1
|
Barış Moğul C, Duran MB, Caner V, Türk NŞ, Tuncay ÖL. The PD-L1 Promoter Methylation Predicts Gene And Protein Expression Levels in Urothelial Carcinoma. Gene 2025; 959:149503. [PMID: 40228759 DOI: 10.1016/j.gene.2025.149503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 04/07/2025] [Accepted: 04/11/2025] [Indexed: 04/16/2025]
Abstract
We aimed to clarify the role of PD-L1 promoter methylation in bladder cancer by analyzing PD-L1 methylation and mRNA expression in FFPE samples, along with PD-L1 mRNA and protein levels in urine samples from bladder urothelial carcinoma patients. We analyzed PD-L1 promoter methylation in 43 bladder urothelial carcinoma tissue samples and 41 non-malignant bladder tissues using methylation-sensitive high-resolution melting analysis to assess two CpG islands (cg15837913, cg19724470). PD-L1 mRNA expression in tissues and urine samples, along with PD-L1 protein levels in urine, were evaluated. The bladder urothelial carcinoma group showed significantly higher methylation rates for cg19724470 and cg15837913 compared to controls (P = 0.016, P = 0.049 respectively). In the patient group, tissue PD-L1 mRNA expression was 15.22 times higher and urinary PD-L1 mRNA expression 6.56 times higher in the cg19724470 unmethylated group compared to the methylated group. Urinary PD-L1 protein concentration was twice as high in the cg19724470 unmethylated group compared to the methylated group. In the patients, tissue PD-L1 mRNA expression was 4.58 times higher and urinary PD-L1 mRNA expression 2.58 times higher in the cg15837913 unmethylated group compared to the methylated group. Moreover, the urinary PD-L1 protein concentration was 1.7 times higher in the cg15837913 unmethylated group than in the methylated group (P = 0.036). A positive correlation was observed between tissue PD-L1 mRNA and both urine PD-L1 mRNA and protein levels and between urine PD-L1 mRNA and protein levels. This study suggests that PD-L1 methylation may be a key epigenetic regulator influencing PD-L1 expression and disease pathogenesis in bladder urothelial carcinoma.
Collapse
Affiliation(s)
- Cansu Barış Moğul
- Department of Medical Biology, School of Medicine, Pamukkale University, Denizli, Turkey.
| | - Mesut Berkan Duran
- Department of Urology, School of Medicine, Pamukkale University, Denizli, Turkey.
| | - Vildan Caner
- Department of Medical Genetics, School of Medicine, Pamukkale University, Denizli, Turkey; Sapiens Genetics Diagnostic Center, İstanbul, Turkey.
| | - Nilay Şen Türk
- Department of Medical Pathology, School of Medicine, Pamukkale University, Denizli, Turkey.
| | - Ömer Levent Tuncay
- Department of Urology, School of Medicine, Pamukkale University, Denizli, Turkey
| |
Collapse
|
2
|
Han HL, Su JY, Zhao XH, Hou DD, Li YM. Peptide-Based Strategies in PLGA-Enhanced Tumor Therapy. J Pept Sci 2025; 31:e70020. [PMID: 40269479 DOI: 10.1002/psc.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/22/2025] [Accepted: 04/04/2025] [Indexed: 04/25/2025]
Abstract
Peptide-based therapeutics have gained attention in cancer treatment because of their good specificity, low toxicity, and ability to modulate immune responses. However, challenges such as enzymatic degradation and poor bioavailability limit their clinical application. Peptide-functionalized poly(lactic-co-glycolic acid) (PLGA) systems have emerged as a transformative platform in cancer therapy that offers unique advantages, including enhanced stability, sustained release, and precise delivery of therapeutic agents. This review highlights the synergistic integration of peptides with PLGA and addresses key challenges of peptide-based therapeutics. The application of peptide-functionalized PLGA systems encompasses a diverse range of strategies for cancer therapy. In chemotherapy, peptides disrupt critical tumor pathways, induce apoptosis, and inhibit angiogenesis, demonstrating their versatility in targeting various aspects of tumor progression. In immunotherapy, peptides act as antigens to stimulate robust immune responses or as immune checkpoint inhibitors to restore T cell activity, overcoming tumor immune evasion. These systems also harness the enhanced permeability and retention effect, facilitating preferential accumulation in tumor tissues while leveraging tumor microenvironment (TME)-responsive mechanisms, such as pH-sensitive or enzyme-triggered drug release, to achieve controlled, localized delivery. Collectively, peptide-functionalized PLGA systems represent a promising, versatile approach for precise cancer therapy that integrates innovative delivery strategies with highly specific, potent therapeutic agents.
Collapse
Affiliation(s)
- Hong-Lin Han
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, China
| | - Jing-Yun Su
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, China
| | - Xiao-Huan Zhao
- SINOPEC key Laboratory of Research and Application of Medical and Hygienic Materials, SINOPEC (Beijing) Research Institute of Chemical Industry co., ltd, Beijing, China
| | - Dan-Dan Hou
- SINOPEC key Laboratory of Research and Application of Medical and Hygienic Materials, SINOPEC (Beijing) Research Institute of Chemical Industry co., ltd, Beijing, China
| | - Yan-Mei Li
- Key Laboratory of Bioorganic Phosphorus Chemistry and Chemical Biology (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, China
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
- Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
3
|
Zhang J, Huang Q, Yang H, Shi X, Su Y, Xia J, Li Y, Liu X. Lipid Coating of Mesoporous Silica Nanoparticles Leads to Efficient Antigen Delivery to Lymph Nodes for Cancer Vaccination. ACS APPLIED BIO MATERIALS 2025; 8:4294-4302. [PMID: 40310259 DOI: 10.1021/acsabm.5c00403] [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: 05/02/2025]
Abstract
The enrichment of antigens in lymph nodes and the ensuing antigen presentation constitute crucial steps in determining the efficacy of tumor vaccines. However, antigen delivery is restricted by enzyme degradation, immune system clearance, and the difficulty of crossing biological barriers. In this study, mesoporous silica nanoparticles (MSNPs) were prepared for antigen loading and were further coated with a phospholipid bilayer membrane (named silicasomes) to improve the delivery efficiency to lymph nodes. Our results showed that silicasomes exhibited superior lymph node enrichment compared to the bare MSNPs following subcutaneous injection. Moreover, their efficacy as a tumor vaccine was validated in the B16-OVA tumor model by loading the ovalbumin antigens (OVA257-264). Besides, the toll-like receptor 4 (TLR4) agonist monophosphoryl lipid A (MPLA), a component of bacterial lipopolysaccharides, was incorporated into the phospholipid membrane on the surface of silicasomes as an adjuvant. The silicasome-OVA257-264 with the addition of MPLA exhibited a more potent antitumor effect, triggering the infiltration of specific T cells into the tumor. These results demonstrated that lipid coating on MSNPs significantly enhanced their delivery efficiency to lymph nodes and enabled synergistic immune activation of tumor antigens and adjuvants, highlighting their potential as effective vehicles for cancer immunotherapy.
Collapse
Affiliation(s)
- Jia Zhang
- School of Life Sciences, Faculty of Medicine, Tianjin University, Tianjin 300072, China
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Basic and Clinical Application of Functional Nucleic Acids, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
| | - Qiang Huang
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Basic and Clinical Application of Functional Nucleic Acids, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
- Department of Urology, The First Hospital of China Medical University, Institute of Urology, China Medical University, Shenyang 110001, China
| | - Honghong Yang
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Basic and Clinical Application of Functional Nucleic Acids, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
| | - Xiayu Shi
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Basic and Clinical Application of Functional Nucleic Acids, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
| | - Yang Su
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Basic and Clinical Application of Functional Nucleic Acids, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
- Hangzhou Institute for Advanced Study (UCAS), Hangzhou, Zhejiang 310000, China
| | - Junlong Xia
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Basic and Clinical Application of Functional Nucleic Acids, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang 310014, China
| | - Yuting Li
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Basic and Clinical Application of Functional Nucleic Acids, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
| | - Xiangsheng Liu
- Zhejiang Cancer Hospital, The Key Laboratory of Zhejiang Province for Basic and Clinical Application of Functional Nucleic Acids, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, China
| |
Collapse
|
4
|
Wang J, Ye F, Chai H, Jiang Y, Wang T, Ran X, Xia Q, Xu Z, Fu Y, Zhang G, Wu H, Guo G, Guo H, Ruan Y, Wang Y, Xing D, Xu X, Zhang Z. Advances and applications in single-cell and spatial genomics. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1226-1282. [PMID: 39792333 DOI: 10.1007/s11427-024-2770-x] [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: 08/20/2024] [Accepted: 10/10/2024] [Indexed: 01/12/2025]
Abstract
The applications of single-cell and spatial technologies in recent times have revolutionized the present understanding of cellular states and the cellular heterogeneity inherent in complex biological systems. These advancements offer unprecedented resolution in the examination of the functional genomics of individual cells and their spatial context within tissues. In this review, we have comprehensively discussed the historical development and recent progress in the field of single-cell and spatial genomics. We have reviewed the breakthroughs in single-cell multi-omics technologies, spatial genomics methods, and the computational strategies employed toward the analyses of single-cell atlas data. Furthermore, we have highlighted the advances made in constructing cellular atlases and their clinical applications, particularly in the context of disease. Finally, we have discussed the emerging trends, challenges, and opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Jingjing Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Fang Ye
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Haoxi Chai
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China
| | - Yujia Jiang
- BGI Research, Shenzhen, 518083, China
- BGI Research, Hangzhou, 310030, China
| | - Teng Wang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
| | - Xia Ran
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China
| | - Qimin Xia
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China
| | - Ziye Xu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Yuting Fu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guodong Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hanyu Wu
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Guoji Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Zhejiang Provincial Key Lab for Tissue Engineering and Regenerative Medicine, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Hongshan Guo
- Bone Marrow Transplantation Center of the First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Institute of Hematology, Zhejiang University, Hangzhou, 310000, China.
| | - Yijun Ruan
- Life Sciences Institute and The Second Affiliated Hospital, Zhejiang University, Hangzhou, 310058, China.
| | - Yongcheng Wang
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Dong Xing
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
- Beijing Advanced Innovation Center for Genomics (ICG), Peking University, Beijing, 100871, China.
| | - Xun Xu
- BGI Research, Shenzhen, 518083, China.
- BGI Research, Hangzhou, 310030, China.
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen, 518083, China.
| | - Zemin Zhang
- Biomedical Pioneering Innovation Center (BIOPIC) and School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
5
|
LIANG HAISU, YAN WEI, LIU ZHI, HE YUNBO, HU JIAO, SHU ZHIWEI, LI HUIHUANG, OTHMANE BELAYDI, REN WENBIAO, QUAN CHAO, QIU DONGXU, CHEN MINFENG, XIONG WEI, ZHANG BINGNAN, LIU PEIHUA. Immunomodulatory behavior of CircRNAs in tumor microenvironment. Oncol Res 2025; 33:1105-1119. [PMID: 40296917 PMCID: PMC12034001 DOI: 10.32604/or.2024.054623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/13/2024] [Indexed: 04/30/2025] Open
Abstract
Circular RNAs (circRNAs) are a type of non coding RNA that possess unique single stranded circular structures formed through reverse splicing mechanisms. Due to the lack of a free end that is typically susceptible to degradation by nucleases, circular RNAs exhibit resistance to ribonuclease R, making them highly stable in eukaryotic cells. The complex relationship between circRNA dysregulation and various pathophysiological conditions, especially cancer. Tumor microenvironment (TME) is a collective term for various components surrounding tumors and is an important factor affecting tumor development. Simultaneous infiltration of TME by different types of immune cells; These immune cells interact with the TME, collectively forming the so-called "tumor immune microenvironment". The complex interactions between tumor cells and TME profoundly affect the behavior of malignant tumors, and circRNAs derived from tumor cells and TME cell components have become important mediators of immune response and evasion within the TME. CircRNAs can directly or indirectly regulate immune cells, thereby modulating anti-tumor immunity. This review highlights how circRNAs, especially those encapsulated in extracellular vesicles like exosomes, influence the differentiation, chemotaxis, and anti-tumor immune functions of immune cells within the TME. Metabolic reprogramming plays an important role in this process. The process of circRNAs regulating tumor immunity is affected by multiple factors, such as hypoxia and viral infection. This review emphasizes the roles of the interaction between circRNAs and the TME in tumor immune regulation and prospects the guiding significance of circRNAs in tumor immune checkpoint therapy.
Collapse
Affiliation(s)
- HAISU LIANG
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410000, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - WEI YAN
- Department of Urology, Shimen Hospital of TCM, Changde, 415300, China
| | - ZHI LIU
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410000, China
- Department of Urology, The Second Affiliated Hospital, Guizhou Medical University, Kaili, 556000, China
| | - YUNBO HE
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410000, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410000, China
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - JIAO HU
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - ZHIWEI SHU
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - HUIHUANG LI
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - BELAYDI OTHMANE
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - WENBIAO REN
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410000, China
- George Whipple Lab for Cancer Research, University of Rochester Medical Institute, Rochester, NY 14627, USA
| | - CHAO QUAN
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - DONGXU QIU
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - MINFENG CHEN
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - WEI XIONG
- NHC Key Laboratory of Carcinogenesis, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - BINGNAN ZHANG
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410000, China
| | - PEIHUA LIU
- Department of Urology, Xiangya Hospital, Central South University, Changsha, 410000, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410000, China
| |
Collapse
|
6
|
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
|
7
|
Chen C, Wen L, Chen G, Yang F, Chen Z, Ji J, Gu J. Pan-cancer analysis of ITGB3 as a potential prognostic and immunological biomarker. Discov Oncol 2025; 16:522. [PMID: 40220261 PMCID: PMC11993531 DOI: 10.1007/s12672-025-02300-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
Integrin β3 (ITGB3) acts as a crucial regulator and target within the tumor immune microenvironment (TIME) and is highly expressed in the TIME of various tumors. The TIMER, TCGA, GTEx, and CCLE databases were utilized to comprehensively analyze the differential expression of ITGB3 in tumor tissues. Kaplan-Meier analysis, forest plots, and univariate and multivariate Cox regression were used to assess the genetic alterations, clinicopathological characteristics, and prognostic value of ITGB3. Additionally, the R software package was used to evaluate the relationship between ITGB3 expression and immune cell infiltration, immunomodulatory genes, and immune checkpoints, and potential signaling pathways were examined through differential expression and enrichment analysis. We found that the high expression of ITGB3 is a significant risk factor for six types of cancer, including adrenocortical carcinoma (ACC), and is closely associated with a lower survival rate. Anti-tumor immune cells (CD8 + T cells, CD4 + Th1 cells, and NKT cells) were significantly reduced. By contrast, pro-tumor immune cells (Tregs and CD4 + Th2 cells), immune checkpoints (CTLA4 and PD-CD1), and negatively regulated co-stimulators of T-cell activation (CTLA4, PD-CD1, and IL10) were significantly elevated in most types of cancer with high ITGB3 expression. Overall, our preliminary results indicate that ITGB3 plays an important role in immunosuppression in the tumor microenvironment. Elevated levels of ITGB3 inhibit tumor immunity, facilitate tumor immune escape, and affect patient prognosis, and it may be a prognostic biomarker.
Collapse
Affiliation(s)
- Changshun Chen
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming, 650032, China
| | - Lei Wen
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming, 650032, China
| | - Ge Chen
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming, 650032, China
| | - Fei Yang
- Department of Orthopedics, Nanchong Central Hospital, Nanchong, 637000, China
| | - Zhong Chen
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming, 650032, China
| | - Jianhua Ji
- Department of Orthopedics and Trauma Surgery, Affiliated Hospital of Yunnan University, Kunming, 650032, China.
| | - Jinyi Gu
- Clinical Laboratory of Affiliated Hospital of Yunnan University, Kunming, 650032, China.
| |
Collapse
|
8
|
Kulkarni AD, Mukarrama T, Barlow BR, Kim J. Recent advances in non-invasive in vivo tracking of cell-based cancer immunotherapies. Biomater Sci 2025; 13:1939-1959. [PMID: 40099377 PMCID: PMC11980607 DOI: 10.1039/d4bm01677g] [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: 03/19/2025]
Abstract
Immunotherapy has been at the forefront of cancer treatment research in recent years due to an increased understanding of the immune system's role in cancer and the substantial benefits it has demonstrated compared to conventional treatment methods. In particular, immune cell-based approaches utilizing T cells, natural killer (NK) cells, macrophages, and more have shown great potential as cancer treatments. While these treatments hold promise, there are still numerous issues that limit their clinical translation, including a lack of understanding of their mechanisms and inconsistent responses to treatment. Traditionally, tissue or blood samples are collected as a means of monitoring treatment progression. However, these in vitro diagnostics are invasive and provide limited information about the real-time status of the treatment or its long-term effectiveness. To address these limitations, novel non-invasive imaging modalities have been developed. These include optical imaging, X-ray computed tomography (CT), magnetic resonance imaging (MRI), positron emission tomography (PET) and single-photon emission computed tomography (SPECT), and photoacoustic (PA) imaging. This review focuses on methods for tracking cell-based cancer immunotherapies using these in vivo imaging modalities, thereby enhancing real-time monitoring of their therapeutic effect and predictions of their long-term efficacy.
Collapse
Affiliation(s)
- Anika D Kulkarni
- Department of Biomedical Engineering, University of California, Davis, Davis, 95616, USA.
| | - Tasneem Mukarrama
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, 95817, USA
| | - Brendan R Barlow
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, 95817, USA
| | - Jinhwan Kim
- Department of Biomedical Engineering, University of California, Davis, Davis, 95616, USA.
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, 95817, USA
| |
Collapse
|
9
|
Nowak JA, Cho E, Davis MA, Zheng S, Bell L, Sha F, Magdalenski JS, Farha OK, Teplensky MH. Strengthening Antisense Oligonucleotide-Mediated Anti-Tumor Immunity via Metal-Organic Framework Nanoparticles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.28.645811. [PMID: 40235985 PMCID: PMC11996403 DOI: 10.1101/2025.03.28.645811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Overexpression of checkpoint proteins, such as programmed death ligand one (PD-L1), prevents immune recognition and enables cancer growth. Current monoclonal antibodies that block PD-L1 tend to be fragile, unable to penetrate tumors, and target cancer at later stages, thus leading to inconsistent patient outcomes. Antisense oligonucleotides (ASOs) provide an alternative to decrease PD-L1 expression, but require frequent high dosing due to fast degradation, rapid clearance, and poor cell uptake. To overcome these issues, we harnessed biocompatible metal-organic framework (MOF) nanoparticles, porous nanomaterials comprising metal nodes and organic linkers, to deliver ASOs. Encapsulating ASOs into MOFs enhances their stability and protection during intracellular delivery, leading to reduced PD-L1 expression and downstream immune recognition. Herein, we synthesized three distinct PD-L1-specific ASOs and loaded them individually into zirconium-based nano-sized NU-1000 MOFs, averaging ∼80% encapsulation efficiency. Release of encapsulated ASOs was sustained up to 7 days ex cellulo . MOF encapsulation increased ASO potency and reduced PD-L1 expression ∼ 3-fold and 2-fold in triple negative breast cancer EMT6 and melanoma B16-F10 cells, respectively. We evaluated the impact of MOF-delivered ASOs on PD-L1-expressing immune cells, where we observed ca. 12-fold increases in dendritic cell co-stimulatory marker expression, and amplified T cell activation and proliferation compared to untreated cells (4-fold and 10-fold, respectively). Notably, these changes drove a 3-fold increase in tumor caspase-3 expression, a key mediator for apoptosis. This research highlights how MOFs can be harnessed to bypass ASO limitations without requiring sequence modifications, and offers a broadly applicable platform for improved oligonucleotide delivery for various genes of interest.
Collapse
|
10
|
Gao Y, Huo Y, Wang L, Ruan J, Chen L, Li H, Hong G. Relative expression orderings based prediction of treatment response to Anti-PD-1 immunotherapy in advanced melanoma. Sci Rep 2025; 15:10235. [PMID: 40133499 PMCID: PMC11937249 DOI: 10.1038/s41598-025-94931-0] [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: 11/07/2024] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Programmed cell death protein 1 (PD-1) plays a critical role in immune tolerance and evasion within the tumor microenvironment, and anti-PD-1 immunotherapy has shown efficacy in treating advanced melanoma. However, response rates vary significantly among patients, necessitating the identification of reliable biomarkers to predict treatment efficacy. Based on within-sample relative expression orderings, we analyzed RNA sequencing data from melanoma patients to construct a predictive model comprising gene pairs associated with treatment response. The model's performance was validated across multiple independent datasets and assessed for correlations with immune infiltration and survival outcomes. The constructed 15-pair model achieved a prediction accuracy of 100% in training datasets and 89.47% in validation sets. Validation in melanoma patients lacking treatment response data revealed significant differences between predicted responders and non-responders across datasets, with the model being an independent prognostic factor. Increased immune cell infiltration was observed in responders, correlating with higher expression levels of key immune checkpoint genes. The relative expression orderings-based model shows promise as a tool for predicting responses to anti-PD-1 therapy in melanoma patients, supporting personalized treatment strategies.
Collapse
Affiliation(s)
- Yaru Gao
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, 341000, China
| | - Yue Huo
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, 341000, China
| | - Lingli Wang
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou, 341000, China
| | - Jiayi Ruan
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou, 341000, China
| | - Lanzhen Chen
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou, 341000, China
| | - Hongdong Li
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou, 341000, China.
| | - Guini Hong
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
11
|
Giri S, Lamichhane G, Pandey J, Khadayat R, K. C. S, Devkota HP, Khadka D. Immune Modulation and Immunotherapy in Solid Tumors: Mechanisms of Resistance and Potential Therapeutic Strategies. Int J Mol Sci 2025; 26:2923. [PMID: 40243502 PMCID: PMC11989189 DOI: 10.3390/ijms26072923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Understanding the modulation of specific immune cells within the tumor microenvironment (TME) offers new hope in cancer treatments, especially in cancer immunotherapies. In recent years, immune modulation and resistance to immunotherapy have become critical challenges in cancer treatments. However, novel strategies for immune modulation have emerged as promising approaches for oncology due to the vital roles of the immunomodulators in regulating tumor progression and metastasis and modulating immunological responses to standard of care in cancer treatments. With the progress in immuno-oncology, a growing number of novel immunomodulators and mechanisms are being uncovered, offering the potential for enhanced clinical immunotherapy in the near future. Thus, gaining a comprehensive understanding of the broader context is essential. Herein, we particularly summarize the paradoxical role of tumor-related immune cells, focusing on how targeted immune cells and their actions are modulated by immunotherapies to overcome immunotherapeutic resistance in tumor cells. We also highlight the molecular mechanisms employed by tumors to evade the long-term effects of immunotherapeutic agents, rendering them ineffective.
Collapse
Affiliation(s)
- Suman Giri
- Asian College for Advance Studies, Purbanchal University, Satdobato, Lalitpur 44700, Nepal;
| | - Gopal Lamichhane
- Department of Nutritional Sciences, Oklahoma State University, Stillwater, OK 74078, USA;
| | - Jitendra Pandey
- Department of Chemistry, University of Hawai’i at Manoa, 2545 McCarthy Mall, Honolulu, HI 96822, USA;
| | - Ramesh Khadayat
- Patan Hospital, Patan Academic of Health Sciences, Lagankhel, Lalitpur 44700, Nepal;
| | - Sindhu K. C.
- Department of Pharmacology, Chitwan Medical College, Tribhuwan University, Bharatpur-05, Chitwan 44200, Nepal;
| | - Hari Prasad Devkota
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Oehonmachi 5-1, Chuo-ku, Kumamoto 862-0973, Japan;
- Headquarters for Admissions and Education, Kumamoto University, Kurokami, 2-39-1, Chuo-ku, Kumamoto 860-8555, Japan
| | - Dipendra Khadka
- NADIANBIO Co., Ltd., Wonkwang University School of Medicine, Business Incubation Center R201-1, Iksan 54538, Jeonbuk, Republic of Korea
- KHAS Health Pvt. Ltd., Dhangadhi-04, Kailali 10910, Nepal
| |
Collapse
|
12
|
Virazels M, Lusque A, Brayer S, Genais M, Dufau C, Milhès J, Filleron T, Pagès C, Sibaud V, Mortier L, Dereure O, Ayyoub M, Fabre A, Andrieu-Abadie N, Pancaldi V, Colacios C, Meyer N, Ségui B, Montfort A. TNF signature in advanced melanoma patients treated with immune checkpoint inhibitors: Results from the MELANFα clinical study. Int J Cancer 2025. [PMID: 40098565 DOI: 10.1002/ijc.35416] [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: 09/19/2024] [Revised: 02/14/2025] [Accepted: 03/04/2025] [Indexed: 03/19/2025]
Abstract
Resistance to immune checkpoint inhibitors (ICI) in cancer patients is not fully understood, and predictive biomarkers are lacking. MELANFα (NCT03348891) is an open-label, prospective, multicenter cohort of 60 patients with advanced melanoma receiving ICI (bitherapy: ipilimumab + nivolumab; monotherapy: pembrolizumab or nivolumab). The primary objective was to evaluate whether changes in plasma TNF between baseline (W0) and week 12 (W12) identified patients with non-progressive disease at W12. Secondary and exploratory objectives were to assess the association between plasma TNF, tumor response, and changes in circulating T cells. Plasma TNF increased along therapy, but its W12/W0 fold change was not associated with non-progressive disease at W12. However, plasma TNF levels at W12 were significantly higher in non-responders than in responders across therapies (p = .0129). The remodeling of circulating T cell subpopulations was mostly triggered by bitherapy. Increased proportions of circulating central memory and effector memory CD8 T cells after bitherapy were positively and negatively associated with response to treatment, respectively. In this cohort, circulating T cells from responders and non-responders also displayed distinct molecular characteristics. Indeed, responders showed an increased proportion of CD8 T cells with low enrichment of TNF-related pathways and high cytotoxic potential, while non-responders displayed increased proportions of circulating CD8 EM T cells enriched for TNF-related pathways and directed toward cytokine expression. In conclusion, our study shows that elevated plasma TNF and enriched TNF pathways in T cells are associated with poorer clinical outcomes, reinforcing the notion that TNF may dampen ICI efficacy.
Collapse
Affiliation(s)
- Mathieu Virazels
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France
- Equipe Labellisée Fondation ARC pour la recherche sur le cancer, Toulouse, France
| | - Amélie Lusque
- Biostatistics & Health Data Science Unit, Oncopole Claudius Regaud, Toulouse, France
- Institut Universitaire du Cancer (IUCT-O), Toulouse, France
| | - Stéphanie Brayer
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France
- Equipe Labellisée Fondation ARC pour la recherche sur le cancer, Toulouse, France
- Service d'Oncodermatologie, IUCT-O, CHU de Toulouse, Toulouse, France
| | - Matthieu Genais
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France
- Equipe Labellisée Fondation ARC pour la recherche sur le cancer, Toulouse, France
| | - Carine Dufau
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France
- Equipe Labellisée Fondation ARC pour la recherche sur le cancer, Toulouse, France
| | - Jean Milhès
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France
- Equipe Labellisée Fondation ARC pour la recherche sur le cancer, Toulouse, France
| | - Thomas Filleron
- Biostatistics & Health Data Science Unit, Oncopole Claudius Regaud, Toulouse, France
- Institut Universitaire du Cancer (IUCT-O), Toulouse, France
| | - Cécile Pagès
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France
- Equipe Labellisée Fondation ARC pour la recherche sur le cancer, Toulouse, France
- Institut Universitaire du Cancer (IUCT-O), Toulouse, France
- Service d'Oncodermatologie, IUCT-O, CHU de Toulouse, Toulouse, France
| | - Vincent Sibaud
- Institut Universitaire du Cancer (IUCT-O), Toulouse, France
- Service d'Oncodermatologie, IUCT-O, CHU de Toulouse, Toulouse, France
| | | | - Olivier Dereure
- Department of Dermatology, University of Montpellier, Montpellier, France
| | - Maha Ayyoub
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France
- Equipe Labellisée Fondation ARC pour la recherche sur le cancer, Toulouse, France
- Institut Universitaire du Cancer (IUCT-O), Toulouse, France
| | - Amandine Fabre
- Institut Universitaire du Cancer (IUCT-O), Toulouse, France
- Clinical Research Department, Oncopole Claudius Regaud, Toulouse, France
| | - Nathalie Andrieu-Abadie
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France
- Equipe Labellisée Fondation ARC pour la recherche sur le cancer, Toulouse, France
| | - Vera Pancaldi
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France
| | - Céline Colacios
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France
- Equipe Labellisée Fondation ARC pour la recherche sur le cancer, Toulouse, France
| | - Nicolas Meyer
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France
- Equipe Labellisée Fondation ARC pour la recherche sur le cancer, Toulouse, France
- Institut Universitaire du Cancer (IUCT-O), Toulouse, France
- Service d'Oncodermatologie, IUCT-O, CHU de Toulouse, Toulouse, France
| | - Bruno Ségui
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France
- Equipe Labellisée Fondation ARC pour la recherche sur le cancer, Toulouse, France
| | - Anne Montfort
- Unité Mixte de Recherche INSERM 1037, CNRS 5071, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse (CRCT), Toulouse, France
- Equipe Labellisée Fondation ARC pour la recherche sur le cancer, Toulouse, France
| |
Collapse
|
13
|
Sawant A, Shi F, Cararo Lopes E, Hu Z, Abdelfattah S, Baul J, Powers JR, Hinrichs CS, Rabinowitz JD, Chan CS, Lattime EC, Ganesan S, White EP. Immune Checkpoint Blockade Delays Cancer Development and Extends Survival in DNA Polymerase Mutator Syndromes. Cancer Res 2025; 85:1130-1144. [PMID: 39786467 PMCID: PMC11907192 DOI: 10.1158/0008-5472.can-24-2589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/01/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
Mutations in the exonuclease domains of the replicative nuclear DNA polymerases POLD1 and POLE are associated with increased cancer incidence, elevated tumor mutation burden (TMB), and enhanced response to immune checkpoint blockade (ICB). Although ICB is approved for treatment of several cancers, not all tumors with elevated TMB respond, highlighting the need for a better understanding of how TMB affects tumor biology and subsequently immunotherapy response. To address this, we generated mice with germline and conditional mutations in the exonuclease domains of Pold1 and Pole. Engineered mice with Pold1 and Pole mutator alleles presented with spontaneous cancers, primarily lymphomas, lung cancer, and intestinal tumors, whereas Pold1 mutant mice also developed tail skin carcinomas. These cancers had highly variable tissue type-dependent increased TMB with mutational signatures associated with POLD1 and POLE mutations found in human cancers. The Pold1 mutant tail tumors displayed increased TMB; however, only a subset of established tumors responded to ICB. Similarly, introducing the mutator alleles into mice with lung cancer driven by mutant Kras and Trp53 deletion did not improve survival, whereas passaging these tumor cells in vitro without immune editing and subsequently implanting them into immunocompetent mice caused tumor rejection in vivo. These results demonstrated the efficiency by which cells with antigenic mutations are eliminated in vivo. Finally, ICB treatment of mutator mice earlier, before observable tumors had developed delayed cancer onset, improved survival and selected for tumors without aneuploidy, suggesting the potential of ICB in high-risk individuals for cancer prevention. Significance: Treating high-mutation burden mice with immunotherapy prior to cancer onset significantly improves survival, raising the possibility of utilizing immune checkpoint blockade for cancer prevention, especially in individuals with increased risk.
Collapse
Affiliation(s)
- Akshada Sawant
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, New Jersey
| | - Fuqian Shi
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
| | | | - Zhixian Hu
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, New Jersey
| | - Somer Abdelfattah
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
| | - Jennele Baul
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
| | - Jesse R. Powers
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, New Jersey
| | | | - Joshua D. Rabinowitz
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, New Jersey
| | - Chang S. Chan
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
- Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Edmund C. Lattime
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
- Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Shridar Ganesan
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
- Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Eileen P. White
- Rutgers Cancer Institute, Rutgers University, New Brunswick, New Jersey
- Ludwig Princeton Branch, Ludwig Institute for Cancer Research, Princeton University, Princeton, New Jersey
- Department of Molecular Biology and Biochemistry, Piscataway, New Jersey
| |
Collapse
|
14
|
Duan J, Chen T, Li Q, Zhang Y, Lu T, Xue J, Sun Y, Gao L, Zhang Y. Protein arginine methyltransferase 6 enhances immune checkpoint blockade efficacy via the STING pathway in MMR-proficient colorectal cancer. J Immunother Cancer 2025; 13:e010639. [PMID: 40086819 PMCID: PMC11907083 DOI: 10.1136/jitc-2024-010639] [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: 09/22/2024] [Accepted: 02/26/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND The emergence of immunotherapy has revolutionized the paradigm of cancer treatment with immune checkpoint blockades (ICB) in solid cancers, including colorectal cancer (CRC). However, only a small subset of CRC patients harboring deficient mismatch repair (dMMR) or microsatellite instability-high (MSI-H) benefits from ICB therapy. A very limited response to ICB therapy has been achieved in MMR-proficient CRC, representing a significant challenge limiting the clinical application of immunotherapy. MMR is the critical DNA repair pathway that maintains genomic integrity by correcting DNA mismatches, which is mediated by the MutSα or MutSβ complex consisting of MSH2 with MSH6 and MSH3, respectively. Given that MMR status directs effective immune response, we sought to determine whether targeting MMR capacity boosts ICB efficacy. METHODS Azoxymethane/dextran sodium sulfate (AOM/DSS)-induced CRC and xenograft model were used to evaluate the function of PRMT6 and response to PRMT6 inhibitor EPZ020411 and combination therapy of PD1 and EPZ020411. Biochemical assays were performed to elucidate the underlying mechanism of PRMT6-mediated MSH2 methylation and immune evasion. RESULTS We have identified PRMT6 as a crucial regulator of MMR capacity via MSH2 dimethylation at R171 and R219. Such a modification abrogates its MMR capacity and prevents the recruitment of MSH3 and MSH6. PRMT6 loss or inhibition triggers cytosolic DNA accumulation and cGAS-STING signaling activation, leading to enhanced immune response in PRMT6-deficient colon tumors or xenografts. Pharmacological inhibition of PRMT6 using EPZ020411 promotes mutagenesis and destabilizes MutSα or MutSβ assembly, and prolonged EPZ020411 exposure maintains an MSI-like phenotype in microsatellite stability (MSS) cells. EPZ020411 treatment sensitizes ICB efficacy of MSS cells, but not MSI cells in vivo. Similar effects have been observed in MSS colon tumors induced by AOM/DSS. CONCLUSIONS Our study provides a preclinical proof of concept to overcome resistance to immunotherapy by targeting PRMT6 in CRC with MSS.
Collapse
Affiliation(s)
- Jinlin Duan
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Department of Pathology, Tongren Hospital Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Tao Chen
- Department of Biliary-Pancreatic Surgery, Shanghai Jiao Tong University, Shanghai, China
| | - Qiwei Li
- Department of Biliary-Pancreatic Surgery, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Zhang
- Department of Clinical Laboratory, Shanghai Sixth People's Hospital, Shanghai, China
- Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Ting Lu
- Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Junyan Xue
- Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Yang Sun
- Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Ling Gao
- Department of General Surgery, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yonglong Zhang
- Laboratory of Targeted Therapy and Precision Medicine, Department of Clinical Laboratory, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
- Department of General Surgery, Shanghai 6th Peoples Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
15
|
Qian C, Sun Y, Yue Y. Construction and Validation of a T Cell Exhaustion-Related Prognostic Signature in Cholangiocarcinoma. Int J Genomics 2025; 2025:8823837. [PMID: 40226355 PMCID: PMC11991809 DOI: 10.1155/ijog/8823837] [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: 11/25/2024] [Accepted: 01/24/2025] [Indexed: 04/15/2025] Open
Abstract
Objective: T cell exhaustion (TEX) is a critical determinant of immune resistance. This study was performed to investigate the key genes linked to TEX in cholangiocarcinoma (CCA) and construct a TEX-associated gene signature to forecast the prognosis of patients with CCA. Methods: Based on the expression data acquired from the E-MTAB-6389 dataset, the TEX-related modules and module genes were identified using weighted coexpression network analysis (WGCNA). Subsequently, a TEX-related prognostic signature was built by using the univariate and least absolute shrinkage and selection operator (LASSO) Cox regression analysis. The immune cell infiltration in each CCA sample was evaluated using the single-sample gene set enrichment analysis (ssGSEA) package, followed by single-cell RNA sequencing (scRNA-seq) analysis. Furthermore, the expression of TEX-related genes in the gene signature was experimentally validated in CCA cells by quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) and western blot analysis. Results: A total of 15 TEX-associated modules and 23 module genes were identified. Then, a four-gene signature related to TEX was established, containing Palladin, Cytoskeletal Associated Protein (PALLD), Member RAS Oncogene Family (RAB31), ADAM Metallopeptidase With Thrombospondin Type 1 Motif 2 (ADAMTS2), and WISP1, which could predict prognosis of patients with CCA. Moreover, neutrophils, endothelial cells, B cells, and T cells exhibited significant infiltration in CCA samples, and these four TEX-related genes were both significantly positively correlated with T cells, endothelial cells, and B cells while negatively correlated with neutrophils. Moreover, a total of 13 cell types were annotated after scRNA-seq analysis. Notably, RAB31 was mainly highly expressed in monocytes, macrophages, DC2 (Dendritic Cells 2), and DC3 (Dendritic Cells 3), and PALLD, ADAMTS2, and WISP1 were mainly overexpressed in fibroblasts. Furthermore, experimental validation revealed that the expression levels of PALLD, RAB31, ADAMTS2, and WISP1 were consistent with the trend results of bioinformatics analysis. Conclusion: A prognostic signature was developed by four TEX-related genes, including PALLD, RAB31, ADAMTS2, and WISP1, which might be a powerful predictor for the prognosis of patients with CCA. These TEX-related genes were related to the infiltration of neutrophils, endothelial cells, B cells, and T cells in CCA.
Collapse
Affiliation(s)
- Changshi Qian
- Department of Hepatopancreatobiliary Surgery, Affiliated Hospital of Yanbian University, Yanji, China
| | - Yuqiao Sun
- Department of Hepatopancreatobiliary Surgery, Affiliated Hospital of Yanbian University, Yanji, China
| | - Yihuai Yue
- Department of Surgery, Medical College of Yanbian University, Yanji, China
| |
Collapse
|
16
|
Vinjamuri S, Pant V. Demystifying the Role of Immuno PET-CT in Non-Small Cell Lung Cancer: Clinical Value and Research Trends. Semin Nucl Med 2025; 55:212-220. [PMID: 40016063 DOI: 10.1053/j.semnuclmed.2025.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 03/01/2025]
Abstract
The management of Lung cancer, especially non-small cell lung cancer has undergone a paradigm shift recently with the advent of new treatment approaches including focused radiotherapy as well as evolution of a newer class of immunotherapy agents. Treatment efficacy and survival rates have improved and it is now even more important that patients are selected for appropriate interventions on the basis of a comprehensive assessment including a range of imaging as well as in-vitro tests such as immunohistochemistry. A new class of tracers targeting programmed cell death such as PD1 and PDL1 (broadly classed as Immuno PET) are being increasingly used in the molecular characterisation of patients deemed resistant to standard treatment approaches and being considered for additional interventions such as immunotherapy. In this review, we review the latest evidence in the field and propose a summary of clinical usefulness and provide a review of the research trends in this exciting and evolving field.
Collapse
Affiliation(s)
- Sobhan Vinjamuri
- Department of Nuclear Medicine, Royal Liverpool University Hospital, Liverpool, UK.
| | - Vineet Pant
- Department of Nuclear Medicine, Royal Liverpool University Hospital, Liverpool, UK
| |
Collapse
|
17
|
Shi R, Sun J, Zhou Z, Shi M, Wang X, Gao Z, Zhao T, Li M, Shu Y. Integration of multiple machine learning approaches develops a gene mutation-based classifier for accurate immunotherapy outcomes. NPJ Precis Oncol 2025; 9:54. [PMID: 40011681 DOI: 10.1038/s41698-025-00842-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 02/17/2025] [Indexed: 02/28/2025] Open
Abstract
In addition to traditional biomarkers like PD-(L)1 expression and tumor mutation burden (TMB), more reliable methods for predicting immune checkpoint blockade (ICB) response in cancer patients are urgently needed. This study utilized multiple machine learning approaches on nonsynonymous mutations to identify key mutations that are most significantly correlated to ICB response. We proposed a classifier, Gene mutation-based Predictive Signature (GPS), to categorize patients based on their predicted response and clinical outcomes post-ICB therapy. GPS outperformed conventional predictors when validated in independent cohorts. Multi-omics analysis and multiplex immunohistochemistry (mIHC) revealed insights into tumor immunogenicity, immune responses, and the tumor microenvironment (TME) in lung adenocarcinoma (LUAD) across different GPS groups. Finally, we validated distinct responses of different GPS samples to ICB in an ex-vivo tumor organoid-PBMC co-culture model. Overall, our findings highlight a simple, robust classifier for accurate ICB response prediction, which could reduce costs, shorten testing times, and facilitate clinical implementation.
Collapse
Affiliation(s)
- Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jing Sun
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- The First Clinical Medical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhaokai Zhou
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meiqi Shi
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xin Wang
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Zhaojia Gao
- Department of Thoracic Surgery, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Tianyu Zhao
- Institute and Clinic for Occupational, Social and Environmental Medicine, LMU University Hospital Munich, Munich, Germany
| | - Minglun Li
- Department of Radiation Oncology, Lueneburg Municipal Hospital, Lueneburg, Germany
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
18
|
Qi J, Zhou M, Yang N, Ma H, He M, Wu G, Ge C, Jin L, Cheng L, Liao W, Ren H, Lei C. TUBA1B as a novel prognostic biomarker correlated with immunosuppressive tumor microenvironment and immunotherapy response. Front Pharmacol 2025; 16:1517887. [PMID: 39968182 PMCID: PMC11832512 DOI: 10.3389/fphar.2025.1517887] [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: 10/27/2024] [Accepted: 01/03/2025] [Indexed: 02/20/2025] Open
Abstract
Background: Tubulin alpha 1b (TUBA1B) is a key microtubule protein essential for maintaining cellular structure and function. This protein contributes significantly to cytoskeletal formation and is implicated in various diseases. Despite its fundamental roles, TUBA1B's impact on tumor prognosis and the tumor immune microenvironment across cancer types remains inadequately understood. Methods To elucidate TUBA1B's role in cancer prognosis and immune response, we conducted a comprehensive analysis, integrating data from established databases such as The Cancer Genome Atlas, Genotype Tissue Expression, Cancer Cell Lineage Encyclopedia, Human Protein Atlas, Kaplan-Meier Plotter, cBioPortal, TIMER, and ImmuCellAI, along with a large-scale clinical study and immunotherapy cohort. We also conducted in vitro functional assays to assess TUBA1B's functional role in tumor cells, allowing for a detailed examination of its relationship with cancer prognosis and immune modulation. Results: Our findings indicate that TUBA1B expression is dysregulated across multiple cancers, correlating strongly with poor survival outcomes and advanced pathological stages. Functional enrichment analyses further revealed that TUBA1B regulates key cell cycle processes, driving tumor proliferation, migration, and invasion. It also influences immune functions within both the innate and adaptive immune systems, affecting immune-related signaling pathways. These insights underscore TUBA1B's multifaceted role in cancer progression and immune response. Conclusion: This study highlights TUBA1B's potential as a human oncogene with substantial roles in tumorigenesis and immune regulation. Elevated TUBA1B levels are associated with an immunosuppressive tumor microenvironment, impacting cancer progression and treatment outcomes. Targeting TUBA1B may offer promising therapeutic avenues for enhancing cancer treatment, offering new perspectives for innovative anti-tumor strategies with high clinical impact.
Collapse
Affiliation(s)
- Juntao Qi
- Rehabilitation Medicine Department, Hunan Aerospace Hospital, Hunan Normal University, Changsha, Hunan, China
- Research Center of Clinical Medicine, Shenzhen Hospital of Shanghai University of Traditional Chinese Medicine, Shenzhen, China
| | - Mingming Zhou
- Department of Critical Care Medicine, Chongqing University Affiliated Cancer Hospital, Chongqing, China
| | - Na Yang
- Laboratory of Oncology and Immunology, School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou, China
| | - Huiyun Ma
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Min He
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Gujie Wu
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Chang Ge
- School of Medicine, Wuhan University, Wuhan, China
| | - Liuyin Jin
- School of Medicine, Wuhan University, Wuhan, China
| | - Lin Cheng
- Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Liao
- Department of Otolaryngology and Head and Neck Surgery, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Hefei Ren
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Caiyun Lei
- Rehabilitation Medicine Department, Hunan Aerospace Hospital, Hunan Normal University, Changsha, Hunan, China
| |
Collapse
|
19
|
Huang JB, Zhou ZY, Lu J, Zhu JY, Lai B, Mao SX, Cao JQ. Inflammatory burden index as a prognostic marker in patients with advanced gastric cancer treated with neoadjuvant chemotherapy and immunotherapy. Front Immunol 2025; 15:1471399. [PMID: 39906738 PMCID: PMC11790653 DOI: 10.3389/fimmu.2024.1471399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 12/23/2024] [Indexed: 02/06/2025] Open
Abstract
Background Blood inflammation index has been shown to correlate with the prognosis of patients with gastric cancer. However, few studies have compared the efficacy of existing blood inflammatory markers in predicting the prognosis of patients with locally advanced gastric cancer in combination with neoadjuvant chemotherapy and immunotherapy. Objective The objective of this study was to compare the prognostic value of existing commonly used blood inflammatory index in patients with advanced gastric cancer treated with neoadjuvant chemotherapy combined with immunotherapy. Methods The clinicopathological data of patients with advanced gastric cancer from three centers in China were analyzed retrospectively. Univariate COX regression analysis was used to analyze the independent risk factors of poor tumor regression and overall survival (OS) in this part of patients, and the predictive value of different inflammatory indexes on prognosis was compared by C-index index. Finally, Inflammatory burden index(IBI) was grouped by X-tile software, and Kaplan-Meier method was used to compare the survival difference between groups. Results A total of 163 patients were enrolled in this study. The median age was 63 years(56-68). The median cycle of neoadjuvant therapy was 4(3-4). The median survival time was 85.1%(1 years), 65.6%(2 years), and 47.4%(3 years).Univariate analysis showed that IBI was an independent risk factor for non-TR(residual tumor cells>50%) (HR=1.08,95%CI:1.00-1.45,p<0.001)and OS(HR=1.04,95%CI:1.03-1.05,p<0.001). IBI is the best predictor of OS (C-index: 0.82, 95% CI: 0.78-0.87) among all inflammatory indexes. The IBI cutoff value was 52.1. It was found that the high IBI group had a higher incidence of postoperative complications(32.1%vs14.3%, p=0.001), the proportion of non-TR patients was significantly higher than that of the low IBI group(64.3%vs35.7%, p =0.001), and the high IBI group had a significantly lower OS((47.6% vs 87.6%, p < 0.001). Conclusion IBI is the best inflammatory index to predict the prognosis of advanced gastric cancer treated with neoadjuvant chemotherapy combined with immunotherapy, which will help guide patients' treatment decisions. This result still needs to be verified by large prospective studies.
Collapse
Affiliation(s)
- Jiao-Bao Huang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhi-Yong Zhou
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jun Lu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Ji-Yun Zhu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Bin Lai
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Sheng-Xun Mao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jia-Qing Cao
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
20
|
Fan T, Xiao C, Deng Z, Li S, Tian H, Zheng Y, Zheng B, Li C, He J. Signatures of H3K4me3 modification predict cancer immunotherapy response and identify a new immune checkpoint-SLAMF9. Respir Res 2025; 26:17. [PMID: 39815269 PMCID: PMC11734478 DOI: 10.1186/s12931-024-03093-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/31/2024] [Indexed: 01/18/2025] Open
Abstract
H3 lysine 4 trimethylation (H3K4me3) modification and related regulators extensively regulate various crucial transcriptional courses in health and disease. However, the regulatory relationship between H3K4me3 modification and anti-tumor immunity has not been fully elucidated. We identified 72 independent prognostic genes of lung adenocarcinoma (LUAD) whose transcriptional expression were closely correlated with known 27 H3K4me3 regulators. We constructed three H3K4me3 modification patterns utilizing the expression profiles of the 72 genes, and patients classified in each pattern exhibited unique tumor immune infiltration characteristics. Using the principal component analysis (PCA) of H3K4me3-related patterns, we constructed a H3K4me3 risk score (H3K4me3-RS) system. The deep learning analysis using 12,159 cancer samples from 26 cancer types and 725 cancer samples from 5 immunotherapy cohorts revealed that H3K4me3-RS was significantly correlated with cancer immune tolerance and sensitivity. Importantly, this risk-score system showed satisfactory predictive performance for the ICB therapy responses of patients suffering from several cancer types, and we identified that SLAMF9 was one of the immunosuppressive phenotype and immunotherapy resistance-determined genes of H3K4me3-RS. The mice melanoma model showed Slamf9 knockdown remarkably restrained cancer progression and enhanced the efficacy of anti-CTLA-4 and anti-PD-L1 therapies by elevating CD8 + T cell infiltration. This study provided a new H3K4me3-associated biomarker system to predict tumor immunotherapy response and suggested the preclinical rationale for investigating the roles of SLAMF9 in cancer immunity regulation and treatment.
Collapse
Affiliation(s)
- Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuofeng Li
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bo Zheng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
21
|
Xu X, Fa L, Sun X, Yang F, Liu Y, Song J, Zhao Y, Dong J. Integrative analysis of ferroptosis in the hypoxic microenvironment of gastric cancer unveils the immune landscape and personalized therapeutic strategies. Front Oncol 2025; 14:1499580. [PMID: 39871942 PMCID: PMC11769819 DOI: 10.3389/fonc.2024.1499580] [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: 09/21/2024] [Accepted: 12/06/2024] [Indexed: 01/29/2025] Open
Abstract
Background Ferroptosis is a cell death mode caused by excessive accumulation of lipid peroxides caused by disturbance of intracellular metabolic pathway, which is closely related to iron and cholesterol metabolism homeostasis. Its regulation within the hypoxic metabolic tumor microenvironment (TME) has the potential to improve the effectiveness of tumor immunotherapy. The predictive role of ferroptosis in gastric cancer (GC) hypoxia TME, particularly in relation to TME immune cell infiltration, has not been fully explained. Methods By analyzing the mRNA expression data of ferroptosis and hypoxia-related genes, a prediction model was constructed to evaluate further the predictive value of immune cell infiltration, clinical characteristics, and immunotherapy efficacy of gastric cancer, and the essential genes were validated. Results Two distinct molecular states of ferroptosis-hypoxia were identified in GC. Notably, patients with high ferroptosis-hypoxia risk scores (FHRS) displayed significant levels of hypoxia and epithelial-mesenchymal transition (EMT), which were associated with unfavorable prognosis, increased chemoresistance, and heightened immunosuppression. Conclusions This study demonstrates that ferroptosis under hypoxic conditions significantly affects the modulation of the tumor immune microenvironment. The FHRS can independently predict prognosis in gastric cancer. Assessing the molecular status of ferroptosis-hypoxia in individual patients will help in selecting more suitable immunotherapy regimens by providing a better understanding of TME characteristics and predicting immunotherapeutic outcomes.
Collapse
Affiliation(s)
- Xiao Xu
- Department of Radiation Oncology, Qingdao People’s Hospital Group (Jiaozhou), Jiaozhou Central Hospital of Qingdao, Qingdao, China
| | - Liangling Fa
- Department of Pathology, Qingdao People’s Hospital Group (Jiaozhou), Jiaozhou Central Hospital of Qingdao, Qingdao, China
| | - Xiaoxiao Sun
- Department of Radiation Oncology, Qingdao People’s Hospital Group (Jiaozhou), Jiaozhou Central Hospital of Qingdao, Qingdao, China
| | - Fangfang Yang
- Cancer Precision Medical Center, Qingdao University, Qingdao, China
| | - Yongrui Liu
- Department of Oncology, Linyi Cancer Hospital, Linyi, China
| | - Jifu Song
- Department of Radiation Oncology, Qingdao People’s Hospital Group (Jiaozhou), Jiaozhou Central Hospital of Qingdao, Qingdao, China
| | - Yongli Zhao
- Department of Radiation Oncology, Qingdao People’s Hospital Group (Jiaozhou), Jiaozhou Central Hospital of Qingdao, Qingdao, China
| | - Jigang Dong
- Department of Radiation Oncology, Qingdao People’s Hospital Group (Jiaozhou), Jiaozhou Central Hospital of Qingdao, Qingdao, China
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| |
Collapse
|
22
|
Zhu L, Leng D, Guo Z, Zhao Y, Leung KT, Dai Y, Li J, Zhao Q. Self-catalyzed nitric oxide nanocomplexes induce ferroptosis for cancer immunotherapy. J Control Release 2025; 377:524-539. [PMID: 39580079 DOI: 10.1016/j.jconrel.2024.11.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 11/25/2024]
Abstract
Ferroptosis, triggered by membrane lipid peroxidation (LPO) and diminished antioxidants, can be induced by intracellular iron (II, Fe2+). However, the role of nitric oxide (NO) in causing Fe2+ overload for ferroptosis remains uncertain. This study reveals that NO can stimulate endogenous Fe2+ release by upregulating heme oxygenase 1 (HMOX1) expression. Here, ferritin heavy chain (FHC) siRNA and hyaluronic acid (HA)-modified Arg-stabilized zinc peroxide (AZOSH), a non-ferrous-based nanoagent, is synthesized to trigger ferroptosis by inducing intracellular Fe2+ overload. AZOSH, a self-catalyzed NO nanocomplex, effectively generates NO through a reaction of self-supplied Arginine (Arg) and hydrogen peroxide (H2O2), which promotes glutathione (GSH) consumption to downregulate glutathione peroxidase 4 (GPX4) expression and produces peroxynitrite (ONOO-) to enhance LPO. Meanwhile, NO promotes endo/lysosomal escape of siRNA by damaging membrane structures. Moreover, AZOSH significantly triggers Fe2+ overload through the synergistic effects of NO-activated HMOX1 expression and FHC siRNA-mediated ferritin sequestration. Additionally, the released Zn2+ from AZOSH induces oxidative stress by inhibiting mitochondrial function, further promoting ferroptosis. Consequently, AZOSH-mediated ferroptosis exhibits a strong cellular immunogenic response for T-cell activation and infiltration. Importantly, the integration of AZOSH with an anti-PD-1 antibody results in notable antitumor efficacy in vivo. Therefore, this study provides a novel concept of NO-induced ferroptosis, highlighting its role in enhancing PD-1-based immunotherapeutic efficacy.
Collapse
Affiliation(s)
- Lipeng Zhu
- School of Life Sciences, Central South University, Changsha 510006, China
| | - Dongliang Leng
- MoE Frontiers Science Center For Precision Oncology, Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, SAR 999078, China
| | - Ziang Guo
- MoE Frontiers Science Center For Precision Oncology, Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, SAR 999078, China
| | - Yuetao Zhao
- School of Life Sciences, Central South University, Changsha 510006, China
| | - Kam-Tong Leung
- Department of Paediatrics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Yeneng Dai
- MoE Frontiers Science Center For Precision Oncology, Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, SAR 999078, China.
| | - Junnan Li
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan Province, Department of Pharmacy, School of Medicine, Hunan Normal University, Changsha 410013, China.
| | - Qi Zhao
- MoE Frontiers Science Center For Precision Oncology, Cancer Centre, Faculty of Health Sciences, University of Macau, Macau, SAR 999078, China.
| |
Collapse
|
23
|
Su J, Li Y, Tan S, Cheng T, Luo Y, Zhang L. Pretreatment neutrophil-to-lymphocyte ratio is associated with immunotherapy efficacy in patients with advanced cancer: a systematic review and meta-analysis. Sci Rep 2025; 15:446. [PMID: 39747391 PMCID: PMC11695637 DOI: 10.1038/s41598-024-84890-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/27/2024] [Indexed: 01/04/2025] Open
Abstract
This study aimed to systematically investigate the value of the pre-treatment neutrophil-to-lymphocyte ratio (NLR) in prognosticating the outcome of patients with advanced cancer receiving immunotherapy. We searched Embase, PubMed, Web of Science, and Cochrane Library to identify studies about cancer patients with immunotherapy until November 29, 2024. Retrospective or prospective cohort studies with pretreatment NLR data were included. The odds ratio (OR) and 95% confidence interval (CI) were calculated to evaluate the predictive value of NLR in prognosis and immunotherapy efficacy. The random effect model was applied for meta-analysis and the risk of bias was assessed by Egger test and the Grading of Recommendations Assessment, Development and Evaluation (GRADE) method. A total of 129 articles involving 18780 cases were finally selected. Most cases were advanced cancers with the median follow-up period ranged 2-48.6 months. The high pretreatment NLR level was associated with the significantly reduced OS (HR (95%CI) = 2.26 (2.03, 2.53)), PFS (HR (95% CI) = 1.83 (1.69, 1.98)), ORR (OR (95%CI) = 0.53 (0.46, 0.61)) and DCR (OR (95% CI) = 0.36 (0.29, 0.43)) in patients with advanced cancer receiving immunotherapy. The quality of evidence was low, attributed to the serious risk of bias and incon¬sistency. An elevated NLR before immunotherapy was significantly associated with poor clinical outcomes in patients with advanced cancer.
Collapse
Affiliation(s)
- Jialin Su
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Tongzipo Rd 283#, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan, 411201, People's Republic of China
| | - Yuning Li
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Tongzipo Rd 283#, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan, 411201, People's Republic of China
| | - Shuhua Tan
- School of Life and Health Sciences, Hunan University of Science and Technology, Xiangtan, 411201, People's Republic of China
| | - Tianli Cheng
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Tongzipo Rd 283#, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China
| | - Yongzhong Luo
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Tongzipo Rd 283#, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China
| | - Lemeng Zhang
- Thoracic Medicine Department 1, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Tongzipo Rd 283#, Yuelu District, Changsha, 410013, Hunan Province, People's Republic of China.
| |
Collapse
|
24
|
Wang J, Hu X, Wang Y, A R, Li X, Sun Y, Guan Z, Li X, Wu Y, Wang J, Zhao F, Liu Y, Wang H, Yu H, Wang T, Zhu M, Li X, Zhang D, Chen W, Han Z, Sun X. Development and characterisation of [ 18F]TTDP, a novel T cell immunoglobulin and ITIM domain tracer, in humanised mice and non-human primates. Eur J Nucl Med Mol Imaging 2025; 52:416-426. [PMID: 39297961 DOI: 10.1007/s00259-024-06911-7] [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: 05/28/2024] [Accepted: 08/28/2024] [Indexed: 09/21/2024]
Abstract
PURPOSE The T cell immunoglobulin and ITIM domain (TIGIT) blockade immunotherapy response is directly associated with individual differences of TIGIT expression on tumour-infiltrating lymphocytes (TILs) in tumour immune microenvironment (TIME) of non-small cell lung cancer (NSCLC). Here, we developed a TIGIT-targeted PET tracer to evaluate its feasibility in predicting immunotherapy efficacy, aiming to manage NSCLC patients accurately. METHODS We synthesised a 18F-labeled TIGIT-targeted D-peptide, [18F]TTDP, and investigated the specificity of [18F]TTDP both to murine TIGIT and human TIGIT by a series of in vitro and in vivo assays. [18F]TTDP PET imaging was performed in humanised immune system (HIS) mice models bearing NSCLC patient-derived xenografts (PDXs) to evaluate the predictive value of FDA-approved combination immunotherapy of atezolizumab plus tiragolumab. Lastly, rhesus macaque was applied for [18F] TTDP PET to explore the tracer's in vivo distribution and translational potential in non-human primates. RESULTS [18F]TTDP showed high specificity for both murine TIGIT and human TIGIT in vitro and in vivo. The HIS NSCLC PDX platform was successfully established for [18F]TTDP PET imaging, and tumour uptake of [18F]TTDP was significantly correlated with the TIGIT expression of TILs in the TIME. [18F]TTDP PET imaging, in predicting treatment response to the combination immunotherapy in NSCLC HIS-PDX models, showed a sensitivity of 83.33% and a specificity of 100%. In addition, [18F]TTDP PET also showed cross-species consistency of the tracer biodistribution between non-human primate and murine animals, and no adverse events were observed. CONCLUSION The combined implementation of the [18F]TTDP and HIS-PDX model creates a state-of-the-art preclinical platform that will impact the identification and validation of TIGIT-targeted PET image-guided diagnosis, treatment response prediction, beneficial patient screening, novel immunotherapies, and ultimately the outcome of NSCLC patients. We first provided in vivo biodistribution of [18F]TTDP PET imaging in rhesus macaque, indicating its excellent translational potential in the clinic.
Collapse
Affiliation(s)
- Jing Wang
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinxin Hu
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yueqi Wang
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Rong A
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaoqian Li
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ying Sun
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhengqi Guan
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaona Li
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yongyi Wu
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jiannan Wang
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Fangyu Zhao
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yang Liu
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongbin Wang
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hong Yu
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Tianyi Wang
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Mengyuan Zhu
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinyu Li
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Duoyi Zhang
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China
| | - Wei Chen
- Department of Nuclear Medicine & Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhaoguo Han
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China.
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China.
| | - Xilin Sun
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Harbin Medical University, Xiangan N Street, Songbei District, Harbin, 150028, Heilongjiang, China.
- NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy, Molecular Imaging Research Center (MIRC) of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
25
|
Forschner A, Nanz L, Maczey-Leber Y, Amaral T, Flatz L, Leiter U. Response and outcome of patients with melanoma skin metastases and immune checkpoint inhibition. Int J Cancer 2025; 156:145-153. [PMID: 39032035 DOI: 10.1002/ijc.35103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/22/2024]
Abstract
It is known, that different metastatic organ systems respond differently to immune checkpoint inhibitors (ICIs). In this study, we aimed to investigate the extent to which skin/subcutaneous metastases respond to ICI or targeted therapies (TTs) and whether the response rate differs from that of distant metastases in the same patient. Patients with melanoma diagnosed between January 2021 and September 2023 with at least one skin/subcutaneous metastasis who had received therapy with ICI or TT in an advanced setting were included in the analysis. Best overall response (BOR) was classified according to the revised response evaluation criteria in solid tumors (RECIST). The BOR of skin metastases and visceral metastases to ICI and TT was compared using the chi-square test. Skin metastases treated with ICI a first-line setting showed an overall response rate (ORR) of 44.1%. In contrast, visceral metastases had a higher ORR of 51.1%. However, the difference was not statistically significant (p = .77). Regarding TT, the ORR for skin metastases was 57.1%, compared to 38.5% for visceral metastases (p = .59). Interestingly, the ORR for skin/subcutaneous metastases was notably lower with ICI compared to visceral metastases, in contrast to patients who underwent TT. Skin metastases showed a poorer response to ICI than visceral metastases. Therefore, careful monitoring is recommended to detect non-response early in patients with skin metastases as skin metastases may have a worse response than TT. A larger cohort is needed for a comprehensive analysis and confirmation of our results.
Collapse
Affiliation(s)
- Andrea Forschner
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Lena Nanz
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Yves Maczey-Leber
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Teresa Amaral
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Lukas Flatz
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tübingen, Tübingen, Germany
| | - Ulrike Leiter
- Center for Dermatooncology, Department of Dermatology, Eberhard Karls University of Tübingen, Tübingen, Germany
| |
Collapse
|
26
|
Huang S, Qin X, Fu S, Hu J, Jiang Z, Hu M, Zhang B, Liu J, Chen Y, Wang M, Liu X, Chen Z, Wang L. STAMBPL1/TRIM21 Balances AXL Stability Impacting Mesenchymal Phenotype and Immune Response in KIRC. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405083. [PMID: 39527690 PMCID: PMC11714167 DOI: 10.1002/advs.202405083] [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: 05/10/2024] [Revised: 10/09/2024] [Indexed: 11/16/2024]
Abstract
Kidney renal clear cell carcinoma (KIRC) is recognized as an immunogenic tumor, and immunotherapy is incorporated into its treatment landscape for decades. The acquisition of a tumor mesenchymal phenotype through epithelial-to-mesenchymal transition (EMT) is associated with immune evasion and can contribute to immunotherapy resistance. Here, the involvement of STAM Binding Protein Like 1 (STAMBPL1) is reported in the development of mesenchymal and immune evasion phenotypes in KIRC cells. Mechanistically, STAMBPL1 elevated protein abundance and surface accumulation of TAM Receptor AXL through diminishing the TRIM21-mediated K63-linked ubiquitination and subsequent lysosomal degradation of AXL, thereby enhancing the expression of mesenchymal genes while suppressing chemokines CXCL9/10 and HLA/B/C. In addition, STAMBPL1 enhanced PD-L1 transcription via facilitating nuclear translocation of p65, and knockdown (KD) of STAMBPL1 augmented antitumor effects of PD-1 blockade. Furthermore, STAMBPL1 silencing and the tyrosine kinase inhibitor (TKI) sunitinib also exhibited a synergistic effect on the suppression of KIRC. Collectively, targeting the STAMBPL1/TRIM21/AXL axis can decrease mesenchymal phenotype and potentiate anti-tumor efficacy of cancer therapy.
Collapse
Affiliation(s)
- Shiyu Huang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Xuke Qin
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Shujie Fu
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Juncheng Hu
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Zhengyu Jiang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Min Hu
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Banghua Zhang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Hubei Key Laboratory of Digestive System DiseaseWuhan430060China
| | - Jiachen Liu
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Central LaboratoryRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Yujie Chen
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Minghui Wang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Xiuheng Liu
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Zhiyuan Chen
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| | - Lei Wang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubei430060China
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubei430060China
| |
Collapse
|
27
|
Tang C, Dong Z, Yan S, Liu B, Wang Z, Cheng L, Liu F, Sun H, Du Y, Pan L, Zhou Y, Jin Z, Zhao L, Wu N, Chang L, Xu X. Microdroplet-enhanced chip platform for high-throughput immunotherapy marker screening from extracellular vesicle RNAs and membrane proteins. Biosens Bioelectron 2025; 267:116748. [PMID: 39276441 DOI: 10.1016/j.bios.2024.116748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/31/2024] [Accepted: 09/03/2024] [Indexed: 09/17/2024]
Abstract
Extracellular vesicles (EVs) are considered as promising candidates for predicting patients who respond to immunotherapy. Nevertheless, simultaneous detection of multiple EVs markers still presents significant technical challenges. In this work, we developed a high-throughput microdroplet-enhanced chip (MEC) platform, which utilizes thousands of individual microchambers (∼pL) as reactors, accelerating the detection efficiency of the CRISPR/Cas systems and increasing the sensitivity by up to 100-fold (aM level). Ten biomarkers (including 5 RNAs and 5 proteins) from patients' EVs are successfully detected on one chip, and the comprehensive markers show increased accuracy (AUC 0.911) than the individual marker for the efficacy prediction of immunotherapy. This platform provides a high-throughput yet sensitive strategy for screening immunotherapy markers in clinical.
Collapse
Affiliation(s)
- Chuanhao Tang
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China; Department of Medical Oncology, Peking University International Hospital, Beijing, 102206, China
| | - Zaizai Dong
- School of Engineering Medicine, Beihang University, Beijing, 100191, China; Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China.
| | - Shi Yan
- State Key Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Bing Liu
- State Key Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Zhiying Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Long Cheng
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, China
| | - Feng Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Hong Sun
- Translational Medicine Center, Beijing Chest Hospital, Capital Medical University, Beijing Key Laboratory in Drug Resistant Tuberculosis Research, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Yimeng Du
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Lu Pan
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Yuhao Zhou
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Zhiyuan Jin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Libo Zhao
- Echo Biotech Co., Ltd, Beijing, 102206, China
| | - Nan Wu
- State Key Laboratory of Molecular Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Thoracic Surgery II, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| | - Lingqian Chang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China; School of Biomedical Engineering, Anhui Medical University, Hefei, 230032, China.
| | - Xiaojie Xu
- Department of Genetic Engineering, Beijing Institute of Biotechnology, Beijing, 100071, China.
| |
Collapse
|
28
|
Wei J, Li W, Zhang P, Guo F, Liu M. Current trends in sensitizing immune checkpoint inhibitors for cancer treatment. Mol Cancer 2024; 23:279. [PMID: 39725966 DOI: 10.1186/s12943-024-02179-5] [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: 09/05/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically transformed the treatment landscape for various malignancies, achieving notable clinical outcomes across a wide range of indications. Despite these advances, resistance to immune checkpoint blockade (ICB) remains a critical clinical challenge, characterized by variable response rates and non-durable benefits. However, growing research into the complex intrinsic and extrinsic characteristics of tumors has advanced our understanding of the mechanisms behind ICI resistance, potentially improving treatment outcomes. Additionally, robust predictive biomarkers are crucial for optimizing patient selection and maximizing the efficacy of ICBs. Recent studies have emphasized that multiple rational combination strategies can overcome immune checkpoint resistance and enhance susceptibility to ICIs. These findings not only deepen our understanding of tumor biology but also reveal the unique mechanisms of action of sensitizing agents, extending clinical benefits in cancer immunotherapy. In this review, we will explore the underlying biology of ICIs, discuss the significance of the tumor immune microenvironment (TIME) and clinical predictive biomarkers, analyze the current mechanisms of resistance, and outline alternative combination strategies to enhance the effectiveness of ICIs, including personalized strategies for sensitizing tumors to ICIs.
Collapse
Grants
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- ZYJC21043 the 1.3.5 Project for Disciplines of Excellence, West China Hospital, Sichuan University
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
- 2023YFS0111 Social Development Science and Technology Project of Sichuan Province on Science and Technology
Collapse
Affiliation(s)
- Jing Wei
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Wenke Li
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Pengfei Zhang
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China
| | - Fukun Guo
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Ming Liu
- Department of Medical Oncology, Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, 610041, China.
| |
Collapse
|
29
|
Zhang H, Lu B, Lu X, Saeed A, Chen L. Current transcriptome database and biomarker discovery for immunotherapy by immune checkpoint blockade. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.09.627506. [PMID: 39713380 PMCID: PMC11661151 DOI: 10.1101/2024.12.09.627506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Immune checkpoint blockade (ICB) has revolutionized the current immuno-oncology and significantly improved clinical outcome for cancer treatment. Despite the advancement in clinics, only a small subset of patients derives immune response to the ICB therapy. Therefore, a robust predictive biomarker that identifies potential candidate becomes increasingly crucial in delivering this technology to the public. In this review, we first discuss the biomarkers that focus on tumor genome, tumor microenvironment and tumor-host interaction. Then, we compare existing databases for biomarker discovery for ICB response. We also present IOhub - an interactive web portal that incorporates 36 bulk and 10 single-cell transcriptome datasets for benchmark analysis of the current biomarkers. Finally, we highlight the trending interest in antibody drug conjugate and combination treatment and their use in precision immuno-oncology.
Collapse
|
30
|
Nu er lan STE, Yu B, Yang Y, Shen Y, Xu B, Zhan Y, Liu C. Discover Mutational Differences Between Lung Adenocarcinoma and Lung Squamous Cell Carcinoma and Search for More Effective Biomarkers for Immunotherapy. Cancer Manag Res 2024; 16:1759-1773. [PMID: 39678041 PMCID: PMC11645897 DOI: 10.2147/cmar.s491661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/01/2024] [Indexed: 12/17/2024] Open
Abstract
Purpose Lung cancer is a severe malignant tumor. This study aims to more comprehensively characterize lung cancer patients and identify combination markers for immunotherapy. Patients and Methods We gathered data from 166 lung cancer patients at the Cancer Hospital Affiliated with Xinjiang Medical University. The collected samples underwent NGS sequencing using a panel of 616 genes associated with cancer. Subsequently, data analysis was conducted to identify markers that are more suitable for lung cancer immunotherapy. Results In this study, the most common variant genes in LUAD were TP53, EGFR, MST1, KMT2C, RBM10, LRP1B. Meanwhile, the highest mutation frequency genes in LUSC samples were TP53, KMT2D, LRP1B, FAT1, MST1, KMT2C. Mutation frequencies, tumor mutation burden (TMB), PD-L1 expression, and mutant-allele tumor heterogeneity (MATH) values differed between LUAD and LUSC, with LUSC exhibiting higher values than LUAD. Irrespective of LUAD or LUSC, patients with TMB≥10 demonstrated better immunotherapy efficacy compared to patients with TMB<10. Similarly, when PD-L1≥50%, whether in LUAD or LUSC, the immunotherapy effect was superior to that of patients with PD-L1<50%. Combining TMB≥10 and PD-L1≥50% as immunotherapy markers, in both LUAD and LUSC, resulted in a very favorable immunotherapy effect, with the overall response rate (ORR) reaching 100%. Conclusion We observed distinct mutation patterns and clinical factors between LUAD and LUSC, and noted that patients with TMB≥10 and PD-L1≥50% exhibited enhanced immunotherapy effects. Combining TMB≥10 and PD-L1≥50% proved to be a more effective predictor of immunotherapy efficacy.
Collapse
Affiliation(s)
- Sai te er Nu er lan
- Department of Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, People’s Republic of China
| | - Bo Yu
- Beijing USCI Medical Laboratory, Beijing, 100195, People’s Republic of China
| | - Yan Yang
- Department of Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, People’s Republic of China
| | - Yanli Shen
- Department of Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, People’s Republic of China
| | - Bing Xu
- Beijing USCI Medical Laboratory, Beijing, 100195, People’s Republic of China
| | - Yiyi Zhan
- Department of Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, People’s Republic of China
| | - Chunling Liu
- Department of Pulmonary Medicine, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830001, People’s Republic of China
| |
Collapse
|
31
|
Zhu J, Zhang W, Wang Z, Wang Y, Li J, Wang Y, Xu F, Chen Y. Mass-tagged self-assembled nanoprobe reveals the transport of PD-L1 from cancer cells to tumor-educated platelets. Anal Chim Acta 2024; 1331:343312. [PMID: 39532409 DOI: 10.1016/j.aca.2024.343312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/23/2024] [Accepted: 10/06/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND The expression level of immune checkpoint proteins detected by tissue biopsy is currently used as a predictive biomarker for immune checkpoint blockade (ICB) therapy. However, tissue biopsy is susceptible to invasive sample collection procedures, significant sampling heterogeneity, and the difficulty of repeated sampling. Therefore, liquid biopsy of blood samples is becoming an alternative choice for immune checkpoint protein detection. Among various vesicles in blood, platelets can obtain cancer information to form a specific group called tumor-educated platelets (TEPs). The platelet-derived proteins in TEPs may have a predictive potential in ICB therapy. RESULTS In this study, a photo-cleavable mass-tagged self-assembled (SAMT) nanoprobe with signal amplification was developed for the quantitative detection of PD-L1. The SAMT probe was assembled by photo-cleavable mass tags, PD-L1 aptamer, and amphiphilic polymer. After binding with PD-L1 on the platelet, the probe can release mass tags with UV light exposure. The amount of the mass tag, representing that of PD-L1, was subsequently determined by mass spectrometry. The assay sensitivity can be greatly improved by up to four orders of magnitude, achieving a detection limit of 10 fM. This assay was subsequently applied to cancer cells and platelet samples from non-small cell lung cancer (NSCLC) patients. The patients with higher tumor stages, higher degrees of lymph node invasion, and better ICB response had higher PD-L1 levels on platelets. Further investigation revealed that PD-L1 on the platelets was transported from cancer cells, providing evidence for the existence of TEPs. SIGNIFICANCE The SAMT probe can amplify the signal of the target molecule into that of multiple mass tags, achieving ultrasensitive ICB protein quantitative detection in platelets. Moreover, the employed SAMT assay not only revealed PD-L1 transport from cancer cells to platelets but also confirmed the presence of TEPs.
Collapse
Affiliation(s)
- Jianhua Zhu
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China; The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, China
| | - Wenjun Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Zhongcheng Wang
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Yan Wang
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Jiapu Li
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Yunjing Wang
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Feifei Xu
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Yun Chen
- School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China; State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing, 211166, China; Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Nanjing, 211166, China.
| |
Collapse
|
32
|
Wang Y, Ju X, Hua R, Chen J, Dai X, Liu L, Wang G, Bai Y, Hu H, Li X. Deep learning analysis of histopathological images predicts immunotherapy prognosis and reveals tumour microenvironment features in non-small cell lung cancer. Br J Cancer 2024; 131:1833-1845. [PMID: 39455880 PMCID: PMC11589918 DOI: 10.1038/s41416-024-02856-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is one of the leading causes of cancer mortality worldwide. Immune checkpoint inhibitors (ICIs) have emerged as a crucial treatment option for patients with advanced NSCLC. However, only a subset of patients experience clinical benefit from ICIs. Therefore, identifying biomarkers that can predict response to ICIs is imperative for optimising patient selection. METHODS Hematoxylin and eosin (H&E) images of NSCLC patients were obtained from the local cohort (n = 106) and The Cancer Genome Atlas (TCGA) (n = 899). We developed an ICI-related pathological prognostic signature (ir-PPS) based on H&E stained histopathology images to predict prognosis in NSCLC patients treated with ICIs using deep learning. To accomplish this, we employed a modified ResNet model (ResNet18-PG), a widely-used deep learning architecture well-known for its effectiveness in handling complex image recognition tasks. Our modifications include a progressive growing strategy to improve the stability of model training and the use of the AdamW optimiser, which enhances the optimisation process by adjusting the learning rate based on training dynamics. RESULTS The deep learning model, ResNet18-PG, achieved an area under the receiver operating characteristic curve (AUC) of 0.918 and a recall of 0.995 on the local cohort. The ir-PPS effectively risk-stratified NSCLC patients. Patients in the low-risk group (n = 40) had significantly improved progression-free survival (PFS) after ICI treatment compared to those in the high-risk group (n = 66, log-rank P = 0.004, hazard ratio (HR) = 3.65, 95%CI: 1.75-7.60). The ir-PPS demonstrated good discriminatory power for predicting 6-month PFS (AUC = 0.750), 12-month PFS (AUC = 0.677), and 18-month PFS (AUC = 0.662). The low-risk group exhibited increased expression of immune checkpoint molecules, cytotoxicity-related genes, an elevated abundance of tumour-infiltrating lymphocytes, and enhanced activity in immune stimulatory pathways. CONCLUSIONS The ir-PPS signature derived from H&E images using deep learning could predict ICIs prognosis in NSCLC patients. The ir-PPS provides a novel imaging biomarker that may help select optimal candidates for ICIs therapy in NSCLC.
Collapse
Affiliation(s)
- Youyu Wang
- Department of Thoracic Surgery, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Institute of Thoracic Oncology and Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xueming Ju
- Department of Ultrasound, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Rong Hua
- Department of Respiratory and Critical Care Medicine, Sixth People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Ji Chen
- Department of Medical Oncology, The Seventh People's Hospital of Chengdu, Chengdu, Sichuan, China
| | - Xiaoqin Dai
- Department of Traditional Chinese Medicine, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Lunxu Liu
- Institute of Thoracic Oncology and Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Guifang Wang
- Department of Respiratory and Critical Care Medicine, Sixth People's Hospital of Chengdu, Chengdu, Sichuan, China.
| | - Yifeng Bai
- Department of Oncology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Honglin Hu
- Department of Oncology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Xiaohua Li
- Department of Respiratory and Critical Care Medicine, Sixth People's Hospital of Chengdu, Chengdu, Sichuan, China.
| |
Collapse
|
33
|
Ma X, Mao M, Liu Z, Liang C, He J, Qu Y, Xu L, Cheng R, Zhuang W, Lei Y, Nie W, Yuan L, Pang DW, Xie HY. AND-Gate Logic Förster Resonance Energy Transfer/Magnetic Resonance Tuning Nanoprobe for Programmable Antitumor Immunity Imaging. J Am Chem Soc 2024; 146:31873-31884. [PMID: 39504515 DOI: 10.1021/jacs.4c11072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
Simultaneous detection of different biomarkers related to the spatiotemporally dynamic immune events is of particular importance for the accurate evaluation of antitumor immune effects. Here, we have developed an AND-gate logic dual resonance energy transfer nanoprobe (named DRET) for dynamic monitoring of programmed CD8+ T cell activation and tumor cell apoptosis. Immunotherapy-induced granzyme B secretion from CD8+ T cells and the subsequent caspase-3 release from apoptotic tumor cells individually activate one of the tiers of the "AND-gate" logic DRET. The resulting fluorescence recovery and magnetic resonance T1 enhancement can be used for precise immunomodulatory drug screening, early efficacy prediction, and immune stratification. Particularly, not only "Responders" can be distinguished from "Non-responders", but also "Acquired resistance" can be identified from "Maintain responders", providing a novel approach to put forward the accurate evaluation of antitumor immunity.
Collapse
Affiliation(s)
- Xianbin Ma
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Mingchuan Mao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Zhenya Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Chao Liang
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Jiaqi He
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Yun Qu
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Luzheng Xu
- Medical and Health Analysis Center, Peking University, Beijing 100191, P. R. China
| | - Ran Cheng
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Wanru Zhuang
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Yao Lei
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| | - Weidong Nie
- School of Life Science, Beijing Institute of Technology, Beijing 100081, P. R. China
| | - Lan Yuan
- Medical and Health Analysis Center, Peking University, Beijing 100191, P. R. China
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Nankai University, Tianjin 300071, P. R. China
| | - Hai-Yan Xie
- State Key Laboratory of Natural and Biomimetic Drugs, Chemical Biology Center, School of Pharmaceutical Sciences, Peking University, Beijing 100191, P. R. China
| |
Collapse
|
34
|
Nkune NW, Abrahamse H. Possible integration of artificial intelligence with photodynamic therapy and diagnosis: A review. J Drug Deliv Sci Technol 2024; 101:106210. [DOI: 10.1016/j.jddst.2024.106210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
35
|
Liu Y, Xu C, Zhang L, Xu G, Yang Z, Xiang L, Jiao K, Chen Z, Zhang X, Liu Y. Syndecan-1 inhibition promotes antitumor immune response and facilitates the efficacy of anti-PD1 checkpoint immunotherapy. SCIENCE ADVANCES 2024; 10:eadi7764. [PMID: 39259785 PMCID: PMC11389782 DOI: 10.1126/sciadv.adi7764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 08/02/2024] [Indexed: 09/13/2024]
Abstract
Tumor cell-originated events prevent efficient antitumor immune response and limit the application of anti-PD1 checkpoint immunotherapy. We show that syndecan-1 (SDC1) has a critical role in the regulation of T cell-mediated control of tumor growth. SDC1 inhibition increases the permeation of CD8+ T cells into tumors and triggers CD8+ T cell-mediated control of tumor growth, accompanied by increased proportions of progenitor-exhausted and effector-like CD8+ T cells. SDC1 deficiency alters multiple signaling events in tumor cells, including enhanced IFN-γ-STAT1 signaling, and augments antigen presentation and sensitivity to T cell-mediated cytotoxicity. Combinatory inhibition of SDC1 markedly potentiates the therapeutic effects of anti-PD1 in inhibiting tumor growth. Consistently, the findings are supported by the data from human tumors showing that SDC1 expression negatively correlates with T cell presence in tumor tissues and the response to immune checkpoint blockade therapy. Our findings suggest that SDC1 inhibits antitumor immunity, and that targeting SDC1 may promote anti-PD1 response for cancer treatment.
Collapse
Affiliation(s)
- Yun Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
| | - Chen Xu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
| | - Li Zhang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
| | - Guiqin Xu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
| | - Zhaojuan Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
| | - Lvzhu Xiang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
| | - Kun Jiao
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
| | - Zehong Chen
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
| | - Xiaoren Zhang
- Affiliated Cancer Hospital and Institute, Guangzhou Medical University, Guangzhou, China
| | - Yongzhong Liu
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200032, China
| |
Collapse
|
36
|
Zhao C, Huang Y, Zhang H, Liu H. CD24 affects the immunosuppressive effect of tumor-infiltrating cells and tumor resistance in a variety of cancers. Discov Oncol 2024; 15:399. [PMID: 39222166 PMCID: PMC11369128 DOI: 10.1007/s12672-024-01284-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Cluster of differentiation 24 (CD24) is a highly glycosylated glycosylphosphatidylinositol (GPI)-anchored surface protein, expressed in various tumor cells, as a "don't eat me" signaling molecule in tumor immune. This study aimed to investigate the potential features of CD24 in pan-cancer. METHODS The correlations between 22 immune cells and CD24 expression were using TIMER analysis. R package "ESTIMATE" was used to predict the proportion of immune and stromal cells in pan-cancer. Spearman's correlation analysis was performed to evaluate the relationships between CD24 expression and immune checkpoints, chemokines, mismatch repair, tumor mutation burden and microsatellite instability, and qPCR and western blot were conducted to assess CD24 expression levels in liver hepatocellular carcinoma (LIHC). In addition, loss of function was performed for the biological evaluation of CD24 in LIHC. RESULTS CD24 expression was positively correlated with myeloid cells, including neutrophils and myeloid-derived suppressor cells, in various tumors, such as BLCA, HNSC-HPV, HNSC, KICH, KIRC, KIRP, TGCT, THCA, THYM, and UCEC. In contrast, anti-tumor NK cells and NKT cells showed a negative association with CD24 expression in BRCA-Her2, ESCA, HNSC-HPV, KIRC, THCA, and THYM. The top three tumors with the highest correlation between CD24 and ImmuneScore were TGCT, THCA, and SKCM. Functional enrichment analysis revealed CD24 expression was negatively associated with various immune-related pathways. Immune checkpoints and chemokines also exhibited inverse correlations with CD24 in CESC, CHOL, COAD, ESCA, READ, TGCT, and THCA. Additionally, CD24 was overexpressed in most tumors, with high CD24 expression in BRCA, LIHC, and CESC correlating with poor prognosis. The TIDE database indicated tumors with high CD24 expression, particularly melanoma, were less responsive to PD1/PD-L1 immunotherapy. Finally, CD24 knockdown resulted in impaired proliferation and cell cycle progression in LIHC. CONCLUSION CD24 participates in regulation of immune infiltration, influences patient prognosis and serves as a potential tumor marker.
Collapse
Affiliation(s)
- Chunmei Zhao
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Ying Huang
- Department of Clinical Laboratory, Qidong People's Hospital/Affiliated Qidong Hospital of Nantong University, Nantong, Jiangsu, China
| | - Haotian Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu Province, China
| | - Huimin Liu
- Department of Clinical Laboratory, Affiliated Nantong Hospital 3 of Nantong University, Nantong Third People's Hospital, Nantong, Jiangsu, China.
| |
Collapse
|
37
|
Consalvo F, De Simone M, Scarpa A, Acerra A, Salzano FA, Fineschi V, Santurro A. Challenges and Complications in the Management of Advanced Oropharyngeal Carcinoma: Role of Post-Mortem Diagnosis and Future Perspectives. J Clin Med 2024; 13:5198. [PMID: 39274413 PMCID: PMC11396599 DOI: 10.3390/jcm13175198] [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: 08/09/2024] [Revised: 08/30/2024] [Accepted: 08/31/2024] [Indexed: 09/16/2024] Open
Abstract
Oropharyngeal squamous-cell carcinoma (OPSCC) poses significant challenges in diagnosis, treatment, and management and has important medico-legal and forensic implications. In particular, the management of OPSCC and its treatment-related complications can often be challenging. In cases with advanced OPSCC, a loco-regional extension of the tumor can contribute to the destruction of oral cavity tissues, while the radiotherapy treatment can induce profound changes in tissue morphology and structure. These changes, which resemble tumor neoplasms and endovascular effects, are related to a higher risk of fatal bleeding, as reported in the case study illustrated, in which a hemorrhage occurred from a lingual artery, originating from an ulcerative, necrotic, hemorrhagic lesion on the tongue. Bleeding complications in OPSCC and prolonged radiotherapy are associated with high mortality and require comprehensive management strategies to improve survival and quality of life. Autopsy investigations, contributing to the definition of post-mortem diagnosis, can provide valuable insights into the pathogenetic mechanisms underlying bleeding and guide therapeutic decisions and preventive measures. The integration of autopsy and histopathological investigation into clinical practice should be considered as a necessary support to optimize the management of complications in advanced OPSCC patients, emphasizing the importance of a patient-centered approach and continued research.
Collapse
Affiliation(s)
- Francesca Consalvo
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Salerno, Italy; (F.C.); (A.A.); (F.A.S.); (A.S.)
- BrainLab s.r.l., Mercato San Severino, 84085 Salerno, Italy
- Unit of Legal Medicine, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84081 Salerno, Italy
| | - Matteo De Simone
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Salerno, Italy; (F.C.); (A.A.); (F.A.S.); (A.S.)
- BrainLab s.r.l., Mercato San Severino, 84085 Salerno, Italy
| | - Alfonso Scarpa
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Salerno, Italy; (F.C.); (A.A.); (F.A.S.); (A.S.)
| | - Alfonso Acerra
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Salerno, Italy; (F.C.); (A.A.); (F.A.S.); (A.S.)
- Unit of Otolaryngology, University Hospital “San Giovanni di Dio e Ruggi, D’Aragona”, 84081 Salerno, Italy
| | - Francesco Antonio Salzano
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Salerno, Italy; (F.C.); (A.A.); (F.A.S.); (A.S.)
- Unit of Otolaryngology, University Hospital “San Giovanni di Dio e Ruggi, D’Aragona”, 84081 Salerno, Italy
| | - Vittorio Fineschi
- Department of Anatomical, Histological, Forensic and Orthopaedic Sciences, Sapienza University of Rome, 00161 Rome, Italy;
| | - Alessandro Santurro
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Salerno, Italy; (F.C.); (A.A.); (F.A.S.); (A.S.)
- Unit of Legal Medicine, University Hospital “San Giovanni di Dio e Ruggi D’Aragona”, 84081 Salerno, Italy
| |
Collapse
|
38
|
Patel SP, Guadarrama E, Chae YK, Dennis MJ, Powers BC, Liao CY, Ferri WA, George TJ, Sharon E, Ryan CW, Othus M, Lopez G, Blanke CD, Kurzrock R. SWOG 1609 cohort 48: anti-CTLA-4 and anti-PD-1 for advanced gallbladder cancer. Cancer 2024; 130:2918-2927. [PMID: 38358334 PMCID: PMC11309904 DOI: 10.1002/cncr.35243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/28/2023] [Accepted: 01/04/2024] [Indexed: 02/16/2024]
Abstract
INTRODUCTION Most patients with advanced gallbladder cancer are treated with multiagent chemotherapy. Immune checkpoint inhibitors offer the possibility of a durable response with less toxicity. This prospective, multicenter, open-label study was designed to evaluate the anticancer activity of nivolumab plus ipilimumab in patients with advanced gallbladder cancer. METHODS Nineteen patients with advanced gallbladder cancer refractory to ≥1 previous therapy received nivolumab 240 mg intravenously every 2 weeks and ipilimumab 1 mg/kg intravenously every 6 weeks until disease progression or unacceptable toxicity. The primary end point was confirmed radiographic overall response rate (ORR) (complete response [CR] + partial response [PR] confirmed on subsequent scan); secondary end points included unconfirmed overall response, clinical benefit rate (confirmed and unconfirmed responses + stable disease >6 months), progression-free survival, overall survival, and toxicity. RESULTS The confirmed ORR was 16% (CR, n = 1 [5%]; PR, n = 2 [11%]); all were microsatellite stable, and the confirmed CR had undetectable programmed death-ligand 1 by immunohistochemistry. The unconfirmed ORR and clinical benefit rates were both 32%. The median duration of response was 14.8 months (range, 4-35.1+ months). The 6-month progression-free survival was 26% (95% CI, 12-55). The median overall survival was 7.0 months (95% CI, 3.9-19.1). The most common toxicities were fatigue (32%), anemia (26%), and anorexia (26%). Aspartate aminotransferase elevation was the most common grade 3/4 toxicity (11%). There was 1 possibly related death (sepsis with attendant hepatic failure). CONCLUSIONS Ipilimumab plus nivolumab was well tolerated and showed modest efficacy with durable responses in previously treated patients with advanced gallbladder cancer. CLINICAL TRIAL REGISTRATION NCT02834013 (ClincialTrials.gov). PLAIN LANGUAGE SUMMARY This prospective study assessed the efficacy and safety of nivolumab plus ipilimumab in 19 patients with advanced gallbladder cancer refractory to previous therapy. The combination demonstrated modest efficacy with a 16% confirmed overall response rate, durable responses, and manageable toxicities, suggesting potential benefits for this challenging patient population.
Collapse
Affiliation(s)
- Sandip P. Patel
- Division of Medical Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | | | - Young Kwang Chae
- Division of Medical Oncology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Michael J. Dennis
- Division of Medical Oncology, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Benjamin C. Powers
- Division of Hematology/Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Chih-Yi Liao
- Division of Hematology/Oncology, University of Chicago Comprehensive Cancer Center, Chicago, IL, USA
| | - William A. Ferri
- Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Thomas J. George
- Division of Hematology/Oncology, University of Florida Health Cancer Center, Gainesville, FL, USA
| | - Elad Sharon
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, Cancer Therapy Evaluation Program, Bethesda, MD, USA
| | - Christopher W. Ryan
- Division of Hematology and Oncology, Oregon Health and Science University Knight Cancer Institute, Portland, OR, USA
| | - Megan Othus
- SWOG Statistics and Data Management Center/Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Gabby Lopez
- SWOG Statistics and Data Management Center/Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Charles D. Blanke
- SWOG Group Chair’s Office, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Razelle Kurzrock
- Division of Medical Oncology, Medical College of Wisconsin Froedtert Cancer Center, Milwaukee, WI, USA
| |
Collapse
|
39
|
Yang Y, Chen XQ, Jia YX, Ma J, Xu D, Xiang ZL. Circ-0044539 promotes lymph node metastasis of hepatocellular carcinoma through exosomal-miR-29a-3p. Cell Death Dis 2024; 15:630. [PMID: 39191749 DOI: 10.1038/s41419-024-07004-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 08/06/2024] [Accepted: 08/15/2024] [Indexed: 08/29/2024]
Abstract
Lymph node metastasis (LNM) is a common invasive feature of hepatocellular carcinoma (HCC) associated with poor clinical outcomes. Through microarray profiling and bioinformatic analyses, we identified the circ-0044539-miR-29a-3p-VEGFA axis as a potential key factor in the progression of HCC LNM. In HCC cells and nude mice, circ-0044539 downregulation or miR-29a-3p upregulation was associated with small tumor size, PI3K-AKT-mTOR pathway inactivation, and downregulation of the key LNM factors (HIF-1α and CXCR4). Furthermore, circ-0044539 was also responsible for exosomal miR-29a-3p secretion. Exosomal miR-29a-3p was then observed to migrate to the LNs and downregulate High-mobility group box transcription factor 1 (Hbp1) in Polymorphonuclear Myeloid-derived suppressor cells (PMN-MDSCs), inducing the formation of a microenvironment suitable for tumor colonization. Overall, circ-0044539 promotes HCC cell LNM abilities and induces an immune-suppressive environment in LNs through exosomes, highlighting its potential as a target for HCC LNM and HCC immunotherapy.
Collapse
Affiliation(s)
- Yi Yang
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Xue-Qin Chen
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Ya-Xun Jia
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Jie Ma
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Di Xu
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zuo-Lin Xiang
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
- Department of Radiation Oncology, Shanghai East Hospital Ji'an hospital, Ji'an City, Jiangxi Province, 343000, China.
| |
Collapse
|
40
|
Shu Z, Dwivedi B, Switchenko JM, Yu DS, Deng X. PD-L1 deglycosylation promotes its nuclear translocation and accelerates DNA double-strand-break repair in cancer. Nat Commun 2024; 15:6830. [PMID: 39122729 PMCID: PMC11316045 DOI: 10.1038/s41467-024-51242-8] [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: 08/30/2022] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Resistance to radiotherapy is a major barrier during cancer treatment. Here using genome-scale CRISPR/Cas9 screening, we identify CD274 gene, which encodes PD-L1, to confer lung cancer cell resistance to ionizing radiation (IR). Depletion of endogenous PD-L1 delays the repair of IR-induced DNA double-strand breaks (DSBs) and PD-L1 loss downregulates non-homologous end joining (NHEJ) while overexpression of PD-L1 upregulates NHEJ. IR induces translocation of PD-L1 from the membrane into nucleus dependent on deglycosylation of PD-L1 at N219 and CMTM6 and leads to PD-L1 recruitment to DSBs foci. PD-L1 interacts with Ku in the nucleus and enhances Ku binding to DSB DNA. The interaction between the IgC domain of PD-L1 and the core domain of Ku is required for PD-L1 to accelerate NHEJ-mediated DSB repair and produce radioresistance. Thus, PD-L1, in addition to its immune inhibitory activity, acts as mechanistic driver for NHEJ-mediated DSB repair in cancer.
Collapse
Affiliation(s)
- Zhen Shu
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Bhakti Dwivedi
- Bioinformatics and Systems Biology Shared Resource, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Jeffrey M Switchenko
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA, USA
| | - David S Yu
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Xingming Deng
- Department of Radiation Oncology, Emory University School of Medicine and Winship Cancer Institute of Emory University, Atlanta, GA, USA.
| |
Collapse
|
41
|
Chang TG, Cao Y, Sfreddo HJ, Dhruba SR, Lee SH, Valero C, Yoo SK, Chowell D, Morris LGT, Ruppin E. LORIS robustly predicts patient outcomes with immune checkpoint blockade therapy using common clinical, pathologic and genomic features. NATURE CANCER 2024; 5:1158-1175. [PMID: 38831056 PMCID: PMC11962634 DOI: 10.1038/s43018-024-00772-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 04/24/2024] [Indexed: 06/05/2024]
Abstract
Despite the revolutionary impact of immune checkpoint blockade (ICB) in cancer treatment, accurately predicting patient responses remains challenging. Here, we analyzed a large dataset of 2,881 ICB-treated and 841 non-ICB-treated patients across 18 solid tumor types, encompassing a wide range of clinical, pathologic and genomic features. We developed a clinical score called LORIS (logistic regression-based immunotherapy-response score) using a six-feature logistic regression model. LORIS outperforms previous signatures in predicting ICB response and identifying responsive patients even with low tumor mutational burden or programmed cell death 1 ligand 1 expression. LORIS consistently predicts patient objective response and short-term and long-term survival across most cancer types. Moreover, LORIS showcases a near-monotonic relationship with ICB response probability and patient survival, enabling precise patient stratification. As an accurate, interpretable method using a few readily measurable features, LORIS may help improve clinical decision-making in precision medicine to maximize patient benefit. LORIS is available as an online tool at https://loris.ccr.cancer.gov/ .
Collapse
Affiliation(s)
- Tian-Gen Chang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Yingying Cao
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Hannah J Sfreddo
- Department of Surgery and Cancer Immunogenomics Research Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Saugato Rahman Dhruba
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Se-Hoon Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute of Health Science and Technology, Sungkyunkwan University, Seoul, South Korea
| | - Cristina Valero
- Department of Surgery and Cancer Immunogenomics Research Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Seong-Keun Yoo
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diego Chowell
- The Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luc G T Morris
- Department of Surgery and Cancer Immunogenomics Research Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
42
|
Bergstrom EN, Alexandrov LB. Enhanced precision in immunotherapy. NATURE CANCER 2024; 5:1136-1138. [PMID: 39134713 DOI: 10.1038/s43018-024-00802-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/30/2024]
Affiliation(s)
- Erik N Bergstrom
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Ludmil B Alexandrov
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA, USA.
- Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
- Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
43
|
Wang K, Peng B, Xu R, Lu T, Chang X, Shen Z, Shi J, Li M, Wang C, Zhou X, Xu C, Chang H, Zhang L. Comprehensive analysis of PPP4C's impact on prognosis, immune microenvironment, and immunotherapy response in lung adenocarcinoma using single-cell sequencing and multi-omics. Front Immunol 2024; 15:1416632. [PMID: 39026674 PMCID: PMC11254641 DOI: 10.3389/fimmu.2024.1416632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Background Elevated PPP4C expression has been associated with poor prognostic implications for patients suffering from lung adenocarcinoma (LUAD). The extent to which PPP4C affects immune cell infiltration in LUAD, as well as the importance of associated genes in clinical scenarios, still requires thorough investigation. Methods In our investigation, we leveraged both single-cell and comprehensive RNA sequencing data, sourced from LUAD patients, in our analysis. This study also integrated datasets of immune-related genes from InnateDB into the framework. Our expansive evaluation employed various analytical techniques; these included pinpointing differentially expressed genes, constructing WGCNA, implementing Cox proportional hazards models. We utilized these methods to investigate the gene expression profiles of PPP4C within the context of LUAD and to clarify its potential prognostic value for patients. Subsequent steps involved validating the observed enhancement of PPP4C expression in LUAD samples through a series of experimental approaches. The array comprised immunohistochemistry staining, Western blotting, quantitative PCR, and a collection of cell-based assays aimed at evaluating the influence of PPP4C on the proliferative and migratory activities of LUAD cells. Results In lung cancer, elevated expression levels of PPP4C were observed, correlating with poorer patient prognoses. Validation of increased PPP4C levels in LUAD specimens was achieved using immunohistochemical techniques. Experimental investigations have substantiated the role of PPP4C in facilitating cellular proliferation and migration in LUAD contexts. Furthermore, an association was identified between the expression of PPP4C and the infiltration of immune cells in these tumors. A prognostic framework, incorporating PPP4C and immune-related genes, was developed and recognized as an autonomous predictor of survival in individuals afflicted with LUAD. This prognostic tool has demonstrated considerable efficacy in forecasting patient survival and their response to immunotherapeutic interventions. Conclusion The involvement of PPP4C in LUAD is deeply intertwined with the tumor's immune microenvironment. PPP4C's over-expression is associated with negative clinical outcomes, promoting both tumor proliferation and spread. A prognostic framework based on PPP4C levels may effectively predict patient prognoses in LUAD, as well as the efficacy of immunotherapy strategy. This research sheds light on the mechanisms of immune interaction in LUAD and proposes a new strategy for treatment.
Collapse
Affiliation(s)
- Kaiyu Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Peng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ran Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tong Lu
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaoyan Chang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhiping Shen
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiaxin Shi
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Meifeng Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chenghao Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiang Zhou
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chengyu Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hao Chang
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Linyou Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
44
|
Wu XH, Wang JQ, Wang MD, Xiao T, Wang Y, Niu JY, Wang L, Hou DY, Fu B, Liu Z, Wang H, Xu W. Bispecific fibrous glue synergistically boosts vascular normalization and antitumor immunity for advanced renal carcinoma therapy. Biomaterials 2024; 308:122550. [PMID: 38581762 DOI: 10.1016/j.biomaterials.2024.122550] [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: 01/11/2024] [Revised: 03/21/2024] [Accepted: 03/23/2024] [Indexed: 04/08/2024]
Abstract
Immune checkpoint blockade therapy represented by programmed cell death ligand 1 (PD-L1) inhibitor for advanced renal carcinoma with an objective response rate (ORR) in patients is less than 20%. It is attributed to abundant tumoral vasculature with abnormal structure limiting effector T cell infiltration and drug penetration. We propose a bispecific fibrous glue (BFG) to regulate tumor immune and vascular microenvironments simultaneously. The bispecific precursor glue peptide-1 (pre-GP1) can penetrate tumor tissue deeply and self-assemble into BFG in the presence of neuropilin-1 (NRP-1) and PD-L1. The resultant fibrous glue is capable of normalizing tumoral vasculature as well as restricting immune escape. The pre-GP1 retains a 6-fold higher penetration depth than that of antibody in the multicellular spheroids (MCSs) model. It also shows remarkable tumor growth inhibition (TGI) from 19% to 61% in a murine advanced large tumor model compared to the clinical combination therapy. In addition, in the orthotopic renal tumor preclinical model, the lung metastatic nodules are reduced by 64% compared to the clinically used combination. This pre-GP1 provides a promising strategy to control the progression and metastasis of advanced renal carcinoma.
Collapse
Affiliation(s)
- Xiu-Hai Wu
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, China; CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China; NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
| | - Jia-Qi Wang
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, China; CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Man-Di Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China; Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53719, USA
| | - Ting Xiao
- Henan Institute of Advanced Technology, Zhengzhou University, No.100 Science Avenue, Zhengzhou, 450052, China
| | - Yu Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China; Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53719, USA
| | - Jia-Yuan Niu
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China; Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Lu Wang
- NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
| | - Da-Yong Hou
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, China; NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
| | - Bo Fu
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, China; CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China; NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China
| | - Zimo Liu
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST) No. 11 Beiyitiao, Zhongguancun, Beijing, 100190, China.
| | - Wanhai Xu
- Department of Urology, Harbin Medical University Cancer Hospital, Harbin, 150081, China; NHC Key Laboratory of Molecular Probes and Targeted Diagnosis and Therapy, Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
45
|
Liu J, Xu X, Zhong H, Yu M, Abuduaini N, Zhang S, Yang X, Feng B. Glycosylation and Its Role in Immune Checkpoint Proteins: From Molecular Mechanisms to Clinical Implications. Biomedicines 2024; 12:1446. [PMID: 39062019 PMCID: PMC11274725 DOI: 10.3390/biomedicines12071446] [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: 05/16/2024] [Revised: 06/21/2024] [Accepted: 06/22/2024] [Indexed: 07/28/2024] Open
Abstract
Immune checkpoint proteins have become recent research hotspots for their vital role in maintaining peripheral immune tolerance and suppressing immune response function in a wide range of tumors. Therefore, investigating the immunomodulatory functions of immune checkpoints and their therapeutic potential for clinical use is of paramount importance. The immune checkpoint blockade (ICB) is an important component of cancer immunotherapy, as it targets inhibitory immune signaling transduction with antagonistic antibodies to restore the host immune response. Anti-programmed cell death-1 (PD-1) and anti-cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) monoclonal antibodies are two main types of widely used ICBs that drastically improve the survival and prognosis of many patients with cancer. Nevertheless, the response rate of most cancer types remains relatively low due to the drug resistance of ICBs, which calls for an in-depth exploration to improve their efficacy. Accumulating evidence suggests that immune checkpoint proteins are glycosylated in forms of N-glycosylation, core fucosylation, or sialylation, which affect multiple biological functions of proteins such as protein biosynthesis, stability, and interaction. In this review, we give a brief introduction to several immune checkpoints and summarize primary molecular mechanisms that modulate protein stability and immunosuppressive function. In addition, newly developed methods targeting glycosylation on immune checkpoints for detection used to stratify patients, as well as small-molecule agents disrupting receptor-ligand interactions to circumvent drug resistance of traditional ICBs, in order to increase the clinical efficacy of immunotherapy strategies of patients with cancer, are also included to provide new insights into scientific research and clinical treatments.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Bo Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200000, China; (J.L.); (X.X.); (H.Z.); (M.Y.); (N.A.); (S.Z.); (X.Y.)
| |
Collapse
|
46
|
Guo YA, Kulshrestha T, Chang MM, Kassam I, Revkov E, Rizzetto S, Tan AC, Tan DS, Tan IB, Skanderup AJ. Transcriptome Deconvolution Reveals Absence of Cancer Cell Expression Signature in Immune Checkpoint Blockade Response. CANCER RESEARCH COMMUNICATIONS 2024; 4:1581-1596. [PMID: 38722600 PMCID: PMC11203396 DOI: 10.1158/2767-9764.crc-23-0442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/16/2024] [Accepted: 05/07/2024] [Indexed: 06/28/2024]
Abstract
Immune checkpoint therapy (ICB) has conferred significant and durable clinical benefit to some patients with cancer. However, most patients do not respond to ICB, and reliable biomarkers of ICB response are needed to improve patient stratification. Here, we performed a transcriptome-wide meta-analysis across 1,486 tumors from ICB-treated patients and tumors with expected ICB outcomes based on microsatellite status. Using a robust transcriptome deconvolution approach, we inferred cancer- and stroma-specific gene expression differences and identified cell-type specific features of ICB response across cancer types. Consistent with current knowledge, stromal expression of CXCL9, CXCL13, and IFNG were the top determinants of favorable ICB response. In addition, we identified a group of potential immune-suppressive genes, including FCER1A, associated with poor response to ICB. Strikingly, PD-L1 expression in stromal cells, but not cancer cells, is correlated with ICB response across cancer types. Furthermore, the unbiased transcriptome-wide analysis failed to identify cancer-cell intrinsic expression signatures of ICB response conserved across tumor types, suggesting that cancer cells lack tissue-agnostic transcriptomic features of ICB response. SIGNIFICANCE Our results challenge the prevailing dogma that cancer cells present tissue-agnostic molecular markers that modulate immune activity and ICB response, which has implications on the development of improved ICB diagnostics and treatments.
Collapse
Affiliation(s)
- Yu Amanda Guo
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, #02-01 Genome, Singapore 138672, Republic of Singapore
| | - Tanmay Kulshrestha
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, #02-01 Genome, Singapore 138672, Republic of Singapore
| | - Mei Mei Chang
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, #02-01 Genome, Singapore 138672, Republic of Singapore
| | - Irfahan Kassam
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, #02-01 Genome, Singapore 138672, Republic of Singapore
| | - Egor Revkov
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, #02-01 Genome, Singapore 138672, Republic of Singapore
- School of Computing, National University of Singapore, Computing 1, 13 Computing Drive, Singapore 117417, Republic of Singapore
| | - Simone Rizzetto
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, #02-01 Genome, Singapore 138672, Republic of Singapore
| | - Aaron C. Tan
- Department of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Republic of Singapore
| | - Daniel S.W. Tan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, #02-01 Genome, Singapore 138672, Republic of Singapore
- Department of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Republic of Singapore
| | - Iain Beehuat Tan
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, #02-01 Genome, Singapore 138672, Republic of Singapore
- Department of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Republic of Singapore
| | - Anders J. Skanderup
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), 60 Biopolis Street, #02-01 Genome, Singapore 138672, Republic of Singapore
- School of Computing, National University of Singapore, Computing 1, 13 Computing Drive, Singapore 117417, Republic of Singapore
- Department of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Republic of Singapore
| |
Collapse
|
47
|
Zhao L, Ren Y, Zhang G, Zheng K, Wang J, Sha H, Zhao M, Huang R, Kang D, Su X, Wu Y, Zhang W, Lai R, Li L, Mei R, Wang Y, Tian Y, Wang F, Liu B, Zou Z. Single-arm study of camrelizumab plus apatinib for patients with advanced mucosal melanoma. J Immunother Cancer 2024; 12:e008611. [PMID: 38908858 PMCID: PMC11328654 DOI: 10.1136/jitc-2023-008611] [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] [Accepted: 03/29/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND Previous studies have suggested the potential synergistic antitumor activity when combining immune checkpoint inhibitors with anti-angiogenic agents in various solid tumors. We aimed to assess the efficacy and safety of camrelizumab (a humanized programmed cell death-1 antibody) plus apatinib (a vascular endothelial growth factor receptor tyrosine kinase inhibitor) for patients with advanced mucosal melanoma (MM), and explore-related biomarkers. METHODS We conducted a single-center, open-label, single-arm, phase II study. Patients with unresectable or recurrent/metastatic MM received camrelizumab and apatinib. The primary endpoint was the confirmed objective response rate (ORR). RESULTS Between April 2019 and June 2022, 32 patients were enrolled, with 50.0% previously received systemic therapy. Among 28 patients with evaluable response, the confirmed ORR was 42.9%, the disease control rate was 82.1%, and the median progression-free survival (PFS) was 8.05 months. The confirmed ORR was 42.9% (6/14) in both treatment-naïve and previously treated patients. Notably, treatment-naïve patients had a median PFS of 11.89 months, and those with prior treatment had a median PFS of 6.47 months. Grade 3 treatment-related adverse events were transaminase elevation, rash, hyperbilirubinemia, proteinuria, hypertension, thrombocytopenia, hand-foot syndrome and diarrhea. No treatment-related deaths were observed. Higher tumor mutation burden (TMB), increased T-cell receptor (TCR) diversity, and altered receptor tyrosine kinase (RTK)/RAS pathway correlated with better tumor response. CONCLUSION Camrelizumab plus apatinib provided promising antitumor activity with acceptable toxicity in patients with advanced MM. TMB, TCR diversity and RTK/RAS pathway genes were identified as potential predictive biomarkers and warrant further validation. TRIAL REGISTRATION NUMBER Chinese Clinical Trial Registry, ChiCTR1900023277.
Collapse
Affiliation(s)
- Lianjun Zhao
- The Comprehensive Cancer Center of Nanjing Drum
Tower Hospital, Affiliated Hospital of Medical School, Nanjing University,
Nanjing, China
- Clinical Cancer Institute of Nanjing
University, Nanjing,
China
| | - Yu Ren
- The Comprehensive Cancer Center of Nanjing Drum
Tower Hospital, Affiliated Hospital of Medical School, Nanjing University,
Nanjing, China
- Clinical Cancer Institute of Nanjing
University, Nanjing,
China
| | - Guiying Zhang
- The Comprehensive Cancer Center of Nanjing Drum
Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing
University of Chinese Medicine, Nanjing, China
| | - Kelin Zheng
- The Comprehensive Cancer Center of Nanjing Drum
Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing
University of Chinese Medicine, Nanjing, China
| | - Jiayu Wang
- The Comprehensive Cancer Center of Nanjing Drum
Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing
University of Chinese Medicine, Nanjing, China
| | - Huizi Sha
- The Comprehensive Cancer Center of Nanjing Drum
Tower Hospital, Affiliated Hospital of Medical School, Nanjing University,
Nanjing, China
- Clinical Cancer Institute of Nanjing
University, Nanjing,
China
| | - Mengke Zhao
- The Comprehensive Cancer Center of Nanjing Drum
Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Rong Huang
- The Comprehensive Cancer Center of Nanjing Drum
Tower Hospital, Affiliated Hospital of Medical School, Nanjing University,
Nanjing, China
| | - Donglin Kang
- The Comprehensive Cancer Center of Nanjing Drum
Tower Hospital Clinical College of Nanjing Medical University, Nanjing, China
| | - Xinyu Su
- The Comprehensive Cancer Center of Nanjing Drum
Tower Hospital, Affiliated Hospital of Medical School, Nanjing University,
Nanjing, China
| | - Yirong Wu
- The Comprehensive Cancer Center of Nanjing Drum
Tower Hospital, Affiliated Hospital of Medical School, Nanjing University,
Nanjing, China
| | - Wangling Zhang
- The Comprehensive Cancer Center of Nanjing Drum
Tower Hospital, Affiliated Hospital of Medical School, Nanjing University,
Nanjing, China
| | - Ruihe Lai
- Department of Nuclear Medicine of Nanjing Drum
Tower Hospital, Affiliated Hospital of Medical School, Nanjing University,
Nanjing, China
| | - Lin Li
- Department of Pathology of Nanjing Drum Tower
Hospital, Affiliated Hospital of Medical School, Nanjing University,
Nanjing, China
| | - Rui Mei
- Jiangsu Hengrui Pharmaceuticals Co.,
Ltd, Shanghai,
China
| | - Yitao Wang
- Jiangsu Hengrui Pharmaceuticals Co.,
Ltd, Shanghai,
China
| | - You Tian
- Jiangsu Hengrui Pharmaceuticals Co.,
Ltd, Shanghai,
China
| | - Fufeng Wang
- Geneseeq Research institute, Nanjing Geneseeq
Technology Inc, Nanjing,
China
| | - Baorui Liu
- The Comprehensive Cancer Center of Nanjing Drum
Tower Hospital, Affiliated Hospital of Medical School, Nanjing University,
Nanjing, China
- Clinical Cancer Institute of Nanjing
University, Nanjing,
China
| | - Zhengyun Zou
- The Comprehensive Cancer Center of Nanjing Drum
Tower Hospital, Affiliated Hospital of Medical School, Nanjing University,
Nanjing, China
- Clinical Cancer Institute of Nanjing
University, Nanjing,
China
| |
Collapse
|
48
|
Wei Y, Zhang Z, Xue T, Lin Z, Chen X, Tian Y, Li Y, Jing Z, Fang W, Fang T, Li B, Chen Q, Lan T, Meng F, Zhang X, Liang X. In Situ Synthesis of an Immune-Checkpoint Blocker from Engineered Bacteria Elicits a Potent Antitumor Response. ACS Synth Biol 2024; 13:1679-1693. [PMID: 38819389 DOI: 10.1021/acssynbio.3c00569] [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: 06/01/2024]
Abstract
Immune-checkpoint blockade (ICB) reinvigorates T cells from exhaustion and potentiates T-cell responses to tumors. However, most patients do not respond to ICB therapy, and only a limited response can be achieved in a "cold" tumor with few infiltrated lymphocytes. Synthetic biology can be used to engineer bacteria as controllable bioreactors to synthesize biotherapeutics in situ. We engineered attenuated Salmonella VNP20009 with synthetic gene circuits to produce PD-1 and Tim-3 scFv to block immunosuppressive receptors on exhausted T cells to reinvigorate their antitumor response. Secreted PD-1 and Tim-3 scFv bound PD-1+ Tim-3+ T cells through their targeting receptors in vitro and potentiated the T-cell secretion of IFN-γ. Engineered bacteria colonized the hypoxic core of the tumor and synthesized PD-1 and Tim-3 scFv in situ, reviving CD4+ T cells and CD8+ T cells to execute an antitumor response. The bacteria also triggered a strong innate immune response, which stimulated the expansion of IFN-γ+ CD4+ T cells within the tumors to induce direct and indirect antitumor immunity.
Collapse
Affiliation(s)
- Yuting Wei
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Zhirang Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Tianyuan Xue
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Zhongda Lin
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Xinyu Chen
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan 523710, China
| | - Yishi Tian
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Yuan Li
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Zhangyan Jing
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Wenli Fang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Tianliang Fang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Baoqi Li
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Qi Chen
- Department of Physiology, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, China
| | - Tianyu Lan
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Fanqiang Meng
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Xudong Zhang
- Shenzhen Key Laboratory for Systems Medicine in Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Xin Liang
- Department of Physiology, Guangdong Provincial Key Laboratory of Medical Molecular Diagnostics, Guangdong Medical University, Dongguan 523808, China
| |
Collapse
|
49
|
Zhou R, Lu D, Mi J, Wang C, Lu W, Wang Z, Li X, Wei C, Zhang H, Ji J, Zhang Y, Zhang D, Wang F. Disulfidptosis-related genes serve as potential prognostic biomarkers and indicate tumor microenvironment characteristics and immunotherapy response in prostate cancer. Sci Rep 2024; 14:14107. [PMID: 38898043 PMCID: PMC11187134 DOI: 10.1038/s41598-024-61679-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/08/2024] [Indexed: 06/21/2024] Open
Abstract
Disulfidptosis, a newly identified programmed cell death pathway in prostate cancer (PCa), is closely associated with intracellular disulfide stress and glycolysis. This study aims to elucidate the roles of disulfidptosis-related genes (DRGs) in the pathogenesis and progression of PCa, with the goal of improving diagnostic and therapeutic approaches. We analyzed PCa datasets and normal tissue transcriptome data from TCGA, GEO, and MSKCC. Using consensus clustering analysis and LASSO regression, we developed a risk scoring model, which was validated in an independent cohort. The model's predictive accuracy was confirmed through Kaplan-Meier curves, receiver operating characteristic (ROC) curves, and nomograms. Additionally, we explored the relationship between the risk score and immune cell infiltration, and examined the tumor microenvironment and somatic mutations across different risk groups. We also investigated responses to immunotherapy and drug sensitivity. Our analysis identified two disulfidosis subtypes with significant differences in survival, immune environments, and treatment responses. According to our risk score, the high-risk group exhibited poorer progression-free survival (PFS) and higher tumor mutational burden (TMB), associated with increased immune suppression. Functional enrichment analysis linked high-risk features to key cancer pathways, including the IL-17 signaling pathway. Moreover, drug sensitivity analysis revealed varied responses to chemotherapy, suggesting the potential for disulfidosis-based personalized treatment strategies. Notably, we identified PROK1 as a crucial prognostic marker in PCa, with its reduced expression correlating with disease progression. In summary, our study comprehensively assessed the clinical implications of DRGs in PCa progression and prognosis, offering vital insights for tailored precision medicine approaches.
Collapse
Affiliation(s)
- Rongbin Zhou
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed By the Province and Ministry, Guangxi Medical University, No. 22, Shuangyong Road, Qingxiu District, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Dingjin Lu
- Department of Urology, People's Hospital of Beihai, Beihai, 536000, Guangxi, China
| | - Junhao Mi
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed By the Province and Ministry, Guangxi Medical University, No. 22, Shuangyong Road, Qingxiu District, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Chengbang Wang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Wenhao Lu
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed By the Province and Ministry, Guangxi Medical University, No. 22, Shuangyong Road, Qingxiu District, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Zuheng Wang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiao Li
- School of Life Sciences, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Chunmeng Wei
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Huiyong Zhang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jin Ji
- Department of Urology, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Department of Urology, Naval Medical Center, Naval Medical Univiersiy, 338 Huaihai West Road, Shanghai, 200433, China
| | - Yifeng Zhang
- Department of Urology, Naval Medical Center, Naval Medical Univiersiy, 338 Huaihai West Road, Shanghai, 200433, China.
| | - Duobing Zhang
- Department of Urology, Suzhou Hospital of Anhui Medical University, 616 The Third Bianyang Road, Yongqiao District, Suzhou, 234000, Anhui, China.
| | - Fubo Wang
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co-Constructed By the Province and Ministry, Guangxi Medical University, No. 22, Shuangyong Road, Qingxiu District, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- School of Life Sciences, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, 530021, Guangxi, China.
- School of Public Health, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
50
|
Zhan Y, Ma S, Zhang T, Zhang L, Zhao P, Yang X, Liu M, Cheng W, Li Y, Wang J. Identification of a novel monocyte/macrophage-related gene signature for predicting survival and immune response in acute myeloid leukemia. Sci Rep 2024; 14:14012. [PMID: 38890346 PMCID: PMC11189543 DOI: 10.1038/s41598-024-64567-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 06/11/2024] [Indexed: 06/20/2024] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematological tumor with poor immunotherapy effect. This study was to develop a monocyte/macrophage-related prognostic risk score (MMrisk) and identify new therapeutic biomarkers for AML. We utilized differentially expressed genes (DEGs) in combination with single-cell RNA sequencing to identify monocyte/macrophage-related genes (MMGs). Eight genes were selected for the construction of a MMrisk model using univariate Cox regression analysis and LASSO regression analysis. We then validated the MMrisk on two GEO datasets. Lastly, we investigated the immunologic characteristics and advantages of immunotherapy and potential targeted drugs for MMrisk groups. Our study identified that the MMrisk is composed of eight MMGs, including HOPX, CSTB, MAP3K1, LGALS1, CFD, MXD1, CASP1 and BCL2A1. The low MMrisk group survived longer than high MMrisk group (P < 0.001). The high MMrisk group was positively correlated with B cells, plasma cells, CD4 memory cells, Mast cells, CAFs, monocytes, M2 macrophages, Endothelial, tumor mutation, and most immune checkpoints (PD1, Tim-3, CTLA4, LAG3). Furthermore, drug sensitivity analysis showed that AZD.2281, Axitinib, AUY922, ABT.888, and ATRA were effective in high-risk MM patients. Our research shows that MMrisk is a potential biomarker which is helpful to identify the molecular characteristics of AML immunology.
Collapse
Affiliation(s)
- Yun Zhan
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, 550004, People's Republic of China
- Guizhou Province Institute of Hematology, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Sixing Ma
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, 550004, People's Republic of China
- Department of Vascular Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Tianzhuo Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
- Guizhou Province Institute of Hematology, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Luxin Zhang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
- Guizhou Province Institute of Hematology, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Peng Zhao
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
- Guizhou Province Institute of Hematology, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Xueying Yang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
- Guizhou Province Institute of Hematology, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Min Liu
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
- Guizhou Province Institute of Hematology, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Weiwei Cheng
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
- Guizhou Province Institute of Hematology, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Ya Li
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
- Guizhou Province Institute of Hematology, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China
| | - Jishi Wang
- Department of Hematology, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China.
- Department of Clinical Medical School, Guizhou Medical University, Guiyang, 550004, People's Republic of China.
- Guizhou Province Institute of Hematology, Guizhou Province Hematopoietic Stem Cell Transplantation Center, Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, People's Republic of China.
| |
Collapse
|