1
|
Ying H, Chen Y, Hong Y, Ying K, Li S, Zhang Y, Mei T, Song X, He Y, Yao C, Yu F. L3-SMI as a predictor of overall survival in oesophageal cancer patients receiving PD-1 inhibitors combined with chemotherapy. Ann Med 2025; 57:2440114. [PMID: 39665392 PMCID: PMC11639058 DOI: 10.1080/07853890.2024.2440114] [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: 07/22/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Programmed death ligand-1 (PD-1), as an immunotherapy target, has been increasingly used in tumour therapies. But as reactions and outcomes to PD-1 inhibitors combined with chemotherapy vary individually, it is primarily important to identify an ideal indicator for predicting the therapeutic effectiveness in individual patients. Oesophageal cancer (EC) patients often have difficulty eating due to tumour blockage of the oesophagus, leading to malnutrition and muscle loss. Sarcopenia is one of the influencing factors for poor prognosis in tumour patients, but its role in PD-1 inhibitors combined with chemotherapy of EC patients is not fully clarified. In this study, we aimed to explore the prognostic significance of Sarcopenia measured by CT in EC patients treated with PD-1 antibody combined with chemotherapy. METHODS The third lumbar skeletal muscle mass index (L3-SMI) was obtained from 83 EC patients before and 3 months after administration of PD-1 inhibitors combined with chemotherapy using conventional CT scans. RESULTS Baseline L3-SMI and 3-month L3-SMI values were found not suitable for predicting the overall survival (OS) of EC patients (p = 0.32 & p = 0.055). Longitudinal change in L3-SMI (ΔL3-SMI) during PD-1 inhibitors combined with chemotherapy was identified as a relevant marker of OS in univariable analysis (HR: 0.98, 95% CI: 0.96-1.00, p = 0.042) and multivariable analysis (HR: 0.96, 95% CI: 0.93-0.99, p = 0.02). L3-SMI-positive patients generally had better OS (p = 0.041). CONCLUSION Excessive muscle loss rather than muscle loss before and after administration of PD-1 inhibitors combined with chemotherapy is an important prognostic factor for therapeutic outcomes and OS in EC patients.
Collapse
Affiliation(s)
- Huiya Ying
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuhao Chen
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yiwen Hong
- WenZhou Medical University, Wenzhou, Zhejiang, China
| | - Kanglei Ying
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shiyu Li
- WenZhou Medical University, Wenzhou, Zhejiang, China
| | - Yuxuan Zhang
- WenZhou Medical University, Wenzhou, Zhejiang, China
| | - Tianhao Mei
- WenZhou Medical University, Wenzhou, Zhejiang, China
| | - Xian Song
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuanhang He
- WenZhou Medical University, Wenzhou, Zhejiang, China
| | - Chenrui Yao
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Fujun Yu
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
2
|
Zhang L, Li MJ, Li XP, Yang B, Xiao T, Wang P, Zhang WD. Respiratory microbiota diversity as a predictive biomarker for the efficacy of PD‑1 blockades in patients with advanced non‑small cell lung cancer: A retrospective exploratory study. Oncol Lett 2025; 29:251. [PMID: 40201032 PMCID: PMC11977453 DOI: 10.3892/ol.2025.14997] [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: 09/07/2024] [Accepted: 03/04/2025] [Indexed: 04/10/2025] Open
Abstract
Despite advancements in immunotherapy, particularly regarding programmed cell death protein 1 (PD-1)/programmed death-ligand 1 blockades, the clinical outcomes in non-small cell lung cancer (NSCLC) remain variable with limited predictive biomarkers currently available. The present study investigated respiratory microbiota diversity as a potential biomarker to predict the efficacy of PD-1 blockades in patients with advanced NSCLC. A retrospective analysis was conducted on 60 patients treated with PD-1 blockades from May 2019 to May 2023. Clinical data were collected and respiratory microbiota from deep induced sputum specimens were analyzed using 16S rRNA gene sequencing. An index of respiratory microbiota α diversity was applied and exploratory analysis was performed accordingly. The objective response rate (ORR) and disease control rate among the 60 patients receiving PD-1 blockades was 23.3% (95% CI, 13.4-36.0%) and 58.3% (95% CI, 44.9-70.9%), respectively. Analysis of prognostic data of patients with advanced NSCLC receiving PD-1 blockades monotherapy demonstrated a median progression-free survival of 3.4 months (95% CI, 2.54-4.26) and a median overall survival (OS) of 12.3 months (95% CI, 6.29-18.31). Patients were stratified into high and low α diversity groups based on the Shannon diversity index of respiratory microbiota. The ORR was increased in the high diversity group (26.7%) compared with that of the low diversity group (20.0%), although the difference was not statistically significant (P=0.542). Notably, the high diversity group demonstrated a longer median PFS (3.9 vs. 2.8 months; P=0.017) and median OS (16.8 vs. 6.8 months; P=0.016) compared with that of the low diversity group. These findings suggested that PD-1 blockades demonstrate promising therapeutic activity for patients with previously treated advanced NSCLC in clinical practice. Respiratory microbiota α diversity might serve as a potential biomarker to predict the efficacy of PD-1 blockades monotherapy in patients with advanced NSCLC in the future. Therefore, further prospective studies are warranted to validate these findings and to explore the underlying mechanisms by which respiratory microbiota might modulate the immune response to cancer therapy.
Collapse
Affiliation(s)
- Liang Zhang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin 300190, P.R. China
| | - Ming-Jiang Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin 300190, P.R. China
| | - Xiao-Ping Li
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin 300190, P.R. China
| | - Bo Yang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin 300190, P.R. China
| | - Ting Xiao
- Department of Pharmacy, College of Pharmacy and Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300071, P.R. China
| | - Ping Wang
- Department of Radiotherapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300202, P.R. China
| | - Wei-Dong Zhang
- Department of Thoracic Surgery, Tianjin First Central Hospital, Tianjin 300190, P.R. China
| |
Collapse
|
3
|
Tareco Bucho TM, Pascucci L, Mancò G, Taraschi A, Macchia MC, Tinari N, Brocco D, Beets-Tan RGH, Pizzi AD, Trebeschi S. RECIST progression: Patterns among target, non-target, non-measurable and new lesions progression. Eur J Radiol 2025; 186:112038. [PMID: 40096772 DOI: 10.1016/j.ejrad.2025.112038] [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: 01/14/2025] [Revised: 02/17/2025] [Accepted: 03/06/2025] [Indexed: 03/19/2025]
Abstract
OBJECTIVES This study investigates whether different types of RECIST-defined tumor progression reflect distinct biological processes or variations of the same progression. MATERIALS AND METHODS We collected retrospective data from N = 223 patients (mean age 67 ± 16 years; 151 men) undergoing immunotherapy per standard-of-care, whose treatment decisions did not follow RECIST. Imaging data were gathered for up to two years post-baseline, continuously evaluating RECIST-defined progression types. We analyzed co-occurrence patterns using odds ratios of progression events and assessed prognostic impacts with time-varying Cox regression models. RESULTS Of 223 patients, 115 (52%) experienced at least one type of progression. Most patients (57%) experienced multiple types of progression. Most patients with only one type of progression did not have follow-up scans to determine if other types of progression would have occurred later. In general, all progression types are likely to co-occur, supported by significant positive odds ratios. New lesions were more frequent early in treatment, while other types showed no clear temporal trends. Non-measurable progression had the strongest association with worse survival (HR, 2.33; CI: 1.45-3.74; p < 0.001), followed by new lesions (HR, 1.73; CI: 1.05-2.86; p = 0.03) and target lesion progression (HR, 1.66; CI: 1.07-2.58; p = 0.02). Non-target lesion progression was rare (9%) and not prognostic. CONCLUSION RECIST-defined progression types often co-occur, suggesting they represent variations of a shared treatment failure process rather than independent events. Future developments should focus on quantitative metrics for progression.
Collapse
Affiliation(s)
- Teresa M Tareco Bucho
- Department of Radiology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Universiteitssingel 40, 6229ER Maastricht, the Netherlands
| | - Luca Pascucci
- Department of Radiology, Santissima Annunziata Hospital, University "G. D'Annunzio", Via dei Vestini, 66100 Chieti, Italy
| | - Giovanni Mancò
- Department of Radiology, Santissima Annunziata Hospital, University "G. D'Annunzio", Via dei Vestini, 66100 Chieti, Italy
| | - Alessio Taraschi
- Department of Radiology, Santissima Annunziata Hospital, University "G. D'Annunzio", Via dei Vestini, 66100 Chieti, Italy
| | - Maria Claudia Macchia
- Department of Radiology, Santissima Annunziata Hospital, University "G. D'Annunzio", Via dei Vestini, 66100 Chieti, Italy
| | - Nicola Tinari
- Department of Medical, Oral & Biotechnological Sciences, University "G. D'Annunzio", Via dei Vestini, 66100 Chieti, Italy
| | - Davide Brocco
- Department of Medical, Oral & Biotechnological Sciences, University "G. D'Annunzio", Via dei Vestini, 66100 Chieti, Italy
| | - Regina G H Beets-Tan
- GROW School for Oncology and Reproduction, Maastricht University, Universiteitssingel 40, 6229ER Maastricht, the Netherlands; Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands; Faculty of Health Sciences, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark
| | - Andrea Delli Pizzi
- Department of Radiology, Santissima Annunziata Hospital, University "G. D'Annunzio", Via dei Vestini, 66100 Chieti, Italy; Department of Innovative Technologies in Medicine and Dentistry, University "G. D'Annunzio", Via dei Vestini, 66100 Chieti, Italy; ITAB Institute for Advanced Biomedical Technologies, University "G. D'Annunzio", Via Luigi Polacchi 11, 66100 Chieti, Italy
| | - Stefano Trebeschi
- Department of Radiology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, the Netherlands; GROW School for Oncology and Reproduction, Maastricht University, Universiteitssingel 40, 6229ER Maastricht, the Netherlands.
| |
Collapse
|
4
|
Pourvaziri A, Mroueh N, Cochran RL, Srinivas Rao S, Kambadakone A. Beyond Conventional CT: Role of Dual-Energy CT in Monitoring Response to Therapy in Abdominal Malignancies. Radiol Imaging Cancer 2025; 7:e240142. [PMID: 40249270 DOI: 10.1148/rycan.240142] [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: 04/19/2025]
Abstract
In the era of precision medicine, imaging plays a critical role in evaluating treatment response to various oncologic therapies. For decades, conventional morphologic assessments using cross-sectional imaging have been the standard for monitoring the effectiveness of systemic and locoregional therapies in patients with cancer. However, the development of new functional imaging tools has widened the scope of imaging from mere response assessment to patient selection and outcome prediction. Dual-energy CT (DECT), known for its superior material differentiation capabilities, shows promise in enhancing treatment response evaluation. DECT-based iodine quantification methods are increasingly being investigated as surrogates for assessing tumor vascularity and physiology, which is particularly important in patients undergoing emerging targeted therapies. The purpose of this review article is to discuss the current and emerging role of DECT in assessing treatment response in patients with malignant abdominal tumors. Keywords: CT-Dual Energy, Transcatheter Tumor Therapy, Tumor Response, Iodine Uptake, Therapeutic Response © RSNA, 2025.
Collapse
Affiliation(s)
- Ali Pourvaziri
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114
| | - Nayla Mroueh
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114
| | - Rory L Cochran
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114
| | - Shravya Srinivas Rao
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114
| | - Avinash Kambadakone
- Department of Radiology, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114
| |
Collapse
|
5
|
Zhong J, Mao S, Chen H, Wang Y, Yin Q, Cen Q, Lu J, Yang J, Hu Y, Xing Y, Liu X, Ge X, Jiang R, Song Y, Lu M, Chu J, Zhang H, Zhang G, Ding D, Yao W. Node-RADS: a systematic review and meta-analysis of diagnostic performance, category-wise malignancy rates, and inter-observer reliability. Eur Radiol 2025; 35:2723-2735. [PMID: 39505734 PMCID: PMC12021726 DOI: 10.1007/s00330-024-11160-1] [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: 04/27/2024] [Revised: 08/02/2024] [Accepted: 09/26/2024] [Indexed: 11/08/2024]
Abstract
OBJECTIVE To perform a systematic review and meta-analysis to estimate diagnostic performance, category-wise malignancy rates, and inter-observer reliability of Node Reporting and Data System 1.0 (Node-RADS). METHODS Five electronic databases were systematically searched for primary studies on the use of Node-RADS to report the possibility of cancer involvement of lymph nodes on CT and MRI from January 1, 2021, until April 15, 2024. The study quality was assessed by modified Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) and Quality Appraisal of Diagnostic Reliability (QAREL) tools. The diagnostic accuracy was estimated with bivariate random-effects model, while the pooled category-wise malignancy rates were obtained with random-effects model. RESULTS Six Node-RADS-CT studies and three Node-RADS-MRI studies covering nine types of cancer were included. The study quality was mainly damaged by inappropriate index test and unknown timing according to QUADAS-2, and unclear blindness during the rating process according to QAREL. The area under hierarchical summary receiver operating characteristic curve (95% conventional interval) was 0.92 (0.89-0.94) for Node-RADS ≥ 3 as positive and 0.91 (0.88-0.93) for Node-RADS ≥ 4 as positive, respectively. The pooled malignancy rates (95% CIs) of Node-RADS 1 to 5 were 4% (0-10%), 31% (9-58%), 55% (34-75%), 89% (73-99%), and 100% (97-100%), respectively. The inter-observer reliability of five studies was interpreted as fair to substantial. CONCLUSION Node-RADS presented a promising diagnostic performance with an increasing probability of malignancy along higher category. However, the evidence for inter-observer reliability of Node-RADS is insufficient, and may hinder its implementation in clinical practice for lymph node assessment. KEY POINTS Question Node-RADS is designed for structured reporting of the possibility of cancer involvement of lymph nodes, but the evidence supporting its application has not been summarized. Findings Node-RADS presented diagnostic performance with AUC of 0.92, and malignancy rates for categories 1-5 ranged from 4% to 100%, while the inter-observer reliability was unclear. Clinical relevance Node-RADS is a useful tool for structured reporting of the possibility of cancer involvement of lymph nodes with high diagnostic performance and appropriate malignancy rate for each category, but unclear inter-observer reliability may hinder its implementation in clinical practice.
Collapse
Affiliation(s)
- Jingyu Zhong
- Department of Imaging, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Shiqi Mao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Haoda Chen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yibin Wang
- Department of Urology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Qian Yin
- Department of Pathology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Qingqing Cen
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Junjie Lu
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, California, 94305, USA
| | - Jiarui Yang
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, 02215, USA
| | - Yangfan Hu
- Department of Imaging, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Yue Xing
- Department of Imaging, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Xianwei Liu
- Department of Imaging, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Xiang Ge
- Department of Imaging, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Run Jiang
- Department of Pharmacovigilance, SciClone Pharmaceuticals (Holdings) Ltd., Shanghai, 200020, China
| | - Yang Song
- MR Research Collaboration Team, Siemens Healthineers Ltd., Shanghai, 200126, China
| | - Minda Lu
- MR Application, Siemens Healthineers Ltd., Shanghai, 200126, China
| | - Jingshen Chu
- Department of Science and Technology Development, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Huan Zhang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University of Medicine, Shanghai, 200025, China
| | - Guangcheng Zhang
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Defang Ding
- Department of Imaging, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China.
| | - Weiwu Yao
- Department of Imaging, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China.
| |
Collapse
|
6
|
Buhtoiarov IN, Minkov M, Vali R, Abla O. Disease response criteria in Langerhans cell histiocytosis: a global view. Int J Hematol 2025:10.1007/s12185-025-03989-z. [PMID: 40287907 DOI: 10.1007/s12185-025-03989-z] [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: 10/31/2024] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025]
Abstract
Langerhans cell histiocytosis (LCH) is an inflammatory myeloid neoplasm with heterogeneous presentations. The discovery of BRAFV600E and other MAPK pathway mutations drastically transformed the treatment landscape, especially for high-risk LCH and CNS-LCH. While treatment strategies for children and adults are somewhat similar, response assessment methodologies remain highly dichotomized.Currently, separate treatment response criteria exist for children and adults, especially in therapeutic trials. Considering the rapid evolution of targeted MAPK-inhibitor therapies, along with ultrasensitive detection of minimal residual disease biomarkers (e.g., circulating BRAFV600E-encoding DNA) and sophisticated imaging tools (18F-FDG-PET and whole-body MRI), harmonization of response criteria in LCH is clearly warranted. The Histiocyte Society Global LCH Treatment Response Harmonization Task Force, a collaborative network of pediatric and adult LCH experts, is set to propose updated pediatric LCH treatment response criteria, which will also serve as the foundation for a universal response assessment tool for pediatric and adult LCH. In this review, we focus on the past, present, and likely future of response assessment in LCH patients, and discuss needs that remain unmet in the targeted therapy era.
Collapse
Affiliation(s)
- Ilia N Buhtoiarov
- Division of Pediatric Hematology, Oncology, and Blood and Marrow Transplantation, Pediatric Institute, Cleveland Clinic Foundation, 9500 Euclid Ave, R3-118, Cleveland, OH, 44195, USA.
| | - Milen Minkov
- Department of Pediatric Hematology/Oncology, University Clinic of Pediatrics, Medical University of Vienna, Vienna, Austria
| | - Reza Vali
- Department of Diagnostic & Interventional Radiology, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Oussama Abla
- Division of Hematology/Oncology, Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| |
Collapse
|
7
|
Li S, Tang J, Chen R, Li Y, Liu S, Chu X, Li L, Liao W. Immunotherapy may promote the occurrence of radiation-induced brain injury in NSCLC patients with brain metastases undergoing radiotherapy: a retrospective propensity score-matching and inverse probability of treatment weighting study. Clin Transl Oncol 2025:10.1007/s12094-025-03928-8. [PMID: 40287912 DOI: 10.1007/s12094-025-03928-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025]
Abstract
PURPOSE The combination of radiotherapy (RT) and immunotherapy can significantly improve the prognosis of non-small cell lung cancer (NSCLC) patients. However, concerns about whether the synergistic effect may increase the risk of radiation-induced brain injury (RIBI) remain controversial. This study aims to explore the impact of immune checkpoint inhibitors (ICIs) on the occurrence of RIBI in patients with NSCLC brain metastases (BMs). METHODS This study retrospectively enrolled NSCLC patients with BMs undergoing RT between January 2017 and December 2023. Patients were stratified into groups based on PD-1/PD-L1 checkpoint inhibitors, administration, with confounding factors controlled via propensity score matching (PSM) and inverse probability of treatment weighting (IPTW). Final cohorts included an RT-only group (n = 54) and an RT + ICIs group (n = 28). RIBI incidence and progression-free survival (PFS) were compared between groups. RESULTS After 1:1 PSM analysis, the incidence of RIBI in the RT + ICIs group was significantly higher than that in the RT-only group (17.9% vs 42.9%, P = 0.042). Further IPTW analysis showed that the incidence of RIBI in the RT + ICIs group was significantly higher than that in the RT-only group (24.8% vs 47.8%, P = 0.033). Regarding the impact on PFS, there was no statistical difference between the two groups in both PSM and IPTW (P > 0.05). CONCLUSIONS Immunotherapy combined with RT may increase the occurrence of RIBI in patients with NSCLC BMs. The mechanism underlying this phenomenon requires further investigation.
Collapse
Affiliation(s)
- Sai Li
- Department of Radiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - JingYi Tang
- Department of Radiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Ruiting Chen
- Department of Radiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Yong Li
- Department of Gastroenterology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Shulin Liu
- Department of Radiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Xianjing Chu
- Department of Oncology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Lang Li
- Department of Radiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Weihua Liao
- Department of Radiology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China.
- FuRong Laboratory, Changsha, 410078, Hunan, China.
| |
Collapse
|
8
|
Jiao W, Zhao L, Mei J, Zhong J, Yu Y, Bi N, Zhang L, Wang L, Fu X, Wang J, Lu S, Liu L, Gao S. Clinical practice guidelines for perioperative multimodality treatment of non-small cell lung cancer. Chin Med J (Engl) 2025:00029330-990000000-01521. [PMID: 40246578 DOI: 10.1097/cm9.0000000000003635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Lung cancer is currently the most prevalent malignancy and the leading cause of cancer deaths worldwide. Although the early stage non-small cell lung cancer (NSCLC) presents a relatively good prognosis, a considerable number of lung cancer cases are still detected and diagnosed at locally advanced or late stages. Surgical treatment combined with perioperative multimodality treatment is the mainstay of treatment for locally advanced NSCLC and has been shown to improve patient survival. Following the standard methods of neoadjuvant therapy, perioperative management, postoperative adjuvant therapy, and other therapeutic strategies are important for improving patients' prognosis and quality of life. However, controversies remain over the perioperative management of NSCLC and presently consensus and standardized guidelines are lacking for addressing critical clinical issues in multimodality treatment. METHODS The working group consisted of 91 multidisciplinary experts from thoracic surgery, medical oncology, radiotherapy, epidemiology, and psychology. This guideline was developed using the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) system. The clinical questions were collected and selected based on preliminary open-ended questionnaires and subsequent discussions during the Guideline Working Group meetings. PubMed, Web of Science, Cochrane Library, Scopus, and China National Knowledge Infrastructure (CNKI) were searched for available evidence. The GRADE system was used to evaluate the quality of evidence and grade the strengths of recommendations. Finally, the recommendations were developed through a structured consensus-building process. RESULTS The Guideline Development Group initially collected a total of 62 important clinical questions. After a series of consensus-building conferences, 24 clinical questions were identified and corresponding recommendation were ultimately developed, focusing on neoadjuvant therapy, perioperative management, adjuvant therapy, postoperative psychological rehabilitation, prognosis assement, and follow-up protocols for NSCLC. CONCLUSIONS This guideline puts forward reasonable recommendations focusing on neoadjuvant therapy, perioperative management, adjuvant therapy, postoperative psychological rehabilitation, prognosis assessment, and follow-up protocol of NSCLC. It standardizes perioperative multimodality treatment and provides guidance for clinical practice among thoracic surgeons, medical oncologists, and radiotherapists, aiming to reduce postoperative recurrence, improve patient survival, accelerate recovery, and minimize postoperative complications such as atelectasis.
Collapse
Affiliation(s)
- Wenjie Jiao
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Liang Zhao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jiandong Mei
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jia Zhong
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yongfeng Yu
- Department of Medical Oncology, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Nan Bi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100021, China
| | - Lan Zhang
- Psychological Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lvhua Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong 518117, China
| | - Xiaolong Fu
- Department of Radiation Oncology, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Jie Wang
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Shun Lu
- Department of Medical Oncology, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Lunxu Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
9
|
Joerger M, Diem S, Wyss N, Koster KL, Besse L, Hess D, Metaxas Y, Flynn MC, Aeppli S, Abou MT, Philip El Saadan T, Mane S, Hiendlmeyer E, von Moos R, Flatz L. Open-label nonrandomized phase IB study to characterize the safety and recommended dose of tinostamustine in combination with nivolumab in patients with advanced melanoma (ENIgMA). Melanoma Res 2025:00008390-990000000-00204. [PMID: 40265641 DOI: 10.1097/cmr.0000000000001040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Tinostamustine is a first-in-class alkylating deacetylase inhibitor that facilitates access to cancer cell DNA, resulting in its damage and counteracting DNA repair systems. We hypothesize that the addition of tinostamustine to immune checkpoint inhibitors (ICIs) improves melanoma treatment. This open-label, nonrandomized phase IB study characterized dose-limiting toxicity (DLT) and the recommended dose (RD) of 2-weekly intravenous tinostamustine at escalating doses of 15 and 30 mg/m2 when administered with 2-weekly nivolumab 3 mg/kg added in cycle 2 in patients with melanoma. We included 17 patients (four at 15 mg/m2 and 13 at 30 mg/m2 tinostamustine). A total of 13/17 (77%) patients were ICI-resistant, 7/17 (41%) had unfavorable melanoma subtypes. No DLT was identified. Tinostamustine RD was 30 mg/m2 every 2 weeks. One patient experienced grade 2 nivolumab-associated immune-related pneumonitis. Tinostamustine-associated grade 3 leukocytopenia was documented in one patient, grade 2 leukocytopenia in five patients, and grade 1 thrombocytopenia in three patients. Treatment discontinuation occurred in one patient for nivolumab-associated immune-related pneumonitis and in another patient for tumor-related hemorrhage. A total of 7/13 (54%) evaluable patients had at least stable disease as best treatment response, including 3/13 (23%) patients with a confirmed partial response. Median progression-free survival was 8.3 weeks [95% confidence interval (CI): 2.4-15.4 weeks), median overall survival was 19.1 weeks (95% CI: 2.4-41 weeks). Two-weekly intravenous tinostamustine at an immune-modulatory dose of 30 mg/m2 is safe when coadministered with nivolumab 3 mg/kg and resulted in 54% disease stabilization and 23% confirmed partial responses in patients with predominantly ICI-resistant, advanced melanoma.
Collapse
Affiliation(s)
- Markus Joerger
- Department of Medical Oncology and Hematology, Cantonal Hospital, St.Gallen
- Medical School, University Hospital, Basel
| | - Stefan Diem
- Department of Medical Oncology and Hematology, Cantonal Hospital, St.Gallen
- Department of Internal Medicine, Regional Hospital, Grabs
| | - Nina Wyss
- Department of Dermatology, Institute of Immunobiology, St.Gallen
- Department of Dermatology
| | - Kira-Lee Koster
- Department of Medical Oncology and Hematology, Cantonal Hospital, St.Gallen
| | - Lenka Besse
- Department of Oncology and Hematology, Laboratory of Experimental Oncology, Cantonal Hospital, St.Gallen, Switzerland
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Dagmar Hess
- Department of Medical Oncology and Hematology, Cantonal Hospital, St.Gallen
| | - Yannis Metaxas
- Department of Oncology/Hematology, Spital Thurgau, Frauenfeld
| | - Marie-Claire Flynn
- Department of Medical Oncology and Hematology, Cantonal Hospital, St.Gallen
| | - Stefanie Aeppli
- Department of Medical Oncology and Hematology, Cantonal Hospital, St.Gallen
| | | | | | - Shrunal Mane
- Department of Dermatology, University Hospital, Tübingen, Germany
| | - Elke Hiendlmeyer
- Clinical Trial Unit (CTU), Cantonal Hospital, St.Gallen, Switzerland
| | | | - Lukas Flatz
- Department of Dermatology, Institute of Immunobiology, St.Gallen
- Department of Dermatology
- Department of Dermatology, University Hospital, Tübingen, Germany
| |
Collapse
|
10
|
Liu M, Gao Y, Li L, Ge L, Yao L, Chu X, Wu F, Tian J. Anti-TIGIT inhibitors plus PD-1 or PD-L1 inhibitors versus PD-1 or PD-L1 inhibitors for first-line treatment of advanced non-small cell lung cancer. Cochrane Database Syst Rev 2025; 4:CD015888. [PMID: 40226904 PMCID: PMC11995688 DOI: 10.1002/14651858.cd015888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
OBJECTIVES This is a protocol for a Cochrane Review (intervention). The objectives are as follows: To evaluate the effectiveness and safety of anti-TIGIT inhibitors in combination with PD-1 inhibitors or PD-L1 inhibitors compared to treatment with PD-1 inhibitors or PD-L1 inhibitors in the first-line treatment of advanced NSCLC. To assess the effectiveness and safety of anti-TIGIT inhibitors in combination with chemotherapy and PD-1 inhibitors or PD-L1 inhibitors compared to treatment with chemotherapy plus PD-1 inhibitors or PD-L1 inhibitors in the first-line treatment of advanced NSCLC.
Collapse
Affiliation(s)
- Ming Liu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou city, China
| | - Ya Gao
- Department of Medical Data, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- Institute for Medical Dataology, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lun Li
- Department of Breast Surgery, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Long Ge
- Evidence-Based Social Science Research Centre, School of Public Health, Lanzhou University, Lanzhou city, China
| | - Liang Yao
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Xiajing Chu
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Fanqi Wu
- Department of Respirator, Lanzhou University Second Hospital, Lanzhou, China
| | - Jinhui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou city, China
| |
Collapse
|
11
|
Cui T, Sun L, Guo X, Cheng C, Zhang N, Zhou S, Chu Q, Xing C, Liang S, Liu Y, Ji C, Li X, Tao S, Gu X, Ma K, Wu C, Chu J, Fu Y, Han S, Zhang Y, Ye J, Liu Y, Wang J, Liu L. Tumor-Derived CD109 Orchestrates Reprogramming of Tumor-Associated Macrophages to Dampen Immune Response. J Hepatol 2025:S0168-8278(25)00231-4. [PMID: 40220905 DOI: 10.1016/j.jhep.2025.03.035] [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: 07/08/2024] [Revised: 03/26/2025] [Accepted: 03/31/2025] [Indexed: 04/14/2025]
Abstract
BACKGROUND & AIMS Despite remarkable advancements in immunotherapy, poor responsiveness in intrahepatic cholangiocarcinoma (iCCA) remains a persistent challenge. Here, we explored the immunosuppressive-related secreted proteins derived from iCCA and the underlying regulatory mechanisms in tumor immune microenvironment (TIME) remodeling, with the aim of developing new targets to inhibit tumor growth and improve the efficacy of immunotherapy. METHODS Proteomic analysis, single-cell transcriptomics, CyTOF, RNA sequencing, and mass spectrometry were conducted to identify the key secreted protein involved in immune suppression and elucidate the underlying biological mechanisms. RESULTS We revealed that tumor-derived soluble CD109 (sCD109) is associated with the immunosuppressive TIME, where elevated sCD109 promotes the enrichment of CD73+ TAMs, hindering T cell immune response. Mechanistically, sCD109 upregulates CD73 mRNA transcription by activating the FcγRI/SYK/NFκB signaling pathway. Meanwhile, sCD109 is internalized into the cytoplasm of macrophages and inhibit the degradation of CD73 protein by binding to the E3 ligase TRIM21, competing with CD73 for its binding site. Dual blockade of CD109 and PD-L1 can improve the infiltration and function of lymphocyte, significantly prolonging the anti-tumor response. CONCLUSIONS Our findings reveal sCD109 as a 'secreted immune checkpoint' that reprograms the TIME and suggest that CD109 inhibition is a valuable strategy to sensitize the effectiveness of iCCA immunotherapy. IMPACT AND IMPLICATIONS Poor response to tumor immunotherapy in patients with intrahepatic cholangiocarcinoma (iCCA) has long been a challenge for clinicians. In this study, we used multiomics approaches to elucidate that tumor cells secrete soluble CD109, which reprograms macrophages, leading to the accumulation of CD73+ macrophages in the tumor immune microenvironment (TIME). This effect significantly inhibits T cell proliferation and the immune response of CD8+ T cells, thereby impairing the efficacy of immunotherapy. In preclinical studies, we demonstrated that targeting CD109 in mice can markedly improve the immunosuppressive TIME, sensitizing iCCA cells to anti-PD-L1 immunotherapy. These findings represent a crucial step toward developing more effective therapies for iCCA and have significant implications for clinicians, scientists, and drug developers in the field.
Collapse
Affiliation(s)
- Tianming Cui
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Linmao Sun
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Xinyu Guo
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Cheng Cheng
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Ning Zhang
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Shuo Zhou
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Qi Chu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Changjian Xing
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Shuhang Liang
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China; Department of Gastrointestinal Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Yufeng Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Changyong Ji
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Xianying Li
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Shengwei Tao
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Xuetian Gu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Kun Ma
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Chenghui Wu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Junhui Chu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Yumin Fu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Sitao Han
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Yuchen Zhang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Jiareng Ye
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Yao Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China.
| | - Jiabei Wang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China.
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China; Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China; Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China.
| |
Collapse
|
12
|
Dioguardi Burgio M, Ronot M, Vilgrain V. ESR Essentials: assessing the radiological response of liver metastases to systemic therapy-practice recommendations by the European Society of Gastrointestinal and Abdominal Radiology. Eur Radiol 2025:10.1007/s00330-025-11540-1. [PMID: 40185923 DOI: 10.1007/s00330-025-11540-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Revised: 02/15/2025] [Accepted: 02/21/2025] [Indexed: 04/07/2025]
Abstract
The liver is a common site for metastatic spread, especially in advanced colorectal, breast, and pancreatic cancers. Imaging evaluation of liver metastases after systemic treatments like chemotherapy, targeted therapy, or immunotherapy is essential to distinguish treatment response from disease progression. The widely used response evaluation criteria in solid tumours (RECIST 1.1) focus on lesion size changes to evaluate treatment response. However, newer therapies, mainly targeted therapy and immunotherapy, often induce changes beyond size reduction, such as tumour necrosis, fibrosis, cystic transformation, calcifications, and modifications at the liver-tumour interface. These morphological and enhancement changes can be evaluated on CT and MRI and may better reflect the biological response in specific clinical settings. Overall, RECIST 1.1 criteria are recommended for assessing the radiological response of liver metastases after systemic treatment. The use of alternative radiological criteria validated on CT (such as Chun or Choi criteria) is recommended in specific clinical settings (e.g. metastatic colorectal cancer or metastatic gastrointestinal stromal tumours). Additionally, CT and MR modifications that reflect fibrosis, necrosis, calcifications, and haemorrhage can serve as ancillary indicators of tumoural response. These alternative criteria and radiological findings should be systematically assessed, particularly in liver metastases with minimal size changes, to better identify responders. KEY POINTS: RECIST 1.1 is the standard for evaluating tumour response in solid tumours and is recommended for the assessment of liver metastases after systemic therapy. CT attenuation, enhancement, and liver/tumour interface may correlate better with tumoural response compared to size reduction. CT and MR changes suggesting necrosis, fibrosis, calcifications, and haemorrhage can be used as additional indicators of tumoural response.
Collapse
Affiliation(s)
- Marco Dioguardi Burgio
- Université Paris Cité, Inserm, Centre de recherche sur l'inflammation, Paris, France.
- Department of Radiology, Hôpital Beaujon, AP-HP.Nord, Clichy, France.
| | - Maxime Ronot
- Université Paris Cité, Inserm, Centre de recherche sur l'inflammation, Paris, France
- Department of Radiology, Hôpital Beaujon, AP-HP.Nord, Clichy, France
| | - Valérie Vilgrain
- Université Paris Cité, Inserm, Centre de recherche sur l'inflammation, Paris, France
- Department of Radiology, Hôpital Beaujon, AP-HP.Nord, Clichy, France
| |
Collapse
|
13
|
Platt JR, Pennycook S, Muthoo CE, Westwood AC, Frood R, Beggs AD, Scarsbrook A, Seligmann JF, Tolan DJM. Colon cancer biology and treatment in the era of precision oncology: A primer for Radiologists. Eur J Radiol 2025; 185:112000. [PMID: 39978239 DOI: 10.1016/j.ejrad.2025.112000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/07/2025] [Accepted: 02/12/2025] [Indexed: 02/22/2025]
Abstract
In the era of precision oncology, systemic therapies for colon cancer are becoming increasingly biomarker-led, with implications for patients in the neoadjuvant, adjuvant and metastatic settings. As the landscape for colon cancer treatment evolves and becomes more complex, it is important that all members of the multidisciplinary team keep abreast of developments to ensure the most effective care is delivered to patients. As core members of the colorectal multidisciplinary team, Radiologists play a central role throughout the patient journey. This review serves as an educational summary of current and emerging treatment pathways in colon cancer, standards for biomarker testing, mechanisms of action for key drugs, important treatment-related complications, relevant tumour biology that underpins patterns of disease and treatment response, and the specific implications systemic therapies have for cancer imaging and Radiologists. We also highlight the increasing role for radiology in patient stratification and the importance of imaging biomarkers. It is crucial that Radiologists understand the current landscape of colon cancer treatment and emerging strategies on the horizon in clinical trials. Only through engagement across the wider multidisciplinary team will we deliver true personalised medicine for patients with colon cancer.
Collapse
Affiliation(s)
- James R Platt
- Division of Oncology, Leeds Institute of Medical Research at St James's, School of Medicine, University of Leeds, Leeds, UK.
| | - Stephanie Pennycook
- Department of Medical Oncology, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| | - Chand E Muthoo
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| | - Alice C Westwood
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St. James's, School of Medicine, University of Leeds, Leeds, UK.
| | - Russell Frood
- Leeds Institute of Clinical Trials Research, School of Medicine, University of Leeds, Leeds, UK.
| | - Andrew D Beggs
- Department of Cancer and Genomics, University of Birmingham, Birmingham, UK.
| | - Andrew Scarsbrook
- Leeds Institute of Medical Research at St James's, School of Medicine, University of Leeds, Leeds, UK.
| | - Jenny F Seligmann
- Division of Oncology, Leeds Institute of Medical Research at St James's, School of Medicine, University of Leeds, Leeds, UK.
| | - Damian J M Tolan
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| |
Collapse
|
14
|
Floudas CS, Goswami M, Donahue RN, Pastor DM, Redman JM, Brownell I, Turkbey EB, Cordes LM, Steinberg SM, Manu M, Francis DC, Lamping E, Marté JL, Kackley M, Krauss E, Manukyan M, Jochems C, Schlom J, Gulley JL, Strauss J. Novel Combination Immunotherapy and Clinical Activity in Patients With HPV-Associated Cancers: A Nonrandomized Clinical Trial. JAMA Oncol 2025; 11:394-399. [PMID: 39976981 PMCID: PMC11843463 DOI: 10.1001/jamaoncol.2024.6998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 12/20/2024] [Indexed: 02/23/2025]
Abstract
IMPORTANCE Patients who experience progression of advanced human papillomavirus (HPV)-associated cancers and who have previously received first-line systemic treatment have a poor prognosis and limited therapeutic options. OBJECTIVE To assess the clinical activity of the combination of the HPV type 16 therapeutic vaccine PDS0101, the tumor-targeting interleukin 12 antibody-drug conjugate PDS01ADC, and the bifunctional anti-programmed cell death ligand 1 (PD-L1)/transforming growth factor β (TGF-β) bintrafusp alfa in advanced HPV-associated cancers. DESIGN, Setting, and Participants This nonrandomized clinical trial was phase 1/2 and investigator initiated, and was conducted at a single US cancer research center between June 2020 and July 2022. Patients with advanced or metastatic HPV-associated cancers were eligible, including patients who were both immune checkpoint blockade (ICB) naive and ICB resistant. The cutoff date for data analysis was May 13, 2024. INTERVENTION Patients received 1 mL of PDS0101 subcutaneously every 4 weeks for 6 doses then every 12 weeks for 2 additional doses, PDS01ADC, 16.8 µg/kg, subcutaneously every 4 weeks or PDS01ADC, 8 µg/kg, subcutaneously every 2 weeks, and bintrafusp alfa, 1200 mg, intravenously every 2 weeks. MAIN OUTCOMES AND MEASURES Objective response rate (ORR) by Response Evaluation Criteria in Solid Tumors version 1.1 in ICB-naive patients. RESULTS Of the 50 eligible patients, 26 (52%) were men and the median age was 56 years (range, 28-80 years). The median (IQR) follow-up was 37.7 (30.6-42.0) months. Fourteen patients (28%) were ICB naive, with an ORR of 35.7% (95% CI, 12.8%-64.9%), and median overall survival (OS) 42.4 months (95% CI, 8.3 months-not estimable); in ICB-resistant patients, the ORR was 16.7% (6 of 36 patients; 95% CI, 6.4%-32.8%) and median OS was 15.8 months (95% CI, 9.0-21.3 months). Among patients with HPV-16-positive tumors (37 patients [74%]), in the ICB-naive group (8 patients [21.6%]) the ORR was 62.5% (95% CI, 24.5%-91.5%) and a median OS measure was not reached. Grade 3 and 4 treatment-related adverse events occurred in 26 of 50 patients (52%). There were no treatment-related deaths. CONCLUSIONS AND RELEVANCE In this trial, the combination of PDS0101, PDS01ADC, and bintrafusp alfa showed an acceptable safety profile and promising antitumor activity and improved OS in patients with HPV-16-positive cancers, in both ICB-naive and ICB-resistant patients, warranting further evaluation of the combination of PDS0101 and PDS01ADC with simultaneous PD-L1/TGF-β inhibition in these populations. Trial Registration ClinicalTrials.gov Identifier: NCT04287868.
Collapse
Affiliation(s)
- Charalampos S. Floudas
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Meghali Goswami
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Renee N. Donahue
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Danielle M. Pastor
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jason M. Redman
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Isaac Brownell
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland
| | - Evrim B. Turkbey
- Radiology and Imaging Sciences, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Lisa M. Cordes
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Michell Manu
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Deneise C. Francis
- Office of Research Nursing, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Elizabeth Lamping
- Office of Research Nursing, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jennifer L. Marté
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Mary Kackley
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Elizabeth Krauss
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Manuk Manukyan
- Clinical Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Caroline Jochems
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Jeffrey Schlom
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - James L. Gulley
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Julius Strauss
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
15
|
Xue C, Dong Z, Tan K, Zhang X, Yu Y, Wang S, Zheng J, Cui H. Pembrolizumab as an effective treatment for diffuse malignant peritoneal mesothelioma with long‑term survival: A case report and literature review. Oncol Lett 2025; 29:187. [PMID: 40070788 PMCID: PMC11894505 DOI: 10.3892/ol.2025.14933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 01/14/2025] [Indexed: 03/14/2025] Open
Abstract
Primary diffuse malignant peritoneal mesothelioma (MPEM) is a malignant disease without standard treatments recommended. Recently, immunotherapy has revolutionized the field of tumor therapy. According to current clinical evidence, advanced MPEM may gain potential clinical benefits from immune checkpoint inhibitors. The present study reported a 61-year-old female patient with persistent low fever as the initial symptom, who was eventually diagnosed with MPEM. This patient obtained significant clinical benefits from pembrolizumab, with disappearance of symptoms, a lasting stable disease response with a progression-free survival of 10.0 months and a long overall survival of 26.2 months. The application of pembrolizumab was explored as an emerging effective treatment for patients with MPEM. In addition, the clinical characteristics, diagnosis, treatment, pathogenesis and target regulation in MPEM were discussed and previous studies were reviewed. Further evidence is needed from future extensive clinical trials.
Collapse
Affiliation(s)
- Chongxiang Xue
- School of Clinical Medicine, Beijing University of Chinese Medicine, Beijing 100029, P.R. China
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Zhe Dong
- Center for Coronary Artery Disease, Division of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, P.R. China
| | - Kexin Tan
- Department of Traditional Chinese Medicine/Integrative Medicine, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Changsha, Hunan 410006, P.R. China
| | - Xu Zhang
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, P.R. China
| | - Yixuan Yu
- Intensive Care Unit, Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, Zhejiang 322000, P.R. China
| | - Shuo Wang
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Jiabin Zheng
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| | - Huijuan Cui
- Department of Integrative Oncology, China-Japan Friendship Hospital, Beijing 100029, P.R. China
| |
Collapse
|
16
|
Nelles C, Gräf M, Bernard P, Persigehl T, Große Hokamp N, Zopfs D, Maintz D, Kreuzberg N, Wolf J, Bröckelmann PJ, Lennartz S. Real-world response assessment of immune checkpoint inhibition: comparing iRECIST and RECIST 1.1 in melanoma and non-small cell lung cancer patients. Eur Radiol 2025; 35:2084-2093. [PMID: 39294304 PMCID: PMC11914328 DOI: 10.1007/s00330-024-11060-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/12/2024] [Accepted: 08/18/2024] [Indexed: 09/20/2024]
Abstract
OBJECTIVES To compare immune response evaluation criteria in solid tumors (iRECIST) and response evaluation criteria in solid tumors (RECIST) 1.1 for response assessment of immune checkpoint inhibitor (ICI) therapy in a real-world setting in patients with melanoma and non-small cell lung cancer (NSCLC). METHODS Two-hundred fifty-two patients with melanoma and NSCLC who received CTLA-4 inhibitor ipilimumab or PD-1 inhibitors nivolumab or pembrolizumab and who underwent staging CT of the chest and abdomen were retrospectively included. Treatment response evaluation according to the RECIST 1.1 and iRECIST guidelines was performed for all patients. Response patterns, as well as overall response rate (ORR), disease control rate (DCR), and time to progression (TTP), were compared between RECIST 1.1 and iRECIST. RESULTS Out of 143 patients with progressive disease (PD) according to RECIST 1.1, 48 (33.6%) did not attain confirmation of progression (iCPD) as per iRECIST and six patients who were treated beyond RECIST 1.1 progression reached PD at a later point in time in iRECIST, resulting in a significant difference in TTP between iRECIST and RECIST 1.1 (618.3 ± 626.9 days vs. 538.1 ± 617.9 days, respectively (p < 0.05)). The number of non-responders as per RECIST 1.1 was 79, whereas it was 60 when using iRECIST. ORR was 28.5% for RECIST 1.1 and 34.1% for iRECIST, and corresponding DCR of 67.4% for RECIST 1.1 and 74.6% for iRECIST. CONCLUSION iRECIST was more suitable than RECIST 1.1 for capturing atypical response patterns to ICI therapy in patients with melanoma and NSCLC, resulting in differences in the assessment of treatment response. CLINICAL RELEVANCE STATEMENT Compared to RECIST 1.1, iRECIST may improve patient care and treatment decisions for patients with NSCLC or melanoma who are treated with immune checkpoint inhibitors in clinical routine. KEY POINTS RECIST 1.1 may incorrectly assess atypical treatment patterns to immune checkpoint inhibitors. iRECIST better captured atypical response patterns compared to RECIST 1.1. iRECIST was more suitable for assessing response to immune checkpoint inhibitors in non-small cell lung carcinoma and melanoma.
Collapse
Affiliation(s)
- Christian Nelles
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Moritz Gräf
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Pascale Bernard
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Thorsten Persigehl
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nils Große Hokamp
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - David Zopfs
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - David Maintz
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Nicole Kreuzberg
- Department of Dermatology and Venereology, Skin Cancer Center, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jürgen Wolf
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Paul J Bröckelmann
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Simon Lennartz
- Institute for Diagnostic and Interventional Radiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
| |
Collapse
|
17
|
Hopkinson G, Taylor J, Wadsley J, Darekar A, Messiou C, Koh DM. Tumour measurements on imaging for clinical trial: A national picture of service provision. BJC REPORTS 2025; 3:19. [PMID: 40148514 PMCID: PMC11950641 DOI: 10.1038/s44276-025-00131-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 02/20/2025] [Accepted: 03/02/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Radiological response evaluation metrics such as RECIST 1.1 inform critical endpoints in oncology trials. The UK was the 6th highest recruiter into oncology trials worldwide between 1999 and 2022, with almost 9000 oncology trials registered during the same period. However, the provision of tumour measurements for oncology trials is often ad hoc and patchy across the NHS. The aim of this work was to understand the barriers to providing an effective imaging tumour measurement service, gain insight into service delivery models and consider the successes and challenges from the perspective of both service providers and end users. METHODS An electronic survey was distributed to those who provide tumour measurement response review for clinical trials (service providers) and those that request and use such measurements in trial activities (service users). RESULTS Responses from 35 sites demonstrated substantial variation in service provision across the UK. Despite workforce pressures, service is largely delivered through radiologists with a minority utilising radiographer role extension. Only 20% of the service providers had dedicated training and 29% received robust financial reimbursement. DISCUSSION Service variation is likely a consequence of limited training, education and infrastructure to support robust service, compounded by increasing radiology workload and workforce pressures.
Collapse
Affiliation(s)
- Georgina Hopkinson
- Department of Radiology, The Royal Marsden NHS Foundation Trust, London, UK.
| | - Jonathan Taylor
- Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | | | - Angela Darekar
- University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Christina Messiou
- Department of Radiology, The Royal Marsden NHS Foundation Trust, London, UK
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Dow-Mu Koh
- Department of Radiology, The Royal Marsden NHS Foundation Trust, London, UK
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| |
Collapse
|
18
|
Alonso-Gordoa T, Anguera G, Domínguez-Esteban M, Reig Ò, Martínez-Barros H, Molina-Cerrillo J, Cruz P, Maroto P. Expert consensus on patterns of progression in kidney cancer after adjuvant immunotherapy and subsequent treatment strategies. Cancer Treat Rev 2025; 136:102925. [PMID: 40186886 DOI: 10.1016/j.ctrv.2025.102925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/21/2025] [Accepted: 03/21/2025] [Indexed: 04/07/2025]
Abstract
Immunotherapy has changed the management of localized clear cell renal cell carcinoma (ccRCC) since the approval of adjuvant pembrolizumab, which demonstrated significant improvements in disease-free survival (DFS) and overall survival (OS) in patients at intermediate and high risk of recurrence. This new approach impacts rescue strategies in patients who relapse after local treatment and during or after adjuvant pembrolizumab. Nevertheless, there is currently no robust scientific evidence on therapeutic decision-making in this clinical situation, representing an area for further debate and research. In this article, a group of experts from the Genitourinary Alliance for Research and Development (GUARD) have reviewed the available scientific evidence to establish the basis for therapeutic decision-making in patients with ccRCC who progress after adjuvant treatment with immunotherapy. Despite the lack of randomized clinical trials in this setting, this group of experts recommends classifying patients according to relapse volume (oligometastatic vs. polymetastatic), time to relapse and certain molecular characteristics. Rescue treatments beyond relapse should be individualized and might include locoregional treatments such as surgery or radiotherapy as well as antiangiogenic therapies in patients defined as resistant to immunotherapy.
Collapse
Affiliation(s)
| | - Georgia Anguera
- Medical- Oncology Department, Santa Creu i Sant Pau Hospital, Barcelona, Spain.
| | | | - Òscar Reig
- Laboratory of Translational Genomics and Targeted Therapies in Solid Tumours, Fundació de Recerca Clínic Barcelona - Institut d'Investigacions Biomèdiques August Pi i Sunyer (FRCB-IDIBAPS), Medicine Department, Barcelona University, Medical Oncology Department, Clínic Hospital, Barcelona, Spain.
| | | | | | - Patricia Cruz
- Medical Oncology Department, Ciudad Real General University Hospital, Ciudad Real, Spain.
| | - Pablo Maroto
- Medical Oncology Department, Santa Creu i Sant Pau Hospital, Barcelona, Spain.
| |
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] [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
|
Bartolomucci A, Nobrega M, Ferrier T, Dickinson K, Kaorey N, Nadeau A, Castillo A, Burnier JV. Circulating tumor DNA to monitor treatment response in solid tumors and advance precision oncology. NPJ Precis Oncol 2025; 9:84. [PMID: 40122951 PMCID: PMC11930993 DOI: 10.1038/s41698-025-00876-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
Circulating tumor DNA (ctDNA) has emerged as a dynamic biomarker in cancer, as evidenced by its increasing integration into clinical practice. Carrying tumor specific characteristics, ctDNA can be used to inform treatment selection, monitor response, and identify drug resistance. In this review, we provide a comprehensive, up-to-date summary of ctDNA in monitoring treatment response with a focus on lung, colorectal, and breast cancers, and discuss current challenges and future directions.
Collapse
Affiliation(s)
- Alexandra Bartolomucci
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Monyse Nobrega
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Tadhg Ferrier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Kyle Dickinson
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Nivedita Kaorey
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Amélie Nadeau
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Alberto Castillo
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Department of Pathology, McGill University, Montreal, QC, Canada.
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
21
|
Drobniak A, Puskulluoglu M, Stokłosa Ł, Pacholczak-Madej R. Exploring the efficacy of nivolumab and ipilimumab in renal cell carcinoma: insights from a district hospital cohort study. Rep Pract Oncol Radiother 2025; 30:34-43. [PMID: 40242417 PMCID: PMC11999007 DOI: 10.5603/rpor.104389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 12/10/2024] [Indexed: 04/18/2025] Open
Abstract
Background Nivolumab and ipilimumab combination is recommended as a first-line treatment for metastatic renal cell carcinoma (mRCC) in patients without life-threatening symptoms. This study aims to assess the efficacy and safety of this treatment regimen administered in the one-day chemotherapy unit of a district hospital. Materials and methods We conducted a retrospective study involving 36 patients diagnosed with mRCC who had received combined immunotherapy at the Department of Chemotherapy, District Hospital in Sucha Beskidzka, Poland. We evaluated treatment response and adverse events (AEs). Laboratory parameters were recorded, and we calculated neutrophil-lymphocyte ratios (NLR), platelet-lymphocyte ratios (PLR), and lymphocyte-monocyte ratios (LMR) at baseline, after 3 months of treatment, and prior to disease progression. Results After a median follow-up of 11 months (7.5-17.5 months), the median overall survival was not reached (NR, 6.7-NR), while the median progression-free survival was 11.5 months (6.7-NR). The objective response rate was 30.6% (n = 11), and the disease-control rate was 66.7% (n = 24). Hemoglobin and eosinophil levels varied at three checkpoints, without differences in NLR, PLR, and LMR. AEs of any grade were observed in 23 patients (63.9%) with a median onset time of 3 months (2-4 months), and serious AEs in 13.8% of patients (n = 5). Conclusions Our analysis suggests that the combination of nivolumab and ipilimumab for mRCC has an acceptable toxicity profile and can be effectively managed in a district hospital's outpatient clinic. This approach requires close patient monitoring and collaboration with other hospital departments to ensure patient safety and treatment efficacy.
Collapse
Affiliation(s)
- Artur Drobniak
- Department of Chemotherapy, The District Hospital, Sucha Beskidzka, Poland
| | - Mirosława Puskulluoglu
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow Branch, Krakow, Poland
| | - Łukasz Stokłosa
- Department of Chemotherapy, The District Hospital, Sucha Beskidzka, Poland
- Department of Chemotherapy, The Specialistic Hospital, Nowy Targ, Poland
- Department of Thoracic Surgery, Pulmonary Hospital, Zakopane, Poland
| | - Renata Pacholczak-Madej
- Department of Chemotherapy, The District Hospital, Sucha Beskidzka, Poland
- Department of Gynecological Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Krakow Branch, Krakow, Poland
- Department of Anatomy, Jagiellonian University, Medical College, Krakow, Poland
| |
Collapse
|
22
|
Cheng Y, Jiang ZP, Chen XB, Lu KY, Liu ZY, Shao D. Prognostic Value of 18F-FDG PET/CT Metabolic Parameters in Resectable Non-small Cell Lung Cancer Treated With Neoadjuvant Immunotherapy Plus Chemotherapy. Clin Nucl Med 2025:00003072-990000000-01624. [PMID: 40108723 DOI: 10.1097/rlu.0000000000005863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Accepted: 02/18/2025] [Indexed: 03/22/2025]
Abstract
OBJECTIVE This study investigates the predictive value of 18F-FDG PET/CT metabolic parameters in patients with non-small cell lung cancer (NSCLC) undergoing neoadjuvant immunotherapy plus chemotherapy. METHODS We conducted a retrospective analysis of clinical data from 131 patients with pathologically confirmed NSCLC who were deemed resectable after 3 cycles of neoadjuvant immunotherapy plus chemotherapy. Pretreatment and post-treatment PET metabolic parameters were evaluated. CT assessments based on immune response evaluation criteria in solid tumors (iRECIST) were compared with PET/CT assessments using the response criteria in solid tumors (PERCIST). ROC curve analysis and Kaplan-Meier survival analysis, including univariate and Cox multivariate analyses, were employed to assess the prognostic value of PET metabolic parameters after treatment. RESULTS The PET/CT assessment based on PERCIST showed high consistency with prognosis, while the CT assessment based on iRECIST demonstrated low consistency. Statistically significant differences were observed between the iRECIST and PERCIST criteria (P<0.001). ROC curve analysis revealed significant differences in post-treatment PET metabolic parameters (postSUVmax, postSUVmean, postSUVpeak, postMTV, and postTLG) as well as the percentage changes in metabolic parameters before and after treatment(Δ) (ΔSUVmax, ΔSUVmean, ΔSUVpeak, ΔMTV, and ΔTLG) (P<0.05). Optimal cutoff values enabled stratification into high-risk and low-risk groups. Univariate analysis showed significantly higher survival in the low-risk group for all parameters except ΔMTV (P=0.311), while Cox multivariate analysis identified ΔSUVmax as the most predictive. CONCLUSIONS The PERCIST is more accurate than iRECIST in evaluating prognosis for NSCLC neoadjuvant immunotherapy plus chemotherapy. PET metabolic parameters, particularly ΔSUVmax, effectively predict prognosis and support clinical decision-making.
Collapse
Affiliation(s)
- You Cheng
- Department of PET Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Zhen-Peng Jiang
- Department of Nuclear Medicine, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Xiao-Bo Chen
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Kai-Yu Lu
- Department of PET Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Zai-Yi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Dan Shao
- Department of PET Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
23
|
Wu G, Chen X, Luo R, Koh YX, Lim TKH, Chew V, Zhou J, Fan J, Gao Q, Zhu K, Shi R. Histopathologic Grading of Residual Tumor Predicts Survival of Intrahepatic Cholangiocarcinoma Patients Treated With Neoadjuvant Therapy: Major Pathologic Response and Its Clinical Significance. Am J Surg Pathol 2025:00000478-990000000-00491. [PMID: 40103370 DOI: 10.1097/pas.0000000000002359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Neoadjuvant therapy (NAT) is increasingly used to treat patients with initially unresectable intrahepatic cholangiocarcinoma (iCCA). A histopathologic grading system for residual tumors that can predict patient survival is lacking in the literature. This retrospective study enrolled 151 iCCA patients who received NAT. The percentage of residual viable tumor (%RVT) extent was calculated by RVT surface area/total tumor bed area ×100 and scored in 5% increments. Kaplan-Meier and Cox regression analyses were used to investigate its correlations with recurrence-free survival (RFS) and overall survival (OS). Tumor regression grading by the College of American Pathologists (CAP) and MD Anderson (MDA) methodologies were also validated. A 10% RVT-based tumor regression score (TRS) showed a significant correlation with both OS and RFS. TRS and major pathologic response (mPR) were therefore defined as follows: TRS 1/mPR, tumor with 0 to 10% RVT; TRS 2, more than 10% RVT. Patients graded as TRS 1/mPR had superior OS (P=0.006) and RFS (P<0.001) compared with those with TRS 2 in univariate analysis. In a multivariate analysis including ypTNM stages, lymphovascular invasion, and perineural invasion, TRS 1/mPR was also found to be an independent prognostic factor for both OS (hazard ratio [HR]: 0.226; 95% CI: 0.053-0.966, P=0.045) and RFS (HR: 0.474; 95% CI: 0.231-0.974, P=0.042). As for the CAP and MDA grading methodologies, they were found to correlate with RFS (CAP: P=0.002; MDA: P=0.001), but not with OS (CAP: P=0.181; MDA: P=0.09). Our study revealed that a TRS of ≤10% RVT significantly correlates with longer OS and RFS and can be suggested as an mPR in iCCA. This indicator is easily applicable, prognostically relevant, and could be further validated in future prospective clinical trials.
Collapse
Affiliation(s)
- Gaohua Wu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute
| | - Xiufen Chen
- Department of Anatomical Pathology, Singapore General Hospital
| | - Rongkui Luo
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ye Xin Koh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Centre
| | | | - Valerie Chew
- Translational Immunology Institute (TII), SingHealth-DukeNUS Academic Medical Centre
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute
| | - Kai Zhu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute
| | - Ruoyu Shi
- Department of Pathology and Laboratory Medicine, Kandang Kerbau Women's and Children's Hospital, Singapore
| |
Collapse
|
24
|
Mou YH, Zhang J, Shen H, Yu J, Han L, Li H, Li QF. Multiple pseudoprogressions during ongoing immunotherapy-based treatment of advanced gastric neuroendocrine carcinoma: A case report and review of literature. World J Gastrointest Oncol 2025; 17:102804. [PMID: 40092963 PMCID: PMC11866236 DOI: 10.4251/wjgo.v17.i3.102804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/24/2024] [Accepted: 01/15/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Gastric neuroendocrine carcinomas (NECs) exhibit aggressive features, such as rapid growth, higher rate of metastasis, and a generally unfavorable prognosis compared to gastric adenocarcinoma. As a result, therapeutic options for NECs remain limited, contributing to the poor prognosis of patients. Immunotherapy has emerged as a promising treatment strategy and demonstrated the potential to partially improve the survival and prognosis of patients with NECs. Nevertheless, the unique clinical response termed pseudoprogression (PsP) has garnered considerable attention in the context of immunotherapy. CASE SUMMARY Presented here is a case of NEC recurrence five and a half months after radical gastric surgery. The 45-year-old male patient underwent combination treatment involving a PD-1 blocker and tyrosine kinase inhibitors and encountered two instances of PsP during treatment. The patient ultimately achieved a durable treatment response without altering his treatment regimens, resulting in a substantial therapeutic benefit. CONCLUSION This case report aimed to provide the authors' experience with the diagnosis of PsP and treatment strategies for PsP in ongoing immunotherapy.
Collapse
Affiliation(s)
- Yan-Hua Mou
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, Hubei Province, China
- Institute of Oncology, Hubei University of Arts and Science, Xiangyang 441021, Hubei Province, China
| | - Juan Zhang
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, Hubei Province, China
- Institute of Oncology, Hubei University of Arts and Science, Xiangyang 441021, Hubei Province, China
| | - Hui Shen
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, Hubei Province, China
- Institute of Oncology, Hubei University of Arts and Science, Xiangyang 441021, Hubei Province, China
| | - Jing Yu
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, Hubei Province, China
- Institute of Oncology, Hubei University of Arts and Science, Xiangyang 441021, Hubei Province, China
| | - Lan Han
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, Hubei Province, China
- Institute of Oncology, Hubei University of Arts and Science, Xiangyang 441021, Hubei Province, China
| | - Hui Li
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, Hubei Province, China
- Institute of Oncology, Hubei University of Arts and Science, Xiangyang 441021, Hubei Province, China
| | - Qing-Feng Li
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang 441021, Hubei Province, China
- Institute of Oncology, Hubei University of Arts and Science, Xiangyang 441021, Hubei Province, China
| |
Collapse
|
25
|
Ahmed J, Knisely A, Torrado C, Stephen B, Yang Y, Song J, Alshawa A, Zarifa A, Jhingran A, Koay EJ, Morris VK, Javle M, Wolff RA, Meric-Bernstam F, Pant S, Rodon J, Naing A. A phase I/II trial of avelumab combinations with ivuxolimab, utomilumab, and radiation therapy in patients with advanced gastrointestinal malignancies. Oncologist 2025; 30:oyaf032. [PMID: 40139261 PMCID: PMC11942784 DOI: 10.1093/oncolo/oyaf032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/30/2024] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Checkpoint agonists utomilumab (4-1BB agonist) and ivuxolimab (OX40 agonist) enhance Teffector cell function. Preclinical studies suggest that combining these drugs with avelumab (anti-PD-L1 antibody) can potentially synergize this effect. In addition, tissue abscopal effects of radiation therapy may improve antigen presentation, complementing PD-L1 blockade. We conducted a single institution, open-label, multi-arm, non-randomized, phase 1/2 clinical trial of avelumab in combination with ivuxolimab, with or without utomilumab, and radiation therapy in patients with advanced solid tumors. Herein, we present a subgroup analysis in patients with gastrointestinal (GI) tumors (pancreatic, colon, gastric, and hepatocellular). METHODS The primary objectives of this study were to assess safety, tolerability, and dose-limiting toxicities. The secondary objectives were to evaluate efficacy including response rate, progression free survival (PFS), as determined by immune-related Response Criteria in Solid Tumors (irRECIST) and overall survival (OS). RESULTS Thirty-one patients with pancreatic (n = 21), colorectal (n = 8), hepatocellular (n = 1), and gastric (n = 1) cancers were included in this study. The most common treatment-related adverse events (TRAEs) were chills (13%), diarrhea (10%), colitis (10%), fatigue (6%), and fever (6%). There were 3 instances of grade 3 diarrhea and colitis (10%) without any other grade ≥ 3 TRAEs Among the 24 patients evaluable for response, 9 (37.5%) had immune-related stable disease (irSD) and 14 (58.3%) had immune-related progressive disease (irPD). One patient had clinical progression without radiological confirmation. The median PFS was 2 months. Median OS was 5.6 months. CONCLUSION Combining avelumab with co-stimulatory checkpoint agonists produces modest activity without added safety concerns in patients with advanced GI malignancies (ClinicalTrials.gov Identifier: NCT03217747).
Collapse
Affiliation(s)
- Jibran Ahmed
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Anne Knisely
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77230, United States
| | - Carlos Torrado
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Bettzy Stephen
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Yali Yang
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Juhee Song
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Anas Alshawa
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Abdulrazzak Zarifa
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Anuja Jhingran
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Eugene J Koay
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Van Karlyle Morris
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Milind Javle
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Robert A Wolff
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Shubham Pant
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Jordi Rodon
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| |
Collapse
|
26
|
Nair U, Rakestraw E, Beasley GM, O’Connor MH. Opportunities for Discovery Using Neoadjuvant Immune Checkpoint Blockade in Melanoma. Int J Mol Sci 2025; 26:2427. [PMID: 40141071 PMCID: PMC11942238 DOI: 10.3390/ijms26062427] [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: 02/04/2025] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Treatment of resectable advanced-stage melanoma with neoadjuvant immunotherapy is rapidly becoming the new standard of care due to significant improvements in event-free survival (EFS) compared to surgery first followed by immunotherapy. The level of responsiveness seen in patients receiving immune checkpoint inhibitors (ICIs) must be mechanistically understood not only for the standardization of treatment but also to advance the novel concept of personalized cancer immunotherapy. This review aims to elucidate markers of the tumor microenvironment (TME) and blood that can predict treatment outcome. Interestingly, the canonical proteins involved in the molecular interactions that immunotherapies aim to disrupt have not been consistent indicators of treatment response, which amplifies the necessity for further research on the predictive model. Other major discussions surrounding neoadjuvant therapy involve the higher-level investigation of ICI efficacy due to the ability to examine a post-treatment tumor molecularly and pathologically, which this review will also cover. As neoadjuvant ICI becomes the standard of care in advanced melanoma treatment, further research aiming to identify more predictive biomarkers of treatment response to advance medical decision-making and patient care should continue to be sought after.
Collapse
Affiliation(s)
| | | | - Georgia M. Beasley
- Department of Surgery, Duke University, Durham, NC 27710, USA; (U.N.); (E.R.); (M.H.O.)
| | | |
Collapse
|
27
|
Akula V, Chen L, Acikgoz Y, Klein K, Yavuz BG, Cevik L, Demir T, Manne A, Sahin I, Kaseb A, Hasanov E. Neoadjuvant immune checkpoint inhibitors for hepatocellular carcinoma. NPJ Precis Oncol 2025; 9:60. [PMID: 40050446 PMCID: PMC11885445 DOI: 10.1038/s41698-025-00846-4] [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: 09/23/2024] [Accepted: 02/24/2025] [Indexed: 03/09/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of liver cancer. HCC treatment is challenging; surgical resection is the primary curative treatment for early-stage disease, but recurrence rates are high. Immune checkpoint inhibitors (ICIs) are a promising neoadjuvant treatment that can reduce recurrence rates and mortality after surgery and achieve complete/partial responses. Clinical trials provide strong evidence for the efficacy and safety of ICI monotherapy for neoadjuvant HCC treatment.
Collapse
Affiliation(s)
- Vinita Akula
- Department of Internal Medicine, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Lily Chen
- Department of Internal Medicine, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Yusuf Acikgoz
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Katherine Klein
- Department of Internal Medicine, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | - Betul Gok Yavuz
- Department of Medicine, University of Missouri, Columbia, MO, USA
| | - Lokman Cevik
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Tarik Demir
- Division of Hematology and Oncology Developmental Therapeutics Institute, Northwestern University, Chicago, IL, USA
| | - Ashish Manne
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Ilyas Sahin
- Division of Hematology & Oncology, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Ahmed Kaseb
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elshad Hasanov
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA.
| |
Collapse
|
28
|
Lee SM, Hamid O, Jotte R, Zakharia Y, Medina T, Gillespie-Twardy A, Mehmi I, Chandra S, Watson G, Ward P, Chaney M, Lu H, Berndt J, O’Connor BP, Rathi K, Shaikh E, Cowey CL. Phase II Open-Label Trial of Brentuximab Vedotin with Pembrolizumab in PD-1-Pretreated Metastatic Non-Small Cell Lung Cancer and Metastatic Cutaneous Melanoma. Clin Cancer Res 2025; 31:848-859. [PMID: 39786430 PMCID: PMC11873802 DOI: 10.1158/1078-0432.ccr-24-1478] [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: 05/15/2024] [Revised: 08/09/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025]
Abstract
PURPOSE Brentuximab vedotin (BV) is hypothesized to selectively deplete T regulatory cells that express CD30 and resensitize tumors to anti-PD-1 therapy. This study evaluated responses to BV + pembrolizumab after PD-1 therapy and explored corresponding biomarkers. PATIENTS AND METHODS A total of 55 patients with metastatic non-small cell lung cancer and 58 patients with metastatic cutaneous melanoma received ≥1 dose of BV + pembrolizumab. Patients had received a median of 2.0 prior lines of systemic therapies (range, 1-7). The primary endpoint was confirmed objective response rate (ORR). Exploratory endpoints included overall survival (OS) and biomarker analysis in blood and tumor. RESULTS For the secondary refractory metastatic non-small cell lung cancer cohort (RECIST v1.1), the ORR was 14%, median progression-free survival (PFS) was 5.85 months, and median OS was 14.4 months. For the secondary refractory metastatic cutaneous melanoma cohort (immune RECIST), the ORR was 24%, median PFS was 4.44 months, and median OS was 21.9 months. Overall, the median duration of OS follow-up was 17.2 months (95% confidence interval, 14.62-22.87). No new safety signals were identified. No treatment-related grade 5 toxicity was seen. Longitudinal immune phenotyping in peripheral blood demonstrated a transient decrease in T regulatory cells. Paired tumor biopsies from baseline and cycle 3 day 1 showed a trend of increased CD8 T-cell infiltration, especially in responding patients. CONCLUSIONS BV + pembrolizumab in solid tumor malignancies resulted in clinically meaningful, durable responses with encouraging OS and PFS rates supportive of the immunomodulatory activity of this combination. Stronger antitumor activity was observed in secondary refractory cohorts. The safety profile of this combination was consistent with the individual drug risk profiles.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Female
- Male
- Middle Aged
- Melanoma/drug therapy
- Melanoma/pathology
- Melanoma/mortality
- Aged
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/mortality
- Adult
- Skin Neoplasms/drug therapy
- Skin Neoplasms/pathology
- Skin Neoplasms/mortality
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Lung Neoplasms/secondary
- Lung Neoplasms/mortality
- Aged, 80 and over
- Brentuximab Vedotin/therapeutic use
- Brentuximab Vedotin/administration & dosage
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Melanoma, Cutaneous Malignant
Collapse
Affiliation(s)
| | - Omid Hamid
- The Angeles Clinic and Research Institute, A Cedars-Sinai Affiliate, Los Angeles, California
| | - Robert Jotte
- Rocky Mountain Cancer Centers, Lone Tree, Colorado
- US Oncology Network, The Woodlands, Texas
| | | | | | | | - Inderjit Mehmi
- The Angeles Clinic and Research Institute, A Cedars-Sinai Affiliate, Los Angeles, California
| | | | - Graham Watson
- US Oncology Network, The Woodlands, Texas
- Virginia Oncology Associates, Norfolk, Virginia
| | - Patrick Ward
- US Oncology Network, The Woodlands, Texas
- Oncology Hematology Care, Cincinnati, Ohio
| | | | | | | | | | | | | | - Charles Lance Cowey
- US Oncology Network, The Woodlands, Texas
- Texas Oncology – Baylor University, Dallas, Texas
| |
Collapse
|
29
|
Lin S, Ma Z, Yao Y, Huang H, Chen W, Tang D, Gao W. Automatic machine learning accurately predicts the efficacy of immunotherapy for patients with inoperable advanced non-small cell lung cancer using a computed tomography-based radiomics model. Diagn Interv Radiol 2025; 31:130-140. [PMID: 39817633 PMCID: PMC11880869 DOI: 10.4274/dir.2024.242972] [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] [Accepted: 11/18/2024] [Indexed: 01/18/2025]
Abstract
PURPOSE Patients with advanced non-small cell lung cancer (NSCLC) have varying responses to immunotherapy, but there are no reliable, accepted biomarkers to accurately predict its therapeutic efficacy. The present study aimed to construct individualized models through automatic machine learning (autoML) to predict the efficacy of immunotherapy in patients with inoperable advanced NSCLC. METHODS A total of 63 eligible participants were included and randomized into training and validation groups. Radiomics features were extracted from the volumes of interest of the tumor circled in the preprocessed computed tomography (CT) images. Golden feature, clinical, radiomics, and fusion models were generated using a combination of various algorithms through autoML. The models were evaluated using a multi-class receiver operating characteristic curve. RESULTS In total, 1,219 radiomics features were extracted from regions of interest. The ensemble algorithm demonstrated superior performance in model construction. In the training cohort, the fusion model exhibited the highest accuracy at 0.84, with an area under the curve (AUC) of 0.89-0.98. In the validation cohort, the radiomics model had the highest accuracy at 0.89, with an AUC of 0.98-1.00; its prediction performance in the partial response subgroup outperformed that in both the clinical and radiomics models. Patients with low rad scores achieved improved progression-free survival (PFS); (median PFS 16.2 vs. 13.4, P = 0.009). CONCLUSION autoML accurately and robustly predicted the short-term outcomes of patients with inoperable NSCLC treated with immune checkpoint inhibitor immunotherapy by constructing CT-based radiomics models, confirming it as a powerful tool to assist in the individualized management of patients with advanced NSCLC. CLINICAL SIGNIFICANCE This article highlights that autoML promotes the accuracy and efficiency of feature selection and model construction. The radiomics model generated by autoML predicted the efficacy of immunotherapy in patients with advanced NSCLC effectively. This may provide a rapid and non-invasive method for making personalized clinical decisions.
Collapse
Affiliation(s)
- Siyun Lin
- Huadong Hospital, Fudan University, Department of Thoracic Surgery, Shanghai, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
| | - Zhuangxuan Ma
- Huadong Hospital, Fudan University, Department of Radiology, Shanghai, China
| | - Yuanshan Yao
- Shanghai Chest Hospital, Shanghai JiaoTong University School of Medicine, Department of Thoracic Surgery, Shanghai, China
| | - Hou Huang
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Shanghai, China
| | - Wufei Chen
- Huadong Hospital, Fudan University, Department of Radiology, Shanghai, China
| | - Dongfang Tang
- Huadong Hospital, Fudan University, Department of Thoracic Surgery, Shanghai, China
| | - Wen Gao
- Huadong Hospital, Fudan University, Department of Thoracic Surgery, Shanghai, China
| |
Collapse
|
30
|
Fottner C, Apostolidis L, Krug S, Rinke A, Grün B, Michl P, Gress TM, Wagner DC, Roth W, Mettler E, Topsch J, Ruckes C, Galle PR, Weber MM. Activity and Safety of Avelumab in High-Grade Neuroendocrine Tumors and Poorly Differentiated Neuroendocrine Carcinomas Progressive after Chemotherapy (AveNEC Trial). Clin Cancer Res 2025; 31:860-867. [PMID: 39786465 DOI: 10.1158/1078-0432.ccr-24-2461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/08/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
PURPOSE Neuroendocrine neoplasms grade 3 (NEN G3) are rare tumors with poor prognosis and no established second-line therapy. The role of immune checkpoint blockade in these aggressive tumors remains unclear. PATIENTS AND METHODS The phase II AveNEC study evaluated the effect of avelumab (AVE, 10 mg/kg i.v. every 2 weeks) in 60 patients with well-differentiated high-grade neuroendocrine tumors (N = 22) or poorly differentiated neuroendocrine carcinomas (N = 38) progressing after ≥1 prior chemotherapy (excluding Merkel cell carcinoma and small cell lung cancer). RESULTS The best overall response according to immune-related Response Evaluation Criteria in Solid Tumors (iRECIST) was partial response (PR) in three (5%) and stable disease (SD) in nine (15%) patients, with a disease control rate at 16 weeks of 15% (3 PRs; 6 SDs) and a median duration of response of 4.3 months. Six (10%) patients achieved SD or PR for >6 months and two for >1 year. Response rates were similar regardless of differentiation, Ki-67 expression, or primary localization. The median progression-free survival was 1.9 months, and the overall survival was 6.6 months. After a median follow-up of 3.6 years, only four (7%) patients were still alive; 1- and 2-year survival rates were 33% and 17%, respectively. Responders had a significantly longer overall survival of 30.2 months compared with 4.8 months in nonresponders. AVE was well tolerated, with few treatment-related grade 3/4 adverse events, and the quality of life remained stable during treatment. CONCLUSIONS In patients with progressive high-grade NEN G3, AVE was well tolerated and provided disease control with significant clinical benefit in 15% of heavily pretreated patients.
Collapse
Affiliation(s)
- Christian Fottner
- Unit of Endocrinology, I Medical Department, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Leonidas Apostolidis
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Sebastian Krug
- UKH-Universitätsklinikum Halle (Saale), Halle (Saale), Germany
| | - Anja Rinke
- Department of Gastroenterology, UKGM-Uniklinikum Giessen und Marburg-Standort Marburg, Marburg, Germany
| | - Barbara Grün
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) Heidelberg, University Hospital Heidelberg, Heidelberg, Germany
| | - Patrick Michl
- Department of Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas M Gress
- Department of Gastroenterology, UKGM-Uniklinikum Giessen und Marburg-Standort Marburg, Marburg, Germany
| | - Daniel-Christoph Wagner
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Wilfried Roth
- Institute of Pathology, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Esther Mettler
- Unit of Endocrinology, I Medical Department, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Jana Topsch
- Interdisciplinary Centre for Clinical Trials, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Christian Ruckes
- Interdisciplinary Centre for Clinical Trials, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Peter R Galle
- Department of Internal Medicine I, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Matthias M Weber
- Unit of Endocrinology, I Medical Department, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
31
|
Badarna M, Keidar Z, Arnon-Sheleg E. Current and Future Perspective of PET/CT in Response Assessment of Malignant Pleural Mesothelioma. Semin Nucl Med 2025; 55:252-263. [PMID: 40021361 DOI: 10.1053/j.semnuclmed.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 02/06/2025] [Indexed: 03/03/2025]
Abstract
Malignant pleural mesothelioma (MPM) is a rare but aggressive cancer characterized by its unique growth patterns, presenting substantial diagnostic challenges. With the shift toward immunotherapy for MPM treatment, assessing therapeutic responses has become increasingly complex. Recent studies indicate that FDG PET/CT may provide more effective response criteria compared to traditional CT-based methods. This review emphasizes the important role of PET/CT in offering deep insights into the disease state and monitoring treatment responses. It also addresses the challenges associated with current imaging criteria, particularly the nonspecificity of FDG uptake that may represent inflammatory responses following treatments or procedures rather than tumor activity. Furthermore, the review discusses the potential of emerging radiopharmaceuticals and advanced volumetric assessments, discussing their implications for improving diagnostic accuracy and treatment evaluation in MPM.
Collapse
Affiliation(s)
- Manar Badarna
- Department of Nuclear Medicine, Rambam Healthcare Campus, Haifa, Israel
| | - Zohar Keidar
- Department of Nuclear Medicine, Rambam Healthcare Campus, Haifa, Israel; Rappaport Faculty of Medicine, Technion - The Israeli Institute of Technology, Haifa, Israel.
| | - Elite Arnon-Sheleg
- Departments of Nuclear Medicine and Diagnostic Radiology, Galilee Medical Center, Nahariya, and the Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
32
|
Huang Z, Karaboué A, Zeng L, Lecoeuvre A, Zhang L, Li XM, Qin H, Danino G, Yang F, Malin MS, Deng L, Rigal M, Liu H, Chen X, Xu Q, Grimaldi L, Collon T, Wang J, Adam R, Yang N, Duchemann B, Zhang Y, Lévi F. Overall survival according to time-of-day of combined immuno-chemotherapy for advanced non-small cell lung cancer: a bicentric bicontinental study. EBioMedicine 2025; 113:105607. [PMID: 39983330 PMCID: PMC11893323 DOI: 10.1016/j.ebiom.2025.105607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/27/2025] [Accepted: 02/05/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND Circadian rhythms regulate immune cell activity, influencing responses to vaccines, and immune checkpoint inhibitors (ICIs). Early time-of-day administration (ToDA) of singe-agent ICIs has been associated with improved overall survival (OS) in patients with metastatic "immunotherapy sensitive" cancers. However, the impact of ToDA on OS in patients receiving combination therapy with ICIs and chemotherapy for advanced non-small cell lung cancer (NSCLC) remains unclear. METHODS This retrospective study included patients from oncology units in Paris, France (Cohort 1) and Hunan, China (Cohort 2) who received first-line immuno-chemotherapy for stage IIIC or IV NSCLC between January 2018 and October 2023. The primary outcome was OS. The median ToDA of the initial four ICI infusions was computed for each patient. Hazard ratio (HR) for death or progression were determined using cut-off times ranging from 10:30 to 13:00. Kaplan Meier and Cox models were used to estimate OS and progression-free survival (PFS) adjusting for main patient characteristics. FINDINGS The study included 713 patients (Cohort 1, n = 165; Cohort 2, n = 548). Pembrolizumab was the most common ICI (51%), which was used with either pemetrexed-carboplatin/cisplatin (49%) or paclitaxel-carboplatin (51%). The optimal ToDA cut-off was 11:30, with patients receiving immuno-chemotherapy before 11:30 showing significantly improved OS (33.0 months [95% CI, 27.5-41.0] vs 19.5 months [18.0-22.5]; p < 0.0001). Multivariable analysis confirmed that earlier ToDA was associated with better OS (adjusted HR = 0.47 [95% CI, 0.37-0.60]). ToDA significantly impacted OS in each cohort and for PFS and response rates in each cohort and the pooled data. INTERPRETATION This sizeable bi-continental study provided real-world evidence that morning administration of standard first-line immuno-chemotherapy was associated with improved clinical outcomes compared to afternoon dosing in patients with NSCLC. Randomised trials are required to validate this finding and inform recommendations for clinical practice. FUNDING National Natural Science Foundation of China (82222048, 82003206, 82173338, and 82102747).
Collapse
Affiliation(s)
- Zhe Huang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China; Department of Pathology, School of Basic Medical Science, Central South University, Changsha, 410013, China
| | - Abdoulaye Karaboué
- Research Unit "Chronotherapy, Cancer, Transplantation", Faculty of Medicine, Paris-Saclay University, Hospital Paul Brousse, Villejuif, 94800, France; Medical Oncology Unit, GHT Paris Grand Nord-Est, Le Raincy-Montfermeil, Montfermeil, 93770, France
| | - Liang Zeng
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Adrien Lecoeuvre
- Clinical Research Unit, Paris Saclay University and Assistance Publique-Hôpitaux de Paris, Bicêtre Hospital, Le Kremlin Bicêtre, 94275, France
| | - Lemeng Zhang
- Department of Thoracic Oncology, Hunan Cancer Hospital, Changsha, 410013, China
| | - Xiao-Mei Li
- Research Unit "Chronotherapy, Cancer, Transplantation", Faculty of Medicine, Paris-Saclay University, Hospital Paul Brousse, Villejuif, 94800, France
| | - Haoyue Qin
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Gabrielle Danino
- Thoracic and Medical Oncology Unit, Avicenne Hospital, Assistance Publique - Hôpitaux de Paris, Bobigny, 93000, France
| | - Feng Yang
- Early Clinical Trial Centre, Office of National Drug Clinical Trial Institution, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Marie-Sara Malin
- Pharmacology Unit, Avicenne Hospital, Assistance Publique - Hôpitaux de Paris, Bobigny, 93000, France
| | - Li Deng
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Marte Rigal
- Pharmacology Unit, Avicenne Hospital, Assistance Publique - Hôpitaux de Paris, Bobigny, 93000, France
| | - Hong Liu
- Department of Dermatology, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Centre of Skin Health and Disease, Xiangya Clinical Research Centre for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Xiang Chen
- Department of Dermatology, Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Centre of Skin Health and Disease, Xiangya Clinical Research Centre for Cancer Immunotherapy, Xiangya Hospital, Central South University, Changsha, 410013, China
| | - Qinqin Xu
- Department of Medical Oncology, Qinghai Provincial People's Hospital, Xining, 810000, China
| | - Lamiae Grimaldi
- Clinical Research Unit, Paris Saclay University and Assistance Publique-Hôpitaux de Paris, Bicêtre Hospital, Le Kremlin Bicêtre, 94275, France
| | - Thierry Collon
- Medical Oncology Unit, GHT Paris Grand Nord-Est, Le Raincy-Montfermeil, Montfermeil, 93770, France
| | - Jing Wang
- Early Clinical Trial Centre, Office of National Drug Clinical Trial Institution, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - René Adam
- Research Unit "Chronotherapy, Cancer, Transplantation", Faculty of Medicine, Paris-Saclay University, Hospital Paul Brousse, Villejuif, 94800, France; Department of Hepatobiliary Surgery, Cancer and Transplantation, AP-HP Paul Brousse Hospital, Paris-Saclay University, Villejuif, 94800, France
| | - Nong Yang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Boris Duchemann
- Research Unit "Chronotherapy, Cancer, Transplantation", Faculty of Medicine, Paris-Saclay University, Hospital Paul Brousse, Villejuif, 94800, France; Thoracic and Medical Oncology Unit, Avicenne Hospital, Assistance Publique - Hôpitaux de Paris, Bobigny, 93000, France
| | - Yongchang Zhang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China; Department of Pathology, School of Basic Medical Science, Central South University, Changsha, 410013, China; Early Clinical Trial Centre, Office of National Drug Clinical Trial Institution, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China; Furong Laboratory, Changsha, 410013, China.
| | - Francis Lévi
- Research Unit "Chronotherapy, Cancer, Transplantation", Faculty of Medicine, Paris-Saclay University, Hospital Paul Brousse, Villejuif, 94800, France; Gastro-Intestinal and Medical Oncology Department, Paul Brousse Hospital, Assistance Publique-Hôpitaux de Paris, Villejuif, 94800, France; Department of Statistics, University of Warwick, Coventry, CV4 7AL, United Kingdom.
| |
Collapse
|
33
|
Xiao A, Li X, Wang C, Fakih M. Clinical Outcomes of Elective Early Discontinuation of Immunotherapy Based on Objective Response in Microsatellite Instability-High Metastatic Colorectal Cancer. Clin Colorectal Cancer 2025; 24:32-38.e1. [PMID: 39256132 DOI: 10.1016/j.clcc.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024]
Abstract
BACKGROUND Patients with microsatellite-high (MSI-H) metastatic colorectal cancers (CRC) may experience long-lasting benefit from immune checkpoint inhibitors (ICI) upon stopping therapy. However, optimal timing and patient selection criteria for early treatment withdrawal remain undefined. In this single-center retrospective study, we characterized the clinical response and associated survival outcomes of patients who received elective early versus late treatment discontinuation. METHODS We retrospectively analyzed patients with MSI-H metastatic CRC treated with ICI therapy from May 2015 to April 2024. Early ICI discontinuation was defined as treatment withdrawal before 2 years, and late ICI discontinuation as after 2 years. Response was assessed using Response Evaluation Criteria in Solid Tumors. Progression-free survival (PFS) and overall survival (OS) were estimated using the Kaplan Meier method. Efficacy outcomes between early and late ICI discontinuation groups were compared using a log-rank test. RESULTS Of 36 patients with MSI-H metastatic CRC, 12 underwent elective early ICI discontinuation and 9 experienced late ICI discontinuation. After a median follow-up of 32 months post-treatment, 91.7% (11/12) in the early discontinuation group remain off therapy without progression. PFS and OS outcomes between the early and late discontinuation groups were similarly favorable (P = .88 and P = .85, respectively), despite a 12-month difference in median duration of ICI therapy (13.3 and 25.6 months, respectively). The most common reason for elective early treatment discontinuation was clinical remission (n = 10), defined as a complete response, or a partial response with negative PET and/or ctDNA testing. CONCLUSIONS Early ICI discontinuation guided by response criteria resulted in low rates of recurrence. Survival outcomes between early and late ICI discontinuation groups were comparable, suggesting that treatment duration can be individualized based on clinical response without compromising favorable long-term prognosis.
Collapse
Affiliation(s)
- Annie Xiao
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Xiaochen Li
- Department of Computational and Quantitative Medicine, Division of Biostatistics, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Chongkai Wang
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Marwan Fakih
- Department of Medical Oncology and Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA.
| |
Collapse
|
34
|
Coschi CH, Ding K, Tong J, Tu D, O'Callaghan C, Leighl NB, Vera-Badillo F, Juergens RA, Hao D, Seymour L, Renouf DJ, Chen E, Gaudreau PO, Fung AS. Effects of cannabinoids on immune checkpoint inhibitor response: CCTG pooled analysis of individual patient data. Immunotherapy 2025; 17:257-268. [PMID: 40184324 DOI: 10.1080/1750743x.2025.2485012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/24/2025] [Indexed: 04/06/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) benefit patients across various tumor types. ICIs block cancer and T-cell interactions whereas cannabinoids may inhibit T-cell activation, reducing lysis of tumor cells. Interactions between cannabinoid use and dual ICI treatment remain unknown. METHODS Individual patient data from 4 Canadian Cancer Trials Group (CCTG) trials of patients treated with dual ICI ± chemotherapy (n = 684) were pooled. Cochran - Mantel - Haenszel and log-rank tests (stratified by trial/treatment arms) correlated cannabinoid use with clinicopathologic characteristics, Best Overall Response (BOR)/iBOR per RECIST 1.1/iRECIST, Progression-Free Survival (PFS)/iPFS, Overall Survival (OS) and immune-related adverse events (irAEs). RESULTS Sixty-five (9.5%) patients took cannabinoids at any time on trial, 32 (4.7%) of which were using cannabinoids at baseline. By multivariate analysis, cannabinoid use at baseline was significantly associated with improved iPFS (0.05), but not iBOR (p = 0.15), PFS (p = 0.12), OS (p = 0.35) or incidence of grade 1/2 or 3/4 irAEs (p = 0.96 and 0.65 respectively). Results were not significantly different with cannabinoid use at any time on trial. CONCLUSION Improved iPFS with cannabinoid use in patients treated with durvalumab plus tremelimumab ± chemotherapy did not translate into OS benefits. This study supports the safe use of cannabinoids in the context of combination ICI therapy.
Collapse
Affiliation(s)
| | - Keyue Ding
- Canadian Cancer Trials Group, Kingston, ON, Canada
| | - Justin Tong
- Kingston Health Sciences Centre, Kingston, ON, Canada
| | - Dongsheng Tu
- Canadian Cancer Trials Group, Kingston, ON, Canada
| | | | | | | | | | - Desiree Hao
- Arthur J.E. Child Comprehensive Cancer Centre and Cumming School of Medicine, Calgary, AB, Canada
| | | | | | - Eric Chen
- Princess Margaret Cancer Centre, Toronto, ON, Canada
| | | | - Andrea S Fung
- Arthur J.E. Child Comprehensive Cancer Centre, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
35
|
Nelson B, Naing A, Fu S, Sheth R, Murthy R, Piha-Paul S. Potentiating intratumoral therapy with immune checkpoint inhibitors: shifting the paradigm of multimodality therapeutics. IMMUNO-ONCOLOGY TECHNOLOGY 2025; 25:101040. [PMID: 39981134 PMCID: PMC11841068 DOI: 10.1016/j.iotech.2024.101040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized oncology, yielding remarkable and durable responses in various cancers. However, a significant proportion of patients develop resistance to ICIs. The tumor microenvironment plays a critical role in immunotherapy resistance, characterized by immune cell composition, regulatory factors, and tumor mutational burden. Intratumoral immunotherapy, involving direct injection of immune-activating agents into tumors, holds promise for converting immunologically 'cold' tumors into responsive 'hot' tumors. This review explores the rationale for combining intratumoral therapies (ITs) with ICIs, highlighting their complementary mechanisms of action and clinical effects. Notable IT approaches include oncolytic viruses, toll-like receptor agonists, and stimulator of interferon gene agonists with ICIs. Overall, combining ITs with ICIs offers a rational strategy to potentiate antitumor immune response and overcome resistance. Further research is needed to optimize the combination strategies, identify biomarkers of response, and establish the safety and efficacy of these novel therapeutic approaches.
Collapse
Affiliation(s)
- B.E. Nelson
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, USA
| | - A. Naing
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, USA
| | - S. Fu
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, USA
| | - R.A. Sheth
- Department of Interventional Radiology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - R. Murthy
- Department of Interventional Radiology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - S. Piha-Paul
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
36
|
Li Y, Liang F, Li Z, Zhang X, Wu A. Neoadjuvant Immunotherapy for Patients With Microsatellite Instability-High or POLE-Mutated Locally Advanced Colorectal Cancer With Bulky Tumors: New Optimization Strategy. Clin Colorectal Cancer 2025; 24:18-31.e2. [PMID: 39095269 DOI: 10.1016/j.clcc.2024.07.001] [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: 04/28/2024] [Revised: 07/01/2024] [Accepted: 07/01/2024] [Indexed: 08/04/2024]
Abstract
OBJECTIVE To evaluate the efficacy and safety of neoadjuvant immunotherapy for patients with microsatellite instability-high (MSI-H) or DNA polymerase ε (POLE)-mutated locally advanced colorectal cancer (LACRC) with bulky tumors. PATIENTS: We retrospectively reviewed 22 consecutive patients with MSI-H or POLE-mutated LACRC with bulky tumors (>8 cm in diameter) who received preoperative programmed death-1 blockade, with or without CapOx chemotherapy. MAIN OUTCOME MEASURES: Pathological complete response (pCR), clinical complete response (cCR), toxicity, R0 resection rate, and complications were evaluated. Survival outcomes were analyzed using the Kaplan-Meier method. Multiplex immunofluorescence analysis were performed before and after treatment. RESULTS: The incidence of immune-related adverse events (irAEs) was 36.4% (8/22). Five of 22 patients presented with surgical emergencies, most commonly perforation or obstruction. The 22 patients underwent a median 4 (1-8) cycles. Two patients were evaluated as cCR and underwent a watch and wait strategy. The R0 resection rate was 100.0% (20/20) and pCR rate was 70.0% (14/20). Twelve of 14 cT4b patients (85.7%) avoided multivisceral resection, and 10 of them achieved pCR or cCR. In the two patients with POLE mutations, one each achieved pCR and cCR. No Grade III/IV postoperative complications occurred. The median follow-up was 16.0 months. Two-year event-free and overall survival for the whole cohort was both 100%. CONCLUSIONS: Preoperative immunotherapy is the optimal option for MSI-H or POLE-mutated LACRC with bulky tumors, especially cT4b. Preoperative immunotherapy in patients with T4b CRC can reduce multivisceral resection and achieve high CR rate.
Collapse
Affiliation(s)
- Yingjie Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Unit III, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| | - Fei Liang
- Department of Biostatistics, Clinical Research Unit, Institute of Clinical Science Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhongwu Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Pathology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiaoyan Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Radiology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Aiwen Wu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Gastrointestinal Cancer Center, Unit III, Peking University Cancer Hospital & Institute, Beijing 100142, China.
| |
Collapse
|
37
|
Guigal-Stephan N, Lockhart B, Moser T, Heitzer E. A perspective review on the systematic implementation of ctDNA in phase I clinical trial drug development. J Exp Clin Cancer Res 2025; 44:79. [PMID: 40022112 PMCID: PMC11871688 DOI: 10.1186/s13046-025-03328-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/13/2025] [Indexed: 03/03/2025] Open
Abstract
Circulating tumour DNA (ctDNA) represents an increasingly important biomarker for the screening, diagnosis and management of patients in clinical practice in advanced/metastatic disease across multiple cancer types. In this context, ctDNA-based comprehensive genomic profiling is now available for patient management decisions, and several ctDNA-based companion diagnostic assays have been approved by regulatory agencies. However, although the assessment of ctDNA levels in Phase II-III drug development is now gathering momentum, it remains somewhat surprisingly limited in the early Phase I phases in light of the potential opportunities provided by such analysis. In this perspective review, we investigate the potential and hurdles of applying ctDNA testing for the inclusion and monitoring of patients in phase 1 clinical trials. This will enable more informed decisions regarding patient inclusion, dose optimization, and proof-of-mechanism of drug biological activity and molecular response, thereby supporting the evolving oncology drug development paradigm. Furthermore, we will highlight the use of cost-efficient, agnostic genome-wide techniques (such as low-pass whole genome sequencing and fragmentomics) and methylation-based methods to facilitate a more systematic integration of ctDNA in early clinical trial settings.
Collapse
Affiliation(s)
- Nolwen Guigal-Stephan
- Translational Medicine, Institut de Recherches Servier, 22 route 128, Gif-sur-Yvette, Saclay, 91190, France.
| | - Brian Lockhart
- Translational Medicine, Institut de Recherches Servier, 22 route 128, Gif-sur-Yvette, Saclay, 91190, France
| | - Tina Moser
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstrasse 6, Graz, 8010, Austria
| | - Ellen Heitzer
- Institute of Human Genetics, Diagnostic & Research Center for Molecular BioMedicine, Medical University of Graz, Neue Stiftingtalstrasse 6, Graz, 8010, Austria.
| |
Collapse
|
38
|
Ayeni A, Evbuomwan O, Vangu MDTW. The Role of [ 18F]FDG PET/CT in Monitoring of Therapy Response in Lung Cancer. Semin Nucl Med 2025; 55:175-189. [PMID: 40021362 DOI: 10.1053/j.semnuclmed.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 03/03/2025]
Abstract
Lung cancer remains a leading cause of cancer deaths worldwide, with an all stage 5-year relative survival rate of less than 30%. Multiple treatment strategies are available and continue to evolve, with therapy primarily tailored to the type and stage of the disease. Accurate monitoring of therapy response is crucial for optimizing treatment outcomes. PET/CT imaging with [18F]FDG has become the standard of care across various phases of lung cancer management due to its ability to assess metabolic activity. This review underscores the pivotal role of [18F]FDG PET/CT in evaluating therapy response in lung cancer, particularly in non-small cell lung cancer (NSCLC). It examines conventional response criteria and their adaptations in the era of immunotherapy, highlighting the value of integrating metabolic imaging with established criteria to improve treatment assessment and guide clinical decisions. The potential of non-[18F]FDG PET tracers targeting diverse biological pathways to provide deeper insights into tumor biology, therapy response and predictive outcomes is also explored. Additionally, the emerging role of radiomics in enhancing treatment efficacy assessment and improving patient management is briefly highlighted. Despite the challenges in the routine clinical application of various metabolic response criteria, [18F]FDG PET/CT remains a crucial tool in monitoring therapy response in lung cancer. Ongoing advancements in therapeutic strategies, radiopharmaceuticals, and imaging techniques continue to drive progress in lung cancer management, promising improved patient outcomes.
Collapse
Affiliation(s)
- Akinwale Ayeni
- Division of Nuclear Medicine, Department of Radiation Sciences, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa; Nuclear Medicine, Klerksdorp/Tshepong Hospital Complex, Klerksdorp, North West Province, South Africa; Division of Nuclear Medicine, Department of Radiation Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa.
| | - Osayande Evbuomwan
- Department of Nuclear Medicine, Faculty of Health Sciences, University of The Free State, Bloemfontein, South Africa
| | - Mboyo-Di-Tamba Willy Vangu
- Division of Nuclear Medicine, Department of Radiation Sciences, Faculty of Health Sciences, University of The Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
39
|
Ge H, Liu C, Shen C, Hu D, Zhao X, Wang Y, Ge H, Qin R, Ma X, Wang Y. The effectiveness and safety of RC48 alone or in combination with PD-1 inhibitors for locally advanced or metastatic urothelial carcinoma: a multicenter, real-world study. J Transl Med 2025; 23:243. [PMID: 40022107 PMCID: PMC11871675 DOI: 10.1186/s12967-025-06237-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/11/2025] [Indexed: 03/03/2025] Open
Abstract
BACKGROUND RC48 is an antibody-drug conjugate (ADC) specifically targeting HER2. Phase II and III clinical trials have proven its significant anti-tumor effect against locally advanced or metastatic urothelial carcinoma (la/mUC). This study aims to further assess the effectiveness and safety of RC48 for patients with la/mUC and provide insights for further clinical practice. METHODS Retrospective analysis for 42 patients with la/mUC who underwent RC48 alone or in combination with PD-1 inhibitors therapy between 18 October 2022 and 1 May 2024 were conducted to assess effectiveness and safety of RC48. Descriptive statistics were used to summarize baseline characteristics, treatment-related adverse events, etc. Cox proportional risk model and the Kaplan-Meier method were applied to analyze patients' survival. RESULTS We observed a median progression-free survival (mPFS) of 6.2 months, although median overall survival (mOS) has not been reached so far. An objective response rate (ORR) of 54.8% and a disease control rate (DCR) of 83.3% was also observed. Patients with first-line therapy, second- or later-line therapy and neoadjuvant therapy were observed disease remission with ORRs of 47.7%, 40.0% and 100.0%, respectively. The most common treatment-related adverse events (TRAEs) include hypoesthesia and elevated transaminases which affect over 90.0% of patients and mostly grade 1-2 in severity, and no treatment-related fatalities were found. CONCLUSIONS This multicenter, real-world study confirms that RC48 alone or in combination with PD-1 inhibitors exerted a promising effectiveness and manageable safety for first-line, second- and post-line, and neoadjuvant therapy with la/mUC.
Collapse
Affiliation(s)
- Huaixi Ge
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Changxue Liu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Chengquan Shen
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ding Hu
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xinzhao Zhao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yanhua Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Huimin Ge
- Department of Critical Care Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ruize Qin
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaocheng Ma
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yonghua Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China.
- Urinary Diseases Clinical Medical Research Center of Qingdao, Qingdao, Shandong, China.
- Shandong Province Medical and Health Key Laboratory of Urology, Qingdao, Shandong, China.
| |
Collapse
|
40
|
Hua Y, Wang A, Xie C, Agrafiotis AC, Zhang P, Li B. Clinical-genomic nomogram for predicting sensitivity to second-line immunotherapy for advanced non-small cell lung cancer. Transl Lung Cancer Res 2025; 14:526-537. [PMID: 40114939 PMCID: PMC11921187 DOI: 10.21037/tlcr-2024-1249] [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: 12/22/2024] [Accepted: 02/18/2025] [Indexed: 03/22/2025]
Abstract
Background The introduction of immune checkpoint inhibitors (ICIs) has significantly improved the outcomes of patients with advanced non-small cell lung cancer (NSCLC). However, ICIs only benefit a subset of patients. The study aimed to identify genomic biomarkers and construct models to predict the response to second-line ICI therapy. Methods We retrospectively collected clinical data and genetic testing results from patients with NSCLC treated with second-line ICI at a single medical center between August 2018 and June 2021. We reanalyzed the raw sequence data of clinical genetic testing and defined the common detection region among the different testing panels. Immunotherapy sensitivity was evaluated using the immune-based Response Evaluation Criteria in Solid Tumors. Results We included 102 patients as a training cohort and 46 as a test cohort. In the training cohort, we examined the relationship between ICI response and the mutation status of 343 genes. Mutations in the EGFR gene were significantly more common in the resistant group than in the sensitive group (41.0% vs. 20.6%; P=0.04), while mutations in the EP300 gene were associated with greater sensitivity to ICIs (39.7% vs. 15.4%; P=0.01). A nomogram was built based on clinical variables, genomic data, and programmed death-ligand 1 (PD-L1) expression. The total nomogram points were significantly higher in the sensitive group than in the resistance group in both cohorts, and the areas under the receiver operating characteristic curve were 0.780 in the training cohort and 0.720 in the test cohort. The higher nomogram points also indicated better progression-free survival. Conclusions Based on real-world clinical settings, the clinical genomic nomogram, which involved limited input variables that were economical and easy to obtain, demonstrated a good ability to predict the response to second-line ICI treatment in advanced NSCLC.
Collapse
Affiliation(s)
- Ying Hua
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Radiation Oncology, Tianjin Medical University, Tianjin, China
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ai Wang
- Department of Oncology, Heze Hospital of Traditional Chinese Medicine, Heze, China
| | - Chao Xie
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Apostolos C Agrafiotis
- Department of Thoracic Surgery, Saint-Pierre University Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Pinlang Zhang
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Baosheng Li
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Radiation Oncology, Tianjin Medical University, Tianjin, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
41
|
Fung T, Samlowski W, Meoz R. Effectiveness and Toxicity of Cemiplimab Therapy for Advanced Cutaneous Squamous Cell Skin Cancer in a Community Oncology Practice. Cancers (Basel) 2025; 17:823. [PMID: 40075670 PMCID: PMC11899135 DOI: 10.3390/cancers17050823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND The immune checkpoint inhibitor cemiplimab has significant clinical activity in unresectable and metastatic cutaneous squamous cell carcinomas. There are limited real-world data available to assess the outcome of cemiplimab treatment in patients in a community practice setting. METHODS We conducted a retrospective analysis of treatment outcomes following cemiplimab treatment (350 mg IV every 3 weeks) of squamous cell skin cancer. An exploratory analysis was performed to evaluate patient subsets, including patients with locally advanced disease, regional or distant metastases, and "too numerous to count" primaries. Another small group of patients who did not respond to the initial four doses of cemiplimab were evaluated following added radiotherapy. RESULTS Of the 36 patients treated, 22 (61.1%) achieved complete remission, 10 (27.8%) experienced a partial response, 3 (8.3%) had stable disease, and 1 (2.8%) developed progressive disease. The median progression-free survival for the entire cohort was over 33 months. Overall, cemiplimab was well-tolerated, with no hospitalizations due to treatment-related toxicity. CONCLUSIONS Cemiplimab produced complete remissions in over 60% of patients with locally advanced and metastatic squamous cell skin cancers, allowing elective treatment discontinuation. Addition of radiotherapy in cemiplimab-refractory patients appeared to increase tumor responsiveness. In contrast, patients with TNTC primary tumors frequently develop new primary skin cancers. Thus, improved treatment options for this patient subset are still needed.
Collapse
Affiliation(s)
- Tina Fung
- Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV 89102, USA;
| | - Wolfram Samlowski
- Kirk Kerkorian School of Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV 89102, USA;
- Comprehensive Cancer Centers of Nevada, Las Vegas, NV 89148, USA
| | - Raul Meoz
- Comprehensive Cancer Centers of Nevada, Las Vegas, NV 89148, USA
| |
Collapse
|
42
|
Laktionov K, Smolin A, Stroyakovskiy D, Moiseenko V, Dvorkin M, Andabekov T, Cheng Y, Liu B, Kozlov V, Odintsova S, Dvoretsky S, Mochalova A, Urda M, Yi T, Li X, László U, Müller V, Bogos K, Fadeeva N, Musaev G, Liu Q, Kirtbaya D, Shi J, Gladkov O, Narimanov M, Semiglazova T, Khasanova A, Chovanec J, Andrašina I, Szabová A, Rosinská O, Sudekova D, Zsolt PS, Ran F, Sun M, Jiang O, Chen R, Zhao E, Liu C, Tan W, Pirmagomedov A, Poddubskaya E, Kislov N, Shumskaya I, Sorokina I, Zinkina-Orikhan A, Linkova Y, Fogt S, Liaptseva D, Siliutina A, Basova O, Kryukov F. Prolgolimab with chemotherapy as first-line treatment for advanced non-squamous non-small-cell lung cancer. Eur J Cancer 2025; 217:115255. [PMID: 39879779 DOI: 10.1016/j.ejca.2025.115255] [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: 08/23/2024] [Revised: 12/06/2024] [Accepted: 01/17/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Prolgolimab is an IgG1 anti-PD-1 monoclonal antibody with the Fc-silencing 'LALA' mutation. The phase III DOMAJOR study assessed efficacy and safety of prolgolimab in combination with pemetrexed and platinum-based chemotherapy vs placebo in combination with pemetrexed and platinum-based chemotherapy as first-line treatment for advanced non-small cell lung cancer (NSCLC). METHODS 292 patients with advanced non-squamous NSCLC were randomized 1:1 to receive 4 cycles of pemetrexed, platinum-based drug and either prolgolimab (3 mg/kg Q3W) or placebo followed by prolgolimab/placebo with pemetrexed until disease progression or toxicity (≤36 months). The primary endpoint was overall survival (OS). RESULTS After a median follow-up of 18 months, the median OS was not reached (95 % CI, 22.28 - NA) in the prolgolimab-combination group vs 14.6 months (95 % CI, 11.73 - 19.15) in the placebo-combination group (HR, 0.51; 95 % CI, 0.35 - 0.73, p = 0.0001). The OS improvement was independent of PD-L1 status. Median progression-free survival (PFS) per Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST 1.1) was 7.7 months in the prolgolimab-combination group and 5.5 months in the placebo-combination group (HR, 0.65; 95 % CI, 0.49 - 0.85, p = 0.0004). The only adverse events that were reported in at least 10 % of the patients that were significantly more frequent in the prolgolimab-combination group were blood creatinine increased and dyspnoea. CONCLUSION Among patients with advanced NSCLC the addition of prolgolimab to pemetrexed and a platinum-based drug increased OS and PFS, with no new safety concerns. This benefit was retained in patients with PD-L1 negative tumors. (ClinicalTrials.gov, NCT03912389).
Collapse
Affiliation(s)
- K Laktionov
- FSBI "N.N. Blokhin National Medical Research Center of Oncology" of the Russian Ministry of Health, Moscow, Russia
| | - A Smolin
- FSBI "Academician N.N. Burdenko Main Military Clinical Hospital", MoH, Moscow, Russia
| | | | - V Moiseenko
- SBHI "N.P. Napalkov St. Petersburg Clinical Scientific and Practical Center for Specialized Types of Medical Care (Oncology)", St. Petersburg, Russia
| | - M Dvorkin
- BHI "Clinical Oncology Dispensary", Omsk, Russia
| | - T Andabekov
- Limited Liability Company AV Medical Group, St. Petersburg, Russia
| | - Y Cheng
- Jilin Cancer Hospital, Changchun, China
| | - B Liu
- Harbin Medical University Cancer Hospital, Harbin, China
| | - V Kozlov
- SBHI "Novosibirsk Regional Clinical Oncology Dispensary", Novosibirsk, Russia
| | - S Odintsova
- JSC Modern Medical Technologies, St. Petersburg, Russia
| | - S Dvoretsky
- FSBEI of Higher Education "Academician I.P. Pavlov First St. Petersburg State Medical University", St. Petersburg, Russia
| | - A Mochalova
- JSC Medsi Group of Companies, Moscow, Russia
| | - M Urda
- F.D. Roosevelt Faculty Hospital with Polyclinic in Banska Bystrica (Fakultná nemocnica s poliklinikou F. D. Roosevelta), Banska Bystrica, Slovakia
| | - T Yi
- Xiangyang Central Hospital, Xiangyang, China
| | - X Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - U László
- Mátrai Gyógyintézet, Mátraháza, Hungary
| | - V Müller
- Semmelweis Egyetem Pulmonológiai Klinika, Budapest, Hungary
| | - K Bogos
- Országos Korányi Pulmonológiai Intézet IV. Tüdőbelosztály, Budapest, Hungary
| | - N Fadeeva
- SBHI "Chelyabinsk Regional Clinical Center of Oncology and Nuclear Medicine", Chelyabinsk, Russia
| | - G Musaev
- FSBI "National Medical and Surgical Center named after N.I. Pirogov", MoH, Moscow, Russia
| | - Q Liu
- The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - D Kirtbaya
- SBHI "Oncology Dispensary No. 2", Sochi, Russia
| | - J Shi
- Linyi Cancer Hospital, Linyi, China
| | - O Gladkov
- Limited Liability Company EVIMED, Chelyabinsk, Russia
| | - M Narimanov
- SBHI "Podolsk Regional Clinical Hospital", Podolsk, Russia
| | - T Semiglazova
- FSBI "N.N. Petrov National Medical Research Center for Oncology", MoH, St. Petersburg, Russia
| | - A Khasanova
- SAHI "M.Z. Sigal Republican Clinical Oncology Dispensary of the Ministry of Health of the Republic of Tatarstan", Kazan, Russia
| | - J Chovanec
- Nemocnica s poliklinikou Sv. Jakuba, Bardejov, Slovakia
| | - I Andrašina
- Východoslovenský onkologický ústav, a.s. Nemocničná lekáreň VITA, Košice, Slovakia
| | - A Szabová
- Nemocnica na okraji mesta, n.o. Onkologická ambulancia, Partizánske, Slovakia
| | - O Rosinská
- Nemocnica Komárno s.r.o. Oddelenie klinickej onkológie, Komárno, Slovakia
| | - D Sudekova
- Fakultná nemocnica s poliklinikou Žilina Klinická a radiačná onkológia, Žilina, Slovakia
| | - P-S Zsolt
- Fejér Megyei Szent György Egyetemi Oktatókórház Pulmonológia, Székesfehérvár, Hungary
| | - F Ran
- Hubei Cancer Hospital, Wuhan, China
| | - M Sun
- Jinan Central Hospital, Jinan, Russia
| | - O Jiang
- The Second People's Hospital of Neijiang, Neijiang, China
| | - R Chen
- Liuzhou People's Hospital, Liuzhou, China
| | - E Zhao
- Cangzhou Hospital of Integrated TCM-WM, Cangzhou, Hebei, China
| | - C Liu
- Third Affiliated Hospital of Xinjiang Medical University, Wulumuqi, China
| | - W Tan
- Weifang People's Hospital, Weifang, China
| | - A Pirmagomedov
- SBHI "Municipal Clinical Hospital No. 1", Nalchik, Russia
| | - E Poddubskaya
- FSAEI of Higher Education I.M. Sechenov First Moscow Medical University, MoH, Moscow, Russia
| | - N Kislov
- SBHI "Regional Clinical Oncology Hospital", Yaroslavl, Russia
| | - I Shumskaya
- SBHI "Nizhny Novgorod Regional Clinical Oncology Dispensary", Nizhny Novgorod, Russia
| | - I Sorokina
- JSC Biocad, St. Petersburg, Russia; MCSC named after A.S. Loginov, Moscow, Russia.
| | | | | | - S Fogt
- JSC Biocad, St. Petersburg, Russia
| | | | | | - O Basova
- JSC Biocad, St. Petersburg, Russia
| | | |
Collapse
|
43
|
Rosnev S, Sterner B, Schiele P, Kolling S, Martin M, Flörcken A, Erber B, Wittenbecher F, Kofla G, Kurreck A, Lang TJL, von Einem JC, de Santis M, Pelzer U, Stintzing S, Bullinger L, Klinghammer K, Geisel D, Ochsenreither S, Frentsch M, Na IK. Reduced monocytic IL10 expression in PD1 inhibitor-treated patients is a harbinger of severe immune-related adverse events. Eur J Cancer 2025; 217:115252. [PMID: 39848112 DOI: 10.1016/j.ejca.2025.115252] [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: 08/29/2024] [Revised: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
BACKGROUND Despite remarkable clinical efficacy, little is known about the system-wide immunological alterations provoked by PD1 blockade. Dynamics of quantitative immune composition and functional repertoire during PD1 blockade could delineate cohort-specific patterns of treatment response and therapy-induced toxicity. METHODS We longitudinally assessed therapy-induced effects on the immune system in fresh whole blood using flow cytometry-based cell quantifications, accompanied by analyses of effector properties of all major immune populations upon cell-type specific stimulations. 43 cancer patients undergoing PD1 blockade were recruited with assessments performed pre-treatment and before cycles 2/4/6, which resulted in the collection of more than 30,000 cytometric data values. RESULTS We observed no intrinsic immune pattern correlating with clinical outcome before PD1 blockade initiation, but cohort-specific immune alterations emerged during therapy. The most striking evolving changes in therapy responders were an increase in activated T and NK cell subsets, which showed high IFNγ and TNFα expression upon ex vivo stimulation. Patients affected by severe immune-related adverse events (s-irAE) presented with an analogously increased number of activated CD4 + and CD8 + T cells compared to patients with no/mild irAE, but lacked the functional divergences observed between responders versus non-responders. Instead, their monocytes showed discriminatory functional deficits with less IL10 production upon stimulation, which led to an abrogated inhibition of T cell proliferation in vitro and thus may account for the observed T cell expansion in patients with s-irAE. CONCLUSION Our holistic explorative approach allowed the delineation of clinically relevant cohorts by treatment-triggered immune changes, potentially enabling better patient stratification and further revealed new mechanistic insights into the pathogenesis of s-irAE.
Collapse
Affiliation(s)
- Stanislav Rosnev
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health Berlin, Germany
| | - Baldur Sterner
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health Berlin, Germany
| | - Phillip Schiele
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health Berlin, Germany
| | - Stefan Kolling
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health Berlin, Germany; Berlin School of Integrative Oncology, Berlin, Germany
| | - Markus Martin
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Anne Flörcken
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; German Cancer Consortium (DKTK), Berlin, Germany
| | - Barbara Erber
- Department of Urology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Friedrich Wittenbecher
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany
| | - Grzegorz Kofla
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Annika Kurreck
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Tonio Johannes Lukas Lang
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jobst C von Einem
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Maria de Santis
- Department of Urology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Urology, Medical University of Vienna, Vienna, Austria
| | - Uwe Pelzer
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sebastian Stintzing
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Lars Bullinger
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; German Cancer Consortium (DKTK), Berlin, Germany
| | - Konrad Klinghammer
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Dominik Geisel
- Department of Radiology, Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sebastian Ochsenreither
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; German Cancer Consortium (DKTK), Berlin, Germany; Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Marco Frentsch
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Berlin Institute of Health Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health Berlin, Germany; Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Il-Kang Na
- Department of Hematology, Oncology and Cancer Immunology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; German Cancer Consortium (DKTK), Berlin, Germany; Berlin Institute of Health (BIH), Berlin, Germany; Experimental and Clinical Research Center, A Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Center for Molecular Medicine, Berlin, Germany.
| |
Collapse
|
44
|
Liu J, Farrow M, Seymour L, Desai J, Loong HH, Ivy P, Koyoma T, Cook N, Blagden S, Garralda E, Massard C, Tolcher AW, Adashek JJ, Zhang L, Zhao S, Shen L, Kurzrock R, El-Deiry WS, Subbiah V, Joshua AM. Accelerating the Future of Oncology Drug Development: The Role of Consortia in the Delivery of Precision Oncology Early Phase Clinical Trials. J Clin Oncol 2025; 43:735-747. [PMID: 39808749 DOI: 10.1200/jco-24-01534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/15/2024] [Accepted: 11/14/2024] [Indexed: 01/16/2025] Open
Abstract
PURPOSE Over the past 15 years, the landscape of early phase clinical trials (EPCTs) has undergone a remarkable expansion in both quantity and intricacy. The proliferation of sites, trials, sponsors, and contract research organizations has surged exponentially, marking a significant shift in research conduct. However, EPCT operations suffer from numerous inefficiencies, such as cumbersome start-up processes, which are particularly critical when drug safety and the recommended phase II dose need to be established in a timely manner. Networks and consortia may overcome some of these challenges of enrolling suitable patients and streamlining start-up, particularly when distance and disease trajectory come into play. DESIGN In this article, we provide an overview of EPCT consortia in adult oncology across different continents assembled through systematic review of the literature and snowball sampling methodology. We illustrate their scope, structure, funding, and achievements. RESULTS Fifteen EPCT consortia were identified including two in the United States, three in Europe, five in Asia-Pacific, two intercontinental consortia, and three within private oncology networks. These consortia vary in their scope, funding, and structure from government-funded models such as the National Cancer Institute Experimental Therapeutics Clinical Trials Networks through charitably funded and private research organizations. EPCT consortia play a role in collaborative research, molecular tumor boards to provide patient-centric biomarker-matched treatments, and streamlining trial conduct to improve timelines and cost efficiency. CONCLUSION The growth in EPCT activity and complexity has resulted in expansion in the number of EPCT consortia globally. By actively engaging with regulatory bodies and pharmaceutical and contract research organization industries, consortia have an opportunity to address the evolving challenges faced in this field and to accelerate the translation of scientific discoveries into clinical practice.
Collapse
Affiliation(s)
- Jia Liu
- The Kinghorn Cancer Centre, St Vincent's Hospital, Sydney, NSW, Australia
- Faculty of Medicine & Health, School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Max Farrow
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Lesley Seymour
- Canadian Cancer Trials Group, Queens University, Kingston, ON, Canada
| | - Jayesh Desai
- Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Herbert H Loong
- The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - Percy Ivy
- National Cancer Institute, Bethesda, MD
| | | | - Natalie Cook
- The Christie NHS Foundation Trust and University of Manchester, Manchester, United Kingdom
| | - Sarah Blagden
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Elena Garralda
- Research Unit for Molecular Therapy of Cancer (UITM), Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Oncology Department, Hospital Universitario Vall d'Hebron (HUVH), Barcelona, Spain
- Phase I Unit-NEXT Oncology, Hospital Quirón Salud, Barcelona, Spain
| | - Christophe Massard
- Département d'Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy, Université Paris Saclay, Villejuif, France
| | | | - Jacob J Adashek
- START Center for Cancer Research, San Antonio, TX
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD
| | - Li Zhang
- Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Provincial Clinical Research Centre for Cancer, Guangzhou, China
| | - Shen Zhao
- Sun Yat-sen University Cancer Centre, State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Guangdong Provincial Clinical Research Centre for Cancer, Guangzhou, China
| | - Lin Shen
- Peking University Cancer Hospital & Institute, Beijing, China
| | - Razelle Kurzrock
- Medical College of Wisconsin, Milwaukie, WI
- Worldwide Innovative Network (WIN) Consortium in Cancer Personalized Medicine, Paris, France
| | - Wafik S El-Deiry
- Medical College of Wisconsin, Milwaukie, WI
- Worldwide Innovative Network (WIN) Consortium in Cancer Personalized Medicine, Paris, France
- The Legorreta Cancer Center at Brown University, Providence, RI
| | | | - Anthony M Joshua
- The Kinghorn Cancer Centre, St Vincent's Hospital, Sydney, NSW, Australia
- Faculty of Medicine & Health, School of Clinical Medicine, University of New South Wales, Sydney, NSW, Australia
- Garvan Institute of Medical Research, Sydney, NSW, Australia
| |
Collapse
|
45
|
Luke JJ, Pinato DJ, Juric D, LoRusso P, Hosein PJ, Desai AM, Haddad R, de Miguel M, Cervantes A, Kim WS, Marabelle A, Zhang Y, Rong Y, Yuan X, Champiat S. Phase I dose-escalation and pharmacodynamic study of STING agonist E7766 in advanced solid tumors. J Immunother Cancer 2025; 13:e010511. [PMID: 39979069 PMCID: PMC11842995 DOI: 10.1136/jitc-2024-010511] [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: 09/24/2024] [Accepted: 01/28/2025] [Indexed: 02/22/2025] Open
Abstract
E7766 is a novel stimulator of interferon genes (STING) agonist, capable of potent activation of immune cells and generating strong antitumor response in preclinical murine tumor models. Here we present the safety, efficacy, and biomarker results of the first-in-human phase I/Ib study of intratumoral E7766 in patients with advanced solid tumors. Eligible patients with relapsing/refractory cancers (n=24) were enrolled in dose-escalating cohorts to receive intratumoral injections of E7766 from 75 to 1000 µg. The most frequent treatment-related treatment-emergent adverse events were chills (50.0%; 85.7%), fever (40.0%; 85.7%), and fatigue (30.0%; 35.7%) in patients who received non-visceral and visceral injections, respectively. Eight patients (33.3%) achieved stable disease as their best response per modified Response Evaluation Criteria In Solid Tumors version 1.1 with variability between injected and non-injected lesions. Plasma levels of IFN-α, IFN-β, IFN-γ, TNF-α, IL-6, IP-10, MCP1, and MIP1b transiently increased in all evaluable patients within 10 hours postinjection, then dropped to baseline levels. Levels of blood and tumor gene expression increased in most interferon-related and STING genes tested. Further increases in programmed death ligand 1 and cluster of differentiation 8 expression at both the RNA and protein levels were also observed in some patients across dose levels. In total, E7766 generated on-target pharmacodynamic effects in patients with solid tumors. Further exploration in a homogeneous patient population is necessary to assess efficacy.
Collapse
Affiliation(s)
- Jason J Luke
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
- University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - David J Pinato
- Imperial College London, London, UK
- University of Piemonte Orientale, Novara, Italy
| | - Dejan Juric
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | | | - Peter J Hosein
- University of Miami Leonard M. Miller School of Medicine, Miami, Florida, USA
| | - Anupam M Desai
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Robert Haddad
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - María de Miguel
- START Madrid-HM CIOCC, Centro Integral Oncológico Clara Campal, Hospital Universitario HM Sanchinarro, Madrid, Spain
| | - Andrés Cervantes
- INCLIVA Biomedical Research Institute, University of Valencia, Valencia, Spain
- Instituto de Salud Carclos III. CIBERONC, Madrid, Spain
| | - Won Seog Kim
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
46
|
Cook AL, Sur S, Dobbyn L, Watson E, Cohen JD, Ptak B, Lee BS, Paul S, Hsiue E, Popoli M, Vogelstein B, Papadopoulos N, Bettegowda C, Gabrielson K, Zhou S, Kinzler KW, Wyhs N. Identification of nonsense-mediated decay inhibitors that alter the tumor immune landscape. eLife 2025; 13:RP95952. [PMID: 39960487 PMCID: PMC11832170 DOI: 10.7554/elife.95952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025] Open
Abstract
Despite exciting developments in cancer immunotherapy, its broad application is limited by the paucity of targetable antigens on the tumor cell surface. As an intrinsic cellular pathway, nonsense-mediated decay (NMD) conceals neoantigens through the destruction of the RNA products from genes harboring truncating mutations. We developed and conducted a high-throughput screen, based on the ratiometric analysis of transcripts, to identify critical mediators of NMD in human cells. This screen implicated disruption of kinase SMG1's phosphorylation of UPF1 as a potential disruptor of NMD. This led us to design a novel SMG1 inhibitor, KVS0001, that elevates the expression of transcripts and proteins resulting from human and murine truncating mutations in vitro and murine cells in vivo. Most importantly, KVS0001 concomitantly increased the presentation of immune-targetable human leukocyte antigens (HLA) class I-associated peptides from NMD-downregulated proteins on the surface of human cancer cells. KVS0001 provides new opportunities for studying NMD and the diseases in which NMD plays a role, including cancer and inherited diseases.
Collapse
Affiliation(s)
- Ashley L Cook
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Surojit Sur
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Laura Dobbyn
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
| | - Evangeline Watson
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Joshua D Cohen
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Blair Ptak
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
| | - Bum Seok Lee
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
| | - Suman Paul
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Emily Hsiue
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Maria Popoli
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Bert Vogelstein
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
- Howard Hughes Medical Institute, Johns Hopkins University School of MedicineBaltimoreUnited States
- Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Nickolas Papadopoulos
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Chetan Bettegowda
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Neurosurgery, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Kathy Gabrielson
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Shibin Zhou
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Kenneth W Kinzler
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
- Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Nicolas Wyhs
- Ludwig Center for Cancer Genetics and Therapeutics, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Oncology, Johns Hopkins Medical InstitutionsBaltimoreUnited States
- Sidney Kimmel Cancer Center, Johns Hopkins University School of MedicineBaltimoreUnited States
| |
Collapse
|
47
|
Liang H, Liang W, Tian J, Zheng S, Su Y, Huang P, Chen R, Guan Z, Cai Q. Safety and efficacy of neoadjuvant therapy with tislelizumab plus chemotherapy for locally advanced head and neck squamous cell carcinoma: a single-arm, retrospective study. Cancer Immunol Immunother 2025; 74:108. [PMID: 39932534 PMCID: PMC11813832 DOI: 10.1007/s00262-025-03953-0] [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: 11/07/2024] [Accepted: 01/21/2025] [Indexed: 02/14/2025]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is a very common type of head and neck cancer, and the 5-year overall survival (OS) rate is only 50%. Inhibitors targeting the programmed death 1 (PD-1) pathway have gained widespread clinical adoption. However, the relationship between the infiltration characteristics of immune cells in the tumor immune microenvironment (TIME) and the effect of immunotherapy on HNSCC remains to be explored. METHODS Patients diagnosed with HNSCC who received Tislelizumab combined with chemotherapy were reviewed between February 2021 and March 30, 2024, in our single center. The laryngoscopy, magnetic resonance imaging (MRI), and pathologic response were evaluated to the efficacy of neoadjuvant therapy with Tislelizumab plus chemotherapy treatment. Treatment-related adverse events (TRAEs) were evaluated according to the Common Terminology Criteria for Adverse Events version 5.0. RESULT Our analysis involved 42 patients who received Tislelizumab combined with chemotherapy. A total of 18 patients underwent surgical treatment following the completion of immunotherapy combined with chemotherapy. Among them, 6 patients achieved pCR (33%). The 1-year OS rate of the 42 patients enrolled in the study was 95.05%, and the 1-year PFS rate was 89.86%. There was a significant positive correlation between the lymphocyte density in HNSCC prior to the administration of neoadjuvant PD-1 inhibitor therapy combined with chemotherapy and the immunotherapy efficacy. Compared with pretreatment, the neutrophil-to-lymphocyte ratio (NLR) was significantly decreased, and the lymphocyte density was significantly increased in HNSCC patients after immunotherapy. CONCLUSIONS The integration of neoadjuvant PD-1 inhibitor therapy with chemotherapy has been demonstrated to be a safe and effective strategy, can improve the tumor response rate and survival rate, and is a valuable treatment for patients with HNSCC. Furthermore, the study suggests that an elevated NLR within the HNSCC tumor microenvironment could potentially serve as a biomarker indicative of diminished immunotherapy efficacy.
Collapse
Affiliation(s)
- Haifeng Liang
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
| | - Wenting Liang
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
| | - Jiawang Tian
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
| | - Shibei Zheng
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
- Department of Otorhinolaryngology, Head and Neck Surgery, Shenzhen Baoan District People's Hospital, Shenzhen, China
| | - Yangzhou Su
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
| | - Piao Huang
- Department of Pathology, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China
| | - Renhui Chen
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China.
| | - Zhong Guan
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China.
| | - Qian Cai
- Department of Otolaryngology-Head and Neck, Sun Yat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, China.
- Department of Otolaryngology-Head and Neck, Zhujiang Hospital of Southern Medical University, Guangzhou, China.
| |
Collapse
|
48
|
Yanai M, Sakamoto T, Nonaka T, Nakada T, Matsuoka S, Moriyama S, Yamamoto N, Teruya Y, Funaki Y, Harada T, Kinoshita N, Yamaguchi K, Kodani M, Yamasaki A. Pseudo-hyperprogression of Malignant Pleural Mesothelioma Treated with Nivolumab: A Case Report. Intern Med 2025:4807-24. [PMID: 39924236 DOI: 10.2169/internalmedicine.4807-24] [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] [Indexed: 02/11/2025] Open
Abstract
Currently, immune checkpoint inhibitors (ICIs) are the standard treatment for malignant pleural mesothelioma (MPM). The characteristic responses to ICI treatment include pseudoprogression (PP) and hyperprogressive disease (HPD), which require attention. Reports on PP of MPM are rare. We herein report a 59-year-old woman with MPM whose pleural lesions rapidly increased in size after initiating nivolumab treatment. This was initially judged as HPD but was subsequently diagnosed as PP owing to a marked reduction in tumor size. This case highlights the importance and difficulty of differentiating between PP and true progression, including HPD, during treatment of MPM with ICIs.
Collapse
Affiliation(s)
- Masaaki Yanai
- Division of Respiratory Medicine and Rheumatology, Tottori University Hospital, Japan
- Cancer Center, Tottori University Hospital, Japan
| | - Tomohiro Sakamoto
- Division of Respiratory Medicine and Rheumatology, Tottori University Hospital, Japan
| | - Takafumi Nonaka
- Division of Respiratory Medicine and Rheumatology, Tottori University Hospital, Japan
| | - Tatsuya Nakada
- Division of Respiratory Medicine and Rheumatology, Tottori University Hospital, Japan
| | - Shuichi Matsuoka
- Division of Respiratory Medicine and Rheumatology, Tottori University Hospital, Japan
| | - Shiro Moriyama
- Division of Respiratory Medicine and Rheumatology, Tottori University Hospital, Japan
| | - Natsumi Yamamoto
- Division of Respiratory Medicine and Rheumatology, Tottori University Hospital, Japan
| | - Yasuhiko Teruya
- Division of Respiratory Medicine and Rheumatology, Tottori University Hospital, Japan
| | - Yoshihiro Funaki
- Division of Respiratory Medicine and Rheumatology, Tottori University Hospital, Japan
| | - Tomoya Harada
- Division of Respiratory Medicine and Rheumatology, Tottori University Hospital, Japan
| | - Naoki Kinoshita
- Division of Respiratory Medicine and Rheumatology, Tottori University Hospital, Japan
| | - Kosuke Yamaguchi
- Division of Respiratory Medicine and Rheumatology, Tottori University Hospital, Japan
| | - Masahiro Kodani
- Division of Respiratory Medicine and Rheumatology, Tottori University Hospital, Japan
- Cancer Center, Tottori University Hospital, Japan
| | - Akira Yamasaki
- Division of Respiratory Medicine and Rheumatology, Tottori University Hospital, Japan
| |
Collapse
|
49
|
Azadinejad H, Farhadi Rad M, Shariftabrizi A, Rahmim A, Abdollahi H. Optimizing Cancer Treatment: Exploring the Role of AI in Radioimmunotherapy. Diagnostics (Basel) 2025; 15:397. [PMID: 39941326 PMCID: PMC11816985 DOI: 10.3390/diagnostics15030397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Radioimmunotherapy (RIT) is a novel cancer treatment that combines radiotherapy and immunotherapy to precisely target tumor antigens using monoclonal antibodies conjugated with radioactive isotopes. This approach offers personalized, systemic, and durable treatment, making it effective in cancers resistant to conventional therapies. Advances in artificial intelligence (AI) present opportunities to enhance RIT by improving precision, efficiency, and personalization. AI plays a critical role in patient selection, treatment planning, dosimetry, and response assessment, while also contributing to drug design and tumor classification. This review explores the integration of AI into RIT, emphasizing its potential to optimize the entire treatment process and advance personalized cancer care.
Collapse
Affiliation(s)
- Hossein Azadinejad
- Department of Immunology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah 6714869914, Iran;
| | - Mohammad Farhadi Rad
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah 6715847141, Iran
| | - Ahmad Shariftabrizi
- Department of Radiology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA;
| | - Arman Rahmim
- Department of Radiology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 0B4, Canada
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Hamid Abdollahi
- Department of Radiology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, BC V5Z 0B4, Canada
| |
Collapse
|
50
|
Altan M, Lopes G, Hiltermann TJN, Govindan R, Villaruz LC, Calvo E, Edelman MJ, Furqan M, Neal J, Felip E, Carlisle JW, Heymach JV, O’Cearbhaill RE, Zauderer M, Chisamore M, Corigliano E, Eleftheriadou I, Zajic S, Jenkins B, Goodison S, Suchindran S, Ramos-Hernandez N, Tarek N, Schoenfeld AJ. Safety and Tolerability of Letetresgene Autoleucel (GSK3377794): Pilot Studies in Patients with Advanced Non-Small Cell Lung Cancer. Clin Cancer Res 2025; 31:529-542. [PMID: 39576208 PMCID: PMC11788651 DOI: 10.1158/1078-0432.ccr-24-1591] [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: 05/24/2024] [Revised: 09/11/2024] [Accepted: 11/20/2024] [Indexed: 02/04/2025]
Abstract
PURPOSE The study aims to evaluate the safety, tolerability, and antitumor response of letetresgene autoleucel (lete-cel), genetically modified autologous T cells expressing a T-cell receptor specific for New York esophageal squamous cell carcinoma 1 (NY-ESO-1)/LAGE-1a shared epitope, alone or in combination with pembrolizumab, in HLA-A*02-positive (HLA-A*02:01, HLA-A*02:05, and/or HLA-A*02:06) patients with NY-ESO-1- and/or LAGE-1a-positive non-small cell lung cancer. PATIENTS AND METHODS Study 208749 was a single-arm study of lete-cel alone. Study 208471 was a multiarm study of lete-cel alone or in combination with pembrolizumab in patients with advanced or recurrent non-small cell lung cancer. RESULTS More than 2,500 patients were screened for target expression. In the multiarm study, 738 (45%) of 1,638 tested patients were HLA-A*02-positive. NY-ESO-1 and LAGE-1a testing was positive in 12% (62/525) and 4% (15/348) of tested patients, respectively. Forty-one patients positive for HLA-A*02 and antigen expression were screened in the single-arm study. Overall, 43 patients underwent leukapheresis and 18 received lete-cel across studies. Lete-cel demonstrated a manageable safety profile. No fatal treatment-related serious adverse events (AE) were reported in either study. Cytopenias and cytokine release syndrome were the most common treatment-emergent AEs. Combining pembrolizumab with lete-cel did not seem to increase toxicity over lete-cel alone. Limited antitumor activity was observed; one of 18 patients had a durable response persisting for 18 months. Pharmacokinetic data showed similar T-cell expansion in all patients. CONCLUSIONS Extensive HLA-A*02 and antigen expression testing was performed to identify potential participants. Lete-cel was generally well tolerated and had no unexpected AEs. Antitumor activity was observed in a limited number of patients.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/therapy
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/genetics
- Male
- Female
- Middle Aged
- Aged
- Pilot Projects
- Lung Neoplasms/therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/genetics
- Antibodies, Monoclonal, Humanized/administration & dosage
- Adult
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/genetics
- HLA-A2 Antigen/immunology
- HLA-A2 Antigen/genetics
- T-Lymphocytes/immunology
- Treatment Outcome
- Aged, 80 and over
- Cancer Vaccines/adverse effects
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Neoplasm Staging
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Membrane Proteins
Collapse
Affiliation(s)
- Mehmet Altan
- Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | | | | | - Ramaswamy Govindan
- Washington University School of Medicine in St. Louis, St. Louis, Missouri
| | | | - Emiliano Calvo
- START Madrid-CIOCC, Centro Integral Oncologico Clara Campal, Madrid, Spain
| | | | - Muhammad Furqan
- Carver College of Medicine, University of Iowa, Iowa City, Iowa
| | - Joel Neal
- Stanford Cancer Institute, Stanford University, Palo Alto, California
| | - Enriqueta Felip
- Vall d’Hebron University Hospital and Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | | | - John V. Heymach
- Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|