1
|
Jia Z, Zhang Y, Cao L, Wang J, Liang H. Research hotspots and trends of immunotherapy and melanoma: A bibliometric analysis during 2014-2024. Hum Vaccin Immunother 2025; 21:2464379. [PMID: 40012099 PMCID: PMC11869780 DOI: 10.1080/21645515.2025.2464379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 01/22/2025] [Accepted: 02/05/2025] [Indexed: 02/28/2025] Open
Abstract
Over the last decade, the increasing global prevalence of melanoma has sparked growing interest in immunotherapies, which show significant potential against this form of skin cancer. This research aims to offer a framework to guide future studies and inspire new research directions. In this study, we used the Web of Science Core Collection to collect papers on immunotherapy and melanoma published between 2014 and 2024. With Excel and visualization tools like VOSviewer, COOC 13.2, Citespace, and Bibliometrix (R-Tool of R-Studio), we analyzed the data to spot trends and new focuses in the research. Our findings indicate a substantial surge in research activity concerning immunotherapy and melanoma between 2014 and 2024. The USA and China emerged as leading contributors, engaging in extensive and close collaborative efforts with European counterparts. Furthermore, seven of the top 10 research institutions are located in the USA, with the MD Anderson Cancer Center in Texas being the most productive. In addition, the Journal of Cancer Immunotherapy is the journal with the most articles published in the field. Professor Georgina V. Long from the Melanoma Institute at the University of Sydney was one of the most productive scholars. Keyword analysis shows that immune checkpoint inhibitors, tumor microenvironment and targeted therapies are key areas of interest for the research community. This paper uses bibliometric analysis to outline research trends and key points in immunotherapy and melanoma from 2014 to 2024, which helps understand the current research and guides future research directions.
Collapse
Affiliation(s)
- Zixuan Jia
- Department of Urology, People’s Hospital of Longhua, Shenzhen, Guangdong, China
- School of Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Youao Zhang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, Guangdong, China
- The First School of Clinical Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Luyan Cao
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, Scotland
| | - Jieyan Wang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, Guangdong, China
| | - Hui Liang
- Department of Urology, People’s Hospital of Longhua, Shenzhen, Guangdong, China
| |
Collapse
|
2
|
Chen DP, Shen CR, Lin WT, Chu YC. Exploring the link between Co-stimulatory gene polymorphisms and clinical manifestations in Graves' ophthalmopathy. Exp Eye Res 2025; 257:110423. [PMID: 40379199 DOI: 10.1016/j.exer.2025.110423] [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: 12/06/2024] [Revised: 05/11/2025] [Accepted: 05/14/2025] [Indexed: 05/19/2025]
Abstract
Graves' ophthalmopathy (GO) is an autoimmune disorder that affects orbital tissues in approximately 30 % of Graves' disease patients. Single nucleotide polymorphisms (SNPs), particularly in immune-related genes, play a crucial role in the development of GO. This study investigates the association between SNPs in co-stimulatory molecules and specific clinical characteristics of GO, including laterality, orbital pain, swelling, diplopia, exophthalmos, redness, and eyelid retraction. Forty-one patients newly diagnosed with GO were analyzed. Genomic DNA was extracted from their blood, and 98 SNPs were amplified using PCR and sequenced. Candidate SNPs, selected based on prior research, were analyzed using chi-square tests and genetic models to assess genotype and allele frequency differences related to clinical manifestations. CD28 SNPs rs3181096 (C vs. T: p = 0.001) and rs3181098 (G vs. A: p = 0.002) were found to show protective effects against eyelid inflammation, while the A-allele of rs200353921(p = 0.005) increased the risk of eyelid inflammation. PDCD1 SNPs rs36084323 (C vs. T: p = 0.004) and rs41386349 (G vs. A: p = 0.005) were linked to diplopia and eyelid inflammation, respectively. The T-allele of rs6705653 in PDCD1 was found to increase the risk of diplopia (p = 0.001) but decreased the risk of eyelid retraction (p = 0.002). Other SNPs, including rs2227982 (p = 0.003) and rs2227981 (G vs. A: p = 0.001), were also associated with diplopia and eyelid retraction, highlighting the complex genetic influences on the clinical manifestations of GO. Furthermore, interactions between age, gender, and SNPs were observed in relation to GO clinical features. These findings highlight the potential regulatory roles of these genes in influencing immune responses and orbital inflammation in GO. Understanding these genetic influences could help identify predictive markers and novel therapeutic targets for GO management.
Collapse
Affiliation(s)
- Ding-Ping Chen
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chia-Rui Shen
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan; Department of Medical Biotechnology and Laboratory Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wei-Tzu Lin
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yen-Chang Chu
- Department of Ophthalmology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
3
|
Almonte AA, Thomas S, Zitvogel L. Microbiota-centered interventions to boost immune checkpoint blockade therapies. J Exp Med 2025; 222:e20250378. [PMID: 40261296 PMCID: PMC12013646 DOI: 10.1084/jem.20250378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/24/2025] Open
Abstract
Immune checkpoint blockade therapies have markedly advanced cancer treatment by invigorating antitumor immunity and extending patient survival. However, therapeutic resistance and immune-related toxicities remain major concerns. Emerging evidence indicates that microbial dysbiosis diminishes therapeutic response rates, while a diverse gut ecology and key beneficial taxa correlate with improved treatment outcomes. Therefore, there is a growing understanding that manipulating the gut microbiota could boost therapy efficacy. This review examines burgeoning methods that target the gut microbiome to optimize therapy and innovative diagnostic tools to detect dysbiosis, and highlights challenges that remain to be addressed in the field.
Collapse
Affiliation(s)
- Andrew A. Almonte
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
| | - Simon Thomas
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Kremlin-Bicêtre, France
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS) 1428, Villejuif, France
| |
Collapse
|
4
|
Saravia J, Chi H. Immunometabolism of regulatory T cells in cancer. Oncogene 2025; 44:2011-2024. [PMID: 40468052 PMCID: PMC12167712 DOI: 10.1038/s41388-025-03458-1] [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: 02/08/2025] [Revised: 05/06/2025] [Accepted: 05/19/2025] [Indexed: 06/18/2025]
Abstract
Regulatory T (Treg) cells play critical roles in maintaining immune tolerance and tissue homeostasis, but impede anti-tumor immunity. Recent work has established how Treg cells metabolically adapt within the tumor microenvironment (TME), and these adaptations frequently provide a functional advantage over effector T cells. Further, enhanced Treg cell function in the TME may contribute to the limited efficacy of current immunotherapies, especially immune checkpoint blockade (ICB). Here, we review recent progress in understanding mechanisms of Treg cell heterogeneity and function in tumors, with a particular focus on cellular metabolism as an underlying factor by which Treg cells are uniquely poised to thrive in the TME and contribute to tumorigenesis. We describe how cellular metabolism and nutrient or metabolic communication shape Treg cell lineage identity and function in the TME. We also discuss the interplay between ICB and Treg cell metabolism and function, and highlight current strategies targeting Treg cell metabolism specifically in the TME. Understanding metabolic control of intratumoral Treg cells provides excellent opportunities to uncover new or combination therapies for cancer.
Collapse
Affiliation(s)
- Jordy Saravia
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
5
|
Ma C, Yu X, Zhang X, Su L, Jiang O, Cui R. Combination of radiotherapy and ICIs in advanced hepatocellular carcinoma: A systematic review of current evidence and future prospects (Review). Oncol Lett 2025; 30:342. [PMID: 40438865 PMCID: PMC12117537 DOI: 10.3892/ol.2025.15088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 03/24/2025] [Indexed: 06/01/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a global health concern because of its rising prevalence and high fatality rates. Conventional treatments for advanced HCC (aHCC) have limited success, emphasizing the need for novel treatment options. Radiotherapy (RT) treatments, such as stereotactic body radiation and proton therapy, improve local tumor management via precision targeting. Moreover, immune checkpoint inhibitors (ICIs) that target the programmed cell death protein 1(PD-1)/PD ligand 1 (PD-L1) and cytotoxic T lymphocyte associated protein 4 (CTLA-4) pathways have promise for systemic antitumor effectiveness. The combination of RT and ICIs takes advantage of their complementary mechanisms: RT kills immunogenic cells and controls the tumor microenvironment to increase antigen presentation, whereas ICIs enhance and maintain antitumor immune responses. This combination enhances tumor regression and immune response in aHCC, improving response rate and progression-free survival with manageable safety. The present review aimed to summarize the rationale for combining RT + ICIs in patients with aHCC and clinical outcomes, as well as ways to enhance this combination technique. The combination of these models is a promising technique for improving outcomes for patients with aHCC and warrants further investigation.
Collapse
Affiliation(s)
- Cheng Ma
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| | - Xinlin Yu
- Department of Oncology, The Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Xialin Zhang
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Lihong Su
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| | - Ou Jiang
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| | - Ran Cui
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| |
Collapse
|
6
|
Kakimi K, Sugie T. Why combine and why neoadjuvant? Tumor immunological perspectives on chemoimmunotherapy in triple-negative breast cancer. Breast Cancer 2025; 32:676-688. [PMID: 40327275 DOI: 10.1007/s12282-025-01707-5] [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: 02/24/2025] [Accepted: 04/18/2025] [Indexed: 05/07/2025]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype characterized by limited targeted therapies and high recurrence rates. While immune checkpoint inhibitors (ICIs) have shown promise, their efficacy as monotherapy is limited. Clinically, ICIs demonstrate significant benefit primarily when combined with chemotherapy, particularly in the neoadjuvant setting for early-stage TNBC, which yields superior outcomes compared to adjuvant therapy. This review elucidates the tumor immunological principles underlying these observations. We discussed how the suppressive tumor microenvironment (TME), progressive T cell exhaustion, and associated epigenetic scarring constrain ICI monotherapy effectiveness. Crucially, we highlight the immunological advantages of the neoadjuvant approach: the presence of the primary tumor provides abundant antigens, and intact tumor-draining lymph nodes (TDLNs) act as critical sites for ICI-mediated priming and expansion of naïve and precursor exhausted T cells. This robust activation within TDLNs enhances systemic anti-tumor immunity and expands the T cell repertoire, a process less effectively achieved in the adjuvant setting after tumor resection. These mechanisms provide a strong rationale for the improved pathological complete response (pCR) rates and event-free survival observed with neoadjuvant chemoimmunotherapy, as demonstrated in trials like KEYNOTE-522. We further explore the implications for adjuvant therapy decisions based on treatment response, the challenges of ICI resistance, the need for predictive biomarkers, management of immune-related adverse events (irAEs), and future therapeutic directions. Understanding the dynamic interplay between chemotherapy, ICIs, T cells, and the TME, particularly the role of TDLNs in the neoadjuvant context, is essential for optimizing immunotherapy strategies and improving outcomes for patients with TNBC.
Collapse
Affiliation(s)
- Kazuhiro Kakimi
- Department of Immunology, Kindai University Faculty of Medicine, 377-2 Onohigashi, Osakasayama, Osaka, 589-8511, Japan.
- Chemotherapy Center, Kansai Medical University Kori Hospital, 8-45 Korihondori, Neyagawa, Osaka, 572-8551, Japan.
| | - Tomoharu Sugie
- Chemotherapy Center, Kansai Medical University Kori Hospital, 8-45 Korihondori, Neyagawa, Osaka, 572-8551, Japan.
| |
Collapse
|
7
|
Yonese I, Yasuda Y, Takemura K, Toide M, Soma T, Yoneoka Y, Fujiwara R, Ito M, Oguchi T, Numao N, Yamamoto S, Yuasa T, Koga F, Yonese J. Impact of IRAES on the outcomes of pembrolizumab therapy in patients with MUC: A comprehensive analysis of severity and the type and number of affected organs. Urol Oncol 2025; 43:441.e1-441.e10. [PMID: 39971635 DOI: 10.1016/j.urolonc.2025.02.003] [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: 10/20/2024] [Revised: 01/26/2025] [Accepted: 02/01/2025] [Indexed: 02/21/2025]
Abstract
BACKGROUND Immune-related adverse events (irAEs) are reportedly associated with favorable outcomes in patients with metastatic urothelial carcinoma (mUC) receiving pembrolizumab. Previous studies on this topic focused on the severity of irAEs. The type and number of organs affected by irAEs may also be associated with the therapeutic outcomes. METHODS The present, retrospective study included 146 patients with mUC receiving pembrolizumab between January 2018 and March 2022. The primary endpoints were the overall response rate (ORR), progression-free survival (PFS), and overall survival (OS) associated with the type and number of organs affected by irAEs and the severity of the symptoms. IrAEs were graded using the Common Terminology Criteria for Adverse Events version 5.0. The treatment response was assessed using the Response Evaluation Criteria in Solid Tumors version 1.1. Cox proportional hazards was used to assess for any association between the variables and survival. Time-dependent analysis was used to assess the status of irAEs as a prognostic factor. RESULTS IrAEs ≥ grade (G) 2 were observed in 48 (33%) patients, of whom 9 (6%) had multiple irAEs. IrAEs ≥G2 were significantly associated with a higher ORR (57% vs. 22% for CONCLUSIONS The severity and number of irAEs and the organs affected by them appeared to be relevant to the therapeutic efficacy of pembrolizumab in patients with mUC.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Male
- Female
- Retrospective Studies
- Middle Aged
- Aged
- Treatment Outcome
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/therapeutic use
- Carcinoma, Transitional Cell/drug therapy
- Carcinoma, Transitional Cell/pathology
- Carcinoma, Transitional Cell/mortality
- Aged, 80 and over
- Severity of Illness Index
- Adult
- Urologic Neoplasms/drug therapy
- Urologic Neoplasms/pathology
- Urologic Neoplasms/mortality
- Urinary Bladder Neoplasms/drug therapy
- Urinary Bladder Neoplasms/pathology
Collapse
Affiliation(s)
- Ichiro Yonese
- Departments of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan; Department of Urology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Yosuke Yasuda
- Departments of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Kosuke Takemura
- Departments of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masahiro Toide
- Department of Urology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Takahiko Soma
- Department of Urology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Yusuke Yoneoka
- Departments of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryo Fujiwara
- Departments of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masaya Ito
- Department of Urology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Tomohiko Oguchi
- Departments of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Noboru Numao
- Departments of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Shinya Yamamoto
- Departments of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takeshi Yuasa
- Departments of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Fumitaka Koga
- Department of Urology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Junji Yonese
- Departments of Genitourinary Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
8
|
Zhang D, Li J, Liang Y, Li T, Ji M, Liu X, Li W, Chen P, Zhang J, Yang Z, Wang L, Chen H. Extracellular HSP90-Facilitated Degradation of Extracellular and Membrane Proteins by Bifunctional Small Molecules. J Med Chem 2025; 68:12641-12657. [PMID: 40497783 DOI: 10.1021/acs.jmedchem.5c00379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2025]
Abstract
Lysosome-targeting chimeras represent a promising strategy for degrading extracellular and membrane proteins via the lysosomal pathway, but the available receptor options remain limited. Herein, we report a novel strategy utilizing extracellular heat shock protein 90 (eHSP90) to facilitate lysosomal degradation of target proteins through bifunctional small molecules, termed extracellular heat shock protein 90-targeting chimeras (eHSPTACs). By connecting an HSP90 ligand to a target protein ligand, eHSPTACs effectively induced the internalization and subsequent lysosomal degradation of extracellular Alexa Fluor 488-labeled α-DNP antibody and membrane programmed cell death-ligand 1 (PD-L1). Notably, dPDL1-4 selectively degraded membrane PD-L1 in tumor cells over normal cells, leveraging the elevated expression of eHSP90 in cancer cells. Moreover, dPDL1-4 demonstrated robust in vivo degradation of membrane PD-L1 and significant tumor growth suppression in the B16F10 syngeneic mouse model. Overall, eHSPTACs offer a general platform for tumor-selective degradation of extracellular and membrane proteins, providing a new therapeutic avenue.
Collapse
Affiliation(s)
- Dongli Zhang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, Department of Chemical Biology, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Jie Li
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, Department of Chemical Biology, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Yaqi Liang
- Department of Medicinal Chemistry, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Tao Li
- Department of Medicinal Chemistry, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Ming Ji
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, Department of Chemical Biology, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Xinmin Liu
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, Department of Chemical Biology, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Wenxin Li
- Department of Medicinal Chemistry, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Pengxiao Chen
- Department of Medicinal Chemistry, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Jiamin Zhang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, Department of Chemical Biology, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Zhengduo Yang
- Department of Pathology, Tianjin Union Medical Center, Tianjin 300121, China
| | - Lulu Wang
- The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics; Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, Department of Chemical Biology, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - He Chen
- Department of Medicinal Chemistry, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
9
|
Dalloul Z, Briend JG, Diawara M, Taylor C, Ouellette RJ. Leveraging liquid biopsy to uncover resistance mechanisms and guide personalized immunotherapy. Transl Oncol 2025; 59:102445. [PMID: 40561796 DOI: 10.1016/j.tranon.2025.102445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 06/04/2025] [Accepted: 06/10/2025] [Indexed: 06/28/2025] Open
Abstract
Cancer therapy has been revolutionized by immune checkpoint inhibitors (ICIs) that create a new paradigm among cancer immunotherapies. These agents restore the immune system capacity to fight cancer through blocking the action of major immune checkpoint proteins that suppress immune responses such as programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), which tumors exploit for immune escape. Another advancement is the absence of known biomarkers involves ICIs, which can further be translated into new liquid tests like liquid biopsies, indicating the immune status of the patient. Liquid biopsy is a minimally invasive method for identifying tumor-derived components such as circulating tumor cells, cell-free DNA and extracellular vesicles (EVs) from body fluids for analysis. Researchers increasingly use liquid biopsy for biomarkers discovery and patient stratification into clinical applications. Even though immunotherapy has great advances, still there are obstacles faced while using ICIs. Many patients fail to respond to the treatment because of the heterogeneous mechanisms of resistance. Immunotherapy resistance is a dynamic interplay between tumors and their surrounding stroma. To understand this variability, humanized mice models are increasingly used for mirroring human immune responses. Such models offer insight into cancer immunotherapy when human immune system is engrafted into mice, and EV and biomarker profiles are established in those models. EVs reflect differences in tumor characteristics and the immune landscape around tumors to develop personalized strategies to enhance ICI efficacy. This may improve patient prognosis, giving reason to hope for better, more effective treatments.
Collapse
Affiliation(s)
- Zeinab Dalloul
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| | | | - Mariama Diawara
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| | - Catherine Taylor
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada
| | - Rodney J Ouellette
- Atlantic Cancer Research Institute, Moncton, New Brunswick, Canada; Department of Chemistry and Biochemistry, Université de Moncton, Moncton, New Brunswick, Canada; Dr Georges L. Dumont University Hospital, Vitalite Health Network, Moncton, New Brunswick, Canada; Beatrice Hunter Cancer Research Institute, Halifax, Nova Scotia, Canada.
| |
Collapse
|
10
|
Cappelli LC, Perin J, Zeger S, Jones M, Bingham CO, Shah AA. Two clinical subgroups of immune checkpoint inhibitor-induced inflammatory arthritis determined by latent class analysis. Semin Arthritis Rheum 2025; 74:152773. [PMID: 40540778 DOI: 10.1016/j.semarthrit.2025.152773] [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: 03/31/2025] [Revised: 05/30/2025] [Accepted: 06/10/2025] [Indexed: 06/22/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) for cancer treatment can cause inflammatory arthritis (IA). Since ICI-IA has a unique pathogenesis, applying categories of traditional IA may be of limited use. METHODS Participants were ≥18 years old, treated with anti-PD-1, anti-PD-L1, and/or anti-CTLA-4 agents, and had ICI-IA diagnosed by a rheumatologist. We clustered patients using latent class analysis (LCA) applied with phenotypic data from the baseline rheumatology visit. The Bayesian Information Criteria (BIC) was used to select the number of phenotypes. We compared demographics, cancer type and treatments, and IA clinical features and treatments between the estimated phenotypes. Finally, we explored differences in cytokine levels and the presence of shared epitope between the groups. RESULTS Twenty variables were used to estimate latent classes. Two distinct phenotypes were indicated by the BIC; 77 patients were estimated to be the first phenotype and 49 in the second phenotype. The statistically significant features that distinguished the phenotypes included higher levels of all components of the CDAI, more stiffness, and more small and upper extremity joint involvement for phenotype 2. Patients in phenotype 2 were marginally more likely to require steroids during their course. There were no significant differences in cancer type, stage or ICI treatment between the phenotype groups. Baseline levels of VEGF-A were higher in phenotype 2. CONCLUSIONS Two separate phenotypes of ICI-IA were identified using LCA, the second having a more severe polyarthritis at baseline and involving the upper extremities. These subgroups provide an opportunity to identify corresponding biomarkers to predict disease outcomes.
Collapse
Affiliation(s)
- Laura C Cappelli
- Johns Hopkins University School of Medicine, Division of Rheumatology, USA.
| | - Jamie Perin
- Johns Hopkins Bloomberg School of Public Health, Department of Biostatistics, USA
| | - Scott Zeger
- Johns Hopkins University School of Medicine, Division of Rheumatology, USA; Johns Hopkins Bloomberg School of Public Health, Department of Biostatistics, USA
| | - Michelle Jones
- Johns Hopkins University School of Medicine, Division of Rheumatology, USA
| | - Clifton O Bingham
- Johns Hopkins University School of Medicine, Division of Rheumatology, USA
| | - Ami A Shah
- Johns Hopkins University School of Medicine, Division of Rheumatology, USA
| |
Collapse
|
11
|
Ibrahim A, Shalabi L, Gadelmawla AF, Hasan MT, Al-Asmar R, Alsheyab Y, Pavan Lingamsetty SS, Yousef AM, Al-Shammari AS, Eltelt AM, Azzam AY, Shahzad M, Sayed SBH, Alsalman O, Hamad B, Al-Qudimat AR. Efficacy and safety of immune checkpoint inhibitors for advanced or metastatic esophageal squamous cell carcinoma: A network meta-analysis. Eur J Pharmacol 2025; 1002:177797. [PMID: 40543743 DOI: 10.1016/j.ejphar.2025.177797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 06/02/2025] [Accepted: 06/02/2025] [Indexed: 06/28/2025]
Abstract
BACKGROUND Combining immune checkpoint inhibitors (ICIs) with chemotherapy has significantly transformed cancer treatment, offering better options for esophageal squamous cell carcinoma (ESCC). This comprehensive network meta-analysis evaluates the efficacy and safety of various ICIs in patients with advanced ESCC. METHODS A systematic literature search of randomized controlled trials (RCTs) was performed across major databases up to March 2025, focusing on studies comparing ICIs (as monotherapy or combined with chemotherapy) against chemotherapy alone. Efficacy and safety outcomes were synthesized using a frequentist network meta-analysis model to generate indirect and direct treatment comparisons. RESULTS Thirteen trials involving 6997 patients revealed that combining ICIs with chemotherapy generally outperformed chemotherapy alone. Specifically, the combination of toripalimab and chemotherapy demonstrated the most significant improvement in overall survival (HR: 0.58, 95 % CI: 0.43-0.78, p = 0.0003; SUCRA: 0.86). Meanwhile, Sintilimab combined with chemotherapy provided the greatest benefit in progression-free survival (HR = 0.56, 95 % CI [0.46; 0.68], p-value <0.0001; SUCRA: 0.87). Tislelizumab alone was noted for its effectiveness in enhancing overall response rates (RR = 2.08, p = 0.0012). Nivolumab was the most effective in lowering the risk of grade 3 or greater adverse events compared to chemotherapy (RR = 0.28, 95 % CI: 0.21-0.39, p < 0.0001; SUCRA: 1). CONCLUSION Combining ICIs with chemotherapy revealed superior efficacy in advanced or metastatic ESCC compared to chemotherapy alone. These findings support using novel immunotherapeutic agents to enhance patient outcomes, warranting further research to optimize treatment regimens and manage adverse events effectively.
Collapse
Affiliation(s)
- Ahmed Ibrahim
- Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Laila Shalabi
- Faculty of Medicine, Gharyan University, Gharyan, Libya.
| | | | | | | | | | | | | | | | - Ahmed M Eltelt
- Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Ahmed Y Azzam
- Montefiore-Einstein Cerebrovascular Research Lab, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| | | | | | | | | | - Ahmad R Al-Qudimat
- Surgical research section, Department of surgery, Hamad Medical Corporation, Doha, Qatar; Public Health College, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
12
|
Cunningham MM, Romero R, Alvarez C, Saxena Beem S, Schwartz TA, Ishizawar RC. Neutrophil to Lymphocyte Ratio as a Biomarker for the Prediction of Cancer Outcomes and Immune-Related Adverse Events in a CTLA-4-Treated Population. Cancers (Basel) 2025; 17:2011. [PMID: 40563660 PMCID: PMC12190284 DOI: 10.3390/cancers17122011] [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: 04/09/2025] [Revised: 05/23/2025] [Accepted: 06/06/2025] [Indexed: 06/28/2025] Open
Abstract
Background/Objectives: Immune-related adverse events (irAEs) triggered by immune checkpoint inhibitor therapy (ICI) have been paradoxically associated with both significant morbidity and improved cancer outcomes. While predictive markers for irAEs have been studied in the PD-1 blockade, less is known for their role in CTLA-4 inhibition. This study aims to fill this gap by evaluating NLR and irAE incidence in a CTLA-4-treated population. Methods: This study is a single-center retrospective cohort study investigating 111 patients treated with CTLA-4 inhibition (ipilimumab) to assess associations for baseline low NLR values with cancer outcomes and irAE type and incidence. Patient charts were manually reviewed by a single physician, and unclear clinical events were assessed by a second physician reviewer. Results: In this cohort, the occurrence of more than one irAE presentation was associated with an improved cancer outcome, OR 1.48 (1.02, 2.15). When stratified by organ-specific manifestation, only endocrinologic irAEs were associated with improved cancer outcome, OR 2.82 (1.19, 6.67). A low baseline NLR was statistically significantly associated with an increased incidence of irAEs of any type, OR 4.34 (1.73, 10.9). Conclusions: These data show that irAE occurrence in cancer patients treated with CTLA-4 inhibition is associated with improved cancer outcomes, similar to that previously seen with PD-1 inhibition. It also suggests that the NLR may serve as a practical peripheral biomarker to predict both cancer response and odds of irAEs in patients treated with CTLA-4 inhibition. This low-cost and widely available tool could provide additional information for modeling cancer outcomes.
Collapse
Affiliation(s)
- Michael M. Cunningham
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, University of North Carolina, Chapel Hill, NC 27514, USA;
| | - Rachel Romero
- Atrium Health Rheumatology, 200 Medical Park Drive, Suite 330, Concord, NC 28025, USA;
| | - Carolina Alvarez
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA; (C.A.); (T.A.S.)
| | - Shruti Saxena Beem
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA; (C.A.); (T.A.S.)
| | - Todd A. Schwartz
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA; (C.A.); (T.A.S.)
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Rumey C. Ishizawar
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, University of North Carolina, Chapel Hill, NC 27514, USA;
- Thurston Arthritis Research Center, University of North Carolina, Chapel Hill, NC 27599, USA; (C.A.); (T.A.S.)
| |
Collapse
|
13
|
Kraehenbuehl L, Winkelbeiner N, Turko P, Staeger R, Ghosh A, Kaiser V, Stadler PC, Nordmann TM, Brüggen MC, Levesque MP, Contassot E, French LE, Dummer R, Meier-Schiesser B. Immune-Related Cutaneous Adverse Events Display Distinct Clinical and Molecular Characteristics, Depending on Immune Checkpoints Targeted. Cancers (Basel) 2025; 17:1992. [PMID: 40563643 PMCID: PMC12190265 DOI: 10.3390/cancers17121992] [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: 05/01/2025] [Revised: 05/28/2025] [Accepted: 06/09/2025] [Indexed: 06/28/2025] Open
Abstract
Background/Objectives: Immune-related cutaneous adverse events (ircAEs) are common complications of cancer immunotherapy and provide insight into immune-related adverse events (irAEs) more broadly. To enhance our molecular understanding, we characterized ircAEs resulting from single-agent (PD1) and combined immunotherapy regimens (P+C). Clinically, maculopapular rash (MPR) and toxic epidermal necrolysis (TEN) resemble ircAEs, providing a valuable basis for investigations. Methods: To investigate the transcriptome and immune infiltrates in ircAEs, we conducted transcriptomic analyses and multiplexed immunohistochemistry on skin biopsies from patients receiving PD1 and P+C, as well as those with MPR, TEN, and healthy controls. Results: Principal component analysis revealed distinct transcriptomic clustering between ircAEs, MPR, and TEN. Specifically, PD1 ircAEs exhibited a gene expression profile similar to TEN, with upregulation of Type-I-response-related genes (e.g., CXCL9 Log2FC 5.34, p < 0.0001, CXCL10 Log2FC 6.03, p < 0.0001), while P+C ircAEs more closely resembled MPR. Immune infiltrates differed significantly between all groups (p = 0.002 by PERMANOVA for all groups). CD4 T-cells were abundant in the dermis of ircAEs from any type of immunotherapy. However, PD1 stained positive in 1.07% of CD4 cells with PD1 monotherapy, compared to 0.3%, 0.4%, and 0.08% in P+C, MPR, and TEN, respectively. Conclusions: This study identified distinct molecular and cellular signatures in ircAEs depending on the type of immune checkpoint blockade. aPD1-associated ircAEs share similarities with the cytotoxic profile of TEN, while P+C more closely mirrored MPR. These findings support the need for tailored management strategies for ircAEs, emphasizing personalized therapeutic approaches to minimize treatment interruptions while preserving the efficacy of cancer immunotherapy.
Collapse
Affiliation(s)
- Lukas Kraehenbuehl
- Department of Dermatology and Allergology, Kantonsspital Aarau, 5001 Aarau, Switzerland; (L.K.); (R.D.)
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland; (P.T.); (R.S.); (V.K.); (T.M.N.); (M.-C.B.); (M.P.L.)
- Faculty of Medicine, Dermatology and Venereology, University of Zurich, 8006 Zurich, Switzerland; (N.W.); (A.G.)
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, Meyer Cancer Center, New York, NY 10065, USA
| | - Nicola Winkelbeiner
- Faculty of Medicine, Dermatology and Venereology, University of Zurich, 8006 Zurich, Switzerland; (N.W.); (A.G.)
| | - Patrick Turko
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland; (P.T.); (R.S.); (V.K.); (T.M.N.); (M.-C.B.); (M.P.L.)
- Faculty of Medicine, Dermatology and Venereology, University of Zurich, 8006 Zurich, Switzerland; (N.W.); (A.G.)
| | - Ramon Staeger
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland; (P.T.); (R.S.); (V.K.); (T.M.N.); (M.-C.B.); (M.P.L.)
- Faculty of Medicine, Dermatology and Venereology, University of Zurich, 8006 Zurich, Switzerland; (N.W.); (A.G.)
| | - Adhideb Ghosh
- Faculty of Medicine, Dermatology and Venereology, University of Zurich, 8006 Zurich, Switzerland; (N.W.); (A.G.)
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, 8091 Zurich, Switzerland
| | - Vivienn Kaiser
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland; (P.T.); (R.S.); (V.K.); (T.M.N.); (M.-C.B.); (M.P.L.)
- Faculty of Medicine, Dermatology and Venereology, University of Zurich, 8006 Zurich, Switzerland; (N.W.); (A.G.)
| | - Pia-Charlotte Stadler
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, 82152 Munich, Germany;
- Department of Dermatology and Allergy, University Hospital, LMU Munich, 81377 Munich, Germany;
| | - Thierry M. Nordmann
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland; (P.T.); (R.S.); (V.K.); (T.M.N.); (M.-C.B.); (M.P.L.)
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried, 82152 Munich, Germany;
- Department of Dermatology and Allergy, University Hospital, LMU Munich, 81377 Munich, Germany;
| | - Marie-Charlotte Brüggen
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland; (P.T.); (R.S.); (V.K.); (T.M.N.); (M.-C.B.); (M.P.L.)
- Faculty of Medicine, Dermatology and Venereology, University of Zurich, 8006 Zurich, Switzerland; (N.W.); (A.G.)
| | - Mitchell P. Levesque
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland; (P.T.); (R.S.); (V.K.); (T.M.N.); (M.-C.B.); (M.P.L.)
- Faculty of Medicine, Dermatology and Venereology, University of Zurich, 8006 Zurich, Switzerland; (N.W.); (A.G.)
| | - Emmanuel Contassot
- Department of Biomedicine, University of Basel, 4001 Basel, Switzerland;
| | - Lars E. French
- Department of Dermatology and Allergy, University Hospital, LMU Munich, 81377 Munich, Germany;
- Philip Frost Department of Dermatology, University of Miami, Miami, FL 33136, USA
| | - Reinhard Dummer
- Department of Dermatology and Allergology, Kantonsspital Aarau, 5001 Aarau, Switzerland; (L.K.); (R.D.)
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland; (P.T.); (R.S.); (V.K.); (T.M.N.); (M.-C.B.); (M.P.L.)
- Faculty of Medicine, Dermatology and Venereology, University of Zurich, 8006 Zurich, Switzerland; (N.W.); (A.G.)
| | - Barbara Meier-Schiesser
- Department of Dermatology, University Hospital Zurich, 8091 Zurich, Switzerland; (P.T.); (R.S.); (V.K.); (T.M.N.); (M.-C.B.); (M.P.L.)
- Faculty of Medicine, Dermatology and Venereology, University of Zurich, 8006 Zurich, Switzerland; (N.W.); (A.G.)
| |
Collapse
|
14
|
Xu K, Gu T, Su K, Liu X, He B, He J, Chi H, Zhou X, Liu H, Xiao R, Tang X, Ye Q, Zhou X, Liu Y, Xiong J, Wang P, Li H, He K, Guo L, Han Y. Stereotactic body radiation therapy (SBRT) increases anti-PD-1 antitumor activity by enhancing the tumor immune microenvironment in mice with metastatic hepatocellular carcinoma. Discov Oncol 2025; 16:1081. [PMID: 40512454 PMCID: PMC12165918 DOI: 10.1007/s12672-025-02914-4] [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: 07/28/2024] [Accepted: 06/04/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND To explore the effect of radiotherapy on anti-pd-1 anti-tumor activity in metastatic hepatocellular carcinoma. METHODS Patients with metastatic HCC treated with intensity-modulated radiation therapy (IMRT) in combination with immunotherapy (n = 13) were retrospectively analyzed by comparing its efficacy with that of immunotherapy alone (n = 12) as well as untreated (n = 20) patients with metastatic hepatocellular carcinoma. Animal experiment used mouse hepatocellular carcinoma H22 cell metastatic tumor model and were also divided into a control group, a PD-1 antibody group, an SBRT group, and an SBRT combined with a PD-1 antibody group. SBRT treatment is 8 Gy×3 F. The growth curves of body weight, irradiated tumor (the primary tumor), and non-irradiated tumor (secondary tumor) were plotted for each group of tumor-bearing mice. For this study, we used flow cytometry to examine effector CD8 + T cells expression in both irradiated and non-irradiated tumors, the CD4 + T and CD4+/CD8 + T cells ratio in the spleen, and used enzyme-linked immunosorbent assays (ELISA) to analyze the concentrations of IFN-γ and IL-10 in serum. Tumors were additionally stained with immunohistochemistry Ki-67 and TdT-mediated dUTP nick end labeling (TUNEL). We used hematoxylin-eosin (HE) staining of liver, spleen, lungs, kidneys, and heart to assess the anti-tumor activity of each group of tumor-bearing mice and their tolerance to determine the safety of the approach. RESULTS Clinical results: The median survival of IMRT + PD-1 group, PD-1 group, and control group were 17.5 months (95Confidence Interval (CI) 13.2-21.8), 12.5 months (95CI 9.0-16.0), and 5.2 months (95CI 5.5-12.9), respectively (P < 0.001). SBRT combined with PD-1 antibody improved tumor control in both radiated and non-radiated tumors, resulting in a complete cure of the half of mice in animal studies. This was linked to an increased in CD8 + effector T cells infiltration triggered by radiotherapy. HE staining of mice in the SBRT combined with the PD-1 treatment group suggested no damage to the liver, spleen, lungs, kidneys, and heart. CONCLUSIONS This study showed that SBRT, while being well-tolerated, significantly increased anti-PD-1 antitumor activity by enhancing the tumor immune microenvironment in mice with metastatic hepatocellular carcinoma without significant toxic side effects. DISCLAIMER This manuscript was previously submitted as a preprint only in Experimental Hematology & Oncology and has no conflict of interest with this submission.
Collapse
Affiliation(s)
- Ke Xu
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Oncology, Chongqing General Hospital, Chongqing, 401147, China
| | - Tao Gu
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Ke Su
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
- Department of Radiation Oncology, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100000, China
| | - Xin Liu
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Bingsheng He
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Jie He
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Xuancheng Zhou
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Hanlin Liu
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Rui Xiao
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Xue Tang
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Qinni Ye
- Clinical Medical College, Southwest Medical University, Luzhou, 646000, China
| | - Xue Zhou
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Yingpeng Liu
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Jie Xiong
- Department of Public Health, Southwest Medical University, Luzhou, 646000, China
| | - Pan Wang
- Clinical Skills Center, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Han Li
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Kun He
- Clinical Research Institute, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, Sichuan, China.
| | - Lu Guo
- Department of Ophthalmology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
| | - Yunwei Han
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, China.
- Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, 646000, China.
- Academician (Expert) Workstation of Sichuan Province, Luzhou, 646000, China.
- The Affiliated Hospital of Southwest Medical University, 25 Taiping Street, Luzhou, Sichuan, China.
| |
Collapse
|
15
|
Qi X, Zhang M, Zhu J, Wang H. Case report of concurrent ulcerative colitis and bilateral breast cancer: a literature review of the bidirectional association between autoimmune diseases and breast cancer. Front Oncol 2025; 15:1495731. [PMID: 40567613 PMCID: PMC12187860 DOI: 10.3389/fonc.2025.1495731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 05/19/2025] [Indexed: 06/28/2025] Open
Abstract
A 58-year-old female patient with bilateral breast cancer developed unexpected hematochezia at a frequency of approximately 10 episodes per day following adjuvant chemotherapy, with the emergency endoscopy reporting superficial ulcers throughout the entire colon, suggesting a diagnosis of ulcerative colitis (UC). Given the absence of preexisting autoimmune history or detectable autoantibodies, we supposed that the onset of UC was closely related to the chemotherapy. The complex bidirectional relationship between autoimmune rheumatic diseases and cancer continues to be elucidated. Variations in autoimmune disease type, duration, and specific clinical/laboratory features may modulate cancer risk, either increasing or decreasing susceptibility to certain malignancies. These associations could potentially inform type-specific cancer screening strategies. Furthermore, the widespread use of immune checkpoint inhibitors across multiple tumor types, along with their associated inflammatory syndromes, has significant implications for the development and management of autoimmune rheumatic diseases. Herein, we report this case, which could be one of the few bilateral breast cancer cases to be reported with ulcerative colitis, and conducted a literature review of the bidirectional association of breast cancer and autoimmune diseases.
Collapse
Affiliation(s)
- Xiao Qi
- Department of Oncology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin, China
- The Institute of Translational Medicine, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Miao Zhang
- Department of Oncology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin, China
- The Institute of Translational Medicine, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
| | - Jinhong Zhu
- Nankai University School of Medicine, Tianjin, China
| | - Huaqing Wang
- Department of Oncology, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
- Tianjin Cancer Institute of Integrative Traditional Chinese and Western Medicine, Tianjin, China
- The Institute of Translational Medicine, Tianjin Union Medical Center, The First Affiliated Hospital of Nankai University, Tianjin, China
| |
Collapse
|
16
|
Rysz J, Ławiński J, Franczyk B, Gluba-Sagr A. Immune Checkpoint Inhibitors in Clear Cell Renal Cell Carcinoma (ccRCC). Int J Mol Sci 2025; 26:5577. [PMID: 40565041 PMCID: PMC12193005 DOI: 10.3390/ijms26125577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 05/27/2025] [Accepted: 05/27/2025] [Indexed: 06/28/2025] Open
Abstract
Renal cell carcinoma (RCC) accounts for about 403,000 new cases and 175,000 deaths worldwide each year. Clear cell RCC (ccRCC), the most prevalent subtype, is often driven by genetic mutations, such as VHL inactivation, leading to angiogenesis and immune escape. Immune checkpoint inhibitors (ICIs) targeting PD-1, PD-L1, and CTLA-4 have transformed treatment paradigms, yet therapeutic resistance remains a critical challenge. The immunosuppressive nature of the tumor microenvironment (TME) in ccRCC plays a central role in limiting ICI efficacy. Emerging strategies aim to overcome resistance by targeting key components of the TME, including tumor-associated macrophages, regulatory T cells (Tregs), and cytokine signaling. Agents such as nivolumab, pembrolizumab, and ipilimumab have demonstrated the ability to restore T-cell activity and mitigate immune suppression, offering clinical benefit in metastatic ccRCC. However, response rates vary, highlighting the need for rational combination therapies. ICIs combined with VEGF inhibitors have shown promising outcomes in clinical trials, and novel regimens continue to be explored. Risk stratification and personalized treatment selection are increasingly important as the therapeutic landscape evolves. This review synthesizes current advances in immunotherapy for ccRCC, with a focus on mechanisms of resistance and innovative strategies to enhance immune responsiveness. A deeper understanding of TME modulation and strategic combination approaches is essential to improve survival and quality of life for patients with advanced ccRCC.
Collapse
Affiliation(s)
- Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
| | - Janusz Ławiński
- Department of Medicine, Faculty of Medicine, University of Rzeszow, 35-959 Rzeszow, Poland
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
| | - Anna Gluba-Sagr
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland; (J.R.); (B.F.)
| |
Collapse
|
17
|
Acar C, Yüksel HÇ, Şahin G, Açar FP, Karaca B. Immune checkpoint inhibitor resumption after discontinuation due to immune-related adverse events: a nomogram-based analysis of risk factors and outcomes. Expert Rev Anticancer Ther 2025:1-9. [DOI: 10.1080/14737140.2025.2517274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2025] [Accepted: 05/28/2025] [Indexed: 06/21/2025]
|
18
|
Wang X, Fu X, Liu Q, Li J, Ge Y, Zhang J, Wang S, Wang L, Wang D, Sun Y, Gan Y, Sun H, Wang Z, Sun Y, Gao A. Impact of irae characteristics on efficacy of consolidative immunotherapy following chemoradiotherapy in locally advanced NSCLC. BMC Pulm Med 2025; 25:283. [PMID: 40483449 PMCID: PMC12144761 DOI: 10.1186/s12890-025-03742-6] [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: 03/01/2025] [Accepted: 05/27/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND Consolidative PD-L1 inhibitors after concurrent chemoradiotherapy (cCRT) have become standard care in locally advanced non-small cell lung cancer (LA-NSCLC). However, the correlation between immune-related adverse event (irAE) characteristics and patient outcomes remains unclear. METHODS This retrospective study enrolled LA-NSCLC patients who received consolidative PD-L1 inhibitors after CRT at four cancer centers. Patients who received CRT alone were also included for comparison. Associations between irAE characteristics, frequency, timing, affected systems, and severity, and progression-free survival (PFS) and overall survival (OS) were assessed. Tumor immune microenvironment (TIME) features were analyzed to identify subpopulations at higher risk of severe irAEs. RESULTS Among 107 patients, 59 (55.1%) developed irAEs; 89.8% were grade 1-2 and 10.2% grade 3-4. Patients with irAEs had significantly longer PFS than those without. Late-onset, single-system, endocrine, and mild irAEs predicted better PFS. In contrast, patients with severe irAEs had worse survival than those without ICI consolidation. TIME analysis revealed that severe irAEs were associated with higher CD103+CD8+ T cells infiltration. A > 1.545% cutoff for CD103+CD8+ T cells may help identify patients less likely to benefit from PD-L1 inhibitor consolidation. CONCLUSIONS Occurrence of irAEs, particularly late-onset, single-system, or grade 1-2 correlated with greater benefit from consolidative PD-L1 inhibitors in LA-NSCLC. Conversely, severe irAEs predict poorer survival, even compared to no ICI consolidation. Elevated CD103+CD8+ T cell infiltration may serve as a biomarker to identify patients at risk of severe irAEs who may not benefit from immunoconsolidation therapy.
Collapse
Affiliation(s)
- Xiufen Wang
- Phase I Clinical Trail Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong, 250117, China
| | - Xuebing Fu
- Department of Thoracic Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Qiaohong Liu
- Department of Ultrasound, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250013, P. R. China
| | - Juan Li
- Phase I Clinical Trail Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong, 250117, China
| | - Yihui Ge
- Department of Thoracic Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Jian Zhang
- Phase I Clinical Trail Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong, 250117, China
| | - Shuyun Wang
- Department of Thoracic Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Leirong Wang
- Phase I Clinical Trail Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong, 250117, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Dahai Wang
- Phase I Clinical Trail Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong, 250117, China
| | - Yanxin Sun
- Phase I Clinical Trail Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong, 250117, China
| | - Yiling Gan
- Phase I Clinical Trail Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong, 250117, China
| | - Haodong Sun
- Department of Thoracic Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Zhen Wang
- Special Inspection Department, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| | - Yuping Sun
- Phase I Clinical Trail Center, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong, 250117, China.
| | - Aiqin Gao
- Department of Thoracic Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| |
Collapse
|
19
|
Mizuno K, Ito T, Sawada T, Kobayashi T, Iwama S, Mori S, Hase T, Fukami Y, Furusawa K, Yura Y, Morimoto R, Sajiki AF, Ushida H, Kato N, Maruyama S, Murohara T, Katsuno M, Ishii M, Akiyama M, Arima H, Kawashima H, Ando Y. Safety and efficacy of retreatment with immune checkpoint inhibitors after severe immune-related adverse events. Oncologist 2025; 30:oyaf120. [PMID: 40515478 PMCID: PMC12166119 DOI: 10.1093/oncolo/oyaf120] [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: 12/06/2024] [Accepted: 04/22/2025] [Indexed: 06/16/2025] Open
Abstract
BACKGROUND While immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, they can trigger severe immune-related adverse events (irAEs). The safety and efficacy of ICI retreatment after severe irAEs remain poorly understood. METHODS We conducted a retrospective analysis of 1271 patients with malignancies treated with ICIs at a university hospital in Japan between September 2014 and June 2023. We evaluated the incidence and characteristics of severe irAEs, defined as grade ≥3, and the safety and efficacy of ICI retreatment. RESULTS Severe irAEs occurred in 222 patients (17.5%). Patients with single endocrinopathies were excluded, and 46 (28.4%) of the remaining 162 patients underwent ICI retreatment. Upon retreatment, 14 patients (30.4%) experienced recurrent or new grade ≥2 irAEs. One patient who experienced hepatotoxicity (grade 3) at initial ICI treatment developed a recurrence (grade 4). Regarding antitumor response, the objective response rate to retreatment was 28.3% (13/46), with 10.9% achieving complete and 17.4% partial response. The median duration of ICI administration after retreatment was 218 days (95% confidence interval [CI]: 84-399). At 1 year after retreatment, 15.4% (95% CI: 6.8-27.4) of patients discontinued due to irAEs, 44.4% (95% CI: 29.7-58.1) due to disease progression, 6.6% (95% CI: 1.7-16.3) completed planned treatment, and 33.4% (95% CI: 20.3-47.2) continued treatment. CONCLUSIONS ICI retreatment after severe irAEs demonstrated a manageable safety profile and promising efficacy, even in patients with grade ≥3 irAEs. ICI retreatment may be a viable option for patients with limited alternatives, particularly those showing favorable antitumor responses at initial treatment.
Collapse
Affiliation(s)
- Kazuyuki Mizuno
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takanori Ito
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tsunaki Sawada
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomoko Kobayashi
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shintaro Iwama
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shoichiro Mori
- Department of Dermatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsunari Hase
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuki Fukami
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenji Furusawa
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshimitsu Yura
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryota Morimoto
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ai Fujita Sajiki
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Ushida
- Department of Ophthalmology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Noritoshi Kato
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shoichi Maruyama
- Department of Nephrology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Clinical Research Education, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Makoto Ishii
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masashi Akiyama
- Department of Dermatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Arima
- Department of Endocrinology and Diabetes, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuichi Ando
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
20
|
Chanson N, Galvagni A, Ramos-Casals M, Ruiz JI, Suijkerbuijk KPM, Gente K, Kerschen P, Karam JD, Belkhir R, Outh R, Closs-Prophette F, Garcia Morillo JS, Robles-Marhuenda Á, Michot JM, Voisin AL, Messayke S, Laparra A, Robert C, Suarez-Almazor M, Mariette X, Lambotte O, ICIR
. Immune checkpoint inhibitors-associated vasculitis: a heterogeneous condition with possible severe disease course. Rheumatology (Oxford) 2025; 64:3685-3690. [PMID: 39714261 DOI: 10.1093/rheumatology/keae711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Collaborators] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/21/2024] [Accepted: 12/06/2024] [Indexed: 12/24/2024] Open
Abstract
OBJECTIVE To describe presentation, treatment and outcome of immune checkpoint inhibitor (ICI) associated-vasculitis in cancer patients in a multicentre study. METHODS Thanks to the ImmunoCancer International Registry (ICIR), a multidisciplinary network focused on the research of the immune related adverse events related to cancer immunotherapies, patients presenting with a clinical and/or radiological suspicion of vasculitis and histological evidence of vasculitis after being exposed to ICIs were retrospectively identified. RESULTS Twenty-eight cases were identified in the ICIR registry. The median interval between starting ICI treatment and vasculitis diagnosis was 4 months. Small vessel vasculitis was predominant (n = 21), followed by large vessel (n = 4) and medium vessel (n = 3). The small vessel vasculitis included 10 unclassified vasculitis either with limited cutaneous involvement (n = 6) or systemic involvement (n = 4), five IgA vasculitis, three cryoglobulinemic vasculitis, and three ANCA+ vasculitis. At presentation or during the evolution, renal and neurologic manifestations were evidenced in seven cases each (25%). Renal biopsies documented immune glomerulopathies in six cases. Only seven patients (25%) fulfilled the 2022 ACR/EULAR classification criteria (four giant cell arteritis, two EGPA and one GPA). Most patients (90%) required systemic corticosteroid and an additional drug was given in 10 patients (36%). Vasculitis outcome was good: 22 patients had vasculitis complete response, no patient died due to vasculitis. Nine patients (32%) were rechallenged with immunotherapy with only one relapse. CONCLUSION ICI-associated vasculitis are rare, heterogeneous, but can be severe requiring urgent multidisciplinary management with aggressive treatment.
Collapse
Affiliation(s)
- Noemie Chanson
- Department of Internal Medicine and Clinical Immunology, AP-HP, Université Paris Saclay, Bicêtre Hospital, Le Kremlin Bicêtre, France
| | - Alexandre Galvagni
- Department of Internal Medicine and Clinical Immunology, AP-HP, Université Paris Saclay, Bicêtre Hospital, Le Kremlin Bicêtre, France
| | - Manuel Ramos-Casals
- Department of Autoimmune Diseases, ICMiD, Hospital Clinic, Barcelona, Spain
- Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Juan Ignacio Ruiz
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Karijn P M Suijkerbuijk
- Department of Medical Oncology, UMC Utrecht Cancer Center, Utrecht University, Utrecht, the Netherlands
| | - Karolina Gente
- Department of Internal Medicine and Rheumatology, Universitätsklinikum, Heidelberg, Germany
| | - Philippe Kerschen
- Service de Neurologie, Centre Hospitalier de Luxembourg, Luxembourg-Ville, Luxembourg
| | - Jean Denis Karam
- Internal Medicine and RECIF, CHU Amiens Picardie, Amiens, France
| | - Rakiba Belkhir
- Department of Rheumatology, AP-HP, Université Paris Saclay, Bicêtre Hospital, Le Kremlin Bicêtre, France
| | - Rodereau Outh
- Service de Médecine Interne et Générale, Centre Hospitalier de Perpignan, Perpignan, France
| | | | | | | | - Jean-Marie Michot
- Departement D'Innovation Therapeutique et D'Essais Precoces, Institut Gustave Roussy, Universite Paris-Saclay, Villejuif, France
| | - Anne Laure Voisin
- Departement D'Innovation Therapeutique et D'Essais Precoces, Institut Gustave Roussy, Universite Paris-Saclay, Villejuif, France
| | - Sabine Messayke
- Departement D'Innovation Therapeutique et D'Essais Precoces, Institut Gustave Roussy, Universite Paris-Saclay, Villejuif, France
| | - Arianne Laparra
- DIOPP, Institut Gustave Roussy, Universite Paris-Saclay, Villejuif, France
| | - Caroline Robert
- Department of Dermatology, Institut Gustave Roussy, University Paris-Saclay, Villejuif, France
| | - Maria Suarez-Almazor
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xavier Mariette
- Department of Rheumatology, AP-HP, Université Paris Saclay, Bicêtre Hospital, Le Kremlin Bicêtre, France
- University Paris-Saclay; INSERM; CEA, Centre Immunology of Viral Infections and Autoimmune Diseases, IDMIT, UMR1184, Department IBFJ, Le Kremlin-Bicêtre, France
| | - Olivier Lambotte
- Department of Internal Medicine and Clinical Immunology, AP-HP, Université Paris Saclay, Bicêtre Hospital, Le Kremlin Bicêtre, France
- University Paris-Saclay; INSERM; CEA, Centre Immunology of Viral Infections and Autoimmune Diseases, IDMIT, UMR1184, Department IBFJ, Le Kremlin-Bicêtre, France
| | | |
Collapse
Collaborators
Manuel Ramos-Casals, Xavier Mariette, Olivier Lambotte, Marie Kostine, Munther A Khamashta, Leonard Calabrese, Maria Suárez-Almazor, Chiara Baldini, Clifton O Bingham, Jacques-Eric Gottenberg, Timothy R D Radstake, Thierry Schaeverbeke, Hendrik Schulze-Koops, Pilar Brito-Zerón, Alejandra Flores-Chávez, Nihan Acar-Denizli,
Collapse
|
21
|
Schwarzlmueller P, Triebig A, Assié G, Jouinot A, Theurich S, Maier T, Beuschlein F, Kobold S, Kroiss M. Steroid hormones as modulators of anti-tumoural immunity. Nat Rev Endocrinol 2025; 21:331-343. [PMID: 40128599 DOI: 10.1038/s41574-025-01102-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/27/2025] [Indexed: 03/26/2025]
Abstract
Immune evasion is a hallmark of cancer progression but the role of steroid hormones in this evasion has long been underrated. This oversight is particularly notable for glucocorticoids given that exogenous glucocorticoids remain a cornerstone therapy in various oncological treatment regimens, supportive care and treatment of immune-related adverse events caused by immune-checkpoint inhibitors. Cortisol, the main endogenous glucocorticoid in humans, is secreted by the adrenal cortex in response to stress. Additionally, cortisol and its inactive metabolite cortisone can be interconverted to further modulate tissue-dependent glucocorticoid action. In the past 5 years, intratumoural production of glucocorticoids, by both immune and tumour cells, has been shown to support tumour immune evasion. Here, we summarize current progress at the crossroads of endocrinology and immuno-oncology. We outline the known effects of steroid hormones on different immune cell types with a focus on glucocorticoids and androgens. We conclude with options for pharmaceutical intervention, including the engineering of cell-based therapies that resist the immunosuppressive action of steroid hormones. Overall, local steroid production and metabolism are emerging elements of tumour immune suppression that are potentially amenable to therapeutic intervention. Targeting steroid hormones to enhance anticancer therapies could increase their efficacy but will require expertise in endocrine care.
Collapse
Affiliation(s)
| | - Alexandra Triebig
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Guillaume Assié
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
- Department of Endocrinology and National Reference Center for Rare Adrenal Disorders, Hôpital Cochin, Paris, France
| | - Anne Jouinot
- Department of Endocrinology and National Reference Center for Rare Adrenal Disorders, Hôpital Cochin, Paris, France
- Université Paris Cité, Institut Cochin, Paris, France
| | - Sebastian Theurich
- Department of Medicine III and Comprehensive Cancer Center (CCC Munich LMU), LMU University Hospital, Munich, Germany
- Cancer- and Immunometabolism Research Group, Gene Center, Ludwig Maximilian University (LMU), Munich, Germany
- Bavarian Cancer Research Center (BZKF), Munich, Germany
- German Cancer Consortium (DKTK), Munich Site, Heidelberg, Germany
| | - Tanja Maier
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Felix Beuschlein
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ), Zurich, Switzerland
- The LOOP Zurich - Medical Research Center, Zurich, Switzerland
| | - Sebastian Kobold
- German Cancer Consortium (DKTK), Munich Site, Heidelberg, Germany
- Division of Clinical Pharmacology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Matthias Kroiss
- Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany.
- Bavarian Cancer Research Center (BZKF), Munich, Germany.
- Kroiss Endokrinologie & Diabetologie, Schweinfurt, Germany.
| |
Collapse
|
22
|
Kazanas S, Anastasopoulou A, Ziogas D, Gogas H, Angelousi A. An Extremely Rare Case of Immune Checkpoint Inhibitors-Related Hypoparathyroidism and a Critical Literature Review. JCEM CASE REPORTS 2025; 3:luaf097. [PMID: 40356787 PMCID: PMC12066411 DOI: 10.1210/jcemcr/luaf097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Indexed: 05/15/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have been approved to treat a variety of malignancies, including melanoma, improving the prognosis of these patients. However, they also cause a wide spectrum of rare immune-related adverse events (irAEs), not well-described yet. Although many endocrinopathies have been recognized as irAEs, primary hypoparathyroidism has rarely been reported, and thus clinical suspicion remains low. Herein, we describe the case of a 68-year-old female patient with metastatic melanoma who was admitted to the emergency department with acute symptomatic hypocalcemia due to immune-related (ir)-hypoparathyroidism after 2 cycles of nivolumab/ipilimumab. The patient was treated symptomatically, and her calcium levels were normalized. Parathyroid hormone levels were partially restored during the 6 months of follow-up. A literature review was conducted, summarizing all other subjects who developed ir-hypoparathyroidism after exposure to ICI-based regimen. The review identified 10 additional cases of hypoparathyroidism during immunotherapy. Interestingly, all melanoma cases with ir-hypoparathyroidism had received nivolumab/ipilimumab; 3 of them were also screened and detected with positive calcium-sensing receptor (CaSR) antibodies. Primary hypoparathyroidism may acutely manifest with symptomatic hypocalcemia and care providers should be aware of this rare irAE.
Collapse
Affiliation(s)
- Spyridon Kazanas
- First Department of Internal Medicine, Laiko General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens 11527, Greece
| | - Amalia Anastasopoulou
- First Department of Internal Medicine, Laiko General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens 11527, Greece
| | - Dimitrios Ziogas
- First Department of Internal Medicine, Laiko General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens 11527, Greece
| | - Helen Gogas
- First Department of Internal Medicine, Laiko General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens 11527, Greece
| | - Anna Angelousi
- First Department of Internal Medicine, Unit of Endocrinology, Laiko General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens 11527, Greece
| |
Collapse
|
23
|
Lasagna A. Mitigation strategies for gastrointestinal (GI) immune-related adverse events for patients with solid tumors receiving immunotherapy. Immunotherapy 2025; 17:595-603. [PMID: 40478172 DOI: 10.1080/1750743x.2025.2516995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Accepted: 06/04/2025] [Indexed: 06/29/2025] Open
Abstract
Over the past decade, immunotherapy has revolutionized the treatment algorithm for solid tumors. Immune checkpoint inhibitors (ICIs) demonstrated efficacy against several tumor types, but they can favor the development of immune-related adverse events (irAEs). IrAEs can sometimes be life-threatening. In this review, we will briefly analyze the main gastro-intestinal toxicities and focus on potential strategies for mitigating irAEs, particularly through the modification of gut microbiota (GM) composition. Finally, we will briefly dwell on the potential role of artificial intelligence (AI) in the prediction of irAEs.
Collapse
Affiliation(s)
- Angioletta Lasagna
- Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
24
|
Orikasa M, Shibuya T, Ishino H, Omori M, Odakura R, Koma M, Ito K, Maruyama T, Nomura K, Ishikawa D, Hojo M, Nagahara A. Small-Bowel Capsule Endoscopy in Immune-Mediated Enterocolitis: A Case Series of Four Patients. JGH Open 2025; 9:e70201. [PMID: 40538456 PMCID: PMC12176690 DOI: 10.1002/jgh3.70201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2025] [Accepted: 06/07/2025] [Indexed: 06/22/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have emerged as a cornerstone of cancer immunotherapy but are associated with immune-mediated adverse events (imAEs), including gastrointestinal toxicities. Among these, enteritis is a less common but clinically significant complication. However, small intestinal involvement remains under-recognized, and endoscopic findings are not well characterized. We report four cases of imAE enteritis in which small-bowel capsule endoscopy (SBCE) was performed. SBCE revealed variable mucosal abnormalities, including villous atrophy, aphthous ulcers, and extensive erosions; in one case, deep circumferential ulcers with stricture were observed. While one patient responded to corticosteroids, the other three required escalation to biologic therapy due to steroid-refractory disease. These findings suggest that SBCE may play a valuable role in assessing disease extent and predicting treatment responsiveness in imAE enteritis. Early utilization of SBCE could facilitate timely therapeutic decision-making in affected patients.
Collapse
Affiliation(s)
- Masayuki Orikasa
- Department of GastroenterologyJuntendo University School of MedicineTokyoJapan
| | - Tomoyoshi Shibuya
- Department of GastroenterologyJuntendo University School of MedicineTokyoJapan
- Department of Pathophysiological Research and Therapeutics for Gastrointestinal DiseaseJuntendo University School of MedicineTokyoJapan
| | - Hirotaka Ishino
- Department of GastroenterologyJuntendo University School of MedicineTokyoJapan
| | - Masashi Omori
- Department of GastroenterologyJuntendo University School of MedicineTokyoJapan
| | - Rina Odakura
- Department of GastroenterologyJuntendo University School of MedicineTokyoJapan
| | - Masao Koma
- Department of GastroenterologyJuntendo University School of MedicineTokyoJapan
| | - Kentaro Ito
- Department of GastroenterologyJuntendo University School of MedicineTokyoJapan
| | - Takafumi Maruyama
- Department of GastroenterologyJuntendo University School of MedicineTokyoJapan
| | - Kei Nomura
- Department of GastroenterologyJuntendo University School of MedicineTokyoJapan
- Innovative Microbiome Therapy Research CenterJuntendo UniversityTokyoJapan
| | - Dai Ishikawa
- Department of GastroenterologyJuntendo University School of MedicineTokyoJapan
- Department of Pathophysiological Research and Therapeutics for Gastrointestinal DiseaseJuntendo University School of MedicineTokyoJapan
- Innovative Microbiome Therapy Research CenterJuntendo UniversityTokyoJapan
| | - Mariko Hojo
- Department of GastroenterologyJuntendo University School of MedicineTokyoJapan
- Department of Pathophysiological Research and Therapeutics for Gastrointestinal DiseaseJuntendo University School of MedicineTokyoJapan
| | - Akihito Nagahara
- Department of GastroenterologyJuntendo University School of MedicineTokyoJapan
- Department of Pathophysiological Research and Therapeutics for Gastrointestinal DiseaseJuntendo University School of MedicineTokyoJapan
- Innovative Microbiome Therapy Research CenterJuntendo UniversityTokyoJapan
| |
Collapse
|
25
|
Zavgorodneva Z, Khan M, Guber H. Co-Occurrence of Hypophysitis and Thyroiditis Induced by PD-L1 Inhibitor Avelumab: Clinical Insights. JCEM CASE REPORTS 2025; 3:luaf093. [PMID: 40356788 PMCID: PMC12066409 DOI: 10.1210/jcemcr/luaf093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Indexed: 05/15/2025]
Abstract
Various endocrinopathies have been described in patients with malignancies who are receiving treatment with immune checkpoint inhibitors (ICIs). We present the case of a patient who was hospitalized with an acute alteration in mental status and was referred to the endocrinology service for evaluation of abnormal thyroid function tests. Subsequent investigation revealed the presence of acute adrenal insufficiency, which was successfully managed with steroid replacement therapy. This case highlights a unique simultaneous presentation of thyrotoxicosis due to thyroiditis and adrenal insufficiency due to hypophysitis in a patient treated with ICI avelumab for 5 months.
Collapse
Affiliation(s)
- Zhanna Zavgorodneva
- Division of Endocrinology, Diabetes & Metabolism, Veterans Affairs New York Harbor Healthcare System, Brooklyn, NY 11209, USA
- Division of Endocrinology, Diabetes & Metabolism, State University of New York, Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Muzammil Khan
- Division of Endocrinology, Diabetes & Metabolism, Veterans Affairs New York Harbor Healthcare System, Brooklyn, NY 11209, USA
- Division of Endocrinology, Diabetes & Metabolism, State University of New York, Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Helena Guber
- Division of Endocrinology, Diabetes & Metabolism, Veterans Affairs New York Harbor Healthcare System, Brooklyn, NY 11209, USA
| |
Collapse
|
26
|
McManus HD, Long JB, Westvold SJ, Leapman MS, Hurwitz ME, Mitchell AP, Pollack CE, Gross CP, Dinan MA. Off-Label Use of First-Line Immunotherapy for Metastatic Renal Cell Carcinoma. Clin Genitourin Cancer 2025; 23:102330. [PMID: 40186898 DOI: 10.1016/j.clgc.2025.102330] [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: 01/16/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 04/07/2025]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICI) were approved by the Food and Drug Administration (FDA) for patients with metastatic renal cell carcinoma (mRCC) in the second- line setting in 2015 and the first-line (1L) in 2018. Little is known about 1 L ICI use in the off-label (before FDA indication-specific approval) and postapproval settings. PATIENTS AND METHODS We retrospectively analyzed off-label and post-FDA-approval 1 L ICI receipt in a cohort of Medicare beneficiaries ≥66 years old diagnosed with mRCC from 2015 to 2019. Off-label and postapproval 1 L ICI were defined as before or on/after 4/16/2018 (1L ipilimumab/nivolumab approval). Associations between demographic characteristics and 1 L ICI receipt in the off-label and postapproval periods were examined using multivariable logistic regression. RESULTS We identified 23,469 patients, of which 368 (2.4%) off-label and 1,663 (21%) postapproval received 1 L ICI. In the off-label period, patients with co-morbid conditions were more likely to receive 1 L ICI compared to patients with no co-morbidities (3+ conditions, OR = 2.00; 95% CL, 1.31-3.05). In the postapproval period, older patients were less likely to receive 1 L ICI (81+ vs. 66-70, OR = 0.60; 95% CL, 0.52-0.69), and patients who were frail were less likely to receive 1 L ICI (OR = 0.77; 95% CL, 0.69-0.87). There were not significant differences in 1 L ICI receipt based on race/ethnicity. CONCLUSION Older patients and patients with more comorbidities were more likely to receive 1 L ICI off-label, but these differences did not persist after FDA approval. After 1 L ipilimumab/nivolumab approval, patients receiving 1 L ICI were more likely younger, healthy, and receiving dual-ICI regimens.
Collapse
Affiliation(s)
- Hannah D McManus
- Department of Medicine, Duke University School of Medicine, Durham, NC.
| | - Jessica B Long
- Yale Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT; Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Sarah J Westvold
- Yale Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT; Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Michael S Leapman
- Yale Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT; Department of Urology, Yale School of Medicine, New Haven, CT; Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT
| | - Michael E Hurwitz
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Aaron P Mitchell
- Health Outcomes Research Group, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Craig Evan Pollack
- Department of Health Policy and Management, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD and Johns Hopkins School of Nursing, Baltimore, MD
| | - Cary P Gross
- Yale Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT; Department of Internal Medicine, Yale School of Medicine, New Haven, CT; Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT
| | - Michaela A Dinan
- Yale Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT; Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT
| |
Collapse
|
27
|
Cobelas-Cartagena SS, Amigo-Otero E, García-Samblas V, Flores-Raposo J, Rodrigo-Herrero S, Bayo-Calero J. Pembrolizumab-induced myocarditis, myositis, and myasthenia gravis overlap syndrome (IM3OS) treated with Efgartigimod: case report. Immunotherapy 2025; 17:561-566. [PMID: 40510022 DOI: 10.1080/1750743x.2025.2517522] [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: 03/24/2025] [Accepted: 06/05/2025] [Indexed: 06/29/2025] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) has revolutionized the treatment of many malignancies. However, they may be associated with life-threatening immune-mediated adverse effects.In this article, we present the case of a patient diagnosed with stage IIB acral melanoma (pT4aN0M0) who started adjuvant treatment with pembrolizumab, after which he developed severe myocarditis with myositis and myasthenia gravis overlap syndrome (IM3OS) treated with Efgartigimod.The use of Efgartigimod resulted in a remarkable recovery, marking the first reported acute application of this agent for IM3OS and positive anti-acetylcholine receptor antibodies (anti-AChR).
Collapse
Affiliation(s)
| | - Emilio Amigo-Otero
- Cardiology Unit, Hospital Universitario Juan Ramón Jiménez, Huelva, Spain
| | | | | | | | - Juan Bayo-Calero
- Oncology Unit, Hospital Universitario Juan Ramón Jiménez, Huelva, Spain
| |
Collapse
|
28
|
Arabi S, Fadaee M, Kazemi T, Rahmani M. Advancements in colorectal cancer immunotherapy: from CAR-T cells to exosome-based therapies. J Drug Target 2025; 33:749-760. [PMID: 39754507 DOI: 10.1080/1061186x.2024.2449482] [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: 09/27/2024] [Revised: 12/03/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025]
Abstract
Colorectal cancer (CRC) continues to be a major worldwide health issue, with elevated death rates linked to late stages of the illness. Immunotherapy has made significant progress in developing effective techniques to improve the immune system's capacity to identify and eradicate cancerous cells. This study examines the most recent advancements in CAR-T cell treatment and exosome-based immunotherapy for CRC. CAR-T cell therapy, although effective in treating blood cancers, encounters obstacles when used against solid tumours such as CRC. These obstacles include the presence of an immunosuppressive tumour microenvironment and a scarcity of tumour-specific antigens. Nevertheless, novel strategies like dual-receptor CAR-T cells and combination therapy involving cytokines have demonstrated promise in surmounting these obstacles. Exosome-based immunotherapy is a promising approach for targeted delivery of therapeutic drugs to tumour cells, with high specificity and minimal off-target effects. However, there are still obstacles to overcome in the field, such as resistance to treatment, adverse effects associated with the immune system, and the necessity for more individualised methods. The current research is focused on enhancing these therapies, enhancing the results for patients, and ultimately incorporating these innovative immunotherapeutic approaches into the standard treatment protocols for CRC.
Collapse
Affiliation(s)
- Sepideh Arabi
- Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Manouchehr Fadaee
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Mohammadreza Rahmani
- Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
29
|
Brazel D, Kazakova V, Fay M, Bollin K, Mittal K, Reynolds KL, Tsang M. Connecting the Dots: Practical Strategies for Academic and Community Oncology Synergy to Advance Multidisciplinary Management in Immunotherapy Toxicity Care. Am Soc Clin Oncol Educ Book 2025; 45:e473080. [PMID: 40408607 DOI: 10.1200/edbk-25-473080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Immunotherapy has significantly affected cancer treatment and survival rates, accompanied by an increase in immune-related adverse events (irAEs) requiring new management strategies. irAEs can affect various organ systems and have varying severity levels, with higher rates observed when combining immune checkpoint inhibitors. National organizations such as ASCO, the National Comprehensive Cancer Network, the Society for Immunotherapy of Cancer, and the European Society for Medical Oncology have created guidelines for managing irAEs. This chapter expands on these guidelines by discussing practical strategies to improve the multidisciplinary management in irAE care, focusing on the who, what, and how to bridge gaps in care and enhance collaboration between academic and community oncology practices. Effective irAE management involves early recognition and guideline-adherent approaches using a multidisciplinary team, including oncologists, other subspecialists, primary care clinicians, and all care team members. Institutions are developing methods to integrate irAE care into clinical workflows, such as incorporating urgent care clinics and e-consults for efficient irAE management and developing hub-and-spoke models to extend specialized care from academic centers to community hospitals for equitable care delivery. Additionally, effective patient education is critical for improving irAE recognition and health literacy. The new ASCO Community of Practice called the Alliance for Support and Prevention of Immune-Related Adverse Events consortium and patient advocacy group Standing Together to Optimize Research, Interventions, and Education in irAEs initiatives aim to advance irAE clinical care, research, and education through global collaboration, standardized data collection, and improved outreach to patients and caregivers.
Collapse
Affiliation(s)
- Danielle Brazel
- Division of Hematology and Oncology, Scripps Clinic, La Jolla, CA
| | - Vera Kazakova
- Division of Hematology and Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA
| | - Magdalena Fay
- Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Kathryn Bollin
- Division of Hematology and Oncology, Scripps Clinic, La Jolla, CA
| | - Kriti Mittal
- Division of Hematology and Oncology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA
| | - Kerry L Reynolds
- Division of Hematology and Oncology, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Mazie Tsang
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Phoenix, AZ
| |
Collapse
|
30
|
Braverman EL, Mognol GP, Minn AJ, Vignali DAA, Varner JA. One Step Ahead: Preventing Tumor Adaptation to Immune Therapy. Am Soc Clin Oncol Educ Book 2025; 45:e481556. [PMID: 40334183 DOI: 10.1200/edbk-25-481556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Immune checkpoint inhibitors are cancer therapeutics that have shown remarkable success in extending lives in many cancers, including melanoma, MSI-high cancers, and other cancers. However, these therapeutics have not shown benefit for many patients with cancer, especially those with advanced cancer diagnoses. In addition, many patients develop resistance to these therapeutics and/or life-altering adverse events that can include cardiotoxicity, pneumonitis, thyroiditis, pancreatitis, and hepatitis. Extensive efforts to improve cancer care by uncovering mechanisms of resistance to immune therapy in solid tumors have led to identification of new sources of resistance and to the development of new approaches to activate or sustain antitumor immunity. Chronic stimulation of T cells by tumors and by checkpoint inhibitors can lead to a progressive state of T-cell exhaustion. Chronic T-cell activation by the tumor microenvironment (TME) or immune therapeutics can upregulate the expression and function of alternate checkpoints, including the T-cell protein LAG-3. Persistent interferon signaling in the TME can drive epigenetic changes in cancer cells that enable tumors to counter immune activation and disrupt tumor cell elimination. In addition, immune-suppressive macrophages can flood tumors in response to signals from dying tumor cells, further preventing effective immune responses. New clinical developments and/or approvals for therapies that target alternate immune checkpoints, such as the T-cell checkpoint LAG-3; myeloid cell proteins, such as the kinase phosphoinositide 3-kinase gamma isoform; and chronic interferon signaling, such as Jak 1 inhibitors, have been approved for cancer care or shown promise in recent clinical trials.
Collapse
Affiliation(s)
- Erica L Braverman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Pediatrics, Division of Pediatric Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Giuliana P Mognol
- Moores Cancer Center, University of California, San Diego, La Jolla, CA
| | - Andy J Minn
- Institute for Immunology and Immune Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA
| | - Judith A Varner
- Moores Cancer Center, University of California, San Diego, La Jolla, CA
- Department of Pathology, University of California, San Diego, La Jolla, CA
| |
Collapse
|
31
|
Ma H, Zhang S, Jiao P, Ding H, Wang F, Zhao Y, Wu J, Guo Z. Serum IL-6 predicts immunotherapy-related adverse and outcome in advanced gastric and esophageal cancer patients with Anti-PD-1 treatment. Front Immunol 2025; 16:1553882. [PMID: 40519900 PMCID: PMC12163003 DOI: 10.3389/fimmu.2025.1553882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 05/13/2025] [Indexed: 06/18/2025] Open
Abstract
Purpose Immune checkpoint inhibitors (ICIs) significantly prolong the survival of cancer patients. including gastric adenocarcinoma (GAC) and esophageal squamous cell carcinoma (ESCC) patients. Immune-related adverse events (irAEs) are inevitably involved in ICIs treatment sometimes with severe consequences. Extreme caution is necessary for predicting irAEs and precisely screening of appropriate patients. We evaluated the association of interleukin-6 (IL-6) with irAEs and their impacts on ICIs treatment effectiveness in advanced GAC and ESCC patients. Methods This retrospective study analyzed 121 patients treated with ICIs between March 1, 2020 and August 31, 2023 to evaluate the association between serum IL-6 and ICIs treatment effectiveness. The occurrence of irAEs, including grade and category, and effectiveness of immunotherapy, including objective remission rate (ORR), disease control rate (DCR), progression-free survival (PFS) and overall survival (OS), was evaluated. Categorical count data were tested by chi-square test. Nonparametric rank sum tests were performed using Wilcoxon and Kruskal-Wallis test. Survival rate estimation and survival curves were generated using Kaplan-Meier curve and Log-rank test. Univariate and multivariable COX regression analyses were performed to identify independent prognostic factors. Results A total of 121 patients including 79 with GAC and 42 with ESCC patients were randomly divided into TC (n=81) and VC (n=40) groups. Higher serum IL-6 levels were associated with increased incidence of irAEs, the outcome analysis also indicated its association with lower DCR, shorter PFS and shorter OS in TC group. The higher IL-6 related irAEs occurrence and poor prognosis (DCR, PFS) was confirmed in the VC group. Individual tumor analysis showed that higher IL-6 was associated with both irAEs occurrence and poor prognosis (DCR, PFS, OS) in ESCC patients, and with irAEs occurrence and poor prognosis (DCR, PFS) in GAC patients. No statistically significant associations were observed between pathological biomarkers including programmed cell death ligand 1 (PD-L1), mismatch repair (MMR) and human epidermal growth factor receptor 2 (HER2) and either IL-6 levels or irAEs occurrence in both GC and ESCC patients. Conclusion Elevated serum IL-6 levels were associated with the incidence of irAEs, and higher IL-6 levels predicted worse prognosis in GAC and ESCC patients with ICIs treatment.
Collapse
Affiliation(s)
- Hongfang Ma
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Shasha Zhang
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Pengqing Jiao
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Haixia Ding
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Fujun Wang
- Department of Endocrinology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yue Zhao
- Department of Gastroenterology and Hepatology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Jianhua Wu
- Animal Center, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhanjun Guo
- Department of Rheumatology and Immunology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
32
|
Zhang L, Yang M, Zhang Y, Lan J, Chen Q. Unraveling the mechanisms of irAEs in endometrial cancer immunotherapy: insights from FAERS and scRNA-seq data. Sci Rep 2025; 15:18645. [PMID: 40436981 PMCID: PMC12119918 DOI: 10.1038/s41598-025-02723-3] [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: 02/18/2025] [Accepted: 05/15/2025] [Indexed: 06/01/2025] Open
Abstract
Endometrial cancer (EC) is one of the most common malignancies in women. In recent years, immunotherapy has gradually become a significant treatment option. However, the mechanisms underlying immune checkpoint inhibitor (ICI)-related Adverse Events (AEs) remain poorly understood, posing significant challenges for optimizing clinical treatment strategies. This study aims to integrate the FAERS database and single-cell transcriptomic data to investigate potential mechanisms underlying PD-1 inhibitor-related AEs in EC immunotherapy, with a focus on exploring the PD-1-associated cell communication network and its potential compensatory activation pathways. Data related to AEs were extracted from the FAERS database. Disproportionality analyses, including Reporting Odds Ratio (ROR), Proportional Reporting Ratio (PRR), Bayesian Confidence Propagation Neural Network (BCPNN), and Multi-item Gamma Poisson Shrinker (MGPS), were used to quantify signals of immune-related AEs (irAEs) associated with ICIs. We compared the occurrence timing and characteristics of AEs across different drugs. Subsequently, scRNA-seq was performed to analyze the tumor microenvironment of EC, focusing on PD-1-high expressing cell populations. Cell Communication was analyzed and key receptor-ligand pairs were identified. From Q1 2004 to Q3 2024, 21,838,627 drug-related reports were retrieved from FAERS, including 2,202 related to ICIs. ICI-associated irAEs involved 26 organ systems, with general disorders, gastrointestinal disorders, and injury/poisoning as the top System Organ Class (SOC). Fatigue, product use issues, and diarrhea were the most reported Preferred Terms (PTs). PD-1 inhibitors were associated with faster onset of AEs compared to PD-L1 inhibitors and Weibull modeling indicated an early failure-type AE pattern for both treatments. Single-cell analysis further demonstrated that PD-1 was highly expressed in CD8 + cytotoxic T cells and Tfh cells, which communicated with other cells within the tumor microenvironment through key receptor-ligand pairs such as CXCL12-CXCR4 and CXCL16-CXCR6. These findings suggested that PD-1 inhibitors may induce AEs through compensatory activation of the CXCR4 and CXCR6 pathways. This study suggested that PD-1 inhibitors may contribute to irAEs in EC, potentially through compensatory activation of the CXCR4 and CXCR6 pathways. By integrating FAERS and scRNA-seq data, key receptor-ligand interactions were identified, providing preliminary insights that could inform future efforts to optimize immunotherapy efficacy and mitigate AEs. However, further validation through clinical studies and mechanistic research is needed to confirm these findings.
Collapse
Affiliation(s)
- Lu Zhang
- Laboratory of Research and Diagnosis of Gynecological Diseases of Xiamen City, Clinical Medical Research Center for Obstetrics and Gynecology Diseases of Fujian Province, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China
| | - Mengjie Yang
- Laboratory of Research and Diagnosis of Gynecological Diseases of Xiamen City, Clinical Medical Research Center for Obstetrics and Gynecology Diseases of Fujian Province, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China
| | - Yiqian Zhang
- Laboratory of Research and Diagnosis of Gynecological Diseases of Xiamen City, Clinical Medical Research Center for Obstetrics and Gynecology Diseases of Fujian Province, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China
| | - Jianfa Lan
- Laboratory of Research and Diagnosis of Gynecological Diseases of Xiamen City, Clinical Medical Research Center for Obstetrics and Gynecology Diseases of Fujian Province, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| | - Qionghua Chen
- Laboratory of Research and Diagnosis of Gynecological Diseases of Xiamen City, Clinical Medical Research Center for Obstetrics and Gynecology Diseases of Fujian Province, Department of Obstetrics and Gynecology, the First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
33
|
Luo T, Guo J, Xi J, Luo X, Fu Z, Chen W, Huang D, Chen K, Xiao Q, Wei S, Wang Y, Du H, Liu L, Cai S, Dong H. Development and validation of a CT-based radiomics machine learning model for differentiating immune-related interstitial pneumonia. Int Immunopharmacol 2025; 156:114681. [PMID: 40262251 DOI: 10.1016/j.intimp.2025.114681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/20/2025] [Accepted: 04/13/2025] [Indexed: 04/24/2025]
Abstract
INTRODUCTION Immune checkpoint inhibitor-related interstitial pneumonia (CIP) poses a diagnostic challenge due to its radiographic similarity to other pneumonias. We developed a non-invasive model using CT imaging to differentiate CIP from other pneumonias (OTP). METHODS We analyzed CIP and OTP patients after the immunotherapy from five medical centers between 2020 and 2023, and randomly divided into training and validation in 7:3. A radiomics model was developed using random forest analysis. A new model was then built by combining independent risk factors for CIP. The models were evaluated using ROC, calibration, and decision curve analysis. RESULTS A total of 238 patients with pneumonia following immunotherapy were included, with 116 CIP and 122 OTP. After random allocation, the training cohort included 166 patients, and the validation included 72 patients. A radiomics model composed of 11 radiomic features was established using the random forest method, with an AUC of 0.833 for the training cohort and 0.821 for the validation. Univariate and multivariate logistic regression analysis revealed significant differences in smoking history, radiotherapy history, and radiomics score between CIP and OTP (p < 0.05). A new model was constructed based on these three factors and a nomogram was drawn. This model showed good calibration and net benefit in both the training and validation cohorts, with AUCs of 0.872 and 0.860, respectively. CONCLUSION Using the random forest method of machine learning, we successfully constructed a CT-based radiomics CIP differential diagnostic model that can accurately, non-invasively, and rapidly provide clinicians with etiological support for pneumonia diagnosis.
Collapse
Affiliation(s)
- Tingyue Luo
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jingze Guo
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Junjie Xi
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoyu Luo
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Zeyu Fu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Weisheng Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Danhui Huang
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Kaijun Chen
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Qiang Xiao
- Pulmonary and Critical Care Medicine, Shunde Hospital, Southern Medical University, Foshan, Guangdong, China
| | - Shuquan Wei
- Department of Pulmonary and Critical Care Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangdong, China
| | - Yan Wang
- Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| | - Huijuan Du
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Laiyu Liu
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China..
| | - Shaoxi Cai
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China..
| | - Hangming Dong
- Chronic Airways Diseases Laboratory, Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China..
| |
Collapse
|
34
|
Ke ZB, Chen JY, Xue YT, Lin B, Huang Q, Huang XY, Chen DN, Chen SH, Ye XJ, Zheng QS, Wei Y, Xue XY, Xu N. Mechanical signal modulates prostate cancer immune escape by USP8-mediated ubiquitination-dependent degradation of PD-L1 and MHC-1. Cell Death Dis 2025; 16:413. [PMID: 40410130 PMCID: PMC12102395 DOI: 10.1038/s41419-025-07736-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 04/30/2025] [Accepted: 05/13/2025] [Indexed: 05/25/2025]
Abstract
The tumor environment of prostate cancer (PCa) tissues of high Gleason score has been proved to be more immune suppressive and has higher extracellular matrix (ECM) stiffness, but whether ECM mechanical stiffness is the cause of higher ability of invasiveness and immune escape of PCa with high Gleason score remains uncertain. In this study, we showed that higher polyacrylamide hydrogels (PAAG) stiffness resulted in the progression and immune escape of PCa via integrin β1/FAK/YAP axis. The translocation of YAP into cell nucleus to bind to TEAD2 promoted the transcriptional activation of USP8. NBR1 could be ubiquitinated, and then degraded, via interacting with P62/SQSTM1 and through autophagy-lysosome pathway. Increased expression of USP8 promoted the abundance of NBR1 via K63-linked de-ubiquitination and PD-L1 via K48-linked de-ubiquitination in response to high PAAG stiffness. NBR1-mediated selective autophagy accelerated the degradation of MHC-1 of PCa. The USP8 inhibitor presented a potential application value in sensitizing immunotherapy of PCa. Taken together, we identified a USP8-mediated de-ubiquitination mechanism that involves in the process of high PAAG stiffness-mediated high expression of PD-L1 and low expression of MHC-1 of PCa cells, which provided a rationale of immunotherapy sensitization of PCa via USP8 inhibition.
Collapse
Affiliation(s)
- Zhi-Bin Ke
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Jia-Yin Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yu-Ting Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Bin Lin
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Qi Huang
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xu-Yun Huang
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Dong-Ning Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Shao-Hao Chen
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xiao-Jian Ye
- Department of Ultrasonography, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Qing-Shui Zheng
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yong Wei
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Xue-Yi Xue
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| | - Ning Xu
- Department of Urology, Urology Research Institute, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
- Department of Urology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
- Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China.
| |
Collapse
|
35
|
Shi Y, Liu X, Liu A, Fang J, Meng Q, Ding C, Ai B, Gu Y, Zhang C, Zhou C, Wang Y, Shui Y, Yu S, Zhang D, Liu J, Zhang H, Zhou Q, Gao X, Chen M, Zhao J, Zhong W, Xu Y, Wang M. Programmed death-ligand 1 tumor proportion score in predicting the safety and efficacy of PD-1/PD-L1 antibody-based therapy in patients with advanced non-small cell lung cancer: A retrospective, multicenter, observational study. Chin Med J (Engl) 2025:00029330-990000000-01563. [PMID: 40413619 DOI: 10.1097/cm9.0000000000003620] [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: 06/13/2024] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND This study aimed to investigate the safety and efficacy of programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) antibody-based therapy in treating patients with advanced non-small cell lung cancer (NSCLC) according to different PD-L1 expression statuses in a real-world setting. METHODS This retrospective, multicenter, observational study enrolled adult patients who received PD-1/PD-L1 antibody-based therapy in China and met the following criteria: (1) had pathologically confirmed, unresectable stage III-IV NSCLC; (2) had a baseline PD-L1 tumor proportion score (TPS); and (3) had confirmed efficacy evaluation results after PD-1/PD-L1 treatment. Logistic regression, Kaplan-Meier analysis, and Cox regression were used to assess the progression-free survival (PFS), overall survival (OS), and immune-related adverse events (irAEs) as appropriate. RESULTS A total of 409 patients, 65.0% (n = 266) with a positive PD-L1 TPS (≥1%) and 32.8% (n = 134) with PD-L1 TPS ≥50%, were included in this study. Cox regression confirmed that patients with a PD-L1 TPS ≥1% had significantly improved PFS (hazard ratio [HR] 0.747, 95% confidence interval [CI] 0.573-0.975, P = 0.032). A total of 160 (39.1%) patients experienced 206 irAEs, and 27 (6.6%) patients experienced 31 grade 3-5 irAEs. The organs most frequently associated with irAEs were the skin (52/409, 12.7%), thyroid (40/409, 9.8%), and lung (34/409, 8.3%). Multivariate logistic regression revealed that a PD-L1 TPS ≥1% (odds ratio [OR] 1.713, 95% CI 1.054-2.784, P = 0.030) was an independent risk factor for irAEs. Other risk factors for irAEs included pretreatment absolute lymphocyte count >2.5 × 109/L (OR 3.772, 95% CI 1.377-10.329, P = 0.010) and pretreatment absolute eosinophil count >0.2 × 109/L (OR 2.006, 95% CI 1.219-3.302, P = 0.006). Moreover, patients who developed irAEs demonstrated improved PFS (13.7 months vs. 8.4 months, P <0.001) and OS (28.0 months vs. 18.0 months, P = 0.007) compared with patients without irAEs. CONCLUSIONS A positive PD-L1 TPS (≥1%) was associated with improved PFS and an increased risk of irAEs in a real-world setting. The onset of irAEs was associated with improved PFS and OS in patients with advanced NSCLC receiving PD-1/PD-L1-based therapy.
Collapse
Affiliation(s)
- Yuequan Shi
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
- Department of Internal Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Xiaoyan Liu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Anwen Liu
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi 330006, China
| | - Jian Fang
- Department of Thoracic Oncology II, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Qingwei Meng
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang 150081, China
| | - Cuimin Ding
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei 050033, China
| | - Bin Ai
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing 100005, China
| | - Yangchun Gu
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing 100191, China
| | - Cuiying Zhang
- Department of Medical Oncology, Cancer Center, People's Hospital, Hohhot, Inner Mongolia Autonomous Region 750306, China
| | - Chengzhi Zhou
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Disease, National Clinical Research Center of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Yan Wang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100021, China
| | - Yongjie Shui
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Siyuan Yu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Dongming Zhang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Jia Liu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Haoran Zhang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Qing Zhou
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Xiaoxing Gao
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Minjiang Chen
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Jing Zhao
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Wei Zhong
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Yan Xu
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Mengzhao Wang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
36
|
Kim GD, Shin SI, Sun P, Lee JE, Chung C, Kang YE, Kang DH, Park J. Single-cell RNA sequencing of baseline PBMCs predicts ICI efficacy and irAE severity in patients with NSCLC. J Immunother Cancer 2025; 13:e011636. [PMID: 40404203 PMCID: PMC12097017 DOI: 10.1136/jitc-2025-011636] [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: 01/20/2025] [Accepted: 04/29/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have transformed treatment and have provided significant clinical benefits and durable responses for patients with advanced non-small cell lung cancer (NSCLC). However, only a small percentage of patients respond to ICI treatment, and immune-related adverse events (irAEs) leading to treatment discontinuation remain challenging. Despite the recognized need for biomarkers to predict both the efficacy of ICIs and the risk of irAEs, such biomarkers are yet to be clearly identified. METHODS In this study, we performed single-cell RNA sequencing (scRNA-seq) of peripheral blood mononuclear cells (PBMCs) from 33 patients with NSCLC before ICIs treatment. To validate our findings, we reanalyzed public scRNA-seq data, conducted a cytometric bead array (CBA), and supported our findings with T-cell receptor sequencing. RESULTS While the immune response was more pronounced in patients with a favorable prognosis, the hypoxic pathway was more prominent in patients with primary resistance. Lymphocytes such as CD8 T cells, CD4 T cells, and natural killer cells were primarily involved in these pathways, with PRF1 and GZMB expression showing strong associations with favorable prognosis. In contrast, irAEs were mainly linked to myeloid cells, such as monocytes and macrophages. As irAE severity increased, inflammation and the TNF-NFKB1 pathway were more prominent. Specifically, increased expression of IL1B, CXCL8, and CXCL2 in monocytes and TNF in macrophages was closely associated with severe irAE through involvement in these pathways.Notably, the increase of PRF1 and GZMB expression showed a close association with both a favorable prognosis and a reduced severity of irAE, which was validated through CBA analysis. Moreover, the expression of these key markers varied according to prognosis and irAE severity regardless of patient background, such as programmed death-ligand 1 expression levels, tumor histology, or prior treatment regimens. CONCLUSIONS This study identified biological pathways and key biomarkers associated with ICI prognosis and irAE severity using PBMC samples before treatment. These findings provide a foundation for improved therapeutic strategies that enhance clinical outcomes while minimizing ICI treatment-associated risks.
Collapse
Affiliation(s)
- Gyeong Dae Kim
- Life Science, Gwangju Institute of Science and Technology, Gwangju, Buk-gu, Korea (the Republic of)
| | - So-I Shin
- Life Science, Gwangju Institute of Science and Technology, Gwangju, Buk-gu, Korea (the Republic of)
| | - Pureum Sun
- Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Jeong Eun Lee
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Chaeuk Chung
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Yea Eun Kang
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Da Hyun Kang
- Department of Internal Medicine, College of Medicine, Chungnam National University, Daejeon, South Korea
| | - Jihwan Park
- Life Science, Gwangju Institute of Science and Technology, Gwangju, Buk-gu, Korea (the Republic of)
| |
Collapse
|
37
|
Liu L, Wei Y, Chen Q, Liang H. Unexplained lymphadenopathy following adjuvant immunotherapy in a non-small cell lung cancer patient: a literature review and a rare case report. Immunopharmacol Immunotoxicol 2025:1-5. [PMID: 40400058 DOI: 10.1080/08923973.2025.2507130] [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: 02/27/2025] [Accepted: 05/10/2025] [Indexed: 05/23/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have shown significant advantages in the treatment of lung cancer. Several studies have reported immune-related adverse events (irAEs) induced by ICIs. However, in clinical practice, irAEs occasionally cause lymphadenopathy, which can be mistaken for tumor progression, making it more challenging to accurately assess the patient's condition. CASE PRESENTATION This research report documents a rare clinical case of a patient with early-stage non-small cell lung cancer (NSCLC) who developed systemic lymphadenopathy during treatment with a PD-1 ICI following surgical resection. The patient developed widespread lymphadenopathy during postoperative PD-1 antibody maintenance therapy, accompanied by a series of irAEs, including persistent low cortisol levels, sluggish responses, memory loss, and stiffness in distal limbs and shoulder joints. Given the clinical presentation, the possibility of lymph node metastasis from lung cancer could not be entirely excluded. However, lymph node biopsy revealed reactive hyperplasia. After receiving corticosteroid treatment, the enlarged lymph nodes significantly reduced in size, and the associated low cortisol symptoms disappeared. During subsequent follow-up, the patient showed significant improvement and maintained a relatively stable condition. RESULTS In cases of postoperative generalized lymphadenopathy in NSCLC patients, if tumor recurrence is suspected, careful consideration is needed, especially in the era of ICI therapy. Postoperative PD-1 antibody maintenance therapy may induce reactive lymphadenopathy, including mediastinal lymph nodes. It is hypothesized that PD-1 antibodies cause T-cell activation in the lymph nodes.
Collapse
Affiliation(s)
- Lila Liu
- Radiation Oncology, Zhongshan City People's Hospital, Guangdong Medical University, Zhanjiang, China
| | - Ying Wei
- Department of Pulmonary Oncology, Zhongshan City People's Hospital, Zhongshan, China
| | - Qijiu Chen
- Department of Pulmonary Oncology, Zhongshan City People's Hospital, Zhongshan, China
| | - Hanlin Liang
- Department of Pulmonary Oncology, Zhongshan City People's Hospital, Zhongshan, China
| |
Collapse
|
38
|
Enayah M, Al-Aqtash T. Pembrolizumab-Associated Multiorgan Sarcoid-Like Reaction: A Case Report and Review of Literature. Case Rep Med 2025; 2025:9915002. [PMID: 40432711 PMCID: PMC12116203 DOI: 10.1155/carm/9915002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Immune checkpoints are molecules that serve to augment or inhibit the immune response. The treatment landscape for numerous tumors now relies significantly on immune checkpoint inhibitors (ICIs). Pembrolizumab, a subset of ICIs specifically focused on the programmed cell death 1 (PD-1) molecule. By blocking PD-1, these inhibitors enhance the ability of the immune system to fight cancer cells. Although PD-1 inhibitors are critical in cancer treatment, their use can be associated with immune-related adverse events, such as ICI-related sarcoid-like reaction. Case Presentation: This report describes a 49-year-old female patient with stage IIIA breast cancer breast cancer who developed ICI-related sarcoid-like reaction after starting a neoadjuvant chemoimmunotherapy regimen that included Pembrolizumab. After 4 months of ongoing treatment, she started having significant nausea and vomiting. Computed tomography (CT) scans performed during hospitalization revealed multiple pathologically enlarged thoracic lymph nodes, suspicious for disease progression. Initially, the laboratory workup and cultures were unrevealing. However, esophagogastroduodenoscopy and bronchoscopy were performed, showing noncaseating granulomas in both the stomach and thoracic lymph node biopsy samples. The patient was diagnosed with a sarcoidosis-like reaction to Pembrolizumab. Notably, her symptoms rapidly improved upon initiating systemic corticosteroids. Follow-up CT scan showed a significant improvement in lymphadenopathy after discontinuing Pembrolizumab. Conclusion: This case emphasizes the significance of acknowledging sarcoid-like reactions as possible adverse effects of Pembrolizumab. Given the rising utilization of PD-1 inhibitors, it becomes imperative to be mindful of such adverse events. This awareness helps avoid misdiagnosing disease progression and aids in making informed decisions about ongoing treatment with ICIs.
Collapse
Affiliation(s)
- Majd Enayah
- School of Medicine, The University of Jordan, Amman, Jordan
| | | |
Collapse
|
39
|
Bogin V. We Need to Talk About Quality of Life with Cancer Patients: Primum Non Nocere in Oncology. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:918. [PMID: 40428876 PMCID: PMC12113248 DOI: 10.3390/medicina61050918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 05/05/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025]
Abstract
The Hippocratic principle primum non nocere, or "first, do no harm", serves as a vital lens through which to re-evaluate modern oncology practices. While recent advances such as immunotherapy, targeted agents, and precision medicine have transformed cancer care, these treatments are not without risk. Even with improved tolerability, they may still lead to substantial toxicities, particularly in frail patients with advanced cancer. The pursuit of survival often overshadows the patient's quality of life, with aggressive interventions frequently continuing beyond the point of meaningful benefit. This perspective article argues for a more individualized and ethically grounded approach to cancer treatment, emphasizing the careful assessment of each patient's clinical status, values, and goals. By integrating geriatric and palliative assessments, improving shared decision making, and moving away from a default treatment-at-all-costs mindset, clinicians can better align care with what truly matters to patients. Honoring primum non nocere in oncology means not only extending life when appropriate but ensuring that life remains worth living.
Collapse
Affiliation(s)
- Vlad Bogin
- Suite 300, Preston Commons Center, 8117 Preston Road, Dallas, TX 75225, USA
| |
Collapse
|
40
|
Keshavarz Sadegh R, Saleki K, Rezaei N. Immune checkpoint inhibitor (ICI) therapy in central nervous system cancers: State-of-the-art and future outlook. Int Immunopharmacol 2025; 159:114837. [PMID: 40394797 DOI: 10.1016/j.intimp.2025.114837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/28/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Invasive central nervous system (CNS) cancers are an area where the development of breakthrough therapies is urgently needed. For instance, conditions such as glioblastoma multiforme (GBM) are associated with poor clinical prognosis, with the majority of trials offering no improvement to marginally enhanced survival. Unleashing the potential of targeting the immune system in CNS cancers has gained attention in recent years. Inhibition of immune checkpoints such as CTLA-4, PD-1/PD-L1, TIM-3, and LAG-3 has been attempted in recent trials. While potentially offering a notable edge over other immunotherapies, multi-organ adverse events have been found with the administration of immune checkpoint inhibitors (ICIs). The present review captures the state-of-the-art evidence on ICI treatments in different CNS cancers. Also, we discuss the value of combinational therapies involving ICIs as well as next-generation therapeutics such as bispecific antibodies targeting PD-1/LAG-3/TIM-3 and CRISPR-Cas9-edited PD-1-knock-out checkpoint-resistant CAR T-cells.
Collapse
Affiliation(s)
- Roghaye Keshavarz Sadegh
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; USERN MUBabol Office, Universal Scientific Education and Research Network (USERN), Babol, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
41
|
Duan L, Liu G, Huang Z, Chen R, Mo D, Xia Y, Hu J, He M. Development and validation of a nomogram for differentiating immune checkpoint inhibitor-related pneumonitis from pneumonia in patients undergoing immunochemotherapy: a multicenter, real-world, retrospective study. Front Immunol 2025; 16:1495450. [PMID: 40458408 PMCID: PMC12127321 DOI: 10.3389/fimmu.2025.1495450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 04/29/2025] [Indexed: 06/19/2025] Open
Abstract
Background Immune Checkpoint Inhibitor-related Pneumonitis (CIP) exhibits high morbidity and mortality rates in the real world, often coexisting with pneumonia, particularly after immunochemotherapy. We aimed to develop and validate a non-invasive nomogram for differentiating CIP from pneumonia in patients undergoing immunochemotherapy. Methods This study encompassed 237 patients from three hospitals. A multivariate logistic regression analysis was conducted to identify risk factors for CIP. Utilizing the random forest machine learning method, optimal development and validation cohort allocation ratios (in a ratio of 8:2) were determined for the predictive model. The performance of the nomogram was evaluated using calibration, the area under the receiver operating characteristic curve (AUC), and decision curve analysis (DCA). Subsequently respiratory pathogens, management, and outcomes were compared between CIP and No CIP cases. Results Among the 237 patients, 104 were diagnosed with CIP, and 133 were no CIP but pneumonia(No CIP). Smoking status, prior chronic obstructive pulmonary disease (COPD), ground glass opacities, non-specific interstitial pneumonitis, Neutrophil to Lymphocyte Ratio (NLR), pleural effusions, and Oxygen Partial Pressure (PaO2) emerged as non-invasive independent predictors of CIP. The nomogram exhibited good discrimination for both the development and validation cohorts, with AUC values of 0.817 (95% CI, 0.754-0.879) and 0.913 (95% CI, 0.826-0.999), respectively. The calibration curves demonstrated good fit for both the development and validation cohort, as evidenced by the Hosmer-Lemeshow tests (χ² = 3.939, p = 0.863 and χ² = 8.117, p = 0.422, respectively). DCA further highlighted their clinical utility. In CIP patients, the use of gamma globulin/albumin and glucocorticoids was significantly higher than in No CIP patients (39.4% vs 23.3%, p = 0.007; 79.8% vs 12.8%, p < 0.0001, respectively). The proportion of patients requiring mechanical ventilation was also significantly higher in the CIP compared to the No CIP group (21.2% vs 11.3%, p = 0.038). Conclusion The nomogram offers a non-invasive approach to differentiate CIP from pneumonia associated with immunochemotherapy, potentially facilitating early intervention and informed treatment decisions.
Collapse
Affiliation(s)
- Linli Duan
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Guanglu Liu
- Algorithm Development Department 1, GRGBanking Equipment Company Ltd., Guangzhou, China
| | - Zijie Huang
- Thoracic Surgery Department, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Rong Chen
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Di Mo
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuxiao Xia
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiazhu Hu
- Department of Oncology, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mengzhang He
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
42
|
Zheng H, Chen Y, Luo W, Han S, Sun M, Lin M, Wu C, Gao L, Xie T, Kong N. Integration of active ingredients from traditional Chinese medicine with nano-delivery systems for tumor immunotherapy. J Nanobiotechnology 2025; 23:357. [PMID: 40382641 PMCID: PMC12085060 DOI: 10.1186/s12951-025-03378-y] [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: 01/07/2025] [Accepted: 04/07/2025] [Indexed: 05/20/2025] Open
Abstract
Tumor immune escape presents a significant challenge in cancer treatment, characterized by the upregulation of immune inhibitory molecules and dysfunction of immune cells. Tumor immunotherapy seeks to restore normal anti-tumor immune responses to control and eliminate tumors effectively. The active ingredients of traditional Chinese medicine (TCM) demonstrate a variety of anti-tumor activities and mechanisms, including the modulation of immune cell functions and inhibiting tumor-related suppressive factors, thereby potentially enhancing anti-tumor immune responses. Furthermore, nano-delivery systems function as efficient carriers to enhance the bioavailability and targeted delivery of TCM active ingredients, augmenting therapeutic efficacy. This review comprehensively analyzes the impact of TCM active ingredients on the immune system and explores the synergistic application of nano-delivery systems in combination with TCM active ingredients for enhancing tumor immunotherapy.
Collapse
Affiliation(s)
- Hao Zheng
- Department of Neurology, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, Fujian, China
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Yiquan Chen
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, Zhejiang, China
| | - Wei Luo
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Shiqi Han
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, Zhejiang, China
| | - Mengjuan Sun
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, Zhejiang, China
| | - Min Lin
- Department of Neurology, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, Fujian, China
| | - Chenghan Wu
- Department of Neurology, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, Fujian, China
| | - Lili Gao
- Department of Neurology, The Second Affiliated Hospital of Fujian Traditional Chinese Medical University, Fuzhou, Fujian, China.
| | - Tian Xie
- College of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China.
| | - Na Kong
- Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 311121, Zhejiang, China.
| |
Collapse
|
43
|
Lau G, Sangro B, Cheng AL, Kudo M, Kelley RK, Tak WY, Gasbarrini A, Reig M, Lim HY, Tougeron D, De Toni EN, Tam VC, Mody K, Gong J, Crysler OV, Sukeepaisarnjaroen W, Lipatov O, Morimoto M, Archambeaud I, Burgio V, Phuong LTT, Chao Y, Peron JM, Berres ML, Ko YJ, Ran D, Makowsky M, Negro A, Abou-Alfa GK. Immune-mediated adverse events and overall survival with tremelimumab plus durvalumab and durvalumab monotherapy in unresectable HCC: HIMALAYA phase III randomized clinical trial. Hepatology 2025:01515467-990000000-01286. [PMID: 40384092 DOI: 10.1097/hep.0000000000001385] [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: 11/14/2024] [Accepted: 04/07/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND AND AIMS In the global phase III HIMALAYA study in unresectable HCC, STRIDE significantly improved overall survival (OS) versus sorafenib; durvalumab was noninferior to sorafenib. Immune checkpoint inhibitor studies have shown an association between the occurrence of immune-mediated adverse events (imAEs) and improved OS. We assessed potential associations between the occurrence of imAEs and OS, and temporal patterns of imAEs, in HIMALAYA. APPROACH AND RESULTS OS in participants who did and did not experience imAEs and the frequency and timing of imAEs were assessed for STRIDE and durvalumab in the safety analysis set of HIMALAYA. imAEs occurred in 139/388 (35.8%) and 64/388 (16.5%) participants with STRIDE and durvalumab, respectively; most were low grade. OS HRs (95% CI) in participants who experienced imAEs versus those who did not were 0.73 (0.56-0.95) for STRIDE and 1.14 (0.82-1.57) for durvalumab. The 36-month OS rate (95% CI) for STRIDE was 36.2% (28.1-46.7) and 27.7% (22.4-34.2) in participants who did and did not experience imAEs, respectively. The most common imAE category with STRIDE was endocrine events (16.5%). Most imAEs occurred ≤3 months after treatment initiation. CONCLUSIONS Participants who experienced imAEs with STRIDE had a numerical improvement in OS versus those who did not, which was not observed for durvalumab. Long-term OS with STRIDE was observed regardless of imAEs. Most imAEs were low grade, manageable, and occurred in the first 3 months after treatment initiation. Results continue to support the benefits of STRIDE in a diverse population that reflects unresectable HCC globally.
Collapse
Affiliation(s)
- George Lau
- Humanity and Health Clinical Trial Center, Humanity and Health Medical Group, Hong Kong Special Administrative Region, China
| | - Bruno Sangro
- Liver Unit and HPB Oncology Area, Clínica Universidad de Navarra and CIBEREHD, Pamplona, Spain
| | - Ann-Lii Cheng
- Department of Oncology, National Taiwan University Cancer Center and National Taiwan University Hospital, Taipei, Taiwan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Robin Kate Kelley
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Won Young Tak
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Antonio Gasbarrini
- Fondazione Policlinico Universitario Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Maria Reig
- Barcelona Clinic Liver Cancer (BCLC), Liver Unit, Hospital Clinic de Barcelona, IDIBAPS, CIBEREHD, University of Barcelona, Barcelona, Spain
| | - Ho Yeong Lim
- Samsung Medical Center, Sungkyunkwan University, Seoul, Republic of Korea
| | - David Tougeron
- Department of Gastroenterology and Hepatology, Poitiers University Hospital, Poitiers, France
| | - Enrico N De Toni
- Department of Medicine II, University Hospital, LMU Munich, Munich, Germany
| | - Vincent C Tam
- Department of Oncology, Tom Baker Cancer Centre, University of Calgary, Calgary, Alberta, Canada
| | - Kabir Mody
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Jacksonville, Florida, USA
| | - Jun Gong
- Department of Medicine, Division of Hematology and Oncology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Oxana V Crysler
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Wattana Sukeepaisarnjaroen
- Department of Medicine, Faculty of Medicine, Khon Kaen University, Srinagarind Hospital, Khon Kaen, Thailand
| | - Oleg Lipatov
- Department of Oncology, Republican Clinical Oncology Dispensary, Ufa, Russia
| | - Manabu Morimoto
- Hepatobiliary and Pancreatic Oncology, Kanagawa Cancer Center, Yokohama, Japan
| | - Isabelle Archambeaud
- Hépato-Gastro-Entérologie et Assistance Nutritionnelle, Institut des Maladies de l'Appareil Digestif (IMAD), Nantes Université, CHU Nantes, Nantes, France
| | - Valentina Burgio
- Department of Medical Oncology, Vita-Salute University, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Yee Chao
- Department of Oncology, Division of Medical Oncology, Center for Immuno-oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | - Marie-Luise Berres
- Clinic of Gastroenterology, Metabolic Diseases and Internal Medicine Intensive Care, University Hospital RWTH Aachen, Aachen, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf (CIO ABCD), Aachen, Germany
| | - Yoo-Joung Ko
- St Michael's Hospital, Unity Health Toronto, Toronto, Ontario, Canada
| | - Di Ran
- AstraZeneca, Gaithersburg, Maryland, USA
| | - Mallory Makowsky
- AstraZeneca, Gaithersburg, Maryland, USA
- Department of Clinical Development, Erasca, Inc., San Diego, California, USA
| | | | - Ghassan K Abou-Alfa
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Medical College, Cornell University, New York, New York, USA
- Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
44
|
Mao XM, Wang WH. Vitiligo-like rash in a patient with lung cancer caused by sintilimab: A case report. World J Clin Cases 2025; 13:101981. [PMID: 40385299 PMCID: PMC11752433 DOI: 10.12998/wjcc.v13.i14.101981] [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: 10/03/2024] [Revised: 12/14/2024] [Accepted: 12/27/2024] [Indexed: 01/15/2025] Open
Abstract
BACKGROUND This article discusses a case involving a 63-year-old man with non-small cell lung cancer, who was treated with a combination of chemotherapy and immunotherapy. The patient was treated with five cycles of chemotherapy (pemetrexed and carboplatin) combined with sintilimab, a programmed death 1 inhibitor. CASE SUMMARY After the fifth cycle of treatment, the patient developed skin itching and a vitiligo-like rash, which are known side effects of immunotherapy. Despite dermatological consultation and treatment with topical corticosteroids, the rash worsened while the itching subsided. The patient continued with the treatment, and after 15 cycles, the tumor showed a response with a reduction in size. The vitiligo-like rash increased, but the antitumor treatment remained effective. CONCLUSION The case highlights the use of immunotherapy in patients with non-small cell lung cancer and the potential side effect of vitiligo-like rash. The patient's tumor responded well to the treatment, and despite the skin reaction, the treatment was not discontinued due to its effectiveness. The article suggests that further studies are needed to understand the mechanism behind vitiligo in patients with lung cancer receiving immune checkpoint inhibitors and whether the development of vitiligo-like rash after immune checkpoint inhibitor therapy is associated with improved prognosis. The case also underscores the importance of managing immune-related adverse events in the context of effective antitumor treatment.
Collapse
Affiliation(s)
- Xiao-Ming Mao
- Department of Respiratory and Critical Care Medicine, Jiangshan People’s Hospital, Jiangshan 324100, Zhejiang Province, China
| | - Wei-Hua Wang
- Department of Respiratory and Critical Care Medicine, Jiangshan People’s Hospital, Jiangshan 324100, Zhejiang Province, China
| |
Collapse
|
45
|
Li J, Tang Z, Zhang L, Tan N, Liu W. Genetic Causal Relationship Between Systemic Lupus Erythematosus and Malignant Tumors of the Female Reproductive System: A GWAS Analysis in European Populations. Hum Mutat 2025; 2025:7447886. [PMID: 40406544 PMCID: PMC12097853 DOI: 10.1155/humu/7447886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Accepted: 04/29/2025] [Indexed: 05/26/2025]
Abstract
Background: Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease that primarily affects women of reproductive age. Existing studies have demonstrated complex associations between SLE and various diseases, but its genetic relationship with malignant tumors of the female reproductive system has not been fully elucidated. This study is aimed at exploring the potential genetic associations and shared molecular basis between SLE and female reproductive system malignancies using genome-wide association studies (GWASs) and cross-trait analysis. Methods: We selected genetic variants significantly associated with SLE (p < 5 × 10-8) from large-scale GWAS databases as genetic instruments and applied various statistical methods to analyze the associations between SLE and cervical cancer, endometrial cancer, ovarian cancer, vulvar cancer, vaginal cancer, and uterine cancer. The primary analysis was conducted using inverse variance weighting (IVW), supplemented by Egger regression, weighted median, and weighted mode methods. To control for potential confounders, we performed multivariable analysis while including BMI, estradiol, and CRP as covariates. Additionally, cross-trait analysis using the association analysis based on subset (ASSET) method was employed to identify shared genetic variants and their effect directions between SLE and uterine cancer. Results: Genetic association analysis showed a significant negative association between SLE and endometrial cancer (OR = 0.972, 95% CI [0.946-0.998], p = 0.038), suggesting that SLE may be associated with a reduced risk of endometrial cancer. For uterine cancer, the weighted median method also indicated a marginally significant negative association (OR = 0.955, 95% CI [0.912-1.000], p = 0.049). Multivariable analysis further confirmed that the protective association between SLE and endometrial cancer remained significant after controlling for BMI, estradiol, and CRP (OR = 0.96, 95% CI [0.93-0.99], p = 0.014). However, no significant association was observed between SLE and cervical cancer, ovarian cancer, vulvar cancer, or vaginal cancer. Cross-trait analysis identified 193 shared genetic variants between SLE and endometrial cancer and 71 shared variants between SLE and uterine cancer, with rs2442719 and rs3131004 showing consistent effect directions in both comparisons. Conclusion: This study provides genetic epidemiological evidence suggesting that SLE may have a protective effect against endometrial and uterine cancers and identifies potential shared genetic bases. These findings offer new insights into the relationship between SLE and gynecological tumors and may provide references for the prevention and treatment of related diseases.
Collapse
Affiliation(s)
- Jianbin Li
- Department of Rheumatism and Immunity, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Department of Rheumatism and Immunity, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Zhuo Tang
- Department of Rheumatism and Immunity, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Department of Rheumatism and Immunity, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Lei Zhang
- Department of Rheumatism and Immunity, Tianjin Academy of Traditional Chinese Medicine, Tianjin, China
| | - Ning Tan
- Department of Rheumatism and Immunity, Shenzhen Nanshan People's Hospital, Shenzhen, China
| | - Wei Liu
- Department of Rheumatism and Immunity, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Department of Rheumatism and Immunity, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
46
|
Monaca F, Gomez-Randulfe I, Parreira AS, Longo V, Galetta D, Pilotto S, Polidori S, Cantale O, Stefani A, Vita E, Taylor P, Gomes F, Cove-Smith L, Summers Y, Tortora G, Blackhall F, Novello S, Bria E, Califano R. Correlation between irAEs and survival outcomes in patients with ES-SCLC treated with first-line chemoimmunotherapy. Eur J Cancer 2025; 221:115435. [PMID: 40250285 DOI: 10.1016/j.ejca.2025.115435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/04/2025] [Accepted: 04/13/2025] [Indexed: 04/20/2025]
Abstract
INTRODUCTION Chemo-immunotherapy (CT-IO) has improved median overall survival (mOS) for patients with extensive-stage small cell lung cancer (ES-SCLC), but its association with immune-related adverse events (irAEs) remains unclear. While irAEs are often linked to better outcomes in other cancers, their prognostic value in ES-SCLC is not well understood. METHODS We conducted a retrospective analysis of 399 consecutive ES-SCLC patients treated with first-line CT-IO between January 2020 and September 2024 across five European centres. Demographic and clinical data were collected. The impact of irAEs on progression-free survival (PFS) and overall survival (OS) was assessed using time-dependent Cox regression. RESULTS The median follow-up was 15.0 months. The overall response rate was 80.3 %, with a median PFS of 6.0 months (95 % CI 5.7-6.3) and mOS of 10.4 months (95 % CI 9.2-11.6). IrAEs occurred in 30.6 % of patients, most commonly affecting the skin (11.0 %). The median time to onset of irAEs was 171 days. Patients with irAEs had significantly longer mPFS (10.8 vs. 5.3 months, p < 0.001) and mOS (18.8 vs. 7.6 months, p < 0.001) compared to those without. No significant difference was found between patients with grade ≥ 3 (n = 46) and < 3 irAEs (n = 76). Multivariate analysis confirmed that irAEs were associated with improved OS (HR 0.64; 95 % CI 0.51-0.80, p < 0.001) and showed a trend towards longer PFS (p = 0.028). CONCLUSION This is the largest retrospective study to demonstrate that irAEs are associated with improved clinical outcomes in ES-SCLC pts receiving 1 L CT-IO.
Collapse
Affiliation(s)
- Federico Monaca
- Università Cattolica del Sacro Cuore, Rome, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Igor Gomez-Randulfe
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Ana Sofia Parreira
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Vito Longo
- Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Domenico Galetta
- Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Sara Pilotto
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Sara Polidori
- Università Cattolica del Sacro Cuore, Rome, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ornella Cantale
- Department of Oncology, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Torino, Italy
| | - Alessio Stefani
- Università Cattolica del Sacro Cuore, Rome, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Emanuele Vita
- Università Cattolica del Sacro Cuore, Rome, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Paul Taylor
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Fabio Gomes
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Laura Cove-Smith
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Yvonne Summers
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Giampaolo Tortora
- Università Cattolica del Sacro Cuore, Rome, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Fiona Blackhall
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Silvia Novello
- Department of Oncology, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Torino, Italy
| | - Emilio Bria
- Università Cattolica del Sacro Cuore, Rome, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Ospedale Isola Tiberina - Gemelli Isola, Rome, Italy
| | - Raffaele Califano
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Division of Cancer Sciences, The University of Manchester, Manchester, UK.
| |
Collapse
|
47
|
Li S, Zhou X, Feng H, Huang K, Chen M, Lin M, Lin H, Deng Z, Chen Y, Liao W, Zhang Z, Chen J, Guan B, Su T, Feng Z, Shu G, Yu A, Pan Y, Fu L. Deciphering the Immunomodulatory Function of GSN + Inflammatory Cancer-Associated Fibroblasts in Renal Cell Carcinoma Immunotherapy: Insights From Pan-Cancer Single-Cell Landscape and Spatial Transcriptomics Analysis. Cell Prolif 2025:e70062. [PMID: 40375605 DOI: 10.1111/cpr.70062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/13/2025] [Accepted: 05/02/2025] [Indexed: 05/18/2025] Open
Abstract
The heterogeneity of cancer-associated fibroblasts (CAFs) could affect the response to immune checkpoint inhibitor (ICI) therapy. However, limited studies have investigated the role of inflammatory CAFs (iCAFs) in ICI therapy using pan-cancer single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics sequencing (ST-seq) analysis. We performed pan-cancer scRNA-seq and ST-seq analyses to identify the subtype of GSN+ iCAFs, exploring its spatial distribution characteristics in the context of ICI therapy. The pan-cancer scRNA-seq and bulk RNA-seq data are incorporated to develop the Caf.Sig model, which predicts ICI response based on CAF gene signatures and machine learning approaches. Comprehensive scRNA-seq analysis, along with in vivo and in vitro experiments, investigates the mechanisms by which GSN+ iCAFs influence ICI efficacy. The Caf.Sig model demonstrates well performances in predicting ICI therapy response in pan-cancer patients. A higher proportion of GSN+ iCAFs is observed in ICI non-responders compared to responders in the pan-cancer landscape and clear cell renal cell carcinoma (ccRCC). Using real-world immunotherapy data, the Caf.Sig model accurately predicts ICI response in pan-cancer, potentially linked to interactions between GSN+ iCAFs and CD8+ Tex cells. ST-seq analysis confirms that interactions and cellular distances between GSN+ iCAFs and CD8+ exhausted T (Tex) cells impact ICI efficacy. In a co-culture system of primary CAFs, primary tumour cells and CD8+ T cells, downregulation of GSN on CAFs drives CD8+ T cells towards a dysfunctional state in ccRCC. In a subcutaneously tumour-grafted mouse model, combining GSN overexpression with ICI treatment achieves optimal efficacy in ccRCC. Our study provides the Caf.Sig model as an outperforming approach for patient selection of ICI therapy, and advances our understanding of CAF biology and suggests potential therapeutic strategies for upregulating GSN in CAFs in cancer immunotherapy.
Collapse
Affiliation(s)
- Shan Li
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Uro-Oncology Institute of Central South University, Changsha, Hunan, China
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xinwei Zhou
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Haoqian Feng
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Kangbo Huang
- Department of Urology, Sun Yat-Sen University Cancer Center, Guangzhou, China
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Minyu Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Mingjie Lin
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hansen Lin
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zebing Deng
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Uro-Oncology Institute of Central South University, Changsha, Hunan, China
| | - Yuhang Chen
- Department of Geniturinary Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wuyuan Liao
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhengkun Zhang
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Jinwei Chen
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Bohong Guan
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Tian Su
- Department of Pediatric Intensive Care Unit (PICU), Guangdong Provincial People's Hospital Heyuan Hospital, Heyuan, Guangdong, China
| | - Zihao Feng
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Guannan Shu
- Department of Urology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou Institute of Pediatrics, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Anze Yu
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Yihui Pan
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Liangmin Fu
- Department of Urology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
- Uro-Oncology Institute of Central South University, Changsha, Hunan, China
- Department of Urology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, China
- Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Disease, Changsha, Hunan, China
| |
Collapse
|
48
|
Yamamoto T, Ito T, Suzuki T, Mizuno K, Yokoyama S, Yamamoto K, Imai N, Ishizu Y, Honda T, Kawashima H. Steroid initiation dose and duration of steroid reduction for immune checkpoint inhibitor-induced liver injury. Hepatol Res 2025. [PMID: 40369781 DOI: 10.1111/hepr.14200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/23/2025] [Accepted: 04/13/2025] [Indexed: 05/16/2025]
Abstract
AIM Treatment for severe immune checkpoint inhibitor (ICI)-induced liver injury (≥ Grade 3) requires prednisolone (PSL) administration and interruption of the underlying malignancy treatment. If the liver injury improves steadily, a lower initial dose of PSL is beneficial. We aimed to investigate the relationship between the initial dose of PSL and the response to PSL or the duration of PSL dose reduction. METHODS We retrospectively collected clinical data of patients treated with ICIs at Nagoya University Hospital between September 2014 and December 2023. Patients who received PSL for severe ICI-induced liver injury were divided according to the starting dose into group A (0.8 mg/kg/day) and group B (1.0 mg/kg/day). The time to improvement in liver injury and the reduction of the PSL dose to 10 mg/day were compared between the groups. RESULTS Overall, 1271 patients were treated with ICIs, of whom 80 experienced severe ICI-induced liver injury. Of the patients, 29 did not receive steroids, and five used PSL doses of <0.5 mg/kg/day. There were no significant differences in the baseline characteristics or laboratory data between the groups. The time to dose reduction to 10 mg/day PSL was significantly shorter in group A than in group B. The time to improvement in liver injury did not differ between the two groups. CONCLUSION In some patients with severe ICI-induced liver injury, a dose of 0.8 mg/kg/day of PSL is sufficient to achieve a therapeutic effect and shorten the time required to reduce the dose to 10 mg/day.
Collapse
Affiliation(s)
- Takafumi Yamamoto
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takanori Ito
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takaya Suzuki
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuyuki Mizuno
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Japan
| | - Shinya Yokoyama
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kenta Yamamoto
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Norihiro Imai
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoji Ishizu
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Honda
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
49
|
Tang Y, Pang J, Chen Y, Qi Q, Wang H, Sun Y, Gul S, Zhou X, Tang W. Constructing a Prognostic Model for Non-Small-Cell Lung Cancer Risk Based on Genes Characterising the Differentiation of Myeloid-Derived Suppressor Cells. Int J Mol Sci 2025; 26:4679. [PMID: 40429821 PMCID: PMC12111218 DOI: 10.3390/ijms26104679] [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: 04/02/2025] [Revised: 05/10/2025] [Accepted: 05/11/2025] [Indexed: 05/29/2025] Open
Abstract
Cancer is the most common malignancy, with over 2 million new cases and nearly 1.8 million deaths worldwide annually. Non-small-cell lung cancer (NSCLC) is the predominant subtype, accounting for the majority of cases. Myeloid-derived suppressor cells (MDSCs), which originate from monocytes and typically differentiate into macrophages and granulocytes, possess potent immunosuppressive capabilities. MDSCs regulate immune responses in various pathological conditions and are strongly associated with poor prognosis in cancer patients. This study aims to elucidate the complex interplay between MDSCs, immune cells, and tumours in the NSCLC tumour microenvironment (TME). By integrating single-cell RNA sequencing (scRNA-seq) data with bulk RNA sequencing (Bulk RNA-seq) data, we identified MDSCs as the target cell population and used Monocle software (v2.22.0) to infer their developmental trajectories. We identified key genes associated with MDSCs differentiation processes and classified MDSCs into seven distinct states based on their functional roles. Furthermore, we constructed a prognostic risk model based on the impact of MDSCs differentiation on NSCLC prognosis, utilizing Elastic Net regression and multivariate Cox regression analysis of Bulk RNA-seq data. The model's performance and accuracy were validated using both internal and external validation sets. Additionally, we compared risk scores with clinical pathological features and the relationship between risk scores and key immune cells in the immune microenvironment, demonstrating the model's clinical predictive value. We also explored how prognostic genes contribute to poor prognosis in NSCLC. Moreover, small molecule compounds targeting these prognostic genes were screened, and their anti-tumour effects were evaluated as potential therapeutic strategies for NSCLC treatment. This study not only reveals the complex regulatory mechanisms of MDSCs in the NSCLC immune microenvironment but also successfully constructs a prognostic risk model based on MDSCs differentiation states. The model demonstrates excellent clinical performance in predicting patient prognosis, effectively identifying high-risk patients and providing robust support for individualized treatment and immunotherapy decisions. Through association analyses with key immune cells in the immune microenvironment and clinical pathological features, our model can assist clinicians in formulating more precise treatment plans based on patients' immune status and tumour characteristics. Furthermore, we identified small molecule compounds targeting these prognostic genes, providing novel and promising therapeutic targets for NSCLC, which could further enhance treatment efficacy and improve patients' survival quality.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xuhong Zhou
- Laboratory of Molecular Genetics of Aging & Tumour, Medicine School, Kunming University of Science and Technology, Kunming 650032, China; (Y.T.); (J.P.); (Y.C.); (Q.Q.); (H.W.); (Y.S.); (S.G.)
| | - Wenru Tang
- Laboratory of Molecular Genetics of Aging & Tumour, Medicine School, Kunming University of Science and Technology, Kunming 650032, China; (Y.T.); (J.P.); (Y.C.); (Q.Q.); (H.W.); (Y.S.); (S.G.)
| |
Collapse
|
50
|
Han X, Xu J, Cui M, Yun Z, Zhao H, Tian S, Mi S, Hou L. Haematological toxicities with immune checkpoint inhibitors in digestive system tumors: a systematic review and network meta-analysis of randomized controlled trials. Clin Exp Med 2025; 25:157. [PMID: 40360867 PMCID: PMC12075026 DOI: 10.1007/s10238-025-01688-x] [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: 03/09/2025] [Accepted: 04/13/2025] [Indexed: 05/15/2025]
Abstract
This study aims to comprehensively evaluate the hematologic toxicity profiles, toxicity spectrum, and safety rankings of immune checkpoint inhibitors (ICIs) used for digestive system tumors. The PubMed, Cochrane Library, Web of Science, and Embase databases were systematically searched from inception to August 2024 to identify randomized controlled trials (RCTs). The primary outcome was anemia, while secondary outcomes included neutropenia, neutrophil count decreased, thrombocytopenia, platelet count decreased, leukopenia, white blood cell (WBC) count decreased, lymphocyte count decreased, and febrile neutropenia (FN). Subgroup analyses were performed based on tumor type, country category, study phase, ICI regimen, control group, chemotherapy regimen, ICI plus different chemotherapy regimens. Two reviewers independently selected the studies, extracted data according to pre-specified criteria, and assessed the risk of bias using the Cochrane Collaboration risk of bias tool. RevMan 5.4 software was utilized to visualize the risk of bias assessments. Stata 16.0 was used to conduct network meta-analysis, sensitivity analysis and meta-regression. 25 phase II and III RCTs (n = 15216) were included. The general safety of ICIs ranked from high to low for grade 1-5 anemia were as follows: avelumab, nivolumab, pembrolizumab, sintilimab, camrelizumab, and tislelizumab. For grade 3-5 anemia, the general safety profile of the ICIs were as follows, from highest to lowest: avelumab, nivolumab, pembrolizumab, sintilimab, and camrelizumab. Compared to chemotherapy, treatment-related hematologic toxicities with ICIs occurred primarily in grade 1-5 anemia, neutropenia, thrombocytopenia, leukopenia, and WBC count decreased. Taking ICI monotherapy, nivolumab plus ipilimumab were generally safer than taking chemotherapy, one ICI drug with chemotherapy, or two ICI drugs with chemotherapy. In terms of grade 1-5 hematologic toxicities, tislelizumab had the highest risk of neutropenia and leukopenia; the primary treatment-adverse events (AEs) for sintilimab was neutrophil count decreased and WBC count decreased; the primary treatment-related AE associated with nivolumab was platelet count decreased; camrelizumab posed the highest risk for lymphocyte count decreased. In terms of grade 3-5 hematologic toxicities, pembrolizumab was predominantly linked to neutropenia; sintilimab showed the greatest risk for neutrophil count decreased, platelet count decreased, and lymphocyte count decreased; avelumab was most associated with WBC count decreased. FN primarily manifested as grade 3-5, with camrelizumab having the highest risk. Among agents used in gastric or gastroesophageal junction cancer, avelumab demonstrated the most favorable safety profile for anemia. Each treatment regimen has its unique safety profile. Early identification and management of ICI-related hematologic toxicities are essential in clinical practice.Systematic Review Registration: PROSPERO CRD42024571508.
Collapse
Affiliation(s)
- Xinpu Han
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Jing Xu
- Hubei Provincial Hospital of Traditional Chinese Medicine, Hubei, China
| | - Meichen Cui
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Zhangjun Yun
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Hongbin Zhao
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Shaodan Tian
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Suicai Mi
- Xiamen Hospital, Dongzhimen Hospital, Beijing University of Chinese Medicine, Xiamen, China.
| | - Li Hou
- Department of Oncology and Hematology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|