1
|
Abdo L, Batista-Silva LR, Bonamino MH. Cost-effective strategies for CAR-T cell therapy manufacturing. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200980. [PMID: 40291594 PMCID: PMC12022644 DOI: 10.1016/j.omton.2025.200980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
CAR-T cell therapy has revolutionized cancer treatment, with approvals for conditions like acute B-leukemia, large B cell lymphoma (LBCL), follicular lymphoma (FL), mantle cell lymphoma (MCL), and multiple myeloma. However, its high costs limit accessibility. Key factors driving these costs include the need for personalized, autologous treatments, transportation to specialized facilities, reliance on viral vectors requiring advanced laboratories, and lengthy cell expansion processes. To address these challenges, alternative strategies aim to simplify and reduce production complexity. Non-viral vectors, such as Sleeping Beauty, piggyBac, and CRISPR, delivered via nanoparticles or electroporation, present promising solutions. These methods could streamline manufacturing, eliminate the need for viral vectors, and reduce associated costs. Furthermore, shortening cell expansion periods and optimizing protocols could significantly accelerate production. An emerging approach involves using genetically edited T cells from healthy donors to create universal CAR-T products capable of treating multiple patients. Finally, decentralized point-of-care (POC) manufacturing of CAR-T cells minimize logistical expenses, eliminating the need for complex infrastructure, and enabling localized production closer to patients. This innovative strategy holds potential for broadening access and reducing costs, representing a step toward democratizing CAR-T therapy. Combined, these advances could make this groundbreaking treatment more feasible for healthcare systems worldwide.
Collapse
Affiliation(s)
- Luiza Abdo
- Cell and Gene Therapy Program, Research Coordination, National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil
| | - Leonardo Ribeiro Batista-Silva
- Cell and Gene Therapy Program, Research Coordination, National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil
| | - Martín Hernán Bonamino
- Cell and Gene Therapy Program, Research Coordination, National Cancer Institute (INCA), Rio de Janeiro 20231-050, Brazil
- Vice-Presidency of Research and Biological Collections (VPPCB), Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro 21040-900, Brazil
| |
Collapse
|
2
|
Saberiyan M, Gholami S, Ejlalidiz M, Rezaeian Manshadi M, Noorabadi P, Hamblin MR. The dual role of chaperone-mediated autophagy in the response and resistance to cancer immunotherapy. Crit Rev Oncol Hematol 2025; 210:104700. [PMID: 40086769 DOI: 10.1016/j.critrevonc.2025.104700] [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: 02/08/2025] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025] Open
Abstract
Cancer immunotherapy has become a revolutionary strategy in oncology, utilizing the host immune system to fight malignancies. Notwithstanding major progress, obstacles such as immune evasion by tumors and the development of resistance still remain. This manuscript examines the function of chaperone-mediated autophagy (CMA) in cancer biology, focusing on its effects on tumor immunotherapy response and resistance. CMA is a selective degradation mechanism for cytosolic proteins, which is crucial for sustaining cellular homeostasis and regulating immune responses. By degrading specific proteins, CMA can either facilitate tumor progression in stressful conditions, or promote tumor suppression by removing oncogenic factors. This double-edged sword highlights the complexity of CMA in cancer progression and its possible effect on treatment results. Here we clarify the molecular mechanisms by which CMA can regulate the immune response and its possible role as a therapeutic target for improving the effectiveness of cancer immunotherapy.
Collapse
Affiliation(s)
- Mohammadreza Saberiyan
- Student Research Committee, Hormozgan University of Medical Sciences, Bandar Abbas, Iran; Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Sarah Gholami
- Young Researchers and Ellie Club, Babol Branch. Islamic Azad University, Babol, Iran
| | - Mahsa Ejlalidiz
- Medical Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadsadegh Rezaeian Manshadi
- Clinical Research Development Center, Imam Hossein Educational Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parisa Noorabadi
- Department of Internal Medicine, School of Medicine, Urmia University of Medical sciences, Urmia, Iran.
| | - Michael R Hamblin
- Laser Research Centre, University of Johannesburg, Doornfontein, South Africa.
| |
Collapse
|
3
|
Allela OQB, Al-Hussainy AF, Sanghvi G, Roopashree R, Kashyap A, Anand DA, Panigrahi R, Garifulina LM, Taher SG, Alwan M, Jawad M, Mushtaq H. Tumor immune evasion and the Let-7 family: insights into mechanisms and therapies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04283-9. [PMID: 40423803 DOI: 10.1007/s00210-025-04283-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 05/09/2025] [Indexed: 05/28/2025]
Abstract
Tumor immune evasion is a complex and adaptive mechanism that allows cancer cells to escape immune detection and destruction, contributing to malignancy progression and poor therapeutic outcomes. This review article explores the integral role of the let-7 family of microRNAs (miRNAs) in mediating tumor immune evasion, particularly how these regulators influence the tumor microenvironment (TME) and immune cell functionality. The let-7 family, known for its tumor-suppressive roles, modulates key immune checkpoints, including PD-L1, and pathways linked to immune response regulation, such as the STAT3/SOCS axis, impacts macrophage polarization and modulates immune cell function. Dysregulation of let-7 miRNAs can enhance tumor immune evasion through mechanisms such as downregulating major histocompatibility complex (MHC) expressions, promoting immunosuppressive cell populations, and manipulating metabolic pathways, which together establish an immunosuppressive TME. Conversely, specific let-7 members show potential in restoring anti-tumor immunity by reversing immune suppression and improving T cell responses. By synthesizing current research, this article underscores the dual role of let-7 in both promoting and inhibiting tumor immune evasion, suggesting their potential as therapeutic targets and biomarkers in cancer immunotherapy. Future studies on the context-dependent roles and advanced delivery systems for let-7-targeting therapies are crucial for enhancing immunotherapeutic efficacy and improving patient outcomes across malignancies.
Collapse
Affiliation(s)
| | | | - Gaurav Sanghvi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Aditya Kashyap
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, 140401, India
| | - D Alex Anand
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Rajashree Panigrahi
- Department of Microbiology, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | | | - Sada Ghalib Taher
- College of Health and Medical Technology, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | - Mariem Alwan
- Pharmacy college, Al-Farahidi University, Baghdad, Iraq
| | - Mahmood Jawad
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | | |
Collapse
|
4
|
Shi Q, Geng J, Han L, Ji X, Li J, Mu Y, Zhao T, Wang L, Jia H. Engineered Salmonella Carrying siRNA-PD-1 Shrinks orthotopically implanted Bladder Cancer in Rats. J Drug Target 2025:1-12. [PMID: 40418584 DOI: 10.1080/1061186x.2025.2512619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/29/2025] [Accepted: 05/20/2025] [Indexed: 05/28/2025]
Abstract
Background Currently, the application of engineered bacteria in tumor treatment has received increasing attention. It has been proved that Bacillus Calmette-Guerin (BCG) can effectively treat bladder cancer (BC). In addition, immune checkpoint blockade is an effective method for tumor treatment. Programmed cell death protein 1 (PD-1), an important immunosuppressive molecule, binds to programmed death ligand receptor 1 (PD-L1) and inhibits the anti-tumor effects of T cells.Methods The plasmid encoding siRNA-PD-1 was constructed, and the rat BC in situ model was established. After the treatment, morphological changes in tumor tissue were detected by HE staining, and the apoptotic cells in tumor tissue were detected by TUNEL. The expression of related proteins was detected by Western blotting, and the proportion of CD4+ and CD8+ T cells in spleen was detected by flow cytometry.Results We found that the engineered Salmonella significantly inhibited the growth and incidence of tumors and increased the apoptosis of tumors. Importantly, engineered Salmonella carrying siRNA-PD-1 enhanced the anti-tumor immune response by inhibiting PD-1 expression and increased the CD8+ T cell infiltration in tumor tissue, while elevated the ratio of CD8+/CD4+ in spleens.Conclusion We demonstrated that engineered Salmonella siRNA-PD-1 exerted significant anti-tumor effects on BC.
Collapse
Affiliation(s)
- Qizhong Shi
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
- Department of Cardiothoracic Surgery, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Jiaxin Geng
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Lulu Han
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
- Department of Urology, Xinxiang Central Hospital, Xinxiang, Henan 453000, P.R.China
| | - Xingchan Ji
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Jiaoran Li
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Yonghui Mu
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
- Department of Pathology, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Tiesuo Zhao
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| | - Lei Wang
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
- Department of Urology, Xinxiang Central Hospital, Xinxiang, Henan 453000, P.R.China
| | - Huijie Jia
- Xinxiang Engineering Technology Research Center of immune checkpoint drug for Liver-Intestinal Tumors, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, Xinxiang Medical University, Xinxiang, Henan 453000, P.R.China
| |
Collapse
|
5
|
Blümke J, Schameitat M, Verma A, Limbecker C, Arlt E, Kessler SM, Kielstein H, Krug S, Bazwinsky-Wutschke I, Haemmerle M. Innate Immunity and Platelets: Unveiling Their Role in Chronic Pancreatitis and Pancreatic Cancer. Cancers (Basel) 2025; 17:1689. [PMID: 40427186 DOI: 10.3390/cancers17101689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 05/10/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive and lethal forms of cancer, characterized by a highly desmoplastic tumor microenvironment. One main risk factor is chronic pancreatitis (CP). Progression of CP to PDAC is greatly influenced by persistent inflammation promoting genomic instability, acinar-ductal metaplasia, and pancreatic intraepithelial neoplasia (PanIN) formation. Components of the extracellular matrix, including immune cells, can modulate this progression phase. This includes cells of the innate immune system, such as natural killer (NK) cells, macrophages, dendritic cells, mast cells, neutrophils, and myeloid-derived suppressor cells (MDSCs), either promoting or inhibiting tumor growth. On one hand, innate immune cells can trigger inflammatory responses that support tumor progression by releasing cytokines and growth factors, fostering tumor cell proliferation, invasion, and metastasis. On the other hand, they can also activate immune surveillance mechanisms, which can limit tumor development. For example, NK cells are cytotoxic innate lymphoid cells that are able to kill tumor cells, and active dendritic cells are crucial for a functioning anti-tumor immune response. In contrast, mast cells and MDSCs rather support a pro-tumorigenic tumor microenvironment that is additionally sustained by platelets. Once thought to play a role in hemostasis only, platelets are now recognized as key players in inflammation and cancer progression. By releasing cytokines, growth factors, and pro-angiogenic mediators, platelets help shape an immunosuppressive microenvironment that promotes fibrotic remodeling, tumor initiation, progression, metastasis, and immune evasion. Neutrophils and macrophages exist in different functional subtypes that can both act pro- and anti-tumorigenic. Understanding the complex interactions between innate immune cells, platelets, and early precursor lesions, as well as PDAC cells, is crucial for developing new therapeutic approaches that can harness the immune and potentially also the coagulation system to target and eliminate tumors, offering hope for improved patient outcomes.
Collapse
Affiliation(s)
- Juliane Blümke
- Institute of Pathology, Section of Experimental Pathology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| | - Moritz Schameitat
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany
| | - Atul Verma
- Department of Internal Medicine I, Medical Faculty, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Celina Limbecker
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany
| | - Elise Arlt
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany
| | - Sonja M Kessler
- Institute of Pharmacy, Experimental Pharmacology for Natural Sciences, Faculty of Natural Sciences, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
| | - Heike Kielstein
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany
| | - Sebastian Krug
- Department of Internal Medicine I, Medical Faculty, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany
- Department of Internal Medicine IV, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Ivonne Bazwinsky-Wutschke
- Institute of Anatomy and Cell Biology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06108 Halle (Saale), Germany
| | - Monika Haemmerle
- Institute of Pathology, Section of Experimental Pathology, Medical Faculty, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany
| |
Collapse
|
6
|
Lin Y, Lin K, Fu Q, Sun X, Wang H, Su L, Xu Y, Liao C. Co-blocking TIGIT and PVRIG Using a Novel Bispecific Antibody Enhances Antitumor Immunity. Mol Cancer Ther 2025; 24:664-677. [PMID: 39851063 DOI: 10.1158/1535-7163.mct-23-0614] [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: 10/30/2023] [Revised: 11/05/2024] [Accepted: 01/22/2025] [Indexed: 01/25/2025]
Abstract
T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT) and poliovirus receptor-related immunoglobulin domain (PVRIG) are immune checkpoints co-expressed on activated T and NK cells, contributing to tumor immune evasion. Simultaneous blockade of these pathways may enhance therapeutic efficacy, positioning them as promising dual targets for cancer immunotherapy. This study aimed to develop a bispecific antibody (BsAb) to co-target TIGIT and PVRIG. Expression of TIGIT and PVRIG was assessed on tumor-infiltrating lymphocytes from patients with various cancers, including non-small cell lung cancer (n = 63) and colorectal cancer (n = 26). The BsAb was engineered by fusing anti-PVRIG nanobodies to the N terminus of anti-TIGIT antibodies. Functional characterization of the BsAb was performed in vitro and in vivo, including assessments of T- and NK-cell activation and cytotoxicity. Pharmacokinetics and safety profiles were evaluated in cynomolgus monkeys. Statistical analyses were conducted using the Student t test. The results showed that the BsAb effectively blocked TIGIT and PVRIG from binding their respective ligands, CD155 and CD112, leading to significant increases in T-cell activation (2.8-fold; P < 0.05) and NK-cell cytotoxicity (1.8-fold; P < 0.05). In vivo, the BsAb demonstrated potent antitumor activity, both as a monotherapy and in combination with anti-PD-1 or anti-PD-L1, in humanized peripheral blood mononuclear cell-reconstituted and transgenic mouse models. Pharmacokinetic studies in cynomolgus monkeys revealed a favorable profile, with no dose-limiting toxicities observed after four repeated doses of 200 mg/kg. These findings provide compelling preclinical evidence for the therapeutic potential of targeting the TIGIT-PVRIG axis with a BsAb. This approach shows promise for enhancing antitumor immunity and warrants further investigation in clinical trials.
Collapse
Affiliation(s)
- Yuan Lin
- Jiangsu Hengrui Pharmaceuticals Co. Ltd., Lianyungang, China
- Shanghai Shengdi Pharmaceuticals Co. Ltd., Shanghai, China
| | - Kan Lin
- Jiangsu Hengrui Pharmaceuticals Co. Ltd., Lianyungang, China
- Shanghai Shengdi Pharmaceuticals Co. Ltd., Shanghai, China
| | - Qiang Fu
- Jiangsu Hengrui Pharmaceuticals Co. Ltd., Lianyungang, China
- Shanghai Shengdi Pharmaceuticals Co. Ltd., Shanghai, China
| | - Xing Sun
- Jiangsu Hengrui Pharmaceuticals Co. Ltd., Lianyungang, China
- Shanghai Shengdi Pharmaceuticals Co. Ltd., Shanghai, China
| | - Huan Wang
- Jiangsu Hengrui Pharmaceuticals Co. Ltd., Lianyungang, China
- Shanghai Shengdi Pharmaceuticals Co. Ltd., Shanghai, China
| | - Lu Su
- Jiangsu Hengrui Pharmaceuticals Co. Ltd., Lianyungang, China
- Shanghai Shengdi Pharmaceuticals Co. Ltd., Shanghai, China
| | - Yanhui Xu
- Jiangsu Hengrui Pharmaceuticals Co. Ltd., Lianyungang, China
- Shanghai Shengdi Pharmaceuticals Co. Ltd., Shanghai, China
| | - Cheng Liao
- Jiangsu Hengrui Pharmaceuticals Co. Ltd., Lianyungang, China
- Shanghai Shengdi Pharmaceuticals Co. Ltd., Shanghai, China
| |
Collapse
|
7
|
Aden D, Zaheer S, Sureka N, Trisal M, Chaurasia JK, Zaheer S. Exploring immune checkpoint inhibitors: Focus on PD-1/PD-L1 axis and beyond. Pathol Res Pract 2025; 269:155864. [PMID: 40068282 DOI: 10.1016/j.prp.2025.155864] [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: 08/31/2024] [Revised: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 04/19/2025]
Abstract
Immunotherapy emerges as a promising approach, marked by recent substantial progress in elucidating how the host immune response impacts tumor development and its sensitivity to various treatments. Immune checkpoint inhibitors have revolutionized cancer therapy by unleashing the power of the immune system to recognize and eradicate tumor cells. Among these, inhibitors targeting the programmed cell death protein 1 (PD-1) and its ligand (PD-L1) have garnered significant attention due to their remarkable clinical efficacy across various malignancies. This review delves into the mechanisms of action, clinical applications, and emerging therapeutic strategies surrounding PD-1/PD-L1 blockade. We explore the intricate interactions between PD-1/PD-L1 and other immune checkpoints, shedding light on combinatorial approaches to enhance treatment outcomes and overcome resistance mechanisms. Furthermore, we discuss the expanding landscape of immune checkpoint inhibitors beyond PD-1/PD-L1, including novel targets such as CTLA-4, LAG-3, TIM-3, and TIGIT. Through a comprehensive analysis of preclinical and clinical studies, we highlight the promise and challenges of immune checkpoint blockade in cancer immunotherapy, paving the way for future advancements in the field.
Collapse
Affiliation(s)
- Durre Aden
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India.
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| | - Monal Trisal
- Department of Pathology, Hamdard Institute of Medical science and research, Jamia Hamdard, New Delhi, India.
| | | | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| |
Collapse
|
8
|
Nazir A, Hussain FHN, Nadeem Hussain TH, Al Dweik R, Raza A. Therapeutic targeting of the host-microbiota-immune axis: implications for precision health. Front Immunol 2025; 16:1570233. [PMID: 40364844 PMCID: PMC12069365 DOI: 10.3389/fimmu.2025.1570233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 03/24/2025] [Indexed: 05/15/2025] Open
Abstract
The human body functions as a complex ecosystem, hosting trillions of microbes that collectively form the microbiome, pivotal in immune system regulation. The host-microbe immunological axis maintains homeostasis and influences key physiological processes, including metabolism, epithelial integrity, and neural function. Recent advancements in microbiome-based therapeutics, including probiotics, prebiotics and fecal microbiota transplantation, offer promising strategies for immune modulation. Microbial therapies leveraging microbial metabolites and engineered bacterial consortia are emerging as novel therapeutic strategies. However, significant challenges remain, including individual microbiome variability, the complexity of host-microbe interactions, and the need for precise mechanistic insights. This review comprehensively examines the host microbiota immunological interactions, elucidating its mechanisms, therapeutic potential, and the future directions of microbiome-based immunomodulation in human health. It will also critically evaluate challenges, limitations, and future directions for microbiome-based precision medicine.
Collapse
Affiliation(s)
- Asiya Nazir
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | | | | | - Rania Al Dweik
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - Afsheen Raza
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| |
Collapse
|
9
|
Naffaa MM, Al-Ewaidat OA, Gogia S, Begiashvili V. Neoantigen-based immunotherapy: advancing precision medicine in cancer and glioblastoma treatment through discovery and innovation. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002313. [PMID: 40309350 PMCID: PMC12040680 DOI: 10.37349/etat.2025.1002313] [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: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Neoantigen-based immunotherapy has emerged as a transformative approach in cancer treatment, offering precision medicine strategies that target tumor-specific antigens derived from genetic, transcriptomic, and proteomic alterations unique to cancer cells. These neoantigens serve as highly specific targets for personalized therapies, promising more effective and tailored treatments. The aim of this article is to explore the advances in neoantigen-based therapies, highlighting successful treatments such as vaccines, tumor-infiltrating lymphocyte (TIL) therapy, T-cell receptor-engineered T cells therapy (TCR-T), and chimeric antigen receptor T cells therapy (CAR-T), particularly in cancer types like glioblastoma (GBM). Advances in technologies such as next-generation sequencing, RNA-based platforms, and CRISPR gene editing have accelerated the identification and validation of neoantigens, moving them closer to clinical application. Despite promising results, challenges such as tumor heterogeneity, immune evasion, and resistance mechanisms persist. The integration of AI-driven tools and multi-omic data has refined neoantigen discovery, while combination therapies are being developed to address issues like immune suppression and scalability. Additionally, the article discusses the ongoing development of personalized immunotherapies targeting tumor mutations, emphasizing the need for continued collaboration between computational and experimental approaches. Ultimately, the integration of cutting-edge technologies in neoantigen research holds the potential to revolutionize cancer care, offering hope for more effective and targeted treatments.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ola A Al-Ewaidat
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Sopiko Gogia
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Valiko Begiashvili
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| |
Collapse
|
10
|
Irannejadrankouhi S, Mivehchi H, Eskandari-Yaghbastlo A, Nejati ST, Emrahoglu S, Nazarian M, Zahedi F, Madani SM, Nabi-Afjadi M. Innovative nanoparticle strategies for treating oral cancers. Med Oncol 2025; 42:182. [PMID: 40285805 DOI: 10.1007/s12032-025-02728-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025]
Abstract
Conventional therapies for oral squamous cell carcinoma (OSCC), a serious worldwide health problem, are frequently constrained by inadequate targeting and serious side effects. Drug delivery systems (DDS) based on nanoparticles provide a possible substitute by improving drug stability, target accuracy, and lowering toxicity. By addressing issues like irregular vasculature and thick tumor matrices, these methods allow for more effective medication administration. For instance, the delivery of cisplatin via liposomes, as opposed to free drug formulations, results in a 40% improvement in tumor suppression. Likewise, compared to traditional techniques, poly (lactic-co-glycolic acid) (PLGA) nanoparticles can produce up to 2.3 times more intertumoral drug accumulation. These platforms have effectively administered natural substances like curcumin and chemotherapeutics like paclitaxel, enhancing therapeutic results while reducing adverse effects. Despite their promise, several types of nanoparticles have drawbacks. For example, PLGA nanoparticles have scaling issues because of their complicated production, whereas liposomes are quickly removed from circulation. In preclinical investigations, functionalized nanoparticles-like EGFR-targeted gold nanoparticles-improve selectivity and effectiveness by obtaining up to 90% receptor binding. By preferentially accumulating in tumors via the increased permeability and retention (EPR) effect, nanoparticles also improve immunotherapy and radiation. Mechanistically, they increase the death of cancer cells by causing DNA damage, interfering with cell division, and producing reactive oxygen species (ROS). There are still issues with toxicity (such as the buildup of metallic nanoparticles in the liver) and large-scale manufacturing. Nevertheless, developments in multifunctional platforms and stimuli-responsive nanoparticles show promise for getting over these obstacles. These developments open the door to more individualized and successful OSCC therapies.
Collapse
Affiliation(s)
| | - Hassan Mivehchi
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | | | | | - Sahand Emrahoglu
- School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Mohammad Nazarian
- Faculty of Dentistry, Belarusion State Medical University, Minsk, Belarus
| | - Farhad Zahedi
- Institute of Molecular Biophysics, Florida State University, 91 Chieftan Way, Tallahassee, FL, 32306, USA
| | - Seyed Mahdi Madani
- Faculty of Dentistry, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, University of Tarbiat Modares, Tehran, Iran.
| |
Collapse
|
11
|
Imani S, Farghadani R, Roozitalab G, Maghsoudloo M, Emadi M, Moradi A, Abedi B, Jabbarzadeh Kaboli P. Reprogramming the breast tumor immune microenvironment: cold-to-hot transition for enhanced immunotherapy. J Exp Clin Cancer Res 2025; 44:131. [PMID: 40281554 PMCID: PMC12032666 DOI: 10.1186/s13046-025-03394-8] [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: 01/16/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
This review discusses reprogramming the breast tumor immune microenvironment from an immunosuppressive cold state to an immunologically active hot state. A complex interplay is revealed, in which the accumulation of metabolic byproducts-such as lactate, reactive oxygen species (ROS), and ammonia-is shown to impair T-cell function and promote tumor immune escape. It is demonstrated that the tumor microenvironment (TME) is dominated by immunosuppressive cytokines, including interleukin-10 (IL-10), transforming growth factorβ (TGFβ), and IL-35. Notably, IL-35 is produced by regulatory T cells and breast cancer cells. The conversion of conventional T cells into IL-35-producing induced regulatory T cells, along with the inhibition of pro-inflammatory cytokine secretion, contributes to the suppression of anti-tumor immunity. It is further demonstrated that key immune checkpoint molecules-such as PD-1, PDL1, CTLA-4, TIM-3, LAG-3, and TIGIT-are upregulated within the TME, leading to Tcell exhaustion and diminished immune responses. The blockade of these checkpoints is shown to restore T-cell functionality and is proposed as a strategy to convert cold tumors into hot ones with robust effector cell infiltration. The therapeutic potential of chimeric antigen receptor (CAR)T cell therapy is also explored, and targeting specific tumor-associated antigens, such as glycoproteins and receptor tyrosine kinases, is highlighted. It is suggested that CART cell efficacy can be enhanced by combining these cells with immune checkpoint inhibitors and other immunomodulatory agents, thereby overcoming the barriers imposed by the immunosuppressive TME. Moreover, the role of the microbiome in regulating estrogen metabolism and systemic inflammation is reviewed. Alterations in the gut microbiota are shown to affect the TME, and microbiome-based interventions are proposed as an additional means to facilitate the cold-to-hot transition. It is concluded that by targeting the metabolic and immunological pathways that underpin immune suppression-through combination strategies involving checkpoint blockade, CART cell therapies, and microbiome modulation-the conversion of the breast TME from cold to hot can be achieved. This reprogramming is anticipated to enhance immune cell infiltration and function, thereby improving the overall efficacy of immunotherapies and leading to better clinical outcomes for breast cancer patients.
Collapse
Affiliation(s)
- Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China.
| | - Reyhaneh Farghadani
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, Subang Jaya, 47500, Selangor Darul Ehsan, Malaysia
| | - Ghazaal Roozitalab
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Mazaher Maghsoudloo
- Key Laboratory of Epigenetics and Oncology, The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Mahdieh Emadi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Atefeh Moradi
- Department of Life Sciences and System Biology, University of Turin, Turin, Italy
| | - Behnaz Abedi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Parham Jabbarzadeh Kaboli
- Department of Biochemistry, Faculty of Medicine, Medical University of Warsaw, Warsaw, 02-091, Poland.
| |
Collapse
|
12
|
Wang Z, He H, Liao X, Yuan L, Sun S, Xu C, Yang X, Zang Q, Peng X, Chen J, Guo X. Discovery of Dual PD-L1/HDAC3 Inhibitors for Tumor Immunotherapy. J Med Chem 2025; 68:8046-8064. [PMID: 40230281 DOI: 10.1021/acs.jmedchem.4c02529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Targeting programmed cell death protein-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) pathway has been considered as one of the most promising strategies for tumor immunotherapy. However, single-target PD-1/PD-L1 inhibitors frequently exhibit limited efficacy, highlighting the urgent need for new therapies. Herein, a series of dual PD-L1/HDAC3 inhibitors were developed through a pharmacophore fusion strategy for the first time. Among them, compound PH3 was identified as the most promising dual PD-L1/HDAC3 inhibitor, with potent PD-1/PD-L1 inhibitory activity (IC50 = 89.4 nM) and selective HDAC3 inhibitory activity (IC50 = 107 nM). Moreover, PH3 exhibited superior in vitro antitumor activities and in vitro immune activation effects. Additionally, PH3 showed potent and dose-dependent antitumor efficacy in the B16-F10 melanoma mouse model without obvious toxicity. Furthermore, PH3 increased the infiltration of CD3+CD8+ and CD3+CD4+ cells in the tumor microenvironment. Collectively, PH3 represented a novel dual PD-L1/HDAC3 inhibitor deserving further investigation as a tumor immunotherapy agent.
Collapse
Affiliation(s)
- Zhijie Wang
- Shenzhen Key Laboratory of Viral Oncology, Ministry of Science and Innovation, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - HaiQi He
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Xiaotong Liao
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Lin Yuan
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Shuding Sun
- Academy of Chinese Medicine Sciences, Henan University of Chinese Medicine, Zhengzhou, Henan 450046, China
| | - Chenglong Xu
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Xixiang Yang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Qinru Zang
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Xiaopeng Peng
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Key Laboratory for Tissue Engineering of Jiangxi Province, School of Pharmacy, Gannan Medical University, Ganzhou 341000, PR China
| | - Jianjun Chen
- School of Pharmaceutical Sciences, Guangdong Provincial Key Laboratory of New Drug Screening, Southern Medical University, Guangzhou 510515, China
| | - Xia Guo
- Shenzhen Key Laboratory of Viral Oncology, Ministry of Science and Innovation, Shenzhen Hospital, Southern Medical University, Shenzhen 518100, China
| |
Collapse
|
13
|
Zalin M, Patel S, Coggins C, Rai V. The Role of Monoclonal Antibodies as Therapeutics in HPV-Related Head and Neck Cancers: An Updated Review. Antibodies (Basel) 2025; 14:37. [PMID: 40407689 PMCID: PMC12101214 DOI: 10.3390/antib14020037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/17/2025] [Accepted: 04/22/2025] [Indexed: 05/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES The increasing prevalence of human papillomavirus (HPV)-positive oropharyngeal squamous cell carcinoma (OPSCC) has necessitated a revaluation of therapeutic strategies. HPV-driven OPSCC differs from HPV-negative OPSCC due to its distinct molecular signatures, increased radiosensitivity, and better prognoses. However, despite these differences, treatment strategies have remained largely uniform, resulting in minimal reductions in morbidity and exposing HPV-positive patients to unnecessary toxicity. Monoclonal antibodies (mAbs) have become a promising therapeutic option due to their ability to target treatment with fewer systemic side effects. Immune checkpoint inhibitors (ICIs) such as pembrolizumab have shown efficacy in enhancing the immune response against tumors, while EGFR inhibitors like cetuximab offer an alternative modality. Current clinical trials aim to refine dosing regimens and identify combination strategies that may enhance therapeutic outcomes. RESULTS Despite promising evidence, several challenges hinder the widespread adoption of mAbs as a standard treatment for HPV-positive OPSCC in clinical practice. This review examines the current role of mAbs in HPV-positive OPSCC treatment, highlighting their limitations and future research directions. CONCLUSIONS Further studies are needed to optimize patient selection, establish standardized treatment protocols, and investigate the long-term benefits of mAb-based therapies in this patient population.
Collapse
Affiliation(s)
- Michael Zalin
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (M.Z.); (S.P.); (C.C.)
| | - Shaan Patel
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (M.Z.); (S.P.); (C.C.)
| | - Carter Coggins
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (M.Z.); (S.P.); (C.C.)
| | - Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, Pomona, CA 91766, USA
| |
Collapse
|
14
|
Kuno S, Pakpian N, Muanprasat C. The potential role of PD-1/PD-L1 small molecule inhibitors in colorectal cancer with different mechanisms of action. Eur J Pharmacol 2025; 992:177351. [PMID: 39922421 DOI: 10.1016/j.ejphar.2025.177351] [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: 09/10/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer-related death worldwide, with increasing incidence in younger ages highlighting the need for new or alternative therapy, of which is immune checkpoint inhibitors. Antibody-based immune checkpoint inhibitors targeting the interaction between programmed cell death protein 1 (PD-1) and programmed death-ligand 1 (PD-L1) have revolutionized cancer treatment, including CRC. However, the low response rate in CRC highlights the need for additional research and innovative therapies. Small molecule inhibitors have risen as another strategy worth exploring, considering their potential to target a wide array of PD-1/PD-L1-related pathways. This review focuses on the potential of small molecule inhibitors targeting the PD-1/PD-L1 axis in CRC. Exploring various classes of small molecule inhibitors, including those that directly block the PD-1/PD-L1 interaction and others that target upstream regulators or downstream signaling pathways involved in PD-1/PD-L1-mediated immune suppression. Additionally, modulation of post-transcriptional and post-translational processes, thereby influencing the expression, stability, or localization of PD-1/PD-L1 proteins to enhance antitumor immunity, provides a multifaceted treatment approach. By disrupting these pathways, these inhibitors can restore immune system activity against tumor cells, offering new hope for overcoming resistance and improving outcomes in CRC patients who do not respond to conventional immune checkpoint inhibitors (ICIs). Integrating these small molecules into CRC treatment strategies could represent a promising advancement in the battle against the challenging disease.
Collapse
Affiliation(s)
- Suhaibee Kuno
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Nattaporn Pakpian
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand
| | - Chatchai Muanprasat
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Samut Prakan, Thailand.
| |
Collapse
|
15
|
Kamrani S, Naseramini R, Khani P, Razavi ZS, Afkhami H, Atashzar MR, Nasri F, Alavimanesh S, Saeidi F, Ronaghi H. Mesenchymal stromal cells in bone marrow niche of patients with multiple myeloma: a double-edged sword. Cancer Cell Int 2025; 25:117. [PMID: 40140850 PMCID: PMC11948648 DOI: 10.1186/s12935-025-03741-x] [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: 10/24/2024] [Accepted: 03/08/2025] [Indexed: 03/28/2025] Open
Abstract
Multiple myeloma (MM) is a hematological malignancy defined by the abnormal proliferation and accumulation of plasma cells (PC) within the bone marrow (BM). While multiple myeloma impacts the bone, it is not classified as a primary bone cancer. The bone marrow microenvironment significantly influences the progression of myeloma and its treatment response. Mesenchymal stromal cells (MSCs) in this environment engage with myeloma cells and other bone marrow components via direct contact and the secretion of soluble factors. This review examines the established roles of MSCs in multiple facets of MM pathology, encompassing their pro-inflammatory functions, contributions to tumor epigenetics, effects on immune checkpoint inhibitors (ICIs), influence on reprogramming, chemotherapy resistance, and senescence. This review investigates the role of MSCs in the development and progression of MM.
Collapse
Affiliation(s)
- Sina Kamrani
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Reza Naseramini
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pouria Khani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Zahra Sadat Razavi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Mohammad Reza Atashzar
- Department of Immunology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Farzad Nasri
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sajad Alavimanesh
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Farzane Saeidi
- Department of Medical Genetics, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Hossein Ronaghi
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran.
| |
Collapse
|
16
|
Yueh PF, Chiang IT, Weng YS, Liu YC, Wong RCB, Chen CY, Hsu JBK, Jeng LB, Shyu WC, Hsu FT. Innovative dual-gene delivery platform using miR-124 and PD-1 via umbilical cord mesenchymal stem cells and exosome for glioblastoma therapy. J Exp Clin Cancer Res 2025; 44:107. [PMID: 40134003 PMCID: PMC11934454 DOI: 10.1186/s13046-025-03336-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 02/18/2025] [Indexed: 03/27/2025] Open
Abstract
Addressing the challenges of identifying suitable targets and effective delivery strategies is critical in pursuing therapeutic solutions for glioblastoma (GBM). This study focuses on the therapeutic potential of microRNA-124 (miR-124), known for its tumor-suppressing properties, by investigating its ability to target key oncogenic pathways in GBM. The results reveal that CDK4 and CDK6-cyclin-dependent kinases that promote cell cycle progression-are significantly overexpressed in GBM brain samples, underscoring their role in tumor proliferation and identifying them as critical targets for miR-124 intervention. However, delivering miRNA-based therapies remains a major obstacle due to the instability of RNA molecules and the difficulty in achieving targeted, efficient delivery. To address these issues, this research introduces an innovative, non-viral dual-gene delivery platform that utilizes umbilical cord mesenchymal stem cells (UMSCs) and their exosomes to transport miR-124 and programmed cell death protein-1 (PD-1). The efficacy of this dual-gene delivery system was validated using an orthotopic GBM model, which closely mimics the tumor microenvironment seen in patients. Experimental results demonstrate that the UMSC/miR-124-PD-1 complex and its exosomes successfully induce apoptosis in GBM cells, significantly inhibiting tumor growth. Notably, these treatments show minimal cytotoxic effects on normal glial cells, highlighting their safety and selectivity. Moreover, the study highlights the immunomodulatory properties of UMSC/miR-124-PD-1 and its exosomes, enhancing the activation of immune cells such as T cells and dendritic cells, while reducing immunosuppressive cells populations like regulatory T cells and myeloid-derived suppressor cells. The orchestrated dual-gene delivery system by UMSCs and exosomes showcased targeted tumor inhibition and positive immune modulation, emphasizing its potential as a promising therapeutic approach for GBM.
Collapse
Affiliation(s)
- Po-Fu Yueh
- Institute of Traditional Medicine, School of Medicine, National Yang Ming Chiao Tung University, 112, Taipei, Beitou, Taiwan (ROC)
| | - I-Tsang Chiang
- Research assistant Center, Show Chwan Memorial Hospital, 500, Changhua, Taiwan (ROC)
- Department of Medical Imaging and Radiological Sciences, Central Taiwan University of Science and Technology, 406, Taichung, Taiwan (ROC)
- Department of Radiation Oncology, Show Chwan Memorial Hospital, Changhua, Taiwan (ROC)
| | - Yueh-Shan Weng
- Department of Biological Science and Technology, China Medical University, 406, Taichung, Taiwan (ROC)
| | - Yu-Chang Liu
- Department of Medical Imaging and Radiological Sciences, Central Taiwan University of Science and Technology, 406, Taichung, Taiwan (ROC)
- Department of Radiation Oncology, Chang Bing Show Chwan Memorial Hospital, 505, Lukang, Changhua, Taiwan (ROC)
- LOHAS Naturopathic Medical Center, Chang Bing Show Chwan Memorial Hospital, Lukang, Taiwan (ROC)
- Evidence-Based Medicine Center, Chang Bing Show Chwan Memorial Hospital, Lukang, Taiwan (ROC)
- Center of Quality Management, Chang Bing Show Chwan Memorial Hospital, Lukang, Taiwan (ROC)
| | - Raymond C B Wong
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, 3002, East Melbourne, VIC, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, 3002, East Melbourne, VIC, Australia
| | - Cheng-Yu Chen
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taipei, Taiwan (ROC)
- Department of Medical Imaging, Taipei Medical University Hospital, 112, Taipei, Taiwan (ROC)
| | - Justin Bo-Kai Hsu
- Department of Computer Science and Engineering, Yuan Ze University, 320, Taoyuan, Taiwan (ROC)
| | - Long-Bin Jeng
- Organ Transplantation Center, China Medical University Hospital, 404, Taichung, Taiwan (ROC).
- Cell Therapy Center, China Medical University Hospital, 404, Taichung, Taiwan (ROC).
- School of Medicine, China Medical University, 404, Taichung, Taiwan (ROC).
| | - Woei-Cherng Shyu
- Graduate Institute of Biomedical Sciences, China Medical University, 404, Taichung, Taiwan (ROC).
- Neuroscience and Brain Disease Center, China Medical University, 404, Taichung, Taiwan (ROC).
- Translational Medicine Research Center, Drug Development Center, Department of Neurology, China Medical University & Hospital, 404, Taichung, Taiwan (ROC).
| | - Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, 406, Taichung, Taiwan (ROC).
- Department of Life Sciences, National Central University, 320, Taoyuan, Taiwan (ROC).
| |
Collapse
|
17
|
Tang Z, Chen C, Zhou C, Liu Z, Li T, Zhang Y, Feng Y, Gu C, Li S, Chen J. Insights into tumor-derived exosome inhibition in cancer therapy. Eur J Med Chem 2025; 285:117278. [PMID: 39823808 DOI: 10.1016/j.ejmech.2025.117278] [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: 10/02/2024] [Revised: 01/11/2025] [Accepted: 01/11/2025] [Indexed: 01/20/2025]
Abstract
Exosomes are critical mediators of cell-to-cell communication in physiological and pathological processes, due to their ability to deliver a variety of bioactive molecules. Tumor-derived exosomes (TDEs), in particular, carry carcinogenic molecules that contribute to tumor progression, metastasis, immune escape, and drug resistance. Thus, TDE inhibition has emerged as a promising strategy to combat cancer. In this review, we discuss the key mechanisms of TDE biogenesis and secretion, emphasizing their implications in tumorigenesis and cancer progression. Moreover, we provide an overview of small-molecule TDE inhibitors that target specific biogenesis and/or secretion pathways, highlighting their potential use in cancer treatment. Lastly, we present the existing obstacles and propose corresponding remedies for the future development of TDE inhibitors.
Collapse
Affiliation(s)
- Ziwei Tang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Cheng Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chen Zhou
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, Gainesville, FL, 32610, United States
| | - Zhouyan Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tong Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Ye Zhang
- School of Petrochemical Engineering, Changzhou University, Changzhou, 213164, China.
| | - Yanyan Feng
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chenglei Gu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shijia Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jichao Chen
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
18
|
Dai D, Wang S, Li J, Zhao Y. Exploring radiation resistance-related genes in pancreatic cancer and their impact on patient prognosis and treatment. Front Immunol 2025; 16:1524798. [PMID: 40103813 PMCID: PMC11914796 DOI: 10.3389/fimmu.2025.1524798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Background Pancreatic cancer is a highly lethal disease with increasing incidence worldwide. Despite surgical resection being the main curative option, only a small percentage of patients are eligible for surgery. Radiotherapy, often combined with chemotherapy, remains a critical treatment, especially for locally advanced cases. However, pancreatic cancer's aggressiveness and partial radio resistance lead to frequent local recurrence. Understanding the mechanisms of radiotherapy resistance is crucial to improving patient outcomes. Methods Pancreatic cancer related gene microarray data were downloaded from GEO database to analyze differentially expressed genes before and after radiotherapy using GEO2R online tool. The obtained differentially expressed genes were enriched by GO and KEGG to reveal their biological functions. Key genes were screened by univariate and multivariate Cox regression analysis, and a risk scoring model was constructed, and patients were divided into high-risk group and low-risk group. Subsequently, Kaplan-Meier survival analysis was used to compare the survival differences between the two groups of patients, further analyze the differential genes of the two groups of patients, and evaluate their sensitivity to different drugs. Results Our model identified 10 genes associated with overall survival (OS) in pancreatic cancer. Based on risk scores, patients were categorized into high- and low-risk groups, with significantly different survival outcomes and immune profile characteristics. High-risk patients showed increased expression of pro-inflammatory immune markers and increased sensitivity to specific chemotherapy agents, while low-risk patients had higher expression of immune checkpoints (CD274 and CTLA4), indicating potential sensitivity to targeted immunotherapies. Cross-dataset validation yielded consistent AUC values above 0.77, confirming model stability and predictive accuracy. Conclusion This study provides a scoring model to predict radiotherapy resistance and prognosis in pancreatic cancer, with potential clinical application for patient stratification. The identified immune profiles and drug sensitivity variations between risk groups highlight opportunities for personalized treatment strategies, contributing to improved management and survival outcomes in pancreatic cancer.
Collapse
Affiliation(s)
- Dong Dai
- Department of Nuclear Medicine, Tianjin Cancer Hospital Airport Hospital, National Clinical Research Center for Cancer, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for China, Tianjin, China
| | - Sen Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jiaze Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Yu Zhao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
19
|
Rajesh A, Gurusamy DS, Manikkam R. Evaluating the Tumor Burden, Histological Changes, and Immune Landscape of Breast Cancer Post-neoadjuvant Chemotherapy: Insights From 50 Cases. Cureus 2025; 17:e80258. [PMID: 40061852 PMCID: PMC11890627 DOI: 10.7759/cureus.80258] [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] [Accepted: 03/08/2025] [Indexed: 03/21/2025] Open
Abstract
Breast cancer is a heterogeneous disease with variable responses to neoadjuvant chemotherapy (NACT). Evaluating the histopathological and immune changes in post-NACT breast cancer specimens is crucial for understanding treatment response and guiding further management. This study aims to assess tumor burden using the Residual Cancer Burden (RCB) index, examine histological alterations, evaluate immune activity through tumor-infiltrating lymphocytes (TILs), and analyze proliferative capacity via Ki-67 expression in post-NACT breast cancer specimens. A cross-sectional study of 50 modified radical mastectomy (MRM) specimens post-NACT was conducted. The histopathological analysis included tumor regression changes, stromal and cellular alterations, and nodal involvement. Immune response was assessed by quantifying TILs, and proliferation was measured using the Ki-67 index. Statistical correlations were made between clinicopathological parameters, TILs, and Ki-67 expression. Residual disease was detected in 39 cases (78%), and 11 cases (22%) had no residual disease. Among the 39 cases with residual disease, the majority were classified as RCB II (22 cases, 56%), 16 cases (41%) were classified as RCB III, and one case (3%) was classified as RCB I. Common histological changes post-NACT included fibrosis in 31 cases (62%), necrosis in 19 cases (38%), and infiltration by foamy histiocytes in 16 cases (32%). Malignant epithelial cells more frequently exhibited foamy cytoplasm (16 cases (41%) vs. two cases (5%); p=0.0003), hyperchromatic nucleus (26 cases (67%) vs. six cases (15%); p=0.0001), and prominent nucleoli (26 cases (67%) vs. four cases (10%); p=0.0001) compared to benign cells. Among the 39 cases with residual disease, low TIL and high Ki-67 expression were observed in 20 cases (51%), while 12 cases (32%) showed high TIL and low Ki-67. Residual tumors with high TIL and high Ki-67 (four cases, 10%) and low TIL and low Ki-67 (three cases, 8%) were less common. A significant inverse relationship was found between TIL levels and Ki-67 expression (p=0.0002), as tumors with low TIL were more likely to have high Ki-67 expression (20 cases, 51%), whereas those with high TIL more frequently exhibited low Ki-67 expression (12 cases, 32%). Post-NACT evaluation of tumor burden, immune landscape, and proliferation provides valuable prognostic insights. Integrating RCB, TILs, and Ki-67 into routine pathological assessment may aid patient stratification and guide personalized treatment strategies. Further large-scale studies are needed to validate these findings and improve therapeutic decision-making in breast cancer management.
Collapse
Affiliation(s)
- Arasi Rajesh
- Pathology, Government Medical College, Tuticorin, IND
| | | | | |
Collapse
|
20
|
Li S, Feng M, Wang F, Liu D, Li M, Dai J, Yang Y, Chai Y, Chen W. Mycobacterium tuberculosis infection may increase the degrees of malignancy in lung adenocarcinoma. Front Immunol 2025; 16:1537520. [PMID: 40061944 PMCID: PMC11885956 DOI: 10.3389/fimmu.2025.1537520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 05/13/2025] Open
Abstract
Background The early diagnosis and management of lung adenocarcinoma co-existing with tuberculosis (LAC-TB) presents significant challenges in clinical settings. This is compounded by a paucity of robust clinical evidence elucidating the interactions between these two conditions. Methods This study included 14 patients diagnosed with LAC-TB, with an equal distribution among those with pulmonary tuberculosis (TB) and those with peripheral lymph node TB. Controls included patients with simple TB and those with lung adenocarcinoma (LAC). Histopathologic examinations confirmed typical changes in each group. Immunohistochemistry analyzed immune markers, focusing on PD-L1, while genomic analysis identified differential mutant genes. Results Pathological evaluations showed that LAC-TB and LAC groups expressed TTF-1 and Napsin A in their adenocarcinoma specimens. Notably, a higher proportion of patients in the LAC-TB group had a Ki-67 proliferation index of ≥10%. Subsequent Molecular analyses revealed significant differences in RALGAPA1 gene expression, with the LAC-TB group also exhibiting a greater median count of missense mutations, single nucleotide polymorphisms, and overall mutations, suggesting a higher malignancy level than the LAC group. Additionally, the LAC-TB group showed an increased tumor mutational burden, indicating a potentially better response to immunotherapy. Immunohistochemical assessments indicated that Mycobacterium tuberculosis (MTB) infection correlated with reduced infiltration of T cells and CD4+ T cells, alongside an upregulation of PD-L1 expression in LAC. Notably, PD-L1 was strongly expressed in the TB granuloma and surrounding areas. Conclusion Our findings suggest that MTB infection may increase the malignancy of LAC, with the pronounced expression of PD-L1 in granuloma regions constituting a pivotal mechanism underlying this relationship.
Collapse
Affiliation(s)
- Shanshan Li
- Department of Pathology, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| | - Mengru Feng
- Department of Pathology, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| | - Fenghua Wang
- Department of Pathology, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| | - Dongxu Liu
- Department of Pathology, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| | - Mingyan Li
- Department of Pathology, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| | - Jinlong Dai
- Department of Pathology, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| | - Yan Yang
- Department of Radiology, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| | - Yinghui Chai
- Department of Clinical Laboratory, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| | - Wen Chen
- Department of Pathology, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| |
Collapse
|
21
|
Jiramonai L, Liang XJ, Zhu M. Extracellular Vesicle-Based Strategies for Tumor Immunotherapy. Pharmaceutics 2025; 17:257. [PMID: 40006624 PMCID: PMC11859549 DOI: 10.3390/pharmaceutics17020257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/26/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Immunotherapy is one of the most promising approaches for cancer management, as it utilizes the intrinsic immune response to target cancer cells. Normally, the human body uses its immune system as a defense mechanism to detect and eliminate foreign objects, including cancer cells. However, cancers develop a 'switch off' mechanism, known as immune checkpoint proteins, to evade immune surveillance and suppress immune activation. Therefore, significant efforts have been made to develop the strategies for stimulating immune responses against cancers. Among these, the use of extracellular vesicles (EVs) to enhance the anti-tumor immune response has emerged as a particularly promising approach in cancer management. EVs possess several unique properties that elevate the potency in modulating immune responses. This review article provides a comprehensive overview of recent advances in this field, focusing on the strategic usage of EVs to overcome tumor-induced immune tolerance. We discuss the biogenesis and characteristics of EVs, as well as their potential applications in medical contexts. The immune mechanisms within the tumor microenvironment and the strategies employed by cancers to evade immune detection are explored. The roles of EVs in regulating the tumor microenvironment and enhancing immune responses for immunotherapy are also highlighted. Additionally, this article addresses the challenges and future directions for the development of EV-based nanomedicine approaches, aiming to improve cancer immunotherapy outcomes with greater precision and efficacy while minimizing off-target effects.
Collapse
Affiliation(s)
- Luksika Jiramonai
- Chinese Academy of Sciences (CAS), Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS), Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengliang Zhu
- Chinese Academy of Sciences (CAS), Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
22
|
Zhang B, Leung PC, Cho WCS, Wong CK, Wang D. Targeting PI3K signaling in Lung Cancer: advances, challenges and therapeutic opportunities. J Transl Med 2025; 23:184. [PMID: 39953539 PMCID: PMC11829425 DOI: 10.1186/s12967-025-06144-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 01/14/2025] [Indexed: 02/17/2025] Open
Abstract
Lung cancer remains the leading cause of cancer-related mortality globally, necessitating the continual exploration of novel therapeutic targets. The phosphoinositide 3-kinase (PI3K) signaling pathway plays a pivotal role in oncogenic processes, including cell growth, survival, metabolism and immune modulation. This comprehensive review delineates the distinct roles of PI3K subtypes-PI3Kα, PI3Kβ, PI3Kγ and PI3Kδ-in lung cancer pathogenesis and progression. We evaluate the current landscape of PI3K inhibitors, transitioning from non-selective early-generation compounds to isoform-specific agents, highlighting their clinical efficacy, resistance mechanisms and potential combination strategies. Furthermore, the intricate interplay between PI3K signaling and the tumor immune microenvironment is explored, elucidating how PI3K modulation can enhance immunotherapeutic responses. Metabolic reprogramming driven by PI3K signaling is also dissected, revealing vulnerabilities that can be therapeutically exploited. Despite promising advancements, challenges such as therapeutic resistance and adverse effects underscore the need for personalized medicine approaches and the development of next-generation inhibitors. This review underscores the multifaceted role of PI3K in lung cancer and advocates for integrated strategies to harness its full therapeutic potential, paving the way for improved patient outcomes.
Collapse
Affiliation(s)
- Bitian Zhang
- Institute of Chinese Medicine, State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | - Ping-Chung Leung
- Institute of Chinese Medicine, State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China
| | | | - Chun-Kwok Wong
- Institute of Chinese Medicine, State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China.
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China.
- Li Dak Sum Yip Yio Chin R & D Centre for Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, China.
| | - Dongjie Wang
- Institute of Chinese Medicine, State Key Laboratory of Research on Bioactivities and Clinical Applications of Medicinal Plants, The Chinese University of Hong Kong, Hong Kong, China.
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China.
| |
Collapse
|
23
|
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
|
24
|
Cozac-Szőke AR, Cozac DA, Negovan A, Tinca AC, Vilaia A, Cocuz IG, Sabău AH, Niculescu R, Chiorean DM, Tomuț AN, Cotoi OS. Immune Cell Interactions and Immune Checkpoints in the Tumor Microenvironment of Gastric Cancer. Int J Mol Sci 2025; 26:1156. [PMID: 39940924 PMCID: PMC11818890 DOI: 10.3390/ijms26031156] [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: 01/06/2025] [Revised: 01/26/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Gastric cancer (GC) ranks as the fifth most prevalent malignant neoplasm globally, with an increased death rate despite recent advancements in research and therapeutic options. Different molecular subtypes of GC have distinct interactions with the immune system, impacting the tumor microenvironment (TME), prognosis, and reaction to immunotherapy. Tumor-infiltrating lymphocytes (TILs) in the TME are crucial for preventing tumor growth and metastasis, as evidenced by research showing that patients with GC who have a significant density of TILs have better survival rates. But cancer cells have evolved a variety of mechanisms to evade immune surveillance, both sialic acid-binding immunoglobulin-like lectin 15 (Siglec-15) and Programmed Death-Ligand 1 (PD-L1) playing a pivotal role in the development of an immunosuppressive TME. They prevent T cell activation and proliferation resulting in a decrease in the immune system's capacity to recognize and eliminate malignant cells. These immune checkpoint molecules function via different but complementary mechanisms, the expression of Siglec-15 being mutually exclusive with PD-L1 and, therefore, providing a different therapeutic approach. The review explores how TILs affect tumor growth and patient outcomes in GC, with particular emphasis on their interactions within the TME and potential targeting of the PD-L1 and Siglec-15 pathways for immunotherapy.
Collapse
Affiliation(s)
- Andreea-Raluca Cozac-Szőke
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.-R.C.-S.); (A.H.S.); (R.N.); (D.M.C.)
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Dan Alexandru Cozac
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.-R.C.-S.); (A.H.S.); (R.N.); (D.M.C.)
- Emergency Institute for Cardiovascular Diseases and Transplantation Targu Mures, 540142 Targu Mures, Romania
| | - Anca Negovan
- Department of Clinical Science-Internal Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Andreea Cătălina Tinca
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Alexandra Vilaia
- Department of Infectious Diseases I, Doctoral School of Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Iuliu-Gabriel Cocuz
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Adrian Horațiu Sabău
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.-R.C.-S.); (A.H.S.); (R.N.); (D.M.C.)
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Raluca Niculescu
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.-R.C.-S.); (A.H.S.); (R.N.); (D.M.C.)
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Diana Maria Chiorean
- Doctoral School of Medicine and Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.-R.C.-S.); (A.H.S.); (R.N.); (D.M.C.)
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| | - Alexandru Nicușor Tomuț
- Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Ovidiu Simion Cotoi
- Pathophysiology Department, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Targu Mures, 540142 Targu Mures, Romania; (A.C.T.); (I.-G.C.); (O.S.C.)
- Pathology Department, Mures Clinical County Hospital, 540011 Targu Mures, Romania
| |
Collapse
|
25
|
Fan T, Xiao C, Deng Z, Li S, Tian H, Zheng Y, Zheng B, Li C, He J. Signatures of H3K4me3 modification predict cancer immunotherapy response and identify a new immune checkpoint-SLAMF9. Respir Res 2025; 26:17. [PMID: 39815269 PMCID: PMC11734478 DOI: 10.1186/s12931-024-03093-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/31/2024] [Indexed: 01/18/2025] Open
Abstract
H3 lysine 4 trimethylation (H3K4me3) modification and related regulators extensively regulate various crucial transcriptional courses in health and disease. However, the regulatory relationship between H3K4me3 modification and anti-tumor immunity has not been fully elucidated. We identified 72 independent prognostic genes of lung adenocarcinoma (LUAD) whose transcriptional expression were closely correlated with known 27 H3K4me3 regulators. We constructed three H3K4me3 modification patterns utilizing the expression profiles of the 72 genes, and patients classified in each pattern exhibited unique tumor immune infiltration characteristics. Using the principal component analysis (PCA) of H3K4me3-related patterns, we constructed a H3K4me3 risk score (H3K4me3-RS) system. The deep learning analysis using 12,159 cancer samples from 26 cancer types and 725 cancer samples from 5 immunotherapy cohorts revealed that H3K4me3-RS was significantly correlated with cancer immune tolerance and sensitivity. Importantly, this risk-score system showed satisfactory predictive performance for the ICB therapy responses of patients suffering from several cancer types, and we identified that SLAMF9 was one of the immunosuppressive phenotype and immunotherapy resistance-determined genes of H3K4me3-RS. The mice melanoma model showed Slamf9 knockdown remarkably restrained cancer progression and enhanced the efficacy of anti-CTLA-4 and anti-PD-L1 therapies by elevating CD8 + T cell infiltration. This study provided a new H3K4me3-associated biomarker system to predict tumor immunotherapy response and suggested the preclinical rationale for investigating the roles of SLAMF9 in cancer immunity regulation and treatment.
Collapse
Affiliation(s)
- Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuofeng Li
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bo Zheng
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
26
|
Yalamandala BN, Huynh TMH, Lien HW, Pan WC, Iao HM, Moorthy T, Chang YH, Hu SH. Advancing brain immunotherapy through functional nanomaterials. Drug Deliv Transl Res 2025:10.1007/s13346-024-01778-5. [PMID: 39789307 DOI: 10.1007/s13346-024-01778-5] [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] [Accepted: 12/17/2024] [Indexed: 01/12/2025]
Abstract
Glioblastoma (GBM), a highly aggressive brain tumor, poses significant treatment challenges due to its highly immunosuppressive microenvironment and the brain immune privilege. Immunotherapy activating the immune system and T lymphocyte infiltration holds great promise against GBM. However, the brain's low immunogenicity and the difficulty of crossing the blood-brain barrier (BBB) hinder therapeutic efficacy. Recent advancements in immune-actuated particles for targeted drug delivery have shown the potential to overcome these obstacles. These particles interact with the BBB by rapidly and reversibly disrupting its structure, thereby significantly enhancing targeting and penetrating delivery. The BBB targeting also minimizes potential long-term damage. At GBM, the particles demonstrated effective chemotherapy, chemodynamic therapy, photothermal therapy (PTT), photodynamic therapy (PDT), radiotherapy, or magnetotherapy, facilitating tumor disruption and promoting antigen release. Additionally, components of the delivery system retained autologous tumor-associated antigens and presented them to dendritic cells (DCs), ensuring prolonged immune activation. This review explores the immunosuppressive mechanisms of GBM, existing therapeutic strategies, and the role of nanomaterials in enhancing immunotherapy. We also discuss innovative particle-based approaches designed to traverse the BBB by mimicking innate immune functions to improve treatment outcomes for brain tumors.
Collapse
Affiliation(s)
- Bhanu Nirosha Yalamandala
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Thi My Hue Huynh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Hui-Wen Lien
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Wan-Chi Pan
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Hoi Man Iao
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Thrinayan Moorthy
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Yun-Hsuan Chang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Shang-Hsiu Hu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan.
| |
Collapse
|
27
|
Tang S, McGinnis R, Cao Z, Baker, Jr. JR, Xu Z, Wang S. Ultrasound-Guided Histotripsy Triggers the Release of Tumor-Associated Antigens from Breast Cancers. Cancers (Basel) 2025; 17:183. [PMID: 39857965 PMCID: PMC11764245 DOI: 10.3390/cancers17020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: There is increasing evidence to indicate that histotripsy treatment can enhance the host anti-tumor immune responses both locally at the targeting tumor site as well as systemically from abscopal effects. Histotripsy is a non-invasive ultrasound ablation technology that mechanically disrupts target tissue via cavitation. A key factor contributing to histotripsy-induced abscopal effects is believed to be the release of tumor-specific antigens (TSAs) or tumor-associated antigens (TAAs) that induce a systemic immune response. In this study, we studied the effect of histotripsy treatment on the release of HER2, a well-defined TAA target for cancer immunotherapy. Methods: A range of doses of histotripsy administered to HER2-postive mammary tumor cells in an in vitro cell culture system and an ex vivo tumor were applied. In addition, a single dose of histotripsy was used for an in vivo murine tumor model. The released proteins, and specifically HER2, in both tumor cell-free supernatants and tumor cell pellets were analyzed by a BCA protein assay, an ultra-performance liquid chromatography (UPLC) assay, and Western blot. Results: Our results showed that histotripsy could significantly trigger the release of HER2 proteins in the current study. The level of HER2 proteins was actually higher in tumor cell-free supernatants than in tumor cell pellets, suggesting that HER2 was released from the intracellular domain into the extracellular compartment. Furthermore, proportionally more HER2 protein was released at higher histotripsy doses, indicating free HER2 was histotripsy-dose-dependent. Conclusions: In conclusion, we have qualitatively and quantitatively demonstrated that histotripsy treatment triggers the release of HER2 from the tumor cells into the extracellular compartment. The histotripsy-mediated release of HER2 antigens provides important insights into the mechanism underlying its immunostimulation and suggests the potential of TSA/TAA-based immunotherapies in numerous cancer types.
Collapse
Affiliation(s)
- Shengzhuang Tang
- Department of Internal Medicine, Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (S.T.)
| | - Reliza McGinnis
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (R.M.); (Z.X.)
| | - Zhengyi Cao
- Department of Internal Medicine, Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (S.T.)
| | - James R. Baker, Jr.
- Department of Internal Medicine, Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (S.T.)
| | - Zhen Xu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; (R.M.); (Z.X.)
| | - Suhe Wang
- Department of Internal Medicine, Michigan Nanotechnology Institute for Medicine and Biological Sciences, University of Michigan, Ann Arbor, MI 48109, USA; (S.T.)
| |
Collapse
|
28
|
Drum DL, Jallorina AG, Wan LS, Chang VT, Lee-Wong MF. Non-Genetic Biomarkers in Merkel Cell Carcinoma: Prognostic Implications and Predictive Utility for Response to Anti-PD-(L)1 Immune Checkpoint Inhibitors. Exp Dermatol 2025; 34:e70030. [PMID: 39791602 DOI: 10.1111/exd.70030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 11/11/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
Merkel cell carcinoma (MCC) is a skin cancer that arises due to either Merkel cell polyomavirus infection (MCPyV) or ultraviolet (UV) radiation exposure, presenting primarily in the head and neck region of fair-skinned males. The recent success of PD-(L)1 immune checkpoint inhibitors (ICIs) in locally advanced/metastatic MCC, with an objective response rate (ORR) around 50% and improved survival, as a first-line treatment has moved ICIs to the forefront of therapy for MCC and generated interest in identifying biomarkers to predict clinical response. The MCC tumour microenvironment (TME) contains various components of the adaptive and innate immune system. These components can contribute to tumour immune escape through immunosuppression by preventing entrance of other immune cells or by aiding in the cytotoxic clearance of tumour cells. We aim to combine information from studies of baseline and on-treatment monitoring of the TME to help predict the success of ICIs in MCC. This review enhances the understanding of how CD8 T cells, γδ T cells and macrophages may impact predictions of response rates to ICIs in MCC patients. These immune cells are non-genetic biomarkers that can also be used to determine prognosis in MCC treatment.
Collapse
Affiliation(s)
- David L Drum
- Department of Medicine, California University of Science and Medicine, Colton, California, USA
| | - Anika G Jallorina
- Department of Medicine, California University of Science and Medicine, Colton, California, USA
| | - Leo S Wan
- Department of Medicine, West Virginia School of Osteopathic Medicine, Lewisburg, West Virginia, USA
| | - Victor T Chang
- Department of Medicine, Hematology/Oncology, Rutgers New Jersey School of Medicine, Newark, New Jersey, USA
- Section of Hematology/Oncology, Veterans Administration New Jersey Health Care System, East Orange, New Jersey, USA
| | - Mary F Lee-Wong
- Department of Medicine and Division of Clinical Immunology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Division of Allergy and Immunology, Maimonides Medical Center, Brooklyn, New York, USA
| |
Collapse
|
29
|
Lei Y, Liu J, Bai Y, Zheng C, Wang D. Peptides as Versatile Regulators in Cancer Immunotherapy: Recent Advances, Challenges, and Future Prospects. Pharmaceutics 2025; 17:46. [PMID: 39861694 PMCID: PMC11768547 DOI: 10.3390/pharmaceutics17010046] [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: 11/30/2024] [Revised: 12/25/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
The emergence of effective immunotherapies has revolutionized therapies for many types of cancer. However, current immunotherapy has limited efficacy in certain patient populations and displays therapeutic resistance after a period of treatment. To address these challenges, a growing number of immunotherapy drugs have been investigated in clinical and preclinical applications. The diverse functionality of peptides has made them attractive as a therapeutic modality, and the global market for peptide-based therapeutics is witnessing significant growth. Peptides can act as immunotherapeutic agents for the treatment of many malignant cancers. However, a systematic understanding of the interactions between different peptides and the host's immune system remains unclear. This review describes in detail the roles of peptides in regulating the function of the immune system for cancer immunotherapy. Initially, we systematically elaborate on the relevant mechanisms of cancer immunotherapy. Subsequently, we categorize peptide-based nanomaterials into the following three categories: peptide-based vaccines, anti-cancer peptides, and peptide-based delivery systems. We carefully analyzed the roles of these peptides in overcoming the current barriers in immunotherapy, including multiple strategies to enhance the immunogenicity of peptide vaccines, the synergistic effect of anti-cancer peptides in combination with other immune agents, and peptide assemblies functioning as immune stimulators or vehicles to deliver immune agents. Furthermore, we introduce the current status of peptide-based immunotherapy in clinical applications and discuss the weaknesses and future prospects of peptide-based materials for cancer immunotherapy. Overall, this review aims to enhance comprehension of the potential applications of peptide-based materials in cancer immunotherapy and lay the groundwork for future research and clinical applications.
Collapse
Affiliation(s)
- Yu Lei
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Y.L.); (J.L.); (Y.B.)
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Jiacheng Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Y.L.); (J.L.); (Y.B.)
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Yaowei Bai
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Y.L.); (J.L.); (Y.B.)
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Y.L.); (J.L.); (Y.B.)
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Dongyuan Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
30
|
Eshaq AM, Flanagan TW, Ba Abbad AA, Makarem ZAA, Bokir MS, Alasheq AK, Al Asheikh SA, Almashhor AM, Binyamani F, Al-Amoudi WA, Bawzir AS, Haikel Y, Megahed M, Hassan M. Immune Checkpoint Inhibitor-Associated Cutaneous Adverse Events: Mechanisms of Occurrence. Int J Mol Sci 2024; 26:88. [PMID: 39795946 PMCID: PMC11719825 DOI: 10.3390/ijms26010088] [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: 11/17/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
Immunotherapy, particularly that based on blocking checkpoint proteins in many tumors, including melanoma, Merkel cell carcinoma, non-small cell lung cancer (NSCLC), triple-negative breast (TNB cancer), renal cancer, and gastrointestinal and endometrial neoplasms, is a therapeutic alternative to chemotherapy. Immune checkpoint inhibitor (ICI)-based therapies have the potential to target different pathways leading to the destruction of cancer cells. Although ICIs are an effective treatment strategy for patients with highly immune-infiltrated cancers, the development of different adverse effects including cutaneous adverse effects during and after the treatment with ICIs is common. ICI-associated cutaneous adverse effects include mostly inflammatory and bullous dermatoses, as well as severe cutaneous side reactions such as rash or inflammatory dermatitis encompassing erythema multiforme; lichenoid, eczematous, psoriasiform, and morbilliform lesions; and palmoplantar erythrodysesthesia. The development of immunotherapy-related adverse effects is a consequence of ICIs' unique molecular action that is mainly mediated by the activation of cytotoxic CD4+/CD8+ T cells. ICI-associated cutaneous disorders are the most prevalent effects induced in response to anti-programmed cell death 1 (PD-1), anti-cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), and anti-programmed cell death ligand 1 (PD-L1) agents. Herein, we will elucidate the mechanisms regulating the occurrence of cutaneous adverse effects following treatment with ICIs.
Collapse
Affiliation(s)
- Abdulaziz M. Eshaq
- Department of Epidemiology and Biostatstics, Milken Institute School of Public Health, George Washington University Washington, Washington, DC 20052, USA;
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Thomas W. Flanagan
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA;
| | - Abdulqader A. Ba Abbad
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Zain Alabden A. Makarem
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Mohammed S. Bokir
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Ahmed K. Alasheq
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Sara A. Al Asheikh
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Abdullah M. Almashhor
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Faroq Binyamani
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Waleed A. Al-Amoudi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Abdulaziz S. Bawzir
- Department of Radiology, King Saud Medical City, Riyadh 11533, Saudi Arabia;
| | - Youssef Haikel
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, University of Strasbourg, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Mossad Megahed
- Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany;
| | - Mohamed Hassan
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, University of Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
31
|
Godiyal Y, Maheshwari D, Taniguchi H, Zinzuwadia SS, Morera-Díaz Y, Tewari D, Bishayee A. Role of PD-1/PD-L1 signaling axis in oncogenesis and its targeting by bioactive natural compounds for cancer immunotherapy. Mil Med Res 2024; 11:82. [PMID: 39690423 DOI: 10.1186/s40779-024-00586-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/29/2024] [Indexed: 12/19/2024] Open
Abstract
Cancer is a global health problem and one of the leading causes of mortality. Immune checkpoint inhibitors have revolutionized the field of oncology, emerging as a powerful treatment strategy. A key pathway that has garnered considerable attention is programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1). The interaction between PD-L1 expressed on tumor cells and PD-1 reduces the innate immune response and thus compromises the capability of the body's immune system. Furthermore, it controls the phenotype and functionality of innate and adaptive immune components. A range of monoclonal antibodies, including avelumab, atezolizumab, camrelizumab, dostarlimab, durvalumab, sinitilimab, toripalimab, and zimberelimab, have been developed for targeting the interaction between PD-1 and PD-L1. These agents can induce a broad spectrum of autoimmune-like complications that may affect any organ system. Recent studies have focused on the effect of various natural compounds that inhibit immune checkpoints. This could contribute to the existing arsenal of anticancer drugs. Several bioactive natural agents have been shown to affect the PD-1/PD-L1 signaling axis, promoting tumor cell apoptosis, influencing cell proliferation, and eventually leading to tumor cell death and inhibiting cancer progression. However, there is a substantial knowledge gap regarding the role of different natural compounds targeting PD-1 in the context of cancer. Hence, this review aims to provide a common connection between PD-1/PD-L1 blockade and the anticancer effects of distinct natural molecules. Moreover, the primary focus will be on the underlying mechanism of action as well as the clinical efficacy of bioactive molecules. Current challenges along with the scope of future research directions targeting PD-1/PD-L1 interactions through natural substances are also discussed.
Collapse
Affiliation(s)
- Yogesh Godiyal
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Drishti Maheshwari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Hiroaki Taniguchi
- Department of Experimental Embryology, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, 05-552, Magdalenka, Poland
- African Genome Center, Mohammed VI Polytechnic University, Hay Moulay Rachid, 43150, Ben Guerir, Morocco
| | - Shweta S Zinzuwadia
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA
| | - Yanelys Morera-Díaz
- Clinical Investigation and Biomedical Research Directions, Center for Genetic Engineering and Biotechnology, 11600, Havana, Cuba
| | - Devesh Tewari
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India.
| | - Anupam Bishayee
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| |
Collapse
|
32
|
Ilieva N, Pencheva M, Hadzhiev H, Tashkova D, Daskalova E, Georgiev P, Genova S. Impact of Neoadjuvant Therapy on PD-L1 Expression in Triple-Negative Breast Cancer and Correlation with Clinicopathological Factors. Diagnostics (Basel) 2024; 14:2672. [PMID: 39682581 DOI: 10.3390/diagnostics14232672] [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: 10/17/2024] [Revised: 11/20/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND This study aims to deliver more insights on the impact of neoadjuvant treatment on Pd-L1 expression and to evaluate its correlation with clinicopathological factors. METHODS We reviewed 88 TNBC cases for the period 2021-2023. Data on age, tumor size, stage, and treatment were collected. Histological slides were assessed for subtype, grade, and TILs. A total of 48 received neoadjuvant treatment. HER2 and Ki67 were evaluated via immunohistochemistry. PD-L1 expression was tested on primary and residual tumors. Statistical analysis was performed using IBM SPSS (p < 0.05). RESULTS In this study, PD-L1 positive expression was found in 44.3% of primary tumors, with 52.9% of initially positive cases losing expression post-treatment. TILs were significantly higher in PD-L1-positive tumors (mean 41.79% vs. 27.55%, p = 0.001). A notable correlation was found between PD-L1 expression and Ki-67 proliferation index, with PD-L1-positive tumors having a median Ki-67 of 64.49 compared to 52.86 in negative cases (p = 0.015). Neoadjuvant immunotherapy led to a lower mean residual cancer burden (0.95 vs. 2.55, p = 0.002) compared to chemotherapy alone. Higher Ki-67 levels (≥50%) were associated with better treatment outcomes, showing a mean RCB score of 1.60 versus 3.16 for lower levels (p = 0.022). HER2-negative cases had a higher prevalence of favorable pathological response (54.5%) compared to HER2-low tumors (25%, p = 0.048), because of the strong correlation to high proliferative index. CONCLUSIONS In conclusion, PD-L1 expression in TNBC shows significant discordance post-treatment, highlighting the need for routine testing and further research on predictive biomarkers.
Collapse
Affiliation(s)
- Nevena Ilieva
- Department of General and Clinical Pathology, Faculty of Medicine, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
- Clinical Pathology Department, Complex Oncology Center Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Mina Pencheva
- Department of Medical Physics and Biophysics, Faculty of Pharmacy, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Hristo Hadzhiev
- First Oncological Department, Complex Oncology Center Plovdiv, Bul. Al. Stamboliyski 2A, 4000 Plovdiv, Bulgaria
| | - Desislava Tashkova
- Department of General and Clinical Pathology, Faculty of Medicine, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
- Clinical Pathology Department, Complex Oncology Center Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Elena Daskalova
- Department of Anatomy, Histology and Embryology, Faculty of Medicine, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Petar Georgiev
- Faculty of Medicine, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| | - Sylvia Genova
- Department of General and Clinical Pathology, Faculty of Medicine, Medical University of Plovdiv, Bul. Vasil Aprilov 15A, 4000 Plovdiv, Bulgaria
| |
Collapse
|
33
|
Gao X, Zhang Y, Zhang M, Sun Y. Holliday junction recognition protein (HJURP) could reflect the clinical outcomes of lung adenocarcinoma patients, and impact the choice of precision therapy. Front Genet 2024; 15:1475511. [PMID: 39649097 PMCID: PMC11621083 DOI: 10.3389/fgene.2024.1475511] [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: 08/04/2024] [Accepted: 11/05/2024] [Indexed: 12/10/2024] Open
Abstract
Background Lung adenocarcinoma (LUAD) is the most prevalent subtype of non-small cell lung cancer (NSCLC), characterized by poor prognosis and a high mortality rate. Identifying reliable prognostic biomarkers and potential therapeutic targets is crucial for improving patient outcomes. Methods We conducted a comprehensive analysis of HJURP expression in LUAD using data from four cohorts: TCGA-LUAD (n = 453), GSE31210 (n = 226), GSE68465 (n = 442), and GSE72094 (n = 386). Univariate Cox regression analysis was employed to identify prognostic genes, with Kaplan-Meier survival analysis used to assess the predictive power of HJURP. Functional enrichment analyses were performed using MetaScape and FGSEA, and spatial transcriptomics and single-cell sequencing data were analyzed to explore HJURP's distribution and potential functions. Additionally, correlations between HJURP expression and genetic alterations, immune cell infiltration, and potential therapeutic responses were evaluated. Results HJURP was identified as a significant prognostic biomarker in all four cohorts, with high expression associated with increased risk of overall survival (OS) death (TCGA-LUAD: HR = 1.93, 95% CI: 1.321-2.815, P < 0.001; GSE31210: HR = 2.75, 95% CI: 1.319-5.735, P = 0.007; GSE68465: HR = 1.57, 95% CI: 1.215-2.038, P < 0.001; GSE72094: HR = 2.2, 95% CI: 1.485-3.27, P < 0.001). Functional analyses indicated that HJURP is involved in DNA metabolic processes, cell cycle regulation, and mitotic processes, with significant activation of pathways related to MYC targets, G2M checkpoint, and DNA repair. High HJURP expression was associated with higher mutation frequencies in TP53, CSMD3, TTN, and MUC16, and positively correlated with pro-inflammatory immune cell infiltration and several immune checkpoints, including PD-L1 and PD-L2. Chemotherapeutic agents such as gefitinib and sorafenib were predicted to be effective against high HJURP-expressing tumors. Conclusion HJURP is a pivotal biomarker for LUAD, consistently associated with poor prognosis and advanced disease stages. Its high expression correlates with specific genetic alterations and immune profiles, highlighting its potential as a therapeutic target. Future studies should validate these findings in larger cohorts.
Collapse
Affiliation(s)
| | | | | | - Yuejiao Sun
- Department of Respiratory, The Affiliated Hospital of Jiaxing University, Jiaxing, Zhejiang, China
| |
Collapse
|
34
|
Hu L, Sun C, Yuan K, Yang P. Expression, regulation, and function of PD-L1 on non-tumor cells in the tumor microenvironment. Drug Discov Today 2024; 29:104181. [PMID: 39278561 DOI: 10.1016/j.drudis.2024.104181] [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: 07/14/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024]
Abstract
Antiprogrammed death ligand 1 (PD-L1) therapy is a leading immunotherapy, but only some patients with solid cancers benefit. Overwhelming evidence has revealed that PD-L1 is expressed on various immune cells in the tumor microenvironment (TME), including macrophages, dendritic cells, and regulatory T cells, modulating tumor immunity and influencing tumor progression. PD-L1 can also be located on tumor cell membranes as well as in exosomes and cytoplasm. Accordingly, the dynamic expression and various forms of PD-L1 might explain the therapy's limited efficacy and resistance. Herein a systematic summary of the expression of PD-L1 on different immune cells and their regulatory mechanisms is provided to offer a solid foundation for future studies.
Collapse
Affiliation(s)
- Lingrong Hu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Chengliang Sun
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Kai Yuan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China.
| | - Peng Yang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China; Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
35
|
Zhao C, Zhao J, Zhang Y, Zhu Y, Yang Z, Liu S, Tang Q, Yang Y, Wang H, Shu Y, Dong P, Wu X, Gong W. PTBP3 Mediates IL-18 Exon Skipping to Promote Immune Escape in Gallbladder Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406633. [PMID: 39116343 PMCID: PMC11481411 DOI: 10.1002/advs.202406633] [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: 06/15/2024] [Revised: 07/31/2024] [Indexed: 08/10/2024]
Abstract
Gallbladder cancer (GBC) is the most common malignant tumor of the biliary system, with poor response to current treatments. Abnormal alternative splicing has been associated with the development of a variety of tumors. Combining the GEO database and GBC mRNA-seq analysis, it is found high expression of the splicing factor polypyrimidine region- binding protein 3 (PTBP3) in GBC. Multi-omics analysis revealed that PTBP3 promoted exon skipping of interleukin-18 (IL-18), resulting in the expression of ΔIL-18, an isoform specifically expressed in tumors. That ΔIL-18 promotes GBC immune escape by down-regulating FBXO38 transcription levels in CD8+T cells to reduce PD-1 ubiquitin-mediated degradation is revealed. Using a HuPBMC mouse model, the role of PTBP3 and ΔIL-18 in promoting GBC growth is confirmed, and showed that an antisense oligonucleotide that blocked ΔIL-18 production displayed anti-tumor activity. Furthermore, that the H3K36me3 promotes exon skipping of IL-18 by recruiting PTBP3 via MRG15 is demonstrated, thereby coupling the processes of IL-18 transcription and alternative splicing. Interestingly, it is also found that the H3K36 methyltransferase SETD2 binds to hnRNPL, thereby interfering with PTBP3 binding to IL-18 pre-mRNA. Overall, this study provides new insights into how aberrant alternative splicing mechanisms affect immune escape, and provides potential new perspectives for improving GBC immunotherapy.
Collapse
Affiliation(s)
- Cheng Zhao
- Laboratory of General Surgery and Department of General SurgeryXinhua Hospital affiliated with Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang RoadShanghai200092China
| | - Jing‐wei Zhao
- Laboratory of General Surgery and Department of General SurgeryXinhua Hospital affiliated with Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang RoadShanghai200092China
| | - Yu‐han Zhang
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang RoadShanghai200092China
| | - Yi‐di Zhu
- Laboratory of General Surgery and Department of General SurgeryXinhua Hospital affiliated with Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang RoadShanghai200092China
| | - Zi‐yi Yang
- Laboratory of General Surgery and Department of General SurgeryXinhua Hospital affiliated with Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang RoadShanghai200092China
| | - Shi‐lei Liu
- Laboratory of General Surgery and Department of General SurgeryXinhua Hospital affiliated with Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang RoadShanghai200092China
| | - Qiu‐yi Tang
- Laboratory of General Surgery and Department of General SurgeryXinhua Hospital affiliated with Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang RoadShanghai200092China
| | - Yue Yang
- Laboratory of General Surgery and Department of General SurgeryXinhua Hospital affiliated with Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang RoadShanghai200092China
| | - Hua‐kai Wang
- Laboratory of General Surgery and Department of General SurgeryXinhua Hospital affiliated with Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang RoadShanghai200092China
| | - Yi‐jun Shu
- Laboratory of General Surgery and Department of General SurgeryXinhua Hospital affiliated with Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang RoadShanghai200092China
| | - Ping Dong
- Laboratory of General Surgery and Department of General SurgeryXinhua Hospital affiliated with Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang RoadShanghai200092China
| | - Xiang‐song Wu
- Laboratory of General Surgery and Department of General SurgeryXinhua Hospital affiliated with Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang RoadShanghai200092China
| | - Wei Gong
- Laboratory of General Surgery and Department of General SurgeryXinhua Hospital affiliated with Shanghai Jiao Tong University School of MedicineNo. 1665 Kongjiang RoadShanghai200092China
- Shanghai Key Laboratory of Biliary Tract Disease ResearchNo. 1665 Kongjiang RoadShanghai200092China
| |
Collapse
|
36
|
Pavelescu LA, Enache RM, Roşu OA, Profir M, Creţoiu SM, Gaspar BS. Predictive Biomarkers and Resistance Mechanisms of Checkpoint Inhibitors in Malignant Solid Tumors. Int J Mol Sci 2024; 25:9659. [PMID: 39273605 PMCID: PMC11395316 DOI: 10.3390/ijms25179659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
Predictive biomarkers for immune checkpoint inhibitors (ICIs) in solid tumors such as melanoma, hepatocellular carcinoma (HCC), colorectal cancer (CRC), non-small cell lung cancer (NSCLC), endometrial carcinoma, renal cell carcinoma (RCC), or urothelial carcinoma (UC) include programmed cell death ligand 1 (PD-L1) expression, tumor mutational burden (TMB), defective deoxyribonucleic acid (DNA) mismatch repair (dMMR), microsatellite instability (MSI), and the tumor microenvironment (TME). Over the past decade, several types of ICIs, including cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors, anti-programmed cell death 1 (PD-1) antibodies, anti-programmed cell death ligand 1 (PD-L1) antibodies, and anti-lymphocyte activation gene-3 (LAG-3) antibodies have been studied and approved by the Food and Drug Administration (FDA), with ongoing research on others. Recent studies highlight the critical role of the gut microbiome in influencing a positive therapeutic response to ICIs, emphasizing the importance of modeling factors that can maintain a healthy microbiome. However, resistance mechanisms can emerge, such as increased expression of alternative immune checkpoints, T-cell immunoglobulin (Ig), mucin domain-containing protein 3 (TIM-3), LAG-3, impaired antigen presentation, and alterations in the TME. This review aims to synthesize the data regarding the interactions between microbiota and immunotherapy (IT). Understanding these mechanisms is essential for optimizing ICI therapy and developing effective combination strategies.
Collapse
Affiliation(s)
- Luciana Alexandra Pavelescu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Robert Mihai Enache
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania
| | - Oana Alexandra Roşu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Monica Profir
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Sanda Maria Creţoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Bogdan Severus Gaspar
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Surgery Clinic, Bucharest Emergency Clinical Hospital, 014461 Bucharest, Romania
| |
Collapse
|
37
|
Rahman MA, Ali MM. Recent Treatment Strategies and Molecular Pathways in Resistance Mechanisms of Antiangiogenic Therapies in Glioblastoma. Cancers (Basel) 2024; 16:2975. [PMID: 39272834 PMCID: PMC11394361 DOI: 10.3390/cancers16172975] [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: 07/30/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
Malignant gliomas present great difficulties in treatment, with little change over the past 30 years in the median survival time of 15 months. Current treatment options include surgery, radiotherapy (RT), and chemotherapy. New therapies aimed at suppressing the formation of new vasculature (antiangiogenic treatments) or destroying formed tumor vasculature (vascular disrupting agents) show promise. This study summarizes the existing knowledge regarding the processes by which glioblastoma (GBM) tumors acquire resistance to antiangiogenic treatments. The discussion encompasses the activation of redundant proangiogenic pathways, heightened tumor cell invasion and metastasis, resistance induced by hypoxia, creation of vascular mimicry channels, and regulation of the tumor immune microenvironment. Subsequently, we explore potential strategies to overcome this resistance, such as combining antiangiogenic therapies with other treatment methods, personalizing treatments for each patient, focusing on new therapeutic targets, incorporating immunotherapy, and utilizing drug delivery systems based on nanoparticles. Additionally, we would like to discuss the limitations of existing methods and potential future directions to enhance the beneficial effects of antiangiogenic treatments for patients with GBM. Therefore, this review aims to enhance the research outcome for GBM and provide a more promising opportunity by thoroughly exploring the mechanisms of resistance and investigating novel therapeutic strategies.
Collapse
Affiliation(s)
- Md Ataur Rahman
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | - Meser M Ali
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
38
|
Pei X, Zhang SL, Qiu BQ, Zhang PF, Liu TS, Wang Y. Cancer Cell Secreted Legumain Promotes Gastric Cancer Resistance to Anti-PD-1 Immunotherapy by Enhancing Macrophage M2 Polarization. Pharmaceuticals (Basel) 2024; 17:951. [PMID: 39065799 PMCID: PMC11279811 DOI: 10.3390/ph17070951] [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: 05/27/2024] [Revised: 06/19/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
The interaction between cancer cells and immune cells plays critical roles in gastric cancer (GC) progression and immune evasion. Forced legumain (LGMN) is one of the characteristics correlated with poor prognosis in gastric cancer patients. However, the role of gastric-cancer-secreted LGMN (sLGMN) in modulating the tumor immune microenvironment and the biological effect on the immune evasion of gastric cancer remains unclear. In this study, we found that forced expression of sLGMN in gastric cancer serum correlates with increased M2 macrophage infiltration in GC tissues and predicted resistance to anti-PD-1 immunotherapy. Mechanistically, gastric cancer cells secrete LGMN via binding to cell surface Integrin αvβ3, then activate Integrin αvβ3/PI3K (Phosphatidylinositol-4,5-bisphosphate3-kinase)/AKT (serine/threonine kinase)/mTORC2 (mammalian target of rapamycin complex 2) signaling, promote metabolic reprogramming, and polarize macrophages from the M1 to the M2 phenotype. Either blocking LGMN, Integrin αv, or knocking out Integrin αv expression and abolishing the LGMN/Integrin αvβ3 interaction significantly inhibits metabolic reprogramming and polarizes macrophages from the M1 to the M2 phenotype. This study reveals a critical molecular crosstalk between gastric cancer cells and macrophages through the sLGMN/Integrinαvβ3/PI3K/AKT/mTORC2 axis in promoting gastric cancer immune evasion and resistance to anti-PD-1 immunotherapy, indicating that the sLGMN/Integrinαvβ3/PI3K/AKT/mTORC2 axis may act as a promising therapeutic target.
Collapse
Affiliation(s)
- Xu Pei
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (X.P.); (S.-L.Z.); (P.-F.Z.)
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330030, China;
| | - Shi-Long Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (X.P.); (S.-L.Z.); (P.-F.Z.)
| | - Bai-Quan Qiu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330030, China;
| | - Peng-Fei Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (X.P.); (S.-L.Z.); (P.-F.Z.)
- Department of Medical Oncology, Shanghai Geriatric Medical Center, Shanghai 201104, China
| | - Tian-Shu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (X.P.); (S.-L.Z.); (P.-F.Z.)
- Cancer Center, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, China
- Center of Evidence-Based Medicine, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, China
| | - Yan Wang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; (X.P.); (S.-L.Z.); (P.-F.Z.)
- Cancer Center, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, China
- Center of Evidence-Based Medicine, Zhongshan Hospital Affiliated to Fudan University, Shanghai 200032, China
- Shanghai Medical College, Zhongshan Hospital Immunotherapy Translational Research Center, Shanghai 200032, China
| |
Collapse
|
39
|
Ren X, Wang L, Liu L, Liu J. PTMs of PD-1/PD-L1 and PROTACs application for improving cancer immunotherapy. Front Immunol 2024; 15:1392546. [PMID: 38638430 PMCID: PMC11024247 DOI: 10.3389/fimmu.2024.1392546] [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: 02/28/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024] Open
Abstract
Immunotherapy has been developed, which harnesses and enhances the innate powers of the immune system to fight disease, particularly cancer. PD-1 (programmed death-1) and PD-L1 (programmed death ligand-1) are key components in the regulation of the immune system, particularly in the context of cancer immunotherapy. PD-1 and PD-L1 are regulated by PTMs, including phosphorylation, ubiquitination, deubiquitination, acetylation, palmitoylation and glycosylation. PROTACs (Proteolysis Targeting Chimeras) are a type of new drug design technology. They are specifically engineered molecules that target specific proteins within a cell for degradation. PROTACs have been designed and demonstrated their inhibitory activity against the PD-1/PD-L1 pathway, and showed their ability to degrade PD-1/PD-L1 proteins. In this review, we describe how PROTACs target PD-1 and PD-L1 proteins to improve the efficacy of immunotherapy. PROTACs could be a novel strategy to combine with radiotherapy, chemotherapy and immunotherapy for cancer patients.
Collapse
Affiliation(s)
- Xiaohui Ren
- Department of Respiratory Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Lijuan Wang
- Department of Hospice Care, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Likun Liu
- Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Juan Liu
- Department of Special Needs Medicine, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|