. The function of PD-1-expressing CD8+ and γδ T-cells in chickens may be impaired after developing MD, which may cause MDV-induced immunosuppression.
Collapse
Affiliation(s)
- Jumpei Sato
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Yoshinosuke Motai
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Shunsuke Yamagami
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Shwe Yee Win
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Fumiya Horio
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Hikaru Saeki
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Naoya Maekawa
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Tomohiro Okagawa
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Satoru Konnai
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
- Institute for Vaccine Research and Development, Hokkaido University, Sapporo 001-0021, Japan
- Veterinary Research Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
| | - Kazuhiko Ohashi
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
- Veterinary Research Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
- International Affairs Office, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
| | - Shiro Murata
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo 060-0818, Japan
- Veterinary Research Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo 001-0020, Japan
| |
Collapse
|
29
|
Hameed SA, Kolch W, Brennan DJ, Zhernovkov V. Direct cell interactions potentially regulate transcriptional programmes that control the responses of high grade serous ovarian cancer patients to therapy. Sci Rep 2025; 15:14484. [PMID: 40280979 PMCID: PMC12032223 DOI: 10.1038/s41598-025-98463-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 04/11/2025] [Indexed: 04/29/2025] Open
Abstract
The tumour microenvironment is composed of a complex cellular network involving cancer, stromal and immune cells in dynamic interactions. A large proportion of this network relies on direct physical interactions between cells, which may impact patient responses to clinical therapy. Doublets in scRNA-seq are usually excluded from analysis. However, they may represent directly interacting cells. To decipher the physical interaction landscape in relation to clinical prognosis, we inferred a physical cell-cell interaction (PCI) network from 'biological' doublets in a scRNA-seq dataset of approximately 18,000 cells, obtained from 7 treatment-naive ovarian cancer patients. Focusing on cancer-stromal PCIs, we uncovered molecular interaction networks and transcriptional landscapes that stratified patients in respect to their clinical responses to standard therapy. Good responders featured PCIs involving immune cells interacting with other cell types including cancer cells. Poor responders lacked immune cell interactions, but showed a high enrichment of cancer-stromal PCIs. To explore the molecular differences between cancer-stromal PCIs between responders and non-responders, we identified correlating gene signatures. We constructed ligand-receptor interaction networks and identified associated downstream pathways. The reconstruction of gene regulatory networks and trajectory analysis revealed distinct transcription factor (TF) clusters and gene modules that separated doublet cells by clinical outcomes. Our results indicate (i) that transcriptional changes resulting from PCIs predict the response of ovarian cancer patients to standard therapy, (ii) that immune reactivity of the host against the tumour enhances the efficacy of therapy, and (iii) that cancer-stromal cell interaction can have a dual effect either supporting or inhibiting therapy responses.
Collapse
Affiliation(s)
- Sodiq A Hameed
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland.
| | - Walter Kolch
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland
- Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland
| | - Donal J Brennan
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland
- UCD Gynaecological Oncology Group Catherine McAuley Research Centre, Mater Misericordiae University Hospital, Eccles Street, Dublin, D07 R2WY, Ireland
| | - Vadim Zhernovkov
- Systems Biology Ireland, School of Medicine, University College Dublin, Belfield, Dublin, D04 V1W8, Ireland
| |
Collapse
|
30
|
Huang MY, Xu CC, Chen Q, Zhang YM, Lyu WY, Ye ZH, Li T, Huang MQ, Lu JJ. Ginsenoside Rh2 in combination with IFNγ potentiated the anti-cancer effect by enhancing interferon signaling response in colorectal cancer cells. Acta Pharmacol Sin 2025:10.1038/s41401-025-01557-z. [PMID: 40263567 DOI: 10.1038/s41401-025-01557-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 03/31/2025] [Indexed: 04/24/2025]
Abstract
Interferon gamma (IFNγ) can amplify immune cell-mediated anti-tumor immunity, as well as directly kill cancer cells. Ginsenoside Rh2 (Rh2), a bioactive compound in traditional Chinese medicine, exhibits anti-cancer effects such as inhibiting proliferation and metastasis. Our earlier research found that Rh2 combined with IFNγ enhanced CXCL10 secretion in cancer cells. Here, we explored whether Rh2 and IFNγ exerted more potent anti-cancer activity in vitro and in vivo, along with its mechanisms and clinical value. Our data showed that Rh2 in combination with IFNγ resulted in a remarkably increased cytotoxicity in colorectal cancer cells including HT29, LoVo and T84 cell lines. Consistently, intratumoral injection with Rh2 plus IFNγ further restricted the HT29 tumor growth in vivo, and importantly, it was demonstrated to be safe for mice. Meanwhile, the combo treatment activated the stimulator of interferon genes (STING) pathway in cancer cells, promoting the transcription of downstream type I interferon. RNA sequencing revealed a dramatically transcriptional alteration in cancer cells with combo treatment and indicated that Rh2 further augmented the activation of interferon signaling pathway, compared with the IFNγ alone. Inhibition of janus kinase (JAK) by ruxolitinib could significantly rescue the cell death-triggered by the combo treatment. Then, a gene set named Rh2+IFNγ signature genes (RISG) was defined, which contained top 20 significantly upregulated genes from the combo treatment. Patients who exhibited a favorable response to the immunotherapy had a higher expression of RISG in tumor compared with those who did not respond. And the high expression of RISG was correlated with better clinical outcome in patients with colorectal cancer (CRC) and skin cutaneous melanoma (SKCM). Herein, the combination of Rh2 with IFNγ served as a promising strategy for cancer treatment, and its-derived RISG gene set also exhibited potential value in predicting clinical outcome. Schematic diagram of the anti-cancer effect of Rh2 combined with IFNγ. The schematic diagram illustrated that ginsenoside Rh2 in combination with IFNγ robustly activated the interferon signals in cancer cells, ultimately leading a significant cell death of cancer cells. ISGs, interferon-stimulated genes. Created with BioRender.com.
Collapse
Affiliation(s)
- Mu-Yang Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Chun-Cao Xu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Qian Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Yan-Ming Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Wen-Yu Lyu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Zi-Han Ye
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China
| | - Ting Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao, 999078, China.
| | - Ming-Qing Huang
- College of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, 350000, China.
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, 999078, China.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Macao, 999078, China.
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao, 999078, China.
| |
Collapse
|
31
|
Cuisiniere T, Hajjar R, Oliero M, Calvé A, Fragoso G, Rendos HV, Gerkins C, Taleb N, Gagnon-Konamna M, Dagbert F, Loungnarath R, Sebajang H, Schwenter F, Wassef R, Ratelle R, De Broux É, Richard C, Santos MM. Initial gut microbiota composition is a determining factor in the promotion of colorectal cancer by oral iron supplementation: evidence from a murine model. MICROBIOME 2025; 13:100. [PMID: 40259408 PMCID: PMC12013013 DOI: 10.1186/s40168-025-02101-1] [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: 10/23/2024] [Accepted: 03/26/2025] [Indexed: 04/23/2025]
Abstract
BACKGROUND Colorectal cancer (CRC) development is influenced by both iron and gut microbiota composition. While iron supplementation is routinely used to manage anemia in CRC patients, it may also impact gut microbiota and promote tumorigenesis. In this study, we investigated the impact of initial gut microbiota composition on iron-promoted tumorigenesis. We performed fecal microbiota transplantation (FMT) in ApcMin/+ mice using samples from healthy controls, CRC patients, and mice, followed by exposure to iron sufficient or iron excess diets. RESULTS We found that iron supplementation promoted CRC and resulted in distinct gut microbiota changes in ApcMin/+ mice receiving FMT from CRC patients (FMT-CRC), but not from healthy controls or mice. Oral treatment with identified bacterial strains, namely Faecalibaculum rodentium, Holdemanella biformis, Bifidobacterium pseudolongum, and Alistipes inops, protected FMT-CRC mice against iron-promoted tumorigenesis. CONCLUSIONS Our findings suggest that microbiota-targeted interventions may mitigate tumorigenic effects of iron supplementation in anemic patients with CRC.
Collapse
Affiliation(s)
- Thibault Cuisiniere
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Roy Hajjar
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Department of Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Manon Oliero
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Annie Calvé
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Gabriela Fragoso
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Hervé Vennin Rendos
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Claire Gerkins
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada
- Institut du Cancer de Montréal, Montréal, Québec, Canada
| | - Nassima Taleb
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
| | - Marianne Gagnon-Konamna
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - François Dagbert
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Rasmy Loungnarath
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Herawaty Sebajang
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Frank Schwenter
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Ramses Wassef
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Richard Ratelle
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Éric De Broux
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Carole Richard
- Digestive Surgery Service, Centre Hospitalier de L'Université de Montréal (CHUM), Montréal, Québec, Canada
- Division of General Surgery, Université de Montréal, Montréal, Québec, Canada
| | - Manuela M Santos
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre hospitalier de l', Université de Montréal (CRCHUM), Montréal, Québec, Canada.
- Institut du Cancer de Montréal, Montréal, Québec, Canada.
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
32
|
Zhao CY, Liu F, Dong JM, Du CP, Zhang CL, Wang CY, Zhang XY, Zhou Q, Liu W, Yang AJ, Zhou YN, Dang Y, Shang LN, Wang M, Li M. SDCBP Orchestrated Gastric Cancer Aggression Through Epithelial- Mesenchymal Transition and Macrophages M2 Polarization. Mol Carcinog 2025. [PMID: 40256939 DOI: 10.1002/mc.23923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/13/2025] [Accepted: 03/24/2025] [Indexed: 04/22/2025]
Abstract
Gastric cancer remains a significant global health burden with limited treatment options and high mortality. Syndecan-binding protein (SDCBP), a scaffolding protein involved in tumor differentiation, has attracted attention as a potential therapeutic target in cancers. However, its precise role in gastric cancer progression is not fully understood. In this study, through bioinformatics analysis and gastric cancer samples detection, we discovered that SDCBP was highly expressed in gastric cancer tissues, which was correlated with clinicopathological features such as tumor invasion depth and distant metastasis, and exhibited heterogeneity across histological or molecular subtypes. Elevated SDCBP expression promoted the proliferation, invasion and migration of gastric cancer cells, and modulated epithelial-mesenchymal transition (EMT) via the ERK signaling pathway. Xenograft experiments in mice confirmed that inhibiting SDCBP or ERK signaling could delay cancer progression. We also found that gastric cancer cells with SDCBP knockdown were able to inhibit the M2 polarization of cocultured macrophages, reduce chemotaxis and enhance phagocytosis of macrophages. Therefore, SDCBP plays a crucial role in driving gastric cancer progression. Targeting SDCBP in gastric cancer can partially reverse the malignant phenotype, and SDCBP is expected to be a promising therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Chan-Yuan Zhao
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Feng Liu
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
| | - Jia-Ming Dong
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Cun-Pu Du
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Chen-Li Zhang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
| | - Chen-Yu Wang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
| | - Xiao-Yu Zhang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Quan Zhou
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Wei Liu
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
| | - Ai-Jun Yang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
| | - Yong-Ning Zhou
- Gansu Province Clinical Research Center for Digestive Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yun Dang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Gansu Provincial Maternity and Child-care Hospital/Gansu Provincial Central Hospital, Lanzhou, China
| | - Li-Na Shang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- Medical College of Northwest Minzu University, Lanzhou, China
| | - Min Wang
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
- Experimental Teaching Center of Basic Medicine, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Min Li
- Institute of Pathology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
- The Forensic Identification Unit of Lanzhou University, Lanzhou, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Lanzhou University, Lanzhou, China
| |
Collapse
|
33
|
Paniagua K, Jin YF, Chen Y, Gao SJ, Huang Y, Flores M. Dissection of tumoral niches using spatial transcriptomics and deep learning. iScience 2025; 28:112214. [PMID: 40230519 PMCID: PMC11994907 DOI: 10.1016/j.isci.2025.112214] [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: 07/16/2024] [Revised: 10/05/2024] [Accepted: 03/10/2025] [Indexed: 04/16/2025] Open
Abstract
This study introduces TG-ME, an innovative computational framework that integrates transformer with graph variational autoencoder (GraphVAE) models for dissection of tumoral niches using spatial transcriptomics data and morphological images. TG-ME effectively identifies and characterizes niches in bench datasets and a high resolution NSCLC dataset. The pipeline consists in different stages that include normalization, spatial information integration, morphological feature extraction, gene expression quantification, single cell expression characterization, and tumor niche characterization. For this, TG-ME leverages advanced deep learning techniques that achieve robust clustering and profiling of niches across cancer stages. TG-ME can potentially provide insights into the spatial organization of tumor microenvironments (TME), highlighting specific niche compositions and their molecular changes along cancer progression. TG-ME is a promising tool for guiding personalized treatment strategies by uncovering microenvironmental signatures associated with disease prognosis and therapeutic outcomes.
Collapse
Affiliation(s)
- Karla Paniagua
- Department of Electrical and Computer Engineering, KLESSE School of Engineering and Integrated Design, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Yu-Fang Jin
- Department of Electrical and Computer Engineering, KLESSE School of Engineering and Integrated Design, University of Texas at San Antonio, San Antonio, TX 78249, USA
| | - Yidong Chen
- Greehey Children Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Department of Population Health Science, The University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Shou-Jiang Gao
- Cancer Virology Program, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yufei Huang
- Cancer Virology Program, UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Electrical and Computer Engineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mario Flores
- Department of Electrical and Computer Engineering, KLESSE School of Engineering and Integrated Design, University of Texas at San Antonio, San Antonio, TX 78249, USA
| |
Collapse
|
34
|
Harada S, Sato T, Yoshioka K. Targeted theranostic nanomedicine using targeted CT-imageable particles that release tebentafusp. Jpn J Radiol 2025:10.1007/s11604-025-01782-w. [PMID: 40244495 DOI: 10.1007/s11604-025-01782-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025]
Abstract
PURPOSE A theranostic nanomedicine for CD3+ bispecific antibodies targeting glycoprotein-100 (GP-100) was tested in vivo using two radiation sessions. CT-imageable nanoparticles composed of hyaluronate-alginate and designed to release their contents upon radiation exposure were evaluated in a mouse model of B16-melanoma model in the left hind leg with pulmonary metastases. MATERIALS AND METHODS In session 1, IFN-γ was encapsulated during the Fe polymerization of hyaluronate-alginate nanoparticles. Nine hours after the intravenous injection of 1 × 1010 IFN-γ nanoparticles, enough to observe dose escalation of either 10 or 20 Gy was administered using 140 keV-X-ray to the primary and metastatic tumors. In session 2, tebentafusp was encapsulated using the same method as in session 1. Seventy-two hours after the intravenous injection of 1 × 1010 tebentafusp-loaded nanoparticles, radiation was administered under conditions identical to those in session 1. RESULTS In session 1, IFN-γ-loaded nanoparticles selectively accumulated in the primary tumor and pulmonary metastasis by passing through the coarse endothelium of tumor vasculature, which could be visualized using CT. IFN-γ nanoparticles continuously released IFN-γ, facilitating the formation of the HLA-A*02:01-GP100-complex. In session 2, the tebentafusp-loaded nanoparticles continuously released tebentafusp, leading to the formation of an immunological synapse consisting of HLA-A*02:01-GP100, tebentafusp, and CD3 on T cells. CD3+ T cells release perforin and granzymes, resulting in the cytolysis of the primary tumor and pulmonary metastasis. This effect was synergistic with that of radiation, resulting in Enhancement Factor (EF) more than 1. CONCLUSION Theranostic nanomedicine demonstrated potential as a dual therapeutic and diagnostic strategy for targeting tumors and metastases, with synergistic effects observed when combined with radiation.
Collapse
Affiliation(s)
- Satoshi Harada
- Department of Radiology, School of Medicine, Iwate Medical University, 1-1 1-Chome Idai-Dori, Yahaba, Shiwa, 028-3694, Japan.
| | - Takahiro Sato
- National Institutes for Quantum Science and Technology, Takasaki Ion Accelerators for Advanced Radiation Application, Foundational Quantum Technology Research Directorate, Takasaki Institute for Advanced Quantum Science, 1233 Watanuki, Takasaki, Japan
| | - Kunihiro Yoshioka
- Department of Radiology, School of Medicine, Iwate Medical University, 1-1 1-Chome Idai-Dori, Yahaba, Shiwa, 028-3694, Japan
| |
Collapse
|
35
|
Tsai WT, Cheng CY, Sun HY, Guo BC, Chiang YC, Cheng CF, Pan YH, Wu UI, Wang JT, Sheng WH, Cheng A, Chen YC, Chang SC. Concomitant autoimmunity and late cancers in adult-onset immunodeficiency due to neutralizing anti-IFN-γ autoantibodies. Front Immunol 2025; 16:1526439. [PMID: 40313931 PMCID: PMC12043703 DOI: 10.3389/fimmu.2025.1526439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/24/2025] [Indexed: 05/03/2025] Open
Abstract
Background Opportunistic intramacrophagic infections are well-characterized in adult-onset immunodeficiency associated with neutralizing anti-IFN-γ autoantibodies (nAIGA). Objective Concomitant autoimmune and neoplastic diseases are rarely described. Methods This study included 50 patients diagnosed with adult-onset immunodeficiency due to nAIGA between 2014-2024. Thirty-three were retrospectively included before January 2022, and 17 out of 295 screened patients were enrolled prospectively since January 2022. Ten patients were excluded due to missing records. All patients had regular follow-ups; anti-IFN-γ titers, autoimmune markers and cancer survey were conducted according to the primary physician's evaluation. Results The median age at diagnosis of adult-onset immunodeficiency was 57 years, and 53% were men. Malignancy occurred in 25%; genitourinary cancer predominated (n=4). Most (93%) patients had at least one positive autoimmune marker. Fifty-eight percent of patients were diagnosed with concomitant autoimmune diseases, and women (65%) predominated. Anti-nuclear antibody was positive in 61%, lupus anticoagulant in 50%, whilst autoimmune thyroiditis markers in 43%. Twenty-two percent of patients required long-term immunomodulation including biologic agents such as rituximab and daratumumab. Three patients (8%) died after a median interval of 9.4 years due to sepsis (n=2) and aggressive urothelial cancer (n=1). Most patients had decreasing nAIGA titers over time; two outliers with persistently high neutralizing antibodies developed late-onset malignancies. Conclusion Adult-onset immunodeficiency due to nAIGA is a syndrome associated with concomitant autoimmunity. Chronic infection and autoimmune-mediated inflammation may foster neoplastic changes, but the underlying mechanism is still undetermined. Autoimmune disease and cancer surveillance for patients with nAIGA is advised.
Collapse
Affiliation(s)
- Wan-Ting Tsai
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Internal Medicine, Taipei City Hospital, Zhongxiao Branch, Taipei, Taiwan
| | - Chih-Yun Cheng
- Department of Internal Medicine, National Taiwan University Hospital, Hsinchu Branch, Hsinchu, Taiwan
| | - Hsin-Yun Sun
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Bei-Chia Guo
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ying-Chieh Chiang
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chiao-Feng Cheng
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yi-Hua Pan
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Un-In Wu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jann-Tay Wang
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Wang-Huei Sheng
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Aristine Cheng
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yee-Chun Chen
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Shan-Chwen Chang
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
36
|
Blanco-Domínguez R, Barros L, Carreira M, van der Ploeg M, Condeço C, Marsères G, Ferreira C, Costa C, Ferreira CM, Déchanet-Merville J, de Miranda NFCC, Mensurado S, Silva-Santos B. Dual modulation of cytotoxic and checkpoint receptors tunes the efficacy of adoptive Delta One T cell therapy against colorectal cancer. NATURE CANCER 2025:10.1038/s43018-025-00948-9. [PMID: 40240620 DOI: 10.1038/s43018-025-00948-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 03/13/2025] [Indexed: 04/18/2025]
Abstract
Colorectal cancer (CRC) remains a challenge for current immunotherapies. Vδ1+ γδ T cells offer a promising alternative because of their HLA-I-independent cytotoxicity and natural tissue tropism. We developed Delta One T (DOT) cells, a Vδ1+ γδ T cell-based adoptive cell therapy clinically explored for hematological malignancies but not yet for solid tumors. Here we demonstrate the capacity of DOT cells to target CRC cell lines and patient-derived specimens and organoids in vitro and to control tumor growth in an orthotopic xenograft model of CRC. Notwithstanding, we found tumor-infiltrating DOT cells to exhibit a dysregulated balance of cytotoxic and inhibitory receptors that paralleled that of endogenous Vδ1+ tumor-infiltrating lymphocytes and limited their cytotoxicity. To maximize efficacy, we unveil two strategies, increasing targeting through upregulation of NKG2D ligands upon butyrate administration and blocking the checkpoints TIGIT and PD1, which synergistically unleashed DOT cell cytotoxicity. These findings support DOT cell-based combinatorial approaches for CRC treatment.
Collapse
Affiliation(s)
| | - Leandro Barros
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | | | - Manon van der Ploeg
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Gabriel Marsères
- University of Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, Bordeaux, France
| | - Cristina Ferreira
- Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisbon, Portugal
| | - Carla Costa
- Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisbon, Portugal
| | - Carlos M Ferreira
- Hospital de Santa Maria, Centro Hospitalar Lisboa Norte, Lisbon, Portugal
| | - Julie Déchanet-Merville
- University of Bordeaux, CNRS, ImmunoConcEpT, UMR 5164, Bordeaux, France
- Equipe labelisée LIGUE Contre le Cancer, Bordeaux, France
| | | | - Sofia Mensurado
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
| | - Bruno Silva-Santos
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal.
- Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
37
|
Hu X, Yu F, Peng M, Yang Z, Ouyang Y, Zhang Z, Zhao W, Yi X, Hu H, Huang X, Wang L. Exploring causal relationship between 41 inflammatory cytokines and marginal zone lymphoma: A bidirectional Mendelian randomization study. Open Med (Wars) 2025; 20:20251171. [PMID: 40292253 PMCID: PMC12032980 DOI: 10.1515/med-2025-1171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 04/30/2025] Open
Abstract
Purpose Marginal zone lymphoma (MZL) is a rare subtype of non-Hodgkin lymphoma, and its diagnosis primarily relies on pathological biopsy. The study aims to investigate the causal relationships between 41 inflammatory cytokines and MZL using a two-sample bidirectional Mendelian randomization (MR) approach, providing new insights and methodologies for rapid differential diagnosis and treatment strategies. Methods Causal associations between 41 inflammatory cytokines and MZL were examined using genetic variant data from two large-scale genome-wide association studies. The inverse variance weighting method was employed, and multiple sensitivity analyses, including MR-Egger, weighted median, simple model, and weighted model methods, were conducted to strengthen the robustness of the findings. Results Elevated levels of MIG and IL-10 were associated with an increased risk of MZL (MIG: OR = 1.57, p = 0.035; IL-10: OR = 1.69, p = 0.021), while higher B-NGF levels exhibited a protective effect (OR = 0.46, p = 0.027). Reverse MR analysis revealed a negative correlation between MZL and IFN-γ levels (OR = 0.97, p = 0.015). Conclusions MIG, IL-10, B-NGF, and IFN-γ are potential biomarkers and therapeutic targets for MZL. IFN-γ likely acts as a downstream molecule in MZL pathogenesis, offering novel insights into MZL-related research, clinical diagnosis, and treatment strategies.
Collapse
Affiliation(s)
- Xinhang Hu
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Thoracic Surgery Research Laboratory, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Fenglei Yu
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Thoracic Surgery Research Laboratory, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Muyun Peng
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Thoracic Surgery Research Laboratory, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Zhi Yang
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Thoracic Surgery Research Laboratory, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Yifan Ouyang
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Thoracic Surgery Research Laboratory, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Zhe Zhang
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Thoracic Surgery Research Laboratory, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Wangcheng Zhao
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Thoracic Surgery Research Laboratory, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Xuyang Yi
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Thoracic Surgery Research Laboratory, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Huali Hu
- Department of Thoracic Surgery, Hunan Rehabilitation Hospital, Changsha, 410000, China
| | - Xingchun Huang
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Thoracic Surgery Research Laboratory, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Li Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Hunan Key Laboratory of Early Diagnosis and Precise Treatment of Lung Cancer, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
- Thoracic Surgery Research Laboratory, The Second Xiangya Hospital, Central South University, Changsha, 410000, China
| |
Collapse
|
38
|
Fu L, Li S, Mei J, Li Z, Yang X, Zheng C, Li N, Lin Y, Cao C, Liu L, Huang L, Shen X, Huang Y, Yun J. BIRC2 blockade facilitates immunotherapy of hepatocellular carcinoma. Mol Cancer 2025; 24:113. [PMID: 40223121 PMCID: PMC11995630 DOI: 10.1186/s12943-025-02319-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 04/01/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND The effectiveness of immunotherapy in hepatocellular carcinoma (HCC) is limited, however, the molecular mechanism remains unclear. In this study, we identified baculoviral IAP repeat-containing protein 2 (BIRC2) as a key regulator involved in immune evasion of HCC. METHODS Genome-wide CRISPR/Cas9 screening was conducted to identify tumor-intrinsic genes pivotal for immune escape. In vitro and in vivo models demonstrated the role of BIRC2 in protecting HCC cells from immune killing. Then the function and relevant signaling pathways of BIRC2 were explored. The therapeutic efficacy of BIRC2 inhibitor was examined in different in situ and xenograft HCC models. RESULTS Elevated expression of BIRC2 correlated with adverse prognosis and resistance to immunotherapy in HCC patients. Mechanistically, BIRC2 interacted with and promoted the ubiquitination-dependent degradation of NFκB-inducing kinase (NIK), leading to the inactivation of the non-canonical NFκB signaling pathway. This resulted in the decrease of major histocompatibility complex class I (MHC-I) expression, thereby protecting HCC cells from T cell-mediated cytotoxicity. Silencing BIRC2 using shRNA or inhibiting it with small molecules increased the sensitivity of HCC cells to immune killing. Meanwhile, BIRC2 blockade improved the function of T cells both in vitro and in vivo. Targeting BIRC2 significantly inhibited tumor growth, and enhanced the efficacy of anti-programmed death protein 1 (PD-1) therapy. CONCLUSIONS Our findings suggested that BIRC2 blockade facilitated immunotherapy of HCC by simultaneously sensitizing tumor cells to immune attack and boosting the anti-tumor immune response of T cells.
Collapse
Affiliation(s)
- Lingyi Fu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Shuo Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Jie Mei
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Ziteng Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Xia Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Chengyou Zheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Nai Li
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Yansong Lin
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Chao Cao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Lixuan Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Liyun Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Xiujiao Shen
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Yuhua Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China
| | - Jingping Yun
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China.
- Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, China.
| |
Collapse
|
39
|
Hu S, Wang M. Identification of a deubiquitinating gene-related signature in ovarian cancer using integrated transcriptomic analysis and machine learning framework. Discov Oncol 2025; 16:510. [PMID: 40208475 PMCID: PMC11985714 DOI: 10.1007/s12672-025-02267-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Ovarian carcinoma represents an aggressive malignancy with poor prognosis and limited therapeutic efficacy. While deubiquitinating (DUB) genes are known to regulate crucial cellular processes and cancer progression, their specific roles in ovarian carcinoma remain poorly understood. METHODS We conducted an integrated analysis of single-cell RNA sequencing and bulk transcriptome data from public databases. DUB genes were identified through Genecard database. Using the Seurat package, we performed cell clustering and differential expression analysis. Cell-cell communications were analyzed using CellChat. A DUB-related risk signature (DRS) was developed using machine learning approaches through integration of GEO and TCGA datasets. The prognostic value and immune characteristics of the signature were systematically evaluated. RESULTS Our analysis revealed eight distinct cell subtypes in the tumor microenvironment, including epithelial, fibroblast, myeloid, and Treg cells. DUB-high cells were predominantly found in Treg and myeloid populations, exhibiting elevated expression of tumor-related pathways and enhanced cell-cell communication networks, particularly between fibroblasts and myeloid cells. Conversely, DUB-low cells were enriched in epithelial populations with reduced immune activity. The DRS model demonstrated robust prognostic value across multiple independent cohorts. High-risk patients, as classified by the DRS, showed significantly poorer survival outcomes and distinct immune infiltration patterns compared to low-risk patients. CONCLUSION This study provides comprehensive insights into DUB gene expression patterns across different cell populations in ovarian carcinoma. The established DRS model offers a promising tool for risk stratification and may guide personalized therapeutic strategies. Our findings highlight the potential role of DUB genes in modulating the tumor immune microenvironment and patient outcomes in ovarian carcinoma.
Collapse
Affiliation(s)
- Suwan Hu
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Mengting Wang
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
40
|
Xu Y, Yang S, Rao Q, Gao Y, Zhou G, Zhao D, Shi X, Chai Y, Zhao C. A mechanistic quantitative systems pharmacology model platform for translational efficacy evaluation and checkpoint combination design of bispecific immuno-modulatory antibodies. Front Pharmacol 2025; 16:1571844. [PMID: 40276607 PMCID: PMC12018249 DOI: 10.3389/fphar.2025.1571844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/31/2025] [Indexed: 04/26/2025] Open
Abstract
Over the past 2 decades, tumor immunotherapies have witnessed remarkable advancements, especially with the emergence of immune checkpoint-targeting bispecific antibodies. However, a quantitative understanding of the dynamic cross-talking mechanisms underlying different immune checkpoints as well as the optimal dosing and target design of checkpoint-targeting bispecific antibodies still remain challenging to researchers. To address this challenge, we have here developed a multi-scale quantitative systems pharmacology (QSP) model platform that integrates a diverse array of immune checkpoints and their interactive functions. The model has been calibrated and validated against an extensive collection of multiscale experimental datasets covering 20+ different monoclonal and bispecific antibody treatments at over 60 administered dose levels. Based on high-throughput simulations, the QSP model platform comprehensively screened and characterized the potential efficacy of different bispecific antibody target combination designs, and model-based preclinical population-level simulations revealed target-specific dose-response relationships as well as alternative dosing strategies that can maintain anti-tumor treatment efficacy while reducing dosing frequencies. Model simulations also pointed out that combining checkpoint-targeting bispecific antibodies with monoclonal antibodies can lead to significantly enhanced anti-tumor efficacy. Our mechanistic QSP model can serve as an integrated precision medicine simulation platform to guide the translational research and clinical development of checkpoint-targeting immuno-modulatory bispecific antibodies.
Collapse
Affiliation(s)
- Yiyang Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Siyuan Yang
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Qi Rao
- School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yuan Gao
- QSPMed Technologies, Nanjing, China
| | - Guanyue Zhou
- Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing, China
| | - Dongmei Zhao
- Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing, China
| | - Xinsheng Shi
- Nanjing Sanhome Pharmaceutical Co., Ltd., Nanjing, China
| | - Yi Chai
- Phase I Clinical Trial Unit, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chen Zhao
- School of Pharmacy, Nanjing Medical University, Nanjing, China
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
41
|
Wang D, Huang W, Li G. miR-145-5p regulates hepatocellular carcinoma malignant advancement and immune escape via down-regulation of AcylCoA synthase ACSL4. BIOMOLECULES & BIOMEDICINE 2025; 25:1184-1196. [PMID: 39652084 PMCID: PMC11984366 DOI: 10.17305/bb.2024.11209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/02/2024] [Accepted: 12/02/2024] [Indexed: 04/04/2025]
Abstract
Hepatocellular carcinoma (HCC) exhibits a subtle onset, high incidence rates, and low survival rates, becoming a substantial threat to human health. Hence, it is crucial to discover fresh biomarkers and treatment targets for the early detection and management of HCC. CCK-8, scratch-wound, and transwell assays were used to evaluate the biological properties of HCC cell lines (Huh-7 and Hep3B). Bioinformatics analysis identified the downstream target mRNA of miR-145-5p as acyl-CoA synthetase long-chain family member 4 (ACSL4). RT-qPCR was used to test miR-145-5p and ACSL4 levels. Transwell chambers were used to co-incubate purified CD8+ T cells and HCC cells for 48 h, and the effect of CD8+ T cells on apoptosis in HCC cells was detected by flow cytometry. A subcutaneous graft tumor model was constructed, and ELISA kits were used to assess cytokine levels and CD8+ T cell activation markers. HCC cells showed a decline in miR-145-5p levels and a rise in ACSL4 levels. Overexpression of miR-145-5p hindered HCC cell proliferation, migration, and invasion, while stimulating CD8+ T cell activation. Conversely, overexpression of ACSL4 enhanced the malignant biological properties of HCC cells and reduced the effect of CD8+ T cells, while silencing ACSL4 had the opposite effect. miR-145-5p targeted and downregulated ACSL4, while overexpression of miR-145-5p weakened the promotion of HCC malignant progression caused by ACSL4 overexpression. Additionally, overexpression of miR-145-5p and silencing ACSL4 were effective in inhibiting tumor growth in vivo. In conclusion, miR-145-5p targets and downregulates ACSL4, leading to the inhibition of HCC malignant progression and preventing immune escape in HCC cells.
Collapse
Affiliation(s)
- Dingxue Wang
- Oncology Department, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Wenqi Huang
- Oncology Department, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Gao Li
- Oncology Department, The First Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
42
|
Xia P, Qu C, Xu X, Tian M, Li Z, Ma J, Hou R, Li H, Rückert F, Zhong T, Zhao L, Yuan Y, Wang J, Li Z. Nanobody Engineered and Photosensitiser Loaded Bacterial Outer Membrane Vesicles Potentiate Antitumour Immunity and Immunotherapy. J Extracell Vesicles 2025; 14:e70069. [PMID: 40240911 PMCID: PMC12003094 DOI: 10.1002/jev2.70069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
Bacterial outer membrane vesicles (OMVs) are promising as antitumour agents, but their clinical application is limited by toxicity concerns and unclear mechanisms. We engineered OMVs with cadherin 17 (CDH17) tumour-targeting nanobodies, enhancing tumour selectivity and efficacy while reducing adverse effects. These engineered OMVs function as natural stimulator of interferon genes (STING) agonists, activating the cyclic GMP-AMP synthase (cGAS)-STING pathway in cancer cells and tumour-associated macrophages (TAMs). Loading engineered OMVs with photoimmunotherapy photosensitisers further enhanced tumour inhibition and STING activation in TAMs. Combining nanobody-engineered OMV-mediated photoimmunotherapy with CD47 blockade effectively suppressed primary and metastatic tumours, establishing sustained antitumour immune memory. This study demonstrates the potential of nanobody-engineered OMVs as STING agonists and provides insights into novel OMV-based immunotherapeutic strategies harnessing the innate immune system against cancer. Our findings open new avenues for OMV applications in tumour immunotherapy, offering a promising approach to overcome current limitations in cancer treatment.
Collapse
Affiliation(s)
- Peng Xia
- Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei ProvinceWuhan UniversityWuhanHubeiP. R. China
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Department of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University)ShenzhenGuangdongP. R. China
- Department of ChemistryThe University of ChicagoChicagoIllinoisUSA
| | - Chengming Qu
- Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei ProvinceWuhan UniversityWuhanHubeiP. R. China
| | - Xiaolong Xu
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Department of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University)ShenzhenGuangdongP. R. China
| | - Ming Tian
- Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei ProvinceWuhan UniversityWuhanHubeiP. R. China
| | - Zhifen Li
- School of Chemistry and Chemical EngineeringShanxi Datong UniversityDatongShanxi ProvinceP. R. China
| | - Jingbo Ma
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Department of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University)ShenzhenGuangdongP. R. China
| | - Rui Hou
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical ResearchThe University of Western AustraliaNedlandsWAAustralia
| | - Han Li
- Department of ChemistryThe University of ChicagoChicagoIllinoisUSA
| | - Felix Rückert
- Department of Visceral SurgeryDiakonissen HospitalSpeyerGermany
| | - Tianyu Zhong
- Department of Laboratory MedicineHuadong Hospital, Fudan UniversityShanghaiP. R. China
| | - Liang Zhao
- Department of PathologyNanfang Hospital, Southern Medical UniversityGuangzhouP. R. China
- Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouP. R. China
| | - Yufeng Yuan
- Zhongnan Hospital of Wuhan University, TaiKang Center for Life and Medical Sciences, Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei ProvinceWuhan UniversityWuhanHubeiP. R. China
| | - Jigang Wang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Department of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University)ShenzhenGuangdongP. R. China
- School of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical SciencesSouthern Medical UniversityGuangzhouGuangdongP. R. China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di Herbs, Artemisinin Research Center, Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijingP. R. China
- State Key Laboratory of Antiviral DrugsSchool of PharmacyHenan UniversityKaifengP. R. China
| | - Zhijie Li
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Centre for Geriatrics, Department of Nuclear MedicineShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University)ShenzhenGuangdongP. R. China
| |
Collapse
|
43
|
Chen W, Yang L, Lee VHF, Xu L, Ma L, Ye Z, Xu W, Zhao C, Zheng D, Kiang KMY, Sun S, Qu Y, Zha J, Pang D, Zhang Y, Liang Z, Lin W, Zhang J, Zhang J, Luo M, Xu Z, Li D, Liang X, Leung GKK, Helali AE, Che C, Feng-Ming (Spring) Kong. Indoleamine 2,3-dioxygenase 1-mediated immune suppressive status is positively associated with brain metastasis in patients with non-small cell lung cancer. JOURNAL OF THE NATIONAL CANCER CENTER 2025; 5:179-192. [PMID: 40265090 PMCID: PMC12010389 DOI: 10.1016/j.jncc.2024.12.004] [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: 04/03/2024] [Revised: 12/10/2024] [Accepted: 12/11/2024] [Indexed: 04/24/2025] Open
Abstract
Background Indoleamine 2,3-dioxygenase (IDO1) activity, measured by kynurenine/tryptophan (K:T) ratio, is known for its association with distant metastasis and overall survival (OS) in patients with non-small cell lung cancer (NSCLC). Here, we aimed to examine whether IDO1 activity is correlated with OS in NSCLC patients with brain metastasis (Bramet) and has negative effect on modulating the anti-tumor functions of immune cells. Methods This study was a part of a prospective clinical trial in circulating biomarkers. Blood or tissues from eligible participants were collected for measurement of kynurenine, tryptophan, immune cell subtype, scRNA-seq analysis, and untargeted metabolomics analysis. Results A total of 195 patients were enrolled. The median kynurenine to tryptophan (K:T) ratio was 0.18, with consistent values observed among patients with NSCLC Bramet and those without (0.18 and 0.11, respectively). Notably, student's t-test analysis revealed significantly higher kynurenine concentrations in stage IV patients compared to those in stage I (2.3 vs 1.7 µM, P < 0.001). In patients with Bramet, both kynurenine concentrations and K:T ratios were significantly elevated in comparison with those of extra-cerebral metastasis (2.7 vs 1.9 µM, P < 0.001; 0.12 vs 0.095, P = 0.028; respectively). Single-cell analysis further validated a high level of IDO1 expression in stage IV tumors or Bramet lesions, particularly in macrophages, regulated by chemokines such as CXCL11. Additionally, K:T ratios exhibited significant associations with Treg cell percentages and OS in patients with Bramet (P = 0.039). Treatment with kynurenine led to the upregulation of immune-suppressive molecules, including PD-1, in T cells. Finally, untargeted metabolomics analysis further identified that, apart from the IDO1 metabolic pathway, other metabolites, such as those involved in phospholipid pathways, were also implicated in Bramet. Conclusion IDO1 metabolites may play immune-suppressive roles in NSCLC patients with Bramet.
Collapse
Affiliation(s)
- Weiwei Chen
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Li Yang
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Victor Ho-fun Lee
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Liangliang Xu
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Lingyu Ma
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zhenghao Ye
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Wanli Xu
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Caining Zhao
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Danyang Zheng
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Karrie Mei-Yee Kiang
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Stella Sun
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Yuan Qu
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jiandong Zha
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Dazhi Pang
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Yan Zhang
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zhibing Liang
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Wenchu Lin
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, China
| | - Jinliang Zhang
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Jitian Zhang
- Department of Surgery, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Min Luo
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Zhiyuan Xu
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Ding Li
- Department of Pathology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Xiaoling Liang
- Department of Pathology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Gilberto Ka-Kit Leung
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Aya El Helali
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
| | - Chiming Che
- Department of Chemistry, State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong, China
| | - Feng-Ming (Spring) Kong
- Department of Clinical Oncology, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong, China
- Department of Clinical Oncology, University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| |
Collapse
|
44
|
Mohammadi Barzelighi H, Bakhshi B, Daraei B, Mirzaei A. Investigating the effect of rAzurin loaded mesoporous silica nanoparticles enwrapped with chitosan-folic acid on breast tumor regression in BALB/ C mice. Int J Biol Macromol 2025; 300:139245. [PMID: 39732269 DOI: 10.1016/j.ijbiomac.2024.139245] [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: 06/12/2024] [Revised: 12/18/2024] [Accepted: 12/25/2024] [Indexed: 12/30/2024]
Abstract
This study aimed to examine how mesoporous silica nanoparticles-chitosan-folic acid impacted the release of recombinant Azurin within the tumor environment. The goal was to trigger apoptosis and stimulate immune responses against both transformed and normal cells in BALB/c mice. The study found that the use of rAzu-MSNs-CS-FA, a specific formulation containing mesoporous silica nanoparticles-chitosan-folic acid, resulted in pH-responsive behavior and slower release of rAzurin compared to other groups. This formulation inhibited MCF7 cells at higher concentrations, induced apoptosis in cells, and caused DNA degradation. It also increased the uptake efficiency of rAzurin and stimulated the secretion of TNF-α, INF-γ, and IL-4 while inhibiting the secretion of IL-6. Furthermore, it regulated the expression of specific genes (upregulating tlr3 and downregulating tlr2, 4, and 9). In animal studies with BALB/c mice, the rAzu-MSNs-CS-FA formulation led to tumor regression and decreased tumor volume over 21 days. Overall, this formulation showed promising results in inducing cytotoxic effects against cancer cells, promoting apoptosis, and eliciting appropriate immune responses, suggesting its potential as a valuable therapy for breast cancer.
Collapse
Affiliation(s)
| | - Bita Bakhshi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Bahram Daraei
- Department of Toxicology and Pharmacology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Arezoo Mirzaei
- Department of Bacteriology and Virology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
45
|
Lv Z, Wu J. Research Hotspots of Interferon Gamma in the Treatment of Lung Cancer: A Bibliometric Analysis Based on CiteSpace. J Interferon Cytokine Res 2025; 45:109-118. [PMID: 39874560 DOI: 10.1089/jir.2024.0242] [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] [Indexed: 01/30/2025] Open
Abstract
Interferon-gamma (IFN-γ) is an important cytokine associated with antitumor immunity and has been implicated in the pathogenesis and progression of lung cancer. Nevertheless, no bibliometric analyses have been published in this field to date, and thus we aim to address this gap in knowledge. A search of the Web of Science (WOS) for literature related to the treatment of lung cancer with IFN-γ was conducted from 2002 to 2024. The extracted information from the included articles was subjected to visual analysis, and network diagrams were generated using software such as CiteSpace and VOSviewer. In total, 589 articles related to the treatment of lung cancer with IFN-γ were included in WOS between 2002 and 2024. The number of articles and citation frequency generally showed an increasing trend year by year. The United States and the University of California are the countries and institutions with the largest number of articles. The researcher who made the largest contribution to this field was Xin Cai from China (6). The Journal for ImmunoTherapy of Cancer published the largest number of relevant papers in the field (16 papers, IF = 12.469). The research hotspots in the field of immune escape in recent years have been IFN-γ, mechanism, immune checkpoints, and microtumor inhibitors. The field of IFN-γ treatment of lung cancer is evolving at a rapid pace. The current research focus within this field is on elucidating the mechanism of IFN-γ treatment of lung cancer, investigating the role of immune checkpoint inhibitors, and examining the tumor microenvironment and other pertinent topics.
Collapse
Affiliation(s)
- Zhen Lv
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Jianjun Wu
- Gansu University of Traditional Chinese Medicine, Lanzhou, China
| |
Collapse
|
46
|
Ryba-Stanisławowska M. Unraveling Th subsets: insights into their role in immune checkpoint inhibitor therapy. Cell Oncol (Dordr) 2025; 48:295-312. [PMID: 39325360 PMCID: PMC11996958 DOI: 10.1007/s13402-024-00992-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2024] [Indexed: 09/27/2024] Open
Abstract
T helper (Th) cell subsets play pivotal roles in regulating immune responses within the tumor microenvironment, influencing both tumor progression and anti-tumor immunity. Among these subsets, Th1 cells promote cytotoxic responses through the production of IFN-γ, while Th2 cells and regulatory T cells (Tregs) exert immunosuppressive effects that support tumor growth. Th9 and Th17 cells have context-dependent roles, contributing to both pro-inflammatory and regulatory processes in tumor immunity. Tumor antigen-specific T cells within the tumor microenvironment often exhibit a dysfunctional phenotype due to increased expression of inhibitory receptors such as CTLA-4 and PD-1, leading to reduced antitumor activity. Monoclonal antibodies that block these inhibitory signals-collectively known as immune checkpoint inhibitors (ICIs)-can reactivate these T cells, enhancing their ability to target and destroy cancer cells. Recent advancements have highlighted the critical role of T helper subsets in modulating responses to ICIs, with their interactions remaining a focus of ongoing research. Both positive and negative effects of ICIs have been reported in relation to Th cell subsets, with some effects depending on the type of tumor microenvironment. This review summarizes the crucial roles of different T helper cell subsets in tumor immunity and their complex relationship with immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Monika Ryba-Stanisławowska
- Department of Medical Immunology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 1, Gdańsk, 80-211, Poland.
| |
Collapse
|
47
|
Chen L, Cai B, Ni X, Lin Q, Ke R, Wan X, Huang T, Shan X, Wang B. Temozolomide monotherapy versus combination therapies in melanoma: a meta-analysis of efficacy and safety. Melanoma Res 2025; 35:87-101. [PMID: 39874124 DOI: 10.1097/cmr.0000000000001021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Temozolomide is used in melanoma therapy, but the comparative efficacy and safety of monotherapy vs combination therapies are unclear. This meta-analysis evaluates temozolomide monotherapy vs combination therapies in melanoma patients. PubMed, Embase, and Cochrane Library were searched up to August 2024 for studies comparing temozolomide monotherapy with combination therapies in melanoma. Primary outcomes were 1-year survival and objective response rates (RR); secondary outcomes included hematologic and non-hematologic toxicities. Data were pooled using risk ratios with 95% confidence intervals (CIs). Seven studies were included. Combination therapies improved objective RR over temozolomide monotherapy (risk ratio 0.68, 95% CI: 0.53-0.88). One-year survival did not differ significantly between groups (risk ratio 0.81, 95% CI: 0.59-1.12). Temozolomide monotherapy was associated with reduced incidence of leukopenia (risk ratio 0.54, 95% CI: 0.30-0.95). Adding interferon-alpha (IFN-α) to temozolomide significantly improved 1-year survival (risk ratio 0.54, 95% CI: 0.35-0.84) and objective RR (risk ratio 0.57, 95% CI: 0.42-0.78) compared to temozolomide alone, without significantly increasing toxicity. Combination therapies enhance objective RR over temozolomide monotherapy, with similar 1-year survival. Temozolomide monotherapy offers a better hematologic safety profile. Combining temozolomide with IFN-α significantly improves survival and RR without increasing toxicity. Clinicians should balance efficacy and safety when choosing melanoma treatments.
Collapse
Affiliation(s)
- Lu Chen
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Beichen Cai
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xuejun Ni
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Qian Lin
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ruonan Ke
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaofen Wan
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Tao Huang
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiuying Shan
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Biao Wang
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
48
|
Ling X, Dong Z, He J, Chen D, He D, Guo R, He Q, Li M. Advances in Polymer-Based Self-Adjuvanted Nanovaccines. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409021. [PMID: 40079071 DOI: 10.1002/smll.202409021] [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: 10/02/2024] [Revised: 02/22/2025] [Indexed: 03/14/2025]
Abstract
Nanovaccines, as a new generation of vaccines, have garnered significant interest due to their exceptional potential in enhancing disease prevention and treatment. Their unique features, such as high stability, antigens protection, prolonged retention, and targeted delivery to lymph nodes, immune cells, and tumors, set them apart as promising candidates in the field of immunotherapy. Polymers, with their superior degradability, capacity to mimic pathogen characteristics, and surface functionality that facilitates modifications, serve as ideal carriers for vaccine components. Polymer-based self-adjuvanted nanovaccines have the remarkable ability to augment immune responses. The inherent adjuvant-like properties of polymers themselves offer a pathway toward more efficient exploitation of nanomaterials and the optimization of nanovaccines. This review article aims to summarize the categorization of polymers and elucidate their mechanisms of action as adjuvants. Additionally, it delves into the advantages and limitations of polymer-based self-adjuvanted nanovaccines in disease management and prevention, providing valuable insights for their design and application. This comprehensive analysis could contribute to the development of more effective and tailored nanovaccines for a wide range of diseases.
Collapse
Affiliation(s)
- Xiaoli Ling
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Ziyan Dong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Jiao He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Dong Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Dan He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Rong Guo
- West China College of Basic Medical Sciences and Forensic Science, Sichuan University, Chengdu, 610041, P. R. China
| | - Qin He
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Man Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Centre for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
49
|
Shi Y, Fan G, Yang E, Zhang Y, Ding H, Tian J, Cheng L, Wang H, Hao T, Wang B, Wang Z. Enhanced efficacy of immune checkpoint inhibitors by folate-targeted multifunctional drug through synergistic therapy inducing ferroptosis and immunogenic cell death in bladder cancer. Mater Today Bio 2025; 31:101584. [PMID: 40104638 PMCID: PMC11919378 DOI: 10.1016/j.mtbio.2025.101584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/26/2025] [Accepted: 02/15/2025] [Indexed: 03/20/2025] Open
Abstract
Purpose The research aims to elucidate the anti-tumor mechanism of the composite multifunctional folate-targeted drug DIFP-FA through sonodynamic therapy (SDT), chemodynamic therapy (CDT), and chemotherapy, as well as its potential to augment immune checkpoint blockade (ICB) therapy in bladder cancer (BC). Methods DIFP-FA was synthesized via the W/O/W method. Its targeting efficacy was assessed using immunofluorescence and small animal imaging. Specific mechanisms were investigated through transcriptome sequencing and validation at both cellular and animal levels was conducted. BC patient-derived organoids (PDOs) and patient-derived tumor xenograft (PDX) models, derived from BC tissues resistant to cisplatin-gemcitabine and tislelizumab, were utilized to evaluate the efficacy of DIFP-FA in combination with SDT/CDT and chemotherapy. A humanized BC-PDX model was constructed to explore the synergistic effect of DIFP-FA with ICB therapy. Results DIFP-FA, by incorporating doxorubicin and indocyanine green, leverages specific binding to folate receptors for precise targeting and efficient internalization into BC cells. DIFP-FA exhibits pH and ultrasound (US)-responsive cargo release properties, ensuring spatiotemporally controlled release. DIFP-FA induces reduced GPX4 and SLC7A11 expression and ferroptosis through the combination of SDT/CDT and chemotherapy. It also facilitates the transport and release of DAMPs, leading to immunogenic cell death (ICD). PDOs and PDX experiments demonstrated that DIFP-FA + US enhanced T lymphocyte infiltration in tumor tissues. Moreover, its combination with anti-PD-1 therapy effectively cleared immune-tolerant BC. Conclusions DIFP-FA integrates SDT/CDT with chemotherapy to induce ferroptosis and ICD, efficiently eradicating tumors and activating the immune response, thereby enhancing the efficacy of ICB therapy.
Collapse
Affiliation(s)
- Yibo Shi
- Institute of Urology, The Second Hospital of Lanzhou University, Key Laboratory of Gansu Province for Urological Diseases, Gansu Province Clinical Research Center for Urinary System Disease, 730030, Lanzhou, Gansu, China
| | - Guangrui Fan
- Institute of Urology, The Second Hospital of Lanzhou University, Key Laboratory of Gansu Province for Urological Diseases, Gansu Province Clinical Research Center for Urinary System Disease, 730030, Lanzhou, Gansu, China
| | - Enguang Yang
- Institute of Urology, The Second Hospital of Lanzhou University, Key Laboratory of Gansu Province for Urological Diseases, Gansu Province Clinical Research Center for Urinary System Disease, 730030, Lanzhou, Gansu, China
| | - Yuanfeng Zhang
- Institute of Urology, The Second Hospital of Lanzhou University, Key Laboratory of Gansu Province for Urological Diseases, Gansu Province Clinical Research Center for Urinary System Disease, 730030, Lanzhou, Gansu, China
| | - Hui Ding
- Institute of Urology, The Second Hospital of Lanzhou University, Key Laboratory of Gansu Province for Urological Diseases, Gansu Province Clinical Research Center for Urinary System Disease, 730030, Lanzhou, Gansu, China
| | - Junqiang Tian
- Institute of Urology, The Second Hospital of Lanzhou University, Key Laboratory of Gansu Province for Urological Diseases, Gansu Province Clinical Research Center for Urinary System Disease, 730030, Lanzhou, Gansu, China
| | - Liang Cheng
- The Legorreta Cancer Center at Brown University, Department of Pathology and Laboratory Medicine, The Warren Albert Medical School of Brown University, Brown University Health, Providence, RI, USA
| | - Hanzhang Wang
- The Legorreta Cancer Center at Brown University, Department of Pathology and Laboratory Medicine, The Warren Albert Medical School of Brown University, Brown University Health, Providence, RI, USA
| | - Tianzhi Hao
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Lanzhou University, 730000, Lanzhou, Gansu, China
| | - Baodui Wang
- State Key Laboratory of Applied Organic Chemistry, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Lanzhou University, 730000, Lanzhou, Gansu, China
| | - Zhiping Wang
- Institute of Urology, The Second Hospital of Lanzhou University, Key Laboratory of Gansu Province for Urological Diseases, Gansu Province Clinical Research Center for Urinary System Disease, 730030, Lanzhou, Gansu, China
| |
Collapse
|
50
|
Bonacci RE, McGill M, Le NTA, Barkarar M, Finnegan C, Wilson M, Ajagbe O, Udekwu CC, Gorski K, Manohar J, Sboner A, Ogunwobi OO. Upregulation of the interferon-inducible antiviral gene RSAD2 in neuroendocrine prostate cancer via PVT1 exon 9 dependent and independent pathways. J Biol Chem 2025; 301:108370. [PMID: 40024473 PMCID: PMC11994405 DOI: 10.1016/j.jbc.2025.108370] [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: 07/30/2024] [Revised: 02/19/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025] Open
Abstract
PVT1 exon 9 overexpression is a newly uncovered aberration in prostate cancer (PCa). We have previously demonstrated the exon 9 region of PVT1 is overexpressed in some patient PCa tissues and caused development of neuroendocrine prostate cancer (NEPC) in vitro and in vivo. In this study, we focused on elucidating downstream mechanisms induced by PVT1 exon 9 overexpression with the goal of further understanding its role in NEPC development. RNA-seq analysis of a PVT1 exon 9 overexpressing PCa model revealed significant enrichment of genes responsible for inducing inflammatory processes including RSAD2. We observed RSAD2 overexpression in all NEPC models examined whereas PVT1 exon 9 was overexpressed only in a subset of the NEPC models. We identified two distinct pathways in which RSAD2 is overexpressed: one dependent and one independent on PVT1 exon 9 overexpression. Knockdown of RSAD2 suppressed cell proliferation and migration suggestive of its role as a therapeutic target in NEPC. We identified RSAD2 induces increased cell proliferation, colony formation, and may be involved in the transition between CRPC and NEPC. Distinct differences between PVT1 exon 9-dependent and PVT1 exon 9-independent NEPC models include differences in type II interferon signaling and AR modulation. PVT1 exon 9 binds to RSAD2 protein and disruption of binding significantly impedes downstream interferon gamma secretion by PVT1 exon 9-dependent NEPC cells. These novel findings indicate the importance of these two independent pathways in NEPC, the need to identify relevant NEPC patient populations and study strategies for targeting PVT1 exon 9 and/or RSAD2.
Collapse
Affiliation(s)
- Rachel E Bonacci
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Meghan McGill
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Nu Thuy Anh Le
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Murtaza Barkarar
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Colin Finnegan
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Maya Wilson
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Oluwabusola Ajagbe
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Chinedum C Udekwu
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Kathryn Gorski
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Jyothi Manohar
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Andrea Sboner
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York, USA
| | - Olorunseun O Ogunwobi
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA.
| |
Collapse
|