1
|
Qiao Y, Xie D, Li Z, Cao S, Zhao D. Global research trends on biomarkers for cancer immunotherapy: Visualization and bibliometric analysis. Hum Vaccin Immunother 2025; 21:2435598. [PMID: 39773010 PMCID: PMC11730411 DOI: 10.1080/21645515.2024.2435598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/08/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
The global burden of cancer continues to grow, posing a significant public health challenge. Although cancer immunotherapy has shown significant efficacy, the response rate is not high. Therefore, the objective of our research was to identify the latest research trends and hotspots on biomarkers from 1993 to 2023. Data were collected from the database Web of Science core collection. Bibliometric analysis and visualization were conducted with CiteSpace(6.3.1), VOSviewer (v1.6.20), R-bibliometrix(v4.3.3), and Microsoft Excel(2019). A total of 2686 literatures were retrieved. The sheer annual volume of publications has shown a rapid upward trend since 2015. The United States has generated the most publications and Harvard University ranked as a leading institution. The global biomarker research on immune checkpoint inhibitors (ICIs) revealed regional differences and in-depth explorations should be promoted in developing countries. Although China has become the second largest country in terms of publication, the average citation per paper and the total link strength were both lower than the other countries. The research on biomarkers mainly concentrated upon the following aspects: PD-1/PD-L1, CTLA-4, gene expression, adverse events, total mutational burden (TMB), body mass index (BMI), gut microbiota, cd8(+)/cd4(+) t-cells, and blood-related biomarkers such as lactate dehydrogenase (LDH), neutrophil-lymphocyte ratio (NLR), cytokines. Furthermore, "artificial intelligence" and "machine learning" have become the most important research hotspot over the last 2 y, which will help us to identify useful biomarkers from complex big data and provide a basis for precise medicine for malignant tumors.
Collapse
Affiliation(s)
- Yuan Qiao
- Department of Clinical Pharmacy, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| | - Dong Xie
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhengxiang Li
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Shaohua Cao
- Department of Clinical Pharmacy, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| | - Dong Zhao
- Department of Clinical Laboratory, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| |
Collapse
|
2
|
Li W, Xiao L, Li H, Cui W. Global research trends of immunosenescence and immunotherapy: A bibliometric study. Hum Vaccin Immunother 2025; 21:2469403. [PMID: 39992200 PMCID: PMC11853558 DOI: 10.1080/21645515.2025.2469403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 01/31/2025] [Accepted: 02/15/2025] [Indexed: 02/25/2025] Open
Abstract
Immunosenescence refers to the gradual decline in immune system function with age, increasing susceptibility to infections and cancer in the elderly. The advent of novel immunotherapies has revolutionized the field of cancer treatment. However, the majority of patients exhibit poor re-sponses to immunotherapy, with immunosenescence likely playing a significant role. In recent years, significant progress has been made in understanding the interplay between immunosenescence and immunotherapy. Our research aims to explore the prospects and development trends in the field of immunosenescence and immunotherapy using a bibliometric analysis. Relevant articles were collected from the Web of Science Core Collection (WoSCC) (retrieved on July 20, 2024). Primary bibliometric characteristics were analyzed using the R package "Biblio-metrix," and keyword co-occurrence analysis and visualization were conducted using VOSviewer. A total of 213 English-language original research and review articles spanning 35 years were re-trieved for bibliometric analysis. There was a surge in publications in this field starting in 2017. The United States and China contributed the most articles. Frontiers in Immunology was the most productive journal, while the University of California System was the highest contributing institution. Besse Benjamin from France emerged as the most influential researcher in this field. Popular keywords included "nivolumab," "T cells," "dendritic cells," and "regulatory T cells." The "immunosenescence-associated secretory phenotype" has become a new hotspot, with immune checkpoint inhibitors remaining a central theme in this domain. The field of immunosenescence and immunotherapy is entering a phase of rapid development and will continue to hold significant value in future research.
Collapse
Affiliation(s)
- Wendi Li
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Xiao
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haiyang Li
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Cui
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
3
|
Wei C, Liu M, Zhang W. Programmed cell death protein 1 in cancer cells. Cell Commun Signal 2025; 23:185. [PMID: 40241148 PMCID: PMC12001728 DOI: 10.1186/s12964-025-02155-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
Programmed cell death protein 1 (PD-1) is frequently detected in certain subsets of tumor cells, and our understanding of PD-1 signaling consequences has expanded to include control of tumor growth, stemness and drug resistance. Nonetheless, tumor cell-intrinsic PD-1 has been comparatively underexplored in relation to PD-1 expressed on the surface of immune cells as an immune checkpoint, despite the imperative need to comprehensively elucidate the underlying mechanisms of action for achieving optimal responses in tumor immunotherapy. Here, we review the roles of the regulation and function of tumor-intrinsic PD-1 from its expression to degradation processes. Our primary focus is on unraveling its enigmatic influence on tumorigenesis and progression as proposed by recent findings, while navigating the labyrinthine network of regulatory mechanisms governing its expression and intricate functional interplay. We also discuss how the elucidation of the mechanistic underpinnings of tumor-intrinsic PD-1 expression holds the potential to explain the divergent therapeutic outcomes observed with anti-PD-1-based combination therapies, thereby furnishing indispensable insights crucial for synergistic anti-tumor strategies.
Collapse
Affiliation(s)
- Chunlian Wei
- School of Basic Medical Sciences, Shandong Second Medical University, Weifang, 261053, Shandong, PR China
- Shandong Engineering Researh Center for Smart Materials and Regenerative Medicine, Shandong Second Medical University, Weifang, 261053, Shandong, PR China
| | - Meijun Liu
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, Shandong, PR China
- Shandong Engineering Researh Center for Smart Materials and Regenerative Medicine, Shandong Second Medical University, Weifang, 261053, Shandong, PR China
| | - Weifen Zhang
- School of Pharmacy, Shandong Second Medical University, Weifang, 261053, Shandong, PR China.
- Shandong Engineering Researh Center for Smart Materials and Regenerative Medicine, Shandong Second Medical University, Weifang, 261053, Shandong, PR China.
| |
Collapse
|
4
|
Philips S, Lu P, Fausel C, Wagner T, Jiang G, Shen F, Cantor E, Tran M, Roland LM, Schneider BP. Association of heightened host and tumor immunity with prolonged duration of response to checkpoint inhibition across solid tumors. Sci Rep 2025; 15:13195. [PMID: 40240402 PMCID: PMC12003766 DOI: 10.1038/s41598-025-96925-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 04/01/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer immunotherapy is a beneficial therapy for many cancer types, but predictive pan-tumor biomarkers for clinical benefit are suboptimal. Our study, employing DNA and RNA based analysis, investigated the role of predicted neoantigens in the benefits of immunotherapy within a cohort of 88 patients of European descent with advanced solid tumors. Patients who had a prolonged (> 12 months) duration of immunotherapy exhibited heightened immune responses, characterized by increased levels of predicted neoantigens with strong HLA binding potential, elevated cytotoxic marker levels, and enhanced T cell activity. Furthermore, our analysis revealed associations between prolonged duration of therapy and rare variants, notably within the EPHA8 gene. These variants, exclusive to patients with a prolonged (> 12 months) duration of immunotherapy, suggest potential implications for immunotherapy response. In addition, the evolutionary conservation of these variants across vertebrate species underscores their functional importance in tumor biology and ultimately, treatment outcomes. Despite limitations in sample size and patient homogeneity, our findings emphasize the potential utility of understanding the molecular and immunological mechanisms underlying immunotherapy responses to further refine personalized treatment strategies.
Collapse
Affiliation(s)
- Santosh Philips
- Division of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Pei Lu
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Chris Fausel
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Thomas Wagner
- College of Pharmacy and Health Sciences, Butler University, Indianapolis, IN, USA
| | - Guanglong Jiang
- Division of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fei Shen
- Division of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Erica Cantor
- Division of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mya Tran
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Lauren M Roland
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA
| | - Bryan P Schneider
- Division of Hematology and Oncology, Indiana University School of Medicine, Indianapolis, IN, USA.
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA.
- Division of Hematology/Oncology, Department of Medicine, Indiana University, 535 Barnhill Drive, RT 473, Indianapolis, IN, 46202, USA.
| |
Collapse
|
5
|
Zhang MJ, Wen Y, Sun ZJ. The impact of metabolic reprogramming on tertiary lymphoid structure formation: enhancing cancer immunotherapy. BMC Med 2025; 23:217. [PMID: 40223062 PMCID: PMC11995586 DOI: 10.1186/s12916-025-04037-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 03/26/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Cancer immunotherapy has achieved unprecedented success in the field of cancer therapy. However, its potential is constrained by a low therapeutic response rate. MAIN BODY Tertiary lymphoid structure (TLS) plays a crucial role in antitumor immunity and is associated with a good prognosis. Metabolic reprogramming, as a hallmark of the tumor microenvironment, can influence tumor immunity and promote the formation of follicular helper T cells and germinal centers. However, many current studies focus on the correlation between metabolism and TLS formation factors, and there is insufficient direct evidence to suggest that metabolism drives TLS formation. This review provided a comprehensive summary of the relationship between metabolism and TLS formation, highlighting glucose metabolism, lipid metabolism, amino acid metabolism, and vitamin metabolism. CONCLUSIONS In the future, an in-depth exploration of how metabolism affects cell interactions and the role of microorganisms in TLS will significantly advance our understanding of metabolism-enhanced antitumor immunity.
Collapse
Affiliation(s)
- Meng-Jie Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Yan Wen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Taikang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430079, China.
- Department of Oral Maxillofacial-Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
6
|
Sarathkumara YD, Van Bibber NW, Liu Z, Heslop HE, Rouce RH, Coghill AE, Rooney CM, Proietti C, Doolan DL. Differential antibody response to EBV proteome following EBVST immunotherapy in EBV-associated lymphomas. Blood Adv 2025; 9:1658-1669. [PMID: 39908567 PMCID: PMC11995064 DOI: 10.1182/bloodadvances.2024014937] [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: 10/10/2024] [Revised: 01/03/2025] [Accepted: 01/21/2025] [Indexed: 02/07/2025] Open
Abstract
ABSTRACT Epstein-Barr virus (EBV) is associated with a diverse range of lymphomas. EBV-specific T-cell (EBVST) infusions have shown promise in safety and clinical effectiveness in treating EBV-associated lymphomas; however, not all patients respond to T-cell immunotherapies. To identify EBV antigen-specific antibody responses associated with clinical outcomes, we comprehensively characterized antibody responses to the complete EBV proteome using a custom protein microarray in 56 patients with EBV-associated lymphoma who received EBVST infusions in phase 1 clinical trials. Responders (nonprogressors) and nonresponders (progressors) had distinct antibody profiles against EBV. Twenty-five immunoglobulin G (IgG) antibodies were significantly elevated in higher levels in nonresponders than in responders at 3 months after EBVST infusion. Ten of these remained significant after adjustment for sex, age, and cancer type, including LMP2A (4 variants), BGRF1/BDRF1 (2 variants), LMP1, BKRF2, BKRF4, and BALF5. Random forest analysis identified these 10 IgG antibodies as key predictors of clinical response. Paired analyses using blood samples collected at both before infusion and 3 months after EBVST infusion indicated an increase in the mean antibody level for 6 other anti-EBV antibodies (IgG [BGLF2, LF1, and BGLF3]; IgA [BGLF3, BALF2, and BBLF2/3) in nonresponders. Overall, our findings suggest that these EBV-directed antibodies as potential serological markers for predicting clinical responses to EBVST infusions and as therapeutic targets for immunotherapy in EBV-positive lymphomas. These trials were registered at www.clinicaltrials.gov as #NCT01555892 (Cytotoxic T-Lymphocytes for EBV-positive Lymphoma [GRALE]), #NCT02973113 (Nivolumab With Epstein Barr Virus Specific T Cells [EBVSTS], Relapsed/Refractory EBV Positive Lymphoma [PREVALE]), and #NCT02287311 (Most Closely Matched 3rd Party Rapidly Generated LMP, BARF1, and EBNA1 Specific CTL, EBV-Positive Lymphoma [MABEL]).
Collapse
Affiliation(s)
- Yomani D. Sarathkumara
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Nathan W. Van Bibber
- Cancer Epidemiology Program, Division of Population Sciences, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Zhiwei Liu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, MD
| | - Helen E. Heslop
- Center for Cell and Gene Therapy, Baylor College of Medicine Houston Methodist Hospital and Texas Children’s Hospital, Houston, TX
| | - Rayne H. Rouce
- Center for Cell and Gene Therapy, Baylor College of Medicine Houston Methodist Hospital and Texas Children’s Hospital, Houston, TX
| | - Anna E. Coghill
- Cancer Epidemiology Program, Division of Population Sciences, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | - Cliona M. Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine Houston Methodist Hospital and Texas Children’s Hospital, Houston, TX
| | - Carla Proietti
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Denise L. Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
7
|
Kalemoglu E, Jani Y, Canaslan K, Bilen MA. The role of immunotherapy in targeting tumor microenvironment in genitourinary cancers. Front Immunol 2025; 16:1506278. [PMID: 40260236 PMCID: PMC12009843 DOI: 10.3389/fimmu.2025.1506278] [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: 10/04/2024] [Accepted: 03/19/2025] [Indexed: 04/23/2025] Open
Abstract
Genitourinary (GU) cancers, including renal cell carcinoma, prostate cancer, bladder cancer, and testicular cancer, represent a significant health burden and are among the leading causes of cancer-related mortality worldwide. Despite advancements in traditional treatment modalities such as chemotherapy, radiotherapy, and surgery, the complex interplay within the tumor microenvironment (TME) poses substantial hurdles to achieving durable remission and cure. The TME, characterized by its dynamic and multifaceted nature, comprises various cell types, signaling molecules, and the extracellular matrix, all of which are instrumental in cancer progression, metastasis, and therapy resistance. Recent breakthroughs in immunotherapy (IO) have opened a new era in the management of GU cancers, offering renewed hope by leveraging the body's immune system to combat cancer more selectively and effectively. This approach, distinct from conventional therapies, aims to disrupt cancer's ability to evade immune detection through mechanisms such as checkpoint inhibition, therapeutic vaccines, and adoptive cell transfer therapies. These strategies highlight the shift towards personalized medicine, emphasizing the importance of understanding the intricate dynamics within the TME for the development of targeted treatments. This article provides an in-depth overview of the current landscape of treatment strategies for GU cancers, with a focus on IO targeting the specific cell types of TME. By exploring the roles of various cell types within the TME and their impact on cancer progression, this review aims to underscore the transformative potential of IO strategies in TME targeting, offering more effective and personalized treatment options for patients with GU cancers, thereby improving outcomes and quality of life.
Collapse
Affiliation(s)
- Ecem Kalemoglu
- Department of Internal Medicine, Rutgers-Jersey City Medical Center, Jersey City, NJ, United States
- Department of Basic Oncology, Health Institute of Ege University, Izmir, Türkiye
| | - Yash Jani
- Medical College of Georgia, Augusta, GA, United States
| | - Kubra Canaslan
- Department of Medical Oncology, Dokuz Eylul University, Izmir, Türkiye
| | - Mehmet Asim Bilen
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA, United States
- Department of Urology, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
8
|
Jin CX, Liu YS, Qin HN, Teng YB, Sun R, Ma ZJ, Wang AM, Liu JW. Peripheral inflammatory factors as prognostic predictors for first-line PD-1/PD-L1 inhibitors in advanced non-small cell lung cancer. Sci Rep 2025; 15:11206. [PMID: 40175366 PMCID: PMC11965408 DOI: 10.1038/s41598-024-84469-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 12/24/2024] [Indexed: 04/04/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have significantly improved the efficacy and prognosis of patients with non-small cell lung cancer (NSCLC). However, there remains a lack of optimal predictive biomarkers for assessing the response of ICIs. This study aimed to evaluate peripheral inflammatory factors as potential predictive biomarkers for NSCLC patients treated with ICIs. We retrospectively analyzed the correlation between peripheral inflammatory factors and the efficacy and prognosis of 124 patients with driver gene-negative advanced NSCLC who received first-line ICIs at our center from September 2018 to June 2022. Progression-free survival (PFS) was estimated using the Kaplan-Meier method. The association between the factors and multiple endpoints were investigated using univariate and multivariate analyses. A total of 124 patients were enrolled in this study. The objective response rate (ORR) was 49.2% and the disease control rate (DCR) was 97.6%, respectively. The median PFS was 12.7 months. The ORR differed statistically between groups based on the NLR, SII, with higher ORR observed in patients with an NLR ratio < 0.68, SII at 6 weeks < 531.26, and SII ratio < 0.74 (p < 0.05). The univariate analysis indicated that ECOG 0-1, smoking, NLR at 6 weeks < 2.72, NLR ratio < 0.68, LMR < 1.34, LMR ratio ≥ 1.38, and SII at 6 weeks < 531.26 were associated with longer PFS (p < 0.05). The multivariate analysis revealed that smoking (p = 0.013), baseline LMR (p = 0.015), and SII at 6 weeks (p = 0.010) were independent predictors of PFS. NLR, LMR, and SII maybe biomarkers for predicting the efficacy and prognosis of first-line ICIs therapy in driver gene-negative advanced NSCLC.
Collapse
Affiliation(s)
- Chen-Xing Jin
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Yan-Song Liu
- Department of Anesthesiology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
- Department of Anesthesiology, Obstetrics & Gynecology Hospital of Fudan University, Shanghai, 200011, Shanghai, China
| | - He-Nan Qin
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Yi-Bin Teng
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Rui Sun
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - Zhong-Jing Ma
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China
| | - A-Man Wang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China.
| | - Ji-Wei Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, Liaoning, China.
| |
Collapse
|
9
|
Basak U, Mukherjee S, Chakraborty S, Sa G, Dastidar SG, Das T. In-silico analysis unveiling the role of cancer stem cells in immunotherapy resistance of immune checkpoint-high pancreatic adenocarcinoma. Sci Rep 2025; 15:10355. [PMID: 40133473 PMCID: PMC11937529 DOI: 10.1038/s41598-025-93924-3] [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: 11/06/2024] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Although immune checkpoint (IC) inhibition is a major treatment modality in cancer-immunotherapy, multiple cancers show low response. Our in-silico exploration by mining cancer datasets using R2, available clinical trial data, and Kaplan-Meier analysis from GEPIA depicted that unlike low-responder (LR) cancers, high-responder (HR) cancers furnish higher IC expression, that upon lowering may provide better prognosis. Contrastingly, pancreatic adenocarcinoma (PAAD) demonstrated high IC expression but low immunotherapy-response. Infiltration scores from TIMER2.0 revealed higher pro-tumor immune subsets and cancer-associated fibroblasts (CAFs) while depicting lower anti-tumor immune subsets in PAAD as compared to HR lung adenocarcinoma (LUAD). Additionally, bioinformatic tool cBioportal showed lesser tumor mutational burden, mismatch repair deficiency and greater percent of driver mutations in TP53, KRAS and CDKN2A in PAAD, supporting its higher immunotherapy-resistance than LUAD. Our search for the 'key' immunotherapy response-deciding factor(s) revealed cancer stem cells (CSCs), the known contributors of therapy-resistance and immuno-evasion, to be positively correlated with above-mentioned driver mutations, pro-tumor immune and CAF subsets; and that PAAD furnished higher expression of CSC genes than LUAD. UMAP/tSNE analyses revealed that high CSC signature is positively correlated with immunotherapy-resistance genes and pro-cancer immune cells, while negatively with cytotoxic-T cells in PAAD. Our in-silico study explains the low immunotherapy-response in high IC-expressing PAAD, wherein CSC plays a pivotal role. Further exploration portrayed correlation of CSCs with immunotherapy-resistance in other LR and HR cancers too, substantiating the need for personalized CSC evaluation and targeting for successful immunotherapy outcomes.
Collapse
Affiliation(s)
- Udit Basak
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Sumon Mukherjee
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Sourio Chakraborty
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Gaurisankar Sa
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Shubhra Ghosh Dastidar
- Bose Institute, Unified Academic Campus, EN 80, Sector V, Bidhannagar, Kolkata, 700091, India.
| | - Tanya Das
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India.
| |
Collapse
|
10
|
Liu Y, Chen X, Zhang W, Yu B, Cen Y, Liu Q, Tang Y, Li S. A CXCR4-targeted immunomodulatory nanomedicine for photodynamic amplified immune checkpoint blockade therapy against breast cancer. Acta Biomater 2025:S1742-7061(25)00223-5. [PMID: 40154764 DOI: 10.1016/j.actbio.2025.03.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
The therapeutic efficacy of immune checkpoint blockade (ICB) is critically compromised by inadequate T lymphocyte infiltration, low T cell-induced pro-inflammatory responses, and the accumulation of immunosuppressive cells within the tumor microenvironment (TME). In this work, a chimeric peptide-engineered immunomodulatory nanomedicine (designated as CXNP-CeBM) is developed for photodynamic amplified ICB therapy against breast cancer. CXNP-CeBM is composed of a CXCR4-targeting peptide ((C16)2-KLGASWHRPDK) loaded with the photosensitizer of Ce6 and the PD-1/PD-L1 inhibitor of BMS8. CXNP-CeBM specifically recognizes CXCR4 on breast cancer, thus suppressing CXCR4-mediated signaling pathways and enhancing the intracellular delivery of therapeutic agents. The photodynamic therapy (PDT) of CXNP-CeBM damages primary tumor cells to initiate immunogenic cell death (ICD), leading to the release of high mobility group box 1 (HMGB1) and the exposure of calreticulin (CRT). Simultaneously, the interruption of CXCR4 signaling reduces tumor fibrosis, promotes T-cell infiltration, and decreases the number of immunosuppressive cells, thereby enhancing the immunotherapeutic effect of ICB. Treatment with CXNP-CeBM would activate systemic anti-tumor immunity, leading to effective inhibition of both primary and lung metastatic tumors, while maintaining low systemic toxicity. This work provides a reliable strategy for the delivery of multi-synergistic agents, effectively activating breast cancer immunity through a multifaceted mechanism. STATEMENT OF SIGNIFICANCE: Although immune checkpoint blockade (ICB) has shown great potential for malignant tumor therapy, its efficacy is compromised by immunosuppressive microenvironments. Herein, a CXCR4-targeted immunomodulatory nanomedicine (CXNP-CeBM) was constructed for photodynamic amplified ICB therapy of breast cancer. CXNP-CeBM could selectively deliver photosensitizers and PD-1/PD-L1 inhibitors to breast cancer cells that overexpressed the chemokine receptor CXCR4, while interrupting CXCR4 signaling to reduce tumor fibrosis, promote T-cell infiltration, and decrease the number of immunosuppressive cells. Moreover, CXNP-CeBM induced photodynamic therapy to trigger immunogenic cell death while downregulating the PD-L1 level to destroy immune evasion mechanisms, thus activating immunological cascades to treat both primary and lung metastatic tumors. This study provided a multi-synergistic strategy for breast cancer immunotherapy through a multifaceted mechanism.
Collapse
Affiliation(s)
- Yibin Liu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Xiayun Chen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Wei Zhang
- School of Chemistry and Chemical Engineering, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Baixue Yu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Yi Cen
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Qianqian Liu
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China
| | - Youzhi Tang
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China.
| | - Shiying Li
- Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, PR China.
| |
Collapse
|
11
|
Chen S, Liu R, Duan S, Zhang B, Wang Y, Li X, Zhao Y, Li Z, Zhou Q, Zhang R, Zhang L, Xu X, Jang R, Zhang J, Li Y, Cai X, Zhang L. Ultrasound-guided percutaneous radiofrequency ablation combined with anti-PD-1 for the treatment of prostate cancer: an experimental study. Front Oncol 2025; 15:1527763. [PMID: 40196732 PMCID: PMC11973081 DOI: 10.3389/fonc.2025.1527763] [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: 11/13/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Background This study seeks to investigate the potential synergistic effects of combining ultrasound-guided percutaneous radiofrequency ablation with anti-PD-1 therapy on prostate cancer, utilizing animal models. Methods A mouse model of prostate cancer was established by subcutaneous injection of 1 × 106 Myc-Cap cells on the right side of FVB mice. When the volume of the tumors reached about 400mm3, the mice were randomly divided into four groups and received corresponding intervention treatments. Among them, Group 1 was the blank control group, Group 2 was the simple anti-PD-1 treatment group, Group 3 was the simple radiofrequency ablation group, and Group 4 is the group that received percutaneous radiofrequency ablation combined with anti-PD-1 therapy under ultrasound guidance. The growth of the tumors was observed in mice after treatment in each group, tumor tissues were collected, and the immune status of the mice was analyzed through flow cytometry, immunohistochemistry, immunofluorescence, and other methods. Results Compared with other treatment groups, ultrasound-guided percutaneous radiofrequency ablation combined with anti-PD-1 therapy significantly reduced the weight and volume of the tumors, demonstrating more effective tumor suppression. At the same time, combination therapy can promote the aggregation of T-cells within the tumor and increase the proportion of cytotoxic T-cells, increase the proportion of M1 macrophages and iNOS expression, and decrease the proportion of M2 macrophages and Arg expression in the local area of the tumors. Conclusion Local ablation can improve the therapeutic effect of PD-1 monoclonal antibody. Our preliminary results suggest that ultrasound-guided percutaneous radiofrequency ablation, in combination with anti-PD-1 treatment, produces synergistic effects. These effects may be driven by changes in immune cell populations within the tumor's immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Si Chen
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ruiqing Liu
- Department of Interventional Therapy, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
- Henan Provincial International Joint Laboratory of Ultrasonic Nanotechnology and Artificial Intelligence in Precision Theragnostic Systems, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Shaobo Duan
- Department of Ultrasound, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Henan University People’s Hospital, Zhengzhou, Henan, China
- Henan Provincial International Joint Laboratory of Ultrasonic Nanotechnology and Artificial Intelligence in Precision Theragnostic Systems, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Beibei Zhang
- Henan Provincial International Joint Laboratory of Ultrasonic Nanotechnology and Artificial Intelligence in Precision Theragnostic Systems, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Yuzhou Wang
- Henan Provincial International Joint Laboratory of Ultrasonic Nanotechnology and Artificial Intelligence in Precision Theragnostic Systems, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Xiaoxiao Li
- Henan University People’s Hospital, Henan Provincial People’s Hospital, Henan University, Zhengzhou, Henan, China
| | - Yingying Zhao
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Zesheng Li
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Qi Zhou
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Rui Zhang
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Linlin Zhang
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaoxia Xu
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Ru Jang
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Juan Zhang
- Zhengzhou University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yaqiong Li
- Henan Provincial International Joint Laboratory of Ultrasonic Nanotechnology and Artificial Intelligence in Precision Theragnostic Systems, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Xiguo Cai
- Henan Provincial Clinical Research Center for Rehabilitation Medicine, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Lianzhong Zhang
- Department of Ultrasound, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Henan University People’s Hospital, Zhengzhou, Henan, China
- Henan Provincial International Joint Laboratory of Ultrasonic Nanotechnology and Artificial Intelligence in Precision Theragnostic Systems, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
- Henan Provincial Clinical Research Center for Rehabilitation Medicine, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| |
Collapse
|
12
|
Wang X, Li Y, Li Y, Wang X, Song H, Wang Y, Huang C, Mao C, Wang L, Zhong C, Yu D, Xia Z, Feng Y, Duan J, Liu Y, Ou J, Luo C, Mai W, Hong H, Cai W, Zheng L, Trempe JF, Fon EA, Liao J, Yi W, Chen J. AMPK-dependent Parkin activation suppresses macrophage antigen presentation to promote tumor progression. SCIENCE ADVANCES 2025; 11:eadn8402. [PMID: 40117357 PMCID: PMC11927615 DOI: 10.1126/sciadv.adn8402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 02/18/2025] [Indexed: 03/23/2025]
Abstract
The constrained cross-talk between myeloid cells and T cells in the tumor immune microenvironment (TIME) restricts cancer immunotherapy efficacy, whereas the underlying mechanism remains elusive. Parkin, an E3 ubiquitin ligase renowned for mitochondrial quality control, has emerged as a regulator of immune response. Here, we show that both systemic and macrophage-specific ablations of Parkin in mice lead to attenuated tumor progression and prolonged mouse survival. By single-cell RNA-seq and flow cytometry, we demonstrate that Parkin deficiency reshapes the TIME through activating both innate and adaptive immunities to control tumor progression and recurrence. Mechanistically, Parkin activation by AMP-activated protein kinase rather than PTEN-induced kinase 1 mediated major histocompatibility complex I down-regulation on macrophages via Autophagy related 5-dependent autophagy. Furthermore, Parkin deletion synergizes with immune checkpoint blockade treatment and Park2-/- signature aids in predicting the prognosis of patients with solid tumor. Our findings uncover Parkin's involvement in suppressing macrophage antigen presentation for coordinating the cross-talk between macrophages and T cells.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Jinfeng Laboratory, Chongqing, China
| | - Yiyi Li
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yan Li
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiumei Wang
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Jinfeng Laboratory, Chongqing, China
| | - Hongrui Song
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yingzhao Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chunliu Huang
- Nasopharyngeal Carcinoma Center, The Fifth Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Zhuhai, China
| | - Chengzhou Mao
- Department of Anatomy and Histology, Shenzhen University Medical School, Shenzhen, China
| | - Lixiang Wang
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Jinfeng Laboratory, Chongqing, China
| | - Cheng Zhong
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Di Yu
- Frazer Institute, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
- Ian Frazer Centre for Children’s Immunotherapy Research, Child Health Research Centre, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Zijin Xia
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yongyi Feng
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Jingjing Duan
- Department of Anatomy and Neurobiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yujia Liu
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Juanjuan Ou
- Yu-Yue Pathology Research Center, Chongqing, China
- Centre for Translational Research in Cancer, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, No. 55 South Renmin Road, Third Inpatient Building, Chengdu, China
- Department of Oncology, Fuling Central Hospital of Chongqing City, Chongqing, China
| | - Congzhou Luo
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Wenhao Mai
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Hai Hong
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Weibin Cai
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Limin Zheng
- Ministry of Education Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jean-François Trempe
- Department of Pharmacology & Therapeutics and Centre de Recherche en Biologie Structurale, McGill University, Montréal, Canada
| | - Edward A. Fon
- McGill Parkinson Program, Neurodegenerative Diseases Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada
| | - Jing Liao
- GMU-GIBH Joint School of Life Sciences, Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou, China
| | - Wei Yi
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Jun Chen
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Jinfeng Laboratory, Chongqing, China
- Key Laboratory of Tropical Disease Control of the Ministry of Education, Sun Yat-sen University, Guangzhou, China
- Guangdong Engineering & Technology Research Center for Disease-Model Animals, Laboratory Animal Center, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
13
|
Zhou Y, Han W, Feng Y, Wang Y, Liu X, Sun T, Xu J. Revealing gut microbiota biomarkers associated with melanoma immunotherapy response and key bacteria-fungi interaction relationships: evidence from metagenomics, machine learning, and SHAP methodology. Front Immunol 2025; 16:1539653. [PMID: 40170844 PMCID: PMC11959079 DOI: 10.3389/fimmu.2025.1539653] [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: 12/04/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Introduction The gut microbiota is associated with the response to immunotherapy in cutaneous melanoma (CM). However, gut fungal biomarkers and bacterial-fungal interactions have yet to be determined. Methods Metagenomic sequencing data of stool samples collected before immunotherapy from three independent groups of European ancestry CM patients were collected. After characterizing the relative abundances of bacteria and fungi, Linear Discriminant Analysis Effect Size (LEfSe) analysis, Random Forest (RF) model construction, and SHapley Additive exPlanations (SHAP) methodology were applied to identify biomarkers and key bacterial-fungal interactions associated with immunotherapy responders in CM. Results Diversity analysis revealed significant differences in the bacterial and fungal composition between CM immunotherapy responders and non-responders. LEfSe analysis identified 45 bacterial and 4 fungal taxa as potential biomarkers. After constructing the RF model, the AUC of models built using bacterial and fungal data separately were 0.64 and 0.65, respectively. However, when bacterial and fungal data were combined, the AUC of the merged model increased to 0.71. In the merged model, the following taxa were identified as important biomarkers: Romboutsia, Endomicrobium, Aggregatilinea, Candidatus Moduliflexus, Colwellia, Akkermansia, Mucispirillum, and Rutstroemia, which were associated with responders, whereas Zancudomyces was associated with non-responders. Moreover, the positive correlation interaction between Akkermansia and Rutstroemia is considered a key bacterial-fungal interaction associated with CM immunotherapy response. Conclusion Our results provide valuable insights for the enrichment of responders to immunotherapy in CM patients. Moreover, this study highlights the critical role of bacterial-fungal interactions in CM immunotherapy.
Collapse
Affiliation(s)
- Yuhang Zhou
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Wenjie Han
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Yun Feng
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Yue Wang
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
| | - Xiaolin Liu
- Department of Bioinformatics, Kanghui Biotechnology Co., Ltd., Shenyang, China
| | - Tao Sun
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Breast Medicine, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital, Shenyang, China
| | - Junnan Xu
- Department of Breast Medicine 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Pharmacology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China
- Department of Breast Medicine, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital, Shenyang, China
| |
Collapse
|
14
|
Qin S, Hu Y, Luo H, Chu W, Deng R, Ma J. Metal ions and nanomaterials for targeted bone cancer immunotherapy. Front Immunol 2025; 16:1513834. [PMID: 40165969 PMCID: PMC11955472 DOI: 10.3389/fimmu.2025.1513834] [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: 10/19/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Bone cancer remains a significant challenge in oncology, with limited success in current therapeutic approaches, particularly immunotherapy. Emerging research highlights the potential of integrating metal ions and nanomaterials for targeted immunotherapy in bone cancer. Metal ions, including calcium, magnesium, and zinc, play a significant role in modulating immune responses within the tumor microenvironment, affecting essential pathways necessary for immune activation. Meanwhile, nanomaterials, particularly metallic nanoparticles, offer precise drug delivery and immune system modulation, improving the efficacy of immunotherapeutic agents. This review explores the synergistic effects of metal ion-nanomaterial conjugates, discussing their role in enhancing immune cell activation, particularly T-cells and macrophages, and their potential for controlled drug release. We highlight preclinical advancements in bone cancer treatment using metal ion-responsive nanoparticles, and address current challenges such as biocompatibility and toxicity. Finally, we discuss the future prospects of these technologies in personalized and precision medicine, aiming to revolutionize bone cancer immunotherapy.
Collapse
Affiliation(s)
- Sen Qin
- Department of Orthopedics, The First Affiliated Hospital of YangTze University, Jingzhou, Hubei, China
| | - YaoFeng Hu
- Department of Neurological Care Unit, The First Affiliated Hospital of YangTze University, Jingzhou, Hubei, China
| | - HuaSong Luo
- Department of Orthopedics, The First Affiliated Hospital of YangTze University, Jingzhou, Hubei, China
| | - Wei Chu
- Department of Orthopedics, The First Affiliated Hospital of YangTze University, Jingzhou, Hubei, China
| | - RuCui Deng
- Department of Neurological Care Unit, The First Affiliated Hospital of YangTze University, Jingzhou, Hubei, China
| | - JinLiang Ma
- Department of Orthopedics, The First Affiliated Hospital of YangTze University, Jingzhou, Hubei, China
| |
Collapse
|
15
|
Lin Y, Xie M, Lau HCH, Zeng R, Zhang R, Wang L, Li Q, Wang Y, Chen D, Jiang L, Damsky W, Yu J. Effects of gut microbiota on immune checkpoint inhibitors in multi-cancer and as microbial biomarkers for predicting therapeutic response. MED 2025; 6:100530. [PMID: 39515321 DOI: 10.1016/j.medj.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 04/16/2024] [Accepted: 10/09/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Gut bacteria are related to immune checkpoint inhibitors (ICIs). However, there is inconsistency in ICI-associated species, while the role of non-bacterial microbes in immunotherapy remains elusive. Here, we evaluated the association of trans-kingdom microbes with ICIs by multi-cohort multi-cancer analyses. METHODS We retrieved fecal metagenomes from 1,359 ICI recipients with four different cancers (metastatic melanoma [MM], non-small cell lung carcinoma [NSCLC], renal cell cancer [RCC], and hepatocellular carcinoma) from 12 published datasets. Microbiota composition was analyzed using the Wilcoxon rank test. The performance of microbial biomarkers in predicting ICI response was assessed by random forest. Key responder-associated microbes were functionally examined in vitro and in mice. FINDINGS Trans-kingdom gut microbiota (bacteria, eukaryotes, viruses, and archaea) was significantly different between ICI responders and non-responders in multi-cancer. Bacteria (Faecalibacterium prausnitzii, Coprococcus comes) and eukaryotes (Nemania serpens, Hyphopichia pseudoburtonii) were consistently enriched in responders of ≥2 cancer types or from ≥3 cohorts, contrasting with the depleted bacterium Hungatella hathewayi. Responder-associated species in each cancer were revealed, such as F. prausnitzii in MM and 6 species in NSCLC. These signature species influenced ICI efficacy by modulating CD8+ T cell activity in vitro and in mice. Moreover, bacterial and eukaryotic biomarkers showed great performance in predicting ICI response in patients from discovery and two validation cohorts (MM: area under the receiver operating characteristic curve [AUROC] = 72.27%-80.19%; NSCLC: AUROC = 72.70%-87.98%; RCC: AUROC = 83.33%-89.58%). CONCLUSIONS This study identified trans-kingdom microbial signatures associated with ICI in multi-cancer and specific cancer types. Trans-kingdom microbial biomarkers are potential predictors of ICI response in patients with cancer. FUNDING Funding information is shown in the acknowledgments.
Collapse
Affiliation(s)
- Yufeng Lin
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Mingxu Xie
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Harry Cheuk-Hay Lau
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ruijie Zeng
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Ruyi Zhang
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Luyao Wang
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Qing Li
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China; Department of Anaesthesia and Intensive Care, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yiwei Wang
- Department of Dermatology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Danyu Chen
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Lanping Jiang
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - William Damsky
- Department of Dermatology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Jun Yu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
16
|
Liu D, Liu L, Zhang X, Zhao X, Li X, Che X, Wu G. Decoding driver and phenotypic genes in cancer: Unveiling the essence behind the phenomenon. Mol Aspects Med 2025; 103:101358. [PMID: 40037122 DOI: 10.1016/j.mam.2025.101358] [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: 10/27/2024] [Revised: 01/25/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Gray hair, widely regarded as a hallmark of aging. While gray hair is associated with aging, reversing this trait through gene targeting does not alter the fundamental biological processes of aging. Similarly, certain oncogenes (such as CXCR4, MMP-related genes, etc.) can serve as markers of tumor behavior, such as malignancy or prognosis, but targeting these genes alone may not lead to tumor regression. We pioneered the name of this class of genes as "phenotypic genes". Historically, cancer genetics research has focused on tumor driver genes, while genes influencing cancer phenotypes have been relatively overlooked. This review explores the critical distinction between driver genes and phenotypic genes in cancer, using the MAPK and PI3K/AKT/mTOR pathways as key examples. We also discuss current research techniques for identifying driver and phenotypic genes, such as whole-genome sequencing (WGS), RNA sequencing (RNA-seq), RNA interference (RNAi), CRISPR-Cas9, and other genomic screening methods, alongside the concept of synthetic lethality in driver genes. The development of these technologies will help develop personalized treatment strategies and precision medicine based on the characteristics of relevant genes. By addressing the gap in discussions on phenotypic genes, this review significantly contributes to clarifying the roles of driver and phenotypic genes, aiming at advancing the field of targeted cancer therapy.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Lei Liu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xiaoman Zhang
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xinming Zhao
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Xiaorui Li
- Department of Oncology, Cancer Hospital of Dalian University of Technology, Shenyang, 110042, China.
| | - Xiangyu Che
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Guangzhen Wu
- Department of Urology, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
17
|
Jing D, Zhang J, Li Z, Yan W, Guo Y. Nanomotors activating both cGAS-STING pathway and immune checkpoint blockade for tumor therapy and bioimaging. Talanta 2025; 284:127258. [PMID: 39586211 DOI: 10.1016/j.talanta.2024.127258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 11/27/2024]
Abstract
Cellular innate immune response is closely related to cGAS-STING pathway and PD-1/PD-L1 immune checkpoint blockade. The lack of tissue penetration of STING agonists and nanomedicines in conventional approaches reduces their immunotherapeutic efficacy. At the same time, because the cGAS-STING signaling pathway is silent in many breast cancer cells, it cannot play its role. To address these challenges, here, we developed a silica nanomotor based on bubble propulsion. Its hollow structure was packed with the photosensitizer Ce6 molecule. Under 808 nm laser irradiation, Ce6 produced 1O2, which lead to intracellular DNA damage and further activated the cGAS-STING pathway, stimulating the maturation of DC cells, and enhancing the tumor infiltration of CD8+ T cells. The nanomotor had an asymmetrical structure. One side of the nanomotor was modified with Pt nanoparticle. This asymmetric modification can catalyze H2O2 in the environment, producing an asymmetric concentration of O2, which realized the bubble driving nanomotor movement and enhances penetration into breast cancer cells of nanomotor. The other side of the nanomotor was modified the LXL-1 aptamer, triphenylphosphine and peptide CLP002. Peptide CLP002 specifically bound residues of PD-L1 interaction with PD-1, blocked the mutual binding between PD-1 and PD-L1, and further improved the immune response ability of tumor infiltrating T cells. In this study, we developed a multi-pronged immunotherapy strategy of intelligent target finding, breaking through the physiological barrier through kinetic energy, accurately intervening the target and bioimaging, providing a new idea for breast cancer cells targeted therapy.
Collapse
Affiliation(s)
- Dan Jing
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Ji Zhang
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Ziyi Li
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Wennan Yan
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Yingshu Guo
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China.
| |
Collapse
|
18
|
Sandhanam K, Tamilanban T, Bhattacharjee B, Manasa K. Exploring miRNA therapies and gut microbiome-enhanced CAR-T cells: advancing frontiers in glioblastoma stem cell targeting. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2169-2207. [PMID: 39382681 DOI: 10.1007/s00210-024-03479-9] [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: 03/21/2024] [Accepted: 09/20/2024] [Indexed: 10/10/2024]
Abstract
Glioblastoma multiforme (GBM) presents a formidable challenge in oncology due to its aggressive nature and resistance to conventional treatments. Recent advancements propose a novel therapeutic strategy combining microRNA-based therapies, chimeric antigen receptor-T (CAR-T) cells, and gut microbiome modulation to target GBM stem cells and transform cancer treatment. MicroRNA therapies show promise in regulating key signalling pathways implicated in GBM progression, offering the potential to disrupt GBM stem cell renewal. CAR-T cell therapy, initially successful in blood cancers, is being adapted to target GBM by genetically engineering T cells to recognise and eliminate GBM stem cell-specific antigens. Despite early successes, challenges like the immunosuppressive tumour microenvironment persist. Additionally, recent research has uncovered a link between the gut microbiome and GBM, suggesting that gut dysbiosis can influence systemic inflammation and immune responses. Novel strategies to modulate the gut microbiome are emerging, enhancing the efficacy of microRNA therapies and CAR-T cell treatments. This combined approach highlights the synergistic potential of these innovative therapies in GBM treatment, aiming to eradicate primary tumours and prevent recurrence, thereby improving patient prognosis and quality of life. Ongoing research and clinical trials are crucial to fully exploit this promising frontier in GBM therapy, offering hope to patients grappling with this devastating disease.
Collapse
Affiliation(s)
- K Sandhanam
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu, 603203, Tamil Nadu, India
| | - T Tamilanban
- Department of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology, Chengalpattu, 603203, Tamil Nadu, India.
| | - Bedanta Bhattacharjee
- Department of Pharmacology, Girijananda Chowdhury University-Tezpur Campus, 784501, Assam, India
| | - K Manasa
- Department of Pharmacology, MNR College of Pharmacy, Sangareddy, 502294, Telangana, India
| |
Collapse
|
19
|
Liu B, Jiang M, Wu Y, Zheng P, Gao X, Wang J. Impact of air pollution on the progress-free survival of non-small cell lung cancer patients with anti-PD-1/PD-L1 immunotherapy: A cohort study. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 368:125683. [PMID: 39809379 DOI: 10.1016/j.envpol.2025.125683] [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: 06/12/2024] [Revised: 12/15/2024] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Air pollution is a well-established risk factor for lung cancer, but limited evidence exists on its impact on the treatment of lung cancer. The objective of this study was to investigate the impact of key pollutants on the efficacy of PD-1/PD-L1 inhibitor immunotherapy in non-small cell lung cancer (NSCLC) patients, thereby providing clinicians with evidence to potentially enhance the efficacy of PD-1 therapy and inform policy decisions for cancer care. To this end, we conducted a study involving 361 NSCLC patients who received PD-1/PD-L1 inhibitor immunotherapy, examining the correlation between air pollution exposure and progression-free survival (PFS) following immunotherapy treatment. Their moving-average ambient levels up to 1 year of PM2.5 and its constituents (organic matter (OM), black carbon (BC), nitrate (NO3-), sulfate (SO42-), and ammonium (NH4+)), as well as ozone (O3) were estimated using the Tracking Air Pollution in China dataset. Cox proportional hazards models were adopted to estimate the effects of exposure to each pollutant on PFS risk for NSCLC. 179 patients obtained the progression of NSCLC. While PM2.5 exposure prior to the immunotherapy was not associated with NSCLC progression, long-term exposure to BC and OM, the important organic components of PM2.5, were significantly associated with a higher risk of NSCLC progression with corresponding hazard ratios (HRs, 95% confidence intervals) of 2.42 (1.39, 4.23) and 2.41 (1.40, 4.14) for 1-year moving average, respectively. Short-term exposure to O3 was also associated with PFS with a HR of 1.64 (1.08, 2.50) for 3-month averaged exposure. Monotonic increasing dose-response relationships were further observed for the associations of BC, OM and O3 with PFS. Our findings imply the need of implementing effective measures for targeted reduction in specific sources of PM2.5 constituents (especially BC and OM) and O3 at different time windows to improve the prognosis of NSCLC patients especially for their PFS.
Collapse
Affiliation(s)
- Bin Liu
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Meijie Jiang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Yuhua Wu
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Pai Zheng
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Xu Gao
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Jinghui Wang
- Cancer Research Center, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China; Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China.
| |
Collapse
|
20
|
Vo DK, Trinh KTL. Polymerase Chain Reaction Chips for Biomarker Discovery and Validation in Drug Development. MICROMACHINES 2025; 16:243. [PMID: 40141854 PMCID: PMC11944077 DOI: 10.3390/mi16030243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/28/2025]
Abstract
Polymerase chain reaction (PCR) chips are advanced, microfluidic platforms that have revolutionized biomarker discovery and validation because of their high sensitivity, specificity, and throughput levels. These chips miniaturize traditional PCR processes for the speed and precision of nucleic acid biomarker detection relevant to advancing drug development. Biomarkers, which are useful in helping to explain disease mechanisms, patient stratification, and therapeutic monitoring, are hard to identify and validate due to the complexity of biological systems and the limitations of traditional techniques. The challenges to which PCR chips respond include high-throughput capabilities coupled with real-time quantitative analysis, enabling researchers to identify novel biomarkers with greater accuracy and reproducibility. More recent design improvements of PCR chips have further expanded their functionality to also include digital and multiplex PCR technologies. Digital PCR chips are ideal for quantifying rare biomarkers, which is essential in oncology and infectious disease research. In contrast, multiplex PCR chips enable simultaneous analysis of multiple targets, therefore simplifying biomarker validation. Furthermore, single-cell PCR chips have made it possible to detect biomarkers at unprecedented resolution, hence revealing heterogeneity within cell populations. PCR chips are transforming drug development, enabling target identification, patient stratification, and therapeutic efficacy assessment. They play a major role in the development of companion diagnostics and, therefore, pave the way for personalized medicine, ensuring that the right patient receives the right treatment. While this tremendously promising technology has exhibited many challenges regarding its scalability, integration with other omics technologies, and conformity with regulatory requirements, many still prevail. Future breakthroughs in chip manufacturing, the integration of artificial intelligence, and multi-omics applications will further expand PCR chip capabilities. PCR chips will not only be important for the acceleration of drug discovery and development but also in raising the bar in improving patient outcomes and, hence, global health care as these technologies continue to mature.
Collapse
Affiliation(s)
- Dang-Khoa Vo
- College of Pharmacy, Gachon University, 191 Hambakmoe-ro, Yeonsu-gu, Incheon 21936, Republic of Korea;
| | - Kieu The Loan Trinh
- Bionano Applications Research Center, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 13120, Gyeonggi-do, Republic of Korea
| |
Collapse
|
21
|
Sun W, Huang S, Sun Z, Zhao Q, Li J, Wen B, Liu J, Deng G, Sun C. Exosome Platform with Three-in-One Functionality of mRNA Therapy, Immune Checkpoint Blockade, and Mild Photothermal Therapy for the Treatment of Triple-Negative Breast Cancer. ACS APPLIED MATERIALS & INTERFACES 2025; 17:10328-10341. [PMID: 39905637 DOI: 10.1021/acsami.4c17804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Immune checkpoint blockade (ICB) has shown promising potential for treating triple-negative breast cancer (TNBC), but its efficacy is limited, mainly due to the "cold," suppressive immune tumor microenvironment (TME). Therefore, improving the TME is crucial for enhancing therapeutic effects against TNBC. Here, we presented a multifunctional nanotherapeutic platform (ALEvs@LNPs) designed to combine cytokine-sensitized mild photothermal therapy, ICB, and interleukin-2 (IL2) mRNA therapy to reprogram the TME, thereby improving the efficacy of ICB therapy in TNBC. Tumor cell-derived exosome-camouflaged lipid nanoparticles with antilymphocyte activation gene-3 (LAG3) were developed to codeliver the photothermal agent IR806, IL2 mRNA, and LAG3 inhibitory antibody (anti-LAG3). Mild photothermal therapy facilitated the reprogramming of "cold" tumors into "hot," thereby enhancing the therapeutic effects of ICB. Meanwhile, ICB also promoted cytokine secretion, increasing the sensitivity of tumor cells to heat. Additionally, IL2 mRNA therapy induced T-cell proliferation and activation, further augmenting the efficacy of ICB. Together, these three therapies established a positive feedback loop that enhanced the therapeutic effects of ICB. This multifunctional nanotherapeutic platform effectively reprogrammed the "cold," suppressive immune TME, offering a promising strategy for TNBC treatment.
Collapse
Affiliation(s)
- Wenwen Sun
- Shandong Provincial Hospital, Shandong University, Jinan 250021, P. R. China
- Yantai Yuhuangding Hospital, Yantai 264000, P. R. China
| | - Shiyun Huang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, P. R. China
| | - Zhihong Sun
- Yantai Yuhuangding Hospital, Yantai 264000, P. R. China
| | - Qi Zhao
- Yantai Yuhuangding Hospital, Yantai 264000, P. R. China
| | - Jie Li
- Yantai Yuhuangding Hospital, Yantai 264000, P. R. China
| | - Baoyu Wen
- Yantai Yuhuangding Hospital, Yantai 264000, P. R. China
| | - Jie Liu
- Yantai Yuhuangding Hospital, Yantai 264000, P. R. China
| | - Guanjun Deng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, P. R. China
| | - Chengming Sun
- Yantai Yuhuangding Hospital, Yantai 264000, P. R. China
| |
Collapse
|
22
|
De Lucia A, Mazzotti L, Gaimari A, Zurlo M, Maltoni R, Cerchione C, Bravaccini S, Delmonte A, Crinò L, Borges de Souza P, Pasini L, Nicolini F, Bianchi F, Juan M, Calderon H, Magnoni C, Gazzola L, Ulivi P, Mazza M. Non-small cell lung cancer and the tumor microenvironment: making headway from targeted therapies to advanced immunotherapy. Front Immunol 2025; 16:1515748. [PMID: 39995659 PMCID: PMC11847692 DOI: 10.3389/fimmu.2025.1515748] [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: 10/23/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
Over the past decades, significant progress has been made in the understanding of non-small cell lung cancer (NSCLC) biology and tumor progression mechanisms, resulting in the development of novel strategies for early detection and wide-ranging care approaches. Since their introduction, over 20 years ago, targeted therapies with tyrosine kinase inhibitors (TKIs) have revolutionized the treatment landscape for NSCLC. Nowadays, targeted therapies remain the gold standard for many patients, but still they suffer from many adverse effects, including unexpected toxicity and intrinsic acquired resistance mutations, which lead to relapse. The adoption of immune checkpoint inhibitors (ICIs) in 2015, has offered exceptional survival benefits for patients without targetable alterations. Despite this notable progress, challenges remain, as not all patients respond favorably to ICIs, and resistance to therapy can develop over time. A crucial factor influencing clinical response to immunotherapy is the tumor microenvironment (TME). The TME is pivotal in orchestrating the interactions between neoplastic cells and the immune system, influencing tumor growth and treatment outcomes. In this review, we discuss how the understanding of this intricate relationship is crucial for the success of immunotherapy and survey the current state of immunotherapy intervention, with a focus on forthcoming and promising chimeric antigen receptor (CAR) T cell therapies in NSCLC. The TME sets major obstacles for CAR-T therapies, creating conditions that suppress the immune response, inducing T cell exhaustion. To enhance treatment efficacy, specific efforts associated with CAR-T cell therapy in NSCLC, should definitely focus TME-related immunosuppression and antigen escape mechanisms, by combining CAR-T cells with immune checkpoint blockades.
Collapse
Affiliation(s)
- Anna De Lucia
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Lucia Mazzotti
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Anna Gaimari
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Matteo Zurlo
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Roberta Maltoni
- Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Claudio Cerchione
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Sara Bravaccini
- Department of Medicine and Surgery, “Kore” University of Enna, Enna, Italy
| | - Angelo Delmonte
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Lucio Crinò
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Patricia Borges de Souza
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Luigi Pasini
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Fabio Nicolini
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Fabrizio Bianchi
- Unit of Cancer Biomarker, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Manel Juan
- Department of Immunology, Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Hugo Calderon
- Department of Immunology, Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Chiara Magnoni
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Luca Gazzola
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Paola Ulivi
- Translational Oncology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Massimiliano Mazza
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
23
|
Suri C, Pande B, Suhasini Sahithi L, Swarnkar S, Khelkar T, Verma HK. Metabolic crossroads: unravelling immune cell dynamics in gastrointestinal cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2025; 8:7. [PMID: 40051496 PMCID: PMC11883236 DOI: 10.20517/cdr.2024.164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 03/09/2025]
Abstract
Metabolic reprogramming within the tumor microenvironment (TME) plays a critical role in driving drug resistance in gastrointestinal cancers (GI), particularly through the pathways of fatty acid oxidation and glycolysis. Cancer cells often rewire their metabolism to sustain growth and reshape the TME, creating conditions such as nutrient depletion, hypoxia, and acidity that impair antitumor immune responses. Immune cells within the TME also undergo metabolic alterations, frequently adopting immunosuppressive phenotypes that promote tumor progression and reduce the efficacy of therapies. The competition for essential nutrients, particularly glucose, between cancer and immune cells compromises the antitumor functions of effector immune cells, such as T cells. Additionally, metabolic by-products like lactate and kynurenine further suppress immune activity and promote immunosuppressive populations, including regulatory T cells and M2 macrophages. Targeting metabolic pathways such as fatty acid oxidation and glycolysis presents new opportunities to overcome drug resistance and improve therapeutic outcomes in GI cancers. Modulating these key pathways has the potential to reinvigorate exhausted immune cells, shift immunosuppressive cells toward antitumor phenotypes, and enhance the effectiveness of immunotherapies and other treatments. Future strategies will require continued research into TME metabolism, the development of novel metabolic inhibitors, and clinical trials evaluating combination therapies. Identifying and validating metabolic biomarkers will also be crucial for patient stratification and treatment monitoring. Insights into metabolic reprogramming in GI cancers may have broader implications across multiple cancer types, offering new avenues for improving cancer treatment.
Collapse
Affiliation(s)
- Chahat Suri
- Department of Oncology, Cross Cancer Institute, University of Alberta, Edmonton AB T6G 1Z2, Canada
| | - Babita Pande
- Department of Physiology, All India Institute of Medical Sciences, Raipur 492099, India
| | | | | | - Tuneer Khelkar
- Department of Botany and Biotechnology, Govt. Kaktiya P G College, Jagdalpur 494001, India
| | - Henu Kumar Verma
- Department of Immunopathology, Institute of Lung Health and Immunity, Comprehensive Pneumology Center, Helmholtz Zentrum, Munich 85764, Germany
| |
Collapse
|
24
|
Cheng B, Lv J, Xiao Y, Song C, Chen J, Shao C. Small molecule inhibitors targeting PD-L1, CTLA4, VISTA, TIM-3, and LAG3 for cancer immunotherapy (2020-2024). Eur J Med Chem 2025; 283:117141. [PMID: 39653621 DOI: 10.1016/j.ejmech.2024.117141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/26/2024] [Accepted: 12/03/2024] [Indexed: 01/03/2025]
Abstract
Cancer immunotherapy, leveraging antibodies, excels in targeting efficacy but faces hurdles in tissue penetration, oral delivery, and prolonged half-life, with costly production and risk of adverse immunogenic effects. In contrast, small molecule immuno-oncology agents provide favorable pharmacokinetic properties and benign toxicity profiles. These agents are well-positioned to address the limitations of antibody-based immunotherapies, augment existing treatment modalities, and achieve synergistic effects when combined with antibodies. This review, for the first time, summarizes the recent advances (2020-2024) in small molecule inhibitors targeting PD-1/PD-L1, CTLA4, VISTA, TIM-3, and LAG3, highlighting rational design, benefits, and potential limitations. It also outlines the prospects for small-molecule immunotherapy.
Collapse
Affiliation(s)
- Binbin Cheng
- Central Laboratory, Wenzhou Medical University Lishui Hospital, Lishui People's Hospital, Lishui, Zhejiang 323000, China; Hubei Polytechnic University, Huangshi, Hubei 435003, China
| | - Jinke Lv
- Foshan Clinical Medical School of Guangzhou University of Chinese Medicine, Foshan 528000, China
| | - Yao Xiao
- Wuchang Hospital Affiliated to Wuhan University of Science and Technology, Wuhan Wuchang Hospital, Wuchang 430063, China
| | - Changshan Song
- Foshan Clinical Medical School of Guangzhou University of Chinese Medicine, Foshan 528000, China.
| | - Jianjun Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Chuxiao Shao
- Central Laboratory, Wenzhou Medical University Lishui Hospital, Lishui People's Hospital, Lishui, Zhejiang 323000, China.
| |
Collapse
|
25
|
Sun XF, Liu C, Chen W, Chen MZ, Tian H. N6-methyladenosine (m6A) RNA methylation of LncRNA LINC01214 accelerates the progression of non-small cell lung cancer (NSCLC) by targeting miR-195-5p/ROCK1 axis. Cytotechnology 2025; 77:29. [PMID: 39744313 PMCID: PMC11685359 DOI: 10.1007/s10616-024-00686-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 12/16/2024] [Indexed: 03/08/2025] Open
Abstract
Long non-coding RNA LINC01214 is reported to be up-regulated in non-small cell lung cancer (NSCLC), however, its function in NSCLC has not been elucidated yet. In our study, we verified that LINC01214 was aberrantly higher in the tumor tissues and cell lines than that in the normal controls, and was relevant to the severity and prognosis of NSCLC through using real-time quantitative PCR. Then, 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide assay and flow cytometry illustrated that knocking down LINC01214 restrained cell proliferation and promoted apoptosis in A549 and H1299 cells. Additionally, western blot results confirmed that LINC01214 silence reduced the protein expression of CDK2, CDK6, CyclinD1 and Bcl2, but increased the protein expression of Bax and Caspase-3. Of note, compared to normal cells, NSCLC cells had higher enrichment level of N6-methyladenosine (m6A) modification of LINC01214, while reducing m6A modification of LINC01214 weakened the stability of LINC01214 and diminished its level in A549 and H1299 through down-regulating methyltransferase METTL3 or overexpressing demethylase ALKBH5. Subsequently, molecular experiments proved that LINC01214 acted as a sponge for miR-195-5p to elevate ROCK1 expression in NSCLC. Furthermore, data from functional recovery experiments showed that elevating miR-195-5p also exerted tumor-suppressive effects in NSCLC; meanwhile, the effects were reversed by overexpressing ROCK1 or inhibiting miR-195-5p. In short, m6A modification-mediated up-regulation of LINC01214 advances cell proliferation and tumorigenesis to promote NSCLC progression through inhibiting miR-195-5p to up-regulate ROCK1.
Collapse
Affiliation(s)
- Xiao-Feng Sun
- Department of Cardiovascular Surgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, 150006 China
| | - Chang Liu
- Future Medical Laboratory, The 2nd Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150086 Heilongjiang China
| | - Wei Chen
- Department of Cardiovascular Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150086 Heilongjiang China
| | - Ming-Zhu Chen
- Department of Cardiovascular Surgery, The 4th Affiliated Hospital of Harbin Medical University, Harbin, 150006 China
| | - Hai Tian
- Future Medical Laboratory, The 2nd Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150086 Heilongjiang China
- Department of Cardiovascular Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Road, Nangang District, Harbin, 150086 Heilongjiang China
| |
Collapse
|
26
|
Chen G, Li W, Ge R, Guo T, Zhang Y, Zhou C, Lin M. NUSAP1 Promotes Immunity and Apoptosis by the SHCBP1/JAK2/STAT3 Phosphorylation Pathway to Induce Dendritic Cell Generation in Hepatocellular Carcinoma. J Immunother 2025; 48:46-57. [PMID: 38980111 PMCID: PMC11753460 DOI: 10.1097/cji.0000000000000531] [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: 02/14/2024] [Accepted: 05/29/2024] [Indexed: 07/10/2024]
Abstract
Hepatocellular carcinoma (HCC) is the most common type of liver cancer and is associated with high morbidity and mortality rates. The aims of this study were to investigate the immune-promoting action of nucleolar and spindle-associated protein 1 (NUSAP1) and identify an immunotherapy target for HCC. The Cancer Genome Atlas (TCGA) was used to analyze interaction molecules and immune correlation. The interaction between NUSAP1 and SHC binding and spindle associated 1 (SHCBP1) was examined. The role of the SHCBP1/Janus kinase 2/signal transducer and activator of transcription 3 (SHCBP1/JAK2/STAT3) pathway in this process was explored. After co-culture with HCC cell lines, the differentiation of peripheral blood mononuclear cells (PBMCs) into dendritic cells (DC) was evaluated by measuring the expression of surface factors CD1a and CD86. Pathological tissues from 50 patients with HCC were collected to validate the results of cell experiments. The expression levels of CD1a and CD86 in tissues were also determined. The results show that NUSAP1 interacted with SHCBP1 and was positively correlated with DC. In HCC cell lines, an interaction was observed between NUSAP1 and SHCBP1. It was verified that NUSAP1 inhibited the JAK2/STAT3 phosphorylation pathway by blocking SHCBP1. After co-culture, the levels of CD1a and CD86 in PBMC were elevated. In the clinical specimens, CD1a and CD86 expression levels were significantly higher in the high-NUSAP1 group versus the low-NUSAP1 group. In Summary, NUSAP1 enhanced immunity by inhibiting the SHCBP1/JAK2/STAT3 phosphorylation pathway and promoted DC generation and HCC apoptosis. NUSAP1 may be a target of immunotherapy for HCC.
Collapse
Affiliation(s)
- Guojie Chen
- Medical School of Nantong University, Nantong, Jiangsu, China
- Clinical Laboratory, Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - WenYa Li
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ruomu Ge
- Clinical Laboratory, Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Ting Guo
- Clinical Laboratory, Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Yuhan Zhang
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Chenglin Zhou
- Laboratory Department, Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| | - Mei Lin
- Clinical Laboratory, Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, Jiangsu, China
| |
Collapse
|
27
|
Li X, He G, Jin H, Xiang X, Li D, Peng R, Tao J, Li X, Wang K, Luo Y, Liu X. Ultrasound-Activated Precise Sono-Immunotherapy for Breast Cancer with Reduced Pulmonary Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407609. [PMID: 39680747 PMCID: PMC11791983 DOI: 10.1002/advs.202407609] [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: 07/06/2024] [Revised: 11/01/2024] [Indexed: 12/18/2024]
Abstract
Immune checkpoint inhibitors have demonstrated remarkable efficacy across various cancer types. However, immune-related adverse events (irAEs) pose a significant challenge in immunotherapy, particularly the associated pneumonia as the primary adverse reaction, which can lead to irreversible pulmonary fibrosis. Additionally, monotherapy with programmed death ligand (PD-L1) inhibitors has shown limited effectiveness. Therefore, to improve the response rate of immunotherapy and reduce pulmonary fibrosis, this study designed and prepared an intelligent nanodrug based on dendritic mesoporous silica nanoparticles (DMSNs) loaded with a sono-sensitive agent protoporphyrin IX (PpIX). Additionally, a reactive oxygen species (ROS) sensitive linker is used to attach the immunotherapeutic drug PD-L1 inhibitor (aPD-L1) to DMSNs via covalent bonds. The external ultrasound (US) activates PpIX to generate ROS, which breaks the linker to release aPD-L1 to induce sonodynamic therapy (SDT) and immunotherapy. This sono-immnotherapy approach demonstrated excellent outcomes in tumor inhibition, eliciting immune responses, and reducing pulmonary fibrosis. Overall, this study offers a new, efficient, and safe method for breast cancer treatment, and expands the application of immunotherapy.
Collapse
Affiliation(s)
- Xiang Li
- Department of Thyroid‐Breast SurgeryThe Fourth Affiliated Hospital of Nanjing Medical University298 Nanpu RoadNanjingJiangsu210032P. R. China
| | - Gao He
- Breast Disease CenterThe First Affiliated Hospital of Nanjing Medical University300 Guangzhou RoadNanjingJiangsu210029P. R. China
- The Afffliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical University366 Taihu RoadTaizhouJiangsu225300P. R. China
| | - Hui Jin
- Department of Breast surgeryThe Affiliated Tumor Hospital of Nantong University30 Tongyang north roadNantongJiangsu226361P. R. China
| | - Xinyu Xiang
- Shanghai Engineering Research Center of Pharmaceutical Intelligent EquipmentShanghai Frontiers Science Research Center for Druggability of Cardiovascular Non‐coding RNAInstitute for Frontier Medical Technology School of Chemistry and Chemical Engineering Shanghai University of Engineering ScienceShanghai201620P. R. China
| | - Dong Li
- Shanghai Engineering Research Center of Pharmaceutical Intelligent EquipmentShanghai Frontiers Science Research Center for Druggability of Cardiovascular Non‐coding RNAInstitute for Frontier Medical Technology School of Chemistry and Chemical Engineering Shanghai University of Engineering ScienceShanghai201620P. R. China
| | - Renmiao Peng
- Shanghai Engineering Research Center of Pharmaceutical Intelligent EquipmentShanghai Frontiers Science Research Center for Druggability of Cardiovascular Non‐coding RNAInstitute for Frontier Medical Technology School of Chemistry and Chemical Engineering Shanghai University of Engineering ScienceShanghai201620P. R. China
| | - Jing Tao
- Department of Thyroid‐Breast SurgeryThe Fourth Affiliated Hospital of Nanjing Medical University298 Nanpu RoadNanjingJiangsu210032P. R. China
| | - Xinping Li
- Department of Thyroid‐Breast SurgeryThe Fourth Affiliated Hospital of Nanjing Medical University298 Nanpu RoadNanjingJiangsu210032P. R. China
| | - Kaiyang Wang
- Shanghai Engineering Research Center of Pharmaceutical Intelligent EquipmentShanghai Frontiers Science Research Center for Druggability of Cardiovascular Non‐coding RNAInstitute for Frontier Medical Technology School of Chemistry and Chemical Engineering Shanghai University of Engineering ScienceShanghai201620P. R. China
| | - Yu Luo
- Shanghai Engineering Research Center of Pharmaceutical Intelligent EquipmentShanghai Frontiers Science Research Center for Druggability of Cardiovascular Non‐coding RNAInstitute for Frontier Medical Technology School of Chemistry and Chemical Engineering Shanghai University of Engineering ScienceShanghai201620P. R. China
| | - Xiaoan Liu
- Breast Disease CenterThe First Affiliated Hospital of Nanjing Medical University300 Guangzhou RoadNanjingJiangsu210029P. R. China
| |
Collapse
|
28
|
Yu B, Shao S, Ma W. Frontiers in pancreatic cancer on biomarkers, microenvironment, and immunotherapy. Cancer Lett 2025; 610:217350. [PMID: 39581219 DOI: 10.1016/j.canlet.2024.217350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Pancreatic cancer remains one of the most challenging malignancies to treat due to its late-stage diagnosis, aggressive progression, and high resistance to existing therapies. This review examines the latest advancements in early detection, and therapeutic strategies, with a focus on emerging biomarkers, tumor microenvironment (TME) modulation, and the integration of artificial intelligence (AI) in data analysis. We highlight promising biomarkers, including microRNAs (miRNAs) and circulating tumor DNA (ctDNA), that offer enhanced sensitivity and specificity for early-stage diagnosis when combined with multi-omics panels. A detailed analysis of the TME reveals how components such as cancer-associated fibroblasts (CAFs), immune cells, and the extracellular matrix (ECM) contribute to therapy resistance by creating immunosuppressive barriers. We also discuss therapeutic interventions that target these TME components, aiming to improve drug delivery and overcome immune evasion. Furthermore, AI-driven analyses are explored for their potential to interpret complex multi-omics data, enabling personalized treatment strategies and real-time monitoring of treatment response. We conclude by identifying key areas for future research, including the clinical validation of biomarkers, regulatory frameworks for AI applications, and equitable access to innovative therapies. This comprehensive approach underscores the need for integrated, personalized strategies to improve outcomes in pancreatic cancer.
Collapse
Affiliation(s)
- Baofa Yu
- Taimei Baofa Cancer Hospital, Dongping, Shandong, 271500, China; Jinan Baofa Cancer Hospital, Jinan, Shandong, 250000, China; Beijing Baofa Cancer Hospital, Beijing, 100010, China; Immune Oncology Systems, Inc, San Diego, CA, 92102, USA.
| | - Shengwen Shao
- Institute of Microbiology and Immunology, Huzhou University School of Medicine, Huzhou, Zhejiang, 313000, China.
| | - Wenxue Ma
- Department of Medicine, Sanford Stem Cell Institute, and Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
29
|
Damaj N, Nassar D, Chamaa B, Kattan J. Immunotherapy in thymic epithelial tumors: an attractive dilemma. Invest New Drugs 2025; 43:69-73. [PMID: 39747775 DOI: 10.1007/s10637-024-01497-1] [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: 11/01/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
Thymomas and thymic carcinomas are the most prevalent tumors that develop in the thymus's epithelial tissue. Thymomas are malignant tumors that develop from the epithelial cells of the thymus and frequently include mixed populations of lymphocytes. In contrast, thymic carcinomas are also tumors of the thymic epithelium, but they are characterized by a lack of lymphocytes, exhibit more aggressive behavior, and are associated with a poorer prognosis. Surgical intervention is the primary approach for managing resectable cases, while advanced, unresectable tumors are treated with platinum-based chemotherapy. The recurrence of the disease can happen months to years after initial treatment. Some patients do benefit from biologic therapies, but there is still a significant need for new treatment options. Immune checkpoint inhibitors have proven safe and clinically effective, improving survival in various cancers. However, their use in thymic cancers is currently limited to treating recurrent thymic carcinoma due to potential immune toxicity risks. This manuscript reviews the current applications of immunotherapy for thymic epithelial tumors and discusses strategies to enhance safety and expand treatment options for patients with these cancers.
Collapse
Affiliation(s)
- Nahed Damaj
- Department of Hematology-Oncology, Faculty of Medicine, Hôtel Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon.
| | - Dany Nassar
- Department of Hematology-Oncology, Faculty of Medicine, Hôtel Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Bilal Chamaa
- Department of General Surgery, Faculty of Medicine, Hôtel Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Joseph Kattan
- Department of Hematology-Oncology, Faculty of Medicine, Hôtel Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| |
Collapse
|
30
|
Xie Z, Shao J, Shen Z, Ye Z, Okada Y, Okuzaki D, Okada N, Tachibana M. HDAC1-3 inhibition triggers NEDD4-mediated CCR2 downregulation and attenuates immunosuppression in myeloid-derived suppressor cells. Cancer Immunol Immunother 2025; 74:81. [PMID: 39891718 PMCID: PMC11787094 DOI: 10.1007/s00262-024-03931-y] [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: 09/02/2024] [Accepted: 12/21/2024] [Indexed: 02/03/2025]
Abstract
Myeloid-derived suppressor cells (MDSCs) play a critical role in cancer progression and resistance, thus representing promising targets for immunotherapy. Despite the established role of histone deacetylases (HDACs) in epigenetic regulation of cell fate and function, their specific impact on MDSCs remains elusive. We sought to investigate the effects and underlying mechanisms of HDAC on MDSCs using various HDAC inhibitors. Our results indicate that HDAC1-3 inhibitors reduce CCR2 expression, a chemokine receptor that mediates the migration of monocytic (M-)MDSCs to tumors and attenuated the immunosuppressive activity of MDSCs. In an orthotropic hepatocellular carcinoma (HCC) murine model, HDAC1-3 inhibitors reduced the infiltration of M-MDSCs, increased the number of natural killer cells in tumors, and suppressed tumor growth. Our results also suggest that HDAC1-3 inhibitors potentiate the antitumor effects of anti-programmed cell death protein 1 antibodies. ATAC-seq and RNA-seq analyses revealed 115 genes epigenetically upregulated by HDAC1-3 inhibitors, primarily linked to transcriptional regulation and ubiquitination. We further elucidated that HDAC1-3 inhibitors facilitate CCR2 protein degradation through ubiquitination-mediated by NEDD4 E3 ligase. Our findings reveal a novel mechanism of action of HDAC1-3 inhibitors in MDSCs and suggest a potential synergistic immunotherapy strategy for clinical benefit in HCC.
Collapse
Affiliation(s)
- Zhiqi Xie
- Wuyi First People's Hospital, Affiliated Hospital, School of Medicine, Hangzhou City University, Hangzhou, 310015, China
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Jinjin Shao
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, 310053, China
| | - Zeren Shen
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Zhichao Ye
- Key Laboratory of Drug Safety Evaluation and Research of Zhejiang Province, Center of Safety Evaluation and Research, Hangzhou Medical College, Hangzhou, 310053, China
| | - Yoshiaki Okada
- Laboratory of Clinical Science and Biomedicine, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Daisuke Okuzaki
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Frontier Research Center, Osaka University, Osaka, 565-0871, Japan
| | - Naoki Okada
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan
| | - Masashi Tachibana
- Project for Vaccine and Immune Regulation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, 565-0871, Japan.
- Laboratory for Context-Dependent Cell Immunology, Department of Biomedical Sciences, College of Life Sciences, Ritsumeikan University, 1-1-1 Nojihigashi, Kusatsu, Shiga, 525-8577, Japan.
| |
Collapse
|
31
|
Zheng F, Li Z, Weng R, Ou B, Zhang T, Zhao R, Feng H. Editorial: Mechanism explorations of enhancing immunotherapeutic sensitivity via mediating immune infiltration and programmed cell death in solid tumor microenvironment. Front Immunol 2025; 16:1559657. [PMID: 39958349 PMCID: PMC11825495 DOI: 10.3389/fimmu.2025.1559657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 01/20/2025] [Indexed: 02/18/2025] Open
Affiliation(s)
- Fang Zheng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Medical Robotics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiliang Li
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruoyu Weng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baochi Ou
- Department of Surgery, First Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Tao Zhang
- Human Oncology and Pathogenesis Program (HOPP), Memorial Sloan Kettering Cancer Center (MSK), New York, NY, United States
| | - Ren Zhao
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Medical Robotics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoran Feng
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Institute of Medical Robotics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
32
|
Das L, Das S. A comprehensive insights of cancer immunotherapy resistance. Med Oncol 2025; 42:57. [PMID: 39883235 DOI: 10.1007/s12032-025-02605-8] [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: 12/07/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025]
Abstract
Cancer is a major global health issue that is usually treated with multiple therapies, such as chemotherapy and targeted therapies like immunotherapy. Immunotherapy is a new and alternative approach to treating various types of cancer that are difficult to treat with other methods. Although immune checkpoint inhibitors have shown promise for long-term efficacy, they have limited effectiveness in common cancer types such as breast, prostate, and lung. Some patients do not respond to immunotherapy, while others develop resistance to the treatment over time, which is classified as primary or acquired resistance. Cancer immunotherapy, specifically immune checkpoint inhibitor-based resistance involves multiple factors such as genes, metabolism, inflammation, and angiogenesis. However, cutting-edge research has identified the mechanisms of immunotherapy resistance and possible solutions. Current research may improve biomarker identification and modify treatment strategies, which will lead to better clinical outcomes. This review provides a comprehensive discussion of the current mechanisms of immunotherapy resistance, related biomarker modulation, and strategies to overcome resistance.
Collapse
Affiliation(s)
- Laavanya Das
- Department of Food and Nutrition, Brainware University, 398, Ramkrishnapur Rd, Barasat, Kolkata, West Bengal, 700125, India
| | - Subhadip Das
- Department of In Vivo Pharmacology, TCG Lifesciences Pvt. Ltd, BN 7, Sector V, Salt Lake City, Kolkata, West Bengal, 700091, India.
| |
Collapse
|
33
|
Yin L, Wang R, Ma X, Jiang K, Hu Y, Zhao X, Zhang L, Wang Z, Long T, Lu M, Li J, Sun Y. Exploring the expression of DLL3 in gastroenteropancreatic neuroendocrine neoplasms and its potential diagnostic value. Sci Rep 2025; 15:3287. [PMID: 39865119 PMCID: PMC11770191 DOI: 10.1038/s41598-025-86237-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 01/09/2025] [Indexed: 01/28/2025] Open
Abstract
Delta-like protein (DLL3) is a novel therapeutic target. DLL3 expression in gastroenteropancreatic neuroendocrine tumors (GEP-NECs) is poorly understood, complicating the distinction between well-differentiated neuroendocrine tumors G3 (NET G3) and poorly differentiated NEC. DLL3 immunohistochemistry (IHC) was performed on 248 primary GEP-NECs, correlating with clinicopathological parameters, NE markers, PD-L1, Ki67 index, and prognosis. Achaete-scute complex-like 1 (ASCL1) IHC was performed on some GEP-NECs. DLL3 IHC was conducted on 36 GEP-NETs, 29 gastric adenocarcinomas (GACs), and metastatic tumors (9 lymph node metastases and 19 distant metastases). DLL3 expression rates were 54.8% in GEP-NECs at the primary site, associated with small cell neuroendocrine carcinoma (SCNEC) (p < 0.001), chemotherapy before baseline (p = 0.015), and at least two NE markers (p = 0.048). DLL3 expression in metastatic GEP-NECs was similar to that of primary tumors. Expression rates in NET G1, NET G2, NET G3, and GACs were 0%, 0%, 15.8%, and 0%, respectively, highlighting DLL3 as a powerful tool for identifying poorly differentiated NEC. DLL3 expression was related to ASCL1 in GEP-NECs, especially in SCNEC. It was not correlated with progression-free survival (PFS) or overall survival(OS), regardless of cutoff value (1%, 50%, 75%). In conclusion, DLL3 targeted therapy may offer potential for the treatment of poorly differentiated NEC of the digestive system, although further studies are needed to validate its efficacy.
Collapse
Affiliation(s)
- L Yin
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Pathology, Peking University Cancer Hospital and Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - R Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Pathology, Peking University Cancer Hospital and Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - X Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - K Jiang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Pathology, Peking University Cancer Hospital and Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - Y Hu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Pathology, Peking University Cancer Hospital and Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - X Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Pathology, Peking University Cancer Hospital and Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China
| | - L Zhang
- Neukio Biotherapeutics, Shanghai, 200131, China
| | - Z Wang
- Neukio Biotherapeutics, Shanghai, 200131, China
| | - T Long
- Neukio Biotherapeutics, Shanghai, 200131, China
| | - M Lu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - J Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China.
| | - Y Sun
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Pathology, Peking University Cancer Hospital and Institute, 52 Fucheng Road, Haidian District, Beijing, 100142, China.
| |
Collapse
|
34
|
Wang X, Shen W, Yao L, Li C, You H, Guo D. Current status and future prospects of molecular imaging in targeting the tumor immune microenvironment. Front Immunol 2025; 16:1518555. [PMID: 39911388 PMCID: PMC11794535 DOI: 10.3389/fimmu.2025.1518555] [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: 10/28/2024] [Accepted: 01/02/2025] [Indexed: 02/07/2025] Open
Abstract
Molecular imaging technologies have significantly transformed cancer research and clinical practice, offering valuable tools for visualizing and understanding the complex tumor immune microenvironment. These technologies allow for the non-invasive examination of key components within the tumor immune microenvironment, including immune cells, cytokines, and stromal cells, providing crucial insights into tumor biology and treatment responses. This paper reviews the latest advancements in molecular imaging, with a focus on its applications in assessing interactions within the tumor immune microenvironment. Additionally, the challenges faced by molecular imaging technologies are discussed, such as the need for highly sensitive and specific imaging agents, issues with data integration, and difficulties in clinical translation. The future outlook emphasizes the potential of molecular imaging to enhance personalized cancer treatment through the integration of artificial intelligence and the development of novel imaging probes. Addressing these challenges is essential to fully realizing the potential of molecular imaging in improving cancer diagnosis, treatment, and patient outcomes.
Collapse
Affiliation(s)
- Xiang Wang
- Department of Radiology, First People’s Hospital of Linping District, Hangzhou, China
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Weifen Shen
- Department of Radiology, First People’s Hospital of Linping District, Hangzhou, China
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lingjun Yao
- Department of Radiology, First People’s Hospital of Linping District, Hangzhou, China
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Li
- Department of Radiology, First People’s Hospital of Linping District, Hangzhou, China
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Huiming You
- Department of Radiology, First People’s Hospital of Linping District, Hangzhou, China
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Duancheng Guo
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Scaletti C, Pratesi S, Bellando Randone S, Di Pietro L, Campochiaro C, Annunziato F, Matucci Cerinic M. The B-cells paradigm in systemic sclerosis: an update on pathophysiology and B-cell-targeted therapies. Clin Exp Immunol 2025; 219:uxae098. [PMID: 39498828 PMCID: PMC11754866 DOI: 10.1093/cei/uxae098] [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: 07/30/2024] [Revised: 10/05/2024] [Accepted: 11/04/2024] [Indexed: 11/07/2024] Open
Abstract
Systemic sclerosis (SSc) is considered a rare autoimmune disease in which there are alterations of both the innate and adaptive immune response resulting in the production of autoantibodies. Abnormalities of the immune system compromise the normal function of blood vessels leading to a vasculopathy manifested by Raynaud's phenomenon, an early sign of SSc . As a consequence of this reactive picture, the disease can evolve leading to tissue fibrosis. Several SSc-specific autoantibodies are currently known and are associated with specific clinical manifestations and prognosis. Although the pathogenetic role of these autoantibodies is still unclear, their production by B cells and plasma cells suggests the importance of these cells in the development of SSc. This review narratively examines B-cell dysfunctions and their role in the pathogenesis of SSc and discusses B-cell-targeted therapies currently used or potentially useful for the management of end-organ complications.
Collapse
Affiliation(s)
- Cristina Scaletti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Sara Pratesi
- Flow Cytometry Diagnostic Center and Immunotherapy, University Hospital Careggi, Florence, Italy
| | - Silvia Bellando Randone
- Department of Clinical and Experimental Medicine, Rheumatology Unit, University of Florence, and Scleroderma Unit, University Hospital Careggi, Florence, Italy
| | - Linda Di Pietro
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Flow Cytometry Diagnostic Center and Immunotherapy, University Hospital Careggi, Florence, Italy
| | - Marco Matucci Cerinic
- Unit of Immunology, Rheumatology, Allergy and Rare diseases (UnIRAR), IRCCS San Raffaele Hospital, Milan, Italy
- Vita Salute San Raffaele University, Milan, Italy
| |
Collapse
|
36
|
Schaub J, Tang SC. Beyond checkpoint inhibitors: the three generations of immunotherapy. Clin Exp Med 2025; 25:43. [PMID: 39888507 PMCID: PMC11785663 DOI: 10.1007/s10238-024-01546-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/23/2024] [Indexed: 02/01/2025]
Abstract
Anti-tumor immunotherapy was rediscovered and rejuvenated in the last two decades with the discovery of CTLA-4, PD-1 and PD-L1 and the roles in inhibiting immune function and tumor evasion of anti-tumor immune response. Following the approval of the first checkpoint inhibitor ipilimumab against CTLA-4 in melanoma in 2011, there has been a rapid development of tumor immunotherapy. Furthermore, additional positive and negative molecules among the T-cell regulatory systems have been identified that that function to fine tune the stimulatory or inhibitory immune cells and modulate their functions (checkpoint modulators). Many strategies are being explored to target macrophages, NK-cells, cytotoxic T-cells, fibroblasts, endothelial cells, cytokines and molecules involved in tumor tolerance and microbiome. Similar to agents that target checkpoint modulators, these newer targets have the potential to synergize with other classes of immunotherapeutic agents and importantly may overcome the resistance to other immunotherapies. In order to better understand the mechanism of action of all major classes of immunotherapy, design clinical trials taking advantage of different types of immunotherapeutic agents and use them rationally in clinical practice either in combination or in sequence, we propose the group all immunotherapies into three generations: with CTLA-4, PD-1 and PD-L1 inhibitors as the first generation, agents that target the checkpoint modulators as the second generation, while those that target TME as the third generation. This review discusses all three generations of immunotherapy in oncology, their mechanism of actions, major clinical trial results and indication, strategies for future clinical trial designs and rational clinical applications.
Collapse
Affiliation(s)
- John Schaub
- Woodlands Medical Specialists, 4724 N Davis Hwy, Pensacola, FL, 32503, USA
| | - Shou-Ching Tang
- LSU-LCMC Cancer Center, LSU School of Medicine, 1700 Tulane Avenue, Room 510, New Orleans, LA, 70112, USA.
| |
Collapse
|
37
|
Olawade DB, Clement David-Olawade A, Adereni T, Egbon E, Teke J, Boussios S. Integrating AI into Cancer Immunotherapy-A Narrative Review of Current Applications and Future Directions. Diseases 2025; 13:24. [PMID: 39851488 PMCID: PMC11764268 DOI: 10.3390/diseases13010024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/12/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
BACKGROUND Cancer remains a leading cause of morbidity and mortality worldwide. Traditional treatments like chemotherapy and radiation often result in significant side effects and varied patient outcomes. Immunotherapy has emerged as a promising alternative, harnessing the immune system to target cancer cells. However, the complexity of immune responses and tumor heterogeneity challenges its effectiveness. OBJECTIVE This mini-narrative review explores the role of artificial intelligence [AI] in enhancing the efficacy of cancer immunotherapy, predicting patient responses, and discovering novel therapeutic targets. METHODS A comprehensive review of the literature was conducted, focusing on studies published between 2010 and 2024 that examined the application of AI in cancer immunotherapy. Databases such as PubMed, Google Scholar, and Web of Science were utilized, and articles were selected based on relevance to the topic. RESULTS AI has significantly contributed to identifying biomarkers that predict immunotherapy efficacy by analyzing genomic, transcriptomic, and proteomic data. It also optimizes combination therapies by predicting the most effective treatment protocols. AI-driven predictive models help assess patient response to immunotherapy, guiding clinical decision-making and minimizing side effects. Additionally, AI facilitates the discovery of novel therapeutic targets, such as neoantigens, enabling the development of personalized immunotherapies. CONCLUSIONS AI holds immense potential in transforming cancer immunotherapy. However, challenges related to data privacy, algorithm transparency, and clinical integration must be addressed. Overcoming these hurdles will likely make AI a central component of future cancer immunotherapy, offering more personalized and effective treatments.
Collapse
Affiliation(s)
- David B. Olawade
- Department of Allied and Public Health, School of Health, Sport and Bioscience, University of East London, London E16 2RD, UK
- Department of Research and Innovation, Medway NHS Foundation Trust, Gillingham, Kent ME7 5NY, UK;
- Department of Public Health, York St John University, London E14 2BA, UK
| | | | - Temitope Adereni
- Department of Public Health, University of Dundee, Dundee DD1 4HN, UK;
| | - Eghosasere Egbon
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Life Science Engineering, FH Technikum, 1200 Vienna, Austria;
| | - Jennifer Teke
- Department of Research and Innovation, Medway NHS Foundation Trust, Gillingham, Kent ME7 5NY, UK;
- Department of Surgery, Medway NHS Foundation Trust, Gillingham, Kent ME7 5NY, UK
- Faculty of Medicine, Health and Social Care, Canterbury Christ Church University, Canterbury, Kent CT1 1QU, UK;
| | - Stergios Boussios
- Faculty of Medicine, Health and Social Care, Canterbury Christ Church University, Canterbury, Kent CT1 1QU, UK;
- Faculty of Life Sciences & Medicine, School of Cancer & Pharmaceutical Sciences, King’s College London, Strand, London WC2R 2LS, UK
- Kent Medway Medical School, University of Kent, Canterbury, Kent CT2 7LX, UK
- AELIA Organization, 9th Km Thessaloniki—Thermi, 57001 Thessaloniki, Greece
- Department of Medical Oncology, Medway NHS Foundation Trust, Gillingham, Kent ME7 5NY, UK
| |
Collapse
|
38
|
Dong Y, Cheng A, Zhou J, Guo J, Liu Y, Li X, Chen M, Hu D, Wu J. PRDX2 induces tumor immune evasion by modulating the HDAC3-Galectin-9 axis in lung adenocarcinoma cells. J Transl Med 2025; 23:81. [PMID: 39825365 PMCID: PMC11740609 DOI: 10.1186/s12967-024-05888-z] [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: 05/09/2024] [Accepted: 11/14/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND PRDX2 is significantly expressed in various cancers and is associated with the proliferation of tumor cells. Nonetheless, the precise mechanism of PRDX2 in tumor immunity remains incompletely understood. This study aims to investigate the impact of PRDX2, which is highly expressed in lung adenocarcinoma, on T cells in the tumor immune microenvironment, and its immune action target to promote the immune escape of lung cancer cells, to provide a theoretical basis for lung adenocarcinoma treatment with PRDX2 as the target. METHODS Mouse animal models to verify the effect of Conoidin A treatment on tumor growth and T cell infiltration. Flow cytometry and Western blot verified tumor cell apoptosis in the in vitro co-culture system as well as granzyme B and perforin expression in T cells. RNA-Seq was used to obtain the downstream immune molecule. si-RNA knockdown of Galectin-9 was co-cultured with T cells in vitro. Immunofluorescence and Western blot verified that PRDX2 regulates Galectin-9 expression through HDAC3. RESULTS PRDX2 expression was negatively correlated with CD8+ T cell expression in LUAD patients. Inhibition of PRDX2 significantly enhanced T-cell killing of LUAD cells and reduced tumor load in both in vitro and in vivo models. Mechanistically, Conoidin A or shRNA_PRDX2 decreased Galectin-9 expression by down-regulating the phosphorylation of HDAC3, consequently enhancing the infiltration and function of CD8+ T cells. CONCLUSIONS This study reveals the role of the PRDX2/HDAC3/Galectin-9 axis in LUAD immune escape and indicates Galectin-9 as a promising target for immunotherapy.
Collapse
Affiliation(s)
- Yunjia Dong
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, 232000, China
| | - Anqi Cheng
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan, Anhui, 232000, China
| | - Jiawei Zhou
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, 232000, China
| | - Jianqiang Guo
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, 232000, China
| | - Yafeng Liu
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- Key Laboratory of Industrial Dust Deep Reduction and Occupational Health and Safety of Anhui Higher Education Institutes, Huainan, Anhui, 232000, China
| | - Xuan Li
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, 232000, China
| | - Maoqian Chen
- School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China
- Anhui Occupational Health and Safety Engineering Laboratory, Huainan, Anhui, 232000, China
| | - Dong Hu
- The First Affiliated Hospital of Anhui University of Science and Technology (Huainan First People's Hospital, School of Medicine), Huainan, Anhui, 232000, China.
- Department of Laboratory Medicine, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 232001, China.
| | - Jing Wu
- Joint Research Center for Occupational Medicine and Health of IHM, School of Medicine, Anhui University of Science and Technology, Huainan, Anhui, 232000, China.
| |
Collapse
|
39
|
Jamali Z, Razipour M, Zargar M, Ghasemnejad-Berenji H, Akrami SM. Ovarian cancer extracellular vesicle biomarkers. Clin Chim Acta 2025; 565:120011. [PMID: 39437983 DOI: 10.1016/j.cca.2024.120011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Ovarian cancer (OC) remains a significant women's health concern due to its high mortality rate and the challenges posed by late detection. Exploring novel biomarkers could lead to earlier, more specific diagnoses and improved survival rates for OC patients. This review focuses on biomarkers associated with extracellular vesicles (EVs) found in various proximal fluids, including urine, ascites, utero-tubal lavage fluid of OC patients. We highlight these proximal fluids as rich sources of potential biomarkers. The review explains the roles of EV biomarkers in ovarian cancer progression and discusses EV-related proteins and miRNAs as potential diagnostic or prognostic indicators and therapeutic targets. Finally, we highlighted the limitations of examining proximal fluids as sources of biomarkers and encourage researchers to proactively pursue innovative solutions to overcome these challenges.
Collapse
Affiliation(s)
- Zeinab Jamali
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Razipour
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Zargar
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hojat Ghasemnejad-Berenji
- Reproductive Health Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Seyed Mohammad Akrami
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
40
|
Feng L, Wang X, Gao Z, Tong Y, Yuan X, Wu T, Xia D, Hu Y. Enhancing Chemotherapy Efficacy via an Autologous Erythrocyte-Anchoring Strategy with a Closed-System Drug-Transfer Device. ACS Biomater Sci Eng 2025; 11:429-441. [PMID: 39696880 DOI: 10.1021/acsbiomaterials.4c02128] [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: 12/20/2024]
Abstract
Chemotherapeutic drugs often fail to localize efficiently to tumors when administered intravenously, causing off-target effects. This study proposes an autologous erythrocyte (ER)-anchoring strategy to improve chemotherapy efficacy and reduce side effects. Utilizing a modified hemodialysis instrument, a closed-system drug-transfer device was developed for autologous ER procurement and immunogenicity mitigation. Doxorubicin (DOX) and indocyanine green (ICG) were encapsulated in autologous ERs and then modified with DSPE-PEG-FA. The final product, DOX-ICG@ER-D, was reintroduced into circulation to enhance chemotherapy. These obtained DOX-ICG@ER-D showed good stability, minimal cardiotoxicity, and extended circulation time. Compared to free DOX, DOX-ICG@ER-D had a higher accumulation of DOX in hepatocellular carcinoma and the release of DOX could be controlled by laser irradiation. Tumor-bearing rats treated by these DOX-ICG@ER-D demonstrated improved antitumor efficacy and reduced cardiotoxicity. Thus, this autologous ER-anchoring strategy offers a promising alternative to intravenous chemotherapy in the clinic.
Collapse
Affiliation(s)
- Lingzi Feng
- College of Engineering and Applied Sciences, MOE Key Laboratory of High Performance Polymer Materials & Technology, Nanjing University, Nanjing 210033, China
- Institute for Applied Research in Public Health, School of Public Health, Nantong University, Nantong, Jiangsu 226019, P. R. China
| | - Xiangqian Wang
- Department of Radiotherapy, Nantong Tumor Hospital & Affiliated Tumor Hospital of Nantong University, Nantong 226361, P. R. China
| | - Ziyi Gao
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu 210029, China
| | - Yuqing Tong
- College of Engineering and Applied Sciences, MOE Key Laboratory of High Performance Polymer Materials & Technology, Nanjing University, Nanjing 210033, China
| | - Xiaopeng Yuan
- Department of Radiotherapy, Nantong Tumor Hospital & Affiliated Tumor Hospital of Nantong University, Nantong 226361, P. R. China
| | - Ting Wu
- Department of Radiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu 210029, China
| | - Donglin Xia
- Institute for Applied Research in Public Health, School of Public Health, Nantong University, Nantong, Jiangsu 226019, P. R. China
| | - Yong Hu
- College of Engineering and Applied Sciences, MOE Key Laboratory of High Performance Polymer Materials & Technology, Nanjing University, Nanjing 210033, China
| |
Collapse
|
41
|
Yuzhakova DV, Sachkova DA, Izosimova AV, Yashin KS, Yusubalieva GM, Baklaushev VP, Mozherov AM, Shcheslavskiy VI, Shirmanova MV. Fluorescence Lifetime Imaging of NAD(P)H in Patients' Lymphocytes: Evaluation of Efficacy of Immunotherapy. Cells 2025; 14:97. [PMID: 39851525 PMCID: PMC11764258 DOI: 10.3390/cells14020097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/30/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND The wide variability in clinical responses to anti-tumor immunotherapy drives the search for personalized strategies. One of the promising approaches is drug screening using patient-derived models composed of tumor and immune cells. In this regard, the selection of an appropriate in vitro model and the choice of cellular response assay are critical for reliable predictions. Fluorescence lifetime imaging microscopy (FLIM) is a powerful, non-destructive tool that enables direct monitoring of cellular metabolism on a label-free basis with a potential to resolve metabolic rearrangements in immune cells associated with their reactivity. OBJECTIVE The aim of the study was to develop a patient-derived glioma explant model enriched by autologous peripheral lymphocytes and explore FLIM of the redox-cofactor NAD(P)H in living lymphocytes to measure the responses of the model to immune checkpoint inhibitors. METHODS The light microscopy, FLIM of NAD(P)H and flow cytometry were used. RESULTS The results demonstrate that the responsive models displayed a significant increase in the free NAD(P)H fraction α1 after treatment, associated with a shift towards glycolysis due to lymphocyte activation. The non-responsive models exhibited no alterations or a decrease in the NAD(P)H α1 after treatment. The FLIM data correlated well with the standard assays of immunotherapy drug response in vitro, including morphological changes, the T-cells activation marker CD69, and the tumor cell proliferation index Ki67. CONCLUSIONS The proposed platform that includes tumor explants co-cultured with lymphocytes and the NAD(P)H FLIM assay represents a promising solution for the patient-specific immunotherapeutic drug screening.
Collapse
Affiliation(s)
- Diana V. Yuzhakova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
| | - Daria A. Sachkova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603950 Nizhny Novgorod, Russia
| | - Anna V. Izosimova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
| | - Konstantin S. Yashin
- Department of Neurosurgery, Privolzsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia;
| | - Gaukhar M. Yusubalieva
- Federal Research and Clinical Center, Federal Medical and Biological Agency, 28 Orekhovy Blvd., 115682 Moscow, Russia; (G.M.Y.); (V.P.B.)
- Laboratory of Molecular Mechanisms of Regeneration and Aging, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova St., 119991 Moscow, Russia
| | - Vladimir P. Baklaushev
- Federal Research and Clinical Center, Federal Medical and Biological Agency, 28 Orekhovy Blvd., 115682 Moscow, Russia; (G.M.Y.); (V.P.B.)
- Laboratory of Molecular Mechanisms of Regeneration and Aging, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova St., 119991 Moscow, Russia
| | - Artem M. Mozherov
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
| | - Vladislav I. Shcheslavskiy
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
- R&D Department, Becker&Hickl GmbH, 7-9 Nunsdorfer Ring, 12277 Berlin, Germany
| | - Marina V. Shirmanova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
| |
Collapse
|
42
|
Chen Y, Qi F, Sun C, Jiang P, Xue X, Yang X, Li X, He X, Wang Y, Zhang T. Navigating the landscape of neoadjuvant immunotherapy for NSCLC: progress and controversies. Ther Adv Med Oncol 2025; 17:17588359241312501. [PMID: 39781239 PMCID: PMC11707791 DOI: 10.1177/17588359241312501] [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: 09/25/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
Recently, attention has increasingly centered on non-small-cell lung cancer (NSCLC) with immune checkpoint inhibitors application. Numerous clinical studies have underscored the potential of immunotherapy in treating resectable NSCLC, highlighting its role in improving patient outcomes. However, despite these promising results, there is ongoing debate regarding the efficacy of immunological combination therapy strategies, the prevalence of treatment-related side effects, the identification of predictive biomarkers, and various other challenges within the neoadjuvant context. Careful consideration is essential to maximize the benefits of immunotherapy for patients with resectable NSCLC. This article offers a detailed overview of recent advancements in neoadjuvant immunotherapy for resectable NSCLC. By examining these developments, we aim to provide new perspectives and valuable insights into the benefits and challenges of applying neoadjuvant immunotherapy in clinical settings.
Collapse
Affiliation(s)
- Yuzhu Chen
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Fei Qi
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Chenhao Sun
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Peng Jiang
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xiangyu Xue
- Department of Biochemistry and Molecular Biology, Heilongjiang Provincial Science and Technology Innovation Team in Higher Education Institutes for Infection and Immunity, Harbin Medical University, Harbin, China
| | - Xiaomei Yang
- Beijing Key Laboratory for Tumor Invasion and Metastasis, Department of Biochemistry and Molecular Biology, Capital Medical University, Beijing, China
- Joint Laboratory for Precision Diagnosis and Treatment Translational Research in Malignant Tumors, Gynecologic Oncology Basic and Clinical Research Laboratory, Capital Medical University, Beijing, China
| | - Xiaomi Li
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China
| | - Xin He
- Department of Nephrology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yishuo Wang
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| | - Tongmei Zhang
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest Hospital, Capital Medical University, No. 9 Beiguan Street, Tongzhou District, Beijing 101149, China
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
43
|
Lim MY, Hong S, Nam YD. Understanding the role of the gut microbiome in solid tumor responses to immune checkpoint inhibitors for personalized therapeutic strategies: a review. Front Immunol 2025; 15:1512683. [PMID: 39840031 PMCID: PMC11747443 DOI: 10.3389/fimmu.2024.1512683] [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: 10/17/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025] Open
Abstract
Immunotherapy, especially immune checkpoint inhibitor (ICI) therapy, has yielded remarkable outcomes for some patients with solid cancers, but others do not respond to these treatments. Recent research has identified the gut microbiota as a key modulator of immune responses, suggesting that its composition is closely linked to responses to ICI therapy in cancer treatment. As a result, the gut microbiome is gaining attention as a potential biomarker for predicting individual responses to ICI therapy and as a target for enhancing treatment efficacy. In this review, we discuss key findings from human observational studies assessing the effect of antibiotic use prior to ICI therapy on outcomes and identifying specific gut bacteria associated with favorable and unfavorable responses. Moreover, we review studies investigating the possibility of patient outcome prediction using machine learning models based on gut microbiome data before starting ICI therapy and clinical trials exploring whether gut microbiota modulation, for example via fecal microbiota transplantation or live biotherapeutic products, can improve results of ICI therapy in patients with cancer. We also briefly discuss the mechanisms through which the gut microbial-derived products influence immunotherapy effectiveness. Further research is necessary to fully understand the complex interactions between the host, gut microbiota, and immunotherapy and to develop personalized strategies that optimize responses to ICI therapy.
Collapse
Affiliation(s)
- Mi Young Lim
- Personalized Diet Research Group, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Republic of Korea
| | - Seungpyo Hong
- Department of Molecular Biology, Jeonbuk National University, Jeonju-si, Jeollabuk-do, Republic of Korea
| | - Young-Do Nam
- Personalized Diet Research Group, Korea Food Research Institute, Wanju-gun, Jeollabuk-do, Republic of Korea
| |
Collapse
|
44
|
Harthorn A, Kuo TH, Torres SW, Lobb RR, Hackel BJ. Expression-Dependent Tumor Pretargeting via Engineered Avidity. Mol Pharm 2025; 22:558-572. [PMID: 39704255 DOI: 10.1021/acs.molpharmaceut.4c01177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024]
Abstract
Selective delivery of therapeutic modalities to tumor cells via binding of tumor-selective cell-surface biomarkers has empowered substantial advances in cancer treatment. Yet, tumor cells generally lack a truly specific biomarker that is present in high density on tumor tissue while being completely absent from healthy tissue. Rather, low but nonzero expression in healthy tissues results in on-target, off-tumor activity with detrimental side effects that constrain the therapeutic window or prevent use altogether. Advanced technologies to enhance the selectivity for tumor targeting are sorely needed. We have engineered a binding platform that is quantitatively dependent upon expression levels, via avidity-driven specificity, rather than binarily reliant on the presence or absence of a biomarker. We systematically varied monomeric binding affinity by engineering affibodies to target carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5) and folate receptor 1 (FolR1). Two identical affibody ligands were tethered, with varying polypeptide linker lengths, to a nanobody that binds Alfa peptide to create a bispecific, trivalent protein for use in pretargeted radioligand therapy. Expression-dependent targeting was achieved in both systems: with 110 nM monomeric affinity to CEACAM5 with a two-amino-acid linker or with 250 nM monomeric affinity for FolR1 and a 10 amino acid linker. The latter bispecific, trivalent achieved over 25-fold differentiation between FolR1high and FolR1low cells in a mixed culture. Similar selectivity was achieved in a size-efficient bivalent molecule lacking a central nanobody. Moreover, the avid bivalent affibody molecule exhibited minimal inhibition by soluble antigen, whereas high-affinity bivalent antibody was inhibited by 97 ± 2%, which is indicative of serum inhibition of shed antigen. This work advances design principles for achieving expression-dependent tumor targeting via low-affinity, high-avidity ligands.
Collapse
Affiliation(s)
- Abbigael Harthorn
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Tse-Han Kuo
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Sarah W Torres
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
| | - Roy R Lobb
- MRB Biotherapeutics, Newton Center, Massachusetts 02459, United States
| | - Benjamin J Hackel
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
- Department of Chemical Engineering and Materials Science, University of Minnesota-Twin Cities, Minneapolis, Minnesota 55455, United States
- MRB Biotherapeutics, Newton Center, Massachusetts 02459, United States
| |
Collapse
|
45
|
Muzaffer U, Nisar N, Ali SI, Kareem O, Paul V. Immunotoxicogenomics: Moving from observation to prediction. IMMUNOTOXICOGENOMICS 2025:181-206. [DOI: 10.1016/b978-0-443-18502-1.00007-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
46
|
Luo J, Zhang Q, Wang S, Zheng L, Liu J, Zhang Y, Wang Y, Wang R, Xiao Z, Li Z. Comprehensive Pan-cancer Analysis of CMPK2 as Biomarker and Prognostic Indicator for Immunotherapy. Curr Cancer Drug Targets 2025; 25:209-229. [PMID: 38486392 DOI: 10.2174/0115680096281451240306062101] [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: 10/12/2023] [Revised: 01/30/2024] [Accepted: 02/14/2024] [Indexed: 02/26/2025]
Abstract
BACKGROUND UMP-CMP kinase 2 (CMPK2) is involved in mitochondrial DNA synthesis, which can be oxidized and released into the cytoplasm in innate immunity. It initiates the assembly of NLRP3 inflammasomes and mediates various pathological processes such as human immunodeficiency virus infection and systemic lupus erythematosus. However, the role of CMPK2 in tumor progression and tumor immunity remains unclear. METHODS We identified CMPK2 expression patterns in the Genotype Tissue-Expression (GTEx), The Cancer Genome Atlas (TCGA), and the Cancer Cell Line Encyclopedia (CCLE) databases. Validation was performed using immunohistochemical staining data from the Human Protein Atlas (HPA) database and qPCR experiments. Receiver operating characteristic curve analysis and Kaplan-Meier survival analysis were conducted to assess the clinical relevance of CMPK2 expression. The Estimation of Stromal and Immune Cells in Malignant Tumor Tissues Using Expression Data (ESTIMATE) algorithm and the Tumor IMmune Estimation Resource (TIMER) database were used to evaluate the correlation between CMPK2 and immune infiltration in tumors. The Tumor Immune Syngeneic Mouse (TISMO) database and other public datasets were utilized to assess the impact of CMPK2 on immune therapy response. MEXPRESS and MethSurv databases were employed to investigate the effects of methylation on CMPK2 expression. RESULTS CMPK2 expression was elevated in 23 cancers and decreased in two cancers. Furthermore, CMPK2 expression had a high diagnostic value for 16 cancers. Elevated CMPK2 expression was associated with lower overall survival (OS), disease-specific survival (DSS), and progression- free interval (PFI) in four cancers. Immune microenvironment-related analysis revealed strong associations between CMPK2 expression and immune cell infiltration, as well as immune checkpoint expression across various tumors. Notably, in four mouse immunotherapy cohorts, CMPK2 expression in treated mouse tumors was higher post-treatment. In five clinical immunotherapy cohorts, patients with high CMPK2 expression show better responses to immunotherapy. Moreover, the methylation level of CMPK2 gene was closely correlated to its expression and tumor prognosis. Among these cancers, the clinical and immunological indications of skin cutaneous melanoma (SKCM) are particularly closely related to CMPK2 expression. CONCLUSION Our analysis preliminarily describes the complex function of CMPK2 in cancer progression and immune microenvironment, highlighting its potential as a diagnostic and therapeutic target for immunotherapy.
Collapse
Affiliation(s)
- Jingyuan Luo
- NHC Key Laboratory of Carcinogenesis, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Clinical Medicine, Xiangya School of Medicine of Central South University, Changsha, China
| | - Qianyue Zhang
- Department of Clinical Medicine, Xiangya School of Medicine of Central South University, Changsha, China
| | - Shutong Wang
- NHC Key Laboratory of Carcinogenesis, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Clinical Medicine, Xiangya School of Medicine of Central South University, Changsha, China
| | - Luojie Zheng
- NHC Key Laboratory of Carcinogenesis, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jie Liu
- NHC Key Laboratory of Carcinogenesis, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Clinical Medicine, Xiangya School of Medicine of Central South University, Changsha, China
| | - Yuchen Zhang
- NHC Key Laboratory of Carcinogenesis, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Clinical Medicine, Xiangya School of Medicine of Central South University, Changsha, China
| | - Yingchen Wang
- NHC Key Laboratory of Carcinogenesis, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Department of Clinical Medicine, Xiangya School of Medicine of Central South University, Changsha, China
| | - Ranran Wang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhigang Xiao
- Department of General Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, China
| | - Zheng Li
- NHC Key Laboratory of Carcinogenesis, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
47
|
Ren XH, Guo T, Xu MF, Huang Y, Liao XR, Qi LJ, Cheng SX. A Multiple Targeting Genome Editing System for Remodulation of Circulating Malignant Cells to Eliminate Cancer Immunosuppression and Restore Immune Responses. Adv Healthc Mater 2025; 14:e2401223. [PMID: 39440615 DOI: 10.1002/adhm.202401223] [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: 05/01/2024] [Revised: 09/29/2024] [Indexed: 10/25/2024]
Abstract
Cancer immunotherapy, which aims to eliminate cancer immunosuppression and reactivate anticancer immunity, holds great promise in oncology treatments. However, it is challenging to accurately study the efficacy of immunotherapy based on human-derived cells through animal experiments due to xenogeneic immune rejection. Herein, a personalized and precise strategy to evaluate the effectiveness of immunotherapy using the blood samples of cancer patients is presented. Through the utilization of multiple cancer-targeting delivery system decorated with the epidermal growth factor receptor (EGFR)-specific aptamer CL4 and the AXL-specific aptamer GL21.T to achieve superior efficiency in delivering the genome editing plasmid for MUC1 knockout, effective modulation on the behavior of circulating malignant cells (CMCs) is realized. After genome editing, both mucin 1 (MUC1) and programmed death-ligand 1 (PD-L1) are significantly downregulated in CMCs. The elimination of immunosuppression results in markedly enhanced secretion of pro-inflammatory anticancer cytokines encompassing interleukins 2, 12, and 15 and interferon-γ by immune cells. The study not only provides a strategy to overcome immunosuppression but also yields critical insights for personalized immunotherapy approaches.
Collapse
Affiliation(s)
- Xiao-He Ren
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230011, China
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Tao Guo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei, Anhui, 230011, China
| | - Ma-Fei Xu
- School of Life Sciences, Anhui Medical University, Hefei, Anhui, 230011, China
| | - Yun Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei, Anhui, 230011, China
| | - Xin-Ru Liao
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Li-Jin Qi
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| | - Si-Xue Cheng
- College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, Hubei, 430072, China
| |
Collapse
|
48
|
Ren J, Ying J, Liu H, Hu S, Li J, Zhou D. Stimulator of Interferon Genes Signal in Lung Cancer Regulates Differentiation of Myeloid-Derived Suppressor Cells in the Tumor Microenvironment Via the Interferon Regulatory Factor 3/NF-κB Pathway. J Interferon Cytokine Res 2025; 45:29-37. [PMID: 39772902 DOI: 10.1089/jir.2024.0150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025] Open
Abstract
Background: This study was designed to explore the action mechanism of stimulator of interferon genes (STING) on the differentiation of myeloid-derived suppressor cells (MDSCs) in the tumor microenvironment of lung cancer. Methods: Bioinformatics analysis yielded a potential pathway for STING to regulate MDSC differentiation, the interferon regulatory factor 3 (IRF3)/NF-κB axis. The transfection efficiency of STING overexpression plasmid and small interfering RNA against IRF3 (siIRF3) was examined by quantitative real-time polymerase chain reaction (qRT-PCR). After transfection, A9 cells were co-cultured with extracted bone marrow cells (BMCs). MDSC differentiation, protein expression of the IRF3/NF-κB pathway, and changes in nuclear translocation of NF-κB were analyzed by flow cytometry, Western blot, and immunofluorescence staining experiments. A transplanted tumor mouse model was used for in vivo experiments. After cyclic diadenyl monophosphate (CDA; STING agonist) treatment, changes in MDSC differentiation and protein expression of the IRF3/NF-κB axis in transplanted tumors were verified by immunohistochemical staining, qRT-PCR, and Western blot. Results: Coculture of A9 cells and BMCs promoted MDSC differentiation, inhibited activation of IRF3/NF-κB signal in A9 cells, and boosted nuclear translocation of NF-κB. However, after the upregulation of STING, IRF3/NF-κB signal was activated, while MDSC differentiation and nuclear translocation of NF-κB were inhibited. SiIRF3 reversed the effects of STING overexpression. In vivo, CDA dampened MDSC differentiation and promoted protein expression of the IRF3/NF-κB axis. Conclusion: STING signal in lung cancer cells inhibits MDSC differentiation through activation of the IRF3/NF-κB pathway.
Collapse
Affiliation(s)
- Jiaojiao Ren
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, China
| | - Jun Ying
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, China
| | - Haijian Liu
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, China
| | - Shanshan Hu
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, China
| | - Jiangdong Li
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, China
| | - Danfei Zhou
- Department of Respiratory and Critical Care Medicine, Ningbo No. 2 Hospital, Ningbo, China
| |
Collapse
|
49
|
Cao Y, Yan W, Yi W, Yin Q, Li Y. Bioengineered therapeutic systems for improving antitumor immunity. Natl Sci Rev 2025; 12:nwae404. [PMID: 40114728 PMCID: PMC11925021 DOI: 10.1093/nsr/nwae404] [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: 08/22/2024] [Revised: 09/29/2024] [Accepted: 11/08/2024] [Indexed: 03/22/2025] Open
Abstract
Immunotherapy, a monumental advancement in antitumor therapy, still yields limited clinical benefits owing to its unguaranteed efficacy and safety. Therapeutic systems derived from cellular, bacterial and viral sources possess inherent properties that are conducive to antitumor immunotherapy. However, crude biomimetic systems have restricted functionality and may produce undesired toxicity. With advances in biotechnology, various toolkits are available to add or subtract certain properties of living organisms to create flexible therapeutic platforms. This review elaborates on the creation of bioengineered systems, via gene editing, synthetic biology and surface engineering, to enhance immunotherapy. The modifying strategies of the systems are discussed, including equipment for navigation and recognition systems to improve therapeutic precision, the introduction of controllable components to control the duration and intensity of treatment, the addition of immunomodulatory components to amplify immune activation, and the removal of toxicity factors to ensure biosafety. Finally, we summarize the advantages of bioengineered immunotherapeutic systems and possible directions for their clinical translation.
Collapse
Affiliation(s)
- Ying Cao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wenlu Yan
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wenzhe Yi
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qi Yin
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
| | - Yaping Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Yantai Key Laboratory of Nanomedicine & Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264000, China
| |
Collapse
|
50
|
Bhaliya KR, Anwer M, Munn A, Wei MQ. New horizons in cancer immunotherapy: The evolving role of R848 and R837 (Review). Mol Clin Oncol 2025; 22:4. [PMID: 39563999 PMCID: PMC11574705 DOI: 10.3892/mco.2024.2799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 08/07/2024] [Indexed: 11/21/2024] Open
Abstract
Therapeutic approaches that increase the efficacy and safety of cancer treatments and improve disease outcomes have been developed worldwide. Immunotherapy uses the body's immune system to inhibit cancerous growth in tissues and organs. Various approaches have been developed to effectively control and inhibit cancerous growth, including checkpoint inhibitors, T-cell transfer therapy, monoclonal antibodies, vaccines and immunomodulators. Toll-like receptors (TLRs) target malignant cells by equipping the immune response. In addition, TLR agonists serve a key role in promoting the innate immune system and initiating antigen-specific T-cell responses. Notably, TLRs and TLR agonists have been utilized as monotherapies or in combination for the treatment of cancer. The present study aimed to review the use of R848 and R837 as TLR agonists, and outline their use as key immunomodulators in cancer therapy.
Collapse
Affiliation(s)
- Krupa R Bhaliya
- Menzies Health Institute, School of Medical Science, Griffith University, Southport, Queensland 4215, Australia
| | - Muneera Anwer
- Menzies Health Institute, School of Medical Science, Griffith University, Southport, Queensland 4215, Australia
| | - Alan Munn
- Menzies Health Institute, School of Medical Science, Griffith University, Southport, Queensland 4215, Australia
| | - Ming Q Wei
- Menzies Health Institute, School of Medical Science, Griffith University, Southport, Queensland 4215, Australia
| |
Collapse
|