1
|
Janes LA, Haykal T, Angeles CV. Vaccines in Melanoma: Past, Present, and Future. Surg Oncol Clin N Am 2025; 34:411-421. [PMID: 40413007 DOI: 10.1016/j.soc.2025.01.004] [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] [Indexed: 05/27/2025]
Abstract
While immune checkpoint inhibitor (ICI) immunotherapy has significantly improved the outcomes for advanced melanoma patients, more than 50% of patients do not respond. Cancer vaccines can induce a robust anti-tumor immune response, which may provide the necessary immunomodulation required for ICI efficacy. To date, there has only been modest clinical benefit with melanoma vaccines. Recent advancements with improved vaccine technology including the use of mRNA vaccines have promised to lead better clinical outcomes. This review will discuss the historical use of vaccines for melanoma, approaches to vaccine development, the current ongoing trials, and future directions.
Collapse
Affiliation(s)
- Lindsay A Janes
- Department of Surgery, University of Michigan, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Tarek Haykal
- Department of Medicine, University of Michigan, Rogel Cancer Center, 1500 East Medical Center Drive, Ann Arbor, MI 48109, USA
| | - Christina V Angeles
- Departments of Surgery and Dermatology, University of Michigan, Rogel Cancer Center, 1500 East Medical Center Drive, Cancer Center 6219, Ann Arbor, MI 48109, USA.
| |
Collapse
|
2
|
Effiom D, Cohen S. Nodal Management in Melanoma: Is Nodal Staging Needed, What Is the Value, and What Is the Extent of Surgery? Surg Oncol Clin N Am 2025; 34:343-357. [PMID: 40413003 DOI: 10.1016/j.soc.2025.01.005] [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] [Indexed: 05/27/2025]
Abstract
Historical management of melanoma metastatic to draining nodal basins involved prophylactic lymphadenectomy, a morbid procedure without proven survival benefit. Over the last decade sentinel lymph node biopsy (SLNB) became a key prognostic tool, identifying patients with clinically occult nodal metastases with minimal morbidity. More recently, with the approval of adjuvant therapies for high-risk melanomas, the role of SLNB in the treatment of melanoma has been questioned. However, no robust alternative prognostic tool exists. SLNB therefore remains a critical staging procedure, informing clinical decision making when weighing the risks and benefits of adjuvant treatment and surveillance.
Collapse
Affiliation(s)
- Derek Effiom
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02144, USA; Department of Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02144, USA
| | - Sonia Cohen
- Division of Gastrointestinal and Oncologic Surgery, Department of Surgery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02144, USA; Department of Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit Street, Boston, MA 02144, USA.
| |
Collapse
|
3
|
Seldomridge AN, Weiser R, Holder AM. Systemic Therapy for Melanoma: What Surgeons Need to Know. Surg Oncol Clin N Am 2025; 34:359-374. [PMID: 40413004 DOI: 10.1016/j.soc.2025.01.003] [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] [Indexed: 05/27/2025]
Abstract
Immune checkpoint inhibitors and targeted therapies (BRAF/MEK inhibitors) have transformed the care of patients with stage IV melanoma, now with 5-year overall survival rates around 50%. Surgeons should be acquainted with these drugs, the multidisciplinary considerations of their use, and the unique immune-related adverse events (irAEs) they can cause. In this review, we discuss systemic therapies for cutaneous melanoma, including the biology of immune checkpoint inhibition, treatment indications, and toxicities. We also explain how these irAEs and other toxicities can impact surgical planning and perioperative management.
Collapse
Affiliation(s)
- Ashlee N Seldomridge
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Roi Weiser
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Ashley M Holder
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| |
Collapse
|
4
|
Yin Y, Wang C, Liang H, Ji Y, Zhao D, Wang L, Shi W, Wang X, Gao Y, Liu B. Design, synthesis, and biological evaluation of 2,3-dihydro-1H-pyrrolo[3,4-c]pyridin-1-one derivatives as potent hematopoietic progenitor kinase 1 (HPK1) inhibitors. Bioorg Chem 2025; 161:108554. [PMID: 40347769 DOI: 10.1016/j.bioorg.2025.108554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/14/2025]
Affiliation(s)
- Yuan Yin
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Chengqi Wang
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Huan Liang
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Yanpeng Ji
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Damin Zhao
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Lilong Wang
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Wei Shi
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Xiaojin Wang
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China
| | - Yong Gao
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China.
| | - Baomin Liu
- Chia Tai Tianqing Pharmaceutical Group Co., Ltd, Jiangsu Key Laboratory of Antiviral Drug Research, 1099 Fuying Road, Jiangning District, Nanjing, Jiangsu Province, China.
| |
Collapse
|
5
|
Tang LB, Peng YL, Chen J, Li JT, Zheng MM, Wu L, Lu C, Wei XW, Cai DX, Guo Z, Ren ZR, Lv SD, Deng Y, Chen ZH, Xu CR, Zhou Q. Rechallenge with immune-checkpoint inhibitors in patients with advanced-stage lung cancer. Nat Rev Clin Oncol 2025:10.1038/s41571-025-01029-7. [PMID: 40490476 DOI: 10.1038/s41571-025-01029-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2025] [Indexed: 06/11/2025]
Abstract
Lung cancer remains the leading cause of cancer-related mortality globally, with many patients diagnosed with advanced-stage disease. Treatment in this setting relies on systemic therapies, including chemotherapy, targeted therapy and immunotherapy. Immune-checkpoint inhibitors (ICIs), which promote or restore antitumour immunity by inhibiting immunosuppressive signalling pathways, are currently the most widely used immunotherapies in these patients. However, immune-related adverse events (irAEs) or disease progression often necessitate discontinuation of these agents, leaving many patients with limited subsequent treatment options. In this scenario, ICI rechallenge has emerged as a potential strategy. Despite this potential, evidence for ICI rechallenge after either disease progression or irAEs in patients with non-small-cell lung cancer is limited and evidence for those with small cell lung cancer seems to be non-existent. In this Review, we provide a comprehensive overview of the available data on ICI rechallenge in the context of both disease progression and irAEs, including a summary of current guidance on clinical management and detailed discussions of safety and efficacy. We also highlight important unanswered questions in an attempt to guide future research in this area.
Collapse
Affiliation(s)
- Li-Bo Tang
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ying-Long Peng
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ji Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jia-Ting Li
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Mei-Mei Zheng
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lv Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chang Lu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xue-Wu Wei
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Dong-Xuan Cai
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhi Guo
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zi-Rui Ren
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Si-Di Lv
- School of Art, Soochow University, Suzhou, China
| | - Yu Deng
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhi-Hong Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chong-Rui Xu
- School of Medicine, South China University of Technology, Guangzhou, China
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qing Zhou
- School of Medicine, South China University of Technology, Guangzhou, China.
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
6
|
Padmanaban V, Contreras CM. Role of Surgery for Metastatic Melanoma in the Era of Checkpoint Blockade. Surg Clin North Am 2025; 105:663-679. [PMID: 40412893 DOI: 10.1016/j.suc.2024.11.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2025]
Abstract
Treatment paradigms have shifted with the introduction of checkpoint inhibitors, facilitating less extensive surgical procedures in some scenarios. Resection and adjuvant therapy are key components in patients with locally advanced or limited metastatic disease. Alternately, these patient populations may benefit from peri-operative systemic therapy based on recent clinical trial data. Resection is also integral to administering tumor infiltrating lymphocyte therapy in patients with treatment-refractory, unresectable melanoma. The role of surgery continues to evolve, and ranges the spectrum from resection with curative intent, to palliative interventions intended to effectively manage symptoms or disease-related complications.
Collapse
Affiliation(s)
- Vennila Padmanaban
- Division of Surgical Oncology, Department of Surgery, James Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Carlo M Contreras
- Division of Surgical Oncology, Department of Surgery, James Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
7
|
Wang X, Li Q, Liu J, Xie C, Zou L, Shi Y, Jiang L, Qin X. Harnessing nano-delivery systems to un-cover the challenges for cervical cancer therapy. Int J Pharm 2025; 677:125657. [PMID: 40306445 DOI: 10.1016/j.ijpharm.2025.125657] [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: 02/20/2025] [Revised: 04/17/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Cervical cancer (CC) remains a prevalent malignancy among women, with current therapeutic strategies facing significant challenges in curbing its rising incidence. Nano-delivery systems have emerged as a promising approach to hinder CC progression. This review provides a comprehensive examination of CC pathogenesis and its physiological characteristics while focusing on applying various nano-delivery systems in CC therapy. Specifically, it highlights the potential of both internal (e.g., pH, reactive oxygen species, glutathione) and external (e.g., Photo, magnetism, sound waves, microwaves, electricity) stimuli-responsive nano-delivery platforms to enhance therapeutic efficacy. The challenges of nano-delivery systems in CC therapy, encompassing in vivo stability, biosafety, distribution, and metabolic processes, are addressed, along with potential remedies. Additionally, the review underscores recent preclinical advances in nano-delivery systems for CC therapy. By thoroughly exploring nanomaterial applications, this review provides valuable perspectives for advancing CC treatment and stimulating future research and innovation in this domain.
Collapse
Affiliation(s)
- Xinyu Wang
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China; Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medical, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Qi Li
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medical, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Jianxin Liu
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China; Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medical, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Chunbao Xie
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medical, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China
| | - Liang Zou
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Yi Shi
- School of Food and Biological Engineering, Chengdu University, Chengdu 610106, China; Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medical, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China; Research Unit for Blindness Prevention of Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan 610072, China; Department of Ophthalmology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Lingxi Jiang
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medical, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| | - Xianyan Qin
- Sichuan Provincial Key Laboratory for Human Disease Gene Study and the Center for Medical Genetics, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, School of Medical, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China.
| |
Collapse
|
8
|
Lei S, Gao Y, Wang K, Wu S, Zhu M, Chen X, Zhou W, Chen X, Zhang J, Duan X, Men K. An Implantable Double-Layered Spherical Scaffold Depositing Gene and Cell Agents to Facilitate Collaborative Cancer Immunotherapy. ACS NANO 2025; 19:17653-17673. [PMID: 40304563 DOI: 10.1021/acsnano.5c01366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Gene therapies and adoptive cell therapy (ACT) are promising strategies for cancer immunotherapy. Referring to their different mechanisms, the combination of these two might result in a strategy with potential collaborative and compensatory effects. However, it is challenging to combine gene therapies and ACT that work in a proper logical order. Here, we developed a double-layered spherical scaffold (DLS) to codeliver mRNA and T cells and constructed an implantable hydrogel formulation, named the GD-920 scaffold. With a diameter of 7 mm, this scaffold loaded primary T cells in the inner layer and the Bim mRNA nanocomplex in the outer layer. While maintaining their bioactivities, GD-920 released gene and cell payloads in a controllable and sequential manner. The mRNA complex from the outer layer was first released and induced immunogenic tumor cell death. The produced antigens then migrated into the scaffold with dendritic cells, triggering a tumor-specific immune response. Finally, activated T cells released by the inner layer attacked the tumor tissue via massive infiltration. We showed that in situ implantation of the GD-920 scaffold is capable of effectively inhibiting tumor growth and is far more potent than that of control scaffolds containing a single payload. Our results demonstrated the outstanding potential of this DLS in combining gene and cell therapeutic approaches to cancer immunotherapy.
Collapse
Affiliation(s)
- Sibei Lei
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yan Gao
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kaiyu Wang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shan Wu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Manfang Zhu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Xiaohua Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Weilin Zhou
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiayu Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jin Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xingmei Duan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Ke Men
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
9
|
Yamamoto Y, Kitadani J, Hayata K, Goda T, Tominaga S, Nakai T, Nagano S, Iwamoto R, Kawai M. Promising Treatment Strategy for Primary Malignant Melanoma of the Esophagus by Radical Esophagectomy and Nivolumab as Adjuvant Therapy: A Case Report. Surg Case Rep 2025; 11:25-0027. [PMID: 40356806 PMCID: PMC12068939 DOI: 10.70352/scrj.cr.25-0027] [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: 01/21/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
INTRODUCTION Primary malignant melanoma of the esophagus (PMME) is a rare malignant tumor of the esophagus with very poor prognosis due to high rates of recurrence and metastasis even after radical resection. Recently, however, immune checkpoint inhibitors such as anti-programmed cell death-1 antibodies have been suggested to improve the prognosis of malignant melanoma. This report describes the use of postoperative nivolumab as adjuvant therapy after surgical resection of PMME, with recurrence-free follow-up for more than 1 year. CASE PRESENTATION A 69-year-old man had chest discomfort and tightness in his throat. Upper gastrointestinal endoscopy revealed multiple melanosis and an elevated lesion in the middle esophagus. After histological examination, he was diagnosed as having PMME, so he underwent thoracoscopic subtotal esophagectomy, three-field lymphadenectomy, pedunculated jejunum reconstruction with super-charge and super-drainage, and feeding jejunostomy due to the past history of gastrectomy. The adjuvant therapy using nivolumab (every 2 weeks, 240 mg) for 1 year was completed with no serious side effects, and there was no recurrence for more than 1 year postoperatively. CONCLUSIONS Although cases of PMME treated with adjuvant nivolumab have rarely been reported, the present case suggests that this approach may represent a promising treatment option, similar to cutaneous melanoma.
Collapse
Affiliation(s)
- Yusuke Yamamoto
- Second Department of Surgery, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Junya Kitadani
- Second Department of Surgery, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Keiji Hayata
- Second Department of Surgery, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Taro Goda
- Second Department of Surgery, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Shinta Tominaga
- Second Department of Surgery, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Tomoki Nakai
- Second Department of Surgery, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Shotaro Nagano
- Second Department of Surgery, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Ryuta Iwamoto
- Department of Human Pathology, Wakayama Medical University, Wakayama, Wakayama, Japan
| | - Manabu Kawai
- Second Department of Surgery, Wakayama Medical University, Wakayama, Wakayama, Japan
| |
Collapse
|
10
|
Kong SL, Feng Z, Kim S, Ha EK, Kamel K, Becich M, Luketich JD, Pennathur A. Hyperthermic Intrathoracic Chemoperfusion and the Role of Adjunct Immunotherapy for the Treatment of Pleural Mesothelioma. Biomolecules 2025; 15:678. [PMID: 40427571 PMCID: PMC12108701 DOI: 10.3390/biom15050678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 05/29/2025] Open
Abstract
Pleural mesothelioma (PM) is an aggressive cancer originating from the mesothelial lining of the pleura, with a rising global incidence since the mid-20th century due to asbestos and erionite exposure. PM accounts for 80-90% of all mesothelioma cases and is histologically classified into three subtypes-epithelioid, sarcomatoid, and biphasic- with epithelioid carrying the most favorable prognosis. Despite advances in surgery, chemotherapy, radiotherapy, and immunotherapy, PM prognosis remains poor, necessitating more effective, multimodal strategies. Hyperthermic intrathoracic chemoperfusion (HITHOC) has emerged as a promising adjunct to cytoreductive surgery by delivering heated chemotherapy directly to the pleural cavity, potentially improving survival-especially in patients with epithelioid PM. Combining HITHOC with post-surgical immunotherapy represents a novel approach to enhancing both local and systemic anti-tumor responses and targeting microscopic disease and distant metastases. This review explores surgical outcomes after surgery for PM, the therapeutic synergy of HITHOC and immunotherapy, ongoing clinical trials evaluating this multimodal strategy, and its implications for future patient care.
Collapse
Affiliation(s)
- Susan Luozheng Kong
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.L.K.); (K.K.); (J.D.L.)
| | - Zihan Feng
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - Sangmin Kim
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.L.K.); (K.K.); (J.D.L.)
| | - Edra K. Ha
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.L.K.); (K.K.); (J.D.L.)
| | - Kero Kamel
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.L.K.); (K.K.); (J.D.L.)
| | - Michael Becich
- Department of Bioinformatics, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
| | - James D. Luketich
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.L.K.); (K.K.); (J.D.L.)
| | - Arjun Pennathur
- Department of Cardiothoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.L.K.); (K.K.); (J.D.L.)
- UPMC Hillman Cancer Center Pittsburgh, Pittsburgh, PA 15213, USA
| |
Collapse
|
11
|
Weichenthal M, Ellebaek E, Mangana J, Asher N, Gavrilova I, Kandolf L, Ugurel S, Hausschild A, Meier F, Leiter U, Livingstone E, Gebhardt C, Gutzmer R, Ruhlmann CH, Mahncke-Guldbrandt L, Haslund CA, Kopec S, Teterycz P, Bender M, Poudroux W, Muñoz-Couselo E, Berciano-Guerrero MA, Shalamanova G, DePalo DK, Brozić JM, Chiarion-Sileni V, Arance A, Ziogas D, Robert C, van de Velde AO, Gassama AA, Shapira R, BenBetzalel G, Grynberg S, Ramelyte E, Bertoldo F, DelPrete V, Gaudy-Marqueste C, Mohr P, Dummer R, Ascierto PA, Gogas H, Espinosa E, Lebbé C, Rutkowski P, Haanen J, Schadendorf D, Svane IM. Immune checkpoint inhibition in metastatic or non-resectable melanoma after failure of adjuvant anti-PD-1 treatment. A EUMelaReg real-world evidence study. Eur J Cancer 2025; 220:115339. [PMID: 40090216 DOI: 10.1016/j.ejca.2025.115339] [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: 01/22/2025] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND Adjuvant immune checkpoint inhibition (ICI) with anti-PD-1 antibodies in high-risk resected melanoma has been shown to improve recurrence-free survival. It is unclear whether prior adjuvant anti-PD-1 therapy is associated with altered response to subsequent ICI treatment in the metastatic setting. METHODS Using data from the European Melanoma Registry (EUMelaReg), we analyzed the efficiency of first-line (1L) ICI in non-resectable or metastatic melanoma after failure from prior adjuvant anti-PD-1 treatment. Both single-agent anti-PD-1 and combined anti-PD-1/CTLA-4 (Ipi/Nivo) 1L regimes were included in the analysis. We identified 389 patients receiving 1L ICI with prior adjuvant anti-PD-1 treatment. The control population was selected from a pool of 3390 PD-1-naive cases by 1:1 matching for the type of 1L ICI and various prognostic factors. As outcome measure, overall remission rates (ORR) were calculated and progression-free survival (PFS) was evaluated by Kaplan-Meier and Cox regression analysis. RESULTS Out of 389 patients, 303 (77.9 %) received Ipi/Nivo and 86 (22.1 %) anti-PD-1 in 1L. ORR was significantly lower in pre-treated patients (31.4 %) as compared to anti-PD-1 naive patients (48.8 %; p < 0.0001). Kaplan-Meier analysis showed significantly shorter median PFS for pre-treated patients. This applied to both anti-PD-1 and Ipi/Nivo treatment. Patients with early recurrence from adjuvant treatment (during or up to 12 weeks after end of treatment) showed lower ORR (28.5 %) and shorter PFS (3.1 months) than those who recurred later (37.7 % and 6.1 months, respectively). CONCLUSIONS Patients with metastatic melanoma, previously exposed to anti-PD-1 ICI in the adjuvant setting showed significantly lower ORR and shorter PFS to 1L ICI with either Ipi/Nivo or single-agent anti-PD-1 retreatment.
Collapse
Affiliation(s)
- Michael Weichenthal
- Skin Cancer Center Kiel, University Hospital Schleswig-Holstein, Kiel, Germany.
| | - Eva Ellebaek
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Joanna Mangana
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Nethanel Asher
- Skin Cancer and Melanoma Center at Davidoff Cancer Center, Rabin Medical Center, Israel
| | - Iva Gavrilova
- Oncodermatology Department, National Oncology Center, Sofia, Bulgaria
| | - Lidija Kandolf
- Department of Dermatology, Faculty of Medicine, Military Medical Academy, Belgrade, Serbia
| | - Selma Ugurel
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany
| | - Axel Hausschild
- Skin Cancer Center Kiel, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Friedegund Meier
- Department of Dermatology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Leiter
- Universitäts-Hautklinik Tübingen, Tübingen, Germany.
| | - Elisabeth Livingstone
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany
| | - Christoffer Gebhardt
- Department of Dermatology and Venereology, University Skin Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Ralf Gutzmer
- Department of Dermatology, Johannes Wesling Medical Center, Ruhr University Bochum Campus Minden, Minden, Germany
| | | | | | | | - Sylwia Kopec
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Paweł Teterycz
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Department of Computational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Marc Bender
- Skin Cancer Center, Division of Molecular Cell Biology, Elbe Kliniken Stade-Buxtehude, Buxtehude, Germany
| | | | - Eva Muñoz-Couselo
- Medical oncology department, Vall d'Hebron Hospital, Barcelona-Spain and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| | - Miguel-Angel Berciano-Guerrero
- Medical Oncology Intercenter Unit, Regional and Virgen de la Victoria University Hospitals, IBIMA Plataforma BIONAND, Málaga, Spain
| | | | - Danielle K DePalo
- Department of Cutaneous Oncology, Surgical Oncology Research Fellow, Moffitt Cancer Center, Tampa, FL, United States
| | - Jasmina Marić Brozić
- Department of Oncology, UHC Sestre milosrdnice, School of Medicine Zagreb, Zagreb, Croatia
| | | | - Anna Arance
- Department of Medical Oncology and IDIBAPS, Hospital Clínic Barcelona, Barcelona, Spain
| | - Dimitrios Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Caroline Robert
- Gustave Roussy Cancer Campus and Université Paris Saclay, Villejuif, France
| | | | - Awa Aminata Gassama
- Scientific Administration, Biometrics Department, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Ronnie Shapira
- The Ella Lemelbaum Institute for Immuno-Oncology and Melanoma, Ramat-Gan, Israel
| | - Guy BenBetzalel
- The Ella Lemelbaum Institute for Immuno-Oncology and Melanoma, Ramat-Gan, Israel
| | - Shirly Grynberg
- The Ella Lemelbaum Institute for Immuno-Oncology and Melanoma, Ramat-Gan, Israel
| | - Egle Ramelyte
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland.
| | - Fabio Bertoldo
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Valerio DelPrete
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Caroline Gaudy-Marqueste
- Aix-Marseille Univ, APHM, Hôpital Timone, Dermatology and Skin Cancer Department. Centre de Recherche en Cancérologie de Marseille (CRCM), Inserm, U1068, Marseilles, France
| | - Peter Mohr
- Department of Dermatology, Elbekliniken Buxtehude, Buxtehude, Germany
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Paolo A Ascierto
- Department of Skin Cancers, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Enrique Espinosa
- Service of Oncology, Hospital U. La Paz, U. Autónoma de Madrid, Madrid-CIBERONC, Spain
| | - Celeste Lebbé
- Université Paris Cite, AP-HP Dermato-oncology and CIC, Cancer Institute APHP, INSERM U976, Saint Louis Hospital, Paris, France.
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - John Haanen
- Netherlands Cancer Institut Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Dirk Schadendorf
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, Essen, Germany
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|
12
|
Qureshi Z, Zaheer Z, Asghar Z, Bakhtiar M, Fatima E, Altaf F. Cardiovascular Risk Profile of Nivolumab Anti-cancer Therapy: A Review of Current Literature. Am J Clin Oncol 2025; 48:235-241. [PMID: 40008416 DOI: 10.1097/coc.0000000000001166] [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] [Indexed: 02/27/2025]
Abstract
OBJECTIVES Immune checkpoint inhibitors (ICI) upregulate host antitumor immunity, proving efficacy across diverse tumor types. Currently approved ICI treatment primarily targets the programmed cell death receptor 1 (PD-1) and its ligand PD-L1, and cytotoxic T lymphocyte-antigen 4 (CTLA-4). Nivolumab is a monoclonal antibody that targets the human PD-1 receptor and is an entirely human immunoglobulin G4 (IgG4), approved by the FDA for various cancers like advanced melanoma, metastatic renal cell carcinoma, Hodgkin lymphoma, and advanced lung carcinoma. This review will summarise and discuss the recent literature on cardiotoxicity associated with nivolumab therapy. METHODS We searched online databases like PubMed, Scopus, Google Scholar, and Embase for articles related to Nivolumab. RESULTS Cardiotoxicity with ICI use is most commonly represented as myocarditis. Patients present with complaints of shortness of breath, palpitations, edema, and fatigue. Takotsubo cardiomyopathy, or broken heart syndrome, is characterized by systolic dysfunction of the left ventricle, mimicking a myocardial infarction but without associated coronary ischemia and with minimal elevation of cardiac enzymes. In the CHECKMATE-037 trial, ventricular arrhythmias occurred in <10% of those who received nivolumab. In a retrospective analysis of patients treated with ICI (predominantly nivolumab monotherapy) for lung cancer, 11% of the patients developed major adverse cardiac events, including myocarditis, non-ST-segment elevated myocardial infarction, supraventricular tachycardia, and pericardial disorders. CONCLUSION Close collaboration between cardiology and oncology specialists is crucial for early detection and effective management of cardiac complications, enhancing the safety of nivolumab anticancer therapy.
Collapse
Affiliation(s)
- Zaheer Qureshi
- The Frank H. Netter M.D. School of Medicine at Quinnipiac University, Bridgeport, CT
| | | | - Zoha Asghar
- Department of Medicine, Ziauddin University, Karachi
| | | | - Eeshal Fatima
- Department of Medicine, Services Institute of Medical Sciences, Lahore, Pakistan
| | - Faryal Altaf
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai/BronxCare Health System, New York, NY
| |
Collapse
|
13
|
Wang J, Chen Q, Shan Q, Liang T, Forde P, Zheng L. Clinical development of immuno-oncology therapeutics. Cancer Lett 2025; 617:217616. [PMID: 40054657 PMCID: PMC11930610 DOI: 10.1016/j.canlet.2025.217616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
Immuno-oncology (IO) is one of the fastest growing therapeutic areas within oncology. IO agents work indirectly via the host's adaptive and innate immune system to recognize and eradicate tumor cells. Despite checkpoint inhibitors being only introduced to the market since 2011, they have become the second most approved product category. Current Food and Drug Administration (FDA)-approved classes of IO agents include: immune checkpoint inhibitors (ICIs), chimeric antigen receptor T-cell therapy (CAR-T), bi-specific T-cell engager (BiTE) antibody therapy, T-cell receptor (TCR) engineered T cell therapy, tumor-infiltrating lymphocyte (TIL) therapy, cytokine therapy, cancer vaccine therapy, and oncolytic virus therapy. Cancer immunotherapy has made progress in multiple cancer types including melanoma, non-small cell lung cancer (NSCLC), renal cell carcinoma (RCC), and urothelial carcinoma; however, several cancers remain refractory to immunotherapy. Future directions of IO include exploration in the neoadjuvant/perioperative setting, combination strategies, and optimizing patient selection through improved biomarkers.
Collapse
Affiliation(s)
- Jianxin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Qi Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Qiang Shan
- Department of General Surgery, Haining People's Hospital, Haining, 314400, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Patrick Forde
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA; The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Mays Cancer Center at the University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Lei Zheng
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA; The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Mays Cancer Center at the University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
14
|
Yuan J, Xu B, Su Y, Zhang P, Zhang X, Gong P. Identification of USP39 as a prognostic and predictive biomarker for determining the response to immunotherapy in pancreatic cancer. BMC Cancer 2025; 25:758. [PMID: 40264098 PMCID: PMC12016207 DOI: 10.1186/s12885-025-14096-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 04/07/2025] [Indexed: 04/24/2025] Open
Abstract
Ubiquitin-Specific Protease 39 (USP39) has been implicated in numerous malignancies, however, its pathogenic mechanisms and impact on the tumor immune microenvironment (TIME) remain incompletely characterized. Based on The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) databases, we investigated the diagnostic and prognostic values of USP39 across various cancer types. Additionally, we examined the correlation between USP39 expression and immune-related gene signature, immune cell infiltration pattern, tumor microsatellite instability (MSI), and tumor mutation burden (TMB). This study specifically focused on exploring the clinical relevance and molecular functions of USP39 in pancreatic adenocarcinoma (PAAD), with particularly emphasis on its role in shaping the TIME and modulating responses to immunotherapy. The results demonstrated that evaluated USP39 expression significantly correlated with advanced tumor stage and unfavorable clinical outcomes across multiple cancer types, most notably in PAAD. Functional enrichment analysis indicated that USP39 potentially promotes tumor progression through multiple oncogenic signaling cascades. In vitro experimental validation confirmed that USP39 knockdown inhibited migration and proliferation of pancreatic cancer cells while inducing apoptosis. Additionally, we identified significant positive correlations between USP39 expression and immune checkpoint molecules, particularly prominent in PAAD. Furthermore, we observed associations between USP39 expression and TMB in 16 cancer types and MSI in 11 cancer types, suggesting that heightened USP39 expression may enhance responsiveness to immunotherapeutic interventions. Collectively, our findings establish USP39 as a valuable immune-related biomarker with both diagnostic and prognostic utility across multiple cancer types, especially PAAD, underscoring its potential as a promising therapeutic target for cancer immunotherapy. Clinical trial number Not applicable.
Collapse
Affiliation(s)
- Jiahui Yuan
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Beibei Xu
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yongcheng Su
- Xiamen Key Laboratory for Tumor Metastasis, Cancer Research School of Medicine, Xiamen University, Xiamen, 361102, China
| | - Pingping Zhang
- Department of Gastroenterology, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Xianbin Zhang
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China
| | - Peng Gong
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Guangdong Provincial Key Laboratory of Chinese Medicine Ingredients and Gut Microbiomics, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
15
|
Zhang B, Zhou Q, Zhang P, Xue C, Ke X, Wang Y, Han T, Zhou F, Dong W, Deng L, Jing M, Zhang Y, Liu X, Zhou J. MRI histogram and Visually AcceSAble Rembrandt Images features nomogram to predict PD-L1 levels in glioblastoma. Neurosurg Rev 2025; 48:371. [PMID: 40257668 DOI: 10.1007/s10143-025-03528-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/29/2025] [Accepted: 04/10/2025] [Indexed: 04/22/2025]
Abstract
This study aimd to delineate the prognostic impact of Programmed death-ligand 1 (PD-L1) expression level in glioblastoma (GBM) patients, and investigate the value of using magnetic resonance imaging (MRI) histograms and Visually Accessible Rembrandt Images (VASARI) features to preoperatively predict the expression level of PD-L1. The clinical and imaging data of 124 patients with GBM at our institution between November 2018 and April 2023 were retrospectively analyzed. PD-L1 expression levels in tumor tissue samples were quantified using immunohistochemical staining. The optimal cutoff PD-L1 level was determined using the X-tile program through Kaplan-Meier survival analysis and log-rank test. The MRI histogram and VASARI features of the patients in the high and low PD-L1 expression groups were recorded. The predictive models for PD-L1 expression level were constructed using multivariable binary logistic regression, and a nomogram was generated. The GBM patients with high PD-L1 expression had unfavorable overall survival. The T1-weighted contrast-enhanced histogram features mean, 1st, 5th, 10th, 25th, 50th, and 75th percentiles and the VASARI feature F5 proportion enhancing were statistically significantly different between groups (all p < 0.05). Multivariate logistic regression analysis showed that mean, 5th, 10th, and 50th percentiles, and F5 proportion enhancing were independent risk factors for predicting PD-L1 expression in GBM patients. The logistic regression model based on these 5 features showed a better predictive performance, and the area under the curve, accuracy, sensitivity, specificity were 0.795, 0.726, 0.887, and 0.621, respectively. The nomogram based on MRI histogram and VASARI features can show promise to non-invasively predict the level of PD-L1 expression in GBM and could be helpful in guiding immune checkpoint inhibitors therapy and predicting patient prognosis. Clinical trial number Not applicable.
Collapse
Affiliation(s)
- Bin Zhang
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen, No. 82, Chengguan District, Lanzhou, Gansu, 730030, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Qing Zhou
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen, No. 82, Chengguan District, Lanzhou, Gansu, 730030, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Peng Zhang
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Caiqiang Xue
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen, No. 82, Chengguan District, Lanzhou, Gansu, 730030, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Xiaoai Ke
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen, No. 82, Chengguan District, Lanzhou, Gansu, 730030, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Yige Wang
- Medical Department, Gansu Provincial Hospital, Lanzhou, Gansu, China
| | - Tao Han
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen, No. 82, Chengguan District, Lanzhou, Gansu, 730030, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Fengyu Zhou
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen, No. 82, Chengguan District, Lanzhou, Gansu, 730030, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Wenjie Dong
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen, No. 82, Chengguan District, Lanzhou, Gansu, 730030, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Liangna Deng
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen, No. 82, Chengguan District, Lanzhou, Gansu, 730030, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Mengyuan Jing
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen, No. 82, Chengguan District, Lanzhou, Gansu, 730030, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Yuting Zhang
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen, No. 82, Chengguan District, Lanzhou, Gansu, 730030, China
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
| | - Xianwang Liu
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China
- Department of Imaging, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen, No. 82, Chengguan District, Lanzhou, Gansu, 730030, China.
- Second Clinical School, Lanzhou University, Lanzhou, Gansu, China.
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China.
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, Gansu, China.
| |
Collapse
|
16
|
Chang EL, Liu R, Keyhanian K, Huynh K, Berkenstock M, Bhatti MT, Chen JJ, Chodosh J, Costello F, Dalvin LA, DeLott LB, Dinkin M, Egan RA, Fraser CL, Freitag SK, Gangaputra S, Gordon LK, Guidon AC, Johnson DB, Kombo N, Kramer M, Lee AG, Levy M, Lobo-Chan AM, Mantopoulos D, Papaliodis G, Pless M, Pimkina J, Rubin KM, Sen HN, Shariff A, Subramanian PS, Tsui E, Yoon MK, McDunn J, Rine J, Reynolds KL, Sobrin L, Chwalisz BK. Consensus disease definitions for ophthalmic immune-related adverse events of immune checkpoint inhibitors. J Immunother Cancer 2025; 13:e011049. [PMID: 40199607 PMCID: PMC11979595 DOI: 10.1136/jitc-2024-011049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/20/2025] [Indexed: 04/10/2025] Open
Abstract
Ophthalmic immune-related adverse events (Eye-irAEs) from immune checkpoint inhibitors can cause visual morbidity. The absence of standardized definitions for Eye-irAEs not only impedes the development of evidence-based treatments but also progress in translational research. The objective of this study was to develop consensus guidance for an approach to Eye-irAEs.Four ophthalmic physicians (uveitis specialists and neuro-ophthalmologists) drafted Eye-irAE consensus guidance and definitions, which were reviewed by the multidisciplinary Eye-irAE definition panel. The panel was divided into Group A (Neuro-ophthalmology/Orbital Disease) and Group B (Uveitis/Ocular Surface Disease). A modified Delphi consensus process was used, with two rounds of anonymous ratings by panelists and two meetings to discuss areas of controversy. For each disorder, five diagnostic components were evaluated: symptoms, examination findings, laboratory studies/imaging findings, diagnostic criteria, and treatment. Panelists rated content for usability, appropriateness and accuracy on 9-point scales in electronic surveys and provided free-text comments. Aggregated survey responses were incorporated into revised definitions. Consensus was based on numeric ratings using the RAND Corporation/ University of California Los Angeles Health Services Utilization Study (RAND/UCLA) Appropriateness Method with prespecified definitions.29 panelists from 25 academic medical centers voted on 114 rating scales (66 neuro-ophthalmic/orbital disease components, 48 uveitis/ocular surface disease components); of these, 86.3% (57/66) in Group A and 89.6% (43/48) in Group B reached first-round consensus. After revisions, all items except 6.1% (4/66) in Group A and 1.6% (1/60) in Group B received second-round consensus. Consensus definitions were achieved for 10/11 neuro-ophthalmic/orbital disorders: optic neuritis, inflammatory optic disc edema, arteritic ischemic optic neuropathy, optic perineuritis, orbital inflammation, thyroid eye disease-like orbital inflammation, cavernous sinus syndrome, oculomotor mononeuritis, trochlear mononeuritis, and abducens mononeuritis. Consensus definitions were achieved for 9/10 uveitis/ocular surface disorders: anterior uveitis, intermediate uveitis, posterior uveitis, panuveitis, Vogt-Koyanagi-Harada-like syndrome, sarcoidosis-like syndrome, acute macular neuroretinopathy, dry eye disease, and scleritis.These disease definitions establish a standardized classification for Eye-irAE, highlighting differences between irAEs and other inflammatory disorders. Importantly, diagnostic certainty does not always align directly with the need to treat as an Eye-irAE. Given the consensus from this representative panel group, it is anticipated the definitions will be used broadly across clinical and research settings.
Collapse
Affiliation(s)
- Eileen L Chang
- Department of Ophthalmology, Mass Eye and Ear, Boston, Massachusetts, USA
- Department of Ophthalmology, Weill Cornell Medical College, New York, New York, USA
| | - Renee Liu
- Department of Ophthalmology, Mass Eye and Ear, Boston, Massachusetts, USA
| | - Kiandokht Keyhanian
- Department of Ophthalmology, Mass Eye and Ear, Boston, Massachusetts, USA
- Department of Neurology, Hackensack Meridian Hackensack University Medical Center, Hackensack, New Jersey, USA
| | - Katie Huynh
- Department of Ophthalmology, Mass Eye and Ear, Boston, Massachusetts, USA
| | - Meghan Berkenstock
- Department of Ophthalmology, Johns Hopkins Medicine Wilmer Eye Institute, Baltimore, Maryland, USA
| | - M Tariq Bhatti
- Department of Ophthalmology, Kaiser Permanente Northern California, Oakland, California, USA
| | - John J Chen
- Departments of Ophthalmology and Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - James Chodosh
- Department of Ophthalmology and Visual Sciences, University of New Mexico, Albuquerque, New Mexico, USA
| | - Fiona Costello
- Departments of Clinical Neurosciences and Surgery, University of Calgary Cumming School of Medicine, Calgary, Alberta, Canada
| | - Lauren A Dalvin
- Department of Ophthalmology, Mayo Clinic, Rochester, Minnesota, USA
| | - Lindsey B DeLott
- Department of Ophthalmology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Marc Dinkin
- Departments of Ophthalmology and Neurology, Weill Cornell Medical College, New York, New York, USA
| | - Robert A Egan
- Department of Neurology, PeaceHealth, Bellingham, Washington, USA
| | - Clare L Fraser
- Save Sight Institute, Faculty of Health and Medicine, University of Sydney, Glebe, New South Wales, Australia
| | - Suzanne K Freitag
- Department of Ophthalmology, Mass Eye and Ear, Boston, Massachusetts, USA
| | - Sapna Gangaputra
- Department of Ophthalmology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Lynn K Gordon
- Department of Ophthalmology, David Geffen School of Medicine, Los Angeles, California, USA
| | - Amanda C Guidon
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ninani Kombo
- Department of Ophthalmology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Michal Kramer
- Department of Ophthalmology, Rabin Medical Center, Tel-Aviv University, Tel Aviv, Israel
| | - Andrew G Lee
- Department of Ophthalmology, Houston Methodist Hospital, Houston, Texas, USA
| | - Michael Levy
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Anne-Marie Lobo-Chan
- Department of Ophthalmology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Dimosthenis Mantopoulos
- Department of Ophthalmology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - George Papaliodis
- Department of Ophthalmology, Mass Eye and Ear, Boston, Massachusetts, USA
| | - Misha Pless
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Julia Pimkina
- Division of Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Krista M Rubin
- Department of Medicine, Mass General Cancer Center, Boston, Massachusetts, USA
| | - H Nida Sen
- National Eye Institute, Bethesda, Maryland, USA
| | - Afreen Shariff
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina, USA
| | - Prem S Subramanian
- Department of Ophthalmology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Edmund Tsui
- Department of Ophthalmology, David Geffen School of Medicine, Los Angeles, California, USA
| | - Michael K Yoon
- Department of Ophthalmology, Mass Eye and Ear, Boston, Massachusetts, USA
| | - Jon McDunn
- Project Data Sphere LLC, Cary, North Carolina, USA
| | | | - Kerry L Reynolds
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lucia Sobrin
- Department of Ophthalmology, Mass Eye and Ear, Boston, Massachusetts, USA
| | - Bart K Chwalisz
- Department of Ophthalmology, Mass Eye and Ear, Boston, Massachusetts, USA
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Xu H, Qiao X, Liang J, Qiu L, Xue L, Fang Y, Xiang H, Li X, Chen Y, Ding H. Organomolecular Ferroelectric Nanocatalyst Augments Tumor Immunotherapy by Inducing Apoptosis and Ferroptosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2417422. [PMID: 39995362 DOI: 10.1002/adma.202417422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/06/2025] [Indexed: 02/26/2025]
Abstract
Immunogenic programmed cell death effectively triggers acute inflammatory responses, thereby enhancing antitumor immunity. The advancement of biodegradable nonmetallic dual inducers represents a promising strategy. Herein, a biodegradable organomolecular ferroelectric nanoplatform (C60-TCNQ, CT) is designed to facilitate effective ferroelectric catalysis, thereby augmenting tumor immunotherapy through apoptosis and ferroptosis. CT-mediated ultrasound-triggered ferroelectric catalysis promotes ferroelectric polarization and significantly increases the production of reactive oxygen species, leading to substantial tumor cell apoptosis. Moreover, the polycyano group of CT nanoparticles selectively reacts with cysteine under mild conditions, resulting in redox imbalances and the accumulation of lipid peroxides, which contribute to the induction of ferroptosis in tumor cells. Additionally, the apoptosis and ferroptosis induced by CT stimulate immunogenic cell death progression, eliciting robust immune responses. In vivo evaluation using a bilateral tumor model demonstrates the capacity of CT to sensitize anti-PD-L1 therapy under ultrasound irradiation, achieving an impressive antitumor response rate of 96.2% against malignant melanoma and an 80% inhibition of tumor metastasis. RNA sequencing analysis revealed that treatment with CT resulted in a downregulation of gene signatures associated with the immune-related Jak-Stat signaling pathway. This study opens a novel avenue to developing organomolecular ferroelectric nanomedicines for effective tumor immunotherapy.
Collapse
Affiliation(s)
- Huning Xu
- Department of Ultrasound National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xiaohui Qiao
- Department of Ultrasound National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jing Liang
- Department of Ultrasound National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Luping Qiu
- Department of Ultrasound National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Liyun Xue
- Department of Ultrasound National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yan Fang
- Department of Ultrasound National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Huijing Xiang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Xingguang Li
- Shanghai Key Laboratory of Functional Materials Chemistry, Key Laboratory for Advanced Materials, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Hong Ding
- Department of Ultrasound National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China
| |
Collapse
|
18
|
Okui J, Nagashima K, Matsuda S, Sato Y, Kawakubo H, Takeuchi M, Hirata K, Yamamoto S, Nomura M, Tsushima T, Takeuchi H, Kato K, Kitagawa Y. Investigating the synergistic effects of immunochemotherapy in esophageal squamous cell carcinoma. Esophagus 2025; 22:188-197. [PMID: 39966261 DOI: 10.1007/s10388-025-01113-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Although combinations of immune-checkpoint inhibitors (ICI) with chemotherapy have been approved for esophageal squamous cell carcinoma (ESCC), it remains unclear whether immunochemotherapy (ICT) offers advantages over the simple addition of individual monotherapies. This study aimed to investigate whether ICT exhibits a synergistic effect in patients with advanced ESCC. METHODS Reconstructed individual patient data of 3330 patients were electronically extracted from the Kaplan-Meier (KM) curves of eight randomized-controlled trials (ATTRACTION-3, CheckMate648, KEYNOTE-181, KEYNOTE-590, RATIONALE-302, RATIONALE-306, ESCORT, and ESCORT-1st). The observed progression-free survival (PFS) curve of each constituent monotherapies was used to estimate simulated PFS curves expected under a model of independent drug action. If the observed curve demonstrated significantly better PFS than the simulated curve, the combination of ICI and chemotherapy may have a synergistic effect, implying a superior outcome compared to simply adding the component monotherapy. RESULTS The 1-year, 2-year, and median PFS of the observed and simulated KM curves were 26.3% vs. 24.8%, 14.6% vs. 12.0%, and 6.9 vs. 6.4 months, respectively. The one-sample log-rank test showed no significant differences between the observed and simulated KM curves (p = 0.073). CONCLUSIONS The observed PFS with ICT was comparable to the simulated PFS estimated from the data for each monotherapy. Although it is unclear whether potential synergies exist for ICT, these findings suggest that the benefits of ICI and chemotherapy do not interfere with each other, thereby providing theoretical support for the efficacy of ICT.
Collapse
Affiliation(s)
- Jun Okui
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
- Department of Biostatistics, Keio University School of Medicine, Tokyo, Japan
| | - Kengo Nagashima
- Biostatistics Unit, Clinical and Translational Research Center, Keio University Hospital, Tokyo, Japan
| | - Satoru Matsuda
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan.
| | - Yasunori Sato
- Department of Biostatistics, Keio University School of Medicine, Tokyo, Japan
| | - Hirofumi Kawakubo
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masashi Takeuchi
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Kenro Hirata
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shun Yamamoto
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Motoo Nomura
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Takahiro Tsushima
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Hiroya Takeuchi
- Department of Surgery, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Ken Kato
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
19
|
Gao Y, Huo Y, Wang L, Ruan J, Chen L, Li H, Hong G. Relative expression orderings based prediction of treatment response to Anti-PD-1 immunotherapy in advanced melanoma. Sci Rep 2025; 15:10235. [PMID: 40133499 PMCID: PMC11937249 DOI: 10.1038/s41598-025-94931-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/18/2025] [Indexed: 03/27/2025] Open
Abstract
Programmed cell death protein 1 (PD-1) plays a critical role in immune tolerance and evasion within the tumor microenvironment, and anti-PD-1 immunotherapy has shown efficacy in treating advanced melanoma. However, response rates vary significantly among patients, necessitating the identification of reliable biomarkers to predict treatment efficacy. Based on within-sample relative expression orderings, we analyzed RNA sequencing data from melanoma patients to construct a predictive model comprising gene pairs associated with treatment response. The model's performance was validated across multiple independent datasets and assessed for correlations with immune infiltration and survival outcomes. The constructed 15-pair model achieved a prediction accuracy of 100% in training datasets and 89.47% in validation sets. Validation in melanoma patients lacking treatment response data revealed significant differences between predicted responders and non-responders across datasets, with the model being an independent prognostic factor. Increased immune cell infiltration was observed in responders, correlating with higher expression levels of key immune checkpoint genes. The relative expression orderings-based model shows promise as a tool for predicting responses to anti-PD-1 therapy in melanoma patients, supporting personalized treatment strategies.
Collapse
Affiliation(s)
- Yaru Gao
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, 341000, China
| | - Yue Huo
- School of Public Health and Health Management, Gannan Medical University, Ganzhou, 341000, China
| | - Lingli Wang
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou, 341000, China
| | - Jiayi Ruan
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou, 341000, China
| | - Lanzhen Chen
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou, 341000, China
| | - Hongdong Li
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou, 341000, China.
| | - Guini Hong
- School of Medical and Information Engineering, Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
20
|
Liu C, Liu X, Cao P, Xin H, Li X, Zhu S. Circadian rhythm related genes identified through tumorigenesis and immune infiltration-guided strategies as predictors of prognosis, immunotherapy response, and candidate drugs in skin cutaneous malignant melanoma. Front Immunol 2025; 16:1513750. [PMID: 40191195 PMCID: PMC11968383 DOI: 10.3389/fimmu.2025.1513750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 02/20/2025] [Indexed: 04/09/2025] Open
Abstract
Background Skin cutaneous malignant melanoma (SKCM) is among the most aggressive forms of skin cancer, notorious for its rapid progression and poor prognosis under late diagnosis. This study investigates the role of circadian rhythm-related genes (CRGs) in SKCM addressing a gap in understanding how CRGs affect tumor progression and patient outcomes. Methods An analysis of CRGs expression was conducted on SKCM samples derived from The Cancer Genome Atlas datasets(TCGA). Moreover, a correlation between various subtypes and their clinical features was identified. The study employed various bioinformatics methods, including differential expression analysis, consensus clustering, and survival analysis, to investigate the role of CRGs. The functional consequences of various CRG expression patterns were further investigated using immune infiltration analysis and gene set variation analysis (GSVA). A scoring system based on CRGs was developed to predict overall survival (OS) and treatment responses in SKCM patients. The predictive accuracy of this score system was then tested, and a nomogram was used to improve its clinical usefulness. Results Key findings from this study include significant genetic alterations in circadian rhythm-related genes (CRGs) in skin cutaneous melanoma (SKCM), such as mutations and CNVs. Two molecular subtypes with distinct clinical outcomes and immune profiles were identified. A prognostic model based on six CRGs (CMTM, TNPO1, CTBS, UTRN, HK2, and LIF) was developed and validated with TCGA and GEO datasets, showing high predictive accuracy for overall survival (OS). A high CRGs score correlated with poor OS, immune checkpoint expression, and reduced sensitivity to several chemotherapeutics, including AKT inhibitor VIII and Camptothecin. Conclusions This work provides valuable insights into the circadian regulation of SKCM and underscores the potential of CRGs as biomarkers for prognosis and targets for therapeutic interventions. The novel molecular subtypes and CRGs prognostic scoring model introduced in this study offer significant contributions to the understanding and management of SKCM.
Collapse
Affiliation(s)
- Chengling Liu
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army (PLA), Guilin, China
| | - Xingchen Liu
- Department of Pathology, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Pengjuan Cao
- Department of Endocrinology and Traditional Chinese Medicine, The 924th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army (PLA), Guilin, China
| | - Haiming Xin
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army (PLA), Guilin, China
| | - Xin Li
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army (PLA), Guilin, China
| | - Sailing Zhu
- Center of Burns and Plastic Surgery and Dermatology, The 924th Hospital of Joint Logistics Support Force of the Chinese People's Liberation Army (PLA), Guilin, China
| |
Collapse
|
21
|
Tascón J, Casanova AG, Vicente-Vicente L, López-Hernández FJ, Morales AI. Nephrotoxicity of Immune Checkpoint Inhibitors in Single and Combination Therapy-A Systematic and Critical Review. Biomedicines 2025; 13:711. [PMID: 40149687 PMCID: PMC11940260 DOI: 10.3390/biomedicines13030711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Immune checkpoint inhibitors (ICIs) have generated a revolutionary approach in the treatment of cancer, but their effectiveness has been compromised by immune-related adverse events, including renal damage. Although rare, these effects are relevant because they have been related to poor patient prognoses. The objective of this review was to estimate the current incidence of nephrotoxicity in patients treated with single and double ICI therapies. Methods: A total of 1283 potential articles were identified, which were reduced to 50 after applying the exclusion and inclusion criteria. Results: This study reveals the increase in acute kidney injury associated with these drugs in the last decade and shows that, interestingly, combined therapies with ICIs does not lead to an increase in kidney damage compared with anti-CTLA-4. It also suggests that kidney damage could be underdiagnosed when it comes to interstitial nephritis, because definitive evidence requires a renal biopsy. Conclusions: In perspective, these conclusions could guide clinicians in making decisions for therapy personalization and highlight the need to search for new diagnostic systems that are more sensitive and specific to the type of damage and could replace the biopsy.
Collapse
Affiliation(s)
- Javier Tascón
- Toxicology Unit, Universidad de Salamanca (USAL), 37007 Salamanca, Spain; (J.T.); (A.G.C.); (L.V.-V.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), 37007 Salamanca, Spain
| | - Alfredo G. Casanova
- Toxicology Unit, Universidad de Salamanca (USAL), 37007 Salamanca, Spain; (J.T.); (A.G.C.); (L.V.-V.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), 37007 Salamanca, Spain
| | - Laura Vicente-Vicente
- Toxicology Unit, Universidad de Salamanca (USAL), 37007 Salamanca, Spain; (J.T.); (A.G.C.); (L.V.-V.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), 37007 Salamanca, Spain
| | - Francisco J. López-Hernández
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), 37007 Salamanca, Spain
- Group of Biomedical Research on Critical Care (BioCritic), 47005 Valladolid, Spain
| | - Ana I. Morales
- Toxicology Unit, Universidad de Salamanca (USAL), 37007 Salamanca, Spain; (J.T.); (A.G.C.); (L.V.-V.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), 37007 Salamanca, Spain
- Group of Biomedical Research on Critical Care (BioCritic), 47005 Valladolid, Spain
| |
Collapse
|
22
|
Zhang Y, Liu C, Li Z, Liu Y, Zheng H, Lin Q, Yu L, Boo YJ, Chan BQY, Loh XJ, Wu YL, Song Q, Li P, Chan SY. Microneedle-Mediated Synergistic Photothermal and Chemotherapy for Targeted Melanoma Treatment. ACS APPLIED MATERIALS & INTERFACES 2025; 17:14952-14967. [PMID: 40029948 DOI: 10.1021/acsami.4c20844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Melanoma, a malignant skin tumor originating from melanocytes, is typically treated with surgery in its early stages. However, chemotherapy becomes the primary treatment as the disease progresses to intermediate and advanced stages. The parenteral administration of chemotherapy can cause anxiety, discomfort, and infection risks, especially in immunocompromised cancer patients. Additionally, the circulating drugs can lead to systemic toxicity and side effects. Microneedles (MNs) provide a safer, less invasive alternative to address these issues. Herein, we demonstrated the effectiveness of integrating antimicrobial MNs with combined photothermal and chemotherapy treatment modalities against melanoma, presenting a promising approach to improving cancer treatment outcomes while minimizing associated risks. By leveraging the unique properties of chitosan (CS) and the versatility of poly(vinyl alcohol) (PVA), we fabricated physically cross-linked MNs with inherent antibacterial and antiviral properties. The physical cross-linked network not only accommodated polypyrrole nanoparticles (PPy NPs) for photothermal capabilities but also facilitated drug [doxorubicin hydrochloride (DOX)] loading and release over an extended period. Interestingly, in vitro and in vivo studies revealed that the MNs possess intrinsic antimelanoma properties. Compared to monotherapies, the combination of photothermal therapy and chemotherapy exhibited enhanced effectiveness against melanoma. This research paves the way for safer, more effective cancer treatment strategies.
Collapse
Affiliation(s)
- Yanni Zhang
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Chuyi Liu
- Xiamen University, School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen, Fujian 361102, China
| | - Zhiguo Li
- Xiamen University, School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen, Fujian 361102, China
| | - Yingjia Liu
- Xiamen University, School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen, Fujian 361102, China
| | - Hua Zheng
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi'an, Shaanxi 710129, China
| | - Qianyu Lin
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Luofeng Yu
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Yi Jian Boo
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Benjamin Qi Yu Chan
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
| | - Yun-Long Wu
- Xiamen University, School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, Xiamen, Fujian 361102, China
| | - Qing Song
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
- Strait Institute of Flexible Electronics (SIFE, Future Technologies), Fujian Key Laboratory of Flexible Electronics, Fujian Normal University and Strait Laboratory of Flexible Electronics (SLoFE), Fuzhou, Fujian 350117, China
| | - Peng Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Siew Yin Chan
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Republic of Singapore
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE), & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| |
Collapse
|
23
|
Jiang Y, Bei W, Wang L, Lu N, Xu C, Liang H, Ke L, Ye Y, He S, Dong S, Liu Q, Zhang C, Wang X, Xia W, Zhao C, Huang Y, Xiang Y, Liu G. Efficacy and safety of cadonilimab (PD-1/CTLA-4 bispecific) in combination with chemotherapy in anti-PD-1-resistant recurrent or metastatic nasopharyngeal carcinoma: a single-arm, open-label, phase 2 trial. BMC Med 2025; 23:152. [PMID: 40069710 PMCID: PMC11899053 DOI: 10.1186/s12916-025-03985-4] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND We aimed to evaluate the efficacy and safety of cadonilimab (anti-PD-1 and CTLA-4 bispecific antibody) plus TPC chemotherapy (NAB-paclitaxel, cisplatin or lobaplatin, and capecitabine) in patients with recurrent or metastatic nasopharyngeal carcinoma (RM-NPC) who failed to PD-1 inhibitor-containing regimens. METHODS In this single-arm, open-label, phase 2 study, RM-NPC patients who failed to at least one line of systemic chemotherapy and anti-PD-1 immunotherapy were enrolled and received cadonilimab plus TPC chemotherapy every 3 weeks for up to 6 cycles, followed by cadonilimab plus capecitabine every 3 weeks for a maximum of 2 years. The primary endpoint was the objective response rate (ORR). The secondary endpoints included progression-free survival (PFS), overall survival (OS), duration of response (DoR), and safety. RESULTS Twenty-five patients were enrolled (84% male; median age 44 years (range, 24-60)), with a median follow-up of 10.2 months. The ORR was 68%, with 3 complete responses, 14 partial responses, and 6 stable diseases. The median DoR was 9.1 months (95% CI, 3.8-14.5 months). The median PFS was 10.6 months (95% CI, 5.2-16.0 months). The 12-month OS was 75.6%. Treatment was well tolerated. Grade 3 or 4 treatment-related adverse events occurred in 12 (48%) patients. Fourteen patients (56%) experienced potentially immune-related adverse events (irAEs). One patient experienced a grade 3 immune-related rash and another patient had grade 3 immune-related lipase increased. No treatment-related death occurred. CONCLUSIONS Cadonilimab in combination with TPC chemotherapy demonstrated promising antitumoral efficacy and manageable toxicities in patients with RM-NPC who failed frontline immunotherapy. Further trials are warranted to confirm and expand these findings. TRIAL REGISTRATION This trial was registered at chictr.org.cn (ChiCTR2200067057).
Collapse
Affiliation(s)
- Yaofei Jiang
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
- Department of Oncology, The First Affiliated Hospital of Nanchang University, NanChang, China
| | - Weixin Bei
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Lin Wang
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Nian Lu
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Cheng Xu
- Department of Radiotherapy, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Hu Liang
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Liangru Ke
- Department of Radiology, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Yanfang Ye
- Clinical Research Design Division, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Shuiqing He
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Shuhui Dong
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Qin Liu
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Chuanrun Zhang
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Xuguang Wang
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Weixiong Xia
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Chong Zhao
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Ying Huang
- Department of Radiotherapy, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Yanqun Xiang
- Department of Nasopharyngeal Carcinoma, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, China.
| | - Guoying Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Department of Oncology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
24
|
Ward FJ, Kennedy PT, Al-Fatyan F, Dahal LN, Abu-Eid R. CTLA-4-two pathways to anti-tumour immunity? IMMUNOTHERAPY ADVANCES 2025; 5:ltaf008. [PMID: 40265076 PMCID: PMC12012449 DOI: 10.1093/immadv/ltaf008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 03/02/2025] [Indexed: 04/24/2025] Open
Abstract
Immune checkpoint inhibitor (ICI) therapies have revolutionized cancer therapy and improved patient outcomes in a range of cancers. ICIs enhance anti-tumour immunity by targeting the inhibitory checkpoint receptors CTLA-4, PD-1, PD-L1, and LAG-3. Despite their success, efficacy, and tolerance vary between patients, raising new challenges to improve these therapies. These could be addressed by the identification of robust biomarkers to predict patient outcome and a more complete understanding of how ICIs affect and are affected by the tumour microenvironment (TME). Despite being the first ICIs to be introduced, anti-CTLA-4 antibodies have underperformed compared with antibodies that target the PD-1/PDL-1 axis. This is due to the complexity regarding their precise mechanism of action, with two possible routes to efficacy identified. The first is a direct enhancement of effector T-cell responses through simple blockade of CTLA-4-'releasing the brakes', while the second requires prior elimination of regulatory T cells (TREG) to allow emergence of T-cell-mediated destruction of tumour cells. We examine evidence indicating both mechanisms exist but offer different antagonistic characteristics. Further, we investigate the potential of the soluble isoform of CTLA-4, sCTLA-4, as a confounding factor for current therapies, but also as a therapeutic for delivering antigen-specific anti-tumour immunity.
Collapse
Affiliation(s)
- Frank J Ward
- Medical Sciences and Nutrition, Institute of Medical Sciences, School of Medicine, University of Aberdeen, Aberdeen, United Kingdom
| | - Paul T Kennedy
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Farah Al-Fatyan
- Medical Sciences and Nutrition, Institute of Medical Sciences, School of Medicine, University of Aberdeen, Aberdeen, United Kingdom
| | - Lekh N Dahal
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, United Kingdom
| | - Rasha Abu-Eid
- Medical Sciences and Nutrition, Institute of Dentistry, School of Medicine, Sciences & Nutrition, University of Aberdeen, Aberdeen, United Kingdom
- School of Dentistry, College of Medicine and Health, The University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
25
|
Carneiro BA, Jotte RM, Gabrail NY, Wentzel K, Huang F, Chaturvedi S, Weispfenning A, Hiemeyer F, Morcos PN, Mongay Soler L, Childs BH, Hansen AR. Safety and Efficacy of Copanlisib in Combination with Nivolumab: A Phase Ib Study in Patients with Advanced Solid Tumors. CANCER RESEARCH COMMUNICATIONS 2025; 5:444-457. [PMID: 39927513 PMCID: PMC11907410 DOI: 10.1158/2767-9764.crc-24-0407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 11/01/2024] [Accepted: 02/05/2025] [Indexed: 02/11/2025]
Abstract
PURPOSE Copanlisib in combination with immune checkpoint inhibitors demonstrated synergy and favorable antitumor immune responses in preclinical models. This study evaluated copanlisib plus nivolumab in adults with advanced solid tumors. PATIENTS AND METHODS In this phase Ib, nonrandomized, open-label, dose-escalation study, patients received intravenous nivolumab 240 mg (day 15 of cycle 1 and days 1 and 15 of subsequent cycles) plus intravenous copanlisib (45 or 60 mg on days 1, 8, and 15 of each cycle) in 28-day cycles. The primary objective was to determine the MTD and/or recommended phase II dose of copanlisib plus nivolumab. Secondary objectives were safety, tolerability, and efficacy. Exploratory objectives included evaluation of potentially predictive biomarkers. RESULTS Overall, 16 patients were treated [copanlisib: 45 mg (n = 5); 60 mg (n = 11)]. The most common cancer types at baseline were bladder (25.0%) and oropharyngeal (18.8%) cancers. No dose-limiting toxicities were observed; copanlisib 60 mg was deemed the recommended phase II dose in combination with nivolumab 240 mg. Grade 3 and 4 treatment-emergent adverse events were reported in 56.3% and 12.5% of patients, respectively; one grade 5 event was reported (unrelated to treatment). Overall, 18.8% of patients achieved a partial response. Evaluations of potential biomarkers did not correlate with response, but copanlisib-modulated biomarker changes were observed before nivolumab administration and were consistent and dose-dependent. CONCLUSIONS No new safety concerns were identified with this combination, and preliminary efficacy indicated an antitumor effect. Data supported an immunomodulatory effect of copanlisib, suggesting that this combination may enhance the efficacy of immune checkpoint inhibitors. SIGNIFICANCE The combination of copanlisib and nivolumab was well tolerated and showed antitumor effects in patients with advanced solid tumors. The number of circulating myeloid-derived suppressive cells decreased 24 to 48 hours after treatment with copanlisib. Further investigation of copanlisib and nivolumab is warranted as a novel strategy to enhance the efficacy of checkpoint inhibitors.
Collapse
Affiliation(s)
- Benedito A. Carneiro
- Legorreta Cancer Center at Brown University, Lifespan Cancer Institute, Providence, Rhode Island
| | - Robert M. Jotte
- Rocky Mountain Cancer Centers, Denver, Colorado
- US Oncology Research, Houston, Texas
| | | | - Kristopher Wentzel
- The Angeles Clinic and Research Institute, A Cedars-Sinai Affiliate, Los Angeles, California
| | - Funan Huang
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, New Jersey
| | | | | | | | - Peter N. Morcos
- Bayer HealthCare Pharmaceuticals, Inc., Whippany, New Jersey
| | | | | | | |
Collapse
|
26
|
Wang Y, Liu C, Pang J, Li Z, Zhang J, Dong L. The Extra-Tumoral Vaccine Effects of Apoptotic Bodies in the Advancement of Cancer Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2410503. [PMID: 39871756 PMCID: PMC11878267 DOI: 10.1002/smll.202410503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/16/2025] [Indexed: 01/29/2025]
Abstract
The induction of apoptosis in tumor cells is a common target for the development of anti-tumor therapies; however, these therapies still leave patients at increased risk of disease recurrence. For example, apoptotic tumor cells can promote tumor growth and immune evasion via the secretion of metabolites, apoptotic extracellular vesicles, and induction of pro-tumorigenic macrophages. This paradox of apoptosis induction and the pro-tumorigenic effects of tumor cell apoptosis has begged the question of whether apoptosis is a suitable cancer therapy, and led to further explorations into other immunogenic cell death-based approaches. However, these strategies still face multiple challenges, the most critical of which is the tumor microenvironment. Contrary to the promotion of immune tolerance mediated by apoptotic tumor cells, apoptotic bodies with enriched tumor-related antigens have demonstrated great immunogenic potential, as evidenced by their ability to initiate systemic T-cell immune responses. These characteristics indicate that apoptotic body-based therapies could be ideal "in situ" extra-tumoral tumor vaccine candidates for the treatment of cancers, and further address the current issues with apoptosis-based or immunotherapy treatments. Although not yet tested clinically, apoptotic body-based vaccines have the potential to better treatment strategies and patient outcomes in the future.
Collapse
Affiliation(s)
- Yulian Wang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Chunyan Liu
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Jiayun Pang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Zhenjiang Li
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Junfeng Zhang
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical BiotechnologySchool of Life SciencesNanjing UniversityNanjingJiangsu210023China
- Chemistry and Biomedicine Innovative CenterNanjing UniversityNanjingJiangsu210023China
| |
Collapse
|
27
|
Tang Y, Shi J, Wang L, Zhang Y, Xu L, Sun T. Development of a predictive model for immune‑related adverse events in patients with cancer. Oncol Lett 2025; 29:103. [PMID: 39736926 PMCID: PMC11683521 DOI: 10.3892/ol.2024.14849] [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: 07/18/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
It is crucial to accurately identify patients with cancer at high risk for immune-related adverse events (irAEs) caused by immune checkpoint inhibitors (ICIs). The present retrospective study analyzed the risk factors for irAEs in 992 patients with cancer treated with ICIs at Xi'an International Medical Center Hospital from December 2021 to December 2023. The patients were categorized into one group that experienced irAEs (n=276) and a control group (n=716) based on the occurrence of irAEs. The clinical characteristics of irAEs group (n=276) and control group (n=716) were analyzed to identify the risk factors of irAEs in patients with cancer. Multivariate regression analysis revealed significant differences between the two groups in terms of hypertension, primary cancer, metastasis, targeted drug combination and radiotherapy (P<0.05). A nomogram predictive model for irAEs was developed based on the relevant risk factors. The predictive model for irAEs in patients with cancer yielded an area under the receiver operating characteristic (ROC) curve of 0.672 (95% confidence interval: 0.630-0.714). In the validation set, the Hosmer-Lemeshow goodness-of-fit test demonstrated a favorable fit with a chi-square value of 0.787 and a P-value of 0.978. The developed predictive model can effectively identify high-risk patients with irAEs, facilitate early identification of irAEs, thereby optimizing the management strategies of irAEs, and ultimately improving the quality of life for patients.
Collapse
Affiliation(s)
- Yajuan Tang
- Department of Pharmacy, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, P.R. China
| | - Jinping Shi
- Department of Pharmacy, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, P.R. China
| | - Liping Wang
- Department of Pharmacy, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, P.R. China
| | - Yan Zhang
- Department of Pharmacy, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, P.R. China
| | - Liting Xu
- Department of Pharmacy, Xi'an International Medical Center Hospital, Xi'an, Shaanxi 710100, P.R. China
| | - Tao Sun
- Department of Pharmacy, The Second Affiliated Hospital of Air Force Medical University, Xi'an, Shaanxi 710038, P.R. China
| |
Collapse
|
28
|
Song P, Pan G, Zhang Y, Ni Y, Wang Q, Shi J, Peng Y, Jing R, Luo D. Prospects and Challenges of Immunotherapy for Thyroid Cancer. Endocr Pract 2025; 31:373-379. [PMID: 39631664 DOI: 10.1016/j.eprac.2024.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/21/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
BACKGROUND Thyroid cancer generally boasts a favorable prognosis; however, advanced and refractory cases exhibit aggressive characteristics and resistance to conventional therapies, necessitating the investigation of innovative treatment modalities. Immunotherapy, which harnesses the body's immune system to target cancer cells, has shown considerable promise for specific thyroid cancer subtypes. OBJECTIVE This review article aims to encapsulate the latest advancements in immunotherapy for thyroid cancer, examining its mechanisms, therapeutic efficacy, ongoing challenges, and the potential benefits of combination therapy approaches. METHODS An extensive literature review and critical analysis of clinical trial data were conducted to inform this synthesis. RESULTS The review reveals that immunotherapy strategies, encompassing immune checkpoint inhibitors, CAR-T cell therapy, tumor vaccines, and immunomodulators, are demonstrating efficacy in the treatment of thyroid cancer. Notably, checkpoint inhibitors have been particularly effective in anaplastic and poorly differentiated thyroid cancers, albeit with challenges such as treatment resistance and adverse effects. The application of CAR-T cell therapy, successful in hematologic cancers, provides a novel perspective for thyroid cancer treatment, although its efficacy in solid tumors requires further study. Additionally, research into tumor vaccines and immunomodulators is advancing, with preliminary evidence suggesting their therapeutic potential for thyroid cancer patients. CONCLUSION The recognition of the immune microenvironment's role in treatment responsiveness is pivotal for enhancing the care of thyroid cancer patients. This review underscores the significance of combination therapy as a means to optimize treatment outcomes and charts a course for future research endeavors to broaden the spectrum of effective treatment options available to thyroid cancer patients.
Collapse
Affiliation(s)
- Ping Song
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Gang Pan
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Yu Zhang
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Yeqin Ni
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Qianyu Wang
- The Fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jingjng Shi
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - You Peng
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Ruirui Jing
- Department of Translational Medicine and Clinical Research, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Dingcun Luo
- Department of Surgical Oncology, Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China; The Fourth Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; College of Mathematical Medicine, Zhejiang Normal University, Jinhua, Zhejiang, China.
| |
Collapse
|
29
|
Cheng X, Zhao H, Li Z, Yan L, Min Q, Wu Q, Zhan Q. Integrative analysis of T cell-mediated tumor killing-related genes reveals KIF11 as a novel therapeutic target in esophageal squamous cell carcinoma. J Transl Med 2025; 23:197. [PMID: 39966857 PMCID: PMC11834232 DOI: 10.1186/s12967-025-06178-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 01/25/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are emerging promising agents for the treatment of patients with esophageal squamous cell carcinoma (ESCC), however, there are only a small proportion respond to ICI therapy. Therefore, selecting candidate patients who will benefit the most from these drugs is critical. However, validated biomarkers for predicting immunotherapy response and overall survival are lacking. As the fundamental principle of ICI therapy is T cell-mediated tumor killing (TTK), we aimed to develop a unique TTK-related gene prognostic index (TTKPI) for predicting survival outcomes and responses to immune-based therapy in ESCC patients. METHODS Transcriptomic and clinical information of ESCC patients were from the GSE53625, GSE53624, GSE47404 and TCGA datasets. TTK-related genes were from the TISIDB database. The LASSO Cox regression model was employed to create the TTKPI. The prediction potential of the TTKPI was evaluated using the KM curve and time-dependent ROC curve analysis. Finally, the relationship between TTKPI and immunotherapy efficacy was investigated in clinical trials of ICIs (GSE91061, GSE135222, IMvigor210 cohort). The role of KIF11 in accelerating tumor progression was validated via a variety of functional experiments, including western blot, CCK-8, colony formation, wound healing scratch, and xenograft tumor model. The KIF11 expression was detected by multiplex fluorescent immunohistochemistry on tissue microarray from ESCC patients. RESULTS We constructed the TTKPI based on 8 TTK-related genes. The TTKPI low-risk patients exhibited better overall survival. TTKPI was significantly and positively correlated with the main immune checkpoint molecules levels. Furthermore, the low-risk patients were more prone to reap the benefits of immunotherapy in the cohort undergoing anti-PD-L1 therapy. Moreover, we performed functional experiments on KIF11, which ranked as the most significant prognostic risk gene among the 8 TTK-related genes. Our findings identified that KIF11 knockdown significantly hindered cell proliferation and mobility in ESCC cells. The KIF11 expression was negatively related with CD8+ T cell infiltration in ESCC patient samples. CONCLUSIONS The TTKPI is a promising biomarker for accurately determining survival and predicting the effectiveness of immunotherapy in ESCC patients. This risk indicator can help patients receive timely and precise early intervention, thereby advancing personalized medicine and facilitating precise immuno-oncology research. KIF11 plays a crucial role in driving tumor proliferation and migration and may act as a potential tumor biomarker of ESCC.
Collapse
Affiliation(s)
- Xinxin Cheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Huihui Zhao
- Department of Medical Oncology and Department of Talent Highland, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Zhangwang Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Liping Yan
- Institute of Cytology and Genetics, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Qingjie Min
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Qingnan Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| | - Qimin Zhan
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
- Peking University International Cancer Institute, Beijing, 100142, China.
| |
Collapse
|
30
|
Dadey RE, Cui J, Rajasundaram D, Yano H, Liu C, Cohen JA, Liu AW, Kaplan DH, Workman CJ, Vignali DAA. Regulatory T cells in the tumor microenvironment display a unique chromatin accessibility profile. Immunohorizons 2025; 9:vlae014. [PMID: 39965167 PMCID: PMC11841976 DOI: 10.1093/immhor/vlae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 10/23/2024] [Indexed: 02/20/2025] Open
Abstract
Regulatory T cells (Tregs) are a suppressive CD4+ T cell population that limit the antitumor immune response. In this study, we analyzed the chromatin accessibility of Tregs in the murine tumor microenvironment (TME) to identify tumor-specific accessible peaks and if these are altered over time in the tumor microenvironment, with or without anti-PD-1 immunotherapy. We found that despite little change in chromatin accessibility of Tregs in the tumor over time, Tregs have a distinct chromatin accessibility signature in the TME compared with Tregs in the periphery. This distinct tumor Treg chromatin accessibility profile highlights reduced accessibility at loci important for an CD4+ conventional T cell (CD4+ Foxp3-) effector phenotype. Analysis of chromatin accessibility in Tregs from B16 and MC38 tumor models indicated that Tregs from skin-resident tumors are most similar to naïve skin resident Tregs but still bear key differences attributable to the TME. We also found that Tregs do not alter their transcriptome or chromatin accessibility following immunotherapy. We conclude that although chromatin accessibility in Tregs is somewhat similar to their tissue residency, the TME may drive a unique chromatin accessibility profile. Treg chromatin accessibility in the tumor appears remarkably stable and unaltered by tumor type, over time, or following immunotherapy.
Collapse
Affiliation(s)
- Rebekah E Dadey
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jian Cui
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Dhivyaa Rajasundaram
- Division of Health Informatics, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Hiroshi Yano
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Graduate Program of Microbiology and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Chang Liu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Jonathan A Cohen
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Andrew W Liu
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Daniel H Kaplan
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Creg J Workman
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
- Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, United States
| |
Collapse
|
31
|
Jiramonai L, Liang XJ, Zhu M. Extracellular Vesicle-Based Strategies for Tumor Immunotherapy. Pharmaceutics 2025; 17:257. [PMID: 40006624 PMCID: PMC11859549 DOI: 10.3390/pharmaceutics17020257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/26/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
Immunotherapy is one of the most promising approaches for cancer management, as it utilizes the intrinsic immune response to target cancer cells. Normally, the human body uses its immune system as a defense mechanism to detect and eliminate foreign objects, including cancer cells. However, cancers develop a 'switch off' mechanism, known as immune checkpoint proteins, to evade immune surveillance and suppress immune activation. Therefore, significant efforts have been made to develop the strategies for stimulating immune responses against cancers. Among these, the use of extracellular vesicles (EVs) to enhance the anti-tumor immune response has emerged as a particularly promising approach in cancer management. EVs possess several unique properties that elevate the potency in modulating immune responses. This review article provides a comprehensive overview of recent advances in this field, focusing on the strategic usage of EVs to overcome tumor-induced immune tolerance. We discuss the biogenesis and characteristics of EVs, as well as their potential applications in medical contexts. The immune mechanisms within the tumor microenvironment and the strategies employed by cancers to evade immune detection are explored. The roles of EVs in regulating the tumor microenvironment and enhancing immune responses for immunotherapy are also highlighted. Additionally, this article addresses the challenges and future directions for the development of EV-based nanomedicine approaches, aiming to improve cancer immunotherapy outcomes with greater precision and efficacy while minimizing off-target effects.
Collapse
Affiliation(s)
- Luksika Jiramonai
- Chinese Academy of Sciences (CAS), Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS), Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mengliang Zhu
- Chinese Academy of Sciences (CAS), Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China;
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
32
|
Inoue Y, Yano Y, Kushida S, Hirohata S, Yoon S, Yasutomi E, Hirano H, Kim SK, Yoshida R, Ueda Y, Momose K, Hayashi H, Kado T, Nishi K, Tanaka H, Matono T, Yamamoto A, Tei H, Nishioka C, Yagi Y, Tamura S, Sakane T, Ehara M, Kawano M, Kitadai J, Matsuura T, Shiomi Y, Komatsu S, Fukumoto T, Tada T, Kodama Y. Higher Absolute Lymphocyte Counts and Lower Des-γ-Carboxyprothrombin Levels After Treatment Initiation Are Associated With the Clinical Efficacy of Tremelimumab Plus Durvalumab Combination Therapy for Hepatocellular Carcinoma. JGH Open 2025; 9:e70123. [PMID: 39989844 PMCID: PMC11842866 DOI: 10.1002/jgh3.70123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/05/2025] [Accepted: 02/09/2025] [Indexed: 02/25/2025]
Abstract
Background and Aims Tremelimumab plus durvalumab (Dur/Tre) combination therapy is now a first-line systemic therapy for advanced hepatocellular carcinoma (HCC). Because systemic therapy is not effective in some patients, it is clinically important to identify factors that could predict the response to treatment at an early stage. We investigated the factors associated with the response to Dur/Tre for advanced HCC in a clinical setting. Methods Seventy patients (median age 74 years; 61 men) who received Dur/Tre between March 2023 and September 2024 were analyzed. We examined the factors associated with the treatment response, including pretreatment factors and factors early in treatment. Results The median treatment duration was 77.5 (interquartile range [IQR] 28-187) days. The overall response and disease control rates were 25.8% and 58.1%, respectively. The median (IQR) progression-free survival (PFS) and overall survival (OS) were 82 (61-133) and 415 (337-NA) days, respectively. Multivariable analysis revealed that higher absolute lymphocyte count (ALC) and lower des-γ-carboxyprothrombin (DCP) levels were significantly associated with PFS. Receiver operating characteristic curve analysis showed that the cutoff value for ALC after 4 weeks of treatment in relation to clinical efficacy was 1125/mm3. A log-rank test using the Kaplan-Meier method showed that OS was significantly longer in patients with ALC above the cutoff and in patients whose DCP levels decreased after starting treatment. Conclusion Higher ALC and lower DCP levels after treatment initiation were associated with the clinical efficacy of Dur/Tre for advanced HCC.
Collapse
Affiliation(s)
- Yuta Inoue
- Division of GastroenterologyKobe University Graduate School of MedicineKobe CityHyogoJapan
| | - Yoshihiko Yano
- Division of GastroenterologyKobe University Graduate School of MedicineKobe CityHyogoJapan
| | - Saeko Kushida
- Hyogo Prefectural Hyogo Cancer CenterAkashiHyogoJapan
| | - Seiya Hirohata
- Hyogo Prefectural Kakogawa Medical CenterKakogawaHyogoJapan
| | - Seitetsu Yoon
- Hyogo Prefectural Kakogawa Medical CenterKakogawaHyogoJapan
| | | | | | | | | | | | | | | | | | | | | | - Tomomitsu Matono
- Hyogo Prefectural Harima‐Himeji General Medical CenterHimejiHyogoJapan
| | - Atsushi Yamamoto
- Hyogo Prefectural Harima‐Himeji General Medical CenterHimejiHyogoJapan
| | - Hiroshi Tei
- Kobe City Medical Center General HospitalKobeHyogoJapan
| | | | | | - Shoji Tamura
- Division of GastroenterologyKobe University Graduate School of MedicineKobe CityHyogoJapan
| | - Tatsuya Sakane
- Division of GastroenterologyKobe University Graduate School of MedicineKobe CityHyogoJapan
| | - Mayumi Ehara
- Division of GastroenterologyKobe University Graduate School of MedicineKobe CityHyogoJapan
| | - Miki Kawano
- Division of GastroenterologyKobe University Graduate School of MedicineKobe CityHyogoJapan
| | - Jun Kitadai
- Division of GastroenterologyKobe University Graduate School of MedicineKobe CityHyogoJapan
| | - Takanori Matsuura
- Division of GastroenterologyKobe University Graduate School of MedicineKobe CityHyogoJapan
| | - Yuuki Shiomi
- Division of GastroenterologyKobe University Graduate School of MedicineKobe CityHyogoJapan
| | - Shohei Komatsu
- Division of Hepato‐Biliary‐Pancreatic SurgeryKobe University Graduate School of MedicineKobe CityHyogoJapan
| | - Takumi Fukumoto
- Division of Hepato‐Biliary‐Pancreatic SurgeryKobe University Graduate School of MedicineKobe CityHyogoJapan
| | - Toshifumi Tada
- Division of GastroenterologyKobe University Graduate School of MedicineKobe CityHyogoJapan
| | - Yuzo Kodama
- Division of GastroenterologyKobe University Graduate School of MedicineKobe CityHyogoJapan
| |
Collapse
|
33
|
Foerster Y, Mayer K, Wasserer S, Dechant M, Verkhoturova V, Heyer S, Biedermann T, Persa O. Elevated Neutrophil-to-Lymphocyte Ratio Correlates With Liver Metastases and Poor Immunotherapy Response in Stage IV Melanoma. Cancer Med 2025; 14:e70631. [PMID: 39931836 PMCID: PMC11811709 DOI: 10.1002/cam4.70631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 01/02/2025] [Accepted: 01/16/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Immune checkpoint inhibition (ICI) has revolutionized treatment for metastasized melanoma, but many patients remain unresponsive. Concerning potential adverse events, reliable biomarkers to predict ICI response are needed. In this context, neutrophil-to-lymphocyte ratio (NLR) and derived NLR (dNLR) have emerged. Liver metastases also limit ICI efficacy, correlating with diminished overall survival (OS) and progression-free survival (PFS) and may siphon activated T cells from the systemic circulation, creating an 'immune desert state'. We evaluated the predictive role of NLR and dNLR for ICI response and the impact of liver metastases on systemic immunity and treatment efficacy. PATIENTS AND METHODS In this single-center retrospective study, we included 141 stage IV melanoma patients undergoing ICI. NLR and dNLR were calculated from absolute neutrophil count, absolute lymphocyte count, and white blood cell count. RESULTS Elevated NLR and dNLR were associated with poor response to ICI and inferior PFS. Patients with liver metastases exhibited higher NLR and dNLR levels and showed diminished response to ICI. CONCLUSIONS Elevated baseline NLR and dNLR predict poor response to ICI and PFS in stage IV melanoma. Liver metastases are negative predictors for ICI response, with associated higher NLR and dNLR levels potentially contributing to therapy resistance.
Collapse
Affiliation(s)
- Yannick Foerster
- Department of DermatologyTUM School of Medicine and HealthMünchenGermany
| | - Kristine Mayer
- Department of DermatologyTUM School of Medicine and HealthMünchenGermany
| | - Sophia Wasserer
- Department of DermatologyTUM School of Medicine and HealthMünchenGermany
| | - Marta Dechant
- Department of DermatologyTUM School of Medicine and HealthMünchenGermany
| | | | - Sarah Heyer
- Department of DermatologyTUM School of Medicine and HealthMünchenGermany
| | - Tilo Biedermann
- Department of DermatologyTUM School of Medicine and HealthMünchenGermany
| | - Oana‐Diana Persa
- Department of DermatologyTUM School of Medicine and HealthMünchenGermany
| |
Collapse
|
34
|
Höppener DJ, Grünhagen DJ, Eggermont AMM, van der Veldt AAM, Verhoef C. An Overview of Liver Directed Locoregional Therapies. Hematol Oncol Clin North Am 2025; 39:103-123. [PMID: 39510668 DOI: 10.1016/j.hoc.2024.08.010] [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] [Indexed: 11/15/2024]
Abstract
An overview of all liver-directed locoregional therapies, including surgical resection for melanoma liver metastases (MLMs), is provided. MLM patients are divided by their primary melanoma location; cutaneous, uvea (eye), and mucosal melanoma. If patients with isolated cutaneous MLMs are considered for surgical resection, treatment with systemic therapy should be part of the treatment course. For uveal MLMs, complete surgical or ablative treatment of all MLMs suggests superior results compared with other liver-directed or systemic therapies, based on current evidence, no recommendations for any liver-directed regional therapy in the treatment of mucosal MLMs can be made.
Collapse
Affiliation(s)
- Diederik J Höppener
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Dirk J Grünhagen
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Alexander M M Eggermont
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS, Utrecht, the Netherlands
| | - Astrid A M van der Veldt
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands
| | - Cornelis Verhoef
- Department of Surgical Oncology and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3015 GD, Rotterdam, the Netherlands.
| |
Collapse
|
35
|
Zhou X, Cheng S, Chen Z, Zhang J, Wang J, Li Q, Zhou X. Inhibiting HnRNP L-mediated alternative splicing of EIF4G1 counteracts immune checkpoint blockade resistance in Castration-resistant prostate Cancer. Neoplasia 2025; 60:101109. [PMID: 39724754 PMCID: PMC11731738 DOI: 10.1016/j.neo.2024.101109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 12/28/2024]
Abstract
Immunotherapy with checkpoint inhibitors produced significant clinical responses in a subset of cancer patients who were resistant to prior therapies. However, Castration-resistant prostate cancer (CRPC) is seriously lack of T cell infiltration, which greatly limits the clinical application of immunotherapy, but the mechanism is unclear. In the present study, in silico analyses and experimental data show that HnRNP L was significantly negatively correlated with CD4+ and CD8+ T cells infiltration in patients; besides, we found deficiency of HnRNP L recruites CD4+ and CD8+ T cells infiltration and impairs tumorigenesis. Mechanically, HnRNP L enhanced the translation of c-Myc and then promoted CXCL8 secretion via alternative splicing of EIF4G1. In vivo, inhibition of EIF4G1 by the inhibitor, SBI-0640756, attenuated HnRNP l-induced tumor progression and immunosuppressive activity. And most of all, therapeutic synergy between HnRNP L knockdown and Anti-PD-1 could significantly suppress xenograft prostate cancer growth. In summary, this study revealled the molecular mechanism of HnRNP L regulating the immune infiltration, which provides a new theoretical basis for overcoming the limitation of immunotherapy for CRPC.
Collapse
MESH Headings
- Male
- Humans
- Prostatic Neoplasms, Castration-Resistant/genetics
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/metabolism
- Prostatic Neoplasms, Castration-Resistant/immunology
- Animals
- Mice
- Alternative Splicing
- Immune Checkpoint Inhibitors/pharmacology
- Immune Checkpoint Inhibitors/therapeutic use
- Drug Resistance, Neoplasm/genetics
- Eukaryotic Initiation Factor-4G/genetics
- Eukaryotic Initiation Factor-4G/metabolism
- Heterogeneous-Nuclear Ribonucleoprotein L/genetics
- Heterogeneous-Nuclear Ribonucleoprotein L/metabolism
- Heterogeneous-Nuclear Ribonucleoprotein L/antagonists & inhibitors
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Gene Expression Regulation, Neoplastic
- Disease Models, Animal
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
Collapse
Affiliation(s)
- Xumin Zhou
- General Surgery Center Department of Thyroid Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China; Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China
| | - Shilong Cheng
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China
| | - Zhongjie Chen
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China
| | - Jinming Zhang
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Jiaqi Wang
- Department of Spinal Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China
| | - Qiang Li
- General Surgery Center Department of Thyroid Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China.
| | - Xumin Zhou
- General Surgery Center Department of Thyroid Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China; Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, PR China.
| |
Collapse
|
36
|
Pan S, Wang Z. Antiviral therapy can effectively suppress irAEs in HBV positive hepatocellular carcinoma treated with ICIs: validation based on multi machine learning. Front Immunol 2025; 15:1516524. [PMID: 39931579 PMCID: PMC11807960 DOI: 10.3389/fimmu.2024.1516524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 12/30/2024] [Indexed: 02/13/2025] Open
Abstract
Background Immune checkpoint inhibitors have proven efficacy against hepatitis B-virus positive hepatocellular. However, Immunotherapy-related adverse reactions are still a major challenge faced by tumor immunotherapy, so it is urgent to establish new methods to effectively predict immunotherapy-related adverse reactions. Objective Multi-machine learning model were constructed to screen the risk factors for irAEs in ICIs for the treatment of HBV-related hepatocellular and build a prediction model for the occurrence of clinical IRAEs. Methods Data from 274 hepatitis B virus positive tumor patients who received PD-1 or/and CTLA4 inhibitor treatment and had immune cell detection results were collected from Henan Cancer Hospital for retrospective analysis. Models were established using Lasso, RSF (RandomForest), and xgBoost, with ten-fold cross-validation and resampling methods used to ensure model reliability. The impact of influencing factors on irAEs (immune-related adverse events) was validated using Decision Curve Analysis (DCA). Both uni/multivariable analysis were accomplished by Chi-square/Fisher's exact tests. The accuracy of the model is verified in the DCA curve. Results A total of 274 HBV-related liver cancer patients were enrolled in the study. Predictive models were constructed using three machine learning algorithms to analyze and statistically evaluate clinical characteristics, including immune cell data. The accuracy of the Lasso regression model was 0.864, XGBoost achieved 0.903, and RandomForest reached 0.961. Resampling internal validation revealed that RandomForest had the highest recall rate (AUC = 0.892). Based on machine learning-selected indicators, antiviral therapy and The HBV DNA copy number showed a significant correlation with both the occurrence and severity of irAEs. Antiviral therapy notably reduced the incidence of IRAEs and may modulate these events through regulation of B cells. The DCA model also demonstrated strong predictive performance. Effective control of viral load through antiviral therapy significantly mitigates the occurrence of irAEs. Conclusion ICIs show therapeutic potential in the treatment of HBV-HCC. Following antiviral therapy, the incidence of severe irAEs decreases. Even in cases where viral load control is incomplete, continuous antiviral treatment can still mitigate the occurrence of irAEs.
Collapse
Affiliation(s)
| | - Zibing Wang
- Department of Immunotherapy, The Affiliated Cancer Hospital of Zhengzhou University
& Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
37
|
Xia Y, Huang C, Zhong M, Zhong H, Ruan R, Xiong J, Yao Y, Zhou J, Deng J. Targeting HGF/c-MET signaling to regulate the tumor microenvironment: Implications for counteracting tumor immune evasion. Cell Commun Signal 2025; 23:46. [PMID: 39856684 PMCID: PMC11762533 DOI: 10.1186/s12964-025-02033-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
The hepatocyte growth factor (HGF) along with its receptor (c-MET) are crucial in preserving standard cellular physiological activities, and imbalances in the c-MET signaling pathway can lead to the development and advancement of tumors. It has been extensively demonstrated that immune checkpoint inhibitors (ICIs) can result in prolonged remission in certain patients. Nevertheless, numerous preclinical studies have shown that MET imbalance hinders the effectiveness of anti-PD-1/PD-L1 treatments through various mechanisms. Consequently, clarifying the link between the c-MET signaling pathway and the tumor microenvironment (TME), as well as uncovering the effects of anti-MET treatment on ICI therapy, is crucial for enhancing the outlook for tumor patients. In this review, we examine the impact of abnormal activation of the HGF/c-MET signaling pathway on the control of the TME and the processes governing PD-L1 expression in cancer cells. The review thoroughly examines both clinical and practical evidence regarding the use of c-MET inhibitors alongside PD-1/PD-L1 inhibitors, emphasizing that focusing on c-MET with immunotherapy enhances the effectiveness of treating MET tumors exhibiting elevated PD-L1 expression.
Collapse
Affiliation(s)
- Yang Xia
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Chunye Huang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Min Zhong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Hongguang Zhong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Ruiwen Ruan
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Yangyang Yao
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| | - Jing Zhou
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| |
Collapse
|
38
|
Zhan M, Long Q, He J, Huang L, Wu B, Xu H, Mo L, Xu T. Immune checkpoint inhibitor-induced diabetes mellitus: clinical characteristics and risk factors. Front Immunol 2025; 16:1499074. [PMID: 39925806 PMCID: PMC11802519 DOI: 10.3389/fimmu.2025.1499074] [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: 09/20/2024] [Accepted: 01/09/2025] [Indexed: 02/11/2025] Open
Abstract
Background Emerging evidence indicates that immune checkpoint inhibitor-induced diabetes mellitus (ICI-DM) might be more common than initially reported, and more different clinical pictures associated with ICI-DM were described. Objective The aim of our study was to identify the clinical characteristics and possible predictive factors of ICI-DM. Methods We conducted a retrospective review of patients who received immune checkpoint inhibitors (ICI) at West China Hospital, Sichuan University until June 2023. Patients were reviewed at death or on 7 May 2024. We applied logistic regression to study the associations between clinical characteristics and ICI-DM. Results Our study included 8,199 participants who received ICI between October 2014 and June 2023. Among them, 1,077 patients (13.14%) developed ICI-DM according to diagnostic criteria based on guidelines. By excluding patients influenced by glucocorticoids or immunosuppressants, ICI-DM was observed in 713 of 8,199 (8.70%) patients. In all patients, hypertension, hyperlipidemia, using glucocorticoids or immunosuppressants, lung cancer, and using more than one pathway of ICI were associated with a higher risk of ICI-DM. However, the risk factors for ICI-DM in patients without the influence of glucocorticoids or immunosuppressants were only hypertension, hyperlipidemia, and pancreatic lesions. In all patients and those patients without the influence of glucocorticoids and immunosuppressants, hypertension and hyperlipidemia may increase the risk for ICI-DM. Conclusions This large, real-world cohort demonstrates that the incidence of ICI-DM may be underestimated in previous literature. Blood glucose monitoring is needed in patients receiving ICI therapy. Clinical trial registration https://www.chictr.org.cn, identifier ChiCTR2300075974.
Collapse
Affiliation(s)
- Mei Zhan
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Qinran Long
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jinhan He
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Litao Huang
- Department of Clinical Research Management, West China Hospital, Sichuan University, Chengdu, China
| | - Bin Wu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Haixia Xu
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Li Mo
- Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Xu
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Asashima H, Akao S, Matsumoto I. Emerging roles of checkpoint molecules on B cells. Immunol Med 2025:1-12. [PMID: 39819449 DOI: 10.1080/25785826.2025.2454045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025] Open
Abstract
Immune checkpoint molecules, including both co-inhibitory molecules and co-stimulatory molecules, are known to play critical roles in regulating T-cell responses. During the last decades, immunotherapies targeting these molecules (such as programmed cell death 1 (PD-1), and lymphocyte activation gene 3 (LAG-3)) have provided clinical benefits in many cancers. It is becoming apparent that not only T cells, but also B cells have a capacity to express some checkpoint molecules. These were originally thought to be only the markers for regulatory B cells which produce IL-10, but recent studies suggest that these molecules (especially T-cell immunoglobulin and mucin domain 1 (TIM-1), T cell immunoreceptor with Ig and ITIM domains (TIGIT), and PD-1) can regulate intrinsic B-cell activation and functions. Here, we focus on these molecules and summarize their characteristics, ligands, and functions on B cells.
Collapse
Affiliation(s)
- Hiromitsu Asashima
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Satoshi Akao
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Isao Matsumoto
- Department of Rheumatology, Institute of Medicine, University of Tsukuba, Ibaraki, Japan
| |
Collapse
|
40
|
Zhang Y, Li Y, Zhang Z, Zheng X, Peng H, Tian Z, Sun R, Sun H. CD49a Targeting Enhances NK Cell Function and Antitumor Immunity. Cancer Immunol Res 2025; 13:139-151. [PMID: 39570767 DOI: 10.1158/2326-6066.cir-24-0124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 06/04/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025]
Abstract
Approximately 70% of patients receiving immune checkpoint blockade therapies develop treatment resistance. Thus, there is a need for the identification of additional immunotherapeutic targets. CD49a is a membrane protein expressed on NK cells and T cells. In this study, we found that CD49a was highly expressed on the surface of tumor-infiltrating NK cells in various mouse tumor models and that CD49a+ tumor-infiltrating NK cells were more exhausted than CD49a- tumor-infiltrating NK cells. Furthermore, CD49a or NK-specific CD49a deficiency slowed tumor growth and prolonged survival in several mouse tumor models, primarily through the essential role played by NK cells in antitumor activities. Blockade of CD49a using an mAb suppressed tumor development in mice, and combination treatment with anti-PD-L1 further enhanced antitumor efficacy. Our research reveals CD49a on NK cells as an immunotherapeutic target and highlights the potential clinical applications of CD49a-targeted therapies.
Collapse
Affiliation(s)
- Yu Zhang
- Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, The Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
| | - Yangyang Li
- Hefei TG ImmunoPharma Corporation Limited, Hefei, China
| | - Zhengfeng Zhang
- Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, The Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
| | - Xiaodong Zheng
- Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, The Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
| | - Hui Peng
- Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, The Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
- Division of Life Sciences and Medicine, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China, Hefei, China
- Research Unit of NK Cell Study, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhigang Tian
- Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, The Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
- Research Unit of NK Cell Study, Chinese Academy of Medical Sciences, Beijing, China
| | - Rui Sun
- Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, School of Basic Medical Sciences, The Institute of Immunology and The CAS Key Laboratory of Innate Immunity and Chronic Disease, University of Science and Technology of China, Hefei, China
- Division of Life Sciences and Medicine, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, University of Science and Technology of China, Hefei, China
| | - Haoyu Sun
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Alwhaibi A, Alenazi MA, Alghadeer S, Mansy W, Alsaif RA, Abualreesh NE, Alanazi RJ, Alroumi A, Alanazi SA. A Real-World Comparison of the Safety Profile for Immune Checkpoint Inhibitors in Oncology Patients. J Clin Med 2025; 14:388. [PMID: 39860394 PMCID: PMC11765622 DOI: 10.3390/jcm14020388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/02/2025] [Accepted: 01/05/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Owing to the growing use of immune checkpoint inhibitors (ICIs) in the treatment of cancer, a wide spectrum of toxicity has arisen among cancer patients. Yet, limited ICI toxicity-related research is currently conducted in our region. Methods: This is a retrospective observational study conducted on adult cancer patients who received at least one cycle of ICI single therapy. Toxicity profiles of different ICI monotherapies were described and compared, and their association with different risk factors was assessed. SPSS version 28 was used for statistical analyses, and p < 0.05 was considered statistically significant. Results: A total of 428 patients were treated with anti-PD1 (nivolumab [n = 221, 51.6%] and pembrolizumab [n = 126, 29.5%]) or anti-PD-L1 (atezolizumab [n = 78, 18.2%] and durvalumab [n = 3, 0.7%]). Pneumonia was the most common complication (10%), followed by acute kidney injury (AKI; 8.2%) and hepatitis (7.9%). The proportion of hepatitis cases was significantly higher among atezolizumab compared to nivolumab-, pembrolizumab-, and durvalumab-treated patients (17.95% vs. 7.7% vs. 2.4% vs. 0.0%, respectively; p < 0.001). Gastrointestinal complication (colitis) was detected in 3.3% of patients with a significant difference between treatment groups (4.5%, 1.6%, 1.3%, and 33.3% in nivolumab, pembrolizumab, atezolizumab, and durvalumab, respectively; p = 0.008). Cardiac complications occurred in 1.2% of patients with a significant difference between treatment groups (0.5% in the nivolumab, 3.8% in the atezolizumab, 33.3% in the durvalumab, and none in the pembrolizumab groups (p < 0.001)). Musculoskeletal side effects, including both arthralgia and fatigue, were the most-reported side effects by 39.5% of patients, with significantly higher arthralgia complainers only in nivolumab (7.7%) compared to other treatment groups (0%, 2.6%, and 0% in pembrolizumab, atezolizumab, and durvalumab, respectively, p = 0.007). Hepatic, cardiovascular, hematological, respiratory, renal, gastrointestinal complications, thyroid complications, and dermatological side effects were found to occur on weeks 6, 7.5, 8, 8, 10, 10, 10.5, and 12 after treatment initiation, respectively, with no significant difference between treatment groups. Despite that, hepatitis and AKI tended to occur earlier with atezolizumab (week 2, p = 0.084) and pembrolizumab (week 2, p = 0.062), respectively, compared to their comparators. The female gender and a history of hepatitis were found to increase the odds of hepatic complication with anti-PD1 or anti-PD-L1 use [OR = 2.71; 95% CI 1.07-6.85, OR = 11.14; 95% CI 3.46-35.88, respectively]. Previous exposure to cancer therapy only was found to increase the odds of developing pneumonia among the treated patients [OR = 3.08; 95% CI 1.12-8.85]. Having hematological malignancy influenced the odds of hematological complications positively (either neutropenia or thrombocytopenia) compared to solid malignancies when patients were treated with anti-PD1 or anti-PD-L1 [OR = 17.18; 95% CI 4.06-72.71]. Finally, the female gender was found to positively associate with the odds of nausea/vomiting and fatigue secondary to anti-PD1 or anti-PD-L1 administration [OR = 2.08; 95% CI 1.34-3.21, OR = 1.65; 95% CI 1.09-2.51, respectively]. On the other hand, previous exposure to cancer therapy was found to reduce the risk of having arthralgia with anti-PD1 or anti-PD-L1 administration [OR = 0.344; 95% CI 0.121-0.974]. Conclusions: Treatment with anti-PD1 or anti-PD-L1 was associated with a spectrum of complications and side effects. Several risk factors have been identified to impact their occurrence. ICI toxicities and risk factors influencing their odds should be recognized and considered in clinical practice, as this could help in individualizing therapeutics regimens and avoiding treatment interruption.
Collapse
Affiliation(s)
- Abdulrahman Alwhaibi
- Department of Clinical Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (W.M.); (N.E.A.)
| | - Miteb A. Alenazi
- Pharmacy Department, King Saud University Medical City, Riyadh 11411, Saudi Arabia;
| | - Sultan Alghadeer
- Department of Clinical Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (W.M.); (N.E.A.)
| | - Wael Mansy
- Department of Clinical Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (W.M.); (N.E.A.)
| | - Reem A. Alsaif
- Pharmacy Department, King Abdulaziz Medical City, National Guard Health Affairs, Riyadh 11426, Saudi Arabia;
| | - Nawaf E. Abualreesh
- Department of Clinical Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; (S.A.); (W.M.); (N.E.A.)
| | - Rakan J. Alanazi
- Pharmacy Practice Department, College of Pharmacy, Alfaisal University, Riyadh 11533, Saudi Arabia;
| | - Abdullah Alroumi
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 11461, Saudi Arabia; (A.A.); (S.A.A.)
| | - Saleh A. Alanazi
- College of Pharmacy, King Saud Bin Abdulaziz University for Health Sciences, Riyadh 11461, Saudi Arabia; (A.A.); (S.A.A.)
| |
Collapse
|
42
|
Santiago-Sánchez GS, Fabian KP, Hodge JW. A landscape of checkpoint blockade resistance in cancer: underlying mechanisms and current strategies to overcome resistance. Cancer Biol Ther 2024; 25:2308097. [PMID: 38306161 PMCID: PMC10841019 DOI: 10.1080/15384047.2024.2308097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/17/2024] [Indexed: 02/03/2024] Open
Abstract
The discovery of immune checkpoints and the development of immune checkpoint inhibitors (ICI) have achieved a durable response in advanced-stage cancer patients. However, there is still a high proportion of patients who do not benefit from ICI therapy due to a lack of response when first treated (primary resistance) or detection of disease progression months after objective response is observed (acquired resistance). Here, we review the current FDA-approved ICI for the treatment of certain solid malignancies, evaluate the contrasting responses to checkpoint blockade in different cancer types, explore the known mechanisms associated with checkpoint blockade resistance (CBR), and assess current strategies in the field that seek to overcome these mechanisms. In order to improve current therapies and develop new ones, the immunotherapy field still has an unmet need in identifying other molecules that act as immune checkpoints, and uncovering other mechanisms that promote CBR.
Collapse
Affiliation(s)
- Ginette S. Santiago-Sánchez
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kellsye P. Fabian
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James W. Hodge
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
43
|
Pol S. [Hepatocellular carcinoma (HCC)]. MEDECINE TROPICALE ET SANTE INTERNATIONALE 2024; 4:mtsi.v4i4.2024.614. [PMID: 40070978 PMCID: PMC11892391 DOI: 10.48327/mtsi.v4i4.2024.614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 10/15/2024] [Indexed: 03/14/2025]
Abstract
Primary liver cancers are tumors that develop from different liver cells. Hepatocellular carcinoma (HCC), which develops from hepatocytes, accounts for approximately 75-85% of primary liver cancers.HCC is the 6th leading cause of cancer worldwide and the 3rd leading cause of cancer-related death. Its incidence is low in northern Europe, but high in sub-Saharan Africa and the Far East, where both hepatotropic viruses and exposure to mycotoxins are. It complicates cirrhosis in over 90% of cases and is predominantly male.The prevalence of HCC is increasing due to improved diagnostic techniques and criteria, but also to the persistence of hepatitis B virus (HBV) and hepatitis C virus (HCV) infections in adults. A worldwide increase in the incidence of steatopathy makes it the leading cause of liver disease worldwide, associated with alcohol abuse and/or steatohepatitis associated with metabolic dysfunction (MASH), including type 2 diabetes.Chronic hepatotropic viral infections, cirrhosis and chemical carcinogens combine to produce an annual incidence of 2-5% of hepatocellular carcinoma arising from cirrhosis. This justifies biannual surveillance of known cirrhosis, without which late diagnosis limits therapeutic options.Major advances have been made in curative treatment (liver transplantation, surgery, radiodestruction) and palliative treatment (chemo- or radioembolization, sorafenib chemotherapy or immunotherapy), depending on how early HCC is diagnosed (size, number of hepatic or extrahepatic lesions) and the severity of underlying liver disease and associated comorbidities.
Collapse
Affiliation(s)
- Stanislas Pol
- AP-HP. Centre Université Paris Centre, Groupe hospitalier Cochin Port Royal, Département médical universitaire de Cancérologie et spécialités médico-chirurgicales, Service des maladies du foie, Paris, France; Université Paris Cité, F-75006, Paris, France
| |
Collapse
|
44
|
Chen CB, Hung SI, Chang JWC, Yang CK, Ma DHK, Teng YC, Lu CW, Chen WT, Yang HY, Tsai CC, Wang CL, Chiang PH, Wu J, Tsai YW, Lu LY, Lin YYW, Hui RCY, Hsieh FM, Hsu CK, Lee CN, Chen YJ, Chen CC, Cui Y, Hsu HC, Chang YC, Chang CJ, Lin HC, Chang CJ, Lin YJ, Ku CL, Wang CW, Chung WH. Immune checkpoint inhibitor-induced severe epidermal necrolysis mediated by macrophage-derived CXCL10 and abated by TNF blockade. Nat Commun 2024; 15:10733. [PMID: 39737932 PMCID: PMC11685864 DOI: 10.1038/s41467-024-54180-7] [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: 01/04/2024] [Accepted: 11/05/2024] [Indexed: 01/01/2025] Open
Abstract
Immune checkpoint inhibitors (ICI) represent new anticancer agents and have been used worldwide. However, ICI can potentially induce life-threatening severe cutaneous adverse reaction (SCAR), such as Stevens-Johnson syndrome/toxic epidermal necrolysis (SJS/TEN), hindering continuous ICI therapy. We examine 6 cohorts including 25 ICI-induced SJS/TEN patients and conduct single-cell RNA sequencing (scRNA-seq) analysis, which shows overexpression of macrophage-derived CXCL10 that recruits CXCR3+ cytotoxic T lymphocytes (CTL) in blister cells from ICI-SJS/TEN skin lesions. ScRNA expression profiles and ex vivo blocking studies further identify TNF signaling as a pathway responsible for macrophage-derived CXCL10 and CTL activation. Based on the trajectory analysis, ICI-activated T cells from whole blood are proposed to serve as the initial cells involved in inflammation, that lead to monocytes differentiating into macrophages and increasing their susceptibility to migrate to the lesion sites. Compared with systemic corticosteroids treatment, ICI-induced SJS/TEN patients treated with biologic TNF blockade showed a significantly rapid recovery and no recurrence of SCAR with continuous ICI therapy. Our findings identify that macrophage-eliciting CTL contribute to the pathogenesis of ICI-induced epidermal necrolysis and provide potential therapeutic targets for the management and prevention of SCAR induced by ICI therapy.
Collapse
Affiliation(s)
- Chun-Bing Chen
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Keelung Branch, Keelung, Taiwan
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Chang Gung Immunology Consortium, Chang Gung University, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China
- Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen, China
| | - Shuen-Iu Hung
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - John Wen-Cheng Chang
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Hematology-Oncology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Chan-Keng Yang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Division of Hematology-Oncology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - David Hui-Kang Ma
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Limbal Stem Cell Laboratory, Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Yu-Chuan Teng
- Genomic Medicine Core Laboratory, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chun-Wei Lu
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wei-Ti Chen
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Hsiao-Yin Yang
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Cheng-Chang Tsai
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Chih Liang Wang
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Division of Pulmonary Oncology and Interventional Bronchoscopy, Department of Thoracic Medicine, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Pin-Hsuan Chiang
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Dermatology, New Taipei Municipal TuCheng Hospital, New Taipei City, Taiwan
| | - Jennifer Wu
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Ya-Wen Tsai
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Lai-Ying Lu
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Yang Yu-Wei Lin
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Rosaline Chung-Yee Hui
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Keelung Branch, Keelung, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | | | - Chao-Kai Hsu
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chaw-Ning Lee
- Department of Dermatology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ju Chen
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Dermatology, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chih-Chiang Chen
- Institute of Clinical Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Department of Dermatology, Taipei Veterans General Hospital, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yilei Cui
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Hung-Chih Hsu
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Division of Hematology-Oncology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Ya-Ching Chang
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chih-Jung Chang
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China
- Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen, China
- Medical Research Center, Xiamen Chang Gung Hospital, Xiamen, China
- School of Medicine, Huaqiao University, Quanzhou, China
| | - Ho-Chen Lin
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan
| | - Chee Jen Chang
- Research Services Center for Health Information, Chang Gung University, Taoyuan, Taiwan
- Clinical Informatics and Medical Statistics Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Jr Lin
- Research Services Center for Health Information, Chang Gung University, Taoyuan, Taiwan
- Clinical Informatics and Medical Statistics Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Lung Ku
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan.
- Chang Gung Immunology Consortium, Chang Gung University, Taoyuan, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Laboratory of Human Immunology and Infectious Disease, Graduate Institute of Clinical Medical Sciences, Chang Gung University, Taoyuan, Taiwan.
- Center for Molecular and Clinical and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Division of Infectious Diseases, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan.
| | - Chuang-Wei Wang
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan.
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan.
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan.
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan.
- Chang Gung Immunology Consortium, Chang Gung University, Taoyuan, Taiwan.
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China.
- Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen, China.
- Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Wen-Hung Chung
- Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan.
- Department of Dermatology, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan.
- Department of Dermatology, Chang Gung Memorial Hospital, Keelung Branch, Keelung, Taiwan.
- Cancer Vaccine and Immune Cell Therapy Core Laboratory, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan.
- Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan.
- Chang Gung Immunology Consortium, Chang Gung University, Taoyuan, Taiwan.
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Whole-Genome Research Core Laboratory of Human Diseases, Chang Gung Memorial Hospital, Keelung, Taiwan.
- Immune-Oncology Center of Excellence, Chang Gung Memorial Hospital, Linkou Branch, Taoyuan, Taiwan.
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan.
- Department of Dermatology, Xiamen Chang Gung Hospital, Xiamen, China.
- Xiamen Chang Gung Allergology Consortium, Xiamen Chang Gung Hospital, Xiamen, China.
- Department of Dermatology, Beijing Tsinghua Chang Gung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, China.
- Department of Dermatology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
45
|
Hu K, Jin L, Zhou Y, Xie G. Pharmacologically induced autoimmune encephalitis-disproportionality analysis utilizing FAERS database. Expert Opin Drug Saf 2024:1-9. [PMID: 39714109 DOI: 10.1080/14740338.2024.2446425] [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: 05/30/2024] [Revised: 10/08/2024] [Accepted: 10/18/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Autoimmune encephalitis (AE) is a neuroimmune disorder that presents significant diagnostic challenges. The FDA Adverse Event Reporting System (FAERS) database can help explore the relationship between drugs and AE, but comprehensive studies are lacking. This study aims to analyze the association between drugs and AE using the FAERS database, providing insights for clinical practice and pharmacovigilance. RESEARCH DESIGN AND METHODS Adverse event reports in the FAERS database were analyzed, focusing on the incidence of drug-induced AE, as well as characteristics such as gender and age. Multiple statistical methods were employed to assess the association between drugs and adverse reactions. RESULTS The study revealed that drug-induced AE predominantly occurred in individuals aged 41 and above, with a higher prevalence among female patients. Nivolumab and pembrolizumab were among the drugs most frequently reported for adverse drug reactions. However, only a minority of drug labels mentioned these adverse reactions. CONCLUSION This study underscores the potential risk of drug-induced AE, advocating for close monitoring in clinical practice. Further epidemiological investigations are warranted to elucidate the exact relationship between drugs and these disorders. While the FAERS database provides crucial leads for such research, additional studies and validation are necessary.
Collapse
Affiliation(s)
- Kaikai Hu
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo University, Ningbo, China
| | - Liuyin Jin
- Science and Education Department, Lishui Second People's Hospital, Lishui, China
| | - Yixia Zhou
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo University, Ningbo, China
| | - Guoming Xie
- Department of Neurology, The Affiliated Lihuili Hospital of Ningbo University, Ningbo University, Ningbo, China
| |
Collapse
|
46
|
Hosseininejad-Chafi M, Eftekhari Z, Oghalaie A, Behdani M, Sotoudeh N, Kazemi-Lomedasht F. Nanobodies as innovative immune checkpoint modulators: advancing cancer immunotherapy. Med Oncol 2024; 42:36. [PMID: 39719469 DOI: 10.1007/s12032-024-02588-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/14/2024] [Indexed: 12/26/2024]
Abstract
The immune system relies on a delicate balance between attacking harmful pathogens and preserving the body's own tissues, a balance maintained by immune checkpoints. These checkpoints play a critical role in preventing autoimmune diseases by restraining excessive immune responses while allowing the immune system to recognize and destroy abnormal cells, such as tumors. In recent years, immune checkpoint inhibitors (ICIs) have become central to cancer therapy, enabling the immune system to target and eliminate cancer cells that evade detection. Traditional antibodies, such as IgGs, have been widely used in immune therapies but are limited by their size and complexity. Nanobodies (Nbs), derived from camelid heavy-chain-only antibodies, offer a promising alternative. These small, stable antibody fragments retain the antigen-binding specificity of traditional antibodies but have enhanced solubility and the ability to target otherwise inaccessible epitopes. This review explores the use of Nbs as ICIs, emphasizing their potential in cancer immunotherapy and other immune-related treatments. Their unique structural properties and small size make Nbs highly effective tools for modulating immune responses, representing a novel approach in the evolving landscape of checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Mohammad Hosseininejad-Chafi
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Zohre Eftekhari
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Akbar Oghalaie
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Nazli Sotoudeh
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran.
| |
Collapse
|
47
|
Acar C, Yüksel HÇ, Şahin G, Açar FP, Tünbekici S, Çelebi G, Karaca B. Efficacy and prognostic factors of anti-PD1 and nivolumab-ipilimumab therapy in advanced melanoma patients resistant to prior ICI treatment. Discov Oncol 2024; 15:813. [PMID: 39704850 DOI: 10.1007/s12672-024-01702-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 12/12/2024] [Indexed: 12/21/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have significantly improved the five-year survival rate for advanced melanoma. However, many patients exhibit resistance to ICI therapy. This study evaluated the efficacy and prognostic factors of anti-PD-1 (Group A) and nivolumab-ipilimumab (Group B) therapy in patients with advanced melanoma who were resistant to prior ICI therapy. We conducted a retrospective analysis of 56 patients with advanced melanoma who had previously shown resistance to ICI therapy. In the Group A (who have previously shown resistance to anti-CTLA-4, n = 28), the objective response rate (ORR) was 42.9%, with a disease control rate (DCR) of 53%. In the Group B (previously shown resistance to anti-PD-1, n = 28), the ORR was 17.9%, and the DCR was 25%. The ORR was lower in two subgroups: patients who showed progression or relapse in the the initial radiological assessment of prior ICI therapy (ORR 10.5%) and patients who had previously received ICI in the adjuvant setting (ORR 8.3%). A Royal Marsden Hospital (RMH) score of 2-3 was a predictor of OS in both groups (Group A: HR 3.789, 95% CI 1.356-10.589, p = 0.011; Group B: HR 4.281, 95% CI 1.490-12.300, p = 0.007) and for PFS in the Group B (HR 3.167, 95% CI 1.062-9.442, p = 0.039). Anti-PD-1 therapy demonstrated efficacy following resistance to anti-CTLA-4, whereas combination ICI therapy showed lower response rates in patients resistant to anti-PD-1. Further studies are needed to confirm the RMH scores and other prognostic markers and to evaluate subgroups with lower efficacy of nivolumab-ipilimumab therapy.
Collapse
Affiliation(s)
- Caner Acar
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, Izmir, 35100, Turkey.
| | - Haydar Çağatay Yüksel
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, Izmir, 35100, Turkey
| | - Gökhan Şahin
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, Izmir, 35100, Turkey
| | - Fatma Pinar Açar
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, Izmir, 35100, Turkey
| | - Salih Tünbekici
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, Izmir, 35100, Turkey
| | - Gülçin Çelebi
- Department of Internal Medicine, Ege University Medical Faculty, Izmir, Turkey, 35100
| | - Burçak Karaca
- Division of Medical Oncology, Department of Internal Medicine, Ege University Medical Faculty, Izmir, 35100, Turkey
| |
Collapse
|
48
|
Zou J, Xu B, Gao H, Luo P, Chen T, Duan H. Microbiome in urologic neoplasms: focusing on tumor immunity. Front Immunol 2024; 15:1507355. [PMID: 39703512 PMCID: PMC11655508 DOI: 10.3389/fimmu.2024.1507355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/18/2024] [Indexed: 12/21/2024] Open
Abstract
Urological tumors are an important disease affecting global human health, and their pathogenesis and treatment have been the focus of medical research. With the in - depth study of microbiomics, the role of the microbiome in urological tumors has gradually attracted attention. However, the current research on tumor - associated microorganisms mostly focuses on one type or one site, and currently, there is a lack of attention to the microbiome in the immunity and immunotherapy of urological tumors. Therefore, in this paper, we systematically review the distribution characteristics of the microbiome (including microorganisms in the gut, urine, and tumor tissues) in urologic tumors, the relationship with disease prognosis, and the potential mechanisms of microbial roles in immunotherapy. In particular, we focus on the molecular mechanisms by which the microbiome at different sites influences tumor immunity through multiple "messengers" and pathways. We aim to further deepen the understanding of microbiome mechanisms in urologic tumors, and also point out the direction for the future development of immunotherapy for urologic tumors.
Collapse
Affiliation(s)
- Jun Zou
- Department of Otorhinolaryngology, The Affiliated Fengcheng Hospital of Yichun University, Fengcheng, Jiangxi, China
| | - Baisheng Xu
- Department of Urology, The First People's Hospital of Xiushui, Jiujiang, Jiangxi, China
| | - Hongbing Gao
- Department of Urology, The First People's Hospital of Xiushui, Jiujiang, Jiangxi, China
| | - Peiyue Luo
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Tao Chen
- The First Clinical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Huanglin Duan
- Department of Urology, The First People's Hospital of Xiushui, Jiujiang, Jiangxi, China
| |
Collapse
|
49
|
Lee Y, Won HS, Seo KJ, Na SJ. FDG PET Images of Pseudoprogression After Nivolumab-FOLFOX Chemotherapy in a Gastric Cancer Patient. Clin Nucl Med 2024; 49:1139-1141. [PMID: 39485872 DOI: 10.1097/rlu.0000000000005515] [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: 11/03/2024]
Abstract
ABSTRACT A 60-year-old man diagnosed with gastric adenocarcinoma underwent FDG PET/CT. After completing 9 cycles of combination chemotherapy with nivolumab and FOLFOX, he had multiple new nodular uptakes in the bilateral cervical chains and mediastinum. The pathology of the right neck node confirmed reactive lymph node hyperplasia without evidence of malignancy, suggesting pseudoprogression after immunotherapy. It is worthwhile to report these PET image patterns, as they could significantly influence clinical decision-making.
Collapse
Affiliation(s)
- Yeongjoo Lee
- From the Division of Nuclear Medicine, Department of Radiology
| | - Hye Sung Won
- Division of Medical Oncology, Department of Internal Medicine
| | - Kyung Jin Seo
- Department of Hospital Pathology, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sae Jung Na
- From the Division of Nuclear Medicine, Department of Radiology
| |
Collapse
|
50
|
Lei T, Cai X, Zhang H, Wu X, Cao Z, Li W, Xie X, Zhang B. Bmal1 upregulates ATG5 expression to promote autophagy in skin cutaneous melanoma. Cell Signal 2024; 124:111439. [PMID: 39343115 DOI: 10.1016/j.cellsig.2024.111439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/14/2024] [Accepted: 09/26/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND Skin cutaneous melanoma (SKCM) is a highly aggressive and malignant tumor that arises from the malignant transformation of melanocytes. In light of the limitations of existing treatment modalities, there is a pressing need to identify new drug targets for SKCM. Aryl-hydrocarbon receptor nuclear translocator-like (ARNTL), also known as Bmal1, is a gene that has been linked to the onset and progression of cancer. However, its role in SKCM remains understudied. METHODS The expression of Bmal1 mRNA and protein was detected using TCGA, GTEx, CCLE, and ULCAN databases. Moreover, survival analysis was performed to investigate the association between Bmal1 and immune invasion and gene expression in immune infiltrating cells via CIBERSORT, R programming, TIMER, Sangerbox, Kaplan-Meier. The study also explored the role of proteins associated with Bmal1 by using R programming and databases (STRING and GSEA). Both in vitro and in vivo studies were conducted to examine the potential role of Bmal1 in SKCM. RESULTS Compared to normal tissues, the expression level of Bmal1 was significantly reduced in SKCM. Which has been associated with its poor prognosis. Similarly, its expression in SKCM was substantially correlated with immune infiltration, while biogenic analysis indicated that it could potentially influence the tumor immune microenvironment (TME) by influencing tumor-associated neutrophils (TANs). Moreover, Bmal1 overexpression suppressed the proliferation and invasion of melanoma cells and enhanced apoptosis, migration, and cell colony formation. CONCLUSION This study concluded that Bmal1 is a novel biomarker that functions as both a diagnostic and prognostic indicator for the progression of SKCM.
Collapse
Affiliation(s)
- Tao Lei
- Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Xin Cai
- Guangzhou Medical University, Guangzhou, Guangdong, PR China
| | - Hao Zhang
- The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, People's Republic of China
| | - Xunping Wu
- Guizhou Provincial People's Hospital Central Laboratory, Guiyang 550002, China
| | - Zhimin Cao
- Beijing Tuberculosis and Thoracic Tumor Institute, Beijing 101149, China
| | - Wen Li
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People`s Hospital, China; Key Laboratory of Pulmonary Immune Diseases, National Health Commission, Guiyang 550002, China
| | - Xingming Xie
- Guizhou Institute of Precision Medicine, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, Guizhou, People's Republic of China.
| | - Bangyan Zhang
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People`s Hospital, China; Key Laboratory of Pulmonary Immune Diseases, National Health Commission, Guiyang 550002, China..
| |
Collapse
|