1
|
Kuusisalo S, Iivanainen S, Koivunen JP. Association of anti-PD-(L)1 treatment duration to efficacy in advanced solid tumors: a single center retrospective study. Ann Med 2025; 57:2476729. [PMID: 40091413 PMCID: PMC11915729 DOI: 10.1080/07853890.2025.2476729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 11/28/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are a standard of care in multiple cancers. Only a minority benefits, thus, optimal use and treatment duration remain indistinct. While biomarkers albeit PD-L1 are scarce, declined performance status and cancer-related systemic inflammation detected by blood inflammatory markers such as C-reactive protein (CRP) have been linked to inferior prognosis. MATERIALS AND METHODS We investigated the association of limited anti-PD-(L)1 treatment duration to therapy efficacy in melanoma and non-small cell lung cancer (NSCLC) patients who received therapy in a non-curative setting in Oulu University Hospital 2014-2022. Baseline prognostic factors (e.g. ECOG, CRP, and PD-L1 for NSCLC) were collected. Progression-free (PFS), overall (OS), and IO-free survival were analyzed using the Kaplan-Meier and Cox regression methods. RESULTS 126 patients (NSCLC, n = 72; melanoma, n = 54) were included. Majority (n = 101) were treated in the first line. Objective response rate was 34.9%. The median (m) anti-PD-(L)1 treatment duration was 3.42 months (mo). The mPFS and mOS were 6.8 mo (CI 95% 4.4-9.3) and 19.1 mo (CI 95% 13.3-24.9). Of the baseline factors, ECOG and CRP retained their significance in multivariate analysis for PFS (HR 0.34, CI 95% 0.19-0.59; HR 0.34, CI 95% 0.22-054) and OS (HR 0.38, CI 95% 0.20-0.71; HR 0.29, CI 95% 0.17-0.49). No difference was observed in PFS (HR 1.40, CI 95% 0.68-2.90) or OS (HR 0.69, CI 95% 0.29-1.65) according to treatment duration (3-6mo vs. > 6 mo). Long median IO-free survival (10.2 months; CI 95%, 4.1-16.3) was detected. CONCLUSION We characterized an anti-PD-(L)1 treated advanced NSCLC and melanoma cohort in which treatment benefit occurs irrespective of treatment duration and long-term benefit is observed off-treatment.
Collapse
Affiliation(s)
- Saara Kuusisalo
- Department of Medical Oncology and Radiotherapy and Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Sanna Iivanainen
- Department of Medical Oncology and Radiotherapy and Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Jussi P. Koivunen
- Department of Medical Oncology and Radiotherapy and Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| |
Collapse
|
2
|
Wurcel V, Rojas Rojas M, Urrego-Reyes J, Medrano Rivera D, Acevedo R, Jiang R, Jiang S, Zhang S, Caparros A, Krepler C, Fukunaga-Kalabis M, Younan ND, Alexander D, Hughes R, Weston G. Number needed to treat (NNT) with pembrolizumab as an adjuvant therapy in resected patients with high-risk stage II (IIB and IIC) melanoma and its application to cost of preventing an event (COPE) in Mexico. J Med Econ 2025; 28:346-353. [PMID: 40078077 DOI: 10.1080/13696998.2025.2466365] [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: 12/16/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025]
Abstract
INTRODUCTION Melanoma, responsible for most skin cancer deaths globally, has mortality rates expected to double by 2040. Pembrolizumab is a highly selective antibody approved for melanoma treatment and other cancers. Despite new treatments for melanoma, high treatment costs and long approval times limit patient access to new therapies. To support decision-making regarding metastatic melanoma therapies, a model was developed to calculate the number needed to treat (NNT) and the cost of preventing an event (COPE) using KEYNOTE-716 (NCT03553836) data. METHOD A cost-per-responder model comparing the clinical and economic impacts of pembrolizumab versus best supportive care (BSC) was developed considering a 52.8-month follow-up for recurrence-free survival (RFS) and distant metastasis-free survival (DMFS) in patients with resected high-risk melanoma. KEYNOTE-716 RFS and DMFS survival curves were used to calculate restricted mean survival time (RMST). The RMST was used to calculate NNT (NNTRMST). The NNTRMST calculates the NNT to result in a difference in mean survival time for a death or an event. NNTRMST is subsequently used to quantify COPE outcomes. RESULTS NNT for RFS was 5.3, reflecting the number of patients needed to treat to gain the additional difference observed in the mean RFS for resected high-risk type II (IIB and IIC) melanoma patients treated with pembrolizumab. For DMFS, the NNTRMST was 7.8. The estimated COPE to prevent an RFS or DMFS event was Mexican Peso (Mex $) 9,554,593 (2024) and Mex $13,961,427, respectively. CONCLUSIONS NNT values for RFS and DMFS data were both lower than the published average NNT value for current melanoma therapies. This demonstrated that fewer additional patients need to be treated in order to avoid a recurrence or a distant metastases event, compared to currently available melanoma therapies. The NNT and COPE highlight the clinical and economic impact of introducing pembrolizumab therapy for the treatment of patients in resected high-risk stage II melanoma.
Collapse
MESH Headings
- Humans
- Antibodies, Monoclonal, Humanized/economics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Melanoma/drug therapy
- Melanoma/surgery
- Melanoma/pathology
- Mexico
- Antineoplastic Agents, Immunological/economics
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/administration & dosage
- Skin Neoplasms/drug therapy
- Skin Neoplasms/pathology
- Skin Neoplasms/surgery
- Cost-Benefit Analysis
- Neoplasm Staging
- Chemotherapy, Adjuvant/economics
- Male
- Female
- Disease-Free Survival
- Quality-Adjusted Life Years
- Middle Aged
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shan Jiang
- Merck & Co., Inc, Rahway, New Jersey, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Wang Q, Sun N, Zhang C, Kunzke T, Zens P, Feuchtinger A, Berezowska S, Walch A. Metabolic heterogeneity in tumor cells impacts immunology in lung squamous cell carcinoma. Oncoimmunology 2025; 14:2457797. [PMID: 39924768 PMCID: PMC11812363 DOI: 10.1080/2162402x.2025.2457797] [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: 08/22/2024] [Revised: 12/22/2024] [Accepted: 01/20/2025] [Indexed: 02/11/2025] Open
Abstract
Metabolic processes are crucial in immune regulation, yet the impact of metabolic heterogeneity on immunological functions remains unclear. Integrating metabolomics into immunology allows the exploration of the interactions of multilayered features in the biological system and the molecular regulatory mechanism of these features. To elucidate such insight in lung squamous cell carcinoma (LUSC), we analyzed 106 LUSC tumor tissues. We performed high-resolution matrix-assisted laser desorption/ionization (MALDI) mass spectrometry imaging (MSI) to obtain spatial metabolic profiles, and immunohistochemistry to detect tumor-infiltrating T lymphocytes (TILs). Unsupervised k-means clustering and Simpson's diversity index were employed to assess metabolic heterogeneity, identifying five distinct metabolic tumor subpopulations. Our findings revealed that TILs are specifically associated with metabolite distributions, not randomly distributed. Integrating a validation cohort, we found that heterogeneity-correlated metabolites interact with CD8+ TIL-associated genes, affecting survival. High metabolic heterogeneity was linked to worse survival and lower TIL levels. Pathway enrichment analyses highlighted distinct metabolic pathways in each subpopulation and their potential responses to chemotherapy. This study uncovers the significant impact of metabolic heterogeneity on immune functions in LUSC, providing a foundation for tailoring therapeutic strategies.
Collapse
Affiliation(s)
- Qian Wang
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Na Sun
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Chaoyang Zhang
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Thomas Kunzke
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Philipp Zens
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Sabina Berezowska
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Axel Walch
- Research Unit Analytical Pathology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
4
|
Wang L, Chen SY, Li JL, Dai J, Qin DY, He RQ, Chen G. Anti-inflammatory effects of immunotherapy in clinical treatment and its potential mechanism in alleviating sleeping disorders: A systematic bibliometric study. Hum Vaccin Immunother 2025; 21:2475601. [PMID: 40097368 PMCID: PMC11917172 DOI: 10.1080/21645515.2025.2475601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/15/2025] [Accepted: 03/02/2025] [Indexed: 03/19/2025] Open
Abstract
Sleeping disorders negatively affect cancer patient management, quality of life, and recovery. Immunotherapy, a rising cancer treatment, shows potential to improve sleep quality by reducing inflammation. This study analyzed 255 publications (2000-2024) from the Web of Science Core Collection using bibliometric methods. The US and China dominate research output, with The Mayo Clinic as a key contributor. Core topics are "immunotherapy," "quality of life," and "antibodies." Emerging keywords like "cancer," "encephalitis," and "depression" highlight a shift toward clinical psychology in treating tumors and rare diseases. It is noteworthy that with the rapid expansion of immunotherapy in cancer treatment, clinical trials have shown that it can improve sleep quality in cancer patients by reducing inflammation. As its application in cancer treatment expands, immunotherapy's potential for treating sleep disorders is promising. Future development is expected to improve sleep quality and address clinical issues, offering broad prospects for patient outcomes.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P.R. China
| | - Si-Yan Chen
- Day Chemotherapy Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P.R. China
| | - Jun-Li Li
- Day Chemotherapy Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P.R. China
| | - Jian Dai
- Department of Clinical Psychology, Jiangbin Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Di-Yuan Qin
- Department of Computer Science and Technology, School of Computer and Electronic Information, Guangxi University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P. R. China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, P.R. China
| |
Collapse
|
5
|
Wang X, Yu YJ, Liao C, Liu XR, Yu R, Wang Y. Characterization of the gut microbiota in drug abuse: prediction, prevention, and personalized medicine to benefit affected populations. EPMA J 2025; 16:505-517. [PMID: 40438492 PMCID: PMC12106171 DOI: 10.1007/s13167-025-00402-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/03/2025] [Indexed: 06/01/2025]
Abstract
Drug abuse poses an enormous threat to global public health. Long-term drug abuse can reduce the quality of life of patients and increase the healthcare burden on society. There is growing interest in developing new methods to mitigate the effects of drug abuse. The gut microbiota plays a key role in maintaining homeostasis within the brain-gut-lung axis, which is critical in drug-abusing patients. The microbiota-brain-gut-lung axis refers to the interactions of microbes with the brain, gut, and lung. The effects of drug abuse on the gut microbiota are increasingly recognized, especially the pathogenesis by which the microbiota-brain-gut-lung axis is involved in regulating organ-organ communication, to explore new therapeutic approaches for clinical drug abuse. Currently, in addition to antibiotics, antiviral drugs, anti-tumor drugs, corticosteroids, drugs for the treatment of neurodegenerative diseases, and anesthetics also cause gut microbiota imbalance. This review summarizes the effects of drug abuse on gut microbiota and the important role of the microbiota-brain-gut-lung axis in drug abuse. Identifying changes in the gut microbiota associated with drug abuse and their underlying mechanisms under the principles of predictive, preventive, and personalized medicine (PPPM) is a critical step toward achieving PPPM. These strategies include FMT, probiotic supplements, and engineered bacteria that can benefit sub-healthy individuals with gut dysbiosis caused by drug abuse.
Collapse
Affiliation(s)
- Xin Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China
| | - Ya-Jie Yu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China
| | - Cai Liao
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China
| | - Xiao-Ru Liu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China
| | - Rui Yu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122 Liaoning China
| |
Collapse
|
6
|
Yu EY, Ferrario C, Linch MD, Stoeckle M, Laguerre B, Arranz JA, Todenhöfer T, Fong PC, Piulats JM, Berry W, Emmenegger U, Mourey L, Joshua AM, Mar N, Appleman LJ, Conter HJ, Gravis G, Li XT, Schloss C, Poehlein C, de Bono JS. Pembrolizumab plus Abiraterone Acetate and Prednisone in Patients with Chemotherapy-naïve Metastatic Castration-resistant Prostate Cancer: Results from KEYNOTE-365 Cohort D. Eur Urol Oncol 2025; 8:641-651. [PMID: 38926066 DOI: 10.1016/j.euo.2024.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/07/2024] [Accepted: 05/20/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND AND OBJECTIVE Abiraterone acetate (abiraterone) plus prednisone is approved for the treatment of metastatic castration-resistant prostate cancer (mCRPC). Our aim was to evaluate the efficacy and safety of pembrolizumab plus abiraterone in mCRPC. METHODS In cohort D of the phase 1b/2 KEYNOTE-365 study (NCT02861573), patients were chemotherapy-naïve, had disease progression ≤6 mo before screening, and had either not received prior next-generation hormonal agents for mCRPC or had received prior enzalutamide for mCRPC and had disease progression or became intolerant to enzalutamide. Patients received pembrolizumab 200 mg intravenously every 3 wk plus abiraterone 1000 mg orally once daily and prednisone 5 mg orally twice daily. The primary endpoints were safety, prostate-specific antigen (PSA) response rate, and objective response rate (ORR) according to Response Evaluation Criteria in Solid Tumors version 1.1 (RECIST v1.1) by blinded independent central review (BICR). Secondary endpoints included radiographic progression-free survival (rPFS) according to Prostate Cancer Clinical Trials Working Group 3-modified RECIST v1.1 by BICR and overall survival (OS). KEY FINDINGS AND LIMITATIONS For the 103 patients who were treated, median follow-up was 28 mo (interquartile range 26-31). The confirmed PSA response rate was 56% (58/103 patients). The ORR for patients with RECIST v1.1-measurable disease was 16% (6/37 patients). Median rPFS was 15 mo (95% confidence interval 9.2-22) and median OS was 30 mo (95% confidence interval 23-not reached); the estimated 24-mo OS rate was 58%. In total, 91% of patients experienced treatment-related adverse events, and 39% experienced grade 3-5 events. Grade 3/4 elevation of alanine aminotransferase (ALT) or aspartate aminotransferase (AST) was observed in 12% and 6.8% of patients, respectively. One patient died due to treatment-related myasthenic syndrome. Study limitations include the single-arm design. CONCLUSIONS Pembrolizumab plus abiraterone and prednisone demonstrated antitumor activity and acceptable safety in patients with chemotherapy-naïve mCRPC. Higher incidence of grade 3/4 elevated ALT/AST occurred than was reported for the individual agents. PATIENT SUMMARY For patients with metastatic castratation-resistant prostate cancer, the drug combination of pembrolizumab plus abiraterone and prednisone showed antitumor activity and acceptable safety.
Collapse
MESH Headings
- Humans
- Male
- Prostatic Neoplasms, Castration-Resistant/drug therapy
- Prostatic Neoplasms, Castration-Resistant/pathology
- Prednisone/therapeutic use
- Prednisone/administration & dosage
- Prednisone/pharmacology
- Aged
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/adverse effects
- Abiraterone Acetate/therapeutic use
- Abiraterone Acetate/administration & dosage
- Abiraterone Acetate/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Middle Aged
- Neoplasm Metastasis
- Cohort Studies
- Aged, 80 and over
Collapse
Affiliation(s)
- Evan Y Yu
- Fred Hutchinson Cancer Center and University of Washington, Seattle, WA, USA.
| | | | | | | | | | | | | | - Peter C Fong
- Auckland City Hospital and University of Auckland, Auckland, New Zealand
| | | | | | | | - Loic Mourey
- IUCT-Oncopole Claudius Regaud, Toulouse, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Zheng T, Li X, Zhou L, Jin J. Predictive value of machine learning for PD-L1 expression in NSCLC: a systematic review and meta-analysis. World J Surg Oncol 2025; 23:199. [PMID: 40405177 PMCID: PMC12101016 DOI: 10.1186/s12957-025-03847-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 05/11/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND As machine learning (ML) continuously develops in cancer diagnosis and treatment, some researchers have attempted to predict the expression of programmed death ligand-1 (PD-L1) in non-small cell lung cancer (NSCLC) by ML. However, there is a lack of systematic evidence on the effectiveness of ML. METHODS We conducted a thorough search across Embase, PubMed, the Cochrane Library, and Web of Science from inception to December 14th, 2023.A systematic review and meta-analysis was conducted to assess the value of ML for predicting PD-L1 expression in NSCLC. RESULTS Totally 30 studies with 12,898 NSCLC patients were included. The thresholds of PD-L1 expression level were < 1%, 1-49%, and ≥ 50%. In the validation set, in the binary classification for PD-L1 ≥ 1%, the pooled C-index was 0.646 (95%CI: 0.587-0.705), 0.799 (95%CI: 0.782-0.817), 0.806 (95%CI: 0.753-0.858), and 0.800 (95%CI: 0.717-0.883), respectively, for the clinical feature-, radiomics-, radiomics + clinical feature-, and pathomics-based ML models; in the binary classification for PD-L1 ≥ 50%, the pooled C-index was 0.649 (95%CI: 0.553-0.744), 0.771 (95%CI: 0.728-0.814), and 0.826 (95%CI: 0.783-0.869), respectively, for the clinical feature-, radiomics-, and radiomics + clinical feature-based ML models. CONCLUSIONS At present, radiomics- or pathomics-based ML methods are applied for the prediction of PD-L1 expression in NSCLC, which both achieve satisfactory accuracy. In particular, the radiomics-based ML method seems to have wider clinical applicability as a non-invasive diagnostic tool. Both radiomics and pathomics serve as processing methods for medical images. In the future, we expect to develop medical image-based DL methods for intelligently predicting PD-L1 expression.
Collapse
Affiliation(s)
- Ting Zheng
- Department of Medical Oncology, The First People's Hospital of Linping District, Hangzhou, 311100, Zhejiang Province, China.
| | - Xingxing Li
- Department of Medical Oncology, The First People's Hospital of Linping District, Hangzhou, 311100, Zhejiang Province, China
| | - Li Zhou
- Department of Medical Oncology, The First People's Hospital of Linping District, Hangzhou, 311100, Zhejiang Province, China
| | - Jianjiang Jin
- Department of Medical Oncology, The First People's Hospital of Linping District, Hangzhou, 311100, Zhejiang Province, China
| |
Collapse
|
8
|
Park J, Chen YY, Cao JJ, An J, Chiu Yen RW, Outen JD, Baylin SB, Topper MJ. MYC plus class IIa HDAC inhibition drives mitochondrial dysfunction in non-small cell lung cancer. Cell Rep 2025; 44:115722. [PMID: 40392656 DOI: 10.1016/j.celrep.2025.115722] [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: 11/14/2024] [Revised: 03/11/2025] [Accepted: 04/29/2025] [Indexed: 05/22/2025] Open
Abstract
Despite much progress in targeting the MYC oncoprotein, combination treatment strategies are needed to exploit this molecular vulnerability. To this end, we interrogated transcriptome data from cancer cell lines treated with MYC inhibitors and identified HDAC5 and HDAC9, both class IIa histone deacetylases (HDACs), as potential therapeutic targets. Notably, these therapeutically actionable HDAC isoforms are known augmenters of several hallmarks of cancer. Dual targeting of MYC and class IIa HDACs induces a significant reduction in viability for non-small cell lung cancer (NSCLC) cell lines with high MYC and mitochondrial activity. Additionally, combination treatment induces a robust MYC suppression with mitochondrial reactive oxygen species (ROS) elevation, which has a causal relationship with therapeutic efficacy. Confirmation of in vivo efficacy was pursued in several animal models, with subsequent molecular-correlate derivation confirming the importance of MYC depletion and mitochondrial dysfunction in drug efficacy. Ultimately, we define a therapeutic approach combining MYC- and class IIa HDAC-inhibition to potentiate anti-tumor efficacy in NSCLC.
Collapse
Affiliation(s)
- Jina Park
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ying-Yu Chen
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jennie J Cao
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julia An
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ray-Whay Chiu Yen
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Stephen B Baylin
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Michael J Topper
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
9
|
Keshavarz Sadegh R, Saleki K, Rezaei N. Immune checkpoint inhibitor (ICI) therapy in central nervous system cancers: State-of-the-art and future outlook. Int Immunopharmacol 2025; 159:114837. [PMID: 40394797 DOI: 10.1016/j.intimp.2025.114837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 04/28/2025] [Accepted: 05/07/2025] [Indexed: 05/22/2025]
Abstract
Invasive central nervous system (CNS) cancers are an area where the development of breakthrough therapies is urgently needed. For instance, conditions such as glioblastoma multiforme (GBM) are associated with poor clinical prognosis, with the majority of trials offering no improvement to marginally enhanced survival. Unleashing the potential of targeting the immune system in CNS cancers has gained attention in recent years. Inhibition of immune checkpoints such as CTLA-4, PD-1/PD-L1, TIM-3, and LAG-3 has been attempted in recent trials. While potentially offering a notable edge over other immunotherapies, multi-organ adverse events have been found with the administration of immune checkpoint inhibitors (ICIs). The present review captures the state-of-the-art evidence on ICI treatments in different CNS cancers. Also, we discuss the value of combinational therapies involving ICIs as well as next-generation therapeutics such as bispecific antibodies targeting PD-1/LAG-3/TIM-3 and CRISPR-Cas9-edited PD-1-knock-out checkpoint-resistant CAR T-cells.
Collapse
Affiliation(s)
- Roghaye Keshavarz Sadegh
- Student Research Committee, Qazvin University of Medical Sciences, Qazvin, Iran; USERN Office, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Kiarash Saleki
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; USERN MUBabol Office, Universal Scientific Education and Research Network (USERN), Babol, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Zhang W, Xiong J, Li Y, Nie J, Zhao W, Guo Z, Liu X, Zhang Q, Chen X, Ye L, Chen Z, Wang H, Xu K, Zhao L, Liu Y, Huang L, Li Y, He Y. Efficacy and safety of distinct regimens for individuals with advanced EGFR-mutated non-small-cell lung cancer who progressed on EGFR tyrosine-kinase inhibitors: a systematic review and network meta-analysis. Ther Adv Med Oncol 2025; 17:17588359251338046. [PMID: 40396122 PMCID: PMC12089717 DOI: 10.1177/17588359251338046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 04/08/2025] [Indexed: 05/22/2025] Open
Abstract
Background Targeted therapy with EGFR tyrosine-kinase inhibitors (TKIs) is the preferred first-line treatment for EGFR-mutated advanced non-small-cell lung cancer (NSCLC), but acquired resistance inevitably occurs in almost all responding individuals. Objectives We aimed to comprehensively review the literature to investigate the efficacy and safety of distinct regimens in the subsequent-line setting, thereby identifying the optimal regimen for these TKI-resistant NSCLC patients. Design A systematic review and network meta-analysis (NMA) using a Bayesian framework. Data sources and methods The PubMed, Embase, Cochrane Library databases, and abstracts of ASCO, ESMO, and WCLC were searched from database inception to November 3, 2024, to identify eligible randomized controlled trials (RCTs) that assessed distinct regimens for individuals with advanced EGFR-mutated NSCLC who progressed on TKIs. The outcomes of progression-free survival (PFS), overall survival (OS), objective response rate (ORR), disease control rate (DCR), and grade 3 or higher adverse events (⩾3AEs) were compared and ranked in overall patients and various subgroups among eight regimens by NMA and the surface under the cumulative ranking curve, respectively. The protocol is registered with PROSPERO, CRD42024601619. Results In total, 14 RCTs, involving 3177 participants and 8 treatment regimens (chemotherapy plus ivonescimab (programmed cell death protein 1/vascular endothelial growth factor inhibitor; chemotherapy + ivonescimab (CT + IVO)); CT + amivantamab + lazertinib (CT + AMI + LAZ), CT + immunotherapy + bevacizumab (CT + IO + BEV), CT + AMI, CT + BEV, CT + IO, CT, and IO), were included. Overall, in patients, the most pronounced PFS benefit was observed with the "CT + IVO," followed by "CT + AMI + LAZ," "CT + IO + BEV," and "CT + AMI," ranked second, third, and fourth, respectively. In terms of OS, the regimen of "CT + AMI" ranked the best, followed by "CT + IVO." However, the comparisons of OS among different regimens did not reach statistical significance, possibly due to immature data. The results for ORR and DCR were similar to those for OS, with "CT + AMI" topping the rankings, followed by "CT + AMI + LAZ." In terms of safety, the incidence of ⩾3AEs was highest in "CT + AMI + LAZ," followed by "CT + AMI." In subgroup analysis, "CT + IVO" demonstrates stable PFS benefits across clinicopathological characteristics, ranking first in most subgroups. Due to the unavailability of OS subgroup data in most RCTs, many regimens were missing in the OS subgroup analysis. Conclusion Integrating the results of different clinical outcomes and subgroup analyses, we conclude that "CT + IVO" is the optimal treatment option with an acceptable safety profile for patients with advanced EGFR-mutated NSCLC who have progressed on TKIs. "CT + AMI + LAZ" and "CT + AMI" are alternative subsequent line options as well, with superior efficacy compared to immunotherapy-based or chemotherapy regimens, yet elevated toxicity profiles requiring vigilant management.
Collapse
Affiliation(s)
- Wengang Zhang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jian Xiong
- Department of Pulmonary and Critical Care Medicine, Yueyang Central Hospital, Yueyang, China
| | - Yujie Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jing Nie
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wencheng Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhiyi Guo
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinyue Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qianqian Zhang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuyang Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Li Ye
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhimin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Hao Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Kandi Xu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lishu Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yujin Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lihua Huang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yuhang Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| |
Collapse
|
11
|
Kejamurthy P, Mk J, Kt RD. A novel anti-PD-L1 DNA aptamer, Apta35 enhances non-small cell lung cancer cell cytotoxicity and apoptosis through lung cancer-activated T lymphocytes. Int Immunopharmacol 2025; 155:114621. [PMID: 40209314 DOI: 10.1016/j.intimp.2025.114621] [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: 07/10/2024] [Revised: 01/31/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
The prevalence of Programmed death ligand 1 (PD-L1) expression in the population of NSCLC patients and blocking the PD1/PD-L1 pathway by inhibiting the PD-1 receptor on immune cells or the PD-L1 ligand on tumour and/or immune cells can inhibit tumour growth. EFBALite algorithm that enables efficient and cost-effective selection of aptamers, expediting the process. Here, we present the development, computational validation, and in vitro analysis of NSCLC of DNA aptamers targeting PD-L1. The Gibbs free energy of two anti-PD-L1 aptamers, Apta35 and Apta90 with -3.06 and - 2.4 kcal/mol respectively. The docking score for Apta35 was -272.3 and 1171.765 for HDOCK and ZDOCK respectively. Further, the Apta35 was taken for the in vitro studies as it was more stable and incubated with NCI-H460. Initially, we confirmed the binding of the TAMRA-labelled Apta35 to the NCI-H460 cell surface through microscopic imaging and further confirmed through FACS analysis. Further experimental results showed that the Apta35 treated along with the act-T cells group reduced the percentage of viability (28 ± 3.5), increased toxicity, and reduced count of NCI-H460 cells when compared with the cells treated only with the act-T cells concerning the treatment to 50 nM concentration. In summary, targeting PD-L1 with a specific aptamer provides an innovative strategy for targeting NSCLC. Apta35 aptamer showed no significant toxicity in the BALB/c nude mice while it was injected every 2 days for a total of 12 days of treatment.
Collapse
Affiliation(s)
- Priyatharcini Kejamurthy
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Jaganathan Mk
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Ramya Devi Kt
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India.
| |
Collapse
|
12
|
Ambrosini-Spaltro A, Rengucci C, Capelli L, Chiadini E, Bennati C, Delmonte A, Vecchiarelli S, Limarzi F, Nosseir S, Gallo G, Valli M, Ulivi P, Calistri D. Clinicopathological Features of Non-Small Cell Lung Carcinoma with NRAS Mutation. J Pers Med 2025; 15:199. [PMID: 40423070 DOI: 10.3390/jpm15050199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 04/24/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
(1) Background: NRAS mutations affect fewer than 1% of lung adenocarcinomas. The aim of this study was to describe the clinicopathological features of lung carcinomas with NRAS mutations. (2) Methods: A series of NRAS-mutated lung carcinomas was collected from a molecular diagnostic unit (from four hospitals). The cases were analyzed with next-generation sequencing. A log-rank test for overall survival (OS) was calculated. (3) Results: NRAS mutations were detected in 14/1948 samples (0.72%) of non-small-cell lung carcinomas from 13 patients (8 males, 5 females). NRAS mutations involved codon 61 in the majority (9/13, 69.2%) of cases. The other NRAS mutations affected codon 12 (2/13, 15.4%), codon 13 (1/13, 7.7%), and codon 142 (1/13, 7.7%). In 7/13 cases, co-alterations in additional genes were also present. Pleomorphic/sarcomatoid features were identified in 3/13 (23.1%) cases, in 2/8 (25.0%) histological specimens, and in 2/5 (40.0%) surgical specimens, respectively. Follow-up data were available in 11/13 cases, with 6 patients deceased. By a log-rank test, patients with NRAS mutations in codon 61 had a better outcome (estimated mean of 32.6 ± 7.1 months) compared to those with other NRAS mutations (estimated mean of 8.7 ± 4.4 months), with a significant difference (p = 0.048 for OS). (4) Conclusions: Lung carcinomas with NRAS mutation may display pleomorphic or sarcomatoid features. Mutations in codon 61 showed a more favorable prognosis than those in other codons.
Collapse
Affiliation(s)
| | - Claudia Rengucci
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola, Italy
| | - Laura Capelli
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola, Italy
| | - Elisa Chiadini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola, Italy
| | - Chiara Bennati
- Oncology Unit, Santa Maria Delle Croci Hospital, AUSL Romagna, 48121 Ravenna, Italy
| | - Angelo Delmonte
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola, Italy
| | | | - Francesco Limarzi
- Pathology Unit, Morgani-Pierantoni Hospital, AUSL Romagna, 47121 Forlì, Italy
| | - Sofia Nosseir
- Pathology Unit, Santa Maria Delle Croci Hospital, AUSL Romagna, 48121 Ravenna, Italy
| | - Graziana Gallo
- Pathology Unit, Bufalini Hospital, AUSL Romagna, 47521 Cesena, Italy
| | - Mirca Valli
- Pathology Unit, Infermi Hospital, AUSL Romagna, 47923 Rimini, Italy
| | - Paola Ulivi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola, Italy
| | - Daniele Calistri
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", 47014 Meldola, Italy
| |
Collapse
|
13
|
Biney F, Giroux-Leprieur É, Daniel C, Du Rusquec P, Auliac JB, Assié JB, Anane-Abrous S, Chouaid C. [Influence of smoking on the efficacy of immunotherapy in advanced lung cancers]. Rev Mal Respir 2025:S0761-8425(25)00182-2. [PMID: 40374498 DOI: 10.1016/j.rmr.2025.04.003] [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: 09/01/2024] [Accepted: 04/21/2025] [Indexed: 05/17/2025]
Abstract
INTRODUCTION Lung cancer is the leading cause of death worldwide. It occurs mainly in smokers, but also in 25% of cases in non-smokers. As regards metastatic stages, while immunotherapy has led to improved overall survival, smoking status may potentially influence its effectiveness. The objective of this study was to analyze its effectiveness as first-line treatment for advanced non-small cell lung cancers (NSCLC) according to patient smoking status. METHOD This retrospective study covers all patients with stage IV NSCLC treated with first- line immunotherapy, alone or in combination, between January 2018 and 2019, in three Ile de France centers. The primary and secondary endpoints were, respectively, overall survival and progression-free survival, according to smoking status. RESULTS The analysis included 105 patients (66.7% men) of whom 38% were active smokers and 9.5% non-smokers, with adenocarcinoma in 65.7% of cases. Median OS and PFS were 17, 23.2, and 24.9 months and 4.9, 7.6, and 4 months for smokers, ex-smokers, and non-smokers respectively, without significant differences. CONCLUSION In this study, smoking status did not seem to modify the effectiveness of immunotherapy. Studies on a larger population are called for.
Collapse
Affiliation(s)
- F Biney
- Service de pneumologie, CHSF, Corbeil Essonne, France.
| | - É Giroux-Leprieur
- Service de pneumologie et oncologie thoracique, université Paris-Saclay, UVSQ, EA 4340 BECCOH, AP-HP-hôpital Ambroise-Paré, Boulogne-Billancourt, France
| | | | | | - J B Auliac
- Service de pneumologie, hôpital Intercommunal, Créteil, France
| | - J B Assié
- Service de pneumologie, hôpital Intercommunal, Créteil, France; Centre de recherche des Cordeliers, Inserm, Sorbonne université, université Paris Cité, Functional Genomics of Solid Tumors Laboratory, Paris, France
| | - S Anane-Abrous
- Service de radiothérapie, hôpital Intercommunal, Créteil, France
| | - C Chouaid
- Service de pneumologie, hôpital Intercommunal, Créteil, France
| |
Collapse
|
14
|
Kuang W, Zeng J, Tong L, Liu Q, Sun H, Feng M, Liang D, Wang W, Wang C. Frequency of microsatellite instability in gynecologic cancers and the efficacy of immune checkpoint inhibitors treated: real-world data from a single gynecologic center. Front Immunol 2025; 16:1567824. [PMID: 40416974 PMCID: PMC12098365 DOI: 10.3389/fimmu.2025.1567824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/15/2025] [Indexed: 05/27/2025] Open
Abstract
Objective This study evaluated the incidence of Microsatellite Instability-High (MSI-H) in patients with gynecologic cancers in a single gynecologic center and investigated the effect of immune checkpoint inhibitors (ICIs) in treating MSI-H in advanced or recurrent gynecologic cancers. Methods We conducted a retrospective study of patients diagnosed with gynecological cancers between June 2021 and May 2024. We investigated their clinicopathological information, the results of microsatellite instability (MSI), the immunohistochemistry staining PD-L1 analyses, the molecular classification testing, and the tumor response to treatment with ICIs. Results Among 1333 patients included in the analysis, the frequency of MSI-H was 1.3% (3/223) in cervical cancer, 25.7% (280/1091) in endometrial cancer, and 10.5% (2/19) in ovarian or tubal and peritoneal cancer. When the patients were evaluated by histologic type, the frequency of MSI-H was 26.1% (241/921) in endometrioid adenocarcinoma and 35.1% (20/57) in mixed adenocarcinoma. Molecular classification results for the 1020 cases that successfully underwent the tests were 71 for the POLE mutation (POLEmut) subtype, 271 for MMR-deficiency (MMRd) subtype, 571 for the non-specific molecular profile (NSMP) subtype, and 107 for the p53 abnormality (p53abn) subtype. Thirty-five patients were treated with ICIs for at least one cycle. The objective response rate (ORR) was 34.3% (95% CI, 19.1% to 52.2%). Among the patients who achieved an objective response, the median time to respond was 2.65 months, and the median duration of response had not been reached. The median progression-free survival (PFS) was 9 months (95% CI, 4 to 10), and the median overall survival (OS) had not been reached. Additionally, in the patients with endometrial cancer, the median PFS in MSI-H patients was 5 months versus 3 months in microsatellite stable (MSS) patients (Δ = 2 months; p=0.92), and the median OS in both MSI-H and MSS patients had not been reached (p=0.89). Conclusion This study had shown the MSI-H frequencies for the three major types of gynecological tumors and demonstrated the clinical benefit of treatment with ICIs in patients with advanced or recurrent gynecologic cancer. Among endometrial cancer patients, the effects of immunotherapy may be consistent regardless of MSI status.
Collapse
Affiliation(s)
- Wei Kuang
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| | - Jing Zeng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
- Department of Obstetrics and Gynecology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Lingling Tong
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| | - Qianqi Liu
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| | - Huanxin Sun
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| | - Min Feng
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| | - Dongni Liang
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| | - Wei Wang
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| | - Cheng Wang
- Department of Pathology, West China Second University Hospital of Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
15
|
Goss G, Ciuleanu T, Ramlau R, Renouf DJ, Chu Q, Kalinka E, Sawrycki P, Bramson J, Nelson BH, Crabbé R, LaCasse E, Lo B, Sahlender DA, Crompton P, Brichory F, Piggott L, Schenker M, Juergens R. Xevinapant plus avelumab in advanced solid tumours, with a dose expansion in advanced non-small-cell lung cancer: exploratory biomarker, safety and efficacy analyses from an open-label, nonrandomised phase Ib study. Ther Adv Med Oncol 2025; 17:17588359251332154. [PMID: 40351326 PMCID: PMC12062605 DOI: 10.1177/17588359251332154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 03/18/2025] [Indexed: 05/14/2025] Open
Abstract
Background Xevinapant, an inhibitor of apoptosis protein (IAP) inhibitor, has shown promising activity in combination with anticancer agents, including radiotherapy, and, in preclinical studies, anti-PD-(L)1 antibodies. This, in part, is due to its ability to restore apoptosis and increase antitumour immunity. Objectives We report efficacy, safety and exploratory biomarker analyses of xevinapant plus avelumab (anti-PD-L1) in a two-part, open-label, nonrandomised, phase Ib study. Design Part A assessed patients with advanced solid tumours who received xevinapant (100, 150, 200 or 250 mg/day, with no random allocation, on Days 1-10 and 15-24) in combination with avelumab (10 mg/kg) on Days 1 and 15 in 28-day cycle. Part B assessed patients with advanced non-small-cell lung cancer (NSCLC) who received xevinapant at the recommended phase II dose (RP2D) plus avelumab (maximum 26 cycles). Methods Part A assessed the safety and tolerability of the combination and established the maximum tolerated dose (MTD) and RP2D of xevinapant. Part B assessed the antitumour activity of xevinapant at the RP2D combined with avelumab compared with a historical control (avelumab alone). Exploratory biomarker analyses were also conducted. Results In part A (n = 16), xevinapant 200 mg/day was established as the RP2D with avelumab and the MTD was not reached. The most common treatment-emergent adverse events (TEAEs) irrespective of xevinapant dose were nausea and fatigue (n = 11 (68.8%) each). In part B (n = 38; four patients received prior anti-PD-(L)1 antibody), the objective response rate (ORR) was 10.5% (95% confidence interval (CI), 2.9-24.8; partial response, n = 4) and the most common TEAE was decreased appetite (n = 13 (34.2%)). Levels of plasma IL-10, IL-1β, IL-13 and CD8+ T cells increased during the study, and circulating levels of CD4+ T cells and Tregs increased during cycle 1. Macrophage-related gene expression signatures increased in patients with a partial response or stable disease. Low baseline Ki-67 expression in tumour samples correlated with a partial response. Conclusion The RP2D of xevinapant with avelumab was established; however, the ORR was not superior to the historical control (avelumab alone). The combination had a manageable safety profile in both study parts. Biomarker analyses provide insights into drivers associated with efficacy in patients with NSCLC receiving xevinapant plus avelumab. Trial registration NCT03270176 (https://clinicaltrials.gov/study/NCT03270176). Registered on ClinicalTrials.gov on 29 August 2017.
Collapse
Affiliation(s)
- Glenwood Goss
- Department of Medicine, University of Ottawa, and the Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, ON K1H8L6, Canada
| | - Tudor Ciuleanu
- Department of Oncology, ‘Iuliu Haţieganu’ University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Rodryg Ramlau
- Oncology Department, Poznan University of Medical Sciences, Poznan, Poland
| | - Daniel J. Renouf
- Department of Medicine, Faculty of Medicine, University of British Columbia, BC Cancer, Vancouver, BC, Canada
| | - Quincy Chu
- Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, AB, Canada
| | - Ewa Kalinka
- Department of Oncology, Polish Mother’s Memorial Hospital, Lodz, Poland
| | - Piotr Sawrycki
- Wojewódzki Szpital Zespolony im. L. Rydygiera w Toruniu, Torun, Poland
| | - Jonathan Bramson
- Centre for Discovery in Cancer Research and Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Brad H. Nelson
- Deeley Research Centre, BC Cancer – Victoria, Victoria, BC, Canada
| | | | | | - Bryan Lo
- Division of Anatomical Pathology, The Ottawa Hospital, Ottawa, ON, Canada
| | | | | | | | - Luke Piggott
- Debiopharm International SA, Lausanne, Switzerland
| | - Michael Schenker
- Oncology Center Sf Nectarie, Craiova, Romania
- Medical Oncology, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | - Rosalyn Juergens
- Department of Oncology, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
16
|
Li A, Luo M, Liu X, Wu H, Liu X, Zhang Z, Zhang X. Toll-like receptor 3 activation enhances antitumor immune response in lung adenocarcinoma through NF-κB signaling pathway. Front Immunol 2025; 16:1585747. [PMID: 40406122 PMCID: PMC12095255 DOI: 10.3389/fimmu.2025.1585747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Accepted: 04/21/2025] [Indexed: 05/26/2025] Open
Abstract
Background Toll-like receptor 3 (TLR3) is a pattern recognition receptor known to play a crucial role in the immune response to cancer. However, its effect on the efficacy of immunotherapy in lung adenocarcinoma (LUAD) remains unclear. This study aims to investigate the role of TLR3 in LUAD by examining its expression levels, prognostic significance, and impact on immune signaling pathways. Methods We analyzed the impact of TLR3 expression on the prognosis of lung adenocarcinoma patients using data from the Cancer Genome Atlas (TCGA) database and four additional cohorts (GSE72094, GSE30219, GSE50081 and GSE31210). Functional enrichment analyses were performed to compare molecular features between low and high TLR3 expression groups using gene set variation analysis (GSVA). We also examined the correlation between TLR3 and tumor mutation burden (TMB), immune infiltration, and PD-L1 expression. Further experimental validation was conducted using co-culture systems of LUAD cells and peripheral blood mononuclear cells (PBMCs) with PD1 inhibitors, and Western blot analysis to investigate the involvement of NF-κB signaling. Results TLR3 expression was significantly lower in LUAD tissues compared to normal tissues, with high TLR3 expression correlating with better survival outcomes across multiple cohorts. High TLR3 expression was associated with increased TMB and enhanced immune activation. Patients with high TLR3 expression exhibited higher immune checkpoint expression and immune cell infiltration. Experimental results showed that TLR3 agonists increased the susceptibility of LUAD cells to activated PBMCs under PD1 inhibitor therapy, inhibiting cell proliferation, migration, and invasion. Additionally, TLR3 has a strong positive correlation with MHC molecules and upregulated PD-L1 expression. NF-κB was identified as a key regulator of PD-L1 expression, with TLR3 agonists enhancing NF-κB and PD-L1 activity. Conclusion TLR3 enhances the anti-tumor immune response in LUAD by modulating NF-κB signaling and PD-L1 expression, making it a promising prognostic biomarker and therapeutic target. This study highlights the potential of TLR3 to improve immunotherapy outcomes, providing a comprehensive analysis of its role in LUAD and paving the way for novel therapeutic strategies targeting TLR3-mediated pathways.
Collapse
Affiliation(s)
- Ang Li
- School of Public Health, North China University of Science and Technology, Tangshan, China
- College of Life Science, North China University of Science and Technology, Tangshan, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, North China University of Science and Technology, Tangshan, China
| | - Man Luo
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Xiyao Liu
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Hongjiao Wu
- School of Public Health, North China University of Science and Technology, Tangshan, China
- College of Life Science, North China University of Science and Technology, Tangshan, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, North China University of Science and Technology, Tangshan, China
| | - Xiaoguang Liu
- College of Life Science, North China University of Science and Technology, Tangshan, China
| | - Zhi Zhang
- Affliated Tangshan Gongren Hospital, North China University of Science and Technology, Tangshan, China
| | - Xuemei Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, China
- College of Life Science, North China University of Science and Technology, Tangshan, China
- Hebei Key Laboratory of Occupational Health and Safety for Coal Industry, North China University of Science and Technology, Tangshan, China
| |
Collapse
|
17
|
Canali B, Apolone G, Ascierto PA, De Braud F, Grossi F, Perrone F, Fiorentino F, Di Costanzo A, Candelora L, Patanè G, Zapparelli G, Mezzanotte C, Didoni G, Riccaboni M. Effect of immuno-oncology on clinical and economic outcomes for a selection of cancers in Italy. Expert Rev Pharmacoecon Outcomes Res 2025:1-11. [PMID: 40329477 DOI: 10.1080/14737167.2025.2493130] [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: 10/09/2024] [Revised: 04/03/2025] [Accepted: 04/08/2025] [Indexed: 05/08/2025]
Abstract
OBJECTIVES This study assesses the impact of immune-oncology (IO) drugs' availability on cancer incidence-adjusted mortality rates from melanoma, lung, and renal cancers at population level in Italy between 2008 and 2019. METHODS We conducted a retrospective study on cross-sectional time-series aggregated data collected from publicly available sources and IQVIA proprietary databases. Three fixed-effects regression models were used to estimate how IO availability affects incidence-adjusted mortality for each cancer type. Estimated deaths were compared with deaths in a scenario with no IO drugs availability. Finally, the number of averted deaths was valued using the human capital approach. RESULTS A 1% increase in IO availability reduces incidence-adjusted mortality rates for melanoma, lung, and renal cancers by 0.125% (95% CI: 0.138-0.112; p < 0.01), 0.011% (95% CI: 0.013-0.009; p < 0.01) and 0.005% (95% CI: 0.006-0.003; p < 0.01) between the introduction of the drug in the therapeutic area and 2019. This reduction resulted in total savings of € 49.0 million, € 61.3 million, and € 10.9 million in indirect costs due to premature mortality, respectively. CONCLUSIONS IO drugs introduction in Italy between 2008 and 2019 was associated with a significant decrease in deaths from each cancer and, consequently, in savings in indirect costs related to premature mortality.
Collapse
Affiliation(s)
- Beatrice Canali
- Real World Solutions, IQVIA Solutions Italy S.r.l, Milan, Italy
| | - Giovanni Apolone
- Scientific Directorate, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Filippo De Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesco Grossi
- Department of Medicine and Technological Innovation, Università degli Studi dell'Insubria, Varese, Italy
- Medical Oncology Division, ASST Sette Laghi, Varese, Italy
| | - Francesco Perrone
- Clinical Trial Unit, Istituto Nazionale Tumori, IRCCS, Fondazione G. Pascale, Naples, Italy
| | | | | | - Laura Candelora
- Real World Solutions, IQVIA Solutions Italy S.r.l, Milan, Italy
| | | | | | - Claudia Mezzanotte
- Pricing, Access Strategy & Health Economics, Bristol Myers Squibb, Rome, Italy
| | - Guido Didoni
- Pricing, Access Strategy & Health Economics, Bristol Myers Squibb, Rome, Italy
| | | |
Collapse
|
18
|
Zhang J, Song Z, Zhang Y, Zhang C, Xue Q, Zhang G, Tan F. Recent advances in biomarkers for predicting the efficacy of immunotherapy in non-small cell lung cancer. Front Immunol 2025; 16:1554871. [PMID: 40406096 PMCID: PMC12095235 DOI: 10.3389/fimmu.2025.1554871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 04/18/2025] [Indexed: 05/26/2025] Open
Abstract
Lung cancer continues to be the primary cause of cancer-related deaths globally, with non-small cell lung cancer (NSCLC) accounting for approximately 85% of all instances. Recently, immune checkpoint inhibitors (ICIs) have transformed the treatment approach for NSCLC, however, only a subset of patients experiences significant benefits. Therefore, identifying reliable biomarkers to forecast the efficacy of ICIs is crucial for ensuring the safety and effectiveness of treatments, becoming a major focus of current research efforts. This review highlights the recent advances in predictive biomarkers for the efficacy of ICIs in the treatment of NSCLC, including PD-L1 expression, tertiary lymphoid structures (TLS), tumor-infiltrating lymphocytes (TILs), tumor genomic alterations, transcriptional signatures, circulating biomarkers, and the microbiome. Furthermore, it underscores the pivotal roles of liquid biopsy, sequencing technologies, and digital pathology in biomarker discovery. Special attention is given to the predictive value of TLS, circulating biomarkers, and transcriptional signatures. The review concludes that the integration of multiple biomarkers holds promise for achieving more accurate efficacy predictions and optimizing personalized immunotherapy strategies. By providing a comprehensive overview of the current progress, this review offers valuable insights into biomarker-based precision medicine for NSCLC and outlines future research directions.
Collapse
Affiliation(s)
- Jiacheng Zhang
- 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, China
| | - Zehao Song
- 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, China
| | - Yuanjie Zhang
- 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, China
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chentong Zhang
- 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, China
| | - Qi Xue
- 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, China
| | - Guochao Zhang
- 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, China
| | - Fengwei Tan
- 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, China
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
19
|
Elder AM, Fairchild HR, Kines KT, Cozzens LM, Becks AR, Slansky JE, Anderson SM, Lyons TR. Semaphorin7A and PD-L1 cooperatively drive immunosuppression during mammary involution and breast cancer. Cell Rep 2025; 44:115676. [PMID: 40333186 DOI: 10.1016/j.celrep.2025.115676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 02/06/2025] [Accepted: 04/16/2025] [Indexed: 05/09/2025] Open
Abstract
Postpartum mammary gland remodeling after a pregnancy/lactation cycle is characterized by mechanisms of cell death and inflammation. Here, we show that SEMA7A promotes PD-L1 expression in immune cells of the mammary tissue during involution. These same phenotypes are mimicked in the microenvironment of SEMA7A-expressing tumors, which partially respond to αPD-1/αPD-L1 treatments in vivo. However, cells that remain after treatment are enriched for SEMA7A expression. Therefore, we tested a monoclonal antibody that directly targets SEMA7A-expressing tumors, in part, by reducing SEMA7A-mediated upregulation of PD-L1. In vivo, the SEMA7A monoclonal antibody reduces tumor growth and/or promotes complete regression of mouse mammary tumors, reduces some immunosuppressive phenotypes in the tumor microenvironment, and restores cytotoxic T cells, suggesting that SEMA7A may be a candidate for immune-based therapy for breast cancer patients.
Collapse
Affiliation(s)
- Alan M Elder
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Cancer Biology Graduate Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Heather R Fairchild
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kelsey T Kines
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Cancer Biology Graduate Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Lauren M Cozzens
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Cancer Biology Graduate Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alexandria R Becks
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; Cancer Biology Graduate Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jill E Slansky
- University of Colorado Cancer Center, Aurora, CO, USA; Department of Immunology and Microbiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | | | - Traci R Lyons
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA; University of Colorado Cancer Center, Aurora, CO, USA; Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
20
|
Saito S, Kawashima Y, Tanaka H, Yoshimura N, Tsukita Y, Saito R, Nakagawa T, Inomata M, Nagashima H, Sugawara S. Prospective Multi-Institutional Observational Study of Retreatment with Anti-PD-1/PD-L1 Antibodies in Patients with Non-Small Cell Lung Cancer Previously Treated with Anti-PD-1/PD-L1 Plus Chemotherapy: NJLCG (North Japan Lung Cancer Group) Trial 1901. Cancers (Basel) 2025; 17:1551. [PMID: 40361477 PMCID: PMC12071100 DOI: 10.3390/cancers17091551] [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: 03/31/2025] [Revised: 04/29/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
The emergence of immune checkpoint inhibitors (ICIs) has revolutionized standard therapies for non-small cell lung cancer (NSCLC) [...].
Collapse
Affiliation(s)
- Shin Saito
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai 981-0914, Japan; (S.S.); (S.S.)
| | - Yosuke Kawashima
- Department of Internal Medicine, Matsuzono Daini Hospital, Morioka 020-0103, Japan
| | - Hisashi Tanaka
- Department of Respiratory Medicine, Hirosaki University Hospital, Hirosaki 036-8563, Japan;
| | - Naruo Yoshimura
- Department of Respiratory Medicine, Tohoku Medical and Pharmaceutical University Hospital, Sendai 983-8512, Japan;
| | - Yoko Tsukita
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai 980-8574, Japan;
| | - Ryota Saito
- Department of Respiratory Medicine, Yamanashi Prefectural Central Hospital, Kofu 400-8506, Japan;
| | - Taku Nakagawa
- Department of Thoracic Surgery, Omagari Kosei Medical Center, Daisen 014-0027, Japan;
| | - Minehiko Inomata
- First Department of Internal Medicine, Toyama University Hospital, Toyama 930-0194, Japan;
| | - Hiromi Nagashima
- Department of Respiratory Medicine, Iwate Medical University Hospital, Iwate 028-3695, Japan;
| | - Shunichi Sugawara
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai 981-0914, Japan; (S.S.); (S.S.)
| |
Collapse
|
21
|
Mattson Cypert BK, Menard K, Chu G, McDevitt T, Verona RI, Rupnow B, Packman K. Mechanisms of Response and Resistance to PSMA×CD3 Bispecifics in CD34+ Humanized Mice. Mol Cancer Ther 2025; 24:740-752. [PMID: 40008870 DOI: 10.1158/1535-7163.mct-23-0779] [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: 11/07/2023] [Revised: 05/06/2024] [Accepted: 02/21/2025] [Indexed: 02/27/2025]
Abstract
Prostate cancer is considered immunologically "cold," with low mutational burden, tumor-infiltrating immune cells, and PD-L1 levels, culminating in poor response to immune checkpoint therapies. CD3 bispecific redirection antibodies can elicit T cell-mediated cytotoxicity and hold promise for immune cell recruitment into prostate tumors. CD3 redirection antibodies in solid tumors are still in the early phases of clinical development, and it is not yet understood whether these potential therapies will achieve the high response rates observed in hematologic malignancies or result in durable T-cell responses. In this study, we demonstrated that treatment with a prostate-specific membrane antigen (PSMA)-targeted CD3 redirector resulted in efficacy against LnCaP.AR human prostate xenografts in CD34+ cord blood-humanized mice. Efficacy correlated with T-cell infiltration into tumors with an activated phenotype and also increased PD-L1 expression. Engineered overexpression of PD-L1 in LNCaP.AR tumors resulted in resistance to PSMA×CD3 bispecific antibody treatment, whereas sensitivity was restored in combination with anti-PD-1 antibody pembrolizumab. PSMA×CD3 and anti-PD-1 combination treatment resulted in complete tumor responses in approximately 20% of mice and elicited immune responses that delayed growth of rechallenged tumors. In a second prostate model, patient-derived LuCaP 86.2 xenografts, PSMA×CD3 monotherapy treatment resulted in complete responses in 25% of mice. When PSMA×CD3-treated responder mice were rechallenged with LuCaP 86.2 tumors, partial control of tumor regrowth was associated with the expansion of effector memory T cells. These studies show that PSMA×CD3 treatment elicits antitumor memory T-cell responses and that combination with PD-1 blockade can enhance these effects in tumors with immune-suppressive tumor microenvironments.
Collapse
Affiliation(s)
| | - Krista Menard
- Janssen Research and Development, Spring House, Pennsylvania
| | - Gerald Chu
- Janssen Research and Development, Spring House, Pennsylvania
| | | | - Raluca I Verona
- Janssen Research and Development, Spring House, Pennsylvania
| | - Brent Rupnow
- Janssen Research and Development, Spring House, Pennsylvania
| | | |
Collapse
|
22
|
He J, Liu Y, Wang X, Song R, Zhang J, Li B, Wang H, Yu J, Wang L. The Impact of Radiation Dose to Immune Cells in Stage IV Non-Small Cell Lung Cancer in the Era of Immunotherapy. Clin Lung Cancer 2025; 26:221-227.e1. [PMID: 40055132 DOI: 10.1016/j.cllc.2025.02.006] [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: 07/14/2024] [Revised: 01/17/2025] [Accepted: 02/08/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE Thoracic radiotherapy (RT) is now widely used in the treatment of advanced non-small cell lung cancer (NSCLC) as palliative, consolidative or radical therapy. However, RT adversely impacts the immune system, which can be evaluated by calculating the estimated dose of radiation to immune cells (EDRIC). We evaluated the prognostic impact of the EDRIC in patients with advanced NSCLC who received immunotherapy and thoracic RT. METHODS We retrospectively enrolled 152 stage IV NSCLC patients who had received first-line immunotherapy and thoracic RT. EDRIC was a model developed by Jin et al., calculated using the number of radiotherapy fractions, mean lung dose, mean heart dose, and mean body dose. Spearman's rank correlation was used to assess the correlations between variables. The relationships of EDRIC (≥5.7 Gy vs. <5.7 Gy) with survival were assessed using Kaplan-Meier and Cox proportional hazard models. RESULTS The median PFS and OS were shorter in the EDRIC ≥ 5.7 Gy group (PFS: 10.2 months vs. 18.6 months, P < .0001; OS: 19.8 months vs. 30.2 months, P = .024). In the multivariate model, higher EDRIC was associated with worse PFS (HR = 2.791, P < .0001) and OS (HR = 1.823, P = .028). Additionally, bone metastasis was associated with worse OS (HR = 1.751, P = .022). CONCLUSION EDRIC was an independent predictor for PFS and OS in advanced NSCLC patients receiving immunotherapy and RT. These observations necessitate further exploration into techniques to optimize radiation exposure to the immune system in cancer treatment.
Collapse
Affiliation(s)
- Junyi He
- Shandong University Cancer Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yingxin Liu
- Shandong University Cancer Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiaoqing Wang
- Department of Portal Hypertension, Shandong Public Health Clinical Center, Shandong University, Jinan, Shandong, China
| | - Ruiting Song
- Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jingze Zhang
- Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Butuo Li
- Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Haohua Wang
- Shandong University Cancer Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jinming Yu
- Shandong University Cancer Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Linlin Wang
- Shandong University Cancer Center, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
23
|
Marrone MT, Reuss JE, Crawford A, Neelon B, Liu JO, Brahmer JR, Platz EA. Statin Use With Immune Checkpoint Inhibitors and Survival in Nonsmall Cell Lung Cancer. Clin Lung Cancer 2025; 26:201-209. [PMID: 39818516 PMCID: PMC12037305 DOI: 10.1016/j.cllc.2024.12.008] [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: 11/11/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/18/2025]
Abstract
OBJECTIVE To determine the association between concurrent statin use with immune checkpoint inhibitors (ICIs) and lung cancer-specific and overall mortality in patients with nonsmall cell lung cancer (NSCLC). MATERIALS AND METHODS SEER-Medicare was used to conduct a retrospective study of Medicare beneficiaries ≥65 years of age diagnosed with NSCLC between 2007 and 2017 treated with an ICI. Patients were followed from date of first ICI claim until death, 1 month from last ICI claim, or 12/31/2018, whichever came first. Associations for time-updated statin use and lung cancer-specific mortality, and overall mortality were estimated using Cox models adjusted for demographic, pathological, treatment-related factors, and a propensity score for statin use. RESULTS Among 1,401 patients, concurrent statin use with any ICI was associated with 41% lower risk of lung-cancer specific mortality compared to patients receiving ICI not using a statin (HR = 0.59; 95% CI = 0.35-0.99). Statin use was associated with a similarly lower risk of overall mortality (HR = 0.62; 95% CI = 0.41-0.94). Consistent inverse associations were observed when restricting to PD-1 inhibitors and by statin type. Limited anti-PD-L1 treatment prevented analysis in this subgroup. CONCLUSION Concurrent statin use with ICIs was associated with lower risk of lung cancer-specific and overall mortality in a population-based sample of older patients with NSCLC. Future work is needed to confirm these findings in prospective studies and randomized trials, including evaluating concurrent statin use with frontline ICIs, deciphering the underlying mechanism, and determining the optimal statin-ICI combination that maximize clinical benefit.
Collapse
Affiliation(s)
- Michael T Marrone
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC.
| | - Joshua E Reuss
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Anna Crawford
- Hollings Cancer Center Biostatistics Shared Resource, Medical University of South Carolina, Charleston, SC
| | - Brian Neelon
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC; Hollings Cancer Center Biostatistics Shared Resource, Medical University of South Carolina, Charleston, SC
| | - Jun O Liu
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD
| | - Julie R Brahmer
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins, Baltimore, MD; The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, MD; The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Elizabeth A Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| |
Collapse
|
24
|
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
|
25
|
Ohmoto A, Shigematsu Y, Saito R, Dobashi A, Fujiwara Y, Togashi Y, Yonese J, Inamura K, Takahashi S. Prognosis and tumor microenvironment in pseudohypoxic pheochromocytoma/paraganglioma. Virchows Arch 2025; 486:983-990. [PMID: 39694932 DOI: 10.1007/s00428-024-04009-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 12/02/2024] [Accepted: 12/08/2024] [Indexed: 12/20/2024]
Abstract
Pheochromocytoma and paraganglioma (PPGL) are rare tumors that occur in the adrenal medulla and extra-adrenal tissues, respectively. The prognosis and tumor microenvironment (TME) of pseudohypoxic PPGL as a major entity have not been fully described. Based on the clinical database of 65 patients with PPGL, we assessed the morphological features as well as the immunohistochemistry of pseudohypoxia-related proteins (SDHB and CAIX) and TME-related immune cell markers. Furthermore, we compared the relapse-free survival (RFS) rates in localized patients between the pathological subgroups. Among 50 available specimens, 84% and 30% of the cases exhibited at least one morphological adverse feature including vascular/capsular invasion and a Ki-67 index > 3%, respectively. The SDHB and CAIX positivity rates were 81% and 51%. Concerning the immune cell markers, the CD163-positive cell numbers were higher in hypoxia-associated PPGL composed of SDHB-negative or CAIX-positive cases than in non-hypoxia PPGL (median 66 vs. 23/mm2). Concerning prognosis, RFS rates were significantly lower in cases with Ki-67 indices > 3% and SDHB-negativity than in those with Ki-67 indices ≤ 3% and SDHB-positivity (3-year rate: 64% vs. 100%, P < 0.001; 57% vs. 100%, P = 0.03). In contrast, RFS was comparable between CAIX-positive and CAIX-negative PPGL cases. Our analyses suggested that SDHB-deficient PPGL exhibited a higher incidence of relapse. Furthermore, M2 macrophage infiltration in TME might be crucial in pseudohypoxic PPGL pathogenesis.
Collapse
Affiliation(s)
- Akihiro Ohmoto
- Department of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 1358550, Japan.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, 417 East 68th Street, New York, NY, 10065, USA.
| | - Yasuyuki Shigematsu
- Department of Pathology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 1358550, Japan
- Division of Pathology, Cancer Institute of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 1358550, Japan
| | - Rumiko Saito
- Department of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 1358550, Japan
- Department of Clinical Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 1358550, Japan
| | - Akito Dobashi
- Pathology Project for Molecular Targets, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 1358550, Japan
| | - Yu Fujiwara
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Mount Sinai Beth Israel, 281 First Avenue, New York, NY, 10003, USA
| | - Yuki Togashi
- Pathology Project for Molecular Targets, Cancer Institute, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 1358550, Japan
| | - Junji Yonese
- Department of Genitourinary Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 1358550, Japan
| | - Kentaro Inamura
- Department of Pathology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 1358550, Japan
- Division of Pathology, Cancer Institute of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 1358550, Japan
- Division of Tumor Pathology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi, 3290498, Japan
| | - Shunji Takahashi
- Department of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo, 1358550, Japan.
| |
Collapse
|
26
|
Almuradova E, Izzo D, Gandini S, Gaeta A, Giordano E, Valenza C, Antonarelli G, Trapani D, Curigliano G. From Dose-Finding to Dose-Optimization in Early-Phase oncology clinical trials. Cancer Treat Rev 2025; 136:102906. [PMID: 40157116 DOI: 10.1016/j.ctrv.2025.102906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 04/01/2025]
Abstract
Dose optimization in Phase I oncology trials balances therapeutic efficacy and patient safety. Traditional dose-escalation methods, such as the 3 + 3 design, primarily focus on safety, often resulting in prolonged exposure to subtherapeutic or excessively toxic doses. Additionally, these methods may fail to account for modern therapies' complex pharmacokinetics and pharmacodynamics, including targeted agents and immunotherapies. Contemporary approaches address these gaps by incorporating biomarkers, pharmacokinetic profiling, and patient-reported outcomes to guide personalized dosing strategies. Such methods improve the precision of dose selection and promote individualized cancer care. This review underscores the importance of distinguishing between dose-finding and dose optimization, advocating for designs that integrate patient perspectives and pharmacologic insights from early-phase trials. Additionally, we highlight the challenges of traditional methodologies and the importance of simplifying complex designs without compromising their scientific rigor. By embracing innovative approaches and patient-centered metrics, Phase I trials can evolve beyond safety assessments to expedite the delivery of effective and tailored cancer therapies.
Collapse
Affiliation(s)
- Elvina Almuradova
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Ege University Hospital, Department of Medical Oncology, Izmir, Turkey
| | - Davide Izzo
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Sara Gandini
- Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Aurora Gaeta
- Experimental Oncology, European Institute of Oncology, IRCCS, Milan, Italy
| | - Edoardo Giordano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Carmine Valenza
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Harvard Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Gabriele Antonarelli
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Dario Trapani
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
27
|
Zhao Z, Yang C, Li J. Pemetrexed and platinum with or without pembrolizumab for advanced non-small-cell lung cancer (NSCLC): a systematic review and meta-analysis. Clin Transl Oncol 2025; 27:2024-2036. [PMID: 39402420 DOI: 10.1007/s12094-024-03751-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/01/2024] [Indexed: 04/19/2025]
Abstract
OBJECTIVE This meta-analysis aimed to evaluate the efficacy and safety of combining pemetrexed and platinum with or without pembrolizumab for the treatment of advanced non-small-cell lung cancer (NSCLC). METHODS A systematic search of PubMed, Embase, Cochrane Library, and Web Of Science databases was conducted to identify studies comparing pemetrexed and platinum with or without pembrolizumab in advanced NSCLC. Raw data were extracted from eligible studies to calculate Hazard Ratios (HR) for Progression-Free Survival (PFS) and Overall Survival (OS), as well as rates of adverse events of all grades and those of Grade 3 or higher. RESULTS Eight studies with 1639 patients occurred advanced NSCLC included. The group receiving pembrolizumab in combination with pemetrexed and platinum showed significant benefits in terms of OS (HR 0.63; 95% CI 0.54-0.73; p < 0.00001) and PFS (HR:0.64; 95% CI 0.48-0.85; p = 0.002) compared to the group receiving pemetrexed and platinum alone. However, this benefit was accompanied by a higher incidence of Grade 3 or higher adverse events (OR: 1.55; 95% CI 1.24-1.95; p = 0.0001). CONCLUSION The combination of pemetrexed and platinum with pembrolizumab is recommended as a first-line treatment option for advanced NSCLC due to its significant efficacy benefits. However, the increased risk of Grade 3 or higher adverse events suggests the need for careful consideration and assessment when considering this regimen for second-line or subsequent therapy.
Collapse
Affiliation(s)
- Zichen Zhao
- Lianyungang Clinical Medical College of Nanjing Medical University, Lianyungang, China
| | - Chuchu Yang
- Lianyungang Clinical Medical College of Nanjing Medical University, Lianyungang, China
| | - Jiashu Li
- Lianyungang Clinical Medical College of Nanjing Medical University, Lianyungang, China.
| |
Collapse
|
28
|
Han T, Cheng S, Wang X, Qi Q, Chen J, Wang W, Zhou J, Li Y, Chen K, Li H, Yang F. Prognostic Significance, Radiological, and Metabolic Characteristics of Metastatic Lymph Nodes in Resectable Non-Small Cell Lung Cancer Following Neoadjuvant Chemoimmunotherapy. Thorac Cancer 2025; 16:e70073. [PMID: 40368376 PMCID: PMC12077928 DOI: 10.1111/1759-7714.70073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/12/2025] [Accepted: 04/15/2025] [Indexed: 05/16/2025] Open
Abstract
BACKGROUND Metastatic lymph nodes (mLNs) exhibit different responses to neoadjuvant immunotherapy compared to the primary tumor (PT) in non-small cell lung cancer (NSCLC). Evaluating mLNs' response is crucial for predicting treatment efficacy and prognosis; however, such assessments are currently insufficient. METHODS We enrolled 101 NSCLC patients with mLNs who underwent neoadjuvant chemoimmunotherapy followed by surgery. Survival outcomes and radiological and metabolic changes were analyzed across different lymph node pathological response groups, and a least absolute shrinkage and selection operator (LASSO) logistic regression model was developed to predict mLNs' response. RNA sequencing was performed to characterize the immune microenvironment of lymph nodes with different pathological responses. RESULTS Residual tumors in mLNs were significantly associated with worse recurrence-free survival (p = 0.003) and a trend toward worse overall survival (p = 0.087). Combining the pathological responses of mLNs and PTs improved prognostic stratification. Neither radiological size changes (AUC: 0.621) nor the SUVmax reduction rate (AUC: 0.645) were effective in distinguishing mLNs response. A model combining radiological and metabolic parameters demonstrated fair prediction efficacy (AUC: 0.85). In separate analyses of N1 and N2 nodes, radiological and metabolic changes of N1 mLNs partly reflected their pathologic response (AUC: 0.734; 0.816), unlike in N2 mLNs. RNA sequencing revealed that immune infiltration in responding lymph nodes differed from non-responding ones, with higher CD8+ T cells, NK T cells, B cells, and dendritic cells in the former. CONCLUSION The pathological response of mLNs provides additional prognostic information, but current tools are ineffective for detecting residual tumors. A model integrating radiological and metabolic parameters may offer better prediction.
Collapse
Affiliation(s)
- Tianxiao Han
- Department of Thoracic Surgery, Thoracic Oncology InstitutePeking University People's HospitalBeijingChina
| | - Sida Cheng
- Department of Thoracic Surgery, Thoracic Oncology InstitutePeking University People's HospitalBeijingChina
| | - Xun Wang
- Department of Thoracic Surgery, Thoracic Oncology InstitutePeking University People's HospitalBeijingChina
| | - QingYi Qi
- Department of RadiologyPeking University People's HospitalBeijingChina
| | - Jinchuan Chen
- Department of Nuclear MedicinePeking University People's HospitalBeijingChina
| | - Wenxiang Wang
- Department of Thoracic Surgery, Thoracic Oncology InstitutePeking University People's HospitalBeijingChina
| | - Jian Zhou
- Department of Thoracic Surgery, Thoracic Oncology InstitutePeking University People's HospitalBeijingChina
| | - Yun Li
- Department of Thoracic Surgery, Thoracic Oncology InstitutePeking University People's HospitalBeijingChina
| | - Kezhong Chen
- Department of Thoracic Surgery, Thoracic Oncology InstitutePeking University People's HospitalBeijingChina
| | - Hao Li
- Department of Thoracic Surgery, Thoracic Oncology InstitutePeking University People's HospitalBeijingChina
| | - Fan Yang
- Department of Thoracic Surgery, Thoracic Oncology InstitutePeking University People's HospitalBeijingChina
| |
Collapse
|
29
|
Wang J, Chen Q, Shan Q, Liang T, Forde P, Zheng L. Clinical development of immuno-oncology therapeutics. Cancer Lett 2025; 617:217616. [PMID: 40054657 PMCID: PMC11930610 DOI: 10.1016/j.canlet.2025.217616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
Immuno-oncology (IO) is one of the fastest growing therapeutic areas within oncology. IO agents work indirectly via the host's adaptive and innate immune system to recognize and eradicate tumor cells. Despite checkpoint inhibitors being only introduced to the market since 2011, they have become the second most approved product category. Current Food and Drug Administration (FDA)-approved classes of IO agents include: immune checkpoint inhibitors (ICIs), chimeric antigen receptor T-cell therapy (CAR-T), bi-specific T-cell engager (BiTE) antibody therapy, T-cell receptor (TCR) engineered T cell therapy, tumor-infiltrating lymphocyte (TIL) therapy, cytokine therapy, cancer vaccine therapy, and oncolytic virus therapy. Cancer immunotherapy has made progress in multiple cancer types including melanoma, non-small cell lung cancer (NSCLC), renal cell carcinoma (RCC), and urothelial carcinoma; however, several cancers remain refractory to immunotherapy. Future directions of IO include exploration in the neoadjuvant/perioperative setting, combination strategies, and optimizing patient selection through improved biomarkers.
Collapse
Affiliation(s)
- Jianxin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Qi Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Qiang Shan
- Department of General Surgery, Haining People's Hospital, Haining, 314400, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Patrick Forde
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA; The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Mays Cancer Center at the University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Lei Zheng
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA; The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Mays Cancer Center at the University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
30
|
Tsukita Y, Umezawa R, Nakagawa T, Anbai A, Makiguchi T, Tanaka H, Horii Y, Suzuki A, Morita R, Nogawa H, Yokouchi H, Kimura N, Jingu K, Inoue A, Sugiura H, Miyauchi E. Phase 2 Trial of Combination Radiotherapy and Pembrolizumab Plus Chemotherapy in Patients With Previously Untreated Metastatic NSCLC: NJLCG 1902. JTO Clin Res Rep 2025; 6:100817. [PMID: 40225955 PMCID: PMC11992379 DOI: 10.1016/j.jtocrr.2025.100817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 02/02/2025] [Accepted: 02/24/2025] [Indexed: 04/15/2025] Open
Abstract
Introduction Treatment strategies that enhance the efficacy of immunotherapy are desired. Radiotherapy can enhance immunity, but the utility of adding radiotherapy to immunotherapy plus platinum-doubled chemotherapy in patients with metastatic NSCLC has not been explored. Methods This multicenter, single-arm phase 2 trial evaluated the efficacy and safety of combining radiotherapy with pembrolizumab plus chemotherapy in patients with previously untreated metastatic NSCLC. Patients begin receiving pembrolizumab plus platinum-doublet chemotherapy within 1 week of starting radiotherapy (30 Gy in 10 fractions). The primary end point was the 12-month progression-free survival (PFS) rate. The secondary end points included PFS, overall survival, and toxicity profiles. Results Forty patients were enrolled. In total, 37 and 38 patients were analyzed for efficacy and safety, respectively. The 12-month PFS rate was 44.3% (90% confidence interval [CI]: 30.3-57.3), which met the primary end point. The median PFS was 8.4 months (95% CI: 5.7-22.2), and the median overall survival was 30.1 months (95% CI: 22.3-not reached). Grade 3 or 4 adverse events occurred in 25 patients (65.8%), and one treatment-related death was reported. Pneumonitis was reported in 10 patients (26.3%), including two cases of grade 3 pneumonitis and one case of grade 5. Conclusions Adding radiotherapy to pembrolizumab plus platinum-doublet chemotherapy led to promising efficacy in patients with previously untreated metastatic NSCLC. Although caution should be exercised with regard to pneumonitis, adverse events were tolerable. Further research is needed to confirm the efficacy and safety of this strategy.
Collapse
Affiliation(s)
- Yoko Tsukita
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Rei Umezawa
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Taku Nakagawa
- Department of Thoracic Surgery, Omagari Kosei Medical Center, Daisen, Japan
| | - Akira Anbai
- Department of Radiology, Omagari Kosei Medical Center, Daisen, Japan
| | - Tomonori Makiguchi
- Department of Respiratory Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Hisashi Tanaka
- Department of Respiratory Medicine, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Yosuke Horii
- Division of Pulmonary Medicine, Department of Internal Medicine, Iwate Medical University School of Medicine, Morioka, Japan
| | - Aya Suzuki
- Department of Respiratory Medicine, Miyagi Cancer Center, Natori, Japan
| | - Ryo Morita
- Department of Respiratory Medicine, Akita Kousei Medical Center, Akita, Japan
| | - Hitomi Nogawa
- Department of Respiratory Medicine, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Hiroshi Yokouchi
- Department of Respiratory Medicine, NHO Hokkaido Cancer Center, Sapporo, Japan
| | - Nozomu Kimura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Keiichi Jingu
- Department of Radiation Oncology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Inoue
- Department of Palliative Medicine, Tohoku University School of Medicine, Sendai, Japan
| | - Hisatoshi Sugiura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Eisaku Miyauchi
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
31
|
Huang YE, Zhou S, Chen S, Chen J, Zhou X, Hou F, Liu H, Yuan M, Jiang W. Mutational signature-based biomarker to predict the response of immune checkpoint inhibitors therapy in cancers. Int J Biol Macromol 2025; 308:142585. [PMID: 40154701 DOI: 10.1016/j.ijbiomac.2025.142585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 03/23/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Patients have a limited response rate to immune checkpoint inhibitors (ICIs) therapy. Although several biomarkers have been proposed, their ability to accurately predict the response to ICIs therapy remains unsatisfactory. In addition, mutational signatures were validated to be associated with ICIs therapy. Therefore, we developed a mutational signature-based biomarker (MS-bio) to predict the response to ICIs therapy. Based on differentially mutated genes, we extracted six mutational signatures (single-base substitution (SBS)-A, SBS-B, SBS-C, SBS-D, double-base substitution (DBS)-A, and DBS-B) as MS-bio, and constructed a random forest (RF) model to predict the response. Internal and external validations consistently demonstrated the excellent predictive capability of MS-bio, with an accuracy reaching up to 0.82. Moreover, MS-bio exhibited superior performance compared to existing biomarkers. To further validate the accuracy of MS-bio, we explored its performance in The Cancer Genome Atlas (TCGA) cohort and found that the predicted responders were immunologically "hot". Finally, we found that SBS-C had the highest importance in prediction and was related to T cell differentiation. Overall, here we introduced MS-bio as a novel biomarker for accurately predicting the response to ICIs therapy, thereby contributing to the advancement of precision medicine.
Collapse
Affiliation(s)
- Yu-E Huang
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China; Guizhou Institute of Precision Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China; Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Shunheng Zhou
- School of Computer Sciences, University of South China, Hengyang 421001, China
| | - Sina Chen
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Jiahao Chen
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China
| | - Xu Zhou
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Fei Hou
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Haizhou Liu
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Mengqin Yuan
- Department of Biomedical Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing 211106, China
| | - Wei Jiang
- Fujian Provincial Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Fuzhou 350005, China; National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China.
| |
Collapse
|
32
|
Nogami N, Umemura S, Kozuki T, Zenke Y, Ohtani J, Ishii M, Han S, Noguchi K, Horinouchi H. A phase 1 study of pembrolizumab plus ipilimumab as first-line treatment in Japanese patients with advanced non-small-cell lung cancer. Respir Investig 2025; 63:296-302. [PMID: 40036983 DOI: 10.1016/j.resinv.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 02/11/2025] [Accepted: 02/14/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Part D of the open-label, phase 1 KEYNOTE-011 study (ClinicalTrials.gov, NCT01840579) evaluated the safety and tolerability, pharmacokinetics, and antitumor activity of pembrolizumab plus ipilimumab as first-line treatment in Japanese participants with advanced NSCLC. METHODS Eligible participants were aged ≥20 years with previously untreated stage IIIB/IV NSCLC (any tumor PD-L1 status permitted). Participants received ≤35 doses of pembrolizumab 200 mg every 3 weeks intravenously (days 1 and 22 of 6-week cycles) plus ≤18 doses of ipilimumab 1 mg/kg every 6 weeks intravenously. The primary endpoint was dose-limiting toxicities (DLTs) occurring during the DLT evaluation period (first 6 weeks of study treatment). The secondary endpoint was pembrolizumab pharmacokinetics; exploratory endpoints included evaluation of antitumor activity. Results were summarized descriptively. RESULTS All 6 participants enrolled in Part D received ≥1 dose of pembrolizumab plus ipilimumab. No DLTs were observed. AEs of any cause were reported in 5 participants (83%; grade 3-4, n = 2 [33%]). No fatal AEs occurred. Two participants had AEs leading to discontinuation: grade 3 pneumonia (n = 1; not treatment-related) and grade 2 organizing pneumonia (n = 1; treatment-related). The geometric mean (% coefficient of variation) pembrolizumab maximum and minimum serum concentrations were 72.7 μg/mL (10.1%) and 8.3 μg/mL (16.2%), respectively, at cycle 1 and 86.2 μg/mL (3.6%) and 28.1 μg/mL (23.6%), respectively, at cycle 4. Objective response rate was 50% (95% CI, 12%-88%); 3 participants had PR. CONCLUSION Pembrolizumab plus ipilimumab had a manageable safety profile and showed antitumor activity in Japanese participants with previously untreated advanced NSCLC.
Collapse
Affiliation(s)
- Naoyuki Nogami
- Ehime University Graduate School of Medicine, Department of Community Medicine, Pulmonology and Cardiology, Toon, Japan.
| | - Shigeki Umemura
- National Cancer Center Hospital East, Department of Thoracic Oncology, Kashiwanoha, 6 Chome-5-1, Chiba, Kashiwa, 277-8577, Japan
| | - Toshiyuki Kozuki
- NHO Shikoku Cancer Center, Department of Thoracic Oncology and Medicine, 160 Minamiumemotomachi, Matsuyama, Ehime, 791-0245, Japan
| | - Yoshitaka Zenke
- National Cancer Center Hospital East, Department of Thoracic Oncology, Kashiwanoha, 6 Chome-5-1, Chiba, Kashiwa, 277-8577, Japan
| | - Junko Ohtani
- MSD K.K., 1 Chome-13-12 Kudankita, Chiyoda City, Tokyo, 102-0073, Japan
| | - Mikio Ishii
- MSD K.K., 1 Chome-13-12 Kudankita, Chiyoda City, Tokyo, 102-0073, Japan
| | - Shirong Han
- MSD K.K., 1 Chome-13-12 Kudankita, Chiyoda City, Tokyo, 102-0073, Japan
| | - Kazuo Noguchi
- MSD K.K., 1 Chome-13-12 Kudankita, Chiyoda City, Tokyo, 102-0073, Japan
| | - Hidehito Horinouchi
- National Cancer Center Hospital, Department of Thoracic Oncology, 5 Chome-1-1 Tsukiji, Chuo City, Tokyo, 104-0045, Japan
| |
Collapse
|
33
|
Li L, Pu H, Zhang X, Guo X, Li G, Zhang M. Resistance to PD-1/PD-L1 immune checkpoint blockade in advanced non-small cell lung cancer. Crit Rev Oncol Hematol 2025; 209:104683. [PMID: 40024354 DOI: 10.1016/j.critrevonc.2025.104683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
Lung cancer is one of the most common malignant tumors, of which non-small cell lung cancer (NSCLC) accounts for about 85 %. Although immune checkpoint inhibitors (ICIs), particularly PD-1/PD-L1 inhibitors, have significantly improved the prognosis of patients with NSCLC. There are still many patients do not benefit from ICIs. Primary resistance remains a major challenge in advanced NSCLC. The cancer-immunity cycle describes the process from antigen release to T cell recognition and killing of the tumor, which provides a framework for understanding anti-tumor immunity. The classical cycle consists of seven steps, and alterations at each stage can result in resistance. This review examines the current status of PD-1/PD-L1 blockade in the treatment of advanced NSCLC and explores potential mechanisms of resistance. We summarize the latest clinical trials of PD-1/PD-L1 inhibitors combined with other therapies and explore potential targets for overcoming primary resistance to PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Lijun Li
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Haihong Pu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xiaoxin Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xiaotian Guo
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Guangrui Li
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Minghui Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
34
|
Hu Z, Hu Z, Chen K, Huang H, Zhong X, Wang Y, Chen J, He X, Shi D, Zeng Y, Li J, Zhou X, Wei P. The Spatial Proximity of CD8 + FoxP3 +PD-1 + Cells to Tumor Cells: A More Accurate Predictor of Immunotherapy Outcomes in Advanced Non-Small-Cell Lung Cancer. Curr Oncol 2025; 32:262. [PMID: 40422521 DOI: 10.3390/curroncol32050262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND To optimize precision immunotherapy for advanced NSCLC, comprehensive tumor immune microenvironment (TIME) characterization is crucial for efficacy prediction. METHODS Pretreatment tumor samples from 46 advanced NSCLC patients treated with PD-1/PD-L1 inhibitors were analyzed. The subregional abundance and spatial proximity scores of TIME cell subpopulations in 27 samples were assessed via multiplex immunohistochemistry (mIHC) targeting pan-CK, CD163, CD8, FoxP3, PD-1, and PD-L1. Correlations between the TIME features, clinicopathologic factors, treatment response, and prognosis were evaluated. RESULTS CD8+FoxP3+ cells were identified in NSCLC tissues, predominantly expressing PD-1/PD-L1. The PD-L1 TPS subgroups showed significant immune cell density/proximity differences, but CD8+FoxP3+PD-1+ infiltration was PD-L1 TPS-independent. Responders had higher CD8+FoxP3+PD-1high density (p = 0.0497) and proximity scores (p = 0.0099) than non-responders. The CD8+FoxP3+PD-1+ presence and tumor proximity were essential for favorable outcomes. In low-PD-L1 TPS patients, the CD8+FoxP3+PD-1+ abundance and proximity scores strongly predicted the response (AUC: 0.79 and 0.75 vs. PD-L1 TPS AUC = 0.58). A survival analysis linked the presence and proximity score of CD8+FoxP3+PD-1+ cells to prolonged overall survival (OS) and progression-free survival (PFS). Notably, a low proximity score of CD8+FoxP3+PD-1+ cells emerged as an independent risk factor for a shorter PFS (HR = 6.16, 95% CI: 2.12-17.93, p = 0.001). CONCLUSION The CD8+FoxP3+PD-1+ spatial proximity to tumor cells robustly predicts improved immunotherapy outcomes in advanced NSCLC.
Collapse
Affiliation(s)
- Zijuan Hu
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Zhihuang Hu
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Thoracic Medical Oncology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Keji Chen
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Huixia Huang
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Xinyang Zhong
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Yaxian Wang
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Jiayu Chen
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Xuefeng He
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| | - Di Shi
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
- Institute of Pathology, Fudan University, 270 Dong'an Road, Shanghai 200032, China
| | - Yupeng Zeng
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
- Institute of Pathology, Fudan University, 270 Dong'an Road, Shanghai 200032, China
| | - Jiwei Li
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Xiaoyan Zhou
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
- Institute of Pathology, Fudan University, 270 Dong'an Road, Shanghai 200032, China
| | - Ping Wei
- Cancer Institute, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, 270 Dong'an Road, Shanghai 200032, China
- Department of Pathology, Fudan University Shanghai Cancer Center, 270 Dong'an Road, Shanghai 200032, China
| |
Collapse
|
35
|
Liu B, Zhou J, He W, Xie B, Zhang R, Cheng X, Zhang Y, Xu L, Guo S. Safety of bronchial artery infusion immunotherapy: from comparative analysis in beagle canines to clinical validation. Discov Oncol 2025; 16:621. [PMID: 40285957 PMCID: PMC12033145 DOI: 10.1007/s12672-025-02398-2] [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: 01/01/2025] [Accepted: 04/15/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Despite advancements in systemic chemotherapy and immune checkpoint inhibitors (ICIs), advanced non-small cell lung cancer (NSCLC) continues to exhibit poor prognosis, underscoring an urgent need for safer and more effective therapeutic strategies. This study investigates the safety profile and biological effects of bronchial arterial infusion (BAI)-administered anti-PD-1 monoclonal antibody (aPD-1 mAb) using a preclinical beagle model and a clinical cohort of advanced NSCLC patients. METHODS In preclinical evaluations, male beagles (n = 3/group) were randomized to receive 5 mg/kg aPD-1 mAb via BAI or intravenous routes (Venous group). Safety assessments included longitudinal imaging, biochemical analyses, and histopathological evaluation. Clinically, patients with advanced NSCLC meeting stringent inclusion criteria underwent BAI immunotherapy, with systematic monitoring of adverse events (AEs). RESULTS Both administration routes demonstrated comparable safety in canines, with no evidence of immune-related pneumonitis or structural lung alterations on CT or histology. Transient AEs (e.g., hematoma, lameness) resolved spontaneously. Pharmacokinetic analysis revealed similar systemic drug concentrations and tissue distribution between BAI and Venous groups (all p > 0.05). Biochemical profiling identified isolated mild LDH elevation in one BAI-treated canine. Notably, the BAI group exhibited significantly enhanced systemic IL-2 levels (80.15 ± 5.24 pg/mL vs. 66.47 ± 5.24 pg/mL in Venous groups, p = 0.001) at day 28, paralleled by elevated pulmonary IL-2 expression (626.90 ± 18.49 vs. 559.18 ± 45.61 pg/mg, p = 0.03). In the clinical cohort (n = 17; 94.1% male, mean age 61.6 ± 7.1 years), BAI immunotherapy was well-tolerated with mild AEs including nausea (n = 1), dyspnea (n = 1), atrial fibrillation (n = 1), and puncture-site hematoma (n = 1). No severe immune-related toxicities (e.g., pneumonitis) emerged during follow-up. CONCLUSION Our study suggest the preliminary safety and feasibility of delivering aPD-1 mAb via BAI in both canine models and NSCLC patients.
Collapse
Affiliation(s)
- Bin Liu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Youyi Rd 1, Yuzhong, 400016, Chongqing, China
- Department of Pulmonary and Critical Care Medicine, Zhuzhou Central Hospital, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, No. 116, Changjiang South Road, Tianyuan District, Zhuzhou, 412007, Hunan, China
| | - Jia Zhou
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Youyi Rd 1, Yuzhong, 400016, Chongqing, China
| | - Wei He
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Youyi Rd 1, Yuzhong, 400016, Chongqing, China
| | - Bo Xie
- Department of Medical Administration, Zhuzhou Central Hospital, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412007, Hunan, China
| | - Rui Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Youyi Rd 1, Yuzhong, 400016, Chongqing, China
| | - Xiaocheng Cheng
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Youyi Rd 1, Yuzhong, 400016, Chongqing, China
| | - Yueming Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Youyi Rd 1, Yuzhong, 400016, Chongqing, China
| | - Li Xu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Youyi Rd 1, Yuzhong, 400016, Chongqing, China
| | - Shuliang Guo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Youyi Rd 1, Yuzhong, 400016, Chongqing, China.
| |
Collapse
|
36
|
Zhu X, Yin G, Xu J, Tang X, Yu F. Dendrobium huoshanense polysaccharide inhibits NSCLC proliferation and immune evasion via FXR1-IL-35 axis signaling pathway. J Nat Med 2025:10.1007/s11418-025-01894-7. [PMID: 40259042 DOI: 10.1007/s11418-025-01894-7] [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: 05/14/2024] [Accepted: 03/10/2025] [Indexed: 04/23/2025]
Abstract
Dendrobium huoshanense has received special attention for its advantages in the treatment of lung cancer, but the underlying molecular mechanisms are not yet well understood. First, we obtained 8 active ingredients and 159 effective action targets of Dendrobium huoshanense using network pharmacology, and searching target interactions through STRING, constructing the PPI network and KEGG, GO and Hallmark enrichment analysis. Then, we combined target's enrichment analysis and GSEA enrichment analysis of IL-35, indicating the mechanism of cDHPs for non-small cell lung cancer (NSCLC) may be related to tight junction and NSCLC pathway. Further, FXR1 and ACTR3 were identified as core therapeutic targets, and high expression of FXR1 or ACTR3 was significantly associated with poor prognosis of patients. The analysis of single-cell data also indicated that the percentage of CD4-CTLA4-Treg cells may be increased by the expression of IL-35, resulting in a suppressive immune microenvironment. Next, In vivo experiment, we detected iTr35 by flow cytometry, detected IL-35 level by RT-PCR, Western blotting and ELISA, and detected NK cell activity to explore the immunomodulatory effects and anti-tumor mechanism of cDHPs. After cDHPs administration, the conversion of CD4+ T cells to iTr35 is inhibited, p35 and EBI3 in both protein and mRNA levels, the levels of IL-35 and IL-4 in serum decreased. The levels of IFN-γ, while the activity of NK cells in mice increased, enhancing the anti-tumor immune effect of the organism. Finally, analysis of sequencing data from the immunotherapy cohort of tumor-bearing mice obtained from the TISMO database shows that the combination of cDHPs and PD-1/PD-L1 antibodies improves effector and thus PD-1/PD-L1 antibody efficacy. These findings suggest that cDHPs inhibit NSCLC proliferation and immune escape via the FXR1-IL-35 axis signaling pathway.
Collapse
Affiliation(s)
- Xinying Zhu
- Translational Medicine Center, The Second Affiliated Hospital of Wannan Medical College, Wuhu, 241000, Anhui Province, China
| | - Guoquan Yin
- Clinical Laboratory, Yangzhou Blood Center in Jiangsu Province, Yangzhou, 225007, Jiangsu Province, China
| | - Jiaqian Xu
- Department of Medical Microbiology and Immunology, School of Preclinical Medicine, Wannan Medical College, Wuhu, 241001, Anhui Province, China
| | - Xiaolei Tang
- Translational Medicine Center, The Second Affiliated Hospital of Wannan Medical College, Wuhu, 241000, Anhui Province, China
| | - Fangliu Yu
- Department of Medical Microbiology and Immunology, School of Preclinical Medicine, Wannan Medical College, Wuhu, 241001, Anhui Province, China.
| |
Collapse
|
37
|
Wu Y, Li R, Tan F, Cao J, Bi N. Efficacy of immunotherapy remained in patients with recurrent/metastatic non-small-cell lung cancer after surgery with or without postoperative thoracic radiotherapy: a bi-center retrospective study. Thorac Cancer 2025. [PMID: 40244829 DOI: 10.1111/1759-7714.15384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/04/2024] [Accepted: 05/09/2024] [Indexed: 04/19/2025] Open
Abstract
PURPOSE Since mediastinal lymph node dissection and radiotherapy (RT) have potential unclear impacts on pulmonary lymphatic system, this study aimed to assess the effectiveness of immune checkpoint inhibitors (ICIs) in recurrent/metastatic non-small-cell lung cancer (NSCLC) patients who previously received radical surgery with or without thoracic RT. METHODS Clinical data of patients who underwent pulmonary lobectomy with systematic lymphadenectomy (2000.1.1-2021.7.2) and received immunotherapy after progression were retrospectively analyzed. Efficacy was mainly evaluated based on progression-free survival (PFS) from the start of the ICIs. Toxicity was defined as treatment discontinuation due to immune-related adverse effects (irAEs). RESULTS Ninety-five patients were enrolled in the final cohort and 30 (31.6%) patients received thoracic RT before ICI treatment. ICIs were administered as a first-line systematic treatment in 52.6% of patients. The median follow-up time was 14.7 months (95% confidence interval [CI] 13.3-18.7 months). The median PFS was 12.3 months (95% CI 8.5-36.6 months). Six (6.3%) patients had treatment suspended due to irAEs. Patients who received RT had comparable median PFS with the non-RT group (17.0 months vs. 11.1 months, p = 0.16). Similar toxicity rates were observed. Similar mPFS were reported in the stage III subgroup (RT vs. non-RT, 8.10 vs. 8.45 months, p = 0.86) or the subgroup treated by ICIs as primary systematic therapy (RT vs. non-RT, 13.6 vs. 16.1 months, p = 0.45). CONCLUSIONS ICIs remained effective in recurrent/metastatic NSCLC patients with radical surgery and RT did not significantly compromise therapeutic effects.
Collapse
Affiliation(s)
- Yuqi Wu
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Renda Li
- 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, China
| | - Fengwei Tan
- 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, China
| | - Jianzhong Cao
- Department of Radiation Oncology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Nan Bi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
38
|
Liao S, Sun H, Lu H, Wu J, Wu J, Wu Z, Xi J, Liao W, Wang Y. Neutrophil-to-lymphocyte ratio-based prognostic score can predict outcomes in patients with advanced non-small cell lung cancer treated with immunotherapy plus chemotherapy. BMC Cancer 2025; 25:697. [PMID: 40234811 PMCID: PMC11998248 DOI: 10.1186/s12885-025-13811-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/25/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUD Immune checkpoint inhibitor (ICI) plus chemotherapy has become the standard of care for advanced non-small cell lung cancer (NSCLC). Nonetheless, reliable efficacy biomarkers of ICI plus chemotherapy are lacking. In this research, we sought to explore efficacy biomarkers and construct robust prognostic models in NSCLC patients treated with ICI plus chemotherapy. METHODS We retrospectively analyzed 171 patients with advanced NSCLC treated with ICI plus chemotherapy. Clinical characteristics and peripheral blood inflammatory indexes were collected and prognostic models were constructed to explore efficacy and prognosis biomarkers of ICI plus chemotherapy. RESULTS In the cohort that received first-line ICI plus chemotherapy, pre-treatment neutrophil-to-lymphocyte ratio (NLR) > 3.3 and fibrinogen (FIB) > 3.196 were associated with worse efficacy and were independent risk factors of progression-free survival (PFS). Compared to programmed cell death ligand 1 (PD-L1), the derived NLR-FIB (NF) score had significantly improved accuracy in predicting efficacy and prognosis. In advanced NSCLC patients with targetable oncogenic driver alterations receiving second- or post-line ICI plus chemotherapy, pre-treatment NLR > 3.53 was associated with worse efficacy and was an independent risk factor of PFS and OS; Tyrosine kinase inhibitor (TKI)-PFS > 12 months were independent risk factors of overall survival (OS). Secondary epidermal growth factor receptor (EGFR)-T790M mutation, platelet-to-lymphocyte ratio (PLR) > 196.81 and albumin (ALB) < 40.25 were associated with worse PFS. Based on NLR and TKI-PFS, an NLR-TKI-PFS (NTP) score was constructed with three OS risk prognosis categories: favorable, intermediate, and poor (corresponding to a median OS of 21, 12, and 5.3 months). CONCLUSIONS The noninvasive NF score, combining NLR > 3.3 and FIB > 3.196, was superior to PD-L1 estimated from tumor tissue in predicting the efficacy and prognosis of first-line ICI plus chemotherapy in advanced NSCLC patients. The noninvasive NTP score, combining NLR > 3.53 and TKI-PFS > 12 months, is a valuable tool for predicting OS and PFS in advanced NSCLC patients with targetable oncogenic driver alterations receiving second- or post-line ICI combination therapy.
Collapse
Affiliation(s)
- Shan Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Huiying Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Hao Lu
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Jiani Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Jianhua Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Zhe Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Jingle Xi
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China.
- Cancer Center, the Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan, China.
| | - Yuanyuan Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong, China.
| |
Collapse
|
39
|
Siu LL, Postel-Vinay S, Villanueva-Vázquez R, de Velasco G, Castanon Alvarez E, Kyriakopoulos CE, Johnson M, Ouali K, McMorn S, Angell HK, Ng F, Saran S, Bayat M, Collins T, Roy A, Lambert AW, Cho S, Miller N, Petruzzelli M, Stone J, Massard C. AZD8701, an Antisense Oligonucleotide Targeting FOXP3 mRNA, as Monotherapy and in Combination with Durvalumab: A Phase I Trial in Patients with Advanced Solid Tumors. Clin Cancer Res 2025; 31:1449-1462. [PMID: 39937271 PMCID: PMC11995004 DOI: 10.1158/1078-0432.ccr-24-1818] [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: 06/12/2024] [Revised: 12/13/2024] [Accepted: 02/06/2025] [Indexed: 02/13/2025]
Abstract
PURPOSE AZD8701 uses next-generation antisense oligonucleotide (ASO) technology to selectively reduce human forkhead box P3 (FOXP3) expression in regulatory T cells, reversing their immunosuppressive function. FOXP3 ASO alone or with PD-(L)1 inhibition attenuated tumor growth in mice. We report a phase I study of AZD8701 alone or combined with durvalumab in patients with advanced solid tumors. PATIENTS AND METHODS Eligible patients had solid tumors and received prior standard-of-care treatment, including anti-PD-(L)1 therapy. Patient cohorts were treated with AZD8701 intravenously weekly at escalating doses, either alone (60-960 mg) or combined (240-720 mg) with durvalumab 1,500 mg intravenous every 4 weeks. The primary objective was safety and tolerability, with the aim of determining the MTD. RESULTS Forty-five patients received AZD8701 monotherapy, and 18 received AZD8701 with durvalumab. One dose-limiting toxicity (increased alanine aminotransferase) occurred with AZD8701 960 mg. The most common adverse events related to AZD8701 monotherapy were fatigue (22.2%), asthenia, pyrexia, and increased alanine aminotransferase (20% each); the safety profile was similar when combined with durvalumab. With AZD8701 monotherapy, 24.4% and 15.6% of the patients had stable disease for ≥16 and ≥24 weeks, respectively; one patient treated with AZD8701 720 mg and durvalumab had a partial response. FOXP3 mRNA changes were heterogeneous (8/13 patients showed a reduction), with no clear dose relationship. ASO accumulated in the tumor epithelium and stroma. CONCLUSIONS This study demonstrates the clinical feasibility of ASO therapy, with generally manageable adverse events, FOXP3 knockdown, and ASO delivery to the tumor.
Collapse
Affiliation(s)
| | | | - Rafael Villanueva-Vázquez
- Institut Català d'Oncologia, Early Drug Development Unit, Medical Oncology Department, ICO-Hospitalet, Barcelona, Spain
| | | | | | | | - Melissa Johnson
- Lung Cancer Research, Sarah Cannon Research Institute at Tennessee Oncology, Nashville, Tennessee
| | - Kaïssa Ouali
- Drug Development Department, Institut Gustave Roussy, Villejuif, France
| | | | | | | | | | | | | | | | | | - Song Cho
- AstraZeneca, Gaithersburg, Maryland
| | | | | | | | - Christophe Massard
- DITEP, Institut Gustave Roussy, Villejuif, France
- Faculty of Medicine, Paris Saclay University, Paris, France
- Molecular Radiotherapy Unit 1030, National Institute of Health and Medical Research (INSERM), Paris, France
| |
Collapse
|
40
|
Shigeta N, Murakami S, Yamamoto K, Yokose T, Isaka T, Washimi K, Miyagi Y, Saito H, Ito H, Saito A. Comparison of programmed death ligand-1 expression in preoperative transbronchial lung biopsy and resected specimens in non-small cell lung cancer. J Cancer Res Clin Oncol 2025; 151:139. [PMID: 40227460 PMCID: PMC11996979 DOI: 10.1007/s00432-025-06189-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/31/2025] [Indexed: 04/15/2025]
Abstract
PURPOSE Programmed death-ligand 1 expression is heterogeneous in non-small cell lung cancer, and small specimens may not accurately represent the entire tumor. The current study investigated the discordance in programmed death-ligand 1 expression between preoperative biopsy samples and resected specimens. METHODS We retrospectively collected data of patients with non-small cell lung cancer who underwent surgical resection from May 2022 to June 2024. The programmed death-ligand 1-positive tumor proportion score was evaluated for each case. RESULTS In total, 118 patients were included in this study. Programmed death-ligand 1 expression was discordant between the biopsy and resected specimens in 34 cases (28.8%), and it was underestimated in 25 (21.2%) biopsy specimens. The concordance according to Cohen's kappa was κ = 0.410 (95% confidence interval: 0.243-0.577). The number of discordant cases decreased as the number of tumor cells in biopsy specimens increased. In the group with > 400 tumor cells, agreement rate was 100%. The least absolute shrinkage and selection operator model identified never smoker, small tumor size, clinical stage II-IV and ≤ 200 tumor cells in biopsy specimens as predictors of underestimation. The area under the receiver operating characteristic curve using those four factors was 0.773 (0.663-0.884). CONCLUSIONS Programmed death-ligand 1 expression in biopsy and resected specimens is often discordant, often being underestimated in biopsy specimens. Discordance is more likely when tumor cell counts are low in the biopsy samples. Therefore, caution is advised when treatment decisions are made based on programmed death-ligand 1 assessments of small specimens.
Collapse
Affiliation(s)
- Naoko Shigeta
- Department of Thoracic Surgery, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan
| | - Shuji Murakami
- Department of Thoracic Oncology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan.
| | - Kouji Yamamoto
- Department of Biostatistics, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| | - Tomoyuki Yokose
- Department of Pathology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan
| | - Tetsuya Isaka
- Department of Thoracic Surgery, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan
| | - Kota Washimi
- Department of Pathology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan
| | - Haruhiro Saito
- Department of Thoracic Oncology, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan
| | - Hiroyuki Ito
- Department of Thoracic Surgery, Kanagawa Cancer Center, 2-3-2 Nakao, Asahi-ku, Yokohama, Kanagawa, 241-8515, Japan
| | - Aya Saito
- Department of Surgery, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, Kanagawa, 236-0004, Japan
| |
Collapse
|
41
|
Cui R, Su H, Jiang Y, Yu X, Liu Y. Propensity score analysis of high-dose rate brachytherapy, immune checkpoint inhibitors, and docetaxel in second-line advanced NSCLC treatment. Sci Rep 2025; 15:12650. [PMID: 40221605 PMCID: PMC11993689 DOI: 10.1038/s41598-025-97918-z] [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: 10/17/2024] [Accepted: 04/08/2025] [Indexed: 04/14/2025] Open
Abstract
This study evaluated the efficacy and safety of combining high-dose-rate brachytherapy, immune checkpoint inhibitors, and docetaxel as second-line treatment for advanced NSCLC, given the poor prognosis after first-line therapy. We conducted a single-center, retrospective, propensity score-matched study comparing HDR brachytherapy plus ICIs and docetaxel (study group) versus ICIs plus docetaxel (control group) in patients with advanced NSCLC who progressed after prior treatment without known driver gene mutations or uninvestigated mutation status. After propensity score matching, 21 patients were included in each group. The study group had a higher ORR (42.9% vs. 28.6%). Median OS was 18.6 months for the study group and 12.8 months for the control group (HR 0.45, 95% CI 0.20-0.85, P = 0.042). Median PFS was 8.6 vs. 5.6 months (HR 0.29, 95% CI 0.15-0.55, P < 0.001). The DCR was higher in the study group (71.4% vs. 61.9%). Treatment-related AEs were manageable, with no significant increase in grade 3/4 toxicities in the study group. Results suggest that combining high-dose rate brachytherapy, immune checkpoint inhibitors, and docetaxel may improve survival and response rates in advanced NSCLC after first-line therapy. Prospective randomized trials are necessary to confirm these findings and validate the strategy's effectiveness.
Collapse
Affiliation(s)
- Ran Cui
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan, China
| | - Hong Su
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan, China
| | - Yan Jiang
- Department of Gastroenterology, The People's Hospital of Longchang, Neijiang, Sichuan, China
| | - Xinlin Yu
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan, China
| | - Yu Liu
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan, China.
- Department of Oncology, The Second People's hospital of Neijiang, Neijiang, Sichuan, China.
| |
Collapse
|
42
|
Xiong X, Liu W, Yao C. Development of an alkaliptosis-related lncRNA risk model and immunotherapy target analysis in lung adenocarcinoma. Front Genet 2025; 16:1573480. [PMID: 40264452 PMCID: PMC12011837 DOI: 10.3389/fgene.2025.1573480] [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: 02/09/2025] [Accepted: 03/28/2025] [Indexed: 04/24/2025] Open
Abstract
Background Lung cancer has the highest mortality rate among all cancers worldwide. Alkaliptosis is characterized by a pH-dependent form of regulated cell death. In this study, we constructed a model related to alkaliptosis-associated long non-coding RNAs (lncRNAs) and developed a prognosis-related framework, followed by the identification of potential therapeutic drugs. Methods The TCGA database was utilized to obtain RNA-seq-based transcriptome profiling data, clinical information, and mutation data. We conducted multivariate Cox regression analysis to identify alkaliptosis-related lncRNAs. Subsequently, we employed the training group to construct the prognostic model and utilized the testing group to validate the model's accuracy. Calibration curves were generated to illustrate the discrepancies between predicted and observed outcomes. Principal Component Analysis (PCA) was performed to investigate the distribution of LUAD patients across high- and low-risk groups. Additionally, Gene Ontology (GO) and Gene Set Enrichment Analysis (GSEA) were conducted. Immune cell infiltration and Tumor Mutational Burden (TMB) analyses were carried out using the CIBERSORT and maftools algorithms. Finally, the "oncoPredict" package was employed to predict immunotherapy sensitivity and to further forecast potential anti-tumor immune drugs. qPCR was used for experimental verification. Results We identified 155 alkaliptosis-related lncRNAs and determined that 5 of these lncRNAs serve as independent prognostic factors. The progression-free survival (PFS) and overall survival (OS) rates of the low-risk group were significantly higher than those of the high-risk group. The risk signature functions as a prognostic factor that is independent of other variables. Different stages (I-II and III-IV) effectively predict the survival rates of lung adenocarcinoma (LUAD) patients, and these lncRNAs can reliably forecast these signatures. GSEA revealed that processes related to chromosome segregation and immune response activation were significantly enriched in both the high- and low-risk groups. The high-risk group exhibited a lower fraction of plasma cells and a higher proportion of activated CD4 memory T cells. Additionally, the OS of the low TMB group was significantly lower compared to the high TMB group. Furthermore, drug sensitivity was significantly greater in the high-risk group than in the low-risk group. These lncRNAs may serve as biomarkers for treating LUAD patients. Conclusion In summary, the construction of an alkaliptosis-related lncRNA prognostic model and drug sensitivity analysis in LUAD patients provides new insights into the clinical diagnosis and treatment of advanced LUAD patients.
Collapse
Affiliation(s)
| | | | - Chuan Yao
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, China
| |
Collapse
|
43
|
Srisawat W, Koonyosying P, Muenthaisong A, Sangkakam K, Varinrak T, Rittipornlertrak A, Nambooppha B, Apinda N, Sthitmatee N. mRNA and protein expression of programmed cell death-ligand-1 on canine mammary gland tumour in dogs of Chiang Mai, Thailand. Int J Vet Sci Med 2025; 13:1-11. [PMID: 40206791 PMCID: PMC11980185 DOI: 10.1080/23144599.2025.2483102] [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: 12/05/2024] [Revised: 02/18/2025] [Accepted: 03/04/2025] [Indexed: 04/11/2025] Open
Abstract
Metastasis-related disease is a major cause of death in canine mammary tumours (CMTs). Immunotherapy has been investigated due to the less successful outcomes of systemic therapy. This study aims to examine the expression of Programmed Cell Death Ligand-1 (PD-L1) in canine mammary tumours in dogs of Chiang Mai, Thailand, and determine the relationship between the level of mRNA expression and clinicopathologic characteristics. A total of 28 CMT samples were collected at the Small Animal Hospital, Chiang Mai University. Quantitative reverse transcriptase-polymerase chain reaction (RT-qPCR) and western blot assays were performed. The results revealed that all CMTs in this study expressed PD-L1 mRNA and PD-L1 protein. The mean relative mRNA expression showed no significant differences between groups categorized by age, tumour size, or histopathological findings. However, the mean relative mRNA expression in tumours with a TNM stage >3 was significantly lower compared to those with TNM stage ≤2. In conclusion, this study investigates the expression of PD-L1 mRNA and PD-L1 protein, particularly in malignant CMTs. The findings strongly support the potential for developing effective immunotherapy methods targeting the PD-1/PD-L1 pathway for advanced CMTs in the future. For further conclusive assessment, future studies should focus on refining immunotherapy strategies for CMT cases expressing PD-L1.
Collapse
Affiliation(s)
- Wanwisa Srisawat
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Multidisciplinary Research Institute, Chiang Mai University, Chiang Mai, Thailand
| | - Pongpisid Koonyosying
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Office of Research Administration, Chiang Mai University, Chiang Mai, Thailand
| | - Anucha Muenthaisong
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Office of Research Administration, Chiang Mai University, Chiang Mai, Thailand
| | - Kanokwan Sangkakam
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Thanya Varinrak
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Center of Veterinary Medical Diagnostic and Animal Health Innovation, Chiang Mai University, Chiang Mai, Thailand
| | - Amarin Rittipornlertrak
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Boondarika Nambooppha
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nisachon Apinda
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nattawooti Sthitmatee
- Laboratory of Veterinary Vaccine and Biological Products, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Research Center for Veterinary Bioscience and Veterinary Public Health, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
44
|
Xie D, Yu J, He C, Jiang H, Qiu Y, Fu L, Kong L, Xu H. Predicting the immune therapy response of advanced non-small cell lung cancer based on primary tumor and lymph node radiomics features. Front Med (Lausanne) 2025; 12:1541376. [PMID: 40248083 PMCID: PMC12003267 DOI: 10.3389/fmed.2025.1541376] [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: 12/07/2024] [Accepted: 03/20/2025] [Indexed: 04/19/2025] Open
Abstract
Objective To identify imaging biomarkers of primary tumors and lymph nodes in patients with stage III-IV non-small cell lung cancer (NSCLC) and assess their predictive ability for treatment response (response vs. non-response) to immune checkpoint inhibitors (ICIs) after 6 months. Methods Retrospective analysis of 83 NSCLC patients treated with ICIs. Quantitative imaging features of the maximum primary lung tumors and lymph nodes on contrast-enhanced CT imaging were extracted at baseline (time point 0, TP0) and after 2-3 cycles of immunotherapy (time point 1, TP1). Delta-radiomics features (delta-RFs) were defined as the net changes in radiomics features (RFs) between TP0 and TP1. Interobserver interclass coefficient (ICC) and Pearson correlation analyses were applied for feature selection, and logistic regression (LR) was used to build a model for predicting treatment response. Results Four and five important delta-RFs were selected to construct the nodal and tumor models, respectively. Δ Tumor diameter was used for constructing the clinical prediction model. The predictive efficacy of the nodal model for the treatment response status was higher than that of the tumor and clinical models. In the training set, the AUC values for the three models were 0.96 (95% CI = 0.90-1.00), 0.86 (95% CI = 0.76-0.95), and 0.82 (95% CI = 0.71-0.93), respectively. In the validation set, the AUC values were 0.94 (95% CI = 0.85-1.00), 0.77 (95% CI = 0.56-0.98), and 0.74 (95% CI = 0.48-1.00), respectively. Conclusion The nodal model based on delta-RFs performed well in distinguishing responders from non-responders and could identify patients more likely to benefit from immunotherapy. Finally, the nodal model exhibited a higher classification performance than the tumor model.
Collapse
Affiliation(s)
- Dong Xie
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Jinna Yu
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Cong He
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Han Jiang
- Department of Medical Oncology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Yonggang Qiu
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Linfeng Fu
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Lingting Kong
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| | - Hongwei Xu
- Department of Radiology, Shaoxing Second Hospital Medical Community General Hospital, Shaoxing, China
| |
Collapse
|
45
|
Cho BC, Johnson M, Bar J, Schaefer E, Yoh K, Zer A, Moskovitz M, Lee SH, Moreno V, de Miguel M, Okuma Y, Kim JH, Lee CH, Peguero J, Ansell P, Biesdorf C, Saab R, Freise KJ, Ramies D, Jeng EE, Camidge DR. A phase 1b study of cofetuzumab pelidotin monotherapy in patients with PTK7-expressing recurrent non-small cell lung cancer. Lung Cancer 2025; 202:108492. [PMID: 40086026 DOI: 10.1016/j.lungcan.2025.108492] [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: 10/24/2024] [Revised: 02/24/2025] [Accepted: 03/08/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND Protein tyrosine kinase 7 (PTK7) overexpression in lung cancer is associated with tumor progression. Cofetuzumab pelidotin (Cofe-P) is an antibody-drug conjugate comprising an anti-PTK7 antibody conjugated to a microtubule inhibitor. Herein, we report the results of a phase 1b study evaluating Cofe-P safety, efficacy, and pharmacokinetics in patients with recurrent non-small cell lung cancer (NSCLC). METHODS This signal-seeking phase 1b, open-label, multicenter, single-arm study enrolled patients with PTK7-expressing recurrent NSCLC. Cofe-P was administered at 2.8 mg/kg intravenously once every 3 weeks. The primary endpoint was objective response rate (ORR) and secondary endpoints were duration of response (DOR), progression-free survival (PFS), and overall survival (OS). RESULTS Overall, 65 patients received Cofe-P; 51 had PTK7 expression of ≥90 % tumor cells with ≥2+ staining intensity by immunohistochemistry. All patients reported adverse events (AEs), most commonly alopecia (52 %) and decreased neutrophil count (43 %); 69 % had grade ≥3 AEs, including grade ≥3 neutropenia in 25 %. Treatment-related AEs were reported in 94 % of patients; none were fatal. Overall, ORR was 18 %; in patients with PTK7 expression of ≥90 % tumor cells with ≥2+ staining and non-squamous epidermal growth factor receptor (EGFR) wild type or mutant, or squamous NSCLC, ORR was 21 %, 15 %, and 13 %, respectively. Overall, median DOR was 7.2 months, median PFS was 5.5 months, and median OS was 12.6 months; longest DOR (median 9.0 months), PFS (median 6.8 months), and OS (median 12.6 months) were in patients with non-squamous EGFR-mutant NSCLC. CONCLUSIONS Cofe-P demonstrated a manageable safety profile and preliminary efficacy across NSCLC histologies and EGFR mutation status. These data support PTK7 as a valid therapeutic target for NSCLC.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/metabolism
- Male
- Female
- Middle Aged
- Aged
- Lung Neoplasms/drug therapy
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Lung Neoplasms/metabolism
- Adult
- Neoplasm Recurrence, Local/drug therapy
- Neoplasm Recurrence, Local/pathology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Cell Adhesion Molecules/metabolism
- Receptor Protein-Tyrosine Kinases/metabolism
- Receptor Protein-Tyrosine Kinases/antagonists & inhibitors
- Aged, 80 and over
- Immunoconjugates/therapeutic use
- Immunoconjugates/pharmacokinetics
- Immunoconjugates/adverse effects
- Treatment Outcome
- Antigens, Neoplasm/metabolism
Collapse
Affiliation(s)
- Byoung Chul Cho
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea.
| | | | - Jair Bar
- Jusidman Cancer Center, Institute of Oncology, Sheba Medical Center, Ramat Gan, Israel.
| | - Eric Schaefer
- Section of Medical Oncology, Highlands Oncology Group, Fayetteville, AR, USA.
| | - Kiyotaka Yoh
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| | - Alona Zer
- Oncology Institute, Rambam Health Care Campus, Rappaport Faculty of Medicine, Technion, Haifa, Israel.
| | - Mor Moskovitz
- Thoracic Cancer Service, Rabin Medical Center Davidoff Cancer Center, Beilinson Campus, Petah Tikva, Israel.
| | - Se-Hoon Lee
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea.
| | - Victor Moreno
- START Madrid-FJD, Medical Oncology Department, University Hospital Fundación Jiménez Diaz, Instituto de Investigación Sanitaria FJD, Madrid, Spain.
| | - Maria de Miguel
- START Madrid HM CIOCC, Instituto de Investigación Sanitaria HM Hospitales, Universidad San Pablo CEU, Madrid, Spain.
| | - Yusuke Okuma
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Joo-Hang Kim
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Republic of Korea.
| | - Chun-Hui Lee
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Hu X, Rodday AM, Gurinovich A, Pan S, Salei YV, Lin JH, Byrne MM, Cao Y, Pai L, Parsons SK. Real-world data of immune-related adverse events in lung cancer patients receiving immune-checkpoint inhibitors. Immunotherapy 2025; 17:321-329. [PMID: 40183219 PMCID: PMC12045565 DOI: 10.1080/1750743x.2025.2488728] [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: 07/18/2024] [Accepted: 04/01/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Immune-checkpoint inhibitors (ICIs) have revolutionized lung cancer (LC) treatment; however, immune-related adverse effects (irAEs) may occur. The risk factors of irAEs and the impact of irAEs on patient outcomes in LC remain uncertain. MATERIALS AND METHODS irAEs within 12 months of ICI initiation in LC patients who initiated ICIs 2018-2021 were identified. Cause-specific Cox regression was used to assess risk factors for irAEs with the competing risk of death; a subset analysis was done among non-small cell lung cancer (NSCLC) group. Multivariable Cox regressions were used to evaluate the impact of irAEs on progression-free survival (PFS) and overall survival (OS). RESULTS Of 125 patients, 50 irAEs occurred in 39 patients. Small cell lung cancer (SCLC) histology was associated with a higher risk of irAEs (Hazard ratio (HR) = 2.73, 95% CI [1.17, 6.35], p = 0.020) than NSCLC. In NSCLC subset, programmed death-ligand 1 (PDL1) positivity (HR = 2.68, 95% CI [1.10. 6.53], p = 0.030) was identified as a risk factor. irAEs were not significantly associated with PFS (HR = 0.69, p = 0.204) or OS (HR = 0.72, p = 0.353). CONCLUSION SCLC histology and PDL1 positivity were associated with irAEs, and the occurrence of irAEs showed no impact on survival in LC patients. Future studies are required to validate the findings.
Collapse
Affiliation(s)
- Xiao Hu
- Division of Hematology-Oncology, Tufts Medical Center, Boston, MA, USA
- Department of Medicine, Maine Medical Center, Portland, ME, USA
- Center for Interdisciplinary Population and Health Research, MaineHealth Institute for Research, Portland, ME, USA
| | - Angie Mae Rodday
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, USA
| | - Anastasia Gurinovich
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, USA
| | - Stacey Pan
- Division of Hematology-Oncology, Tufts Medical Center, Boston, MA, USA
| | - Yana V. Salei
- Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Jeffrey H. Lin
- Department of Medicine, Tufts Medical Center, Boston, MA, USA
| | - Margaret M. Byrne
- Department of Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Yu Cao
- Division of Hematology-Oncology, Tufts Medical Center, Boston, MA, USA
| | - Lori Pai
- Division of Hematology-Oncology, Tufts Medical Center, Boston, MA, USA
| | - Susan K. Parsons
- Division of Hematology-Oncology, Tufts Medical Center, Boston, MA, USA
- Institute for Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, MA, USA
| |
Collapse
|
47
|
Nishimyo K, Ikeda S, Fushimi K, Yamazaki T, Ishikawa KB. Systemic treatment patterns and adherence to guidelines in Japanese patients with metastatic non-small cell lung cancer. Future Oncol 2025; 21:1101-1111. [PMID: 40018807 PMCID: PMC11988220 DOI: 10.1080/14796694.2025.2470611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND Non-small-cell-lung cancer (NSCLC) medication use and guideline adherence remain unclear. We investigated treatment patterns and adherence among Japanese patients with NSCLC. RESEARCH DESIGN AND METHODS We analyzed treatment patterns and guideline adherence by age and histology in ≥ 20-year-olds with stage IV NSCLC treated between 2016-2018 using diagnostic procedure combination data. Logistic regression analysis evaluated the impact of various factors on guideline adherence. RESULTS We included 9,722 patients. In < 75-year-olds with nonsquamous NSCLC, first-to third-line treatments comprised 31.8% platinum combination therapy, 26.3% immune checkpoint inhibitors, and 62.5% cytotoxic chemotherapy. In ≥ 75-year-olds, first-line and second-line molecular targeted therapies represented 46.6% and 35.6%, whereas third-line cytotoxic chemotherapy represented 42.3%. In squamous NSCLC, first-line platinum combination therapy was predominant (69.7% and 47.7% for < 75-and ≥75-year-olds). The most common second-line and third-line therapies were immune checkpoint inhibitors (48.6% and 50.8% for < 75-and ≥75-year-olds) and cytotoxic chemotherapy (62.5% and 55.2% for < 75-and ≥75-year-olds), respectively. The highest guideline adherence (90%) was in < 75-year-olds with squamous NSCLC. Age, histology, activities of daily living, and cumulative hospitalizations over the past 18 months influenced treatment adherence. CONCLUSION New NSCLC drug introduction increased regardless of age, suggesting prognosis improvement. More efficient drug application and broader guideline dissemination are required.
Collapse
Affiliation(s)
- Keiko Nishimyo
- Department of Social Medical Sciences, Graduate School of Medicine, International University of Health and Welfare, Minato-ku, Tokyo, Japan
| | - Shunya Ikeda
- Department of Social Medical Sciences, Graduate School of Medicine, International University of Health and Welfare, Minato-ku, Tokyo, Japan
| | - Kiyohide Fushimi
- Department of Health Policy and Informatics, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Tsutomu Yamazaki
- Department of Social Medical Sciences, Graduate School of Medicine, International University of Health and Welfare, Minato-ku, Tokyo, Japan
| | - Koichi B Ishikawa
- Department of Social Medical Sciences, Graduate School of Medicine, International University of Health and Welfare, Minato-ku, Tokyo, Japan
| |
Collapse
|
48
|
Okui J, Nagashima K, Matsuda S, Sato Y, Kawakubo H, Takeuchi M, Hirata K, Yamamoto S, Nomura M, Tsushima T, Takeuchi H, Kato K, Kitagawa Y. Investigating the synergistic effects of immunochemotherapy in esophageal squamous cell carcinoma. Esophagus 2025; 22:188-197. [PMID: 39966261 DOI: 10.1007/s10388-025-01113-y] [Citation(s) in RCA: 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: 09/30/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Although combinations of immune-checkpoint inhibitors (ICI) with chemotherapy have been approved for esophageal squamous cell carcinoma (ESCC), it remains unclear whether immunochemotherapy (ICT) offers advantages over the simple addition of individual monotherapies. This study aimed to investigate whether ICT exhibits a synergistic effect in patients with advanced ESCC. METHODS Reconstructed individual patient data of 3330 patients were electronically extracted from the Kaplan-Meier (KM) curves of eight randomized-controlled trials (ATTRACTION-3, CheckMate648, KEYNOTE-181, KEYNOTE-590, RATIONALE-302, RATIONALE-306, ESCORT, and ESCORT-1st). The observed progression-free survival (PFS) curve of each constituent monotherapies was used to estimate simulated PFS curves expected under a model of independent drug action. If the observed curve demonstrated significantly better PFS than the simulated curve, the combination of ICI and chemotherapy may have a synergistic effect, implying a superior outcome compared to simply adding the component monotherapy. RESULTS The 1-year, 2-year, and median PFS of the observed and simulated KM curves were 26.3% vs. 24.8%, 14.6% vs. 12.0%, and 6.9 vs. 6.4 months, respectively. The one-sample log-rank test showed no significant differences between the observed and simulated KM curves (p = 0.073). CONCLUSIONS The observed PFS with ICT was comparable to the simulated PFS estimated from the data for each monotherapy. Although it is unclear whether potential synergies exist for ICT, these findings suggest that the benefits of ICI and chemotherapy do not interfere with each other, thereby providing theoretical support for the efficacy of ICT.
Collapse
Affiliation(s)
- Jun Okui
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
- Department of Biostatistics, Keio University School of Medicine, Tokyo, Japan
| | - Kengo Nagashima
- Biostatistics Unit, Clinical and Translational Research Center, Keio University Hospital, Tokyo, Japan
| | - Satoru Matsuda
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan.
| | - Yasunori Sato
- Department of Biostatistics, Keio University School of Medicine, Tokyo, Japan
| | - Hirofumi Kawakubo
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Masashi Takeuchi
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Kenro Hirata
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Shun Yamamoto
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Motoo Nomura
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Takahiro Tsushima
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Hiroya Takeuchi
- Department of Surgery, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Ken Kato
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
49
|
Zhang W, Wang J, Liang J, He Z, Wang K, Lin H. RNA methylation of CD47 mediates tumor immunosuppression in EGFR-TKI resistant NSCLC. Br J Cancer 2025; 132:569-579. [PMID: 39900985 PMCID: PMC11920402 DOI: 10.1038/s41416-025-02945-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Although immune checkpoint inhibitors (ICIs) have been successfully utilized in patients with non-small cell lung cancer (NSCLC), EGFR-mutated patients didn't benefit from ICIs. The underlying mechanisms for the poor efficacy of this subgroup remain unclear. METHODS CD8+T cells cytotoxicity, DCs phagocytosis and immunofluorescence assay were applied to examine the immunosuppressive microenvironment of NSCLC. m6A RNA immunoprecipitation, luciferase assay and immunohistochemistry were used to explore the relationship between CD47 and ALKBH5 in EGFR-TKI resistant NSCLC. Autochthonous EGFR-driven lung tumor mouse model and PDXs were performed to explore the therapeutic potential of CD47 antibody and EGFR-TKI combination. RESULTS We found that EGFR-TKI resistance promoted a more immunosuppressive tumor microenvironment and inhibited anti-tumor functions of CD8+ T cells. Mechanistically, the m6A eraser ALKBH5 was inhibited in EGFR-TKI resistant NSCLC, which subsequently upregulates CD47 by catalyzing m6A demethylation and causes immunosuppression. Combined treatment with EGFR-TKI and inhibitors of CD47 enhances antitumor immunity and EGFR-TKI efficacy in vivo. CONCLUSIONS Collectively, our findings reveal the possible underlying mechanism for poor immune response of ICIs in EGFR-TKI resistant NSCLC and provide preclinical evidence that targeted therapy combined with innate immune checkpoint blockade may provide synergistic effects in NSCLC treatment.
Collapse
Affiliation(s)
- Wei Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiawen Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jialu Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhanghai He
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- Department of Pathology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Kefeng Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Department of Thoracic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Huayue Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
- Breast Tumor Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
50
|
Huangfu R, Hou K, Zhao J, Wu S, Ping Y. Toward the benefit and value of immune treatment beyond progression in lung cancer? Insights from a systematic review and meta-analysis. Front Immunol 2025; 16:1547978. [PMID: 40236709 PMCID: PMC11996641 DOI: 10.3389/fimmu.2025.1547978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/13/2025] [Indexed: 04/17/2025] Open
Abstract
Objective Immune treatment beyond progression (ITBP) has emerged as a novel therapeutic strategy in oncology. This systematic review and meta-analysis aim to evaluate the efficacy and safety of ITBP in patients with lung cancer, while also identifying characteristics of populations that may benefit most from this treatment approach. Methods This study adheres to the PRISMA guidelines. We searched PubMed, Embase, and the Cochrane Library for relevant literature on immunotherapy for lung cancer, using self-constructed databases up until February 1, 2024. The study includes real-world data from patients with lung cancer undergoing ITBP, categorized into two groups: non-ITBP (NTBP) and ITBP. Two authors independently conducted literature screening, quality assessment, and data extraction. The primary efficacy indicators include overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and disease control rate (DCR). The safety indicator assessed was the incidence of immune-related adverse events (irAEs). Results We included 9 studies with a total of 5,141 patients with lung cancer, comprising 2,051 patients in the ITBP group and 3,090 in the NTBP group. Patients receiving ITBP showed significantly better outcomes than those receiving NTBP, including superior OS and PFS following treatment beyond progression (OS: hazard ratio (HR) 0.72, 95% confidence interval (CI) 0.68-0.77, P < 0.05; PFS: HR 0.63, 95% CI 0.51-0.78, P < 0.05). Additionally, the ITBP group demonstrated higher ORR and DCR (ORR: odds ratio (OR) 0.48, 95% CI 0.31-0.75, P < 0.05; DCR: OR 0.37, 95% CI 0.24-0.57, P < 0.05). No significant difference in the incidence of irAEs was found between the two groups (OR 1.24, 95% CI 0.83-1.85, P > 0.05). Subgroup analysis revealed that factors such as age, gender, lung cancer subtype, and smoking history significantly influenced OS outcomes in the ITBP group. Conclusion Our findings suggest that ITBP is an effective treatment strategy for patients with lung cancer. Further research should focus on identifying specific patient populations that benefit from ITBP and exploring the potential efficacy of combining ITBP with other therapeutic regimens. Systematic Review Registration https://www.crd.york.ac.uk/prospero/, identifier CRD42024513475.
Collapse
Affiliation(s)
- Rui Huangfu
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Kun Hou
- Department of Pharmacy, Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, China
| | - Jie Zhao
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Shikui Wu
- School of Pharmacy, Inner Mongolia Medical University, Hohhot, China
| | - Yaodong Ping
- Department of Pharmacy, Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, China
- Department of Pharmacy, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| |
Collapse
|