1
|
Nemati M, Hsu CY, Nathiya D, Kumar MR, Oghenemaro EF, Kariem M, Kaur P, Bhanot D, Hjazi A, Azam Saedi T. Gemcitabine: immunomodulatory or immunosuppressive role in the tumor microenvironment. Front Immunol 2025; 16:1536428. [PMID: 40270972 PMCID: PMC12014622 DOI: 10.3389/fimmu.2025.1536428] [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: 11/28/2024] [Accepted: 03/21/2025] [Indexed: 04/25/2025] Open
Abstract
Gemcitabine (GEM), a nucleoside analog chemotherapy agent, has been widely used in the treatment of various cancers. In recent years, there has been growing interest in understanding the immunomodulatory or immunosuppressive effects of GEM. The immunomodulatory roles of GEM could influence the anti-tumor immune responses via several mechanisms, such as modulation of antigen presentation, cytokine production, and immune cell population. Furthermore, there is evidence that GEM enhances the therapeutic efficacy of immunotherapies, including oncolytic viruses, immune checkpoint inhibitors, CAR T-cells, and therapeutic vaccines. On the other hand, accumulating evidence also proposed that GEM may act as an immunosuppressive agent within the tumor microenvironment, resulting in immune evasion of tumor cells and tumor growth. These paradoxical roles of GEM in modifying immune responses highlight the complexity of GEM interaction with immune cells and responses within the tumor microenvironment. This review aims to provide an overview of the immunomodulatory and immunosuppressive effects of GEM within the tumor microenvironment and how GEM affects the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Mahnaz Nemati
- Amir Oncology Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Chou-Yi Hsu
- Thunderbird School of Global Management, Arizona State University, Phoenix, AZ, United States
| | - Deepak Nathiya
- Department of Pharmacy Practice, NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - M. Ravi Kumar
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Delta State, Nigeria
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | - Deepak Bhanot
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Tayebeh Azam Saedi
- Department of Genetics, Faculty of Science, Islamic Azad University, Tonekabon, Iran
| |
Collapse
|
2
|
Arun B, John G, Raman R. MicroRNA Signatures: Illuminating Minimal Residual Disease Monitoring in Juvenile Myelomonocytic Leukemia - A Review. J Hematol 2025; 14:43-55. [PMID: 40336920 PMCID: PMC12056752 DOI: 10.14740/jh1384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/20/2025] [Indexed: 05/09/2025] Open
Abstract
Juvenile myelomonocytic leukemia (JMML) is an aggressive pediatric myelodysplastic/myeloproliferative neoplasm characterized by RAS pathway mutations and significant heterogeneity. Minimal residual disease (MRD) monitoring is crucial for evaluating treatment response and predicting relapse risk. MicroRNA (miRNAs), small non-coding RNAs with pivotal roles in gene regulation, have emerged as promising biomarkers for JMML MRD detection. This review explores the mechanistic role of miRNAs in JMML pathogenesis, emphasizing their diagnostic, prognostic, and therapeutic potential. Dysregulated miRNA profiles correlate with distinct JMML subgroups and disease progression, suggesting utility in non-invasive MRD monitoring. Emerging evidence highlights miR-150-5p as a tumor suppressor targeting STAT5b and its therapeutic potential in ameliorating JMML's aberrant signaling pathways. We compare traditional MRD methods, such as flow cytometry and polymerase chain reaction (PCR), with miRNA-based techniques, underscoring the latter's superior sensitivity, specificity, and non-invasiveness. Recent advances in miRNA profiling technologies, including next-generation sequencing and digital PCR, enable precise detection of residual leukemic cells and support personalized treatment approaches. Despite significant progress, challenges persist in standardizing miRNA-based assays and validating their clinical utility. Ethical considerations, including patient privacy and informed consent, remain critical for integrating miRNA diagnostics into routine care. This review provides a comprehensive synthesis of current knowledge on miRNA signatures in JMML, illuminating their transformative potential in MRD monitoring and paving the way for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Bhavyadharshini Arun
- Hasan Lab, Department of Medical Oncology, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Geofrey John
- Homi Bhabha National Institute, Mumbai, Maharashtra, India
- Department of Radiation Oncology, Advanced Centre for Treatment Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai, Maharashtra, India
| | - Rajeshkumar Raman
- Department of Pharmaceutical Biotechnology, JSS College of Pharmacy, Ooty, The Nilgiris, India
| |
Collapse
|
3
|
Jafari M, Abdoli S, Asgari M, Moghaddam Pour M, Shokrgozar MA, Sharifzadeh Z. Combination Therapy of Oncolytic Newcastle Virus and Lenalidomide Enhanced Cytotoxicity in Prostate Cancer Cells. IRANIAN BIOMEDICAL JOURNAL 2025; 29:9-19. [PMID: 40223343 PMCID: PMC12040631 DOI: 10.61186/ibj.4367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/10/2024] [Indexed: 04/20/2025]
Abstract
Background Despite existing treatments, advanced solid tumors, such as prostate cancer (PCa), require the development of novel anticancer therapies. Oncolytic viruses (OVs) present a potential treatment option for solid tumors. Newcastle disease virus (NDV) is one of the most promising OVs that can replicate within and destroys human cancer cells. This study aimed to evaluate the cytotoxic and apoptotic effects of the NDV strain on human PCa cells in vitro. Additionally, We examined a novel treatment for PCa by combining Lenalidomide (Len) with NDV. Methods NDV strains La Sota, B1, and I2 were tested for cytotoxicity against several cell lines. A safety assessment was conducted in primary cells using peripheral blood mononuclear cells (PBMCs). Also, apoptosis induction was measured using annexin V/7AAD staining. Finally, the cytotoxic effects of NDV alone and in combination with Len, were assessed using MTT. Results The NDV showed cytotoxic effects on tumor cell lines and induced apoptosis in infected prostate cells compared to control cells. The NDV La Sota strain exhibited significant oncolytic capacity, reducing the viability of LNCaP and DU145 cells to less than 40% at specific concentrations, while showing no cytotoxic effects on primary PBMCs. Also, NDV induced apoptosis in the prostate cell line by 60%. Furthermore, Len enhanced the cytotoxicity of PCa cells when combined with NDV. Conclusion Our study confirms the efficacy of oncolytic NDV treatment for PCa, particularly utilizing the La Sota strain. When combined with Len, NDV indicates an enhanced effectiveness in destroying tumor cells. These findings suggest a prospective treatment approach that needs more preclinical and clinical studies to improve outcomes in PCa treatment.
Collapse
Affiliation(s)
- Mahdie Jafari
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Shahriyar Abdoli
- School of Advanced Medical Technologies, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Asgari
- Malaria and Vector Research Group (MVRG), Biotechnology Research Center (BRC), Pasteur Institute of Iran, Tehran, Iran
- Myeloma Center, Winthrop P, Rockefeller Institute, Department of Internal Medicine, University of Arkansas for Medical Science, Little Rock, Arkansas, USA
| | - Masoud Moghaddam Pour
- Poultry Viral Vaccines Research and Production Department, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Extension Organization, Karaj, Iran
| | | | | |
Collapse
|
4
|
Ponterio E, Haas TL, De Maria R. Oncolytic virus and CAR-T cell therapy in solid tumors. Front Immunol 2024; 15:1455163. [PMID: 39539554 PMCID: PMC11557337 DOI: 10.3389/fimmu.2024.1455163] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024] Open
Abstract
Adoptive immunotherapy with T cells, genetically modified to express a tumor-reactive chimeric antigen receptor (CAR), is an innovative and rapidly developing life-saving treatment for cancer patients without other therapeutic opportunities. CAR-T cell therapy has proven effective only in hematological malignancies. However, although by now only a few clinical trials had promising outcomes, we predict that CAR-T therapy will eventually become an established treatment for several solid tumors. Oncolytic viruses (OVs) can selectively replicate in and kill cancer cells without harming healthy cells. They can stimulate an immune response against the tumor, because OVs potentially stimulate adaptive immunity and innate components of the host immune system. Using CAR-T cells along with oncolytic viruses may enhance the efficacy of CAR-T cell therapy in destroying solid tumors by increasing the tumor penetrance of T cells and reducing the immune suppression by the tumor microenvironment. This review describes recent advances in the design of oncolytic viruses and CAR-T cells while providing an overview of the potential combination of oncolytic virotherapy with CAR-T cells for solid cancers. In this review, we will focus on the host-virus interaction in the tumor microenvironment to reverse local immunosuppression and to develop CAR-T cell effector function.
Collapse
Affiliation(s)
- Eleonora Ponterio
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
- Dipartimento di Medicina e Chirurgia Traslazionali, Sezione di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Tobias Longin Haas
- Dipartimento di Medicina e Chirurgia Traslazionali, Sezione di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy
- IIGM - Italian Institute for Genomic Medicine, Candiolo, TO, Italy
| | - Ruggero De Maria
- Dipartimento di Medicina e Chirurgia Traslazionali, Sezione di Patologia Generale, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario “A. Gemelli” - I.R.C.C.S., Rome, Italy
| |
Collapse
|
5
|
Maali A, Noei A, Feghhi-Najafabadi S, Sharifzadeh Z. A Systematic Review on the Dual Role of Interleukin-1 in CAR T-Cell Therapy: Enhancer and Mitigator. IRANIAN BIOMEDICAL JOURNAL 2024; 28:221-34. [PMID: 39891450 PMCID: PMC11829154 DOI: 10.61186/ibj.4444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 10/29/2024] [Indexed: 02/10/2025]
Abstract
Chimeric antigen receptor T-cell therapy is a groundbreaking approach for treating certain hematologic malignancies and solid tumors. However, its application is limited by severe toxicities, particularly CRS and ICANS, dramatically limit its broader application. IL-1 plays a crucial role in both enhancing CAR T-cell efficacy and driving these toxic effects. This review systematically examines the dual functions of IL-1, highlighting its role in promoting CAR T-cell activation and persistence while contributing to CRS and ICANS pathogenesis. Strategies to mitigate IL-1-driven toxicities, including IL-1 receptor antagonists, monoclonal antibodies, IL-1 trapping, and interference with IL-1 production, show promise in reducing adverse effects without compromising therapeutic efficacy. Understanding the complex role of IL-1 in CAR T-cell therapy may lead to optimized treatment strategies, improving safety and expanding clinical applicability. Further research is essential to refine IL-1-targeted interventions and enhance the therapeutic potential of CAR T-cell therapy. Chimeric antigen receptor (CAR) T-cell therapy is a groundbreaking approach for treating certain hematologic malignancies and solid tumors. However, its application is limited by severe toxicities, particularly cytokine release syndrome (CRS) and cell-associated neurotoxicity syndrome (ICANS), dramatically limit its broader application. IL-1 plays a crucial role in both enhancing CAR T-cell efficacy and driving these toxic effects. This review systematically examines the dual functions of IL-1, highlighting its role in promoting CAR T-cell activation and persistence while contributing to CRS and ICANS pathogenesis. Strategies to mitigate IL-1-driven toxicities, including IL-1 receptor antagonists, monoclonal antibodies, IL-1 trapping, and interference with IL-1 production, show promise in reducing adverse effects without compromising therapeutic efficacy. Understanding the complex role of IL-1 in CAR T-cell therapy may lead to optimized treatment strategies, improving safety and expanding clinical applicability. Further research is essential to refine IL-1-targeted interventions and enhance the therapeutic potential of CAR T-cell therapy.
Collapse
Affiliation(s)
- Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Ahmad Noei
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | - Saba Feghhi-Najafabadi
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Student Research Committee, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
6
|
Karadimas T, Huynh TH, Chose C, Zervoudakis G, Clampitt B, Lapp S, Joyce D, Letson GD, Metts J, Binitie O, Mullinax JE, Lazarides A. Oncolytic Viral Therapy in Osteosarcoma. Viruses 2024; 16:1139. [PMID: 39066301 PMCID: PMC11281467 DOI: 10.3390/v16071139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/13/2024] [Accepted: 07/14/2024] [Indexed: 07/28/2024] Open
Abstract
Primary bone malignancies, including osteosarcoma (OS), are rare but aggressive. Current OS treatment, involving surgical resection and chemotherapy, has improved survival for non-metastatic cases but remains ineffective for recurrent or metastatic OS. Oncolytic viral therapy (OVT) is a promising alternative, using naturally occurring or genetically modified viruses to selectively target and lyse cancer cells and induce a robust immune response against remaining OS cells. Various oncolytic viruses (OVs), such as adenovirus, herpes simplex virus, and measles virus, have demonstrated efficacy in preclinical OS models. Combining OVT with other therapeutics, such as chemotherapy or immunotherapy, may further improve outcomes. Despite these advances, challenges in reliability of preclinical models, safety, delivery, and immune response must be addressed to optimize OVT for clinical use. Future research should focus on refining delivery methods, exploring combination treatments, and clinical trials to ensure OVT's efficacy and safety for OS. Overall, OVT represents a novel approach with the potential to drastically improve survival outcomes for patients with OS.
Collapse
Affiliation(s)
- Thomas Karadimas
- Morsani College of Medicine, University of South Florida Health, Tampa, FL 33602, USA; (T.H.H.); (C.C.); (B.C.); (S.L.)
| | - Thien Huong Huynh
- Morsani College of Medicine, University of South Florida Health, Tampa, FL 33602, USA; (T.H.H.); (C.C.); (B.C.); (S.L.)
| | - Chloe Chose
- Morsani College of Medicine, University of South Florida Health, Tampa, FL 33602, USA; (T.H.H.); (C.C.); (B.C.); (S.L.)
| | - Guston Zervoudakis
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL 33612, USA; (G.Z.); (D.J.); (G.D.L.); (J.M.); (O.B.); (J.E.M.); (A.L.)
| | - Bryan Clampitt
- Morsani College of Medicine, University of South Florida Health, Tampa, FL 33602, USA; (T.H.H.); (C.C.); (B.C.); (S.L.)
| | - Sean Lapp
- Morsani College of Medicine, University of South Florida Health, Tampa, FL 33602, USA; (T.H.H.); (C.C.); (B.C.); (S.L.)
| | - David Joyce
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL 33612, USA; (G.Z.); (D.J.); (G.D.L.); (J.M.); (O.B.); (J.E.M.); (A.L.)
| | - George Douglas Letson
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL 33612, USA; (G.Z.); (D.J.); (G.D.L.); (J.M.); (O.B.); (J.E.M.); (A.L.)
| | - Jonathan Metts
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL 33612, USA; (G.Z.); (D.J.); (G.D.L.); (J.M.); (O.B.); (J.E.M.); (A.L.)
| | - Odion Binitie
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL 33612, USA; (G.Z.); (D.J.); (G.D.L.); (J.M.); (O.B.); (J.E.M.); (A.L.)
| | - John E. Mullinax
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL 33612, USA; (G.Z.); (D.J.); (G.D.L.); (J.M.); (O.B.); (J.E.M.); (A.L.)
| | - Alexander Lazarides
- Sarcoma Department, Moffitt Cancer Center, Tampa, FL 33612, USA; (G.Z.); (D.J.); (G.D.L.); (J.M.); (O.B.); (J.E.M.); (A.L.)
| |
Collapse
|
7
|
Anjum S, Naseer F, Ahmad T, Liaquat A, Abduh MS, Kousar K. Co-delivery of oncolytic virus and chemotherapeutic modality: Vincristine against prostate cancer treatment: A potent viro-chemotherapeutic approach. J Med Virol 2024; 96:e29748. [PMID: 38975633 DOI: 10.1002/jmv.29748] [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: 10/31/2023] [Revised: 05/22/2024] [Accepted: 06/04/2024] [Indexed: 07/09/2024]
Abstract
Prostate cancer is a prevalent carcinoma among males, and conventional treatment options are often limited. Cytotoxic chemotherapy, despite its drawbacks, remains a mainstay. We propose a targeted co-delivery approach using nanoscale delivery units for Oncolytic measles virus (OMV) and vincristine (VC) to enhance treatment efficacy. The HA-coated OMV + VC-loaded TCs nanoformulation is designed for targeted oncolytic activity in prostate cancer. The CD44 expression analysis in prostate cancer cell lines indicates a significantly high expression in PC3 cells. The optimization of nanoformulations using Design of Expert (DOE) is performed, and the preparation and characterization of HA-coated OMV + VC-loaded TCs nanoformulations are detailed showing average particle size 397.2 ± 0.01 nm and polydispersity index 0.122 with zeta potential 19.7 + 0.01 mV. Results demonstrate successful encapsulation efficiency with 2.4 × 106 TCID50/Ml and sustained release of OMV and VC from the nanoformulation for up to 72 h. In vitro, assays reveal potent anticancer activity at 10 ± 0.71% cell viability in PC3 cells compared to 73 ± 0.66% in HPrEC and significant morphological changes at 90 µg/ml in dose and time-dependent manner. The co-formulation showed positive cell death 49.5 ± 0.02% at 50 µg PI/ml in PBS and 54.3% cell cycle arrest at the G2/M phase, 8.1% G0/G1 and 5.7% at S phase, with significant mitochondrial membrane potential (MMP) at 50 µg/ml, as assessed by flow cytometry (FACS). The surface-integrating ligand approach enhances the targeted delivery of the oncolytic virus and chemotherapeutic drug, presenting a potential alternative for prostate cancer treatment and suggested that co-administering VC and OMV in a nanoformulation could improve therapeutic outcomes while reducing chemotherapeutic drug doses.
Collapse
Affiliation(s)
- Sadia Anjum
- Department of Biology, University of Hail, Hail, Saudia Arabia
| | - Faiza Naseer
- Department of Biosciences, Shifa Tameer e Millat University, Islamabad, Pakistan
| | - Tahir Ahmad
- Industrial Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Afrose Liaquat
- Shifa College of Medicine, Shifa Tameer-e-Millat University, Islamabad, Pakistan
| | - Maisa S Abduh
- Immune Responses in Different Diseases Research Group, Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdul-Aziz University, Jeddah, Saudia Arabia
| | - Kousain Kousar
- Industrial Biotechnology, Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and Technology, Islamabad, Pakistan
| |
Collapse
|
8
|
Stergiopoulos GM, Concilio SC, Galanis E. An Update on the Clinical Status, Challenges, and Future Directions of Oncolytic Virotherapy for Malignant Gliomas. Curr Treat Options Oncol 2024; 25:952-991. [PMID: 38896326 PMCID: PMC11878440 DOI: 10.1007/s11864-024-01211-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/24/2024] [Indexed: 06/21/2024]
Abstract
OPINION STATEMENT Malignant gliomas are common central nervous system tumors that pose a significant clinical challenge due to the lack of effective treatments. Glioblastoma (GBM), a grade 4 malignant glioma, is the most prevalent primary malignant brain tumor and is associated with poor prognosis. Current clinical trials are exploring various strategies to combat GBM, with oncolytic viruses (OVs) appearing particularly promising. In addition to ongoing and recently completed clinical trials, one OV (Teserpaturev, Delytact®) received provisional approval for GBM treatment in Japan. OVs are designed to selectively target and eliminate cancer cells while promoting changes in the tumor microenvironment that can trigger and support long-lasting anti-tumor immunity. OVs offer the potential to remodel the tumor microenvironment and reverse systemic immune exhaustion. Additionally, an increasing number of OVs are armed with immunomodulatory payloads or combined with immunotherapy approaches in an effort to promote anti-tumor responses in a tumor-targeted manner. Recently completed oncolytic virotherapy trials can guide the way for future treatment individualization through patient preselection, enhancing the likelihood of achieving the highest possible clinical success. These trials also offer valuable insight into the numerous challenges inherent in malignant glioma treatment, some of which OVs can help overcome.
Collapse
Affiliation(s)
| | | | - Evanthia Galanis
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Oncology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
9
|
Crimini E, Boscolo Bielo L, Berton Giachetti PPM, Pellizzari G, Antonarelli G, Taurelli Salimbeni B, Repetto M, Belli C, Curigliano G. Beyond PD(L)-1 Blockade in Microsatellite-Instable Cancers: Current Landscape of Immune Co-Inhibitory Receptor Targeting. Cancers (Basel) 2024; 16:281. [PMID: 38254772 PMCID: PMC10813411 DOI: 10.3390/cancers16020281] [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: 12/11/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
High microsatellite instability (MSI-H) derives from genomic hypermutability due to deficient mismatch repair function. Colorectal (CRC) and endometrial cancers (EC) are the tumor types that more often present MSI-H. Anti-PD(L)-1 antibodies have been demonstrated to be agnostically effective in patients with MSI-H cancer, but 50-60% of them do not respond to single-agent treatment, highlighting the necessity of expanding their treatment opportunities. Ipilimumab (anti-CTLA4) is the only immune checkpoint inhibitor (ICI) non-targeting PD(L)-1 that has been approved so far by the FDA for MSI-H cancer, namely, CRC in combination with nivolumab. Anti-TIM3 antibody LY3321367 showed interesting clinical activity in combination with anti-PDL-1 antibody in patients with MSI-H cancer not previously treated with anti-PD(L)-1. In contrast, no clinical evidence is available for anti-LAG3, anti-TIGIT, anti-BTLA, anti-ICOS and anti-IDO1 antibodies in MSI-H cancers, but clinical trials are ongoing. Other immunotherapeutic strategies under study for MSI-H cancers include vaccines, systemic immunomodulators, STING agonists, PKM2 activators, T-cell immunotherapy, LAIR-1 immunosuppression reversal, IL5 superagonists, oncolytic viruses and IL12 partial agonists. In conclusion, several combination therapies of ICIs and novel strategies are emerging and may revolutionize the treatment paradigm of MSI-H patients in the future. A huge effort will be necessary to find reliable immune biomarkers to personalize therapeutical decisions.
Collapse
Affiliation(s)
- Edoardo Crimini
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy (G.P.); (G.A.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Luca Boscolo Bielo
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy (G.P.); (G.A.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Pier Paolo Maria Berton Giachetti
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy (G.P.); (G.A.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Gloria Pellizzari
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy (G.P.); (G.A.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Gabriele Antonarelli
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy (G.P.); (G.A.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Beatrice Taurelli Salimbeni
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy (G.P.); (G.A.)
| | - Matteo Repetto
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Carmen Belli
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy (G.P.); (G.A.)
| | - Giuseppe Curigliano
- Division of Early Drug Development, European Institute of Oncology, IRCCS, Via Giuseppe Ripamonti 435, 20141 Milan, Italy (G.P.); (G.A.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| |
Collapse
|
10
|
Zhang L, Guo S, Chang S, Jiang G. Revolutionizing Cancer Treatment: Unleashing the Power of Combining Oncolytic Viruses with CAR-T Cells. Anticancer Agents Med Chem 2024; 24:1407-1418. [PMID: 39051583 DOI: 10.2174/0118715206308253240723055019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 06/07/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024]
Abstract
Oncolytic Viruses (OVs) have emerged as a promising treatment option for cancer thanks to their significant research potential and encouraging results. These viruses exert a profound impact on the tumor microenvironment, making them effective against various types of cancer. In contrast, the efficacy of Chimeric antigen receptor (CAR)-T cell therapy in treating solid tumors is relatively low. The combination of OVs and CAR-T cell therapy, however, is a promising area of research. OVs play a crucial role in enhancing the tumor-suppressive microenvironment, which in turn enables CAR-T cells to function efficiently in the context of solid malignancies. This review aims to provide a comprehensive analysis of the benefits and drawbacks of OV therapy and CAR-T cell therapy, with a focus on the potential of combining these two treatment approaches.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - ShuXian Guo
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - ShuYing Chang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| | - Guan Jiang
- Department of Dermatology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, China
| |
Collapse
|
11
|
Shen Z, Liu X, Fan G, Na J, Liu Q, Lin F, Zhang Z, Zhong L. Improving the therapeutic efficacy of oncolytic viruses for cancer: targeting macrophages. J Transl Med 2023; 21:842. [PMID: 37993941 PMCID: PMC10666393 DOI: 10.1186/s12967-023-04709-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/07/2023] [Indexed: 11/24/2023] Open
Abstract
Oncolytic viruses (OVs) for cancer treatment are in a rapid stage of development, and the direct tumor lysis and activation of a comprehensive host immune response are irreplaceable advantages of cancer immunotherapy. However, excessive antiviral immune responses also restrict the spread of OVs in vivo and the infection of tumor cells. Macrophages are functionally diverse innate immune cells that phagocytose tumor cells and present antigens to activate the immune response, while also limiting the delivery of OVs to tumors. Studies have shown that the functional propensity of macrophages between OVs and tumor cells affects the overall therapeutic effect of oncolytic virotherapy. How to effectively avoid the restrictive effect of macrophages on OVs and reshape the function of tumor-associated macrophages in oncolytic virotherapy is an important challenge we are now facing. Here, we review and summarize the complex dual role of macrophages in oncolytic virotherapy, highlighting how the functional characteristics of macrophage plasticity can be utilized to cooperate with OVs to enhance anti-tumor effects, as well as highlighting the importance of designing and optimizing delivery modalities for OVs in the future.
Collapse
Affiliation(s)
- Zhen Shen
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Guixiang Fan
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Jintong Na
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Qiaoqiao Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China
| | - Faquan Lin
- Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| | - Zhikun Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China.
| | - Liping Zhong
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, 530021, China.
- Pharmaceutical College, Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
12
|
Marques da Silva B, Mayne KJ, Zakrocka I. The potential association between influenza vaccination and lower incidence of renal cell carcinoma. Clin Kidney J 2023; 16:1714-1717. [PMID: 37915932 PMCID: PMC10616471 DOI: 10.1093/ckj/sfad180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Indexed: 11/03/2023] Open
Abstract
It is well-established that kidney cancer or renal cell carcinoma (RCC) occurs more commonly in chronic kidney disease (CKD) than in the general population, although the underlying mechanisms are incompletely understood. Beyond hereditary RCC syndromes; smoking, obesity and hypertension are widely known risk factors for RCC, irrespective of CKD. Kidney-specific factors such as episodes of acute kidney injury, nephrolithiasis and cyst formation have also been shown to be associated with RCC development. One potential and less explored factor is the role of viruses in the development of kidney cancer. In this issue of Clinical Kidney Journal, Lin et al. raise the interesting hypothesis that influenza vaccination may be associated with lower incidence of RCC in adults with CKD. We discuss potential mechanisms underlying this interesting observation in the context of immune dysregulation in CKD.
Collapse
Affiliation(s)
- Bernardo Marques da Silva
- Department of Nephrology and Renal Transplantation, Centro Hospitalar Universitário Lisboa Norte, Lisboa, Portugal
| | - Kaitlin J Mayne
- Nuffield Department of Population Health, University of Oxford, Oxford, UK & School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | | |
Collapse
|
13
|
Lahimchi MR, Maroufi F, Maali A. Induced Pluripotent Stem Cell-Derived Chimeric Antigen Receptor T Cells: The Intersection of Stem Cells and Immunotherapy. Cell Reprogram 2023; 25:195-211. [PMID: 37782910 DOI: 10.1089/cell.2023.0041] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy is a promising cell-based immunotherapy applicable to various cancers. High cost of production, immune rejection, heterogeneity of cell product, limited cell source, limited expandability, and relatively long production time have created the need to achieve a universal allogeneic CAR-T cell product for "off-the-shelf" application. Since the innovation of induced pluripotent stem cells (iPSCs) by Yamanaka et al., extensive efforts have been made to prepare an unlimited cell source for regenerative medicine, that is, immunotherapy. In the autologous grafting approach, iPSCs prepare the desired cell source for generating autologous CAR-T cells through more accessible and available sources. In addition, generating iPSC-derived CAR-T cells is a promising approach to achieving a suitable source for producing an allogeneic CAR-T cell product. In brief, the first step is reprogramming somatic cells (accessible from peripheral blood, skin, etc.) to iPSCs. In the next step, CAR expression and T cell lineage differentiation should be applied in different arrangements. In addition, in an allogeneic manner, human leukocyte antigen/T cell receptor (TCR) deficiency should be applied in iPSC colonies. The allogeneic iPSC-derived CAR-T cell experiments showed that simultaneous performance of HLA/TCR deficiency, CAR expression, and T cell lineage differentiation could bring the production to the highest efficacy in generating allogeneic iPSC-derived CAR-T cells.
Collapse
Affiliation(s)
| | - Faezeh Maroufi
- Department of Medical Laboratory Sciences, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Amirhosein Maali
- Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
14
|
Hervás-Corpión I, Alonso MM. Oncolytic viruses as treatment for adult and pediatric high-grade gliomas: On the way to clinical success. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 379:169-188. [PMID: 37541723 DOI: 10.1016/bs.ircmb.2023.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/06/2023]
Abstract
High-grade gliomas (HGGs) are the most common and aggressive primary brain tumors in both adult and pediatric populations. Despite the multimodal treatment modality currently available for HGG, the prognosis is dismal, with a low overall survival rate at two years after diagnosis. In the last decade, oncolytic virotherapy has emerged as a promising and feasible therapeutic tool in management of these tumors due to its oncolytic and immunostimulatory properties. Various oncolytic viruses, such as herpes simplex virus, adenovirus, poliovirus, reovirus, parvovirus and others, have been evaluated in the early stages of the clinical setting with regard to improving the outcome of patients with HGG. In this review, we summarize completed and ongoing clinical trials of oncolytic virotherapy for adult and pediatric malignant gliomas in terms of safety and efficacy, followed by a brief discussion about the current status and future directions of this therapy in the brain tumor field.
Collapse
Affiliation(s)
- Irati Hervás-Corpión
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain; Solid Tumor Program, Center for the Applied Medical Research (CIMA), Pamplona, Navarra, Spain; Department of Pediatrics, Clínica Universidad de Navarra (CUN), Pamplona, Navarra, Spain.
| | - Marta M Alonso
- Health Research Institute of Navarra (IdiSNA), Pamplona, Navarra, Spain; Solid Tumor Program, Center for the Applied Medical Research (CIMA), Pamplona, Navarra, Spain; Department of Pediatrics, Clínica Universidad de Navarra (CUN), Pamplona, Navarra, Spain.
| |
Collapse
|
15
|
Zhang J, He Q, Mao D, Wang C, Huang L, Wang M, Zhang J. Efficacy and adverse reaction management of oncolytic viral intervention combined with chemotherapy in patients with liver metastasis of gastrointestinal malignancy. Front Oncol 2023; 13:1159802. [PMID: 37197423 PMCID: PMC10183573 DOI: 10.3389/fonc.2023.1159802] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/17/2023] [Indexed: 05/19/2023] Open
Abstract
Background The liver is a key target organ for colorectal and gastric cancer metastasis. One of the challenges in the treatment of colorectal and gastric cancers is the management of liver metastasis. This study aimed to investigate the efficacy, adverse effects, and coping strategies of oncolytic virus injection in patients with liver metastases of gastrointestinal malignancies. Methods We prospectively analyzed patients treated at Ruijin Hospital affiliated with Shanghai Jiao Tong University School of Medicine from June 2021 to October 2022. 47 patients with gastrointestinal cancer liver metastasis were included in the study. The data, including clinical manifestations, imaging, tumor markers, postoperative adverse reactions, psychological intervention, dietary guidance, and adverse reaction management were evaluated. Results Oncolytic virus injection was successful in all patients, and no drug injection-related deaths occurred. The adverse effects, such as fever, pain, bone marrow suppression, nausea, and vomiting, were mild and resolved subsequently. Based on the comprehensive intervention of nursing procedures, the postoperative adverse reactions of patients were effectively alleviated and treated. None of the 47 patients had any puncture point infections, and the pain caused by the invasive operation was relieved quickly. After 2 courses of oncolytic virus injection, postoperative liver MRI showed 5 partial remissions, 30 stable diseases, and 12 progressive diseases in target organs. Conclusion Interventions based on nursing procedures can ensure the smooth treatment of recombinant human adenovirus type 5 in patients with liver metastases of gastrointestinal malignant tumors. This is of great importance for clinical treatment and significantly reduces patient complications and improves the patient's quality of life.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Nursing, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianyun He
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dongliang Mao
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Wang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Huang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Medical Center on Aging of Ruijin Hospital (MCARJH), Shanghai Jiao Tong University, School of Medicine, Shanghai, China
- *Correspondence: Mei Wang, ; Lei Huang,
| | - Mei Wang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Mei Wang, ; Lei Huang,
| | - Jun Zhang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|