1
|
Qiao Y, Xie D, Li Z, Cao S, Zhao D. Global research trends on biomarkers for cancer immunotherapy: Visualization and bibliometric analysis. Hum Vaccin Immunother 2025; 21:2435598. [PMID: 39773010 PMCID: PMC11730411 DOI: 10.1080/21645515.2024.2435598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/08/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
The global burden of cancer continues to grow, posing a significant public health challenge. Although cancer immunotherapy has shown significant efficacy, the response rate is not high. Therefore, the objective of our research was to identify the latest research trends and hotspots on biomarkers from 1993 to 2023. Data were collected from the database Web of Science core collection. Bibliometric analysis and visualization were conducted with CiteSpace(6.3.1), VOSviewer (v1.6.20), R-bibliometrix(v4.3.3), and Microsoft Excel(2019). A total of 2686 literatures were retrieved. The sheer annual volume of publications has shown a rapid upward trend since 2015. The United States has generated the most publications and Harvard University ranked as a leading institution. The global biomarker research on immune checkpoint inhibitors (ICIs) revealed regional differences and in-depth explorations should be promoted in developing countries. Although China has become the second largest country in terms of publication, the average citation per paper and the total link strength were both lower than the other countries. The research on biomarkers mainly concentrated upon the following aspects: PD-1/PD-L1, CTLA-4, gene expression, adverse events, total mutational burden (TMB), body mass index (BMI), gut microbiota, cd8(+)/cd4(+) t-cells, and blood-related biomarkers such as lactate dehydrogenase (LDH), neutrophil-lymphocyte ratio (NLR), cytokines. Furthermore, "artificial intelligence" and "machine learning" have become the most important research hotspot over the last 2 y, which will help us to identify useful biomarkers from complex big data and provide a basis for precise medicine for malignant tumors.
Collapse
Affiliation(s)
- Yuan Qiao
- Department of Clinical Pharmacy, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| | - Dong Xie
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhengxiang Li
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Shaohua Cao
- Department of Clinical Pharmacy, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| | - Dong Zhao
- Department of Clinical Laboratory, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| |
Collapse
|
2
|
Kumar KP, Madhusoodanan M, Pangath M, Menon D. Innovative landscapes in intraperitoneal therapy of ovarian cancer. Drug Deliv Transl Res 2025; 15:1877-1906. [PMID: 39888579 DOI: 10.1007/s13346-024-01765-w] [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: 12/09/2024] [Indexed: 02/01/2025]
Abstract
Epithelial ovarian cancer is the most prevalent gynecological malignancy, characterized by high mortality rates due to its late-stage diagnosis and frequent recurrence. The current standard of care for ovarian cancer is a combination of debulking surgery followed by the conventional mode of chemotherapy. Despite significant advances in therapeutic modalities, the overall survival rate of EOC continues to be poor, mainly because low concentrations of the chemotherapeutics reach the peritoneum, which is the primary site of ovarian cancer, leading to disease relapse. Here, intraperitoneal chemotherapy gains advantage due to its ability to deliver the drug molecules directly to the peritoneal cavity and provide localized and sustained effects. This is facilitated by the use of diverse kinds of nano or micron sized delivery systems, which help in transporting drugs, vaccines, antibodies and genes appropriately to the peritoneum for its desired function. This review article delves on how intraperitoneal delivery impacts the therapy of epithelial ovarian cancer spanning the conventional therapeutic modes to the recent nanoinnovations in chemotherapy, immunotherapy and gene therapy.
Collapse
Affiliation(s)
- Krishna Pradeep Kumar
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Maneesha Madhusoodanan
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Meghna Pangath
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India
| | - Deepthy Menon
- Amrita School of Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, Kerala, India.
| |
Collapse
|
3
|
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
|
4
|
Yu T, Wu C, Sahasranaman S, Tian X, Fei Li Y, Tang Z, Yang Y, Wan Y, Zhang Q, Schnell P, Mendoza‐Naranjo A, Abdrashitov R, Hanley WD, Budha N. Clinical Pharmacology Overview of Tislelizumab in Patients With Advanced Tumors With a Focus on Racial Impact. Clin Transl Sci 2025; 18:e70221. [PMID: 40286322 PMCID: PMC12033005 DOI: 10.1111/cts.70221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/30/2025] [Accepted: 02/22/2025] [Indexed: 04/29/2025] Open
Abstract
Tislelizumab, an anti-programmed cell death protein-1 monoclonal antibody, has demonstrated improved survival over the standard of care for multiple cancers. However, tislelizumab's effectiveness across different racial/ethnicity groups warrants further evaluation. This clinical pharmacology overview includes tislelizumab's pharmacokinetic properties, correlations with efficacy and safety, and immunogenicity, with a focus on racial impact. Non-compartmental pharmacokinetic analysis was conducted using data from Asian and White patients enrolled in BGB-A317-001 and BGB-A317-102. Population pharmacokinetic analyses used pooled data from 12 clinical studies to evaluate the impact of intrinsic/extrinsic factors on tislelizumab's pharmacokinetic properties, including race effect. Exposure-efficacy/exposure-safety relationships and immunogenicity assessments were evaluated for the phase III BGB-A317-302/-303 studies. Tislelizumab exhibited dose-proportional pharmacokinetics, and there were no clinically meaningful differences in tislelizumab's pharmacokinetic parameters at 200 mg once every 3 weeks between BGB-A317-001 (n = 12, 83% White patients) and BGB-A317-102 (n = 20, 100% Chinese patients); race was not a significant covariate. No clinically relevant exposure-efficacy/-safety relationships were observed in BGB-A317-302/-303. Incidence of anti-drug antibodies (ADAs) was similar between Asian and White patients. The presence of ADAs was not clinically relevant for tislelizumab's pharmacokinetic properties, efficacy, or safety. There were no differences in tislelizumab's pharmacokinetic or ADA characteristics between Asian and White patients with advanced cancer and no clinically relevant exposure-efficacy/-safety dependency or impact of immunogenicity on efficacy and safety. Data from the extensive clinical program of tislelizumab support the use of tislelizumab across broad patient populations with relevant tumor types.
Collapse
MESH Headings
- Female
- Humans
- Male
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antineoplastic Agents, Immunological/pharmacokinetics
- Antineoplastic Agents, Immunological/administration & dosage
- Antineoplastic Agents, Immunological/adverse effects
- Asian People
- Clinical Trials, Phase III as Topic
- Dose-Response Relationship, Drug
- Neoplasms/drug therapy
- Neoplasms/ethnology
- Neoplasms/immunology
- Neoplasms/pathology
- Treatment Outcome
- White People
Collapse
Affiliation(s)
- Tian Yu
- Clinical Pharmacology and PharmacometricsBeiGene USA, Inc.San CarlosCaliforniaUSA
| | - Chi‐Yuan Wu
- Clinical Pharmacology and PharmacometricsBeiGene USA, Inc.San CarlosCaliforniaUSA
| | | | - Xianbin Tian
- PK SciencesNovartis Biomedical Research, Novartis Pharmaceutical, Inc.East HanoverNew JerseyUSA
| | - Ying Fei Li
- Pharmacometrics, Oncology, Global Drug DevelopmentNovartis Pharmaceutical, Inc.East HanoverNew JerseyUSA
| | - Zhiyu Tang
- Clinical Pharmacology and PharmacometricsBeiGene USA, Inc.San CarlosCaliforniaUSA
| | - Yanfei Yang
- Clinical BA BeiGene (Shanghai) Co., Ltd.ShanghaiChina
| | - Ya Wan
- Scientific Programming, BeiGene (Shanghai) Co., Ltd.ShanghaiChina
| | - Quting Zhang
- Scientific Programming, BeiGene (Shanghai) Co., Ltd.ShanghaiChina
| | - Patrick Schnell
- Global Product SafetyBeiGene USA, Inc.Ridgefield ParkNew JerseyUSA
| | | | | | - William D. Hanley
- Clinical Pharmacology and PharmacometricsBeiGene USA, Inc.San CarlosCaliforniaUSA
| | - Nageshwar Budha
- Clinical Pharmacology and PharmacometricsBeiGene USA, Inc.San CarlosCaliforniaUSA
| |
Collapse
|
5
|
Naffaa MM, Al-Ewaidat OA, Gogia S, Begiashvili V. Neoantigen-based immunotherapy: advancing precision medicine in cancer and glioblastoma treatment through discovery and innovation. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002313. [PMID: 40309350 PMCID: PMC12040680 DOI: 10.37349/etat.2025.1002313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Neoantigen-based immunotherapy has emerged as a transformative approach in cancer treatment, offering precision medicine strategies that target tumor-specific antigens derived from genetic, transcriptomic, and proteomic alterations unique to cancer cells. These neoantigens serve as highly specific targets for personalized therapies, promising more effective and tailored treatments. The aim of this article is to explore the advances in neoantigen-based therapies, highlighting successful treatments such as vaccines, tumor-infiltrating lymphocyte (TIL) therapy, T-cell receptor-engineered T cells therapy (TCR-T), and chimeric antigen receptor T cells therapy (CAR-T), particularly in cancer types like glioblastoma (GBM). Advances in technologies such as next-generation sequencing, RNA-based platforms, and CRISPR gene editing have accelerated the identification and validation of neoantigens, moving them closer to clinical application. Despite promising results, challenges such as tumor heterogeneity, immune evasion, and resistance mechanisms persist. The integration of AI-driven tools and multi-omic data has refined neoantigen discovery, while combination therapies are being developed to address issues like immune suppression and scalability. Additionally, the article discusses the ongoing development of personalized immunotherapies targeting tumor mutations, emphasizing the need for continued collaboration between computational and experimental approaches. Ultimately, the integration of cutting-edge technologies in neoantigen research holds the potential to revolutionize cancer care, offering hope for more effective and targeted treatments.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ola A Al-Ewaidat
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Sopiko Gogia
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Valiko Begiashvili
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| |
Collapse
|
6
|
Rezazadeh‐Gavgani E, Majidazar R, Lotfinejad P, Kazemi T, Shamekh A. Immune Checkpoint Molecules: A Review on Pathways and Immunotherapy Implications. Immun Inflamm Dis 2025; 13:e70196. [PMID: 40243372 PMCID: PMC12004596 DOI: 10.1002/iid3.70196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 03/12/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Today, treating cancer patients with monoclonal antibodies (mAbs), by targeting immune checkpoints, is one of the most outstanding immunotherapeutic methods. Immune checkpoints are special molecules having regulatory role in immune system responses. Once these molecules are presented on cancer cells, these cells will be capable of evading the immune system through their own specific pathways. This Evasion can be prevented by counterbalancing immune system responses with immune checkpoints related antibodies. AIMS The current study aimed to highlight immunotherapy and its methods, describe the immune checkpoints pathways, outline the immune checkpoint inhibitors (ICIs), and recent advances in this field, and sketch an outlook on the best treatment options for the most prevalent cancers. MATERIALS & METHODS This research implemented a narrative review method. A comprehensive literature review on the history, molecular and cellular biology, and the clinical aspects of immune checkpoint molecules was performed to illustrate the pathways involved in various cancers. Also, currently-available and future potential immunotherapies targeting these pathways were extracted from the searched studies. RESULTS The immune checkpoint family consists of many molecules, including CTLA-4, PD-1, PD-L1, LAG-3, TIM-3, and TIGIT. Attempts to modify these molecules in cancer treatment led to the development of therapeutic monoclonal antibodies. Most of these antibodies have entered clinical studies and some of them have been approved by the Food and Drug Administration (FDA) to be used in cancer patients' treatment plans. DISCUSSION With these novel treatments and the combination therapies they offer, there is also hope for better treatment outcomes for the previously untreatable metastatic cancers. In spite of the beneficial aspects of immune checkpoint therapy, similar to other treatments, they may cause side effects in some patients. Therefore, more studies are needed to reduce the probable side effects and uncover their underlying mechanism. CONCLUSION Based on the data shown in this review, there is still a lack of knowledge about the complete properties of ICIs and the possible combination therapies that we may be able to implement to achieve a better treatment response in cancer patients.
Collapse
Affiliation(s)
| | - Reza Majidazar
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
| | - Parisa Lotfinejad
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyTabriz University of Medical SciencesTabrizIran
| | - Tohid Kazemi
- Immunology Research CenterTabriz University of Medical SciencesTabrizIran
- Department of ImmunologyTabriz University of Medical SciencesTabrizIran
| | - Ali Shamekh
- Student Research CommitteeTabriz University of Medical SciencesTabrizIran
- Aging Research InstituteTabriz University of Medical SciencesTabrizIran
| |
Collapse
|
7
|
Naumov SS, Tashireva LA, Krakhmal NV, Vtorushin SV. Evaluation of immune-checkpoint molecules in dMMR/pMMR colorectal cancer by multiplex immunohistochemistry. Clin Transl Oncol 2025; 27:1668-1680. [PMID: 39240301 DOI: 10.1007/s12094-024-03691-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/21/2024] [Indexed: 09/07/2024]
Abstract
PURPOSE Colorectal cancer is the most common malignancy worldwide. A number of pathological and molecular genetic criteria are currently used as predictors of the disease. They include assessment of MMR deficiency or MSI/MSS status, which among others, determine the immunogenicity of the tumor. In this regard, the evaluation of PD-L1, CTLA-4, and LAG-3 immune checkpoint molecules in different tumor compartments according to MMR status deserves special attention. METHODS Multiplex immunohistochemistry was used to evaluate the expression of immune checkpoint molecules in the tumor core and at the invasive margin. RESULTS Data analysis showed the predominance of PD-L1 (p = 0.011), CTLA-4 (p = 0.004), and LAG-3 (p = 0.013) expression at the invasive margin of dMMR carcinomas compared to pMMR samples. Quantitative analysis of TILs population in the tumor core and at the invasive margin allowed establishment of the predominance of CD3+ and CD8+ lymphocytes at the invasive margin of dMMR carcinomas. Study of the CD163+ macrophages population in the same tumor compartments revealed the predominance of the studied TAMs in the core and at the invasive margin of dMMR carcinomas and the predominance of CD163+ macrophages with PD-L1-phenotype in the tumor stroma. CONCLUSION This study revealed a significant predominance of PD-L1, CTLA-4, LAG-3, and CD 3+ ,CD8+ lymphocytes in dMMR colorectal carcinomas. Further research on the immune landscape in different tumor compartments will likely have high prognostic value for CRC patients, as it might expand the criteria for prescribing immunotherapy.
Collapse
Affiliation(s)
| | - Liubov Alexandrovna Tashireva
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences , Tomsk, 634009, Russia
| | - Nadezhda Valerievna Krakhmal
- Siberian State Medical University, Tomsk, 634050, Russia
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences , Tomsk, 634009, Russia
| | - Sergey Vladimirovich Vtorushin
- Siberian State Medical University, Tomsk, 634050, Russia
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences , Tomsk, 634009, Russia
| |
Collapse
|
8
|
Hirao H, Honda M, Tomita M, Li L, Adawy A, Xue W, Hibi T. Intravital Imaging of Immune Responses in the Cancer Microenvironment. Cancer Med 2025; 14:e70899. [PMID: 40257446 PMCID: PMC12010765 DOI: 10.1002/cam4.70899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/18/2025] [Accepted: 04/09/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND To date, many types of immune cells have been identified, but their precise role in cancer immunity remains unclear. Understanding the immune responses involved in cancer and the cancer microenvironment is becoming increasingly important for elucidating disease mechanisms. In recent years, the application of intravital imaging in cancer research has provided new insights into the mechanisms of cancer-specific immune events, including innate and adaptive immunity. RESULTS In this review, we focus on the emerging role of intravital imaging in cancer research and describe how cancer and immune cells can be observed using intravital imaging in vivo. We also discuss new insights gained by this state-of-the-art technique. CONCLUSIONS Intravital imaging is a relatively new field of research that offers significant advantages, including the ability to directly capture cell-cell interactions, pathophysiology, and immune cell dynamics in the cancer microenvironment in vivo.
Collapse
Affiliation(s)
- Hiroki Hirao
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Masaki Honda
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Masahiro Tomita
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Lianbo Li
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Ahmad Adawy
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Weijie Xue
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| | - Taizo Hibi
- Department of Pediatric Surgery and TransplantationKumamoto University Graduate School of Medical SciencesKumamotoJapan
| |
Collapse
|
9
|
Madera G, Hodge A, Roskelly L, Greenbaum C. What Is the Impact of Novel Systemic Anticancer Therapy on Acute Oncology Education and Service Delivery? Semin Oncol Nurs 2025; 41:151842. [PMID: 39986898 DOI: 10.1016/j.soncn.2025.151842] [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: 12/03/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 02/24/2025]
Abstract
OBJECTIVES To discuss the importance of educating healthcare professionals about oncological emergencies linked to novel systemic anticancer therapy (SACT) and the impact on acute oncology (AO) services. METHODS This discussion is based on clinical expertise and informed by current literature. RESULTS Novel SACT, such as immune-checkpoint inhibitors, have more complex toxicity profiles and can be challenging to recognize and treat. An increasing prevalence of toxicity is expected as new drugs are developed and the numbers of patients living with and beyond cancer expand; more data are required to capture the full extent of this. There are knowledge gaps within the healthcare workforce, particularly outside oncology-specialist settings. Focused research in this area will provide direction for targeted educational interventions. CONCLUSIONS Insufficient SACT education is a safety issue; severe toxicities can be fatal but initial symptoms can be subtle and may be missed. We argue that emergency care pathways can help to streamline the appropriate management of patients with SACT toxicity, but awareness of AO issues remains "everyone's business." Continuing clinical education is key to maintaining awareness of newly developed SACT. AO service models may vary, but AO competence assessment passports can provide a standardized method of evidencing AO knowledge and skills. IMPLICATIONS FOR NURSING PRACTICE Oncology nurses, as a highly specialized and knowledgeable part of the healthcare workforce, are key in supporting interprofessional education. By using existing cancer nursing frameworks, this can support the implementation of the AO Passports. The learning and development of AO services in the UK can be transferred internationally.
Collapse
Affiliation(s)
- Gina Madera
- Medical Oncology/Networked Services, The Christie NHS Foundation Trust, Manchester, UK.
| | - Ali Hodge
- Immunotherapy and Acute Oncology, Cancer Services Division, University Hospitals Sussex NHS Foundation Trust, Brighton, UK
| | - Lara Roskelly
- Macmillan Acute Oncology Team, Cancer Division, Frimley Health NHS Foundation Trust, UK
| | - Clare Greenbaum
- Workforce and Education, Greater Manchester Cancer Alliance, The Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|
10
|
Belényesi SK, Patmore S, O'Driscoll L. Extracellular vesicles and the tumour microenvironment. Biochim Biophys Acta Rev Cancer 2025; 1880:189275. [PMID: 39900204 DOI: 10.1016/j.bbcan.2025.189275] [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: 06/11/2024] [Revised: 12/18/2024] [Accepted: 01/29/2025] [Indexed: 02/05/2025]
Abstract
Extracellular vesicles (EVs), tiny packages of information released by cells, are well established as being involved in unwanted cell-to-cell communication in cancer. EVs from cancer cells have been associated with the spread of drug resistance, immune suppression, and metastasis. Additional to cancer cells, the tumour microenvironment (TME) involves many cell types -including immune cells, fibroblasts, and endothelial cells, each of which has a potential role in how tumours grow, spread, and respond (or otherwise) to therapy. This review collates and distils research developments regarding the role of EVs in multi-way communication between cells in the TME. Further research including tailored clinical studies are now warranted to determine how best to prevent this extensive adverse communication occurring and/or how best to exploit it for biomarker discovery and as a therapeutic approach, in the interest of patients and also for economic benefit.
Collapse
Affiliation(s)
- Szilárd-Krisztián Belényesi
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland; Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland
| | - Sean Patmore
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland; Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland
| | - Lorraine O'Driscoll
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Ireland; Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland; Trinity St. James's Cancer Institute, Trinity College Dublin, Ireland.
| |
Collapse
|
11
|
Georgopoulou S, Droney J, Jaganathan PP, Howell P, Doherty AM, Young K, Cruickshank S. Identification of the PROMs used to assess ICI toxicities and HRQoL in patients receiving immune checkpoint inhibitor treatment in cancer care and their suitability: A systematic review. Cancer Treat Rev 2025; 135:102862. [PMID: 40056766 DOI: 10.1016/j.ctrv.2024.102862] [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: 09/02/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 03/10/2025]
Abstract
BACKGROUND The implementation of patient-reported outcome measures (PROMs) in the clinical identification of immunotherapy toxicities is a complex intervention. There has been very little work evaluating the clinical utility and generalisability of PROMs used after immune checkpoint inhibitor (ICI) treatment to date. We reviewed evidence on the use of PROMs assessing toxicities and health-related quality of life in patients treated with ICIs. METHODS PubMed, EMBASE, MEDLINE, PsycInfo, CINAHL, Web of Knowledge, the Cochrane Library were searched (January 2008 - October 2024). Quantitative studies reporting the use of PROMs to identify, assess and manage toxicities at any timepoint and HRQoL associated with ICI treatment in adult patients with cancer were included. A narrative synthesis describes the key characteristics of the PROMs identified. RESULTS 43 studies were included; 12 on melanoma/skin, 12 on lung and 19 on other cancers. Study designs included 20 randomised controlled trials, 14 cohort studies, six cross-sectional studies and three non-randomised interventional trials. The lack of ICI-specific PROMs was highlighted, particularly as the PROMs used lacked sufficient sensitivity for ICI treatments. CONCLUSIONS There is need for an ICI-specific PROM for effective assessment of toxicities and a tailored PROM for assessment of HRQoL. Some suggested key domains by certain studies for ICI-specific PROMs include: (a) ICI-specific items (e.g. certain USD-I and PRO-CTCAE items) to capture symptoms associated with ICI treatments such as rash, myalgia, (b) role, psychological, emotional and social functioning domains within HRQoL assessments and (c) additional patient-reported toxicities not included in existing PROMs. Findings emphasize the importance of using a disease-specific PROM that is applicable, acceptable and sufficiently sensitive to identify toxicities and HRQoL issues across all stages.
Collapse
Affiliation(s)
| | - Joanne Droney
- The Royal Marsden NHS Foundation Trust, London, United Kingdom; Imperial College London, London, United Kingdom
| | | | - Paul Howell
- The Royal Marsden School, London, United Kingdom
| | | | - Kate Young
- The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | | |
Collapse
|
12
|
Strazzabosco G, Liboni A, Pezzi G, Alogna A, Bortolotti D. Insights into Liposomal and Gel-Based Formulations for Dermatological Treatments. Gels 2025; 11:245. [PMID: 40277680 PMCID: PMC12027463 DOI: 10.3390/gels11040245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/14/2025] [Accepted: 03/23/2025] [Indexed: 04/26/2025] Open
Abstract
Dermatological diseases pose a significant challenge due to their chronic nature, complex pathophysiology, and the need for effective, patient-friendly treatments. Recent advancements in liposomal and gel-based formulations have played a crucial role in improving drug delivery, therapeutic efficacy, and patient compliance. Liposomal formulations have garnered considerable attention in dermatology due to their ability to encapsulate both hydrophilic and lipophilic compounds, enabling controlled drug release and enhanced skin penetration. However, challenges such as formulation complexity, stability issues, and regulatory constraints remain. Similarly, gel-based formulations are widely used due to their ease of application, biocompatibility, and ability to retain active ingredients. However, they also face limitations, including restricted penetration depth, susceptibility to microbial contamination, and challenges in achieving sustained drug release. The integration of liposomal and gel-based technologies offers a promising strategy to overcome current challenges and optimize dermatological drug delivery. This review explores both well-established therapies and recent innovations, offering a comprehensive overview of their applications in the treatment of prevalent dermatological conditions. Ultimately, continued research is essential to refine these formulations, expanding their clinical utility and enhancing therapeutic effectiveness in dermatology.
Collapse
Affiliation(s)
- Giovanni Strazzabosco
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (G.S.); (A.L.)
| | - Alessia Liboni
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (G.S.); (A.L.)
| | - Giulia Pezzi
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy; (G.P.); (D.B.)
| | - Andrea Alogna
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy; (G.P.); (D.B.)
| | - Daria Bortolotti
- Department of Environmental and Prevention Sciences, University of Ferrara, 44121 Ferrara, Italy; (G.P.); (D.B.)
| |
Collapse
|
13
|
Basak U, Mukherjee S, Chakraborty S, Sa G, Dastidar SG, Das T. In-silico analysis unveiling the role of cancer stem cells in immunotherapy resistance of immune checkpoint-high pancreatic adenocarcinoma. Sci Rep 2025; 15:10355. [PMID: 40133473 PMCID: PMC11937529 DOI: 10.1038/s41598-025-93924-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Although immune checkpoint (IC) inhibition is a major treatment modality in cancer-immunotherapy, multiple cancers show low response. Our in-silico exploration by mining cancer datasets using R2, available clinical trial data, and Kaplan-Meier analysis from GEPIA depicted that unlike low-responder (LR) cancers, high-responder (HR) cancers furnish higher IC expression, that upon lowering may provide better prognosis. Contrastingly, pancreatic adenocarcinoma (PAAD) demonstrated high IC expression but low immunotherapy-response. Infiltration scores from TIMER2.0 revealed higher pro-tumor immune subsets and cancer-associated fibroblasts (CAFs) while depicting lower anti-tumor immune subsets in PAAD as compared to HR lung adenocarcinoma (LUAD). Additionally, bioinformatic tool cBioportal showed lesser tumor mutational burden, mismatch repair deficiency and greater percent of driver mutations in TP53, KRAS and CDKN2A in PAAD, supporting its higher immunotherapy-resistance than LUAD. Our search for the 'key' immunotherapy response-deciding factor(s) revealed cancer stem cells (CSCs), the known contributors of therapy-resistance and immuno-evasion, to be positively correlated with above-mentioned driver mutations, pro-tumor immune and CAF subsets; and that PAAD furnished higher expression of CSC genes than LUAD. UMAP/tSNE analyses revealed that high CSC signature is positively correlated with immunotherapy-resistance genes and pro-cancer immune cells, while negatively with cytotoxic-T cells in PAAD. Our in-silico study explains the low immunotherapy-response in high IC-expressing PAAD, wherein CSC plays a pivotal role. Further exploration portrayed correlation of CSCs with immunotherapy-resistance in other LR and HR cancers too, substantiating the need for personalized CSC evaluation and targeting for successful immunotherapy outcomes.
Collapse
Affiliation(s)
- Udit Basak
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Sumon Mukherjee
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Sourio Chakraborty
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Gaurisankar Sa
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Shubhra Ghosh Dastidar
- Bose Institute, Unified Academic Campus, EN 80, Sector V, Bidhannagar, Kolkata, 700091, India.
| | - Tanya Das
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India.
| |
Collapse
|
14
|
Huang YE, Zhou S, Chen S, Chen J, Zhou X, Hou F, Liu H, Yuan M, Jiang W. Mutational signature-based biomarker to predict the response of immune checkpoint inhibitors therapy in cancers. Int J Biol Macromol 2025; 308:142585. [PMID: 40154701 DOI: 10.1016/j.ijbiomac.2025.142585] [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: 07/18/2024] [Revised: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Patients have a limited response rate to immune checkpoint inhibitors (ICIs) therapy. Although several biomarkers have been proposed, their ability to accurately predict the response to ICIs therapy remains unsatisfactory. In addition, mutational signatures were validated to be associated with ICIs therapy. Therefore, we developed a mutational signature-based biomarker (MS-bio) to predict the response to ICIs therapy. Based on differentially mutated genes, we extracted six mutational signatures (single-base substitution (SBS)-A, SBS-B, SBS-C, SBS-D, double-base substitution (DBS)-A, and DBS-B) as MS-bio, and constructed a random forest (RF) model to predict the response. Internal and external validations consistently demonstrated the excellent predictive capability of MS-bio, with an accuracy reaching up to 0.82. Moreover, MS-bio exhibited superior performance compared to existing biomarkers. To further validate the accuracy of MS-bio, we explored its performance in The Cancer Genome Atlas (TCGA) cohort and found that the predicted responders were immunologically "hot". Finally, we found that SBS-C had the highest importance in prediction and was related to T cell differentiation. Overall, here we introduced MS-bio as a novel biomarker for accurately predicting the response to ICIs therapy, thereby contributing to the advancement of precision medicine.
Collapse
Affiliation(s)
- Yu-E Huang
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China; Guizhou Institute of Precision Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Shunheng Zhou
- School of Computer Sciences, University of South China, Hengyang 421001, China
| | - Sina Chen
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Jiahao Chen
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Xu Zhou
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Fei Hou
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Haizhou Liu
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Mengqin Yuan
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Wei Jiang
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| |
Collapse
|
15
|
Selvaraj C, Santhosh R, Alothaim AS, Vijayakumar R, Desai D, Safi SZ, Singh SK. Advances in cancer therapy: unveil the immunomodulatory protein involved in signaling pathways as molecular targets. CHEMICAL PAPERS 2025. [DOI: 10.1007/s11696-025-04007-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 03/05/2025] [Indexed: 04/01/2025]
|
16
|
Popovici IA, Kajanto LA, Popovici LR, Augustin IG, Gales LN. Navigating Stomatologic Complications Secondary to Antineoplastic Agents-A Comprehensive Review. Cancers (Basel) 2025; 17:1061. [PMID: 40227621 PMCID: PMC11987939 DOI: 10.3390/cancers17071061] [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: 02/11/2025] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 04/15/2025] Open
Abstract
This review systematically examines the oral complications associated with conventional and novel anti-cancer therapies. It highlights that while molecularly targeted agents including monoclonal antibodies targeting the vascular endothelial growth factor and its receptor, the epidermal growth factor receptor, tyrosine kinase inhibitors, and immune checkpoint inhibitors tend to exhibit a lower overall toxicity profile compared to traditional cytotoxic chemotherapeutics, they are nonetheless linked to significant oral adverse events. These complications encompass inflammatory mucosal reactions known as mucositis, salivary gland dysfunction leading to a sensation of dryness in the mouth, taste alterations referred to as dysgeusia, and, critically, medication-related osteonecrosis of the jaw. In particular, bone-modifying agents such as bisphosphonates and denosumab disrupt bone remodeling and the formation of new blood vessels, thereby increasing the susceptibility to osteonecrosis of the jaw, especially following invasive dental procedures. The review delineates the multifactorial pathogenesis underlying these toxicities, which involves direct cell toxicity, impaired wound healing, and secondary infections. Furthermore, it emphasizes the importance of pre-treatment dental evaluation and preventive strategies including patient education, prophylactic dental care, and the integration of adjunctive therapies such as laser therapy and autologous platelet concentrates to mitigate these adverse effects. The analysis advocates for interdisciplinary collaboration between oncologists and dental professionals to optimize management protocols, enhance treatment adherence, and ultimately improve the quality of life for oncology patients undergoing anti-cancer therapy.
Collapse
Affiliation(s)
- Ion Alexandru Popovici
- Department of Implant Prosthetic Therapy, Faculty of Dentistry, University of Medicine and Pharmacy “Carol Davila” Bucharest, 010221 Bucharest, Romania;
| | - Lidia Anca Kajanto
- Institute of Oncology “Alexandru Trestioreanu”, 022328 Bucharest, Romania (L.N.G.)
| | | | | | - Laurentia Nicoleta Gales
- Institute of Oncology “Alexandru Trestioreanu”, 022328 Bucharest, Romania (L.N.G.)
- Oncology Department, University of Medicine and Pharmacy “Carol Davila” Bucharest, 050474 Bucharest, Romania
| |
Collapse
|
17
|
Drummond-Guy O, Daly J, Wu A, Stewart N, Milne K, Duff C, Nelson BH, Williams KC, Wisnovsky S. Polysialic acid is upregulated on activated immune cells and negatively regulates anticancer immune activity. Front Oncol 2025; 15:1520948. [PMID: 40182033 PMCID: PMC11965634 DOI: 10.3389/fonc.2025.1520948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/20/2025] [Indexed: 04/05/2025] Open
Abstract
Suppression of anticancer immune function is a key driver of tumorigenesis. Identifying molecular pathways that inhibit anticancer immunity is critical for developing novel immunotherapeutics. One such molecule that has recently been identified is the carbohydrate polysialic acid (polySia), whose expression is dramatically upregulated on both cancer cells and immune cells in breast cancer patient tissues. The role of polySia in the anticancer immune response, however, remains incompletely understood. In this study, we profile polySia expression on both healthy primary immune cells and on infiltrating immune cells in the tumour microenvironment (TME). These studies reveal polySia expression on multiple immune cell subsets in patient breast tumors. We find that stimulation of primary T-cells and macrophages in vitro induces a significant upregulation of polySia expression. We subsequently show that polySia is appended to a range of different carrier proteins within these immune cells. Finally, we find that selective removal of polySia can significantly potentiate killing of breast cancer cells by innate immune cells. These studies implicate polySia as a significant negative regulator of anticancer immunity.
Collapse
Affiliation(s)
- Olivia Drummond-Guy
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - John Daly
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Angeline Wu
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Natalie Stewart
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Katy Milne
- Deeley Research Centre, British Columbia (BC) Cancer, Victoria, BC, Canada
| | - Chloe Duff
- Deeley Research Centre, British Columbia (BC) Cancer, Victoria, BC, Canada
| | - Brad H. Nelson
- Deeley Research Centre, British Columbia (BC) Cancer, Victoria, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada
| | - Karla C. Williams
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Simon Wisnovsky
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
18
|
Fu L, Yong JM, Yeh R, Bartlett F, Whitelock JM, Lord MS. Functionalized Cerium Oxide Nanoparticles Enhance Penetration into Melanoma Spheroids In Vivo through Angiogenesis. Adv Healthc Mater 2025:e2405129. [PMID: 40109098 DOI: 10.1002/adhm.202405129] [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: 12/18/2024] [Revised: 02/27/2025] [Indexed: 03/22/2025]
Abstract
Angiogenesis is a crucial step in tumor progression, including melanoma, making anti-angiogenic strategies a widely explored treatment approach. However, both innate and acquired resistance to these therapies suggest that this approach may need re-evaluation. Nanoparticles have gained attention for their potential to enhance drug delivery and retention within tumors via the bloodstream. However, the in vitro screening of nanoparticles is limited by the inability of preclinical models to replicate the complex tumor microenvironment, especially the blood supply. Here, it is demonstrated that melanoma cells embedded in Matrigel spheroids can engraft in and be vascularized by the chorioallantoic membrane (CAM) of fertilized chicken eggs. This model allows for the assessment of nanoparticle toxicity and accumulation in tumor spheroids, as well as functional effects such as angiogenesis. Cerium oxide nanoparticles (nanoceria) and their surface functionalized derivatives are widely explored for biomedical applications due to their ability to modulate oxidative stress and angiogenesis. Here, it is observed that heparin functionalized nanoceria penetrate melanoma spheroids in the CAM and promote spheroid vascularization to a greater extent than nanoceria alone. This study aids in the development of preclinical cancer models for nanoparticle screening and provides new insight into the interplay between nanoparticle surface coatings and biological effects.
Collapse
Affiliation(s)
- Lu Fu
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Joel M Yong
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Robyn Yeh
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Florence Bartlett
- Katherina Gaus Light Microscopy Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, 2052, Australia
| | - John M Whitelock
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Megan S Lord
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
19
|
Al-Sharabass EA, El-Houseini ME, Effat H, Ibrahim SA, Abdellateif MS. The clinical potential of PDL-1 pathway and some related micro-RNAs as promising diagnostic markers for breast cancer. Mol Med 2025; 31:106. [PMID: 40108523 PMCID: PMC11921724 DOI: 10.1186/s10020-025-01137-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Immune checkpoint pathways play important roles in breast cancer (BC) pathogenesis and therapy. METHODS Expression levels of programmed cell death protein 1 (PD-1), cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), programmed death-ligand 1 (PD-L1), Forkhead box P3 (FOXP3), miR-155, and miR-195 were assessed in the peripheral blood of 90 BC patients compared to 30 healthy controls using quantitative real-time PCR (qRt-PCR). The plasma level of soluble MHC class I chain related-protein B (MIC-B) protein was assessed using the enzyme linked immunosorbent assay (ELISA) technique. The data were correlated to the clinico-pathological characteristics of the patients. RESULTS There was a significant increase in the expression levels of PDL-1 [17.59 (3.24-123), p < 0.001], CTLA-4 [23.34 (1.3-1267), p = 0.006], PD-1 [10.25 (1-280), p < 0.001], FOXP3 [11.5 (1-234.8), p = 0.001], miR-155 [87.3 (1.5-910), p < 0.001] in BC patients compared to normal controls. The miR-195 was significantly downregulated in BC patients [0.23 (0-0.98, p < 0.001]. The plasma level of MIC-B was significantly increased in the BC patients [0.941 (0.204-6.38) ng/ml], compared to the control group [0.351 (0.211-0.884) ng/mL, p < 0.00]. PDL-1, CTLA-4, PD-1, and FOXP3 achieved a specificity of 100% for distinguishing BC patients, at a sensitivity of 93.3%, 82.2%, 62.2%, and 71.1% respectively. The combined expression of PDL-1 and CTLA-4 scored a 100% sensitivity and 100% specificity for diagnosing BC (p < 0.001). The sensitivity, specificity, and AUC of miR-155 were 88.9%, 96.7%, and 0.934; respectively (p < 0.001). While those of miR-195 were 73.3%, 60%, and 0.716; respectively (p = 0.001). MIC-B expression showed a 77.8% sensitivity, 80% specificity, and 0.811 AUC at a cutoff of 1.17 ng/ml (p < 0.001). Combined expression of miR-155 and miR-195 achieved a sensitivity of 91.1%, a specificity of 96.7%, and AUC of 0.926 (p < 0.001). Multivariate analysis showed that PDL-1 (OR:13.825, p = 0.004), CTLA-4 (OR: 20.958, p = 0.010), PD-1(OR:10.550, p = 0.044), MIC-B (OR: 17.89, p = 0.003), miR-155 (OR: 211.356, P < 0.001), and miR-195(OR:0.006, P < 0.001) were considered as independent risk factors for BC. CONCLUSIONS The PB levels of PDL-1, CTLA-4, PD-1, FOXP3, MIC-B, miR-155, and miR-195 could be used as promising diagnostic markers for BC patients.
Collapse
Affiliation(s)
| | - Motawa E El-Houseini
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | - Heba Effat
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | | | - Mona S Abdellateif
- Medical Biochemistry and Molecular Biology, Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt.
| |
Collapse
|
20
|
Eichhorn S, Pili R, Epstein JB, Rajesh R, Satheeshkumar PS. Receiving immunotherapy for the treatment of advanced renal cell carcinoma is associated with higher burden of illness, coagulopathy, cardiac arrhythmia, and disparities. Support Care Cancer 2025; 33:285. [PMID: 40088263 DOI: 10.1007/s00520-025-09353-5] [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: 08/06/2024] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND The adverse outcomes and costs of immunotherapy (IT) have yet to be fully explored. Our study aims to assess the association between the use of IT in patients with metastatic renal cell carcinoma (mRCC) and the burden of illness (BOI), including coagulopathy, arrhythmia, and disparities. METHODS The study used US national data to investigate the association between the use of IT in the mRCC and the BOI. The BOI was measured by total charges and length of stay (LOS). Additionally, we examined coagulopathy, arrhythmia, and disparities in these outcomes. This investigation was conducted using generalized linear models (glm). RESULTS Of 28,535 patients who had mRCC, 230 had previously received IT. In the adjusted glm, after accounting for other variables, "IT" was found to be associated with higher total charges- (coeff = 7.67; 95% CI 4.86 - 12.09). There was no association with IT and LOS. Coagulopathy (aOR = 5.61; 95% CI 2.40 - 13.14) and arrhythmia (aOR = 4.34; 95% CI 2.20 - 8.55) were associated with IT treatment. Moreover, compared to males, females had a lower cardiac arrhythmia risk (aOR 0.83, 95% CI 0.72-0.98). Non-whites, compared to Whites, had a higher total charge (1.21, 95% CI 1.13-1.29), higher coagulopathies (aOR 1.25; 95% CI 1.01-1.54), but lower cardiac arrhythmia risk (aOR 0.57; 95% CI 0.47-0.69). CONCLUSION Although IT has become increasingly important in treating mRCC, this is the first time real-world data on the costs, negative consequences, and disparities of IT are examined. The results may have important implications for creating innovative, supportive care models for this population.
Collapse
Affiliation(s)
- Susan Eichhorn
- Boston Medical Center, Boston University School of Medicine, Boston, MA, 02118, USA
| | - Roberto Pili
- Department of Medicine, Division of Hematology and Oncology, University at Buffalo, Buffalo, NY, 14203, USA
| | - Joel B Epstein
- City of Hope Comprehensive Cancer Center, Duarte CA and Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical System, Los Angeles, CA, USA
| | - Richa Rajesh
- Nova Southeastern University, Fort Lauderdale, FL, 33328, USA
| | - Poolakkad S Satheeshkumar
- Department of Medicine, Division of Hematology and Oncology, University at Buffalo, Buffalo, NY, 14203, USA.
| |
Collapse
|
21
|
Tascón J, Casanova AG, Vicente-Vicente L, López-Hernández FJ, Morales AI. Nephrotoxicity of Immune Checkpoint Inhibitors in Single and Combination Therapy-A Systematic and Critical Review. Biomedicines 2025; 13:711. [PMID: 40149687 PMCID: PMC11940260 DOI: 10.3390/biomedicines13030711] [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: 01/08/2025] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Immune checkpoint inhibitors (ICIs) have generated a revolutionary approach in the treatment of cancer, but their effectiveness has been compromised by immune-related adverse events, including renal damage. Although rare, these effects are relevant because they have been related to poor patient prognoses. The objective of this review was to estimate the current incidence of nephrotoxicity in patients treated with single and double ICI therapies. Methods: A total of 1283 potential articles were identified, which were reduced to 50 after applying the exclusion and inclusion criteria. Results: This study reveals the increase in acute kidney injury associated with these drugs in the last decade and shows that, interestingly, combined therapies with ICIs does not lead to an increase in kidney damage compared with anti-CTLA-4. It also suggests that kidney damage could be underdiagnosed when it comes to interstitial nephritis, because definitive evidence requires a renal biopsy. Conclusions: In perspective, these conclusions could guide clinicians in making decisions for therapy personalization and highlight the need to search for new diagnostic systems that are more sensitive and specific to the type of damage and could replace the biopsy.
Collapse
Affiliation(s)
- Javier Tascón
- Toxicology Unit, Universidad de Salamanca (USAL), 37007 Salamanca, Spain; (J.T.); (A.G.C.); (L.V.-V.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), 37007 Salamanca, Spain
| | - Alfredo G. Casanova
- Toxicology Unit, Universidad de Salamanca (USAL), 37007 Salamanca, Spain; (J.T.); (A.G.C.); (L.V.-V.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), 37007 Salamanca, Spain
| | - Laura Vicente-Vicente
- Toxicology Unit, Universidad de Salamanca (USAL), 37007 Salamanca, Spain; (J.T.); (A.G.C.); (L.V.-V.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), 37007 Salamanca, Spain
| | - Francisco J. López-Hernández
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), 37007 Salamanca, Spain
- Group of Biomedical Research on Critical Care (BioCritic), 47005 Valladolid, Spain
| | - Ana I. Morales
- Toxicology Unit, Universidad de Salamanca (USAL), 37007 Salamanca, Spain; (J.T.); (A.G.C.); (L.V.-V.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), 37007 Salamanca, Spain
- Group of Biomedical Research on Critical Care (BioCritic), 47005 Valladolid, Spain
| |
Collapse
|
22
|
Qian Y, Yang D, Lin X, Shen C, Zhang J, Xu J, Zhao Y, Zhu L, Kong H, Zhang M, Zhu Y, Zhou C, He J. Dendrimer-Derived Mimics of Host Defense Peptides Selectively Disrupt Cancer Cell Membranes for Melanoma Therapy. Pharmaceutics 2025; 17:361. [PMID: 40143025 PMCID: PMC11946345 DOI: 10.3390/pharmaceutics17030361] [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: 01/27/2025] [Revised: 02/18/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Melanoma is one of the most common malignancies, posing a significant health threat to patients, particularly in advanced stages due to its high aggressiveness. Chemotherapy agents with biocompatibility and low susceptibility to induce resistance are required for systematic management. Methods: Dendrimer-derived mimics (DMs) of host defense peptides (HDPs), which were constructed by a dendrimer core and optimized ratios of the hydrophobic arm, were used to treat A375 cells and HaCaT cells as the control. Live/dead staining, flow cytometry, and scanning electron microscopy (SEM) were conducted to analyze the anticancer mechanism. Mice with subcutaneous tumors were used to test the antitumor activity and toxicity in vivo. Results: DMs exhibited enhanced activity against A375 cells with remarkable selectivity, which mimics the action of natural HDPs and can cause damage to cell membranes. DMs can effectively inhibit solid tumor growth with minimal systemic toxicity and no adverse effects on healthy tissues. Conclusion: All the findings highlight DMs as promising anticancer candidates with significant potential for systemic melanoma therapy.
Collapse
Affiliation(s)
- Yusheng Qian
- Translational Medical Center for Stem Cell Therapy, Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Q.); (D.Y.); (X.L.); (C.S.); (J.Z.); (H.K.); (M.Z.)
- School of Material Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Danjing Yang
- Translational Medical Center for Stem Cell Therapy, Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Q.); (D.Y.); (X.L.); (C.S.); (J.Z.); (H.K.); (M.Z.)
| | - Xiangyu Lin
- Translational Medical Center for Stem Cell Therapy, Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Q.); (D.Y.); (X.L.); (C.S.); (J.Z.); (H.K.); (M.Z.)
| | - Chenyun Shen
- Translational Medical Center for Stem Cell Therapy, Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Q.); (D.Y.); (X.L.); (C.S.); (J.Z.); (H.K.); (M.Z.)
| | - Jieping Zhang
- Translational Medical Center for Stem Cell Therapy, Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Q.); (D.Y.); (X.L.); (C.S.); (J.Z.); (H.K.); (M.Z.)
| | - Jin Xu
- Laboratory Animal Center of Tongji University, Tongji University, Shanghai 200092, China;
| | - Yan Zhao
- Experimental Teaching Center for Medicine and Life Science, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Z.); (L.Z.)
| | - Ling Zhu
- Experimental Teaching Center for Medicine and Life Science, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Z.); (L.Z.)
| | - Haoran Kong
- Translational Medical Center for Stem Cell Therapy, Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Q.); (D.Y.); (X.L.); (C.S.); (J.Z.); (H.K.); (M.Z.)
| | - Mingyu Zhang
- Translational Medical Center for Stem Cell Therapy, Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Q.); (D.Y.); (X.L.); (C.S.); (J.Z.); (H.K.); (M.Z.)
| | - Yueqian Zhu
- Department of Dermatology, The First Affiliated Hospital of Soochow University, Suzhou 215031, China;
| | - Chuncai Zhou
- School of Material Science and Engineering, Tongji University, 4800 Caoan Road, Shanghai 201804, China
| | - Jing He
- Translational Medical Center for Stem Cell Therapy, Department of Dermatology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200331, China; (Y.Q.); (D.Y.); (X.L.); (C.S.); (J.Z.); (H.K.); (M.Z.)
| |
Collapse
|
23
|
Shatila M, Eshaghi F, Cruz CC, Machado AP, Mathew A, Zhao D, Siddiqui BA, Thomas AS, Chari ST, Wang Y. Differential Disease Behavior of Immune-Mediated Colitis Among Different Types of Immune Checkpoint Inhibition. Target Oncol 2025; 20:339-347. [PMID: 40038186 DOI: 10.1007/s11523-025-01135-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/11/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Immune-checkpoint inhibitors (ICIs) enhance the immune response against cancer but can cause immune-related adverse events, with immune-mediated colitis (IMC) being among the most common. OBJECTIVE We investigated variations in gastrointestinal disease behavior and outcomes among patients receiving different ICI regimens. METHODS This retrospective chart review included patients who received ICIs and developed IMC. Groups were categorized by their last ICI regimen before IMC onset into either programmed cell death protein-1/ligand-1 monotherapy or cytotoxic T-lymphocyte antigen 4 (CTLA-4) monotherapy/combination immunotherapy. Demographic and IMC-related clinical information was collected. RESULTS There were 414 patients included in this study: 169 treated with programmed cell death protein-1/ligand-1 monotherapy and 245 treated with CTLA-4 mono/combination therapy. Patients treated with CTLA-4 therapy had an earlier onset of IMC (median 46 days vs 123 days, p < 0.001). They were more likely to present with fever (p = 0.02), abdominal pain (p = 0.049), or hematochezia (p < 0.001). They also had more severe colitis with 47.3% of patients in the CTLA-4 group presenting with grade ≥3 colitis versus 20.2% in the programmed cell death protein-1/ligand-1 group (p < 0.05). On endoscopy, CTLA-4 mono/combination therapy was associated with increased ulcerative findings (24.4 vs 8.4%, p = 0.002). On histology, the programmed cell death protein-1/ligand-1 group was more likely to have microscopic colitis (13.9 vs 5.8%, p < 0.045). CONCLUSIONS This study provides insight into the effect of ICI type on IMC disease course. Cytotoxic T-lymphocyte antigen 4 inhibition leads to an earlier and more severe IMC onset with distinct endoscopic and histologic features. Further research is needed to refine treatment algorithms and identify the mechanisms underlying the variability in IMC presentation among different ICI regimens.
Collapse
Affiliation(s)
- Malek Shatila
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1466, Houston, TX, 77030, USA
| | - Farzin Eshaghi
- Department of Internal Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Carolina Colli Cruz
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1466, Houston, TX, 77030, USA
| | - Antonio Pizuorno Machado
- Department of Internal Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Antony Mathew
- Department of Internal Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Dan Zhao
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bilal A Siddiqui
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anusha Shirwaikar Thomas
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1466, Houston, TX, 77030, USA
| | - Suresh T Chari
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1466, Houston, TX, 77030, USA
| | - Yinghong Wang
- Department of Gastroenterology, Hepatology and Nutrition, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1466, Houston, TX, 77030, USA.
| |
Collapse
|
24
|
Sheykhhasan M, Ahmadieh-Yazdi A, Heidari R, Chamanara M, Akbari M, Poondla N, Yang P, Malih S, Manoochehri H, Tanzadehpanah H, Mahaki H, Fayazi Hosseini N, Dirbaziyan A, Al-Musawi S, Kalhor N. Revolutionizing cancer treatment: The power of dendritic cell-based vaccines in immunotherapy. Biomed Pharmacother 2025; 184:117858. [PMID: 39955851 DOI: 10.1016/j.biopha.2025.117858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 02/18/2025] Open
Abstract
In the modern time, cancer immunotherapies have increasingly become vital treatment options, joining long-established methods like surgery, chemotherapy, and radiotherapy treatment. Central to this emerging approach are dendritic cells (DCs), which boast a remarkable ability for antigen presentation. This ability is being leveraged to modulate T and B cell immunity, offering a groundbreaking strategy for tackling cancer. However, the percentage of patients experiencing meaningful benefits from this treatment remains relatively low, underscoring the ongoing necessity for further research and development in this field. This review offers a comprehensive analysis of the present-day progress in dendritic cell (DC)-based vaccines and recent efforts to enhance their efficacy. We explore the intricacies of DC function, from antigen capture to T cell stimulation, and discuss the outcomes of both preclinical and clinical trials across various cancer types. While the results are promising, the real-world application of DC-based vaccines is still nascent, posing multiple challenges that need to be overcome. These obstacles include optimizing the methods for DC generation and antigen loading, overcoming the immunosuppressive nature of the tumor microenvironment, and enhancing specificities of the immunologic response through personalized vaccines. The review concludes by emphasizing prospective opportunities for future research and emphasizing the critical need for extensive clinical trials. These trials are essential to validate the effectivity of DC-based vaccines and solidify their role in the broader spectrum of cancer immunotherapy options.
Collapse
Affiliation(s)
- Mohsen Sheykhhasan
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran.
| | - Amirhossein Ahmadieh-Yazdi
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Reza Heidari
- Infectious Diseases Research Center, AJA University of Medical Sciences, Tehran, Iran; Cancer Epidemiology Research Center, AJA University of Medical Sciences, Tehran, Iran; Medical Biotechnology Research Center, AJA University of Medical Sciences, Tehran, Iran
| | - Mohsen Chamanara
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran; Student research committee, AJA University of Medical Sciences, Tehran, Iran
| | - Mohammad Akbari
- Department of Medical School, Faculty of Medical Sciences, Islamic Azad University, Tonekabon Branch, Mazandaran, Iran
| | - Naresh Poondla
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Center for Global Health Research, Saveetha Medical College & Hospital, Chennai, India
| | - Piao Yang
- Department of Molecular Genetics, College of Arts and Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Sara Malih
- Department of Radiology, University of Wisconsin-Madison, Madison, WI, USA; Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hamed Manoochehri
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Hamid Tanzadehpanah
- Antimicrobial Resistance Research Center, Basic Science Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hanie Mahaki
- Vascular & Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Nashmin Fayazi Hosseini
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Ashkan Dirbaziyan
- Department of Microbiology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | | | - Naser Kalhor
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom, Iran
| |
Collapse
|
25
|
Wang TW, Nakanishi M. Immune surveillance of senescence: potential application to age-related diseases. Trends Cell Biol 2025; 35:248-257. [PMID: 39025762 DOI: 10.1016/j.tcb.2024.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/20/2024]
Abstract
Several lines of evidence suggest that the age-dependent accumulation of senescent cells leads to chronic tissue microinflammation, which in turn contributes to age-related pathologies. In general, senescent cells can be eliminated by the host's innate and adaptive immune surveillance system, including macrophages, NK cells, and T cells. Impaired immune surveillance leads to the accumulation of senescent cells and accelerates the aging process. Recently, senescent cells, like cancer cells, have been shown to express certain types of immune checkpoint proteins as well as non-classical immune-tolerant MHC variants, leading to immune escape from surveillance systems. Thus, immune checkpoint blockade (ICB) may be a promising strategy to enhance immune surveillance of senescence, leading to the amelioration of some age-related diseases and tissue dysfunction.
Collapse
Affiliation(s)
- Teh-Wei Wang
- Division of Cancer Cell Biology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Makoto Nakanishi
- Division of Cancer Cell Biology, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| |
Collapse
|
26
|
Westermann R, Cordtz RL, Duch K, Mellemkjaer L, Hetland ML, Magnussen B, Dreyer L. Cancer risk with tocilizumab/sarilumab, abatacept and rituximab treatment in patients with rheumatoid arthritis: a Danish cohort study. Rheumatology (Oxford) 2025; 64:1019-1028. [PMID: 38452297 DOI: 10.1093/rheumatology/keae140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/09/2024] [Accepted: 02/07/2024] [Indexed: 03/09/2024] Open
Abstract
OBJECTIVES To investigate cancer risk in RA patients treated with tocilizumab/sarilumab, abatacept or rituximab compared with those who received TNF inhibitors (TNFi) and compared with biological DMARDs (bDMARD)-naïve RA patients. METHODS Nationwide registry-based cohort study of RA patients who initiated bDMARD treatment with tocilizumab/sarilumab, abatacept, rituximab, and TNFi, as well as bDMARD-naive patients who initiated their second type of conventional synthetic DMARD. Patients were identified in the Danish Rheumatology Quality Register (DANBIO) and followed for cancer from 2006 to 2020. Patients could contribute multiple treatments, with person years, deaths and cancers allocated to each treatment group in a 'latest type of treatment' manner. Inverse probability of treatment weighting and weighted cause-specific Cox models were used to calculate hazard ratios (HRs) for cancer in each tocilizumab/sarilumab, abatacept and rituximab group compared with TNFi-treated and bDMARD-naïve groups, respectively. RESULTS In total, 21 982 treatment initiations, 96 475 person years and 1423 cancers were identified. There were no statistically significant increased HRs for overall cancer in tocilizumab/sarilumab, abatacept or rituximab treatment groups (HRs ranged from 0.7 to 1.1). More than 5 years of abatacept exposure showed a non-significantly increased HR compared with TNFi (HR 1.41, 95% CI 0.74-2.71). For haematological cancers, rituximab treatment showed non-significantly reduced HRs: vs TNFi-treated (HR 0.09; 95% CI 0.00-2.06) and bDMARD-naïve (HR 0.13; 95% CI 0.00-1.89). CONCLUSION Treatment with tocilizumab/sarilumab, abatacept or rituximab in RA patients was not associated with increased risks of cancer compared with TNFi-treated and with bDMARD-naïve RA patients in a real-world setting.
Collapse
Affiliation(s)
- Rasmus Westermann
- Center of Rheumatic Research Aalborg (CERRA), Department of Rheumatology, Aalborg University Hospital, Aalborg University, Aalborg, Denmark
- Clinical Cancer Research Unit, Aalborg University Hospital, Aalborg, Denmark
| | - René Lindholm Cordtz
- Center of Rheumatic Research Aalborg (CERRA), Department of Rheumatology, Aalborg University Hospital, Aalborg University, Aalborg, Denmark
| | - Kirsten Duch
- Center of Rheumatic Research Aalborg (CERRA), Department of Rheumatology, Aalborg University Hospital, Aalborg University, Aalborg, Denmark
- Research Data and Biostatistics, Aalborg University Hospital, Aalborg, Denmark
| | | | - Merete Lund Hetland
- The DANBIO Registry, Center for Rheumatology and Spine Diseases, Centre of Head and Orthopedics, Rigshospitalet, Glostrup, Denmark
- Department of Clinical Medicine, University of Copenhagen Faculty of Health and Medical Sciences, Copenhagen, Denmark
- Copenhagen Center for Arthritis Research (COPECARE), Center for Rheumatology and Spine Diseases, Centre of Head and Orthopedics, Rigshospitalet, Glostrup, Denmark
| | - Bergur Magnussen
- Center of Rheumatic Research Aalborg (CERRA), Department of Rheumatology, Aalborg University Hospital, Aalborg University, Aalborg, Denmark
| | - Lene Dreyer
- Center of Rheumatic Research Aalborg (CERRA), Department of Rheumatology, Aalborg University Hospital, Aalborg University, Aalborg, Denmark
| |
Collapse
|
27
|
Bloom M, Podder S, Dang H, Lin D. Advances in Immunotherapy in Hepatocellular Carcinoma. Int J Mol Sci 2025; 26:1936. [PMID: 40076561 PMCID: PMC11900920 DOI: 10.3390/ijms26051936] [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: 12/20/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Over the past several years, the therapeutic landscape for patients with advanced, unresectable, or metastatic hepatocellular carcinoma has been transformed by the incorporation of checkpoint inhibitor immunotherapy into the treatment paradigm. Frontline systemic treatment options have expanded beyond anti-angiogenic tyrosine kinase inhibitors, such as sorafenib, to a combination of immunotherapy approaches, including atezolizumab plus bevacizumab and durvalumab plus tremelimumab, both of which have demonstrated superior response and survival to sorafenib. Additionally, combination treatments with checkpoint inhibitors and tyrosine kinase inhibitors have been investigated with variable success. In this review, we discuss these advances in systemic treatment with immunotherapy, with a focus on understanding both the underlying biology and mechanism of these strategies and their efficacy outcomes in clinical trials. We also review challenges in identifying predictive biomarkers of treatments and discuss future directions with novel immunotherapy targets.
Collapse
Affiliation(s)
- Matthew Bloom
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA;
| | - Sourav Podder
- Department of Surgery, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA; (S.P.); (H.D.)
| | - Hien Dang
- Department of Surgery, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA; (S.P.); (H.D.)
| | - Daniel Lin
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA;
| |
Collapse
|
28
|
Ezdoglian A, Tsang-A-Sjoe M, Khodadust F, Burchell G, Jansen G, de Gruijl T, Labots M, van der Laken CJ. Monocyte-related markers as predictors of immune checkpoint inhibitor efficacy and immune-related adverse events: a systematic review and meta-analysis. Cancer Metastasis Rev 2025; 44:35. [PMID: 39982537 PMCID: PMC11845441 DOI: 10.1007/s10555-025-10246-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/22/2025] [Indexed: 02/22/2025]
Abstract
The efficacy and off-target effects of immune checkpoint inhibitors (ICI) in cancer treatment vary among patients. Monocytes likely contribute to this heterogeneous response due to their crucial role in immune homeostasis. We conducted a systematic review and meta-analysis to evaluate the impact of monocytes on ICI efficacy and immune-related adverse events (irAEs) in patients with cancer. We systematically searched PubMed, Web of Science, and Embase for clinical studies from January 2000 to December 2023. Articles were included if they mentioned cancer, ICI, monocytes, or any monocyte-related terminology. Animal studies and studies where ICIs were combined with other biologics were excluded, except for studies where two ICIs were used. This systematic review was registered with PROSPERO (CRD42023396297) prior to data extraction and analysis. Monocyte-related markers, such as absolute monocyte count (AMC), monocyte/lymphocyte ratio (MLR), specific monocyte subpopulations, and m-MDSCs were assessed in relation to ICI efficacy and safety. Bayesian meta-analysis was conducted for AMC and MLR. The risk of bias assessment was done using the Cochrane-ROBINS-I tool. Out of 5787 studies identified in our search, 155 eligible studies report peripheral blood monocyte-related markers as predictors of response to ICI, and 32 of these studies describe irAEs. Overall, based on 63 studies, a high MLR was a prognostic biomarker for short progression-free survival (PFS) and overall survival (OS) hazard ratio (HR): 1.5 (95% CI: 1.21-1.88) and 1.52 (95% CI:1.13-2.08), respectively. The increased percentage of classical monocytes was an unfavorable predictor of survival, while low baseline rates of monocytic myeloid-derived suppressor cells (m-MDSCs) were favorable. Elevated intermediate monocyte frequencies were associated but not significantly correlated with the development of irAEs. Baseline monocyte phenotyping may serve as a composite biomarker of response to ICI; however, more data is needed regarding irAEs. Monocyte-related variables may aid in risk assessment and treatment decision strategies for patients receiving ICI in terms of both efficacy and safety.
Collapse
Affiliation(s)
- Aiarpi Ezdoglian
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Michel Tsang-A-Sjoe
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Fatemeh Khodadust
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - George Burchell
- Amsterdam University Medical Library, Amsterdam, The Netherlands
| | - Gerrit Jansen
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Tanja de Gruijl
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam University Medical Center, Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Mariette Labots
- Department of Medical Oncology, Amsterdam University Medical Center, Location Vrije Universiteit, Amsterdam, The Netherlands
| | - Conny J van der Laken
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
29
|
Fretwell EC, Houldsworth A. Oncolytic Virus Therapy in a New Era of Immunotherapy, Enhanced by Combination with Existing Anticancer Therapies: Turn up the Heat! J Cancer 2025; 16:1782-1793. [PMID: 40092697 PMCID: PMC11905400 DOI: 10.7150/jca.102285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 01/20/2025] [Indexed: 03/19/2025] Open
Abstract
Oncolytic viral therapy is a promising treatment for cancer, where 'cold' tumour cells can become 'hot' to the host immune system. However, with few FDA approved therapies, development of new strategies for more cancer types has been slow and relatively unsuccessful in recent years, Combination therapy has been successful for other types of cancer treatment, therefore, may be a viable alternative to improve the efficacy of oncolytic viral therapy which may reduce some of the adverse events of currently used monotherapies, oncolytic virus therapy and chemotherapy being mutually complimentary with each other. Combining oncolytic viruses with immune checkpoint inhibitors provides a significant increase in efficacy when viral therapy was combined with the drug ipilimumab. Phase I and II studies concluded that combination with chemotherapies was safe and effective but did not significantly improve on current monotherapies. Recent experiments suggest that a combination of CAR-T and CAR--M cells is a promising therapeutic approach but needs to advance to clinical testing to observe the human response to the therapy. Viral combination with ipilimumab showed the highest potential for a successful treatment and clinical trials should be advanced to phase III to find conclusive supporting evidence. This review aims to identify and evaluate the potential of currently evolving oncolytic viral therapy with recent advances in genetic engineering providing enhanced oncolytic activity in the tumour, and addressing the lack of host immune responses in 'cold' tumours, with an additional role in enhancing conventional treatment efficacy with combination therapies. The potential of oncolytic viruses to 'turn up the heat' of a tumour microenvironment immunogenicity in combination with other anticancer treatments, provides a promising future for new cancer therapies.
Collapse
Affiliation(s)
| | - Annwyne Houldsworth
- University of Exeter Medical School, Faculty of Health and Life Sciences, Exeter, EX2 4TH, UK
| |
Collapse
|
30
|
Yildirim A, Wei M, Liu Y, Nazha B, Brown JT, Carthon BC, Choi Y, Suh L, Goswamy RV, McClintock GR, Hartman C, Caulfield S, Ciuro J, Goldman JM, Harris WB, Kucuk O, Master VA, Bilen MA. Association of baseline inflammatory biomarkers and clinical outcomes in patients with advanced renal cell carcinoma treated with immune checkpoint inhibitors. Ther Adv Med Oncol 2025; 17:17588359251316243. [PMID: 39943944 PMCID: PMC11815817 DOI: 10.1177/17588359251316243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 01/10/2025] [Indexed: 02/17/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have become the mainstay treatment of metastatic kidney cancer, demonstrating enhanced outcomes and durable responses in select patient subgroups. However, identifying reliable prognostic biomarkers for treatment outcomes remains challenging. Objectives This study aimed to assess the correlation between baseline inflammatory markers and overall survival (OS), progression-free survival (PFS), and clinical benefit (CB) in metastatic kidney cancer patients receiving ICIs. CB was defined as patients achieving stable disease, partial response, or complete response. Design Retrospective, single-center study. Methods A retrospective analysis was conducted on 401 adult patients with advanced kidney cancer treated with ICIs at Emory Winship Cancer Institute between 2018 and 2023. Modified Glasgow Prognostic Score (mGPS), neutrophil-to-lymphocyte (NLR), monocyte-to-lymphocyte (MLR), platelet-to-lymphocyte (PLR), and neutrophil-to-eosinophil ratios (NER) were collected from baseline blood samples. Results Among 401 patients (median age, 66; 71% male; 21% Black/African American), median follow-up was 43.0 months (95% CI, 36.6-51.4). Patients with mGPS scores of 0 had longer OS than those with a score of 1 (hazard ratio (HR), 0.38; 95% CI, 0.23-0.62; p < 0.001) and 2 (HR, 0.37; 95% CI, 0.20-0.67; p = 0.001), and longer PFS compared to patients with mGPS scores of 1 (HR, 0.66; 95% CI, 0.44-0.98; p = 0.039) and 2 (HR, 0.44; 95% CI, 0.29-0.67; p < 0.001). Low baseline NLR was associated with longer PFS (HR, 0.73; 95% CI, 0.54-0.97; p = 0.032). Low baseline MLR correlated with improved OS (HR, 0.60; 95% CI, 0.44-0.83; p = 0.002) and PFS (HR, 0.73; 95% CI, 0.55-0.97; p = 0.031). Similarly, low baseline PLR was associated with higher CB likelihood (odds ratio (OR), 2.20; 95% CI, 1.31-3.69; p = 0.003), and low baseline NER was linked to improved OS (HR, 0.63; 95% CI, 0.46-0.87; p = 0.004), PFS (HR, 0.67; 95% CI, 0.51-0.88; p = 0.003), and higher CB (OR, 2.04; 95% CI, 1.20-3.46; p = 0.008). Conclusion Lower levels of systemic inflammatory markers are associated with more favorable clinical outcomes with ICI treatment. Prospective studies are needed for further validation.
Collapse
Affiliation(s)
- Ahmet Yildirim
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Yuan Liu
- Departments of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | - Bassel Nazha
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Jacqueline T. Brown
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Bradley C. Carthon
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Yujin Choi
- University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lauren Suh
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Rohit V. Goswamy
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Greta R. McClintock
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Caitlin Hartman
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Sarah Caulfield
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Department of Pharmaceutical Services, Emory University School of Medicine, Atlanta, GA, USA
| | - Jordan Ciuro
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Jamie M. Goldman
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Wayne B. Harris
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Omer Kucuk
- Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| | - Viraj A. Master
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
- Department of Urology, Emory University School of Medicine, Atlanta, GA, USA
| | - Mehmet A. Bilen
- Genitourinary Medical Oncology Program, Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, 1365B Clifton Road NE, Suite B4000, Office 4212, Atlanta, GA 30322, USA
- Winship Cancer Institute of Emory University, Atlanta, GA, USA
| |
Collapse
|
31
|
Nedeljković M, Vuletić A, Mirjačić Martinović K. Divide and Conquer-Targeted Therapy for Triple-Negative Breast Cancer. Int J Mol Sci 2025; 26:1396. [PMID: 40003864 PMCID: PMC11855393 DOI: 10.3390/ijms26041396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and malignant type of breast cancer with limited treatment options and poor prognosis. One of the most significant impediments in TNBC treatment is the high heterogeneity of this disease, as highlighted by the detection of several molecular subtypes of TNBC. Each subtype is driven by distinct mutations and pathway aberrations, giving rise to specific molecular characteristics closely connected to clinical behavior, outcomes, and drug sensitivity. This review summarizes the knowledge regarding TNBC molecular subtypes and how it can be harnessed to devise tailored treatment strategies instead of blindly using targeted drugs. We provide an overview of novel targeted agents and key insights about new treatment modalities with an emphasis on the androgen receptor signaling pathway, cancer stem cell-associated pathways, phosphatidylinositol 3-kinase (PI3K)/AKT pathway, growth factor signaling, and immunotherapy.
Collapse
Affiliation(s)
- Milica Nedeljković
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (A.V.); (K.M.M.)
| | | | | |
Collapse
|
32
|
Brauer J, Tumani M, Frey N, Lehmann LH. The cardio-oncologic burden of breast cancer: molecular mechanisms and importance of preclinical models. Basic Res Cardiol 2025; 120:91-112. [PMID: 39621070 PMCID: PMC11790711 DOI: 10.1007/s00395-024-01090-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 02/04/2025]
Abstract
Breast cancer, the most prevalent cancer affecting women worldwide, poses a significant cardio-oncological burden. Despite advancements in novel therapeutic strategies, anthracyclines, HER2 antagonists, and radiation remain the cornerstones of oncological treatment. However, each carries a risk of cardiotoxicity, though the molecular mechanisms underlying these adverse effects differ. Common mechanisms include DNA damage response, increased reactive oxygen species, and mitochondrial dysfunction, which are key areas of ongoing research for potential cardioprotective strategies. Since these mechanisms are also essential for effective tumor cytotoxicity, we explore tumor-specific effects, particularly in hereditary breast cancer linked to BRCA1 and BRCA2 mutations. These genetic variants impair DNA repair mechanisms, increase the risk of tumorigenesis and possibly for cardiotoxicity from treatments such as anthracyclines and HER2 antagonists. Novel therapies, including immune checkpoint inhibitors, are used in the clinic for triple-negative breast cancer and improve the oncological outcomes of breast cancer patients. This review discusses the molecular mechanisms underlying BRCA dysfunction and the associated pathological pathways. It gives an overview of preclinical models of breast cancer, such as genetically engineered mouse models, syngeneic murine models, humanized mouse models, and various in vitro and ex vivo systems and models to study cardiovascular side effects of breast cancer therapies. Understanding the underlying mechanism of cardiotoxicity and developing cardioprotective strategies in preclinical models are essential for improving treatment outcomes and reducing long-term cardiovascular risks in breast cancer patients.
Collapse
Affiliation(s)
- J Brauer
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany
| | - M Tumani
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany
| | - N Frey
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany
| | - L H Lehmann
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
33
|
Ferreira N, Kulkarni A, Agorku D, Midelashvili T, Hardt O, Legler TJ, Ströbel P, Conradi LC, Alves F, Ramos-Gomes F, Markus MA. OrganoIDNet: a deep learning tool for identification of therapeutic effects in PDAC organoid-PBMC co-cultures from time-resolved imaging data. Cell Oncol (Dordr) 2025; 48:101-122. [PMID: 38805131 PMCID: PMC11850476 DOI: 10.1007/s13402-024-00958-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2024] [Indexed: 05/29/2024] Open
Abstract
PURPOSE Pancreatic Ductal Adenocarcinoma (PDAC) remains a challenging disease due to its complex biology and aggressive behavior with an urgent need for efficient therapeutic strategies. To assess therapy response, pre-clinical PDAC organoid-based models in combination with accurate real-time monitoring are required. METHODS We established stable live-imaging organoid/peripheral blood mononuclear cells (PBMCs) co-cultures and introduced OrganoIDNet, a deep-learning-based algorithm, capable of analyzing bright-field images of murine and human patient-derived PDAC organoids acquired with live-cell imaging. We investigated the response to the chemotherapy gemcitabine in PDAC organoids and the PD-L1 inhibitor Atezolizumab, cultured with or without HLA-matched PBMCs over time. Results obtained with OrganoIDNet were validated with the endpoint proliferation assay CellTiter-Glo. RESULTS Live cell imaging in combination with OrganoIDNet accurately detected size-specific drug responses of organoids to gemcitabine over time, showing that large organoids were more prone to cytotoxic effects. This approach also allowed distinguishing between healthy and unhealthy status and measuring eccentricity as organoids' reaction to therapy. Furthermore, imaging of a new organoids/PBMCs sandwich-based co-culture enabled longitudinal analysis of organoid responses to Atezolizumab, showing an increased potency of PBMCs tumor-killing in an organoid-individual manner when Atezolizumab was added. CONCLUSION Optimized PDAC organoid imaging analyzed by OrganoIDNet represents a platform capable of accurately detecting organoid responses to standard PDAC chemotherapy over time. Moreover, organoid/immune cell co-cultures allow monitoring of organoid responses to immunotherapy, offering dynamic insights into treatment behavior within a co-culture setting with PBMCs. This setup holds promise for real-time assessment of immunotherapeutic effects in individual patient-derived PDAC organoids.
Collapse
Affiliation(s)
- Nathalia Ferreira
- Translational Molecular Imaging, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Ajinkya Kulkarni
- Translational Molecular Imaging, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - David Agorku
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Teona Midelashvili
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075, Göttingen, Germany
| | - Olaf Hardt
- Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany
| | - Tobias J Legler
- Department of Transfusion Medicine, University Medical Center Göttingen, Göttingen, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Lena-Christin Conradi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075, Göttingen, Germany
| | - Frauke Alves
- Translational Molecular Imaging, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
- Clinic of Hematology and Medical Oncology, Department of Diagnostic and Interventional Radiology, University Medical Center Göttingen, Göttingen, Germany
| | - Fernanda Ramos-Gomes
- Translational Molecular Imaging, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - M Andrea Markus
- Translational Molecular Imaging, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany.
| |
Collapse
|
34
|
de la Torre-Montero JC, Serra-López J, Alvárez-García R, Battle-Vidal M, Gil-Gómez N, Beorlegui-Murillo P, Pérez-Cárdenas D, Sánchez-Belchiz A, Cruz EM, Peñuelas-Saiz Á. Nutritional Status in Locally Advanced or Metastatic Solid Cancer Patients Treated With Chemotherapy, Radiotherapy, and Immunotherapy in Spanish Outpatient Oncology Units. Semin Oncol Nurs 2025; 41:151801. [PMID: 39794238 DOI: 10.1016/j.soncn.2024.151801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 11/27/2024] [Accepted: 12/09/2024] [Indexed: 01/13/2025]
Abstract
OBJECTIVES Malnutrition is a prevalent condition in cancer patients that significantly impacts patients' clinical outcomes and health-related quality of life (HR-QoL). The outcome was to characterize the nutritional status by describing the prevalence of malnutrition (mild, moderate, or severe) and its risk in outpatient cancer patients. METHODS Multicenter, prospective, cross-sectional, descriptive, two-cohort study conducted on consecutive adult patients with locally advanced or metastatic solid tumors (stages III-IV). The study was conducted in 10 Spanish hospitals distributed all over the Spanish geography, with a recruitment period of 5 months (between April and September 2020). Study patients were divided into two groups according to their cancer therapy: group A, patients who underwent immunotherapy, and group B, patients who received combined therapy (immunotherapy plus chemotherapy and radiotherapy). RESULTS A total of 585 patients were included. The proportion of patients at risk of malnutrition was notably more significant in the combination group (chemotherapy and/or radiotherapy) than in the immunotherapy-only group (28.3% versus 58.5%, respectively, P < .0001). According to this evaluation the highest proportion of patients at risk were those with pancreatic cancer (51 patients; 89.5%), followed by large intestine cancer (52 patients; 55.3%) and lung cancer (56 patients; 29.3%), P < .0001. CONCLUSIONS Patients treated with only immunotherapy seemed to have better nutritional status, which indicated health-related quality of life improvement. Additionally, there was a trend associating nutritional status with tumor location. Treatment strategy, treatment duration, performance status, and treatment location were independently associated with malnutrition. IMPLICATIONS FOR NURSING PRACTICE Integrating nutritional assessment into routine clinical practice will improve the quality of life of oncology patients. An integrative approach to health improves overall results in terms of nutritional status and improved quality of life and shows that daily living activities are more satisfactory for patients with nursing interventions. Nursing interventions are consistent with an educational approach to patients as long as the interventions described in international guidelines are detailed in the framework of the patient care.
Collapse
Affiliation(s)
- Julio C de la Torre-Montero
- Director of the Health Sciences Department, Spanish Society of Oncology Nursing; Universidad Pontificia Comillas; and Fundación San Juan de Dios, Madrid, Spain.
| | - Jorgina Serra-López
- Medical Oncology Department, Spanish Society of Oncology Nursing, Madrid; and Hospital Santa Creu i Sant Pau, Barcelona, Spain
| | - Raquel Alvárez-García
- Medical Oncology Department, Spanish Society of Oncology Nursing, Madrid; and Hospital Vall d'Hebrón, Barcelona, Spain
| | - Magdalena Battle-Vidal
- Medical Oncology Department, Spanish Society of Oncology Nursing, Madrid; and Son Espases University Hospital, Palma de Mallorca, Spain
| | - Nieves Gil-Gómez
- Medical Oncology Department, Spanish Society of Oncology Nursing; and Hospital Universitario Quirón Pozuelo, Madrid, Spain
| | - Patricia Beorlegui-Murillo
- Medical Oncology Department, Spanish Society of Oncology Nursing, Madrid; and Clínica Universitaria de Navarra, Navarra, Spain
| | - Dolores Pérez-Cárdenas
- Medical Oncology Department, Spanish Society of Oncology Nursing; and 12 de Octubre University Hospital, Madrid, Spain
| | - Andrés Sánchez-Belchiz
- Medical Oncology Department, Spanish Society of Oncology Nursing, Madrid; and Virgen del Rocío University Hospital, Sevilla, Spain
| | - Erik Medina Cruz
- Medical Oncology Department, Spanish Society of Oncology Nursing, Madrid; and Hospital Universitario de Canarias, Palmas de Gran Canaria, Spain
| | - Ángeles Peñuelas-Saiz
- Medical Oncology Department, Spanish Society of Oncology Nursing, Madrid; and Hospital Vall d'Hebrón, Barcelona, Spain
| |
Collapse
|
35
|
Das L, Das S. A comprehensive insights of cancer immunotherapy resistance. Med Oncol 2025; 42:57. [PMID: 39883235 DOI: 10.1007/s12032-025-02605-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025]
Abstract
Cancer is a major global health issue that is usually treated with multiple therapies, such as chemotherapy and targeted therapies like immunotherapy. Immunotherapy is a new and alternative approach to treating various types of cancer that are difficult to treat with other methods. Although immune checkpoint inhibitors have shown promise for long-term efficacy, they have limited effectiveness in common cancer types such as breast, prostate, and lung. Some patients do not respond to immunotherapy, while others develop resistance to the treatment over time, which is classified as primary or acquired resistance. Cancer immunotherapy, specifically immune checkpoint inhibitor-based resistance involves multiple factors such as genes, metabolism, inflammation, and angiogenesis. However, cutting-edge research has identified the mechanisms of immunotherapy resistance and possible solutions. Current research may improve biomarker identification and modify treatment strategies, which will lead to better clinical outcomes. This review provides a comprehensive discussion of the current mechanisms of immunotherapy resistance, related biomarker modulation, and strategies to overcome resistance.
Collapse
Affiliation(s)
- Laavanya Das
- Department of Food and Nutrition, Brainware University, 398, Ramkrishnapur Rd, Barasat, Kolkata, West Bengal, 700125, India
| | - Subhadip Das
- Department of In Vivo Pharmacology, TCG Lifesciences Pvt. Ltd, BN 7, Sector V, Salt Lake City, Kolkata, West Bengal, 700091, India.
| |
Collapse
|
36
|
Hushmandi K, Einollahi B, Lee EHC, Sakaizawa R, Glaviano A, Reiter RJ, Saadat SH, Farani MR, Huh YS, Aref AR, Salimimoghadam S, Kumar AP. Bispecific antibodies as powerful immunotherapeutic agents for urological cancers: Recent innovations based on preclinical and clinical evidence. Int J Biol Sci 2025; 21:1410-1435. [PMID: 39990653 PMCID: PMC11844292 DOI: 10.7150/ijbs.96155] [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: 03/10/2024] [Accepted: 08/25/2024] [Indexed: 02/25/2025] Open
Abstract
Conventional immunotherapy has emerged as a key option for cancer treatment. However, its efficacy has been limited in urological cancers, especially prostate cancer, because of the immunosuppressive tumor microenvironment (TME), difficulty in drug delivery, aberrant immune response, and damage to normal cells. Bispecific antibodies (BsAbs) are engineered proteins with two different antigen-binding domains, designed using different technologies and in various formats. BsAb-based tumor immunotherapy has yielded optimistic results in preclinical and clinical investigations of many tumor types, including urological cancers. However, a series of challenges, including tumor heterogeneity, TME, Ab immunogenicity, adverse effects, serum half-life, low response rates, and drug resistance, hamper the application of BsAbs. In this review, we provide insights into the most common BsAb platforms with different mechanisms of action, which are under preclinical and clinical research, along with ways to overcome the challenges in BsAb administration for treating urological cancer.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Reo Sakaizawa
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| | - Russel J. Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, Texas USA
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Marzieh Ramezani Farani
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Incheon 22212, Republic of Korea
| | - Yun Suk Huh
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Incheon 22212, Republic of Korea
| | - Amir Reza Aref
- Department of Vitro Vision, DeepkinetiX Inc., Boston, MA, USA
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
37
|
Salimi Asl A, Davari M, Ghorbani A, Seddighi N, Arabi K, Saburi E. Neoadjuvant immunotherapy and oncolytic virotherapy in HPV positive and HPV negative skin cancer: A comprehensive review. Int Immunopharmacol 2025; 146:113790. [PMID: 39673996 DOI: 10.1016/j.intimp.2024.113790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Skin cancer is the most common new cancer among Caucasians. This cancer has different types, of which non-melanoma skin cancer is the most common type. Various factors affect this disease, one of which is viral infections, including HPV. This virus plays an important role in skin cancer, especially cSCCs. There are various options for the treatment of skin cancer, and today special attention has been paid to treatments based on therapeutic goals, immunotherapy and combination therapy. In this study, we have investigated treatments based on immunotherapy and virotherapy and the effect of HPV virus on the effectiveness of these treatments in skin cancer. Treatments based on virotherapy are performed for a long time in combination with other common treatments such as radiotherapy and chemotherapy in order to have a greater effect and lower its side effects, which include: shortness of breath, tachycardia, lowering blood pressure in the patient. Also, the most important axis of immunotherapy is to focus on PD1-PDL1, despite abundant evidence on the importance of immunotherapy, many studies investigate the use of immunotherapy inhibitors in the adjuvant and neoadjuvant setting in various cancers. Also, previous findings show conflicting evidence of the effect of HPV status on the response to immunotherapy.
Collapse
Affiliation(s)
- Ali Salimi Asl
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Mohsen Davari
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| | - Atousa Ghorbani
- Department of Biology, North Tehran Branch, Islamic Azad University, Tehran, Iran; Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Narjes Seddighi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Kimia Arabi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran.
| | - Ehsan Saburi
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics and Molecular Medicine Department, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
38
|
Miranda I, Jahan N, Shevde LA. The metastatic cascade through the lens of therapeutic inhibition. Cell Rep Med 2025; 6:101872. [PMID: 39706193 PMCID: PMC11866422 DOI: 10.1016/j.xcrm.2024.101872] [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: 06/19/2024] [Revised: 10/21/2024] [Accepted: 11/18/2024] [Indexed: 12/23/2024]
Abstract
Metastasis is a main cause of cancer-related death, and a deeper understanding of the metastatic process will inform more targeted and mechanistic approaches that can abrogate challenges in treatment efficacy and toxicity. Several steps throughout the metastatic cascade, from angiogenesis to secondary tumor formation, offer specific vulnerabilities to therapies that can lead to the decline or cessation of metastatic progression. A deeper understanding of the metastatic cascade also allows combination systemic therapies to be used synergistically. In this review, we describe current treatment modalities in the context of multiple steps of the metastatic cascade. We highlight their mechanisms and present their efficacy across multiple cancers. This work also presents targets within the metastatic cascade in need of more research that can advance the landscape of treatments and lead to the goal of metastatic cancer remission.
Collapse
Affiliation(s)
- Ian Miranda
- Medical Scientist Training Program, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nusrat Jahan
- Division of Hematology and Oncology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Lalita A Shevde
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
39
|
Rebeck ON, Wallace MJ, Prusa J, Ning J, Evbuomwan EM, Rengarajan S, Habimana-Griffin L, Kwak S, Zahrah D, Tung J, Liao J, Mahmud B, Fishbein SRS, Ramirez Tovar ES, Mehta R, Wang B, Gorelik MG, Helmink BA, Dantas G. A yeast-based oral therapeutic delivers immune checkpoint inhibitors to reduce intestinal tumor burden. Cell Chem Biol 2025; 32:98-110.e7. [PMID: 39571582 PMCID: PMC11741927 DOI: 10.1016/j.chembiol.2024.10.013] [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: 03/15/2024] [Revised: 08/09/2024] [Accepted: 10/28/2024] [Indexed: 12/13/2024]
Abstract
Engineered probiotics are an emerging platform for in situ delivery of therapeutics to the gut. Herein, we developed an orally administered, yeast-based therapeutic delivery system to deliver next-generation immune checkpoint inhibitor (ICI) proteins directly to gastrointestinal tumors. We engineered Saccharomyces cerevisiae var. boulardii (Sb), a probiotic yeast with high genetic tractability and innate anticancer activity, to secrete "miniature" antibody variants that target programmed death ligand 1 (Sb_haPD-1). When tested in an ICI-refractory colorectal cancer (CRC) mouse model, Sb_haPD-1 significantly reduced intestinal tumor burden and resulted in significant shifts to the immune cell profile and microbiome composition. This oral therapeutic platform is modular and highly customizable, opening new avenues of targeted drug delivery that can be applied to treat a myriad of gastrointestinal malignancies.
Collapse
Affiliation(s)
- Olivia N Rebeck
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Miranda J Wallace
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jerome Prusa
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jie Ning
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Esse M Evbuomwan
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Sunaina Rengarajan
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Dermatology, John T. Milliken Department of Internal Medicine, Washington University School of Medicine, St. Louis MO 63110, USA
| | - LeMoyne Habimana-Griffin
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Suryang Kwak
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David Zahrah
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jason Tung
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James Liao
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bejan Mahmud
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Skye R S Fishbein
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Erick S Ramirez Tovar
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rehan Mehta
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bin Wang
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mark G Gorelik
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Beth A Helmink
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gautam Dantas
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
40
|
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
|
41
|
Jeon S, Jeon Y, Lim JY, Kim Y, Cha B, Kim W. Emerging regulatory mechanisms and functions of biomolecular condensates: implications for therapeutic targets. Signal Transduct Target Ther 2025; 10:4. [PMID: 39757214 DOI: 10.1038/s41392-024-02070-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/01/2024] [Accepted: 11/06/2024] [Indexed: 01/07/2025] Open
Abstract
Cells orchestrate their processes through complex interactions, precisely organizing biomolecules in space and time. Recent discoveries have highlighted the crucial role of biomolecular condensates-membrane-less assemblies formed through the condensation of proteins, nucleic acids, and other molecules-in driving efficient and dynamic cellular processes. These condensates are integral to various physiological functions, such as gene expression and intracellular signal transduction, enabling rapid and finely tuned cellular responses. Their ability to regulate cellular signaling pathways is particularly significant, as it requires a careful balance between flexibility and precision. Disruption of this balance can lead to pathological conditions, including neurodegenerative diseases, cancer, and viral infections. Consequently, biomolecular condensates have emerged as promising therapeutic targets, with the potential to offer novel approaches to disease treatment. In this review, we present the recent insights into the regulatory mechanisms by which biomolecular condensates influence intracellular signaling pathways, their roles in health and disease, and potential strategies for modulating condensate dynamics as a therapeutic approach. Understanding these emerging principles may provide valuable directions for developing effective treatments targeting the aberrant behavior of biomolecular condensates in various diseases.
Collapse
Affiliation(s)
- Soyoung Jeon
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Yeram Jeon
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Ji-Youn Lim
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea
| | - Yujeong Kim
- Department of Life Science, University of Seoul, Seoul, South Korea
| | - Boksik Cha
- New Drug Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, South Korea.
| | - Wantae Kim
- Department of Life Science, University of Seoul, Seoul, South Korea.
| |
Collapse
|
42
|
Yang G, Tian L, Wang Y. Hyperprogressive disease induced by PD-1 inhibitor monotherapy in lung adenocarcinoma with HER2 exon 20 insertion: report of two cases and review of literature. Discov Oncol 2025; 16:12. [PMID: 39760792 PMCID: PMC11703795 DOI: 10.1007/s12672-025-01749-3] [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: 09/09/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025] Open
Abstract
Monotherapy with anti-programmed cell death protein 1 (PD-1) monoclonal antibody has been approved for the treatment of advanced non-small cell lung cancer with positive programmed cell death-ligand 1 (PD-L1) expression and oncogene wild type, which revealed survival benefit compared with chemotherapy. Nevertheless, certain patients develop rapid progression on anti-PD-1 inhibitor monotherapy. This novel pattern is called hyperprogressive disease (HPD), and the underlying mechanism and molecular characteristics still leaves not clear. Here, we reported two heavily pretreated advanced lung adenocarcinoma cases with HER2 exon 20 insertion who presented HPD after two cycles of anti-PD-1 inhibitor sintilimab monotherapy, and they both carried co-alterations in the PI3K/AKT/mTOR and cell cycle signaling pathway. We speculated that HER2 exon 20 insertion might be viewed as a potential biomarker to avoid single-agent immunotherapy in certain patients with driver mutations, or timely guide proper treatment strategies.
Collapse
Affiliation(s)
- Guangjian Yang
- Department of Respiratory Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People's Republic of China
| | - Linyan Tian
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China
| | - Yan Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People's Republic of China.
| |
Collapse
|
43
|
Ren H, He J, Dong J, Jiang G, Hao J, Han L. Specific BCG-related gene expression levels correlate with immune cell infiltration and prognosis in melanoma. J Leukoc Biol 2024; 117:qiae064. [PMID: 38478636 DOI: 10.1093/jleuko/qiae064] [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: 01/30/2024] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 01/01/2025] Open
Abstract
Melanoma, caused by malignant melanocytes, is known for its invasiveness and poor prognosis. Therapies are often ineffective due to their heterogeneity and resistance. Bacillus Calmette-Guérin (BCG), primarily a tuberculosis vaccine, shows potential in treating melanoma by activating immune responses. In this study, data from The Cancer Genome Atlas and the National Center for Biotechnology Information Gene Expression Omnibus database were utilized to determine pivotal DEGs such as DSC2, CXCR1, BOK, and CSTB, which are significantly upregulated in BCG-treated blood samples and are strongly associated with the prognosis of melanoma. We employ tools like edgeR and ggplot2 for functional and pathway analysis and develop a prognostic model using LASSO Cox regression analysis to predict patient survival. A notable finding is the correlation between BCG-related genes and immune cell infiltration in melanoma, highlighting the potential of these genes as both biomarkers and therapeutic targets. Additionally, the study examines genetic alterations in these genes and their impact on the disease. This study highlights the necessity of further exploring BCG-related genes for insights into melanoma pathogenesis and treatment enhancement, suggesting that BCG's role in immune activation could offer novel therapeutic avenues in cancer treatment.
Collapse
Affiliation(s)
- He Ren
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Rd, Harbin, 150081, Heilongjiang, China
| | - Jiacheng He
- College of Environment and Chemistry Engineering, Yanshan University, 438 W Hebei Rd, Qinhuangdao, 066004, Hebei, China
- Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, 601 W Huangpu Ave, Guangzhou, 510632, Guangdong, China
| | - Jie Dong
- Department of Clinical Laboratory, Guangzhou Twelfth People's Hospital, 1 Tianqiang Rd, Guangzhou, 510620, Guangdong, China
| | - Guoqian Jiang
- Key Laboratory of Measurement Technology and Instrumentation of Hebei Province, 438 W Hebei Rd, Qinhuangdao, 066004, Hebei, China
- School of Electrical Engineering, Yanshan University, 438 W Hebei Rd, Qinhuangdao, 066004, Hebei, China
| | - Jianlei Hao
- Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, 601 W Huangpu Ave, Guangzhou, 510632, Guangdong, China
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, 79 Kangning Rd, Zhuhai, 519000, Guangdong, China
| | - Liang Han
- School of Health, Guangdong Pharmaceutical University, 280 Daxuecheng Outer Ring East Rd, Guangzhou, 510006, Guangdong, China
| |
Collapse
|
44
|
Eshaq AM, Flanagan TW, Ba Abbad AA, Makarem ZAA, Bokir MS, Alasheq AK, Al Asheikh SA, Almashhor AM, Binyamani F, Al-Amoudi WA, Bawzir AS, Haikel Y, Megahed M, Hassan M. Immune Checkpoint Inhibitor-Associated Cutaneous Adverse Events: Mechanisms of Occurrence. Int J Mol Sci 2024; 26:88. [PMID: 39795946 PMCID: PMC11719825 DOI: 10.3390/ijms26010088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/13/2025] Open
Abstract
Immunotherapy, particularly that based on blocking checkpoint proteins in many tumors, including melanoma, Merkel cell carcinoma, non-small cell lung cancer (NSCLC), triple-negative breast (TNB cancer), renal cancer, and gastrointestinal and endometrial neoplasms, is a therapeutic alternative to chemotherapy. Immune checkpoint inhibitor (ICI)-based therapies have the potential to target different pathways leading to the destruction of cancer cells. Although ICIs are an effective treatment strategy for patients with highly immune-infiltrated cancers, the development of different adverse effects including cutaneous adverse effects during and after the treatment with ICIs is common. ICI-associated cutaneous adverse effects include mostly inflammatory and bullous dermatoses, as well as severe cutaneous side reactions such as rash or inflammatory dermatitis encompassing erythema multiforme; lichenoid, eczematous, psoriasiform, and morbilliform lesions; and palmoplantar erythrodysesthesia. The development of immunotherapy-related adverse effects is a consequence of ICIs' unique molecular action that is mainly mediated by the activation of cytotoxic CD4+/CD8+ T cells. ICI-associated cutaneous disorders are the most prevalent effects induced in response to anti-programmed cell death 1 (PD-1), anti-cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), and anti-programmed cell death ligand 1 (PD-L1) agents. Herein, we will elucidate the mechanisms regulating the occurrence of cutaneous adverse effects following treatment with ICIs.
Collapse
Affiliation(s)
- Abdulaziz M. Eshaq
- Department of Epidemiology and Biostatstics, Milken Institute School of Public Health, George Washington University Washington, Washington, DC 20052, USA;
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Thomas W. Flanagan
- Department of Pharmacology and Experimental Therapeutics, LSU Health Sciences Center, New Orleans, LA 70112, USA;
| | - Abdulqader A. Ba Abbad
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Zain Alabden A. Makarem
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Mohammed S. Bokir
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Ahmed K. Alasheq
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Sara A. Al Asheikh
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Abdullah M. Almashhor
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Faroq Binyamani
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Waleed A. Al-Amoudi
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (A.A.B.A.); (Z.A.A.M.); (M.S.B.); (A.K.A.); (A.M.A.); (F.B.); (W.A.A.-A.)
| | - Abdulaziz S. Bawzir
- Department of Radiology, King Saud Medical City, Riyadh 11533, Saudi Arabia;
| | - Youssef Haikel
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, University of Strasbourg, 67000 Strasbourg, France
- Pôle de Médecine et Chirurgie Bucco-Dentaire, Hôpital Civil, Hôpitaux Universitaire de Strasbourg, 67000 Strasbourg, France
| | - Mossad Megahed
- Clinic of Dermatology, University Hospital of Aachen, 52074 Aachen, Germany;
| | - Mohamed Hassan
- Research Laboratory of Surgery-Oncology, Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Institut National de la Santé et de la Recherche Médicale, University of Strasbourg, 67000 Strasbourg, France;
- Department of Operative Dentistry and Endodontics, Dental Faculty, University of Strasbourg, 67000 Strasbourg, France
| |
Collapse
|
45
|
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
|
46
|
Kim MK, Son HN, Hong SW, Park SH, Yang DH, Ye BD, Byeon JS, Myung SJ, Yang SK, Yoon S, Hwang SW. CD8+ cell dominance in immune checkpoint inhibitor-induced colitis and its heterogeneity across endoscopic features. Therap Adv Gastroenterol 2024; 17:17562848241309445. [PMID: 39735350 PMCID: PMC11672375 DOI: 10.1177/17562848241309445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/06/2024] [Indexed: 12/31/2024] Open
Abstract
Background Immune checkpoint inhibitor (ICI)-induced colitis is a significant adverse event associated with ICI therapy, known to be linked to increased cytotoxic T-cell activity. Objectives To compare T-cell subsets based on the endoscopic features of ICI-induced colitis and to compare these findings with those of inflammatory bowel disease (IBD). Design Prospective cohort study. Methods We analyzed patients with ICI-induced colitis, confirmed through both endoscopic and histological evaluation. Biopsy specimens were examined using multiplex immunohistochemistry to assess their immune cell profile. Clinical outcomes were analyzed. Immune cell profiles were compared based on their endoscopic features and contrasted with those of patients with IBD. Results Seventeen patients with ICI-induced colitis were included in the study. All patients showed clinical improvement after treatment, and steroids were administered to 11 patients (64.7%). Based on endoscopic features, the patients were classified as Crohn's disease (CD)-like (n = 3, 17.6%), ulcerative colitis (UC)-like (n = 9, 52.9%), or microscopic colitis (MC)-like (n = 5, 29.4%). In ICI-induced colitis, cytotoxic T cells (Tc cells) were more predominant than helper T cells (Th cells) (p = 0.053), and this trend was most pronounced in the MC-like subtype (p = 0.020). When comparing the number of CD8+ cells infiltrating the crypts, both the UC-like and MC-like subtypes had significantly more infiltrating cells than the CD-like subtype (p = 0.008 and p = 0.016, respectively). In comparison to IBD, IBD exhibited a Th-dominant profile, whereas CD-like ICI-induced colitis had a lower Th cell density than CD (p = 0.032) and UC-like ICI-induced colitis had a higher Tc density than UC (p = 0.045). Conclusion Analysis of T-cell subsets of ICI-induced colitis revealed a Tc-dominant profile, contrasting with the Th dominance observed in patients with IBD. The Tc-dominant profile was evident in UC-like and MC-like subtypes, with significant crypt infiltration by CD8+ cells. Tc may play an important role in the pathophysiology of ICI-induced colitis.
Collapse
Affiliation(s)
- Min Kyu Kim
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hye-Nam Son
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung Wook Hong
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang Hyoung Park
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Inflammatory Bowel Disease Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Dong-Hoon Yang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Byong Duk Ye
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Inflammatory Bowel Disease Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jeong-Sik Byeon
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seung-Jae Myung
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Suk-Kyun Yang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
- Inflammatory Bowel Disease Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Shinkyo Yoon
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
| | - Sung Wook Hwang
- Department of Gastroenterology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul 05505, Republic of Korea
- Inflammatory Bowel Disease Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
47
|
Dadwal S, Dhar S, Baghel K, Mishra A, Mehrotra S, Prajapati VK. From past to present: The evolution of immunotherapy and its modern modalities. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 144:1-32. [PMID: 39978965 DOI: 10.1016/bs.apcsb.2024.10.015] [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
Immunotherapy is emerging as a novel and reliable therapeutic technique for treating diseases such as autoimmunity, HIV/AIDS, allergy and cancers. This approach works by modulating the patient's immune system, activating both the innate and humoral branches to combat life-threatening diseases. The foundation of immunotherapy began with the discovery and development of "serum therapy" by German physiologist Emil Von Behring who received the Nobel Prize in 1901 for his contributions to the treatment of diphtheria. Around the same time, Dr. William Coley expanded the field for cancer treatment by developing the first immune based cure for sarcomas using attenuated strains of bacteria injected directly into patient's tumours. As medical science advanced, a broader understanding of the immune system and its components led to the emergence of different immunotherapeutic techniques. These include adoptive cell transfer therapy, cytokine therapy, cancer vaccines, and antibody-drug conjugates. The chapter provides a comprehensive understanding of the history and the current techniques used in immunotherapy, detailing the principles behind their mechanisms and the types of diseases tackled by each immunotherapeutic technique. By examining the journey from early discoveries to modern advancements, the chapter highlights the transformative impact of immunotherapy on medical science and patient care.
Collapse
Affiliation(s)
- Surbhi Dadwal
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Sarthak Dhar
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Kirti Baghel
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Rajasthan, India
| | - Sanjana Mehrotra
- Department of Human Genetics, Guru Nanak Dev University, Amritsar, Punjab, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, University of Delhi South Campus, Benito Juarez Road, Dhaula Kuan, New Delhi, India.
| |
Collapse
|
48
|
Maldonado-García JL, Fragozo A, Pavón L. Cytokine release syndrome induced by anti-programmed death-1 treatment in a psoriasis patient: A dark side of immune checkpoint inhibitors. World J Clin Cases 2024; 12:6782-6790. [PMID: 39687650 PMCID: PMC11525914 DOI: 10.12998/wjcc.v12.i35.6782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/26/2024] [Accepted: 09/09/2024] [Indexed: 10/24/2024] Open
Abstract
In recent years, cancer immunotherapy has introduced novel treatments, such as monoclonal antibodies, which have facilitated targeted therapies against tumor cells. Programmed death-1 (PD-1) is an immune checkpoint expressed in T cells that regulates the immune system's activity to prevent over-activation and tissue damage caused by inflammation. However, PD-1 is also expressed in tumor cells and functions as an immune evasion mechanism, making it a therapeutic target to enhance the immune response and eliminate tumor cells. Consequently, immune checkpoint inhibitors (ICIs) have emerged as an option for certain tumor types. Nevertheless, blocking immune checkpoints can lead to immune-related adverse events (irAEs), such as psoriasis and cytokine release syndrome (CRS), as exemplified in the clinical case presented by Zhou et al involving a patient with advanced gastric cancer who received sintilimab, a monoclonal antibody targeting PD-1. Subsequently, the patient experienced exacerbation of psoriasis and CRS. The objective of this editorial article is to elucidate potential immunologic mechanisms that may contribute to the development of CRS and psoriasis in patients receiving ICIs. It is crucial to acknowledge that while ICIs offer superior safety and efficacy compared to conventional therapies, they can also manifest irAEs affecting the skin, gastrointestinal tract, or respiratory system. In severe cases, these irAEs can lead to life-threatening complications such as circulatory shock or multiorgan failure. Consequently, it is recommended that patients receiving ICIs undergo regular monitoring to identify and manage these adverse events effectively.
Collapse
Affiliation(s)
- José Luis Maldonado-García
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Coyoacán 04510, Ciudad de México, Mexico
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Mexico City 1134, Ciudad de México, Mexico
| | - Ana Fragozo
- Unidad de Desarrollo e Investigación en Bioterapéuticos, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City 11340, Ciudad de México, Mexico
| | - Lenin Pavón
- Laboratorio de Psicoinmunología, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñiz, Mexico City 11340, Mexico
| |
Collapse
|
49
|
Sastri KT, Gupta NV, Kannan A, Dutta S, Ali M Osmani R, V B, Ramkishan A, S S. The next frontier in multiple sclerosis therapies: Current advances and evolving targets. Eur J Pharmacol 2024; 985:177080. [PMID: 39491741 DOI: 10.1016/j.ejphar.2024.177080] [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: 05/28/2024] [Revised: 10/11/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
Recent advancements in research have significantly enhanced our comprehension of the intricate immune components that contribute to multiple sclerosis (MS) pathogenesis. By conducting an in-depth analysis of complex molecular interactions involved in the immunological cascade of the disease, researchers have successfully identified novel therapeutic targets, leading to the development of innovative therapies. Leveraging pioneering technologies in proteomics, genomics, and the assessment of environmental factors has expedited our understanding of the vulnerability and impact of these factors on the progression of MS. Furthermore, these advances have facilitated the detection of significant biomarkers for evaluating disease activity. By integrating these findings, researchers can design novel molecules to identify new targets, paving the way for improved treatments and enhanced patient care. Our review presents recent discoveries regarding the pathogenesis of MS, highlights their genetic implications, and proposes an insightful approach for engaging with newer therapeutic targets in effectively managing this debilitating condition.
Collapse
Affiliation(s)
- K Trideva Sastri
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India.
| | - N Vishal Gupta
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India.
| | - Anbarasu Kannan
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - Suman Dutta
- Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
| | - Riyaz Ali M Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India
| | - Balamuralidhara V
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Shivarathreeshwara Nagara, Bannimantap, Mysuru, India
| | - A Ramkishan
- Deputy Drugs Controller (India), Central Drugs Standard Control Organization, Directorate General of Health Services, Ministry of Health & Family Welfare, Government of India, India
| | | |
Collapse
|
50
|
Singh DD, Haque S, Kim Y, Han I, Yadav DK. Remodeling of tumour microenvironment: strategies to overcome therapeutic resistance and innovate immunoengineering in triple-negative breast cancer. Front Immunol 2024; 15:1455211. [PMID: 39720730 PMCID: PMC11666570 DOI: 10.3389/fimmu.2024.1455211] [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: 06/26/2024] [Accepted: 10/31/2024] [Indexed: 12/26/2024] Open
Abstract
Triple-negative breast cancer (TNBC) stands as the most complex and daunting subtype of breast cancer affecting women globally. Regrettably, treatment options for TNBC remain limited due to its clinical complexity. However, immunotherapy has emerged as a promising avenue, showing success in developing effective therapies for advanced cases and improving patient outcomes. Improving TNBC treatments involves reducing side effects, minimizing systemic toxicity, and enhancing efficacy. Unlike traditional cancer immunotherapy, engineered nonmaterial's can precisely target TNBC, facilitating immune cell access, improving antigen presentation, and triggering lasting immune responses. Nanocarriers with enhanced sensitivity and specificity, specific cellular absorption, and low toxicity are gaining attention. Nanotechnology-driven immunoengineering strategies focus on targeted delivery systems using multifunctional molecules for precise tracking, diagnosis, and therapy in TNBC. This study delves into TNBC's tumour microenvironment (TME) remodeling, therapeutic resistance, and immunoengineering strategies using nanotechnology.
Collapse
Affiliation(s)
- Desh Deepak Singh
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Youngsun Kim
- Department of Obstetrics and Gynecology, Kyung Hee University Medical Center, Seoul, Republic of Korea
| | - Ihn Han
- Plasma Bioscience Research Center, Applied Plasma Medicine Center, Department of Electrical & Biological Physics, Kwangwoon University, Seoul, Republic of Korea
| | - Dharmendra Kumar Yadav
- Department of Biologics, College of Pharmacy, Hambakmoeiro 191, Yeonsu-gu, Incheon, Republic of Korea
| |
Collapse
|