1
|
Wang C, Zhou L, Kang X, Huang CH, Gao Z, Shen J, Wu S, Wu S, Cai Y, Chen W, Dai S, Chen P. A nanoplasmonic cell-on-a-chip for in situ monitoring of PD-L1 + exosome-mediated immune modulation. Biosens Bioelectron 2025; 277:117293. [PMID: 39999609 PMCID: PMC11996229 DOI: 10.1016/j.bios.2025.117293] [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: 10/03/2024] [Revised: 02/11/2025] [Accepted: 02/20/2025] [Indexed: 02/27/2025]
Abstract
Despite the transformative impact of immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 pathway in cancer therapy, up to 80% of patients fail to respond, necessitating reliable predictive biomarkers to guide treatment decisions. Recent studies highlight the critical role of tumor-derived exosomal PD-L1 in immune evasion, and its potential as a diagnostic and prognostic biomarker in cancer immunotherapy. However, significant challenges remain in elucidating the functional roles of PD-L1+ exosomes in immune suppression, as current methods lack the ability to precisely and simultaneously characterize and monitor exosome secretion and the corresponding immune modulation on site. To address this, we developed an integrated microfluidic platform that combines a digital nanoplasmonic immunoassay with a cell-on-a-chip system, enabling in situ monitoring of PD-L1+ exosome secretion and exosome-mediated T cell immune responses. This nanoplasmonic immunoassay integrated cell-on-a-chip (NIIC) creates a localized co-cultured microenvironment that facilitates exosome-mediated cellular interactions without direct contact. The NIIC employs machine-learning assisted signal processing for highly sensitive detection of both exosomes and cytokines, providing spatial and quantitative analysis of immune modulation in situ. Using this system, we demonstrated that PD-L1+ exosomes from cancer cells significantly suppressed IFN-γ and IL-2 secretion in neighboring T cells, offering direct insights into exosome-mediated immune suppression. The NIIC platform represents a powerful tool for advancing the understanding of exosome-driven immune modulation and holds potential for predicting clinical responses to anti-PD-1/PD-L1 therapies, paving the way for more personalized cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Chuanyu Wang
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849
| | - Lang Zhou
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849
| | - Xuejia Kang
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849; Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Chung-Hui Huang
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Zhuangqiang Gao
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849
| | - Jialiang Shen
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849
| | - Shuai Wu
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849
| | - Siqi Wu
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849
| | - Yuxin Cai
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Siyuan Dai
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849
| | - Pengyu Chen
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849.
| |
Collapse
|
2
|
Garg P, Ramisetty S, Nair M, Kulkarni P, Horne D, Salgia R, Singhal SS. Strategic advancements in targeting the PI3K/AKT/mTOR pathway for Breast cancer therapy. Biochem Pharmacol 2025; 236:116850. [PMID: 40049296 DOI: 10.1016/j.bcp.2025.116850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/17/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
Breast cancer (BC) is a complex disease that affects millions of women worldwide. Its growing impact calls for advanced treatment strategies to improve patient outcomes. The PI3K/AKT/mTOR pathway is a key focus in BC therapy because it plays a major role in important processes like tumor growth, survival, and resistance to treatment. Targeting this pathway could lead to better treatment options and outcomes. The present review explores how the PI3K/AKT/mTOR pathway becomes dysregulated in BC, focusing on the genetic changes like PIK3CA mutations and PTEN loss that leads to its aggravation. Current treatment options include the use of inhibitors targeting PI3K, AKT, and mTOR with combination therapies showing promise in overcoming drug resistance and improving effectiveness. Looking ahead, next-generation inhibitors and personalized treatment plans guided by biomarker analysis may provide more accurate and effective options for patients. Integrating these pathway inhibitors with immunotherapy offers an exciting opportunity to boost anti-tumor responses and improve survival rates. This review offers a comprehensive summary of the current progress in targeting the PI3K/AKT/mTOR pathway in BC. It highlights future research directions and therapeutic strategies aimed at enhancing patient outcomes and quality of life.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Sravani Ramisetty
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Meera Nair
- William J. Brennan High School, San Antonio, TX 78253, USA
| | - Prakash Kulkarni
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sharad S Singhal
- Department of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
3
|
Yan C, Wang G. Advances in research on flavonoids in tumor immunotherapy (Review). Mol Med Rep 2025; 31:150. [PMID: 40211703 PMCID: PMC11995692 DOI: 10.3892/mmr.2025.13515] [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: 09/03/2024] [Accepted: 02/25/2025] [Indexed: 04/16/2025] Open
Abstract
Cancer immunotherapy is an approach used in anti‑tumor treatment; however, its efficacy is limited to specific tumor types that are inherently sensitive to immune system modulation. Expanding the scope of indications and enhancing the efficacy of cancer immunotherapy are key goals for continued advancement. Flavonoids modulate the tumor‑immunosuppressive microenvironment. Integrating flavonoids with immunotherapeutic modalities, including cancer vaccines, immune checkpoint inhibitors and adoptive immune‑cell therapy, has potential in terms of augmenting the therapeutic efficacy of immunotherapy. The present review aimed to summarize flavonoids that enhance cancer immunotherapy, focusing on their underlying mechanisms and the application of nanotechnology to overcome inherent limitations such as poor solubility, low bioavailability, rapid metabolism, and instability under physiological conditions, thereby highlighting the potential of flavonoids in advancing cancer immunotherapy.
Collapse
Affiliation(s)
- Chaoguang Yan
- Department of Oncology, Weifang Chinese Medicine Hospital, Weifang, Shandong 261000 P.R. China
| | - Guangchun Wang
- Department of Oncology, Weifang Chinese Medicine Hospital, Weifang, Shandong 261000 P.R. China
| |
Collapse
|
4
|
Clabeaux CE, Rana HS, Patadia AH, Dertinger JE, Germann C, Allen RC. Oculofacial plastic surgery in the cancer patient: A narrative review. Eur J Ophthalmol 2025; 35:856-865. [PMID: 39648598 DOI: 10.1177/11206721241301808] [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: 12/10/2024]
Abstract
With the advances in systemic therapy and radiotherapy, the life expectancy of patients battling cancer has increased. The oculofacial plastic surgeon should be aware of the potential impacts a patient in remission or one being actively treated may pose. The goal of this review is to discuss the considerations a surgeon should have in this patient population.MethodsThe authors performed a computerized search using PubMed, Embase, and Google Scholar. The search terms used were "chemotherapy AND surgery", "immunotherapy AND surgery", "radiotherapy AND surgery", "nutrition AND surgery", "(oculoplastic OR orbit OR eyelid OR lacrimal OR puncta) AND (chemotherapy OR immunotherapy OR radiotherapy)", "(facial OR facial plastic OR oculoplastic) AND (chemotherapy OR immunotherapy OR radiotherapy)", "(cancer OR malignancy) AND surgery", "(cancer OR malignancy) AND (surgery OR surgical) complications", "wound healing AND (cancer OR malignancy)", "infection AND (cancer OR malignancy)", "(bleeding OR blood loss) AND (cancer OR malignancy) AND surgery", "(chemotherapy OR immunotherapy OR radiotherapy) AND wound healing", "(chemotherapy OR immunotherapy OR radiotherapy) AND (bleeding OR blood loss)", "(chemotherapy OR immunotherapy OR radiotherapy) AND infection".ResultsA total of 89 articles, published from 1993 to 2023 in the English language or with English translations were included. Articles published earlier than 2000 were cited for foundational knowledge. References cited in the identified articles were also used to gather further data for the review.Conclusions and RelevancePatients who are being treated for cancer or are undergoing current treatment for cancer require special considerations. Systemic therapies and radiotherapy impact the physiology of patients and the integrity of tissue in ways that significantly impact surgical interventions. It is imperative for the oculofacial plastic surgeon to have a complete understanding on how a previous or current diagnosis of cancer can influence surgical outcomes.
Collapse
Affiliation(s)
- Carson E Clabeaux
- Department of Ophthalmology, Madigan Army Medical Center, Tacoma, Washington, USA
| | - Harkaran S Rana
- Department of Facial Plastic Surgery, Trauma and Subspecialty Surgeons, Denver, Colorado, USA
| | - Amol H Patadia
- Department of Ophthalmology, Brooke Army Medical Center, San Antonio, Texas, USA
| | - Jake E Dertinger
- Department of Surgery, William Beaumont Army Medical Center, El Paso, Texas, USA
| | - Colby Germann
- Department of Medicine, Upstate Medical University, Syracuse, New York, USA
| | - Richard C Allen
- Department of Oculoplastic Surgery, Texas Oculoplastic Consultants: TOC Eye and Face, Austin, Texas, USA
| |
Collapse
|
5
|
Nejat Dehkordi A, Maddahi M, Vafa P, Ebrahimi N, Aref AR. Salivary biomarkers: a promising approach for predicting immunotherapy response in head and neck cancers. Clin Transl Oncol 2025; 27:1887-1920. [PMID: 39377974 DOI: 10.1007/s12094-024-03742-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 09/21/2024] [Indexed: 04/27/2025]
Abstract
Head and neck cancers, including cancers of the mouth, throat, voice box, salivary glands, and nose, are a significant global health issue. Radiotherapy and surgery are commonly used treatments. However, due to treatment resistance and disease recurrence, new approaches such as immunotherapy are being explored. Immune checkpoint inhibitors (ICIs) have shown promise, but patient responses vary, necessitating predictive markers to guide appropriate treatment selection. This study investigates the potential of non-invasive biomarkers found in saliva, oral rinses, and tumor-derived exosomes to predict ICI response in head and neck cancer patients. The tumor microenvironment significantly impacts immunotherapy efficacy. Oral biomarkers can provide valuable information on composition, such as immune cell presence and checkpoint expression. Elevated tumor mutation load is also associated with heightened immunogenicity and ICI responsiveness. Furthermore, the oral microbiota may influence treatment outcomes. Current research aims to identify predictive salivary biomarkers. Initial studies indicate that tumor-derived exosomes and miRNAs present in saliva could identify immunosuppressive pathways and predict ICI response. While tissue-based markers like PD-L1 have limitations, combining multiple oral fluid biomarkers could create a robust panel to guide treatment decisions and advance personalized immunotherapy for head and neck cancer patients.
Collapse
Affiliation(s)
| | - Moein Maddahi
- Faculty of Density, Yeditepe University, Istanbul, Turkey
| | - Parinaz Vafa
- Faculty of Density, Yeditepe University, Istanbul, Turkey
| | - Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran.
| | - Amir Reza Aref
- Mass General Cancer Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
6
|
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
|
7
|
Wu HH, Leng S, Eisenstat DD, Sergi C, Leng R. Targeting p53 for immune modulation: Exploring its functions in tumor immunity and inflammation. Cancer Lett 2025; 617:217614. [PMID: 40054656 DOI: 10.1016/j.canlet.2025.217614] [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: 01/16/2025] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/18/2025]
Abstract
p53, often referred to as the "guardian of the genome," is a critical regulator of cellular responses to stress. p53 plays a dual role in tumor suppression and immune regulation. In addition to its well-known functions of maintaining genomic stability and inducing apoptosis, p53 orchestrates a complex interaction between innate and adaptive immune responses. This involvement contributes to pathogen clearance, immune surveillance, and immunogenic cell death (ICD). This review explores the influence of p53 on immune dynamics, detailing its effects on macrophages, dendritic cells, natural killer cells (NK), T cells, and B cells. This review explains how mutations in p53 disrupt immune responses, promoting tumor immune evasion, and highlights its regulation of inflammatory cytokines and pattern recognition receptors. Furthermore, p53's role in ICD marks it as a key player in antitumor immunity, which has significant implications for cancer immunotherapy. The review also discusses the role of p53 in inflammation, autoimmune diseases, and chronic infections, revealing its dual function in promoting and suppressing inflammation through interactions with NF-κB signaling. Therapeutically, approaches that target p53, including wild-type p53 reactivation and combination therapies with immune checkpoint inhibitors, show considerable promise. Advances in high-throughput technologies, such as single-cell RNA sequencing and CRISPR screens, provide new insights into the immunological functions of p53, including its role in microbiome-immune interactions and immune senescence. This comprehensive review highlights the importance of incorporating immunological insights from p53 into innovative therapeutic strategies, addressing existing knowledge gaps, and paving the way for personalized medicine.
Collapse
Affiliation(s)
- H Helena Wu
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, T6G 2S2, Canada.
| | - Sarah Leng
- Department of Laboratory Medicine and Pathology (5B4. 09), University of Alberta, Edmonton, AB, T6G 2B7, Canada
| | - David D Eisenstat
- Department of Oncology, Cross Cancer Institute, 11560 University Ave., University of Alberta, Edmonton, Alberta, T6G 1Z2, Canada; Department of Pediatrics, University of Alberta, 11405 - 87 Ave., Edmonton, Alberta, T6G 1C9, Canada; Department of Medical Genetics, University of Alberta, 8613 114 Street, Edmonton, Alberta, T6G 2H7, Canada; Murdoch Children's Research Institute, Department of Paediatrics, University of Melbourne, 50 Flemington Road, Parkville, Victoria, 3052, Australia
| | - Consolato Sergi
- Department of Laboratory Medicine and Pathology (5B4. 09), University of Alberta, Edmonton, AB, T6G 2B7, Canada; Division of Anatomical Pathology, Children's Hospital of Eastern Ontario (CHEO), University of Ottawa, 401 Smyth Road Ottawa, ON, K1H 8L1, Canada
| | - Roger Leng
- 370 Heritage Medical Research Center, Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, T6G 2S2, Canada.
| |
Collapse
|
8
|
Azmal M, Miah MM, Prima FS, Paul JK, Haque ASNB, Ghosh A. Advances and challenges in cancer immunotherapy: Strategies for personalized treatment. Semin Oncol 2025; 52:152345. [PMID: 40305928 DOI: 10.1016/j.seminoncol.2025.152345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 05/02/2025]
Abstract
Cancer immunotherapy has transformed oncology by harnessing the immune system to specifically target cancer cells, offering reduced systemic toxicity compared to traditional therapies. This review highlights key strategies, including adoptive cell transfer (ACT), immune checkpoint inhibitors, oncolytic viral (OV) therapy, monoclonal antibodies (mAbs), and mRNA-based vaccines. ACT reinfuses enhanced immune cells like tumor-infiltrating lymphocytes (TILs) to combat refractory cancers, while checkpoint inhibitors (eg, PD-1 and CTLA-4 blockers) restore T-cell activity. OV therapy uses engineered viruses (eg, T-VEC) to selectively lyse cancer cells, and advanced mAbs improve targeting precision. mRNA vaccines introduce tumor-specific antigens to trigger robust immune responses. Despite significant progress, challenges like immune-related side effects, high costs, and immunosuppressive tumor microenvironments persist. This review underscores the need for combination strategies and precision medicine to overcome these barriers and maximize the potential of immunotherapy in personalized cancer treatment.
Collapse
Affiliation(s)
- Mahir Azmal
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Md Munna Miah
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Fatema Sultana Prima
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Jibon Kumar Paul
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Anm Shah Newaz Been Haque
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Ajit Ghosh
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh.
| |
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] [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
|
Ali A, Azmat U, Khatoon A, Akbar K, Murtaza B, Ji Z, Irshad U, Su Z. From gene editing to tumor eradication: The CRISPR revolution in cancer therapy. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 196:114-131. [PMID: 40250571 DOI: 10.1016/j.pbiomolbio.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/20/2025]
Abstract
Cancer continues to be a significant worldwide health concern, characterized by high rates of occurrence and death. Unfortunately, existing treatments frequently fall short of delivering satisfying therapeutic outcomes. Immunotherapy has ushered in a new era in the treatment of solid tumors, yet its effectiveness is still constrained and comes with unwanted side effects. The advancement of cutting-edge technology, propelled by gene analysis and manipulation at the molecular scale, shows potential for enhancing these therapies. The advent of genome editing technologies, including CRISPR-Cas9, can greatly augment the efficacy of cancer immunotherapy. This review explores the mechanism of CRISPR-Cas9-mediated genome editing and its wide range of tools. The study focuses on analyzing the effects of CRISPR-induced double-strand breaks (DSBs) on cancer immunotherapy, specifically by gene knockdown or knockin. In addition, the study emphasizes the utilization of CRISPR-Cas9-based genome-wide screening to identify targets, the potential of spatial CRISPR genomics, and the extensive applications and difficulties of CRISPR-Cas9 in fundamental research, translational medicine, and clinical environments.
Collapse
Affiliation(s)
- Ashiq Ali
- Department of Histology and Embryology, Shantou University Medical College, Shantou, China.
| | - Urooj Azmat
- Department of Zoology, Wildlife and Fisheries, Faculty of Sciences, University of Agriculture, Faisalabad, 38040, Punjab, Pakistan
| | - Aisha Khatoon
- Department of Pathology, University of Agriculture, Faisalabad, Pakistan
| | - Kaynaat Akbar
- Department of Zoology, Wildlife and Fisheries, Faculty of Sciences, University of Agriculture, Faisalabad, 38040, Punjab, Pakistan
| | - Bilal Murtaza
- School of Bioengineering, Dalian University of Science and Technology Dalian, China
| | - Ziyi Ji
- Department of Histology and Embryology, Shantou University Medical College, Shantou, China
| | - Urooj Irshad
- Department of Zoology, Faculty of Sciences, Superior University, Lahore, Pakistan
| | - Zhongjing Su
- Department of Histology and Embryology, Shantou University Medical College, Shantou, China.
| |
Collapse
|
11
|
Ryba-Stanisławowska M. Unraveling Th subsets: insights into their role in immune checkpoint inhibitor therapy. Cell Oncol (Dordr) 2025; 48:295-312. [PMID: 39325360 PMCID: PMC11996958 DOI: 10.1007/s13402-024-00992-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2024] [Indexed: 09/27/2024] Open
Abstract
T helper (Th) cell subsets play pivotal roles in regulating immune responses within the tumor microenvironment, influencing both tumor progression and anti-tumor immunity. Among these subsets, Th1 cells promote cytotoxic responses through the production of IFN-γ, while Th2 cells and regulatory T cells (Tregs) exert immunosuppressive effects that support tumor growth. Th9 and Th17 cells have context-dependent roles, contributing to both pro-inflammatory and regulatory processes in tumor immunity. Tumor antigen-specific T cells within the tumor microenvironment often exhibit a dysfunctional phenotype due to increased expression of inhibitory receptors such as CTLA-4 and PD-1, leading to reduced antitumor activity. Monoclonal antibodies that block these inhibitory signals-collectively known as immune checkpoint inhibitors (ICIs)-can reactivate these T cells, enhancing their ability to target and destroy cancer cells. Recent advancements have highlighted the critical role of T helper subsets in modulating responses to ICIs, with their interactions remaining a focus of ongoing research. Both positive and negative effects of ICIs have been reported in relation to Th cell subsets, with some effects depending on the type of tumor microenvironment. This review summarizes the crucial roles of different T helper cell subsets in tumor immunity and their complex relationship with immune checkpoint inhibitor therapy.
Collapse
Affiliation(s)
- Monika Ryba-Stanisławowska
- Department of Medical Immunology, Faculty of Medicine, Medical University of Gdańsk, Dębinki 1, Gdańsk, 80-211, Poland.
| |
Collapse
|
12
|
Zheng B, Wang H, Zhai S, Li J, Lu K. Mitochondria-targeted photothermal-chemodynamic therapy enhances checkpoint blockade immunotherapy on colon cancer. Mater Today Bio 2025; 31:101542. [PMID: 40018055 PMCID: PMC11867542 DOI: 10.1016/j.mtbio.2025.101542] [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: 12/02/2024] [Revised: 01/28/2025] [Accepted: 02/01/2025] [Indexed: 03/01/2025] Open
Abstract
Immunotherapy has emerged as a hotspot for cancer treatment. However, the response rate of monotherapy remains relatively low in clinical settings. Photothermal therapy (PTT), which employs light energy to ablate tumors, can also activate tumor-specific immune responses. This effect has been attributed in several studies to the release of damage-associated molecular patterns (DAMPs) triggered by mitochondrial injury. We propose that mitochondria-targeted PTT may better synergize with immunotherapy. Herein, we constructed a multifunctional nanoplatform that enables mitochondria-targeted photothermal-chemodynamic combination therapy by conjugating indocyanine green-thiol (ICG-SH) and mercaptoethyl-triphenylphosphonium (TPP-SH) onto polyvinyl pyrrolidone (PVP)-coated gold-copper nanoparticles (AIT). Upon near-infrared light (NIR) irradiation, AIT ablates cancer cells and amplifies the effect of chemodynamic therapy (CDT), thereby inducing apoptosis in the tumor. The combination of CDT and PTT promotes immunogenic cell death, which could synergize with checkpoint blockade immunotherapy. In a bilateral mouse colon cancer model, we observed complete eradication of light-irradiated primary tumors and significant inhibition of distant untreated tumors in the group treated with AIT plus anti-PD-1 (αPD-1). We found a significant increase in serum levels of pro-inflammatory factors, including interleukin-6 (IL-6), interferon-γ (IFN-γ), and tumor necrosis factor-α (TNF-α), following PTT/CDT/immunotherapy treatment, suggesting effective activation of the immune response. The enhanced immunogenicity caused by AIT with αPD-1 treatment resulted in efficient antigen presentation, as indicated by the increased infiltration of dendritic cells (DCs) into the tumor-draining lymph nodes (LNs). We also observed enhanced infiltration of CD8+ T cells in distant tumors in the AIT with αPD-1 group compared to αPD-1 alone. Hence, mitochondria-targeting represents an effective strategy to potentiate the combination of photothermal, chemodynamic, and immune checkpoint blockade therapies for the treatment of metastatic cancer.
Collapse
Affiliation(s)
- Benchao Zheng
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, PR China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, PR China
| | - Hongbo Wang
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, PR China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, PR China
| | - Shiyi Zhai
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, PR China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, PR China
| | - Jiangsheng Li
- Key Laboratory of Carcinogenesis and Translational Research of Ministry of Education, Key Laboratory for Research and Evaluation of Radiopharmaceuticals of National Medical Products Administration, Department of Nuclear Medicine, Peking University Cancer Hospital, Beijing, 100142, PR China
| | - Kuangda Lu
- Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, PR China
- Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, PR China
| |
Collapse
|
13
|
Mivehchi H, Eskandari-Yaghbastlo A, Ghazanfarpour M, Ziaei S, Mesgari H, Faghihinia F, Zokaei Ashtiani N, Afjadi MN. Microenvironment-based immunotherapy in oral cancer: a comprehensive review. Med Oncol 2025; 42:140. [PMID: 40153139 DOI: 10.1007/s12032-025-02694-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/19/2025] [Indexed: 03/30/2025]
Abstract
Oral cancer, a prevalent form of head and neck malignancy, accounts for 4% of global cancer cases. The most common type, oral squamous cell carcinoma (OSCC), has a survival rate of about 50%. Even though emerging molecular therapies show promise for managing oral cancer, current treatments like surgery, radiotherapy, and chemotherapy have significant side effects. In addition, the complex tumor microenvironment (TME), involving the extracellular matrix (ECM) and cells like fibroblasts and stromal cells like immune cells, promotes tumor growth and inhibits immune responses, complicating treatment. Nonetheless, immunotherapy is crucial in cancer treatment, especially in oral cancers. Indeed, its effectiveness lies in targeting immune checkpoints such as PD-1 and CTLA-4 inhibitors, as well as monoclonal antibodies like pembrolizumab and cetuximab, adoptive cell transfer methods (including CAR-T cell therapy), cytokine therapy such as IL-2, and tumor vaccines. Thus, these interventions collectively regulate tumor proliferation and metastasis by targeting the TME through autocrine-paracrine signaling pathways. Immunotherapy indeed aims to stimulate the immune system, leveraging both innate and adaptive immunity to counteract cancer cell signals and promote tumor destruction. This review will explore how the TME controls tumor proliferation and metastasis via autocrine-paracrine signaling pathways. It will then detail the effectiveness of immunotherapy in oral cancers, focusing on immune checkpoints, targeted monoclonal antibodies, adoptive cell transfer, cytokine therapy, and tumor vaccines.
Collapse
Affiliation(s)
- Hassan Mivehchi
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | | | | | - SeyedMehdi Ziaei
- Faculty of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hassan Mesgari
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Islamic Azad University, Tehran Branch, Tehran, Iran
| | - Farbod Faghihinia
- School of Dentistry, Yasuj University of Medical Sciences, Yasuj, Iran
| | | | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
14
|
Suzuki T, Mizuno K, Yamamoto T, Ito T, Ishikawa T, Honda T, Akamatsu S, Ishii M, Ando Y, Kawashima H. Clinical characteristics of immune checkpoint inhibitor-related pancreatic injury with pancreatitis in patients with advanced malignancies. Dig Liver Dis 2025:S1590-8658(25)00273-7. [PMID: 40155252 DOI: 10.1016/j.dld.2025.02.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/10/2025] [Accepted: 02/24/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND/PURPOSE Immune checkpoint inhibitor (ICIs) therapy can cause immune checkpoint inhibitor-related pancreatic injury (ICI-PI). This study aimed to elucidate the clinical characteristics of ICI-PI and explore treatment approaches. METHODS We retrospectively analyzed data from patients with malignancies treated with ICIs at Nagoya University Hospital between September 2014 and June 2023. ICI-PI and pancreatitis were identified and classified using the Common terminology Criteria for Adverse Events (CTCAE) ver.5.0, and the patients with ICI-PI with/without pancreatitis were analyzed. RESULTS This study included data from 930 patients treated with ICIs. Of these, eight (0.85 %) and 10 (1.08 %) patients were diagnosed with ICI-PI without and with pancreatitis, respectively. Five of the 10 patients received standard pancreatitis treatment, while three received additional glucocorticoid therapy. One patient experienced recurrent pancreatitis and developed a pancreatic pseudocyst, which improved after high-dose glucocorticoid treatment. ICI rechallenge was pursued in five patients. Two patients received ICIs without an interval. CONCLUSIONS ICI-PI with pancreatitis is rare but has become more prevalent with the increasing use of ICIs. Future prospective multicenter studies are needed to confirm these findings and develop standardized diagnostic and treatment protocols.
Collapse
Affiliation(s)
- Takaya Suzuki
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Japan
| | - Kazuyuki Mizuno
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Japan; Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Japan.
| | - Takafumi Yamamoto
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Japan
| | - Takanori Ito
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Japan
| | - Takuya Ishikawa
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Japan
| | - Takashi Honda
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Japan
| | - Shusuke Akamatsu
- Department of Urology, Nagoya University Graduate School of Medicine, Japan
| | - Makoto Ishii
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Japan
| | - Yuichi Ando
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Japan
| |
Collapse
|
15
|
Singh SR, Bhaskar R, Ghosh S, Yarlagadda B, Singh KK, Verma P, Sengupta S, Mladenov M, Hadzi-Petrushev N, Stojchevski R, Sinha JK, Avtanski D. Exploring the Genetic Orchestra of Cancer: The Interplay Between Oncogenes and Tumor-Suppressor Genes. Cancers (Basel) 2025; 17:1082. [PMID: 40227591 PMCID: PMC11988167 DOI: 10.3390/cancers17071082] [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: 02/10/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Cancer is complex because of the critical imbalance in genetic regulation as characterized by both the overexpression of oncogenes (OGs), mainly through mutations, amplifications, and translocations, and the inactivation of tumor-suppressor genes (TSGs), which entail the preservation of genomic integrity by inducing apoptosis to counter the malignant growth. Reviewing the intricate molecular interplay between OGs and TSGs draws attention to their cell cycle, apoptosis, and cancer metabolism regulation. In the present review, we discuss seminal discoveries, such as Knudson's two-hit hypothesis, which framed the field's understanding of cancer genetics, leading to the next breakthroughs with next-generation sequencing and epigenetic profiling, revealing novel insights into OG and TSG dysregulation with opportunities for targeted therapy. The key pathways, such as MAPK/ERK, PI3K/AKT/mTOR, and Wnt/β-catenin, are presented in the context of tumor progression. Importantly, we further highlighted the advances in therapeutic strategies, including inhibitors of KRAS and MYC and restoration of TSG function, despite which mechanisms of resistance and tumor heterogeneity pose daunting challenges. A high-level understanding of interactions between OG-TSGs forms the basis for effective, personalized cancer treatment-something to strive for in better clinical outcomes. This synthesis should integrate foundational biology with translation and, in this case, contribute to the ongoing effort against cancer.
Collapse
Affiliation(s)
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeongsan-si 38541, Republic of Korea;
- Research Institute of Cell Culture, Yeungnam University, Gyeongsan-si 38541, Republic of Korea
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida 201301, India
| | | | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Symbiosis International (Deemed University), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune 411057, India
| | - Prashant Verma
- School of Management, BML Munjal University, NH8, Sidhrawali, Gurugram 122413, India
| | - Sonali Sengupta
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Mitko Mladenov
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia
| | - Nikola Hadzi-Petrushev
- Faculty of Natural Sciences and Mathematics, Institute of Biology, Ss. Cyril and Methodius University, 1000 Skopje, North Macedonia
| | - Radoslav Stojchevski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| | | | - Dimiter Avtanski
- Friedman Diabetes Institute, Lenox Hill Hospital, Northwell Health, New York, NY 10022, USA
- Feinstein Institutes for Medical Research, Manhasset, NY 11030, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY 11549, USA
| |
Collapse
|
16
|
Zhang S, Chen W, Zhou J, Liang Q, Zhang Y, Su M, Zhang Z, Qu J. The Benefits and Safety of Monoclonal Antibodies: Implications for Cancer Immunotherapy. J Inflamm Res 2025; 18:4335-4357. [PMID: 40162076 PMCID: PMC11952073 DOI: 10.2147/jir.s499403] [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: 10/05/2024] [Accepted: 03/06/2025] [Indexed: 04/02/2025] Open
Abstract
Monoclonal antibodies (mAbs) have transformed cancer treatment by providing highly targeted and effective therapies that specifically attack cancer cells, thus reducing the likelihood of adverse events (AEs) in patients. mAbs exert their action through various mechanisms, such as receptor blockade, antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and inhibition of immune checkpoints (eg, PD-1, PD-L1, and CTLA-4). These therapies have led to significant improvements in the treatment of several cancers, including HER2-positive breast cancer, non-small cell lung cancer (NSCLC), and melanoma. The efficacy of mAb therapy in cancer treatment is influenced by various intrinsic and extrinsic factors, such as environmental exposures, psychosocial factors, infection status, ways of life, and tumor microenvironment (TME), all of which can impact immune responses and treatment outcomes. Notably, the therapeutic benefits of mAbs are often accompanied by immune-related AEs (irAEs), which can vary from mild to severe and affect multiple organ systems. The dual nature of mAbs-stimulating antitumor immune responses while also inducing immune-related side effects-presents a notable challenge in clinical practice. This review highlights the importance of proactive strategies for managing irAEs, such as early detection, corticosteroid use, targeted immunosuppressive treatments, and the urgent need for reliable predictive biomarkers to improve treatment outcomes. Advancements in the prevention, prediction, and management of irAEs are essential to enhance the safety and effectiveness of mAb-based therapies, ultimately aiming to improve cancer patient outcomes.
Collapse
Affiliation(s)
- Shuguang Zhang
- Department of Pharmacy, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Wenying Chen
- Department of Pharmacy, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, People’s Republic of China
| | - Jihong Zhou
- Department of Respiratory and Critical Care Medicine, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Qi Liang
- Department of Pharmacy, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Yu Zhang
- Department of Intensive Care Unit, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Ming Su
- Department of Respiratory and Critical Care Medicine, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Zilong Zhang
- Department of Pharmacy, Shenzhen Bao’an Chinese Medicine Hospital, The Seventh Clinical College of Guangzhou University of Chinese Medicine, Shenzhen, People’s Republic of China
| | - Jian Qu
- Department of Pharmacy, the Second Xiangya Hospital, Central South University, Institute of Clinical Pharmacy, Central South University, Changsha, People’s Republic of China
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, People’s Republic of China
| |
Collapse
|
17
|
Murariu MO, Boia ER, Sitaru AM, Mot CI, Negru MC, Brici AC, Zahoi DE, Balica NC. Long-Term Quality of Life and Functional Outcomes in Patients with Total Laryngectomy. Cancers (Basel) 2025; 17:1011. [PMID: 40149346 PMCID: PMC11941457 DOI: 10.3390/cancers17061011] [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: 02/23/2025] [Revised: 03/14/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Laryngeal cancer affects quality of life (QoL), speech, and swallowing. Total laryngectomy (TL) causes severe impairments, while partial laryngectomy (PL) and chemoradiotherapy (CRT) preserve the organ but yield variable outcomes. This study assesses QoL, speech rehabilitation, swallowing, and social reintegration across these treatments. METHODS This prospective observational cohort study was conducted at the ENT Clinic, Victor Babeș University of Medicine and Pharmacy, Timișoara; recruitment was conducted between October 2019 and January 2024. Seventy-five patients diagnosed with laryngeal squamous cell carcinoma (LSCC) were initially enrolled but only 15 patients (20%) completed the 12-month follow-up, with an attrition rate of 80%. Tumor stages ranged from T1 to T4a, with TL patients having a higher proportion of advanced-stage disease (Stage III-IV: 76%) compared to PL (45%) and CRT (50%). Validated instruments, including the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire for Head and Neck Cancer (EORTC QLQ-H&N35), the Voice Handicap Index-30 (VHI-30), the Hospital Anxiety and Depression Scale (HADS), and the Dysphagia Outcome and Severity Scale (DOSS), were used to assess QoL, voice function, swallowing function, and psychological impact. RESULTS At 12 months, the global QoL score from the EORTC QLQ-H&N35 was lowest in TL patients (49.8 ± 10.9), significantly lower than both PL (61.2 ± 9.6, p = 0.002) and CRT (64.1 ± 7.8, p < 0.001). Post hoc Bonferroni analysis confirmed significant pairwise differences between TL vs. PL (p = 0.002) and TL vs. CRT (p < 0.001), while the difference between PL and CRT was non-significant (p = 0.14). TL patients had higher speech-related disability (VHI: 88.3 ± 12.6) and dysphagia prevalence (DOSS: 4.0 ± 1.2), with 16% remaining enteral feeding-dependent. Anxiety (HADS-A: 7.5 ± 2.9) and depression (HADS-D: 9.0 ± 3.2) were highest in TL patients, with 36% meeting clinical depression criteria at 12 months. Multivariable regression identified TL (OR = 3.92, 95% CI: 2.14-5.79, p < 0.001) and advanced tumor stage (OR = 2.85, 95% CI: 1.79-4.21, p = 0.002) as strong predictors of poor QoL. Kaplan-Meier analysis showed no significant OS differences (p = 0.12), but CRT patients had lower DFS (78%) compared to TL (82%) and PL (85%) (p = 0.048). CONCLUSIONS TL patients experience the most significant impairments in QoL, speech, and social reintegration despite rehabilitation. CRT patients show higher recurrence rates but better QoL, while PL offers the best balance of function and survival. These findings highlight the need for long-term survivorship support tailored to treatment type.
Collapse
Affiliation(s)
- Maria Octavia Murariu
- Department of Doctoral Studies, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania;
| | - Eugen Radu Boia
- ENT Department, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (E.R.B.); (C.I.M.); (M.C.N.); (N.C.B.)
| | - Adrian Mihail Sitaru
- Department of Pediatric Surgery, “Louis Turcanu” Emergency Clinical Hospital for Children, Iosif Nemoianu Street 2, 300011 Timisoara, Romania;
| | - Cristian Ion Mot
- ENT Department, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (E.R.B.); (C.I.M.); (M.C.N.); (N.C.B.)
| | - Mihaela Cristina Negru
- ENT Department, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (E.R.B.); (C.I.M.); (M.C.N.); (N.C.B.)
| | | | - Delia Elena Zahoi
- Department of Anatomy and Embryology, “Victor Babes” University of Medicine and Pharmacy Timisoara, 300041 Timisoara, Romania
| | - Nicolae Constantin Balica
- ENT Department, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (E.R.B.); (C.I.M.); (M.C.N.); (N.C.B.)
| |
Collapse
|
18
|
Almawash S. Revolutionary Cancer Therapy for Personalization and Improved Efficacy: Strategies to Overcome Resistance to Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2025; 17:880. [PMID: 40075727 PMCID: PMC11899125 DOI: 10.3390/cancers17050880] [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: 01/06/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a significant public health issue worldwide, standing as a primary contributor to global mortality, accounting for approximately 10 million fatalities in 2020 [...].
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
19
|
Liu J, Wang Y, Song Z, Zhang Y. Nanoengineered immune check point inhibitors delivery for targeted brain cancer treatment: Current status and future perspectives. Biochem Pharmacol 2025; 233:116789. [PMID: 39900203 DOI: 10.1016/j.bcp.2025.116789] [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: 11/18/2024] [Revised: 01/06/2025] [Accepted: 01/30/2025] [Indexed: 02/05/2025]
Abstract
Brain tumors create special difficulties because of their position and the protective covering of blood brain barrier (BBB) that restricts efficient medication access. Treatment alternatives such as surgery and chemotherapy demonstrate poor performance against severe brain tumors. The use of immune checkpoint inhibitors (ICIs) hints at effective cancer therapy; however, their application to brain cancer faces challenges due to inefficient delivery through the BBB and the tumor's suppressive environment. Nanoengineering can increase the transport of ICIs to brain tumors. Numerous nano-delivery systems such as liposomes and micelles have explored ways to avoid the BBB via transcytosis and the EPR mechanism. Functionalization of nanocarriers enhances targeting tumor cells and improves treatment accuracy. New developments involve delivering ICIs together with adjuvants to change the TME and focusing on immune cells such as TAMs and Tregs to boost immunity against tumors. Nanoengineered ICIs have shown effective improvement in animal models by reducing toxicity and enhancing efficacy. Converting these successes into real clinical trials is not easy as they face regulatory concerns and safety challenges. Clinical trials currently examine the use of nanocarriers for treating brain cancer; however, scalability' and 'long-term safety' continue to pose challenges. Future approaches will focus on combining customized medicine with advanced nanotechnology and AI to refine treatment methods. Despite obstacles ahead, nanotechnology-based ICIs offer a hopeful approach to enhance brain cancer efficacy and address existing therapeutic constraints.
Collapse
Affiliation(s)
- Juan Liu
- Department of General Medicine, the Second Hospital of Jilin University, Changchun 130000, China
| | - Yichao Wang
- Department of Obstetrics and Gynecology, the Second Hospital of Jilin University, Changchun 130000, China
| | - Zhidu Song
- Ophthalmology Department, the Second Hospital of Jilin University, Changchun 130000, China
| | - Yukai Zhang
- Neurosurgery Department, the Second Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
20
|
Du F, Wang G, Dai Q, Huang J, Li J, Liu C, Du K, Tian H, Deng Q, Xie L, Zhao X, Zhang Q, Yang L, Li Y, Wu Z, Zhang Z. Targeting novel regulated cell death: disulfidptosis in cancer immunotherapy with immune checkpoint inhibitors. Biomark Res 2025; 13:35. [PMID: 40012016 DOI: 10.1186/s40364-025-00748-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
The battle against cancer has evolved over centuries, from the early stages of surgical resection to contemporary treatments including chemotherapy, radiation, targeted therapies, and immunotherapies. Despite significant advances in cancer treatment over recent decades, these therapies remain limited by various challenges. Immune checkpoint inhibitors (ICIs), a cornerstone of tumor immunotherapy, have emerged as one of the most promising advancements in cancer treatment. Although ICIs, such as CTLA-4 and PD-1/PD-L1 inhibitors, have demonstrated clinical efficacy, their therapeutic impact remains suboptimal due to patient-specific variability and tumor immune resistance. Cell death is a fundamental process for maintaining tissue homeostasis and function. Recent research highlights that the combination of induced regulatory cell death (RCD) and ICIs can substantially enhance anti-tumor responses across multiple cancer types. In cells exhibiting high levels of recombinant solute carrier family 7 member 11 (SLC7A11) protein, glucose deprivation triggers a programmed cell death (PCD) pathway characterized by disulfide bond formation and REDOX (reduction-oxidation) reactions, termed "disulfidptosis." Studies suggest that disulfidptosis plays a critical role in the therapeutic efficacy of SLC7A11high cancers. Therefore, to investigate the potential synergy between disulfidptosis and ICIs, this study will explore the mechanisms of both processes in tumor progression, with the goal of enhancing the anti-tumor immune response of ICIs by targeting the intracellular disulfidptosis pathway.
Collapse
Affiliation(s)
- Fei Du
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Guojun Wang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qian Dai
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Jiang Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Junxin Li
- Department of pharmacy, Zigong Fourth People's Hospital, Zigong, 643000, China
| | - Congxing Liu
- Department of Pharmacy, Chengfei Hospital, Chengdu, 610000, China
| | - Ke Du
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pediatrics, Luzhou Maternal and Child Health Hospital, Luzhou Second People's Hospital, Luzhou, 646000, Sichuan, China
| | - Hua Tian
- School of Nursing, Chongqing College of Humanities, Science & Technology, Chongqing, 401520, China
| | - Qiwei Deng
- Heruida Pharmaceutical Co.,ltd, Haikou, Hainan, 570100, China
| | - Longxiang Xie
- The TCM Hospital of Longquanyi District, Chengdu, 610100, Sichuan, China
| | - Xin Zhao
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qimin Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Lan Yang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhigui Wu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhuo Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
21
|
Yang W, Hicks DR, Ghosh A, Schwartze TA, Conventry B, Goreshnik I, Allen A, Halabiya SF, Kim CJ, Hinck CS, Lee DS, Bera AK, Li Z, Wang Y, Schlichthaerle T, Cao L, Huang B, Garrett S, Gerben SR, Rettie S, Heine P, Murray A, Edman N, Carter L, Stewart L, Almo SC, Hinck AP, Baker D. Design of high-affinity binders to immune modulating receptors for cancer immunotherapy. Nat Commun 2025; 16:2001. [PMID: 40011465 PMCID: PMC11865580 DOI: 10.1038/s41467-025-57192-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 02/14/2025] [Indexed: 02/28/2025] Open
Abstract
Immune receptors have emerged as critical therapeutic targets for cancer immunotherapy. Designed protein binders can have high affinity, modularity, and stability and hence could be attractive components of protein therapeutics directed against these receptors, but traditional Rosetta based protein binder methods using small globular scaffolds have difficulty achieving high affinity on convex targets. Here we describe the development of helical concave scaffolds tailored to the convex target sites typically involved in immune receptor interactions. We employed these scaffolds to design proteins that bind to TGFβRII, CTLA-4, and PD-L1, achieving low nanomolar to picomolar affinities and potent biological activity following experimental optimization. Co-crystal structures of the TGFβRII and CTLA-4 binders in complex with their respective receptors closely match the design models. These designs should have considerable utility for downstream therapeutic applications.
Collapse
Affiliation(s)
- Wei Yang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Derrick R Hicks
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Agnidipta Ghosh
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Tristin A Schwartze
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian Conventry
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Inna Goreshnik
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Aza Allen
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Samer F Halabiya
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Chan Johng Kim
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Cynthia S Hinck
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - David S Lee
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Asim K Bera
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Zhe Li
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Yujia Wang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Thomas Schlichthaerle
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Longxing Cao
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Buwei Huang
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Sarah Garrett
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Stacey R Gerben
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Stephen Rettie
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Piper Heine
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Analisa Murray
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Natasha Edman
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Lauren Carter
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Lance Stewart
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Steven C Almo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Andrew P Hinck
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA.
- Institute for Protein Design, University of Washington, Seattle, WA, USA.
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|
22
|
Kushlinskii NE, Kovaleva OV, Gratchev AN, Alferov AA, Kuzmin YB, Sokolov NY, Tsekatunov DA, Ryzhavskaya IB, Kuznetsov IN, Kushlinskii DN, Mamedli ZZ, Stilidi IS. Assessing the Clinical Relevance of Soluble PD-1 and PD-L1: A Multi-Cohort Study Across Diverse Tumor Types and Prognostic Implications. Biomedicines 2025; 13:500. [PMID: 40002913 PMCID: PMC11852959 DOI: 10.3390/biomedicines13020500] [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: 12/30/2024] [Revised: 02/09/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Immune checkpoint inhibitors targeting the PD-1/PD-L1 pathway have revolutionized cancer immunotherapy, however the clinical relevance of their soluble forms (sPD-1 and sPD-L1) remains less studied. Soluble PD-1 and PD-L1 have been implicated in tumor progression, prognosis, and treatment response across various malignancies. This study aims to provide a comprehensive analysis of sPD-1 and sPD-L1 levels in serum across diverse tumor types, including rare malignancies, and to evaluate their associations with clinicopathological characteristics and prognostic significance. Methods: In this study we analyzed sPD-1 and sPD-L1 levels in serum samples from 675 cancer patients representing a range of malignancies, including ovarian cancer, breast cancer, gastric cancer, colorectal cancer, renal cell carcinoma, and bone tumors. sPD-1 and sPD-L1 concentrations were measured using ELISA. Statistical analyses were performed to evaluate associations between soluble marker concentrations and clinicopathological factors, including tumor stage, size, histological subtype, and survival outcomes. Results: Elevated sPD-L1 levels were observed in several tumor types, including ovarian cancer, renal cell carcinoma, and gastric cancer, where they were associated with features of advanced disease, such as tumor size, stage, and metastases. In contrast, sPD-1 levels showed limited associations, with significant findings solely in gastric cancer and bone tumors, where levels correlated with histological subtype and differentiation. Prognostic analyses identified sPD-L1 as a marker of poor survival outcomes in ovarian cancer and bone tumors, while sPD-1 displayed no consistent prognostic significance. Conclusions: This study identifies the potential of sPD-L1 as a biomarker for tumor progression and prognosis across multiple malignancies. In contrast, sPD-1 showed limited clinical relevance, suggesting the importance of further investigation. These findings contribute to our understanding of soluble immune checkpoint proteins and their integration into personalized oncology strategies.
Collapse
Affiliation(s)
- Nikolay E. Kushlinskii
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 115478 Moscow, Russia; (N.E.K.); (O.V.K.); (A.A.A.); (Y.B.K.); (N.Y.S.); (Z.Z.M.); (I.S.S.)
- Ministry of Health of the Russian Federation, Russian University of Medicine, 127473 Moscow, Russia;
| | - Olga V. Kovaleva
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 115478 Moscow, Russia; (N.E.K.); (O.V.K.); (A.A.A.); (Y.B.K.); (N.Y.S.); (Z.Z.M.); (I.S.S.)
| | - Alexei N. Gratchev
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 115478 Moscow, Russia; (N.E.K.); (O.V.K.); (A.A.A.); (Y.B.K.); (N.Y.S.); (Z.Z.M.); (I.S.S.)
- Laboratory for Tumor Stromal Cells Biology, Institute for Carcinogenesis, N.N. Blokhin Cancer Research Center, Kashirskoye Sh. 24, 115478 Moscow, Russia
| | - Alexander A. Alferov
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 115478 Moscow, Russia; (N.E.K.); (O.V.K.); (A.A.A.); (Y.B.K.); (N.Y.S.); (Z.Z.M.); (I.S.S.)
- Ministry of Health of the Russian Federation, Russian University of Medicine, 127473 Moscow, Russia;
| | - Yurii B. Kuzmin
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 115478 Moscow, Russia; (N.E.K.); (O.V.K.); (A.A.A.); (Y.B.K.); (N.Y.S.); (Z.Z.M.); (I.S.S.)
- Ministry of Health of the Russian Federation, Russian University of Medicine, 127473 Moscow, Russia;
| | - Nikolai Y. Sokolov
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 115478 Moscow, Russia; (N.E.K.); (O.V.K.); (A.A.A.); (Y.B.K.); (N.Y.S.); (Z.Z.M.); (I.S.S.)
| | - Dmitry A. Tsekatunov
- Regional State Budgetary Healthcare Institution of Khabarovsk Territory, Regional Clinical Oncology Center, 680042 Khabarovsk, Russia; (D.A.T.); (I.B.R.); (D.N.K.)
| | - Irina B. Ryzhavskaya
- Regional State Budgetary Healthcare Institution of Khabarovsk Territory, Regional Clinical Oncology Center, 680042 Khabarovsk, Russia; (D.A.T.); (I.B.R.); (D.N.K.)
| | - Igor N. Kuznetsov
- Ministry of Health of the Russian Federation, Russian University of Medicine, 127473 Moscow, Russia;
| | - Dmitry N. Kushlinskii
- Regional State Budgetary Healthcare Institution of Khabarovsk Territory, Regional Clinical Oncology Center, 680042 Khabarovsk, Russia; (D.A.T.); (I.B.R.); (D.N.K.)
| | - Zaman Z. Mamedli
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 115478 Moscow, Russia; (N.E.K.); (O.V.K.); (A.A.A.); (Y.B.K.); (N.Y.S.); (Z.Z.M.); (I.S.S.)
| | - Ivan S. Stilidi
- N.N. Blokhin National Medical Research Center of Oncology, Ministry of Health of the Russian Federation, 115478 Moscow, Russia; (N.E.K.); (O.V.K.); (A.A.A.); (Y.B.K.); (N.Y.S.); (Z.Z.M.); (I.S.S.)
| |
Collapse
|
23
|
Yamaguchi T, Kitahara S, Matsui A, Okamoto J, Muragaki Y, Masamune K. HIFU induces reprogramming of the tumor immune microenvironment in a pancreatic cancer mouse model. Med Mol Morphol 2025:10.1007/s00795-025-00419-1. [PMID: 39870899 DOI: 10.1007/s00795-025-00419-1] [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: 11/13/2024] [Accepted: 01/06/2025] [Indexed: 01/29/2025]
Abstract
This study evaluates the effects of different high-intensity focused ultrasound irradiation (HIFU) methods on local tumor suppression and systemic antitumor effects, including the abscopal effect, in a mouse model of pancreatic cancer. To ascertain the efficacy of the treatment, pancreatic cancer cells were injected into the thighs of mice and HIFU was applied on one side using continuous waves or trigger pulse waves. Then, tumor volume, tissue changes, and immune marker levels were analyzed. Both the irradiation methods suppressed tumor growth, with the trigger pulse wave showing stronger effects and the difference being significant. Tumor suppression was also observed on the non-irradiated side, suggesting an abscopal effect. These effects vary depending on the irradiation method used. We conclude that HIFU induces both local tumor suppression and a systemic immune response, suggesting its potential for combination with immunotherapy for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Toshihiro Yamaguchi
- Faculty of Advanced Techno-Surgery (FATS), Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku, Tokyo, 162-8666, Japan
| | - Shuji Kitahara
- Faculty of Advanced Techno-Surgery (FATS), Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku, Tokyo, 162-8666, Japan.
| | - Aya Matsui
- Department of Vascular Physiology, Kanazawa University Graduate School of Medical Sciences, 13-1 Takaramachi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Jun Okamoto
- SONIRE Therapeutics Inc., Nihonbashi Life Science, Building 2 803, 3-11-5 Nihonbashi Honcho, Chuo-Ku 103-0023, Tokyo, Japan
| | - Yoshihiro Muragaki
- Faculty of Advanced Techno-Surgery (FATS), Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku, Tokyo, 162-8666, Japan
- Center for Advanced Medical Engineering Research and Development, Kobe University, 1-5-1 Minatojima Minamimachi, Chuo-Ku, Kobe, Hyogo, 650-0047, Japan
| | - Ken Masamune
- Faculty of Advanced Techno-Surgery (FATS), Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku, Tokyo, 162-8666, Japan.
| |
Collapse
|
24
|
Li S, Pan Y, Ye R, Wang Y, Li L. Immune checkpoints in B-cell Lymphoma: Still an Unmet challenge from Basic research to clinical practice. Int Immunopharmacol 2025; 146:113717. [PMID: 39673995 DOI: 10.1016/j.intimp.2024.113717] [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: 08/11/2024] [Revised: 11/24/2024] [Accepted: 11/24/2024] [Indexed: 12/16/2024]
Abstract
In the last decade, advancements in immunotherapy knowledge have highlighted CTLA-4, PD-1, LAG-3, TIM-3, and TIGIT, decisive immune checkpoints exhibiting within the tumor microenvironment (TME), as fundamental objects for cancer immunotherapy. The widespread clinical use of immune checkpoint inhibitors (ICls), employing PD-1/PD-L1 or CTLA-4 antibodies to obstruct crucial checkpoint regulators, is noted in treating B-cell lymphoma patients. Nevertheless, the prolonged advantages of the currently employed treatments against CTLA-4, PD-1, and PD-L1 are uncommon among patients. Thus, recent focus has been progressively moved to additional immune checkpoints on T cells, like LAG-3, TIM-3, and TIGIT, which are now seen as reassuring targets for treatment and broadly acknowledged. There are several types of immunecheckpoint molecules expressed by T cells, and inhibitors targeting immune checkpoints can revive and amplify the immune response of T lymphocytes against tumors, a crucial aspect in lymphoma therapy. However, there is little knowledge about their regulation. Herein, we discuss the anti-tumor effects and functions of ICIs in controlling T-cell activity, as well as the progress in combined application with other immunotherapies.
Collapse
Affiliation(s)
- Sijia Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Yuanyuan Pan
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Ruyu Ye
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Yu Wang
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China
| | - Li Li
- Department of Hematology, The Second Hospital of Dalian Medical University, Dalian, PR China.
| |
Collapse
|
25
|
Park SJ, Yang S, Lee S, Joo SH, Park T, Kim DH, Kim H, Park S, Kim JT, Kwack WG, Kang SW, Song YK, Cha JM, Rhee SY, Chung EK. Machine-Learning Parsimonious Prediction Model for Diagnostic Screening of Severe Hematological Adverse Events in Cancer Patients Treated with PD-1/PD-L1 Inhibitors: Retrospective Observational Study by Using the Common Data Model. Diagnostics (Basel) 2025; 15:226. [PMID: 39857110 PMCID: PMC11763827 DOI: 10.3390/diagnostics15020226] [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: 10/04/2024] [Revised: 01/08/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Earlier detection of severe immune-related hematological adverse events (irHAEs) in cancer patients treated with a PD-1 or PD-L1 inhibitor is critical to improving treatment outcomes. The study aimed to develop a simple machine learning (ML) model for predicting irHAEs associated with PD-1/PD-L1 inhibitors. Methods: We utilized the Observational Medical Outcomes Partnership-Common Data Model based on electronic medical records from a tertiary (KHMC) and a secondary (KHNMC) hospital in South Korea. Severe irHAEs were defined as Grades 3-5 by the Common Terminology Criteria for Adverse Events (version 5.0). The predictive model was developed using the KHMC dataset, and then cross-validated against an independent cohort (KHNMC). The full ML models were then simplified by selecting critical features based on the feature importance values (FIVs). Results: Overall, 397 and 255 patients were included in the primary (KHMC) and cross-validation (KHNMC) cohort, respectively. Among the tested ML algorithms, random forest achieved the highest accuracy (area under the receiver operating characteristic curve [AUROC] 0.88 for both cohorts). Parsimonious models reduced to 50% FIVs of the full models showed comparable performance to the full models (AUROC 0.83-0.86, p > 0.05). The KHMC and KHNMC parsimonious models shared common predictive features including furosemide, oxygen gas, piperacillin/tazobactam, and acetylcysteine. Conclusions: Considering the simplicity and adequate predictive performance, our simplified ML models might be easily implemented in clinical practice with broad applicability. Our model might enhance early diagnostic screening of irHAEs induced by PD-1/PD-L1 inhibitors, contributing to minimizing the risk of severe irHAEs and improving the effectiveness of cancer immunotherapy.
Collapse
Affiliation(s)
- Seok Jun Park
- Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.P.); (S.Y.); (S.H.J.); (T.P.); (D.H.K.); (H.K.); (S.P.)
- Institute of Regulatory Innovation Through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seungwon Yang
- Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.P.); (S.Y.); (S.H.J.); (T.P.); (D.H.K.); (H.K.); (S.P.)
- Institute of Regulatory Innovation Through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Suhyun Lee
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Sung Hwan Joo
- Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.P.); (S.Y.); (S.H.J.); (T.P.); (D.H.K.); (H.K.); (S.P.)
- Institute of Regulatory Innovation Through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
| | - Taemin Park
- Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.P.); (S.Y.); (S.H.J.); (T.P.); (D.H.K.); (H.K.); (S.P.)
- Institute of Regulatory Innovation Through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Dong Hyun Kim
- Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.P.); (S.Y.); (S.H.J.); (T.P.); (D.H.K.); (H.K.); (S.P.)
- Institute of Regulatory Innovation Through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Hyeonji Kim
- Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.P.); (S.Y.); (S.H.J.); (T.P.); (D.H.K.); (H.K.); (S.P.)
- Institute of Regulatory Innovation Through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Soyun Park
- Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.P.); (S.Y.); (S.H.J.); (T.P.); (D.H.K.); (H.K.); (S.P.)
- Institute of Regulatory Innovation Through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Jung-Tae Kim
- Department of Pharmacy, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea;
| | - Won Gun Kwack
- Division of Pulmonary, Allergy and Critical Care Medicine, Kyung Hee University Hospital, Seoul 02447, Republic of Korea;
| | - Sung Wook Kang
- Department of Pulmonary and Critical Care Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea;
| | - Yun-Kyoung Song
- College of Pharmacy, The Catholic University of Korea-Sungsim Campus, Bucheon 14662, Gyeonggi-do, Republic of Korea;
| | - Jae Myung Cha
- Division of Gastroenterology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University School of Medicine, Seoul 05278, Republic of Korea
| | - Sang Youl Rhee
- Center for Digital Health, Medical Science Research Institute, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Endocrinology and Metabolism, Kyung Hee University School of Medicine, Seoul 02447, Republic of Korea
| | - Eun Kyoung Chung
- Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.P.); (S.Y.); (S.H.J.); (T.P.); (D.H.K.); (H.K.); (S.P.)
- Institute of Regulatory Innovation Through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
- Department of Pharmacy, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea;
| |
Collapse
|
26
|
Lawal B, Wang Y, Lotfinejad P, Sharma R, Yang C, Annasamudram A, Wang XS. NFATC2 target gene signature correlates with immune checkpoint blockade resistance in melanoma. Am J Cancer Res 2025; 15:311-321. [PMID: 39949941 PMCID: PMC11815363 DOI: 10.62347/eyml2689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/10/2024] [Indexed: 02/16/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs), such as anti-PD-1 and anti-CTLA-4, have significantly advanced melanoma treatment by reactivating the immune system to target cancer cells. However, a substantial portion of patients do not respond or develop resistance, highlighting the need for more effective predictive biomarkers. Dysregulation of transcriptional programs has been implicated in cancer progression and immune evasion, with transcription factors (TFs) playing a crucial role. In this study, we investigated transcriptional gene signatures (TGSs) for their potential to predict ICI resistance in melanoma by analyzing two independent clinical trial datasets. Among the identified TFs, NFATC2 (Nuclear Factor of Activated T Cells 2) was observed to be a promising marker for resistance to anti-PD-1 therapy. NFATC2, a regulator of T cell activation, may be co-opted by melanoma cells to evade immune surveillance. Our analysis indicated that elevated NFATC2 TGS scores were associated with ICI resistance and poorer survival outcomes across multiple melanoma cohorts. Validation in independent datasets further suggested NFATC2's potential predictive value, particularly in patients without liver metastasis or with prior anti-CTLA-4 therapy. Elevated NFATC2 TGS scores also correlated with reduced immune cell infiltration, specifically of CD8+ T cells, increased markers of T cell exhaustion, and higher tumor purity. These findings support NFATC2 TGS as a candidate biomarker for stratifying melanoma patients and potentially informing ICI therapy response. Further research into NFATC2-associated immune evasion mechanisms may offer insights for overcoming resistance to immunotherapy.
Collapse
Affiliation(s)
- Bashir Lawal
- UPMC Hillman Cancer Center, University of PittsburghPittsburgh, PA 15213, USA
- Department of Pathology, University of PittsburghPittsburgh, PA 15261, USA
| | - Yue Wang
- UPMC Hillman Cancer Center, University of PittsburghPittsburgh, PA 15213, USA
- Department of Pathology, University of PittsburghPittsburgh, PA 15261, USA
| | - Parisa Lotfinejad
- UPMC Hillman Cancer Center, University of PittsburghPittsburgh, PA 15213, USA
- Department of Pathology, University of PittsburghPittsburgh, PA 15261, USA
| | - Renu Sharma
- UPMC Hillman Cancer Center, University of PittsburghPittsburgh, PA 15213, USA
- Department of Pathology, University of PittsburghPittsburgh, PA 15261, USA
| | - Chuang Yang
- UPMC Hillman Cancer Center, University of PittsburghPittsburgh, PA 15213, USA
- Department of Pathology, University of PittsburghPittsburgh, PA 15261, USA
| | - Anusha Annasamudram
- UPMC Hillman Cancer Center, University of PittsburghPittsburgh, PA 15213, USA
- Department of Pathology, University of PittsburghPittsburgh, PA 15261, USA
| | - Xiao-Song Wang
- UPMC Hillman Cancer Center, University of PittsburghPittsburgh, PA 15213, USA
- Department of Pathology, University of PittsburghPittsburgh, PA 15261, USA
| |
Collapse
|
27
|
Yuzhakova DV, Sachkova DA, Izosimova AV, Yashin KS, Yusubalieva GM, Baklaushev VP, Mozherov AM, Shcheslavskiy VI, Shirmanova MV. Fluorescence Lifetime Imaging of NAD(P)H in Patients' Lymphocytes: Evaluation of Efficacy of Immunotherapy. Cells 2025; 14:97. [PMID: 39851525 PMCID: PMC11764258 DOI: 10.3390/cells14020097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/30/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND The wide variability in clinical responses to anti-tumor immunotherapy drives the search for personalized strategies. One of the promising approaches is drug screening using patient-derived models composed of tumor and immune cells. In this regard, the selection of an appropriate in vitro model and the choice of cellular response assay are critical for reliable predictions. Fluorescence lifetime imaging microscopy (FLIM) is a powerful, non-destructive tool that enables direct monitoring of cellular metabolism on a label-free basis with a potential to resolve metabolic rearrangements in immune cells associated with their reactivity. OBJECTIVE The aim of the study was to develop a patient-derived glioma explant model enriched by autologous peripheral lymphocytes and explore FLIM of the redox-cofactor NAD(P)H in living lymphocytes to measure the responses of the model to immune checkpoint inhibitors. METHODS The light microscopy, FLIM of NAD(P)H and flow cytometry were used. RESULTS The results demonstrate that the responsive models displayed a significant increase in the free NAD(P)H fraction α1 after treatment, associated with a shift towards glycolysis due to lymphocyte activation. The non-responsive models exhibited no alterations or a decrease in the NAD(P)H α1 after treatment. The FLIM data correlated well with the standard assays of immunotherapy drug response in vitro, including morphological changes, the T-cells activation marker CD69, and the tumor cell proliferation index Ki67. CONCLUSIONS The proposed platform that includes tumor explants co-cultured with lymphocytes and the NAD(P)H FLIM assay represents a promising solution for the patient-specific immunotherapeutic drug screening.
Collapse
Affiliation(s)
- Diana V. Yuzhakova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
| | - Daria A. Sachkova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., 603950 Nizhny Novgorod, Russia
| | - Anna V. Izosimova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
| | - Konstantin S. Yashin
- Department of Neurosurgery, Privolzsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia;
| | - Gaukhar M. Yusubalieva
- Federal Research and Clinical Center, Federal Medical and Biological Agency, 28 Orekhovy Blvd., 115682 Moscow, Russia; (G.M.Y.); (V.P.B.)
- Laboratory of Molecular Mechanisms of Regeneration and Aging, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova St., 119991 Moscow, Russia
| | - Vladimir P. Baklaushev
- Federal Research and Clinical Center, Federal Medical and Biological Agency, 28 Orekhovy Blvd., 115682 Moscow, Russia; (G.M.Y.); (V.P.B.)
- Laboratory of Molecular Mechanisms of Regeneration and Aging, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 32 Vavilova St., 119991 Moscow, Russia
| | - Artem M. Mozherov
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
| | - Vladislav I. Shcheslavskiy
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
- R&D Department, Becker&Hickl GmbH, 7-9 Nunsdorfer Ring, 12277 Berlin, Germany
| | - Marina V. Shirmanova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603005 Nizhny Novgorod, Russia; (D.A.S.); (A.V.I.); (A.M.M.); (M.V.S.)
| |
Collapse
|
28
|
Kulbay M, Tuli N, Mazza M, Jaffer A, Juntipwong S, Marcotte E, Tanya SM, Nguyen AXL, Burnier MN, Demirci H. Oncolytic Viruses and Immunotherapy for the Treatment of Uveal Melanoma and Retinoblastoma: The Current Landscape and Novel Advances. Biomedicines 2025; 13:108. [PMID: 39857692 PMCID: PMC11762644 DOI: 10.3390/biomedicines13010108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 12/25/2024] [Accepted: 12/31/2024] [Indexed: 01/27/2025] Open
Abstract
Intraocular malignant tumors are rare; however, they can cause serious life-threatening complications. Uveal melanoma (UM) and retinoblastoma (RB) are the most common intraocular tumors in adults and children, respectively, and come with a great disease burden. For many years, several different treatment modalities for UM and RB have been proposed, with chemotherapy for RB cases and plaque radiation therapy for localized UM as first-line treatment options. Extraocular extension, recurrence, and metastasis constitute the major challenges of conventional treatments. To overcome these obstacles, immunotherapy, which encompasses different treatment options such as oncolytic viruses, antibody-mediated immune modulations, and targeted immunotherapy, has shown great potential as a novel therapeutic tool for cancer therapy. These anti-cancer treatment options provide numerous advantages such as selective cancer cell death and the promotion of an anti-tumor immune response, and they prove useful in preventing vision impairment due to macular and/or optic disc involvement. Numerous factors such as the vector choice, route of administration, dosing, and patient characteristics must be considered when engineering an oncolytic virus or other forms of immunotherapy vectors. This manuscript provides an in-depth review of the molecular design of oncolytic viruses (e.g., virus capsid proteins and encapsulation technologies, vectors for delivery, cell targeting) and immunotherapy. The most recent advances in preclinical- and clinical-phase studies are further summarized. The recent developments in virus-like drug conjugates (i.e., AU011), oncolytic viruses for metastatic UM, and targeted immunotherapies have shown great results in clinical trials for the future clinical application of these novel technologies in the treatment algorithm of certain intraocular tumors.
Collapse
Affiliation(s)
- Merve Kulbay
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3J1, Canada; (M.K.)
| | - Nicolas Tuli
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H4A 3J1, Canada
| | - Massimo Mazza
- Faculty of Medicine and Health Sciences, McGill University, Montreal, QC H4A 3J1, Canada
| | - Armaan Jaffer
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 2V5, Canada
- Research Excellence Cluster in Vision, University of British Columbia, Vancouver, BC V5Z 3N9, Canada
| | - Sarinee Juntipwong
- Kellogg Eye Center, Department of Ophthalmology and Visual Science, University of Michigan, Ann Arbor, MI 48105, USA
| | - Emily Marcotte
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada;
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Stuti Misty Tanya
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3J1, Canada; (M.K.)
| | - Anne Xuan-Lan Nguyen
- Department of Ophthalmology & Vision Sciences, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Miguel N. Burnier
- Department of Ophthalmology & Visual Sciences, McGill University, Montreal, QC H4A 3J1, Canada; (M.K.)
- McGill University Ocular Pathology and Translational Research Laboratory, McGill University, Montreal, QC H4A 3J1, Canada;
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC H4A 3J1, Canada
| | - Hakan Demirci
- Kellogg Eye Center, Department of Ophthalmology and Visual Science, University of Michigan, Ann Arbor, MI 48105, USA
| |
Collapse
|
29
|
Singer M, Hamdy R, Elsayed TM, Husseiny MI. The Mechanisms and Therapeutic Implications of Metabolic Communication in the Tumor-Immune Microenvironment. METABOLIC DYNAMICS IN HOST-MICROBE INTERACTION 2025:291-315. [DOI: 10.1007/978-981-96-1305-2_12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
|
30
|
Luo B, Liao M, Nie B, Yu Y, Yao Q. Genomic profiles and their associations with microsatellite instability status, tumor mutational burden, and programmed death ligand 1 expression in Chinese patients with colorectal cancer. J Gastrointest Oncol 2024; 15:2460-2472. [PMID: 39816034 PMCID: PMC11732342 DOI: 10.21037/jgo-24-748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/10/2024] [Indexed: 01/18/2025] Open
Abstract
Background Colorectal cancer (CRC) is among the most prevalent malignancies globally, with a rising incidence observed in younger demographics. Despite surgical resection remaining the cornerstone of treatment, metastatic CRC poses significant therapeutic challenges. Immunotherapy, a mode of treatment that leverages the patient's immune system, presents a promising frontier in CRC management, particularly for late-stage cases with limited treatment options. The study was aimed to elucidate the relationships between genetic profiles and predictive biomarkers in CRC patients to inform immunotherapy decisions and improve outcomes. Methods We conducted a large-scale study involving 660 patients with CRC, analyzing genetic profiles and predictive biomarkers for immune checkpoint inhibitors (ICIs) using next-generation sequencing (NGS) and immunohistochemistry (IHC). The study focused on assessing the association between gene mutations and markers such as microsatellite instability (MSI) status, tumor mutational burden (TMB), and programmed death ligand 1 (PD-L1) expression. Results Analysis revealed a diverse mutational landscape in CRC, with TP53 (73.64%), APC (67.58%), and KRAS (46.82%) being the most frequently mutated genes. We observed significant associations between KRAS mutations and co-occurrences with FBXW7, PIK3CA, and SMAD4 mutations, while KRAS mutations were mutually exclusive with TP53 mutations. KRAS mutations were enriched in the PD-L1 tumor proportion score (TPS) ≥1% population (P=0.03), whereas APC mutations were enriched in the PD-L1 TPS <1% population (P=0.10) as compared to their wild types. Additionally, specific mutations such as KRAS p.A146T, PIK3CA p.H1047R, and BRAF p.V600E were significantly associated with higher TMB and MSI-high status, indicating potential benefits from ICI therapy. Conclusions Our findings underscore the importance of genetic profiling in guiding treatment decisions for patients with CRC, particularly in the era of immunotherapy. Understanding the complex interplay between genetic alterations and immune markers is critical for optimizing therapeutic strategies and improving clinical outcomes. Further research is warranted to validate these findings and explore personalized treatment approaches in CRC.
Collapse
Affiliation(s)
- Bo Luo
- Department of General Surgery, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Min Liao
- Department of Traditional Chinese Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Bin Nie
- Department of General Surgery, Chengdu BOE Hospital, Chengdu, China
| | - Yunbao Yu
- Department of General Surgery, Chengdu BOE Hospital, Chengdu, China
| | - Qipeng Yao
- Department of Traditional Chinese Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
31
|
Ma L, Kim MO. Advances in Preventive and Therapeutic Strategies for Oral Cancer: A Short Review. J Cancer Prev 2024; 29:113-119. [PMID: 39790224 PMCID: PMC11706729 DOI: 10.15430/jcp.24.027] [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: 12/02/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
Oral cancer is a major global health concern, with high incidence and mortality rates, especially in high-risk populations. Early diagnosis remains a challenge, and current treatments, such as surgery, radiation, and chemotherapy, have limited effectiveness, particularly in advanced stages. Recent advances in targeted therapies and immunotherapy offer promising alternatives, providing more precise and personalized treatment options. Targeted therapies, such as epidermal growth factor receptor inhibitors, aim to disrupt specific molecular pathways in tumor growth, while immunotherapies, including immune checkpoint inhibitors and chimeric antigen receptor-T cell therapy, enhance the body's immune response to fight cancer. Combination therapies, integrating both targeted and immune strategies, are being explored to overcome the limitations of single-agent treatments. This review highlights the current strategies in the prevention and treatment of oral cancer, discusses emerging therapies, explores future research directions, focusing on optimizing existing treatments, identifying new biomarkers, and developing innovative therapeutic approaches. The potential of personalized medicine and combination therapies offers new hope for improving survival rates and quality of life for oral cancer patients.
Collapse
Affiliation(s)
- Lei Ma
- Department of Animal Science and Biotechnology, Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Korea
| | - Myoung Ok Kim
- Department of Animal Science and Biotechnology, Research Institute for Innovative Animal Science, Kyungpook National University, Sangju, Korea
| |
Collapse
|
32
|
Arafat Hossain M. A comprehensive review of immune checkpoint inhibitors for cancer treatment. Int Immunopharmacol 2024; 143:113365. [PMID: 39447408 DOI: 10.1016/j.intimp.2024.113365] [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: 08/25/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Immunology-based therapies are emerging as an effective cancer treatment, using the body's immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
33
|
Kravtsova-Ivantsiv Y, Goldhirsh G, Ciechanover A. CXCL12 restricts tumor growth by suppressing the Ras, ERK1/2, c-Myc, and the immune checkpoint PD-L1 pathways. Proc Natl Acad Sci U S A 2024; 121:e2416909121. [PMID: 39689179 DOI: 10.1073/pnas.2416909121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/15/2024] [Indexed: 12/19/2024] Open
Abstract
Cytokines constitute a family of proteins that modulate the immune system and are secreted by many cells. CXCL12, along with its receptor CXCR4, are essential players in numerous processes. Dysregulation of their function underlie the mechanism(s) of several pathologies, including malignancies. Here, we demonstrate an unexpected effect of the cytokine and its receptor: In both cells and animal models, CXCL12 restricts tumorigenicity of the human glioblastoma cells U87-MG and U-118, and of a cell line derived from PyMT mouse breast cancer. Overexpression of CXCL12 inhibits activation of the oncogene Ras which results in downregulation of its proliferative signals, such as reduced phosphorylation of the extracellular signal-regulated kinase 1/2 (ERK1/2), inhibition of c-Myc expression, and subsequent inhibition of cell cycle. Furthermore, CXCL12 induces downregulation of the growth differentiation factor 15 (GDF15), insulin-like growth factor-binding protein 6 (IGFBP6), and matrix metalloproteinase-3 (MMP3), which are implicated in sending metastases. Indeed, monitoring cell migration in vitro and generation of metastases in mice demonstrate that CXCL12 slows the migration of U87-MG and PyMT cells. Remarkably, overexpression of CXCL12 also downregulates the cell surface immune checkpoint protein programmed cell death-ligand 1 (PD-L1), resulting in recruitment of cytotoxic CD8 T cells into xenografts accompanied by their shrinkage. Overall, CXCL12 inhibits tumor growth through several distinct mechanisms: inhibition of cell cycle and migration, as well as impairment of immune checkpoint, thereby stimulating a strong host's immune response. The mechanism(s) that renders CXCL12 a tumor-promoting factor in certain cells and a suppressor in others has remained elusive.
Collapse
Affiliation(s)
- Yelena Kravtsova-Ivantsiv
- The Rappaport Faculty of Medicine and Research Institute, and the Rappaport Technion Integrated Cancer Center (R-TICC), Technion-Israel Institute of Technology, Haifa 3109601, Israel
| | - Gilad Goldhirsh
- The Rappaport Faculty of Medicine and Research Institute, and the Rappaport Technion Integrated Cancer Center (R-TICC), Technion-Israel Institute of Technology, Haifa 3109601, Israel
| | - Aaron Ciechanover
- The Rappaport Faculty of Medicine and Research Institute, and the Rappaport Technion Integrated Cancer Center (R-TICC), Technion-Israel Institute of Technology, Haifa 3109601, Israel
| |
Collapse
|
34
|
Choubey RB, Sweta, Vibha, Sharma A, Rai AK. Immunotherapy to CD5, a T-cell antigen having roles from development to peripheral function: Future prospective and challenges. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 144:431-460. [PMID: 39978974 DOI: 10.1016/bs.apcsb.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
CD5 is a pan T-cell marker expressed by all T-cells and a subset of B-cells, i.e., B1a cells. The significance of CD5 is evident from its functions, starting from T-cell development, antigen priming, activation, and effector response to the maintenance of tolerance. Varying CD5 expression and signaling in response to TCR-pMHC complex avidity is associated with thymic selection, competency, and effector response. Altered CD5 expression is associated with immunological and diseased conditions such as CD5-/low infiltrating T-cells in solid tumors, CD5hi T-cells in anergy conditions, CD5-/low phenotype of leukemic T-cells, high CD5 expression by regulatory T-cells, CD5lowphenotype of autoreactive T-cells, etc. A low CD5 expression triggers activation-induced cell death upon antigenic stimulation. There are three forms of CD5: membrane CD5 (mCD5), intracellular CD5 (cCD5) and soluble CD5 (sCD5). mCD5 and cCD5 are generated from conventional and non-conventional mRNA variants, i.e., E1A and E1B, respectively. E1B variant encoding cCD5 is derived from a human endogenous retrovirus segment inserted 8.2 kb upstream to conventional E1A exon. Various conditions, such as leukemia, exposure to hydrocarbon, hypoxia, etc., can trigger E1B transcription and, thus, cCD5 expression. Blocking mCD5 with mAb can restore immune response, effectively targeting cancer. Understanding cCD5, linked to leukemogenesis, can offer new avenues of immunotherapy.
Collapse
Affiliation(s)
- Ranjeet Bahadur Choubey
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Allahabad, UP, India
| | - Sweta
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Allahabad, UP, India
| | - Vibha
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Allahabad, UP, India
| | - Avika Sharma
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Allahabad, UP, India
| | - Ambak Kumar Rai
- Department of Biotechnology, Motilal Nehru National Institute of Technology Allahabad, Allahabad, UP, India.
| |
Collapse
|
35
|
Hong S, Park J, Oh Y, Cho H, Kim K. Nanotechnology-Based Strategies for Safe and Effective Immunotherapy. Molecules 2024; 29:5855. [PMID: 39769944 PMCID: PMC11676242 DOI: 10.3390/molecules29245855] [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: 10/30/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
Cancer immunotherapy using immune checkpoint blockades has emerged as a promising therapeutic approach. However, immunotherapy faces challenges such as low response rates in solid tumors, necessitating strategies to remodel the immune-suppressive tumor microenvironment (TME) into an immune-activated state. One of the primary approaches to achieve this transformation is through the induction of immunogenic cell death (ICD). Herein, we discussed strategies to maximize ICD induction using nanoparticles. In particular, this review highlighted various studies integrating chemotherapy, radiation therapy (RT), photodynamic therapy (PDT), and photothermal therapy (PTT) with nanoparticle-based immunotherapy. The research covered in this review aims to provide valuable insights for future studies on nanoparticle-assisted immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Kwangmeyung Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Republic of Korea; (S.H.); (J.P.); (Y.O.); (H.C.)
| |
Collapse
|
36
|
Kwon WA, Lee MK. Evolving Treatment Landscape of Frontline Therapy for Metastatic Urothelial Carcinoma: Current Insights and Future Perspectives. Cancers (Basel) 2024; 16:4078. [PMID: 39682263 DOI: 10.3390/cancers16234078] [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: 11/06/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
Cisplatin-based chemotherapy has long been the standard first-line (1L) treatment for metastatic urothelial carcinoma (mUC). However, up to 50% of patients with mUC may be ineligible for cisplatin owing to comorbidities, necessitating alternative primary treatment options. Immune checkpoint inhibitors (ICIs) have emerged as a vital alternative for those unable to receive cisplatin. Nevertheless, the prognosis of advanced UC remains dire and challenges persist in optimizing 1L therapy. Recent medical advancements have redirected attention towards innovative drug combinations for the primary treatment of mUC. The combination of enfortumab vedotin (EV) and pembrolizumab has shown significantly improved overall and progression-free survival rates compared to those with chemotherapy alone. This combination can be used as a 1L treatment for patients with mUC who are cisplatin-ineligible or require alternatives to standard chemotherapy. While platinum-based chemotherapy continues to be essential for many patients, the approval of EV and pembrolizumab as 1L treatments for cisplatin-ineligible patients signifies a major breakthrough in primary cancer care. These therapies offer enhanced outcomes in terms of survival and response rates and highlight the increasing relevance of ICI-containing regimens in frontline cancer care. This review provides an exhaustive overview of the current frontline treatment landscape of mUC and explores new therapeutic strategies, with the aim of facilitating clinical decision-making and guiding therapeutic strategies in patients with mUC.
Collapse
Affiliation(s)
- Whi-An Kwon
- Department of Urology, Myongji Hospital, Hanyang University College of Medicine, Goyang-si 10475, Republic of Korea
| | - Min-Kyung Lee
- Department of Internal Medicine, Myongji Hospital, Hanyang University College of Medicine, Goyang-si 10475, Republic of Korea
| |
Collapse
|
37
|
Ahuja S, Zaheer S. The evolution of cancer immunotherapy: a comprehensive review of its history and current perspectives. KOREAN JOURNAL OF CLINICAL ONCOLOGY 2024; 20:51-73. [PMID: 39778508 PMCID: PMC11717579 DOI: 10.14216/kjco.24009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/24/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025]
Abstract
Cancer immunotherapy uses the body's immune system to combat cancer, marking a significant advancement in treatment. This review traces its evolution from the late 19th century to its current status. It began with William Coley's pioneering work using bacterial toxins to stimulate the immune system against cancer cells, establishing the foundational concept of immunotherapy. In the mid-20th century, cytokine therapies like interferons and interleukins emerged, demonstrating that altering the immune response could reduce tumors and highlighting the complex interplay between cancer and the immune system. The discovery of immune checkpoints, regulatory pathways that prevent autoimmunity but are exploited by cancer cells to evade detection, was a pivotal development. Another major breakthrough is CAR-T cell therapy, which involves modifying a patient's T cells to target cancer-specific antigens. This personalized treatment has shown remarkable success in certain blood cancers. Additionally, cancer vaccines aim to trigger immune responses against tumor-specific or associated antigens, and while challenging, ongoing research is improving their efficacy. The historical progression of cancer immunotherapy, from Coley's toxins to modern innovations like checkpoint inhibitors and CAR-T cell therapy, underscores its transformative impact on cancer treatment. As research delves deeper into the immune system's complexities, immunotherapy is poised to become even more crucial in oncology, offering renewed hope to patients globally.
Collapse
Affiliation(s)
- Sana Ahuja
- Department of Pathology, Safdarjung Hospital, Vardhman Mahavir Medical College, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Safdarjung Hospital, Vardhman Mahavir Medical College, New Delhi, India
| |
Collapse
|
38
|
Hamid O, Hamidi N. Enhancing immuno-oncology efficacy with H1-antihistamine in cancer therapy: a review of current research and findings. Curr Med Res Opin 2024; 40:2139-2146. [PMID: 39503414 DOI: 10.1080/03007995.2024.2427323] [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: 08/01/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/11/2024]
Abstract
Cancer remains a major global cause of death, posing significant treatment challenges. The interactions between tumor cells and the tumor microenvironment (TME) are crucial in influencing tumor initiation, progression, metastasis, and treatment response. There has been significant research and clinical interest in targeting the TME as a therapeutic approach in cancer, with advancements being made through drug development. Histamine binds to HRH1 receptors on the TME, which inhibit CD8+ T cell activity, promote tumor growth, and contribute to resistance against immunotherapy. By inhibiting CD8+ T cells, the effectiveness of immunotherapies targeting these cells is reduced. By blocking the HRH1 pathway, H1-antihistamines can mitigate this suppression and enhance the response to immunotherapies that target CD8+ T cells. Therefore, understanding the role of histamine and its potential impact on T cells and the role of H1-antihistamines in improving immune-oncology (I/O) agents' efficacy ultimately could lead to more effective cancer therapies. The objective of this review is to examine the current literature to investigate the potential role of H1-antihistamines on the effectiveness of I/O drugs and their role in enhancing treatment against cancer. We conducted a comprehensive literature search, which included multiple databases including PubMed, Google Scholar, and EMBASE, as well as a search of oncology congresses. Our literature review initially identified thirty studies. Twenty-three of these were excluded for failing to meet inclusion criteria, which varied from study design to the type of antihistamines and patient populations involved. The clinical studies investigated the effect of different generations of H1-antihistamines in combination with I/O treatments on patients' outcomes. The findings from these studies indicated that patients using H1-antihistamines concomitantly with I/O agents experienced longer median overall survival (mOS), progression-free survival (mPFS), or improved survival compared to those who did not use antihistamines. Additionally, these trials differentiated between cationic and non-cationic H1-antihistamines, revealing that users of cationic antihistamines had overall better outcomes in terms of longer mOS and mPFS. The assessed trials were consistent in their comparisons of quantitative and qualitative, efficacy, and safety outcomes.
Collapse
Affiliation(s)
- Oday Hamid
- Department of Oncology, AstraZeneca/University of Michigan College of Pharmacy, Gaithersburg, MD, USA
| | - Negar Hamidi
- Department of Oncology, AstraZeneca/University of Maryland School of Pharmacy, Baltimore, MD, USA
| |
Collapse
|
39
|
Yu H, Lin J, Chen J, Chen L, Zou J, Liu B, Hu D, Xiao Y, Yu L, Sun Y. A surprising complete response to cadonilimab in a primary metastatic cervical cancer: a case report. Front Immunol 2024; 15:1494138. [PMID: 39660134 PMCID: PMC11628523 DOI: 10.3389/fimmu.2024.1494138] [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: 09/10/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
The outcome of patients with recurrent/metastatic cervical cancer (R/M CC) is poor, with a 5-year survival rate of only 10%-20%. Recent advances in immunotherapy renewed its interest in R/M CC treatment. It has been suggested that cadonilimab, a novel bispecific antibody targeting programmed death 1 (PD-1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4), significantly improved the survival outcomes of the R/M CC. In the present study, we reported a programmed death ligand 1 (PD-L1) and human epidermal growth factor receptor 2 (HER-2) positive CC case at stage IV who was treated with cadonilimab and achieved a surprising radiographic complete response (CR) for 10 months, even in the PD-L1 negative metastatic site. Demographic, clinical, histopathological, laboratory, treatment regime and imaging data were recorded. Unfortunately, the patient progressed rapidly during maintenance therapy when cadonilimab was replaced by sintilimab, the monoclonal antibody against PD-1, indicating the more powerful anti-tumor activity of dual blockade immunotherapy. To conclude, cadonilimab offers a promising and effective therapeutic approach for R/M CC. Notably, HER-2 is also expected to be a new reference target for cadonilimab therapy.
Collapse
Affiliation(s)
- Haijuan Yu
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Jie Lin
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Jian Chen
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Lijun Chen
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Jianping Zou
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Bin Liu
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Dan Hu
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Youping Xiao
- Department of Radiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Linhao Yu
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Yang Sun
- Department of Gynecology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| |
Collapse
|
40
|
Ko Y, Park SY, Park JW, Kim JH, Kang HG, Lee JA. CD47 in Osteosarcoma: Correlation with Metastasis and Macrophage-Mediated Phagocytosis. Cells 2024; 13:1862. [PMID: 39594611 PMCID: PMC11592588 DOI: 10.3390/cells13221862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 10/31/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
CD47 is expressed on cell surfaces and acts as a "don't eat me" signal by interacting with signal-regulatory protein-α on the macrophage surface. Some cancer cells express CD47 protein and can evade macrophage phagocytosis. Herein, we evaluated the feasibility of targeting CD47 for osteosarcoma by analyzing its expression patterns, clinicopathological correlations, and immunotherapeutic potential. We performed a retrospective analysis on 24 biopsy samples from patients with osteosarcoma to investigate correlations between CD47 protein positivity and clinicopathological characteristics. CD47 protein expression was detected in 20.8% of the biopsy samples. CD47 positivity correlated with metastasis at diagnosis. Patients with CD47-positive tumors were older than those with CD47-negative tumors. However, CD47 protein expression was not associated with sex, tumor size, or histologic response to preoperative chemotherapy. In vitro, CD47 antibody (B6H12) did not affect osteosarcoma cell viability or apoptosis. In a wound-healing assay, CD47 inhibited the migration of osteosarcoma cells. Differentiated macrophages exhibited higher phagocytic activity against osteosarcoma cells when pretreated with B6H12 compared with the isotype control. Our preliminary data suggest a possible interaction between CD47 protein and macrophage phagocytosis in osteosarcoma metastasis. A better understanding of the role of CD47 is necessary to develop an innovative immunotherapeutic approach against osteosarcoma.
Collapse
Affiliation(s)
- Yunmi Ko
- Department of Pediatrics, National Cancer Center, Goyang 10408, Republic of Korea;
| | - Seog-Yun Park
- Department of Pathology, National Cancer Center, Goyang 10408, Republic of Korea;
| | - Jong Woong Park
- Orthopedic Oncology Clinic, Center for Rare Cancer, National Cancer Center, Goyang 10408, Republic of Korea; (J.W.P.); (J.H.K.); (H.G.K.)
| | - June Hyuk Kim
- Orthopedic Oncology Clinic, Center for Rare Cancer, National Cancer Center, Goyang 10408, Republic of Korea; (J.W.P.); (J.H.K.); (H.G.K.)
| | - Hyun Guy Kang
- Orthopedic Oncology Clinic, Center for Rare Cancer, National Cancer Center, Goyang 10408, Republic of Korea; (J.W.P.); (J.H.K.); (H.G.K.)
| | - Jun Ah Lee
- Department of Pediatrics, National Cancer Center, Goyang 10408, Republic of Korea;
| |
Collapse
|
41
|
Ghosh MK, Kumar S, Begam S, Ghosh S, Basu M. GBM immunotherapy: Exploring molecular and clinical frontiers. Life Sci 2024; 356:123018. [PMID: 39214286 DOI: 10.1016/j.lfs.2024.123018] [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: 08/21/2024] [Accepted: 08/25/2024] [Indexed: 09/04/2024]
Abstract
GBM is the most common, aggressive, and intracranial primary brain tumor; it originates from the glial progenitor cells, has poor overall survival (OS), and has limited treatment options. In this decade, GBM immunotherapy is in trend and preferred over several conventional therapies, due to their better patient survival outcome. This review explores the clinical trials of several immunotherapeutic approaches (immune checkpoint blockers (ICBs), CAR T-cell therapy, cancer vaccines, and adoptive cell therapy) with their efficacy and safety. Despite significant progress, several challenges (viz., immunosuppressive microenvironment, heterogeneity, and blood-brain barrier (BBB)) were experienced that hamper their immunotherapeutic potential. Furthermore, these challenges were clinically studied to be resolved by multiple combinatorial approaches, discussed in the later part of the review. Thus, this review suggests the clinical use and potential of immunotherapy in GBM and provides the holistic recent knowledge and future perspectives.
Collapse
Affiliation(s)
- Mrinal K Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata 700091, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India.
| | - Sunny Kumar
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata 700091, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Sabana Begam
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata 700091, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Sayani Ghosh
- Cancer Biology and Inflammatory Disorder Division, Council of Scientific and Industrial Research-Indian Institute of Chemical Biology (CSIR-IICB), TRUE Campus, CN-6, Sector-V, Salt Lake, Kolkata 700091, India; Academy of Scientific and Innovative Research, Ghaziabad, Uttar Pradesh 201002, India
| | - Malini Basu
- Department of Microbiology, Dhruba Chand Halder College, Dakshin Barasat, South 24 Parganas, PIN-743372, India
| |
Collapse
|
42
|
Garg P, Pareek S, Kulkarni P, Horne D, Salgia R, Singhal SS. Next-Generation Immunotherapy: Advancing Clinical Applications in Cancer Treatment. J Clin Med 2024; 13:6537. [PMID: 39518676 PMCID: PMC11546714 DOI: 10.3390/jcm13216537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024] Open
Abstract
Next-generation immunotherapies have revolutionized cancer treatment, offering hope for patients with hard-to-treat tumors. This review focuses on the clinical applications and advancements of key immune-based therapies, including immune checkpoint inhibitors, CAR-T cell therapy, and new cancer vaccines designed to harness the immune system to combat malignancies. A prime example is the success of pembrolizumab in the treatment of advanced melanoma, underscoring the transformative impact of these therapies. Combination treatments, integrating immunotherapy with chemotherapy, radiation, and targeted therapies, are demonstrating synergistic benefits and improving patient outcomes. This review also explores the evolving role of personalized immunotherapy, guided by biomarkers, genomic data, and the tumor environment, to better target individual tumors. Although significant progress has been made, challenges such as resistance, side effects, and high treatment costs persist. Technological innovations, including nanotechnology and artificial intelligence, are explored as future enablers of these therapies. The review evaluates key clinical trials, breakthroughs, and the emerging immune-modulating agents and advanced delivery systems that hold great promise for enhancing treatment efficacy, reducing toxicity, and expanding access to immunotherapy. In conclusion, this review highlights the ongoing advancements in immunotherapy that are reshaping cancer care, with future strategies poised to overcome current challenges and further extend therapeutic reach.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Siddhika Pareek
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Prakash Kulkarni
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Departments of Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| | - Sharad S. Singhal
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center and National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
43
|
Lica JJ, Pradhan B, Safi K, Jakóbkiewicz-Banecka J, Hellmann A. Promising Therapeutic Strategies for Hematologic Malignancies: Innovations and Potential. Molecules 2024; 29:4280. [PMID: 39275127 PMCID: PMC11397263 DOI: 10.3390/molecules29174280] [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: 08/01/2024] [Revised: 09/04/2024] [Accepted: 09/06/2024] [Indexed: 09/16/2024] Open
Abstract
In this review we explore innovative approaches in the treatment of hematologic cancers by combining various therapeutic modalities. We discuss the synergistic potential of combining inhibitors targeting different cellular pathways with immunotherapies, molecular therapies, and hormonal therapies. Examples include combining PI3K inhibitors with proteasome inhibitors, NF-κB inhibitors with immunotherapy checkpoint inhibitors, and neddylation inhibitors with therapies targeting the tumor microenvironment. Additionally, we discuss the potential use of small molecules and peptide inhibitors in hematologic cancer treatment. These multidimensional therapeutic combinations present promising strategies for enhancing treatment efficacy and overcoming resistance mechanisms. However, further clinical research is required to validate their effectiveness and safety profiles in hematologic cancer patients.
Collapse
Affiliation(s)
- Jan Jakub Lica
- Faculty of Health Science, Powiśle University, 80-214 Gdańsk, Poland
| | - Bhaskar Pradhan
- Department of Biochemistry, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Kawthar Safi
- Department of Biochemistry and Clinical Chemistry, Faculty of Biology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | | | - Andrzej Hellmann
- Department of Hematology and Transplantology, Faculty of Medicine, Medical University of Gdańsk, 80-214 Gdańsk, Poland
| |
Collapse
|
44
|
Peristeri DV, Tarazi M, Casey P, Powell A, Sultan J. Effect of the immune microenvironment on prognosis in oesophageal adenocarcinoma. Best Pract Res Clin Gastroenterol 2024; 72:101954. [PMID: 39645286 DOI: 10.1016/j.bpg.2024.101954] [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: 05/15/2024] [Revised: 09/20/2024] [Accepted: 10/22/2024] [Indexed: 12/09/2024]
Abstract
Oesophageal adenocarcinoma (OAC) is amongst the most lethal cancers worldwide, with poor treatment response leading to low survival rates. Recent improvements have been achieved by including the tumour microenvironment (TME) and patients' immune profiles in treatment decisions. We already know that patients with immune-enriched/inflamed TME have better survival outcomes. However, OAC TME is largely immunosuppressed and appears to be treatment-resistant. Immunotherapeutic strategies are already part of the therapeutic plans in OAC; a greater understanding of the immune microenvironment underlying oesophageal adenocarcinoma is needed if we are to exploit the inherent cancer-fighting capabilities of each patient's immune system. Therefore, implementing the crosstalks between the tumour and its microenvironment (TME) might be the key to improving overall survival. In this review, we discuss accumulated evidence regarding TME and immune checkpoint inhibitors in OAC, as well as recent and ongoing therapeutic attempts to improve patient treatment and outcomes at an individual level.
Collapse
Affiliation(s)
- Dimitra V Peristeri
- Department of Upper GI Surgery, Salford Royal Hospital, Stott Lane, Salford, UK.
| | - Munir Tarazi
- Department of Upper GI Surgery, Salford Royal Hospital, Stott Lane, Salford, UK; Department of Surgery and Cancer, Imperial College London, UK
| | - Patrick Casey
- Department of Upper GI Surgery, Salford Royal Hospital, Stott Lane, Salford, UK
| | - Arfon Powell
- Department of Upper GI Surgery, Salford Royal Hospital, Stott Lane, Salford, UK; Division of Diabetes, Endocrinology, and Gastroenterology, University of Manchester, UK
| | - Javed Sultan
- Department of Upper GI Surgery, Salford Royal Hospital, Stott Lane, Salford, UK; Division of Cancer Sciences, School of Medical Sciences, University of Manchester, UK
| |
Collapse
|
45
|
Zhou Y, Yu H, Chen C, Li A, Zhang X, Qiu H, Du W, Fu S, Zhang L, Hong S. Placebo immune-related adverse events (irAEs): A neglected phenomenon in cancer immunotherapy trials. Eur J Cancer 2024; 208:114203. [PMID: 38981314 DOI: 10.1016/j.ejca.2024.114203] [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: 05/17/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/11/2024]
Abstract
PURPOSE This study aims to investigate the underexplored prevalence of placebo-reported immune-related adverse events (irAEs) in immune checkpoint inhibitor (ICI) trials. METHODS We searched public databases for randomized clinical trials (RCTs) involving ICI versus placebo treatments in patients with malignancies. Study characteristics and irAEs occurrences were extracted for meta-analyses using a random-effects model. MAIN OUTCOMES Proportions of patients reported to experience any grade and grade 3 to 5 placebo irAEs; the risk ratio (RR) of reporting 'false' irAEs in the experiment arm (defined as 'false-irAE ratio', calculated by dividing the proportion of patients documented with irAEs in the placebo arm by that in the experimental arm). RESULTS 47 RCTs with 30,119 patients were analyzed. The pooled proportion of patients reported to experience any grade and grade 3 to 5 irAEs among placebo participants was 22.85 % (17.33 %-29.50 %) and 3.40 % (2.35 %-4.63 %), respectively. The pooled proportion of placebo-treated patients who experienced serious irAEs was 0.67 % (0.03 %-1.91 %). Treatment discontinuation and death due to placebo irAEs occurred in 0.69 % (<0.01 %-1.30 %) and 0.12 % (<0.01 %-0.40 %) of patients, respectively. The false-irAE ratio for any grade and grade 3 to 5 irAEs were 0.49 and 0.28. The false-irAE ratio was significantly higher in RCTs with control arms of placebo plus non-immunotherapy than in those with placebo alone (any grade: 0.57 vs. 0.32, P < 0.001; grade 3 to 5: 0.36 vs. 0.12, P = 0.009). CONCLUSION Our analyses of placebo-treated participants in ICI RCTs document the common occurrence of placebo irAEs. These findings are important for interpreting irAE profiles, avoiding inappropriate therapeutic interventions.
Collapse
Affiliation(s)
- Yixin Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of VIP region, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Hui Yu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Chen Chen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Radiation Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Anlin Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Xuanye Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Huijuan Qiu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of VIP region, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Wei Du
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China
| | - Sha Fu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation of Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou 510120, China; Department of Cellular & Molecular Diagnostics Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, 107 Yanjiang West Road, Guangzhou 510120, China.
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China.
| | - Shaodong Hong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou 510060, China; Guangdong Provincial Clinical Research Center for Cancer, Guangzhou, Guangdong 510060, China; Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou 510060, China.
| |
Collapse
|
46
|
Adiwinata R, Tandarto K, Tanadi C, Waleleng BJ, Haroen H, Rotty L, Gosal F, Rotty L, Hendratta C, Lasut P, Winarta J, Waleleng A, Simadibrata P, Simadibrata M. Immune checkpoint inhibitor colitis, a rising issue in targeted cancer therapy era: A literature review. ROMANIAN JOURNAL OF INTERNAL MEDICINE = REVUE ROUMAINE DE MEDECINE INTERNE 2024; 62:219-230. [PMID: 38595047 DOI: 10.2478/rjim-2024-0015] [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: 11/29/2023] [Indexed: 04/11/2024]
Abstract
Research advances in the oncology treatment field have led to the widespread use of immunotherapy. The usage of immune checkpoint inhibitor (ICI) has improved the survival of cancer patients with metastases. This has also led to the rapidly expanding indications for ICI use. However, ICI usage may lead to toxicity, which may be immune-related, in different organ-specific targets. The immune-related adverse events (irAEs) of ICI may lead to increased morbidity, decreased quality of life, and early termination of ICI. The clinical manifestations of irAEs in the gastrointestinal system are variable, ranging from self-limited to life-threatening or fatal events. In this review article, we would like to focus on discussing ICI-induced colitis, which is one of the most common ICI irAEs in the gastrointestinal tract.
Collapse
Affiliation(s)
- Randy Adiwinata
- 1Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi "Prof. dr. R. D. Kandou" Hospital, Manado, Indonesia
- 2Gastrointestinal Cancer Center, MRCCC Siloam Hospital Semanggi, Jakarta, Indonesia
| | - Kevin Tandarto
- 3Intensive Care Unit, Columbia Asia Hospital, Semarang, Indonesia
| | | | - Bradley Jimmy Waleleng
- 5Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi "Prof. dr. R. D. Kandou" Hospital, Manado, Indonesia
| | - Harlinda Haroen
- 6Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi/Prof. dr. R. D. Kandou Hospital, Manado, Indonesia
| | - Linda Rotty
- 6Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi/Prof. dr. R. D. Kandou Hospital, Manado, Indonesia
| | - Fandy Gosal
- 5Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi "Prof. dr. R. D. Kandou" Hospital, Manado, Indonesia
| | - Luciana Rotty
- 5Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi "Prof. dr. R. D. Kandou" Hospital, Manado, Indonesia
| | - Cecilia Hendratta
- 6Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi/Prof. dr. R. D. Kandou Hospital, Manado, Indonesia
| | - Pearla Lasut
- 6Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi/Prof. dr. R. D. Kandou Hospital, Manado, Indonesia
| | - Jeanne Winarta
- 5Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi "Prof. dr. R. D. Kandou" Hospital, Manado, Indonesia
| | - Andrew Waleleng
- 5Division of Gastroenterology-Hepatology, Department of Internal Medicine, Faculty of Medicine, Universitas Sam Ratulangi "Prof. dr. R. D. Kandou" Hospital, Manado, Indonesia
| | - Paulus Simadibrata
- 2Gastrointestinal Cancer Center, MRCCC Siloam Hospital Semanggi, Jakarta, Indonesia
- 7Abdi Waluyo Hospital, Jakarta, Indonesia
| | - Marcellus Simadibrata
- 8Division of Gastroenterology, Department of Internal Medicine, Faculty of Medicine, Universitas Indonesia, Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| |
Collapse
|
47
|
Ranawat P, Sharma B, Singh P, Kaur T. Exploring Cancer Immunotherapy and the Promise of Cancer Vaccine. ADVANCES IN MEDICAL DIAGNOSIS, TREATMENT, AND CARE 2024:265-310. [DOI: 10.4018/979-8-3693-3976-3.ch008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The goal of immunotherapy is to enhance the immune system by managing the immunological-mediated microenvironment, which makes it possible for immune cells to locate and destroy tumour cells at vital nodes. In the tumor microenvironment, immune responses against tumour cells are reduced when these cells take up immune-regulatory mechanisms. An environment that suppresses the immune system is facilitated by immune cells, including regulatory T cells, regulatory B cells, dendritic cells, and myeloid-derived suppressor cells. In a number of cancer types, adoptive immune cells and immune checkpoint modulators have shown impressive anticancer benefits. Tumour growth is facilitated in large part by immune cells found in the tumour microenvironment (TME). Tumour growth may be stimulated or inhibited by these cells. The ability of the immune system to elude detection by cancer cells offers new possibilities for innovative cancer treatment strategies.
Collapse
|
48
|
Mongeon B, Hébert-Doutreloux J, Surendran A, Karimi E, Fiset B, Quail DF, Walsh LA, Jenner AL, Craig M. Spatial computational modelling illuminates the role of the tumour microenvironment for treating glioblastoma with immunotherapies. NPJ Syst Biol Appl 2024; 10:91. [PMID: 39155294 PMCID: PMC11330976 DOI: 10.1038/s41540-024-00419-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 08/07/2024] [Indexed: 08/20/2024] Open
Abstract
Glioblastoma is the most common and deadliest brain tumour in adults, with a median survival of 15 months under the current standard of care. Immunotherapies like immune checkpoint inhibitors and oncolytic viruses have been extensively studied to improve this endpoint. However, most thus far have failed. To improve the efficacy of immunotherapies to treat glioblastoma, new single-cell imaging modalities like imaging mass cytometry can be leveraged and integrated with computational models. This enables a better understanding of the tumour microenvironment and its role in treatment success or failure in this hard-to-treat tumour. Here, we implemented an agent-based model that allows for spatial predictions of combination chemotherapy, oncolytic virus, and immune checkpoint inhibitors against glioblastoma. We initialised our model with patient imaging mass cytometry data to predict patient-specific responses and found that oncolytic viruses drive combination treatment responses determined by intratumoral cell density. We found that tumours with higher tumour cell density responded better to treatment. When fixing the number of cancer cells, treatment efficacy was shown to be a function of CD4 + T cell and, to a lesser extent, of macrophage counts. Critically, our simulations show that care must be put into the integration of spatial data and agent-based models to effectively capture intratumoral dynamics. Together, this study emphasizes the use of predictive spatial modelling to better understand cancer immunotherapy treatment dynamics, while highlighting key factors to consider during model design and implementation.
Collapse
Affiliation(s)
- Blanche Mongeon
- Sainte-Justine University Hospital Azrieli Research Centre, Montréal, QC, Canada
- Department of Mathematics and Statistics, Université de Montréal, Montréal, QC, Canada
| | | | - Anudeep Surendran
- Center for Advanced Systems Understanding, Görlitz, Germany
- Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - Elham Karimi
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
| | - Benoit Fiset
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
| | - Daniela F Quail
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montréal, QC, Canada
- Department of Medicine, Division of Experimental Medicine, McGill University, Montréal, QC, Canada
| | - Logan A Walsh
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, QC, Canada
- Department of Human Genetics, McGill University, Montréal, QC, Canada
| | - Adrianne L Jenner
- School of Mathematical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Morgan Craig
- Sainte-Justine University Hospital Azrieli Research Centre, Montréal, QC, Canada.
- Department of Mathematics and Statistics, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
49
|
Sunyur AM, Alkhayat D, Mohammad HA, Alahmadi HA, Alharbi LA, Khawaji ZY, Badawi AS. Clinical Patterns and Factors Contributing to Ophthalmic and Otologic Events Associated With Immune Checkpoint Inhibitors: A Narrative Review. Cureus 2024; 16:e66611. [PMID: 39258067 PMCID: PMC11386091 DOI: 10.7759/cureus.66611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2024] [Indexed: 09/12/2024] Open
Abstract
Immune checkpoint inhibitors, which are a type of cancer immunotherapy, have been associated with the development of adverse events related to an overactive immune system caused by the effect of this type of therapy. It affects a wide range of organs, including the ear and eye. Ophthalmic toxicity related to immune checkpoint inhibitors usually occurs bilaterally. Corneal toxicity (mainly dry eye disease) and uveitis are the most commonly reported patterns of toxicity. Other patterns of involvement include optic neuritis, serous retinal detachment, keratitis, ophthalmoplegia, and ocular myasthenia, but are not limited to them. Potential factors contributing to the development of toxicity are age, previous history of ocular immune disease, type, doses, and duration of treatment, and race. Ototoxicity is also reported in the literature, usually manifesting as bilateral, symmetrical/asymmetrical hearing loss. Ear toxicity presenting as ear fullness, tinnitus, and vertigo has also been mentioned in the literature. Hearing loss is often associated with word/speech recognition. An audiogram usually shows a pattern of sensorineural hearing loss. Otitis media has also been reported to be a potential cause of ear toxicity. Immune checkpoint inhibitor toxicity was present more commonly when used along with other anti-neoplastic agents. Ear toxicity, which presumably results from damage to the melanocytes in the ear, often presents with other melanocytotic manifestations, like uveitis and vitiligo. According to the literature, some agents (ipilimumab, nivolumab, atezolizumab, and pembrolizumab) were more commonly associated with toxic effects on the eye and ear and more when combined with each other.
Collapse
Affiliation(s)
- Amal M Sunyur
- Medicine and Surgery, Taibah University, Medina, SAU
| | - Duaa Alkhayat
- Department of Clinical Oncology, Taibah University, Medina, SAU
| | | | | | | | | | | |
Collapse
|
50
|
Baron JM, Widatalla S, Gubens MA, Khalil F. Real-World Biomarker Test Ordering Practices in Non-Small Cell Lung Cancer: Interphysician Variation and Association With Clinical Outcomes. JCO Precis Oncol 2024; 8:e2400039. [PMID: 39208373 PMCID: PMC11371103 DOI: 10.1200/po.24.00039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/14/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
PURPOSE Patients with metastatic or advanced non-small cell lung cancer (NSCLC) need biomarker testing, including, in most cases, anaplastic lymphoma kinase (ALK), epidermal growth factor receptor (EGFR), and PD-L1, to identify options for targeted therapies and to optimally incorporate immune checkpoint inhibitors into therapeutic regimens. We sought to examine real-world patterns of biomarker testing, quantify interphysician practice variation, and correlate testing with clinical outcomes. METHODS We extracted real-world data from a nationwide electronic health record-derived deidentified database from 17,165 patients diagnosed with advanced NSCLC between 2018 and 2021 and receiving care in the community setting. We analyzed data using descriptive analyses, fixed- and mixed-effects logistic regression models, and proportional hazard models. RESULTS Only 67% of all 17,165 patients and 77% of patients with nonsquamous, metastatic NSCLC had ALK, EGFR, and PD-L1 testing within 90 days of diagnosis. Later diagnosis year (2019-2021 compared with 2018) was associated with higher rates of ALK, EGFR, and PD-L1 testing; stage IIIB/C disease (compared with stage IV), squamous histology, and Black or African American race were associated with lower rates. Interphysician variation was substantial with a median odds ratio between physicians (adjusted for patient factors) of 1.78 for ALK, EGFR, and PD-L1 testing. Patients with nonsquamous, metastatic NSCLC had significantly prolonged survival if tested with all three biomarkers (median, 364 days for all three v 180 for none of the three; hazard ratio, 0.67; P < .001). CONCLUSION Rates of biomarker testing appear suboptimal with substantial interphysician variation. Testing correlates with improved survival, although causality cannot be proven from this study. Additional work is needed to address the underlying causes of suboptimal test ordering.
Collapse
Affiliation(s)
- Jason M. Baron
- Roche Diagnostics, Medical and Scientific Affairs, Indianapolis, IN
| | - Sarrah Widatalla
- Roche Diagnostics, Medical and Scientific Affairs, Indianapolis, IN
| | - Matthew A. Gubens
- Division of Hematology and Oncology, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Farah Khalil
- Department of Anatomic Pathology, Moffitt Cancer Center, Tampa, FL
- University of South Florida, Tampa, FL
| |
Collapse
|