1
|
Zhou Z, Yang Y, Chen S, You M. Cost-effectiveness analysis of first-line cadonilimab plus chemotherapy in HER2-negative advanced gastric or gastroesophageal junction adenocarcinoma. Front Immunol 2025; 16:1575627. [PMID: 40433373 PMCID: PMC12106304 DOI: 10.3389/fimmu.2025.1575627] [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: 02/12/2025] [Accepted: 04/24/2025] [Indexed: 05/29/2025] Open
Abstract
Background The COMPASSION-15 trial demonstrated that cadonilimab plus chemotherapy (CAD-CHM) confers clinical benefits over placebo plus chemotherapy (PLA-CHM) as a first-line treatment for human epidermal growth factor receptor 2 (HER2)-negative advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma. However, the introduction of cadonilimab substantially elevates treatment costs, and its cost-effectiveness relative to PLA-CHM remains undetermined. This study evaluates the cost-effectiveness of CAD-CHM compared with PLA-CHM from the perspective of the Chinese healthcare system. Methods A Markov model with three health states was developed to assess the cost-effectiveness of CAD-CHM in HER2-negative advanced G/GEJ adenocarcinoma. Clinical efficacy data were sourced from the COMPASSION-15 trial, while drug costs were calculated based on national tender prices, and additional costs and utility values were extracted from published literature. The analysis encompassed the overall population, as well as subgroups stratified by programmed death ligand 1 (PD-L1) combined positive score (CPS) ≥ 5 and CPS < 5. Outcomes included total costs, quality-adjusted life-years (QALYs), and incremental cost-effectiveness ratios (ICERs). Sensitivity analyses were conducted to evaluate model robustness. Results The ICER of CAD-CHM was $67,378.09 per QALY in the overall population, $48,433.34 per QALY in the PD-L1 CPS ≥ 5 subgroup, and $78,463.86 per QALY in the PD-L1 CPS < 5 subgroup. Key determinants influencing model outcomes included patient weight, cadonilimab cost, and the utility value of progression-free survival. Across all groups, CAD-CHM resulted in an ICER exceeding the willingness-to-pay threshold of $41,511 per QALY, with a 0% probability of cost-effectiveness compared with PLA-CHM. Conclusion From the perspective of the Chinese healthcare system, CAD-CHM is not cost-effective as a first-line treatment for HER2-negative advanced G/GEJ adenocarcinoma, either in the overall population or in subgroups stratified by PD-L1 CPS status, compared with chemotherapy alone.
Collapse
Affiliation(s)
- Zhifeng Zhou
- Department of Pharmacy, Fuzhou University Affiliated Provincial Hospital, Fuzhou, Fujian, China
| | - Yanqing Yang
- Department of Clinical Nutrition, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou, Fujian, China
| | - Shaofang Chen
- Department of Pharmacy, Mindong Hospital Affiliated to Fujian Medical University, Ningde, Fujian, China
| | - Maojin You
- Department of Pharmacy, Mindong Hospital Affiliated to Fujian Medical University, Ningde, Fujian, China
| |
Collapse
|
2
|
Peng C, Gong C, Zhang X, Liu D. A prognostic model for highly aggressive prostate cancer using interpretable machine learning techniques. Front Med (Lausanne) 2025; 12:1512870. [PMID: 40421291 PMCID: PMC12104253 DOI: 10.3389/fmed.2025.1512870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 04/21/2025] [Indexed: 05/28/2025] Open
Abstract
Background Extremely aggressive prostate cancer, including subtypes like small cell carcinoma and neuroendocrine carcinoma, is associated with poor prognosis and limited treatment options. This study sought to create a robust, interpretable machine learning-based model that predicts 1-, 3-, and 5-year survival in patients with extremely aggressive prostate cancer. Additionally, we sought to pinpoint key prognostic factors and their clinical implications through an innovative method. Materials and methods This study retrospectively analyzed data from 1,620 patients with extremely aggressive prostate cancer in the SEER database (2000-2020). Feature selection was performed using the Boruta algorithm, and survival predictions were made using nine machine learning algorithms, including XGBoost, logistic regression (LR), support vector machine (SVM), random forest (RF), k-nearest neighbor (KNN), decision tree (DT), elastic network (Enet), multilayer perceptron (MLP) and lightGBM. Model performance was evaluated using metrics such as AUC, accuracy (F1 score), confusion matrix, and decision curve analysis. Additionally, Shapley Additive Explanations (SHAP) were applied to interpret feature importance within the model, revealing the clinical factors that influence survival predictions. Results Among the nine models, the lightGBM model exhibited the best performance, with an AUC and F1 score of (0.8, 0.809) for 1-year survival prediction, (0.809, 0.751) for 3-year survival prediction, and (0.773, 0.611) for 5-year survival prediction. SHAP analysis revealed that M stage was the most important feature for predicting 1- and 3-year survival, while PSA level had the greatest impact on 5-year survival predictions. The model demonstrated good clinical utility and predictive accuracy through decision curve analysis and confusion matrix. Conclusion The lightGBM model has good predictive power for survival in patients with extremely aggressive prostate cancer. By identifying key clinical factors and providing actionable predictions, the model has the potential to enhance prognostic accuracy and improve patient outcomes.
Collapse
Affiliation(s)
| | | | | | - Duxian Liu
- Department of Pathology, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
3
|
Zhang H, Xia L, Xuzhang W, Li Z, Zhang J, Li F, Cheng C, Wang J, Zong X, Yang X, Lu S. BCL-2 mutant B7H6-CAR-T cells synergized with venetoclax for treating small cell lung cancer. J Immunother Cancer 2025; 13:e010073. [PMID: 40341023 PMCID: PMC12067830 DOI: 10.1136/jitc-2024-010073] [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: 07/10/2024] [Accepted: 04/18/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Patients with small cell lung cancer (SCLC) generally have a poor prognosis, with an exceptionally high proliferative rate and a strong propensity for early metastasis, indicating the urgent need for novel therapies. The development of chimeric antigen receptor (CAR)s targeting solid tumors is limited owing to the lack of target antigens and low efficacy. In this study, we aimed to discover new targets for SCLC CAR-T therapy and develop CAR-T-based combinational treatment against SCLC in preclinical models. METHODS The in vitro antitumor activity of B7H6-specific CAR-T cell was evaluated. Venetoclax-resistant B7H6 CAR-T cell were designed and the synergistic effect of venetoclax and B7-H6 CAR-T cells was tested in vitro and in vivo. RESULT B7H6 is highly expressed in SCLC tumors. CAR-T cell against B7H6 displayed antigen-specific antitumor efficacy. BCL-2(D103E)-expressing CAR-T cells showed resistance to venetoclax-induced apoptosis. The combinational treatment of venetoclax and BCL-2(D103E)-expressing B7H6-targeting showed potent anti-SCLC effect in vitro and in vivo. CONCLUSIONS Our findings suggest that the combination of BCL-2 mutant-expressing B7H6-targeting CAR-T cells and venetoclax could be a promising novel strategy against B7H6-expressing SCLCs and other solid tumors, providing the foundation for CAR-T cells and proapoptotic small molecules therapy in patients with SCLCs in a clinical trial.
Collapse
Affiliation(s)
- Huihui Zhang
- Shanghai Lung Cancer Center, Shanghai Key Laboratory of Thoracic Tumor Biotherapy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Liliang Xia
- Shanghai Lung Cancer Center, Shanghai Key Laboratory of Thoracic Tumor Biotherapy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wendi Xuzhang
- Shanghai Lung Cancer Center, Shanghai Key Laboratory of Thoracic Tumor Biotherapy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ziming Li
- Shanghai Lung Cancer Center, Shanghai Key Laboratory of Thoracic Tumor Biotherapy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junshi Zhang
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Fanlin Li
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Chen Cheng
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jiawen Wang
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xincheng Zong
- Faculty of Art & Science, University of Toronto, Toronto, Ontario, Canada
| | - Xuanming Yang
- Shanghai Lung Cancer Center, Shanghai Key Laboratory of Thoracic Tumor Biotherapy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Sheng Yushou Center of Cell Biology and Immunology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
- Joint International Research Laboratory of Metabolic and Developmental Sciences, Shanghai Jiao Tong University, Shanghai, China
- Department of Gynaecology and Obstetrics, Shanghai Pudong New Area people's Hospital, Shanghai, China
| | - Shun Lu
- Shanghai Lung Cancer Center, Shanghai Key Laboratory of Thoracic Tumor Biotherapy, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Dhaeyer S, Missault E, Surmont V, Vermaelen K, Stevens D. Outcome of temozolomide in relapsed small cell lung cancer: A retrospective single center analysis. Lung Cancer 2025; 203:108539. [PMID: 40279759 DOI: 10.1016/j.lungcan.2025.108539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/17/2025] [Accepted: 04/11/2025] [Indexed: 04/29/2025]
Abstract
OBJECTIVES Extensive-stage small cell lung cancer (SCLC) has a dismal prognosis. Despite initial responsiveness to first-line platinum-etoposide chemotherapy, most patients relapse within six months. Managing disease progression, particularly in platinum-resistant or refractory cases, remains challenging. Topotecan is the only drug approved in the European Union for the second-line treatment of SCLC but is associated with modest clinical activity and high rates of hematological toxicities. Temozolomide, an oral alkylating agent, has been investigated as a viable alternative for treating relapsed SCLC. This study presents the largest real-world cohort of SCLC-patients treated with temozolomide. METHODS We performed a retrospective analysis of patients with relapsed SCLC treated with temozolomide at a single academic hospital in Belgium. Temozolomide was administered at a fixed dose of 250 mg orally once daily on days 1-5 of each 28-day cycle. Data on activity (overall response rate (ORR), disease control rate (DCR), median progression-free survival (mPFS) and median overall survival (mOS)) and safety (treatment related adverse events (TRAE)) were collected. RESULTS Between February 2011 and May 2023, a total of 48 patients with relapsed SCLC were treated with temozolomide of which 47 patients, median age 61 years, were included in this real-world analysis. The majority of the patients were heavily pretreated with 57.4 % having received two or more prior systemic therapies. An objective response was observed in 14.9 % and the DCR was 23.4 %. The median PFS was 1.7 months (95 % CI 1.5-1.9) and the median OS was 3.2 months (95 % CI 2.3-4.1). Grade 3-4 TRAEs occurred in 34 % of the patients. CONCLUSIONS Temozolomide demonstrated modest clinical activity in this real-world effectiveness analysis of patients with relapsed SCLC. Nevertheless, given its comparable response rate and milder toxicity profile compared to topotecan, temozolomide should be considered as a viable alternative to topotecan for treating relapsed SCLC.
Collapse
Affiliation(s)
- Sofie Dhaeyer
- Department of Internal Medicine, Ghent University Hospital, Ghent, Belgium
| | - Elise Missault
- Department of Internal Medicine, Ghent University Hospital, Ghent, Belgium
| | - Veerle Surmont
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Karim Vermaelen
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Dieter Stevens
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium.
| |
Collapse
|
5
|
Stewart DJ, Cole K, Brule S. A Population Survival Kinetics Assessment of Extensive Small Cell Lung Cancer and Rationale for Maintenance Therapy. Curr Oncol 2025; 32:258. [PMID: 40422517 DOI: 10.3390/curroncol32050258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/21/2025] [Accepted: 04/27/2025] [Indexed: 05/28/2025] Open
Abstract
Progression-free survival (PFS) and overall survival (OS) curves generally approximate first-order kinetics. On log-linear plots, convex curves with downward inflection (indicating late acceleration of progression/death) might arise from stopping effective therapies. We digitized published PFS/OS curves for etoposide/platinum-treated extensive small-cell lung cancer (SCLC) and other malignancies and replotted the curves log-linearly. Of 26 SCLC PFS curves, 21 (81%) were highly convex (with a marked late down-turn), and 26 (100%) were moderately or highly convex vs. 35/888 (4%) highly convex and 186 (21%) moderately/highly convex curves for other cancers (p < 0.0001). For SCLC, all 32 OS curves were moderately or highly convex vs. 87/363 (24%) that were moderately/highly convex for other cancers (p < 0.0001). The SCLC PFS curves had an initial downward inflection at a median of 3.1 months (around the completion of first-line chemotherapy), then a second inflection at 5.4 months, with further acceleration of progression. The median PFS half-life was 11.9 months while receiving treatment vs. 1.7 months after the second inflection point. Immunotherapy benefit appeared to be limited to 6-10% of the population. SCLC PFS/OS curves are more often convex than for other cancers, reflecting SCLC chemotherapy sensitivity but rapid progression following the completion of first-line chemotherapy. Effective maintenance strategies are needed.
Collapse
Affiliation(s)
- David J Stewart
- Faculty of Medicine, Department of Medicine, Division of Medical Oncology, University of Ottawa, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
| | - Katherine Cole
- Faculty of Medicine, Department of Medicine, Division of Medical Oncology, University of Ottawa, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
| | - Stephanie Brule
- Faculty of Medicine, Department of Medicine, Division of Medical Oncology, University of Ottawa, 501 Smyth Road, Ottawa, ON K1H 8L6, Canada
| |
Collapse
|
6
|
Das S, Zea MP, Russon MP, Xing Z, Torregrosa-Allen S, Cervantes HE, Harper HA, Elzey BD, Tran EJ. Supinoxin blocks small cell lung cancer progression by inhibiting mitochondrial respiration through DDX5. iScience 2025; 28:112219. [PMID: 40224004 PMCID: PMC11987007 DOI: 10.1016/j.isci.2025.112219] [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: 08/07/2024] [Revised: 12/26/2024] [Accepted: 03/11/2025] [Indexed: 04/15/2025] Open
Abstract
DDX5 is a DEAD-box RNA helicase that is overexpressed and implicated in the progression of several cancers, including small cell lung cancer (SCLC). Our laboratory has demonstrated that DDX5 is essential for the invasive growth of SCLC and mitochondrial respiration. SCLC is an extremely lethal, recalcitrant tumor, and currently lacking effective treatments. Supinoxin (RX 5902), a compound having anti-cancer activity, is a known target of phosphor-DDX5. We now report that Supinoxin inhibits the proliferation of chemo-sensitive and chemo-resistant SCLC lines, H69 and H69AR, respectively. Additionally, Supinoxin mitigates both the growth of H69AR xenograft tumors and SCLC PDX tumors in vivo. Finally, we find that Supinoxin inhibits expression of mitochondrial genes and effectively blocks respiration. These studies suggest that Supinoxin functions in anti-tumor progression by reducing cellular energy levels through DDX5.
Collapse
Affiliation(s)
- Subhadeep Das
- Department of Biochemistry, Purdue University, BCHM A343, 175 S. University Street, West Lafayette, IN 47907-2063, USA
- Purdue University Institute for Cancer Research, Purdue University, Hansen Life Sciences Research Building, Room 141, 201 S. University Street, West Lafayette, IN 47907-2064, USA
| | - Maria P. Zea
- Department of Biochemistry, Purdue University, BCHM A343, 175 S. University Street, West Lafayette, IN 47907-2063, USA
| | - Matthew P. Russon
- Department of Biochemistry, Purdue University, BCHM A343, 175 S. University Street, West Lafayette, IN 47907-2063, USA
| | - Zheng Xing
- Department of Biochemistry, Purdue University, BCHM A343, 175 S. University Street, West Lafayette, IN 47907-2063, USA
| | - Sandra Torregrosa-Allen
- Purdue University Institute for Cancer Research, Purdue University, Hansen Life Sciences Research Building, Room 141, 201 S. University Street, West Lafayette, IN 47907-2064, USA
| | - Heidi E. Cervantes
- Purdue University Institute for Cancer Research, Purdue University, Hansen Life Sciences Research Building, Room 141, 201 S. University Street, West Lafayette, IN 47907-2064, USA
| | - Haley Anne Harper
- Purdue University Institute for Cancer Research, Purdue University, Hansen Life Sciences Research Building, Room 141, 201 S. University Street, West Lafayette, IN 47907-2064, USA
| | - Bennett D. Elzey
- Purdue University Institute for Cancer Research, Purdue University, Hansen Life Sciences Research Building, Room 141, 201 S. University Street, West Lafayette, IN 47907-2064, USA
- Department of Comparative Pathobiology, Purdue University, West Lafayette, IN, USA
| | - Elizabeth J. Tran
- Department of Biochemistry, Purdue University, BCHM A343, 175 S. University Street, West Lafayette, IN 47907-2063, USA
- Purdue University Institute for Cancer Research, Purdue University, Hansen Life Sciences Research Building, Room 141, 201 S. University Street, West Lafayette, IN 47907-2064, USA
| |
Collapse
|
7
|
Jamal A, Aldreiwish AD, Banawas SS, Alqurashi YE, Kamal MA, Ahmad F. The paths toward immunotherapy of esophageal cancer: An overview of clinical trials. Int Immunopharmacol 2025; 151:114261. [PMID: 40015204 DOI: 10.1016/j.intimp.2025.114261] [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: 12/30/2024] [Revised: 01/26/2025] [Accepted: 02/06/2025] [Indexed: 03/01/2025]
Abstract
As the seventh-leading contributor to global cancer-related deaths, esophageal cancer (EC) is one of the most challenging types of cancer. Despite advancements in conventional therapies, including surgery, chemotherapy, and radiotherapy, the five-year survival rate remains low, underscoring the need for the development of more efficacious treatment approaches. Immunotherapy has emerged as a promising treatment approach, offering new hope for EC patients. This review provides an in-depth examination of the latest immunotherapeutic strategies for EC, focusing on immune checkpoint inhibitors, adoptive cell therapy, cancer vaccines, and oncolytic virotherapy. We critically analyze the current clinical data to highlight the progress and pitfalls of each immunotherapeutic approach for EC. Additionally, we explore the potential for combination therapies, which could overcome the resistance often seen with monotherapies. Finally, we discuss the limitations of current treatments and outline key areas for future research to improve patient outcomes and survival.
Collapse
Affiliation(s)
- Azfar Jamal
- Department of Biology, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia; Health and Basic Science Research Centre, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - Allolo D Aldreiwish
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Saeed S Banawas
- Health and Basic Science Research Centre, Majmaah University, Al-Majmaah 11952, Saudi Arabia; Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Yaser E Alqurashi
- Department of Biology, College of Science Al-Zulfi, Majmaah University, Al-Majmaah 11952, Saudi Arabia
| | - Mohammad Azhar Kamal
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Fuzail Ahmad
- Respiratory Care Department, College of Applied Sciences, Almaarefa University, Diriya, Riyadh 13713, Saudi Arabia
| |
Collapse
|
8
|
Dal Maso A, Ferrarini F, Esposito G, Minuzzo SA, Puggia AM, Pezzuto F, Zulato E, Bao LC, De Nuzzo M, Ferro A, Frega S, Pasello G, Calabrese F, Fassan M, Rea F, Guarneri V, Indraccolo S, Bonanno L. Liver kinase B1 expression is associated with improved prognosis and tumor immune microenvironment features in small cell lung cancer. Front Oncol 2025; 15:1552506. [PMID: 40255421 PMCID: PMC12006004 DOI: 10.3389/fonc.2025.1552506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 03/17/2025] [Indexed: 04/22/2025] Open
Abstract
Background Small cell lung cancer (SCLC) is characterized by early metastatic potential and poor prognosis. Liver kinase B1 (LKB1) is a tumor suppressor and a cell metabolism regulator. LKB1 downregulation has been associated with a cold tumor immune microenvironment (TIME). We aimed to analyze the role of LKB1 in SCLC in relation to its association with overall survival (OS) and TIME components. Methods We retrospectively evaluated SCLC patients consecutively treated at our institution from 1996 to 2020 with available tissue. LKB1, PD-L1 on tumor cells and on tumor immune-infiltrating cells, CD8, and FOXP3 were evaluated by immunohistochemistry (IHC), categorized according to predefined cutoffs. The primary endpoint was the description of LKB1 expression, and the secondary endpoints were the association with prognosis and TIME features. Results Tissue samples of 138 out of 481 SCLCs were adequate for molecular analyses. Eighty patients had limited stage (LS) at diagnosis and 58 had extended stage (ES). The median LKB1 IHC score was 4. Patients with IHC score >4 (n = 67) were classified as LKB1-positive. The probability of LKB1 positivity was higher in LS [odds ratio 2.78, 95% confidence interval (95% CI) 1.18-7.14]. At the data cutoff (2 January 2024), 123 patients died. The median OS (mOS) was 14.0 months (95% CI 11.5-19.4). mOS was significantly longer in patients with LKB1-positive expression [32.4 months (95% CI 13.6-62.4) vs. 11.2 months (95% CI 8.7-14.7); p < 0.001]. At multivariate analysis, positive LKB1 expression, LS, and no weight loss at diagnosis were confirmed as independent positive prognostic factors. TIME features were evaluated in 70 patients. Unexpectedly, LKB1-negative samples were more likely to show CD8+ tumor-infiltrating lymphocytes (TILs; p = 0.013). No association with PD-L1 expression nor the presence of FOXP3+ TILs was found. Conclusion LKB1 expression is a potential positive prognostic marker in SCLC. In this series, LKB1 expression was negatively associated with the presence of CD8+ TILs.
Collapse
Affiliation(s)
| | - Federica Ferrarini
- Basic and Translational Oncology, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Giovanni Esposito
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Sonia Anna Minuzzo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Anna Maria Puggia
- Anatomy and Pathological Histology, Veneto Institute of Oncology IOV - IRCCS, Castelfranco Veneto, Italy
| | - Federica Pezzuto
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Elisabetta Zulato
- Basic and Translational Oncology, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Loc Carlo Bao
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Mattia De Nuzzo
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Alessandra Ferro
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Stefano Frega
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
| | - Giulia Pasello
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Matteo Fassan
- Department of Medicine, University of Padua, Padua, Italy
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Federico Rea
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
- Thoracic Surgery Unit, Padova University Hospital, Padova, Italy
| | - Valentina Guarneri
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Stefano Indraccolo
- Basic and Translational Oncology, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Laura Bonanno
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| |
Collapse
|
9
|
Shen L, Zhang Y, Li Z, Zhang X, Gao X, Liu B, Wang Y, Ba Y, Li N, Zhang R, Zhang J, Chen Y, Chen J, Huang M, Fu Y, Liu M, Liu Z, Zhao J, Li W, Wei J, Li C, Xu N, Guo Z, Cao B, Liu L, Nie P, Wan L, Sheng L, Liu Z, He Y, Gu K, Wu G, Wang W, Zhang F, Qiu W, Guo J, Ying J, Pan H, Xu H, Yuan Y, Bai Y, Wang Z, Xu J, Zhao X, Liu H, Zhang X, Dai W, Xu H, Liu M, Xie L, Tang Y, Jin J, Qu X, Fang X, Huang M, Chen H, Zheng Z, Wang Y, Wang D, Li X, Yu G, Liu H, Zhou Y, Zhong D, Zeng S, Kang M, Wang M, Gao Y, Li W, Wang Z, Zhang M, Zhang J, Li Q, Sun S, Zang A, Lin L, Xie M, Zhuang Z, Zhang T, Yao Z, Lu D, Liu W, Hu M, Wang ZM, Li B, Xia M, Zhang J, Ying X, Pardoll DM, Ji J. First-line cadonilimab plus chemotherapy in HER2-negative advanced gastric or gastroesophageal junction adenocarcinoma: a randomized, double-blind, phase 3 trial. Nat Med 2025; 31:1163-1170. [PMID: 39843940 DOI: 10.1038/s41591-024-03450-4] [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: 09/29/2024] [Accepted: 12/04/2024] [Indexed: 01/24/2025]
Abstract
Programmed cell death protein-1 (PD-1) inhibitors plus chemotherapy have been the standard of care in the first-line treatment of advanced gastric or gastroesophageal junction (G/GEJ) adenocarcinoma; however, the survival benefits are modest in patients with low programmed death ligand 1 (PD-L1) expression. Here we investigated the efficacy and safety of cadonilimab (PD-1/cytotoxic T lymphocyte antigen-4 (CTLA-4) bispecific antibody) plus chemotherapy as first-line treatment in G/GEJ adenocarcinoma. The prespecified interim analysis is reported here. This was a randomized, double-blind, placebo-controlled phase 3 study. Eligible patients were adults with untreated, unresectable, locally advanced or metastatic G/GEJ adenocarcinoma. Patients were randomized 1:1 to receive cadonilimab (10 mg kg-1 every 3 weeks) or placebo plus chemotherapy (every 3 weeks). The primary endpoint was overall survival (OS) in the intention-to-treat population (one-sided significance level, P = 0.025). Secondary endpoints included OS in patients with a PD-L1 combined positive score ≥5, progression-free survival, objective response rate, duration of response and safety. As of 18 August 2023, 610 patients from 75 study centers were randomized to cadonilimab (n = 305) or placebo (n = 305). With a median follow-up of 18.7 months, the cadonilimab group had a significantly longer median OS (14.1 versus 11.1 months; hazard ratio (HR) 0.66; 95% confidence interval (CI) 0.54-0.81; P < 0.001) than the placebo group. The primary endpoint was met. The median progression-free survival was 7.0 months versus 5.3 months (HR 0.53, 95% CI 0.44-0.65). The median OS in patients with a PD-L1 combined positive score ≥5 was 15.3 months versus 10.9 months (HR 0.58, 95% CI 0.41-0.82). The objective response rate was 65.2% versus 48.9% with a median duration of response of 8.8 months versus 4.4 months. Grade ≥3 treatment-related adverse events occurred in 65.9% of the cadonilimab group and 53.6% of the placebo group, and the most common were decreased platelet count, decreased neutrophil count and anemia. Most of the immune-related adverse events were grade 1 or 2. No new safety signals were observed. Cadonilimab plus chemotherapy significantly improved OS with a manageable safety profile in patients with advanced G/GEJ adenocarcinoma. ClinicalTrials.gov registration: NCT05008783 .
Collapse
Affiliation(s)
- Lin Shen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yanqiao Zhang
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Ziyu Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaotian Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiangyu Gao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China
| | - Bo Liu
- Department of Gastroenterology, Shandong Cancer Hospital, Jinan, China
| | - Yusheng Wang
- Shanxi Provincial Cancer Hospital, Taiyuan, China
| | - Yi Ba
- Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Ning Li
- Henan Cancer Hospital, Zhengzhou, China
| | - Ruixing Zhang
- The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | | | - Ye Chen
- The First Affiliated Hospital of Henan University of Science and Technology, Luoyang, China
| | - Jian Chen
- Yantai Yuhuangding Hospital, Yantai, China
| | - Mingzhu Huang
- Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yang Fu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mulin Liu
- The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Zheng Liu
- Handan Central Hospital, Handan, China
| | - Jun Zhao
- Changzhi People's Hospital, Changzhi, China
| | - Wei Li
- The First Bethune Hospital of Jilin University, Changchun, China
| | - Jia Wei
- Nanjing Drum Tower Hospital, Nanjing, China
| | - Changzheng Li
- Department of Gastroenterology, Shandong Cancer Hospital, Jinan, China
| | - Nong Xu
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | - Bangwei Cao
- Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lian Liu
- Qilu Hospital of Shandong University, Jinan, China
| | - Peng Nie
- Gansu Wuwei Tumour Hospital, Wuwei, China
| | - Lixin Wan
- Nanyang Central Hospital, Nanyang, China
| | - Lili Sheng
- The First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | | | - Yifu He
- Anhui Provincial Cancer Hospital, Hefei, China
| | - Kangsheng Gu
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guowu Wu
- Meizhou People's Hospital, Meizhou, China
| | - Weibo Wang
- Shandong Provincial Hospital, Jinan, China
| | - Futong Zhang
- Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Wensheng Qiu
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jun Guo
- Xingtai People's Hospital, Xingtai, China
| | - Jieer Ying
- Zhejiang Cancer Hospital, Hangzhou, China
| | - Hongming Pan
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | - Yuan Yuan
- Xuzhou Central Hospital, Xuzhou, China
| | - Yuansong Bai
- The Third Bethune Hospital of Jilin University, Changchun, China
| | - Zhenghua Wang
- The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Jiye Xu
- Zhoukou Central Hospital, Zhoukou, China
| | | | - Hao Liu
- Sichuan Provincial People's Hospital, Chengdu, China
| | - Xizhi Zhang
- Northern Jiangsu People's Hospital, Yangzhou, China
| | - Wenxiang Dai
- The First Affiliated Hospital of The University of South China, Hengyang, China
| | - Hongyan Xu
- Weihai Municipal Hospital, Weihai, China
| | - Ming Liu
- West China Hospital of Sichuan University, Chengdu, China
| | - Lin Xie
- Yunnan Cancer Hospital, Kunming, China
| | - Yong Tang
- Affiliated Cancer Hospital of Xinjiang Medical University, Urumqi, China
| | - Jianying Jin
- Taizhou Hospital of Zhejiang Province, Linhai, China
| | - Xiujuan Qu
- The First Hospital of China Medical University, Shenyang, China
| | - Xuefeng Fang
- The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Mingwei Huang
- Guangxi Medical University Cancer Hospital, Nanning, China
| | - Hao Chen
- The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, China
| | - Zhendong Zheng
- General Hospital of Northern Theater Command, Shenyang, China
| | - Ying Wang
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Daqing Wang
- Hengshui People's Hospital (Harrison International Peace Hospital), Hengshui, China
| | - Xiaoqin Li
- Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Guohua Yu
- Weifang People's Hospital, Weifang, China
| | - Haiyan Liu
- The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Yongjian Zhou
- Fujian Medical University Union Hospital, Fuzhou, China
| | | | - Shan Zeng
- Xiangya Hospital Central South University, Changsha, China
| | - Mafei Kang
- Affiliated Hospital of Guilin Medical University, Guilin, China
| | | | - Yong Gao
- Huai'an First People's Hospital, Huai'an, China
| | - Wenxin Li
- Inner Mongolia Autonomous Region People's Hospital, Hohhot, China
| | - Zejun Wang
- The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, China
| | | | | | - Qingshan Li
- Affiliated Hospital of Chengde Medical University, Chengde, China
| | | | - Aimin Zang
- Affiliated Hospital of Hebei University, Baoding, China
| | - Lizhu Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ming Xie
- Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhixiang Zhuang
- The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Tao Zhang
- Yuebei People's Hospital, Shaoguan, China
| | | | | | - Wei Liu
- Akeso Biopharma Inc., Zhongshan, China
| | | | | | | | | | - Jiajia Zhang
- University of California Los Angeles Health, Los Angeles, CA, USA
| | - Xiangji Ying
- Department of Epidemiology, University of North Carolina Gillings School of Global Public Health, Chapel Hill, NC, USA
| | - Drew M Pardoll
- The Bloomberg~Kimmel Institute for Cancer Immunotherapy and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jiafu Ji
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
10
|
Yang Z, Luo Y, Lu R, Liu X, Liu H, Liu S, Huang C, Tian J, Zhang L. Incidence Rates of Cutaneous Immune-Related Adverse Events in Patients with Lung Cancer: A Systematic Review and Meta-Analysis. Curr Oncol 2025; 32:195. [PMID: 40277752 PMCID: PMC12025845 DOI: 10.3390/curroncol32040195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/26/2025] Open
Abstract
OBJECTIVE Cutaneous immune-related adverse events (cirAEs) represent a prevalent manifestation of adverse reactions linked to immune checkpoint inhibitors (ICIs) therapy, substantially affecting patients' quality of life. This systematic review and meta-analysis aimed to quantify the pooled incidence of cirAEs in this population and strengthen clinical awareness for early recognition and management. METHODS A comprehensive search of PubMed, Embase, CINAHL, Cochrane Library, CBM, CNKI, and Wanfang databases was conducted from inception to December 2022. Literature that reported the incidence of cirAEs in patients with lung cancer receiving ICIs therapy was included. A meta-analysis was conducted using R software, version 4.4.1 to estimate the pooled incidence of cirAEs, and a random-effects model was used for data synthesis. Begg's rank correlation and funnel plots were used to assess publication bias. RESULTS A total of 99 articles involving 23,814 patients with lung cancer receiving ICIs therapy were included, with publication dates ranging from 2012 to 2022. The meta-analysis results reveal that the incidence of cirAEs in patients with lung cancer was 20.26% (95% confidence interval [CI (17.12-23.81)]. Significant differences were observed between all subgroups, including continent, study type, combination therapy, dual ICIs therapy, and diagnostic criteria for cirAEs for Grade 1-2 and Grade 3-4 incidences. CONCLUSIONS The incidence of cirAEs in patients with lung cancer is relatively high, particularly undergoing combined or dual ICIs therapy. To comprehensively characterize cirAEs in patients with lung cancer, large-scale multicenter studies integrating real-world pharmacovigilance data are warranted to establish precise incidence estimates and identify clinically significant risk factors. IMPLICATIONS FOR CLINICAL PRACTICE This review's insights aroused clinical staff's attention and concern about cirAEs, potentially enhancing the quality of life of patients with cancer.
Collapse
Affiliation(s)
- Zhihui Yang
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China; (Z.Y.)
- Evidence Based Nursing and Midwifery Practice PR China: A JBI Centre of Excellence, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China
| | - Yuanyuan Luo
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China; (Z.Y.)
| | - Ruiqi Lu
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China; (Z.Y.)
| | - Xinqi Liu
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China; (Z.Y.)
| | - Hanyu Liu
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China; (Z.Y.)
| | - Suting Liu
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China; (Z.Y.)
| | - Chen Huang
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China; (Z.Y.)
| | - Jinhui Tian
- Evidence Based Medicine Centre, School of Basic Medical Sciences, Lanzhou University, No. 199, Donggang West Road, Lanzhou 730000, China
| | - Lili Zhang
- School of Nursing, Southern Medical University, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China; (Z.Y.)
- Evidence Based Nursing and Midwifery Practice PR China: A JBI Centre of Excellence, No. 1023, South Shatai Road, Baiyun District, Guangzhou 510515, China
| |
Collapse
|
11
|
Ren X, Guo A, Geng J, Chen Y, Wang X, Zhou L, Shi L. Pan-cancer analysis of co-inhibitory molecules revealing their potential prognostic and clinical values in immunotherapy. Front Immunol 2025; 16:1544104. [PMID: 40196117 PMCID: PMC11973099 DOI: 10.3389/fimmu.2025.1544104] [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: 12/12/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Background The widespread use of immune checkpoint inhibitors (anti-CTLA4 or PD-1) has opened a new chapter in tumor immunotherapy by providing long-term remission for patients. Unfortunately, however, these agents are not universally available and only a minority of patients respond to them. Therefore, there is an urgent need to develop novel therapeutic strategies targeting other co-inhibitory molecules. However, comprehensive information on the expression and prognostic value of co-inhibitory molecules, including co-inhibitory receptors and their ligands, in different cancers is not yet available. Methods We investigated the expression, correlation, and prognostic value of co-inhibitory molecules in different cancer types based on TCGA, UCSC Xena, TIMER, CellMiner datasets. We also examined the associations between the expression of these molecules and the extent of immune cell infiltration. Besides, we conducted a more in-depth study of VISTA. Result The results of differential expression analysis, correlation analysis, and drug sensitivity analysis suggest that CTLA4, PD-1, TIGIT, LAG3, TIM3, NRP1, VISTA, CD80, CD86, PD-L1, PD-L2, PVR, PVRL2, FGL1, LGALS9, HMGB1, SEMA4A, and VEGFA are associated with tumor prognosis and immune cell infiltration. Therefore, we believe that they are hopefully to serve as prognostic biomarkers for certain cancers. In addition, our analysis indicates that VISTA plays a complex role and its expression is related to TMB, MSI, cancer cell stemness, DNA/RNA methylation, and drug sensitivity. Conclusions These co-inhibitory molecules have the potential to serve as prognostic biomarkers and therapeutic targets for a broad spectrum of cancers, given their strong associations with key clinical metrics. Furthermore, the analysis results indicate that VISTA may represent a promising target for cancer therapy.
Collapse
Affiliation(s)
- Xiaoyu Ren
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Anjie Guo
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Jiahui Geng
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Yuling Chen
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Xue Wang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Lian Zhou
- Department of Head&Neck Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Lei Shi
- School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
12
|
Long B, Zhou H, Yu Z, Zhu J, Yang H, Huang Z, Wei D, Chen S, Yang X, Zhao X, Zhang W, Yan H, Guan X, Li L, Zhang G, Yu H, Che S, Gao Z, Jiang X, Luo C, Mao J, Zhao D, Li Y, Jiang Z, Jiao Z. Neoadjuvant cadonilimab plus FLOT chemotherapy in locally advanced gastric/gastroesophageal junction adenocarcinoma: A multicenter, phase 2 study. MED 2025; 6:100531. [PMID: 39536755 DOI: 10.1016/j.medj.2024.10.008] [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: 07/15/2024] [Revised: 08/06/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Treatment with cadonilimab and chemotherapy has shown promise as a first-line treatment for gastric or gastroesophageal junction (G/GEJ) adenocarcinoma. However, its application in neoadjuvant settings has not yet been documented. METHODS This multicenter, phase 2 trial (ChiCTR2200066893) was conducted at four hospitals across China. Treatment-naive patients with locally advanced G/GEJ adenocarcinoma (cT3/4, N+, M0) and who were human epidermal growth factor receptor 2 negative received 3-cycle or 4-cycle neoadjuvant treatment of cadonilimab plus FLOT (5-fluorouracil, leucovorin, oxaliplatin, and docetaxel) chemotherapy, followed by gastrectomy and 4-cycle adjuvant FLOT chemotherapy. The primary endpoint was the pathological complete response (pCR) rate. Secondary endpoints included major pathological response (MPR), overall response rate (ORR), disease control rate (DCR), R0 resection rate, downstaging rate, and safety. FINDINGS Between December 23, 2022, and December 15, 2023, 32 of 38 patients completed the scheduled treatment, achieving an R0 resection rate of 100% (32/32). The pCR rate was 21.1% (8/38, 90% confidence interval [CI]: 9.7-32.4), and the MPR rate was 44.7% (17/38, 90% CI: 30.9-58.5). Radiological evaluations were available for 28 of 38 patients by blinded independent central review. The ORR was 60.7% (17/28, 90% CI: 44.7-76.7), and the DCR was 100.0% (28/28, 90% CI: 100.0-100.0). Tumor downstaging occurred in 71.9% of patients (23/32), with consistent efficacy across all populations observed in the subgroup analysis. Grade 3 adverse events occurred in 31.6% of patients without severe safety issues. CONCLUSIONS Neoadjuvant cadonilimab plus FLOT chemotherapy treatment exhibits promising efficacy with manageable toxicities in locally advanced G/GEJ adenocarcinoma, providing preliminary evidence for further investigation. FUNDING This study was funded by Akeso Biopharma.
Collapse
Affiliation(s)
- Bo Long
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Huinian Zhou
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Zeyuan Yu
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Junmin Zhu
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Hanteng Yang
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Zeping Huang
- Lanzhou University Second Hospital, The Oncological Surgery Department, Lanzhou, China
| | - Dengwen Wei
- Sun Yat-sen University Cancer Center Gansu Provincial Cancer Hospital, The Gastrointestinal Surgery Department, Lanzhou, China
| | - Shigong Chen
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Xiaojun Yang
- Gansu Provincial Hospital, The General Surgery Department, Lanzhou, China
| | - Xiaoning Zhao
- Sun Yat-sen University Cancer Center Gansu Provincial Cancer Hospital, The Gastrointestinal Surgery Department, Lanzhou, China
| | - Wenjuan Zhang
- Lanzhou University Second Hospital, The Radiology Department, Lanzhou, China
| | - Hong Yan
- Lanzhou University Second Hospital, The Pathology Department, Lanzhou, China
| | - Xiaoying Guan
- Lanzhou University Second Hospital, The Pathology Department, Lanzhou, China
| | - Long Li
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Gengyuan Zhang
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Hongwei Yu
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Shengfu Che
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Zhongti Gao
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Xiangyan Jiang
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Changjiang Luo
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Jie Mao
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China
| | - Da Zhao
- The First Hospital of Lanzhou University, The Oncology Department, Lanzhou, China
| | - Yumin Li
- Lanzhou University Second Hospital, The Oncological Surgery Department, Lanzhou, China
| | - Zebin Jiang
- Gansu Provincial Hospital, The General Surgery Department, Lanzhou, China
| | - Zuoyi Jiao
- Lanzhou University Second Hospital, The General Surgery Department, Lanzhou, China; Biobank of Tumors from Plateau of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China.
| |
Collapse
|
13
|
Tascón J, Casanova AG, Vicente-Vicente L, López-Hernández FJ, Morales AI. Nephrotoxicity of Immune Checkpoint Inhibitors in Single and Combination Therapy-A Systematic and Critical Review. Biomedicines 2025; 13:711. [PMID: 40149687 PMCID: PMC11940260 DOI: 10.3390/biomedicines13030711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Immune checkpoint inhibitors (ICIs) have generated a revolutionary approach in the treatment of cancer, but their effectiveness has been compromised by immune-related adverse events, including renal damage. Although rare, these effects are relevant because they have been related to poor patient prognoses. The objective of this review was to estimate the current incidence of nephrotoxicity in patients treated with single and double ICI therapies. Methods: A total of 1283 potential articles were identified, which were reduced to 50 after applying the exclusion and inclusion criteria. Results: This study reveals the increase in acute kidney injury associated with these drugs in the last decade and shows that, interestingly, combined therapies with ICIs does not lead to an increase in kidney damage compared with anti-CTLA-4. It also suggests that kidney damage could be underdiagnosed when it comes to interstitial nephritis, because definitive evidence requires a renal biopsy. Conclusions: In perspective, these conclusions could guide clinicians in making decisions for therapy personalization and highlight the need to search for new diagnostic systems that are more sensitive and specific to the type of damage and could replace the biopsy.
Collapse
Affiliation(s)
- Javier Tascón
- Toxicology Unit, Universidad de Salamanca (USAL), 37007 Salamanca, Spain; (J.T.); (A.G.C.); (L.V.-V.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), 37007 Salamanca, Spain
| | - Alfredo G. Casanova
- Toxicology Unit, Universidad de Salamanca (USAL), 37007 Salamanca, Spain; (J.T.); (A.G.C.); (L.V.-V.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), 37007 Salamanca, Spain
| | - Laura Vicente-Vicente
- Toxicology Unit, Universidad de Salamanca (USAL), 37007 Salamanca, Spain; (J.T.); (A.G.C.); (L.V.-V.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), 37007 Salamanca, Spain
| | - Francisco J. López-Hernández
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), 37007 Salamanca, Spain
- Group of Biomedical Research on Critical Care (BioCritic), 47005 Valladolid, Spain
| | - Ana I. Morales
- Toxicology Unit, Universidad de Salamanca (USAL), 37007 Salamanca, Spain; (J.T.); (A.G.C.); (L.V.-V.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain;
- Department of Physiology and Pharmacology, Universidad de Salamanca (USAL), 37007 Salamanca, Spain
- Group of Translational Research on Renal and Cardiovascular Diseases (TRECARD), 37007 Salamanca, Spain
- Group of Biomedical Research on Critical Care (BioCritic), 47005 Valladolid, Spain
| |
Collapse
|
14
|
Li J, Zhou X, Wu L, Ma J, Tan Y, Wu S, Zhu J, Wang Q, Shi Q. Optimal early endpoint for second-line or subsequent immune checkpoint inhibitors in previously treated advanced solid cancers: a systematic review. BMC Cancer 2025; 25:293. [PMID: 39966752 PMCID: PMC11837729 DOI: 10.1186/s12885-025-13712-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND The administration of second-line or subsequent immune checkpoint inhibitors (ICIs) in previously treated patients with advanced solid cancers has been clinically investigated. However, previous clinical trials lacked an appropriate primary endpoint for efficacy assessment. This systematic review aimed to explore the most optimal early efficacy endpoint for such trials. METHODS Phase 2 or 3 clinical trials involving patients with advanced solid cancers with disease progression following standard first-line therapy receiving second-line or subsequent ICI administration, with adequate survival outcome data, were included from PubMed, Embase, Web of Science, and Cochrane Library databases before February 2023. Quality assessment was conducted using the Cochrane tool and Newcastle-Ottawa Quality Assessment Scale for Cohort Studies for randomized controlled trials (RCTs) and non-randomized trials, respectively. Objective response rate (ORR) and progression-free survival (PFS) at 3, 6, and 9 months were investigated as potential early efficacy endpoint candidates for 12-month overall survival (OS), with a strong correlation defined as Pearson's correlation coefficient r ≥ 0.8. RESULTS A total of 64 RCTs comprising 22,725 patients and 106 non-randomized prospective trials involving 10,608 participants were eligible for modeling and external validation, respectively. RCTs examined 15 different cancer types, predominantly non-small-cell lung cancer (NSCLC) (17, 28%), melanoma (9, 14%), and esophageal squamous cell carcinoma (5, 8%). The median sample size of RCTs was 124 patients, and the median follow-up time was 3.2-57.7 months. The ORR (r = 0.38; 95% confidence interval [CI], 0.18-0.54) and PFS (r = 0.42; 95% CI, 0.14-0.64) exhibited weak trial-level correlations with OS. Within ICI treatment arms, the r values of ORR and 3-, 6-, and 9-month PFS with 12-month OS were 0.61 (95% CI, 0.37-0.79), 0.78 (95% CI, 0.62-0.88), 0.84 (95% CI, 0.77-0.90), and 0.86 (95% CI, 0.79-0.90), respectively. External validation of 6-month PFS indicated an acceptable discrepancy between actual and predicted 12-month OS. CONCLUSIONS In non-randomized phase 2 trials on second-line or subsequent ICI therapy in patients with advanced solid cancers, 6-month PFS could serve as an early efficacy endpoint. However, early efficacy endpoints are not recommended in RCTs to replace OS.
Collapse
Affiliation(s)
- Jingqiu Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoding Zhou
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lei Wu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Jiabao Ma
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Yan Tan
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Songke Wu
- Department of Oncology, People'S Hospital of Cangxi County, Guangyuan, China.
| | - Jie Zhu
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.
| | - Qifeng Wang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China.
| | - Qiuling Shi
- Center for Cancer Prevention Research, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, School of Public Health and Management, Chongqing Medical University, Chongqing, China
| |
Collapse
|
15
|
Calles A, Navarro A, Doger de Speville Uribe BG, Colomé EÁ, de Miguel M, Álvarez R, Arregui M, Moreno V, Rocha P, Calvo E, Ramon-Patino J, Corral de la Fuente E, Alcalá-López D, Boix O, Fernández-Pinto M, Rodríguez-Morató J, Palmero R, Nadal E, Jove M, Felip E. Lurbinectedin Plus Pembrolizumab in Relapsed SCLC: The Phase I/II LUPER Study. J Thorac Oncol 2025:S1556-0864(25)00064-4. [PMID: 39938593 DOI: 10.1016/j.jtho.2025.02.005] [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: 11/22/2024] [Revised: 01/30/2025] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
INTRODUCTION SCLC has limited second-line treatment options after chemotherapy. We assessed the efficacy and safety of lurbinectedin combined with pembrolizumab in relapsed SCLC patients who had not received prior immunotherapy, aiming to prevent early progression and achieve sustained responses. METHODS The LUPER trial (NCT04358237) is a phase I/II, single-arm, open-label, multicenter study. Phase I established the recommended phase II dose. The primary endpoint of phase II was the investigator-confirmed objective response rate. Secondary endpoints included duration of response, progression-free survival (PFS), overall survival (OS), and safety. Patients were categorized as platinum-sensitive (chemotherapy-free interval ≥ 90 d) or platinum-resistant (<90 d). RESULTS The recommended phase II dose was 3.2 mg/m2 lurbinectedin and 200 mg pembrolizumab IV every three weeks. Phase II included 28 patients, 50% of whom were platinum-resistant. The objective response rate was 46.4% (95% confidence interval: 27.5-66.1, p < 0.001), including three complete responses, with two complete metabolic responses post-treatment completion at 35 cycles. The median duration of response was 7.8 months, with 40% of patients maintaining responses for 12 months or longer. The median PFS was 4.6 months, and the median OS was 10.5 months. Platinum-sensitive patients had significantly better PFS (8.0 versus 2.8 mo, p = 0.012) and numerically superior OS (15.7 versus 7.1 mo, p = 0.058). Grade 3 or higher treatment-related adverse events occurred in 71.4% of patients, with transient neutropenia being the most common. Immune-related adverse events were consistent with prior pembrolizumab studies. CONCLUSIONS Lurbinectedin plus pembrolizumab reported promising efficacy in relapsed SCLC, particularly for platinum-sensitive patients, with a known and manageable safety profile. These results support further exploration of this combination in SCLC treatment.
Collapse
Affiliation(s)
- Antonio Calles
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain.
| | - Alejandro Navarro
- Medical Oncology Department, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Medica Scientia Innovation Research (MEDSIR), Barcelona (Spain), Ridgewood, New Jersey
| | | | - Enric Álvarez Colomé
- Medical Oncology Department, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - María de Miguel
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Rosa Álvarez
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Marta Arregui
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Víctor Moreno
- START Madrid-FJD, Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Pedro Rocha
- Medical Oncology Department, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Emiliano Calvo
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | - Jorge Ramon-Patino
- START Madrid-CIOCC, Centro Integral Oncológico Clara Campal, Madrid, Spain
| | | | - Daniel Alcalá-López
- Medica Scientia Innovation Research (MEDSIR), Barcelona (Spain), Ridgewood, New Jersey
| | - Olga Boix
- Medica Scientia Innovation Research (MEDSIR), Barcelona (Spain), Ridgewood, New Jersey
| | | | - Jose Rodríguez-Morató
- Medica Scientia Innovation Research (MEDSIR), Barcelona (Spain), Ridgewood, New Jersey
| | - Ramón Palmero
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Spain
| | - Ernest Nadal
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Spain
| | - Maria Jove
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet, Barcelona, Spain
| | - Enriqueta Felip
- Medical Oncology Department, Vall d'Hebron University Hospital and Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
16
|
Nabipur L, Mouawad M, Venketaraman V. Therapeutic Applications of Programmed Death Ligand 1 Inhibitors in Small Cell Lung Cancer. Biomedicines 2025; 13:401. [PMID: 40002814 PMCID: PMC11852381 DOI: 10.3390/biomedicines13020401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/29/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Small cell lung cancer (SCLC) is an aggressive cancer with rapid progression, limited treatment success, and high relapse rates. Chemotherapy and radiation are standard treatments but often result in chemoresistance. PD-L1 inhibitors have gained attention for their role in enhancing tumor immunity. Methods: This review summarizes clinical trials involving PD-L1 inhibitors, such as atezolizumab, durvalumab, pembrolizumab, and nivolumab, in SCLC treatment. Key trials include IMpower133, CASPIAN, KEYNOTE-604, and CheckMate 331, focusing on survival outcomes and treatment efficacy. Results: Studies such as IMpower133 and CASPIAN demonstrate improved overall survival when PD-L1 inhibitors were added to platinum-based chemotherapy. However, outcomes in trials such as KEYNOTE-604 and CheckMate 331 varied, showing the need for refined patient selection. Adverse events (AEs) associated with these treatments were also noted. PD-L1 inhibitors offer promise in SCLC treatment, but efficacy varies across trials and patient groups. Future research should focus on better patient selection and overcoming resistance mechanisms. Addressing immune-related AEs is essential for optimizing treatment strategies.
Collapse
Affiliation(s)
| | | | - Vishwanath Venketaraman
- College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, CA 91766, USA; (L.N.); (M.M.)
| |
Collapse
|
17
|
Shen Y, Liu Z, Chen Y, Shi X, Dong S, Wang B. Candidate Biomarker of Response to Immunotherapy In Small Cell Lung Cancer. Curr Treat Options Oncol 2025; 26:73-83. [PMID: 39841387 DOI: 10.1007/s11864-025-01292-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2025] [Indexed: 01/23/2025]
Abstract
OPINION STATEMENT Small-cell lung cancer accounts for about 15% of lung cancers with an extremely poor prognosis. The incorporation of immunotherapy to platinum-based chemotherapy offers sustained overall survival benefits and become the standard for the first-line setting of extensive-stage small-cell lung cancer. However, only a limited number of patients derive prolonged benefits. Although novel immunomodulatory agents and combination strategies are currently under investigation, identifying patients who are likely to obtain clinical benefits from this therapeutic approach is urgently needed. The modest therapeutic response to immunotherapy can be explained by various mechanisms. Traditional biomarkers do not guide immunotherapeutic decision-making in small-cell lung cancer. Notably, recent progress in the understanding of the molecular typing of small-cell lung cancer based on multi-omics data might bring new sights. This review summarizes the potential biomarkers for small-cell lung cancer immunotherapy based on clinical trials and preclinical studies. Moreover, important constraints in identifying biomarkers for small-cell lung cancer treatment are discussed.
Collapse
Affiliation(s)
- Yili Shen
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, Huzhou, Zhejiang, China
- Huzhou Key Laboratory of Precision Diagnosis and Treatment in Respiratory Diseases, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Zhicong Liu
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, Huzhou, Zhejiang, China
- Huzhou Key Laboratory of Precision Diagnosis and Treatment in Respiratory Diseases, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Yi Chen
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, Huzhou, Zhejiang, China
- Huzhou Key Laboratory of Precision Diagnosis and Treatment in Respiratory Diseases, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Xuefei Shi
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, Huzhou, Zhejiang, China.
- Huzhou Key Laboratory of Precision Diagnosis and Treatment in Respiratory Diseases, Huzhou Central Hospital, Huzhou, Zhejiang, China.
| | - Shunli Dong
- Huzhou Key Laboratory of Precision Diagnosis and Treatment in Respiratory Diseases, Huzhou Central Hospital, Huzhou, Zhejiang, China.
- Department of Central Laboratory, Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, Huzhou, Zhejiang, China.
| | - Bin Wang
- Department of Respiratory Medicine, Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, Huzhou, Zhejiang, China.
- Huzhou Key Laboratory of Precision Diagnosis and Treatment in Respiratory Diseases, Huzhou Central Hospital, Huzhou, Zhejiang, China.
| |
Collapse
|
18
|
Najah Q, Almosilhy NA, Ghanm TIE. Is nivolumab alone or in combination with ipilimumab more effective for treating lung cancer? a meta-analysis. Eur J Clin Pharmacol 2025; 81:269-278. [PMID: 39680077 DOI: 10.1007/s00228-024-03789-0] [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: 07/08/2024] [Accepted: 12/08/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Nivolumab and ipilimumab combination immunotherapy has become a standard treatment option for certain cancers. However, the benefits of combination therapy compared to nivolumab monotherapy in lung cancer patients are not entirely clear. We aimed to evaluate whether nivolumab plus ipilimumab improves clinical outcomes in lung cancer patients compared to nivolumab monotherapy. METHODS A literature search was performed on PubMed, Web of Science, and Scopus from inception until November 2024 to identify relevant randomized controlled trials. The Cochrane risk of bias tool was used to assess the risk of bias, the hazard ratio (HR) was calculated for survival, risk ratios (RRs) were calculated for response rate and safety outcomes, and a random effects model meta-analysis was performed to estimate the safety and efficacy of the treatments. RESULTS Seven trials comprising 2134 patients were included. Compared with patients receiving nivolumab monotherapy, non-small cell lung cancer patients who received combination therapy had better progression-free survival (HR = 0.82, 95% CI 0.71; 0.93, P < 0.01, low certainty), and there were no significant differences in overall survival (HR = 0.95, 95% CI 0.86; 1.0, P = 0.31, moderate certainty), or objective response rate (RR = 1.36, 95% CI 0.91; 2.02, P = 0.14 very low certainty). The combination group had a significantly greater risk of grade 3-4 adverse events (RR = 2.77, 95% CI 1.38; 5.56, P < 0.01, low certainty). CONCLUSION Although combination treatment significantly improved progression-free survival in NSCLC patients, it was also associated with a greater risk of adverse events and treatment-related mortality than nivolumab monotherapy. The current evidence is insufficient for choosing combination treatment over nivolumab monotherapy.
Collapse
Affiliation(s)
- Qasi Najah
- Faculty of Medicine, Elmergib University, Al-Khums, Libya.
- Medical Research Group of Libya, Negida Academy, Arlington, MA, USA.
| | - Nereen A Almosilhy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
| | - Thoria Ibrahim Essa Ghanm
- Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
| |
Collapse
|
19
|
Deng Y, Chen Q, Guo C, Chen J, Li X, Li Z, Zhang Y, Zhao J, Zhou J, Cai J, Yan T, Wang X, Bi X, Huang Z, Zhao H. Comprehensive single-cell atlas of colorectal neuroendocrine tumors with liver metastases: unraveling tumor microenvironment heterogeneity between primary lesions and metastases. Mol Cancer 2025; 24:28. [PMID: 39838423 PMCID: PMC11748842 DOI: 10.1186/s12943-025-02231-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: 09/10/2024] [Accepted: 01/09/2025] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Colorectal neuroendocrine tumors with liver metastases (CRNELM) are associated with a poorer prognosis compared to their nonmetastatic counterparts. A comprehensive understanding of the tumor microenvironment (TME) heterogeneity between primary lesions (PL) and liver metastases (LM) could provide crucial insights for enhancing clinical management strategies for these patients. METHODS We utilized single-cell RNA sequencing to analyze fresh tissue samples from CRNELM patients, aiming to elucidate the variations in TME between PL and LM. Complementary multidimensional validation was achieved through spatial transcriptomics, bulk RNA sequencing, and multiplex immunohistochemistry/immunofluorescence. RESULTS Our single-cell RNA sequencing analysis revealed that LM harboured a higher proportion of CD8 + T cells, CD4 + T cells, NK cells, NKT cells, and B cells exhibiting a stress-like phenotype compared to PL. RGS5 + pericytes may play a role in the stress-like phenotype observed in immune cells within LM. MCs in PL (PL_MCs) and LM (LM_MCs) exhibit distinct activation of tumor-associated signaling pathways. Notably, COLEC11 + matrix cancer-associated fibroblasts (COLEC11_mCAFs) were found to be significantly associated with LM_MCs. Cell communication analysis unveiled potential targetable receptor-ligand interactions between COLEC11_mCAFs and LM_MCs. Multidimensional validation confirmed the prominence of the characteristic stress-like phenotypes, including HSPA6_CD8_Tstr, HSPA6_NK, and COLEC11_mCAFs in LM. Moreover, a higher abundance of COLEC11_mCAFs correlated with poorer survival rates in the neuroendocrine tumor patient cohort. CONCLUSION Overall, our study provides the first single-cell analysis of the cellular and molecular differences between PL and LM in CRNELM patients. We identified distinct cell subsets and receptor-ligand interactions that may drive TME discrepancies and support metastatic tumor growth. These insights highlight potential therapeutic targets and inform strategies for better managing CRNELM patients.
Collapse
Affiliation(s)
- Yiqiao Deng
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qichen Chen
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Chengyao Guo
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jinghua Chen
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xin Li
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zhiyu Li
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yefan Zhang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianjun Zhao
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianguo Zhou
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianqiang Cai
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Yan
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Xiaobing Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Xinyu Bi
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Zhen Huang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Hong Zhao
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
20
|
Buckley MW, Balaji Warner A, Brahmer J, Cappelli LC, Sharfman WH, Fuchs E, Kang H, Forde PM, Gladstone DE, Ambinder R, Kelly RJ, Lipson EJ, Gojo I, Lee EJ, Johnson TP, Saidha S, Llinas R, Ostrow LW, Naidoo J, Probasco JC. Immune-related encephalitis after immune checkpoint inhibitor therapy. Oncologist 2025; 30:oyae186. [PMID: 39066587 PMCID: PMC11783331 DOI: 10.1093/oncolo/oyae186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) have revolutionized cancer treatment but can trigger immune-related encephalitis. We report one of the largest case series of patients with immune-related encephalitis and review of the literature. METHODS Retrospective series of patients with immune-related encephalitis and literature review. RESULTS Fourteen patients with cancer treated with ICI (50% combination therapy) developed immune-related encephalitis. Diagnostic testing revealed cerebral spinal fluid (CSF) lymphocytic pleocytosis (85%) and elevated protein (69%), abnormal brain magnetic resonance imaging(MRI) (33%) or brain FDG-PET (25%), electroencephalogram (EEG) abnormalities (30%), and autoantibodies (31%). Encephalitis treatment included: corticosteroids (86%), intravenous immunoglobulin (IVIg) (36%), plasmapheresis (7%), and rituximab (29%). There were no deaths and 12 patients had significant recovery, although long-term complications were observed. All patients discontinued ICI. Longitudinal follow-up demonstrated anti-cancer response to ICI at 3 months (85%) and 6 months post-ICI initiation (77%). A literature review identified 132 patients with immune-related encephalitis. Most were treated with PD-1 inhibitors (18% combination). Common abnormalities included elevated CSF protein (84%) or pleocytosis (77%), abnormal brain MRI (65%), or autoantibodies (47%). Nearly all were treated with corticosteroids, many required additional therapy with IVIg (26%) or rituximab (12%). Most patients had clinical improvement (81%) but a minority (10%) had a clinical relapse after completing corticosteroid taper. ICIs were resumed in 7 patients (5%), with relapse in 3. CONCLUSIONS AND RELEVANCE Immune-related encephalitis is treatable and improves with corticosteroids in most cases but may require additional immunosuppression. Re-emergence of encephalitis is rare and does not typically result in adverse outcomes, and this should be considered in neurological immune-related adverse event management guidelines.
Collapse
Affiliation(s)
- Monica W Buckley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Department of Neurology, University of Virginia School of Medicine, Charlottesville, VA 22903, United States
| | - Aanika Balaji Warner
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
| | - Julie Brahmer
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
| | - Laura C Cappelli
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - William H Sharfman
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
| | - Ephraim Fuchs
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Hyunseok Kang
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, United States
| | - Patrick M Forde
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
| | - Douglas E Gladstone
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- R.J. Zuckerberg Cancer Center at Hofstra/Northwell Health, Lake Success, NY 11042, United States
| | - Richard Ambinder
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Ronan J Kelly
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Charles A. Sammons Cancer Center, Baylor University Medical Center, Dallas, TX 75246, United States
| | - Evan J Lipson
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
| | - Ivana Gojo
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Edward J Lee
- Maryland Oncology Hematology, Columbia, MD 21044, United States
| | - Tory P Johnson
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Shiv Saidha
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Rafael Llinas
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Lyle W Ostrow
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, United States
| | - Jarushka Naidoo
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD 21287, United States
- Department of Oncology, Johns Hopkins Bayview Medical Center, Baltimore, MD 21224, United States
- Department of Medicine, Beaumont Hospital Dublin and RCSI University of Health Sciences, Dublin, 9, Ireland
| | - John C Probasco
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| |
Collapse
|
21
|
Gao G, Sun M, Yang Z, Li J, Ji H, Yu G. Combining Immunotherapy with Anlotinib in Extensive-Stage Small Cell Lung Cancer: A Multicenter Analysis of Efficacy and Safety. Technol Cancer Res Treat 2025; 24:15330338251329248. [PMID: 40112323 PMCID: PMC11926841 DOI: 10.1177/15330338251329248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/15/2025] [Accepted: 03/05/2025] [Indexed: 03/22/2025] Open
Abstract
BackgroundExtensive-stage small cell lung cancer (ES-SCLC) is a highly aggressive malignancy with poor prognosis. This study aimed to assess the efficacy of combining immunotherapy (IT) with Anlotinib in ES-SCLC patients.MethodsThis study was a multicenter retrospective cohort analysis. Survival outcomes were evaluated using Kaplan-Meier curves and Cox proportional hazards regression models.ResultsA total of 147 patients were included in the analysis. The median overall survival (mOS) for the cohort was 15.5 months (95% CI: 13.9-17.1). Patients in the chemotherapy(CT) plus IT group had an mOS of 17.8 months, compared to 12.6 months in the CT-alone group (p = 0.055). When stratified into CT + IT + Anlotinib, CT + IT, and CT-alone groups, the mOS were 18.5, 16.3, and 12.6 months, respectively, with the CT + IT + Anlotinib group demonstrating significantly improved OS compared to CT-alone (p = 0.044). The ORR and DCR for the entire cohort were 71.4% and 85.7%, respectively. Subgroup analysis revealed ORRs of 74.1% (CT + IT + Anlotinib), 73.9% (CT + IT), and 70.1% (CT-alone), with corresponding DCRs of 92.6%, 91.3%, and 82.5%. Multivariate analysis revealed that radiotherapy (RT, p = 0.003) and IT (p = 0.021) were independent prognostic factors for OS, while liver metastasis (p = 0.023) and RT (p = 0.018) were associated with PFS. Patients receiving RT in combination with CT showed markedly improved OS (17.5 vs 12.5 months; p = 0.002) and PFS (7.3 vs 6.3 months; p = 0.004). The incidence of adverse events was comparable across all groups (p = 0.721).ConclusionThe combined application of Anlotinib with IT and the combination of CT with RT both significantly improved survival outcomes in patients with ES-SCLC while maintaining a favorable safety profile. These findings warrant further investigation in future studies.
Collapse
Affiliation(s)
- Guogang Gao
- Department of Thoracic Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, 70 Heping Road, Weihai 264200, Shandong, China
| | - Meiling Sun
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107 Wenhua Xilu, Jinan 250012, Shandong, China
- Department of Respiratory Medicine, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, 70 Heping Road, Weihai 264200, Shandong, China
| | - Zhongfei Yang
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University Dezhou Hospital, 1166 Dong Fang Hong west road, Dezhou, Shandong 253000, China
| | - Jingyi Li
- Department of Radiation Oncology,Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, 70 Heping Road, Weihai 264200, Shandong, China
| | - Huaijun Ji
- Department of Thoracic Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, 70 Heping Road, Weihai 264200, Shandong, China
| | - Ge Yu
- Department of Thoracic Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, 70 Heping Road, Weihai 264200, Shandong, China
| |
Collapse
|
22
|
Su F, Fan WX, Zhang Y, Zhang XL, Du YY, Li WL, Hu WQ, Zhao J. A systematic review of gastritis as an immune-related adverse event in clinical interventions. Hum Vaccin Immunother 2024; 20:2408852. [PMID: 39434209 PMCID: PMC11497991 DOI: 10.1080/21645515.2024.2408852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/10/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) are crucial in cancer treatment, and the associated immune-related adverse events (irAEs) have garnered significant attention, yet reports on associated immune related gastritis are limited. The diagnosis of immune related gastritis remains predominantly exclusionary, meanwhile its management diverges significantly from that of conventional gastritis. Current guidelines lack standardized grading criteria, and substantial data from large-scale, tertiary clinical studies are absent, therefore we conducted a systematic review of Medline, Web of Science, and Embase databases, identifying 31 articles from 2017 to December 31, 2023, involving 258 patients. Clinical manifestations included epigastric pain (53.1%), mucosal erythema (56.1%), and lymphocyte infiltration (48.6%). Corticosteroid therapy was common (94.7%), with 86.7% experiencing post-treatment improvement. 80% of patients can be diagnosed through endoscopy and pathology, while the remaining 20% may require PET-CT. Hormonal therapy is favored but diverges from standard management. Accurate diagnosis is crucial in managing immune related gastritis effectively.
Collapse
Affiliation(s)
- Fei Su
- Department of Graduate School, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Wen-Xuan Fan
- Department of Graduate School, Shanxi Medical University, Taiyuan, Shanxi, China
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Yan Zhang
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Department of Graduate School, Changzhi Medical College, Changzhi, Shanxi, China
| | - Xiao-Ling Zhang
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Yun-Yi Du
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Wei-Ling Li
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
- Department of Graduate School, Changzhi Medical College, Changzhi, Shanxi, China
| | - Wen-Qing Hu
- Department of Gastrointestinal Surgery, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| | - Jun Zhao
- Department of Oncology, Changzhi People’s Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China
| |
Collapse
|
23
|
Kesari S, Wojcinski A, Pabla S, Seager RJ, Gill JM, Carrillo JA, Wagle N, Park DJ, Nguyen M, Truong J, Takasumi Y, Chaiken L, Chang SC, Barkhoudarian G, Kelly DF, Juarez TM. Pre-radiation Nivolumab plus ipilimumab in patients with newly diagnosed high-grade gliomas. Oncoimmunology 2024; 13:2432728. [PMID: 39572979 PMCID: PMC11587836 DOI: 10.1080/2162402x.2024.2432728] [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: 06/17/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024] Open
Abstract
The limited success of immune checkpoint inhibitors (ICIs) in the adjuvant setting for glioblastoma highlights the need to explore administering ICIs prior to immunosuppressive radiation. To address the feasibility and safety of this approach, we conducted a phase I study in patients with newly diagnosed Grade 3 and Grade 4 gliomas. Patients received nivolumab 300 mg every 2 weeks and ipilimumab 1 mg/kg every 6 weeks until disease progression or unacceptable toxicity. Fifteen patients were treated, with four patients on dexamethasone at treatment initiation and five tumors having MGMT promoter methylated. Treatment began a median of 38 days post-surgery. The most common treatment-related adverse events (AEs) were rash, pruritus, fatigue, nausea, and anorexia. Grade 3 AEs were lipase increased (n = 2), anorexia (n = 1), pruritus (n = 1), and rash (n = 3), and one Grade 4 cerebral edema occurred. Median progression-free survival (mPFS) was 1.3 months and median overall survival (mOS) was 19.3 months (95% CI, 12.9-NA). Three patients deferred conventional radiochemotherapy for over seven months while ten eventually received it. Progressing tumors tended to exhibit higher LAG-3 levels at baseline compared to shrinking tumors. Analysis of paired pre-treatment and post-progression tissue (n = 5) showed trends of up-regulated TGF-β, ERBB2, ERBB3, and ERBB4 signaling pathways, downregulated PPAR signaling, decreased B cell proportions, and increased monocytes proportions in tumors post-treatment. We show nivolumab plus ipilimumab can be safely administered prior to standard radiotherapy for newly diagnosed gliomas and is operationally feasible. Clinicaltrials.gov NCT03425292 registered February 7, 2018.
Collapse
Affiliation(s)
- Santosh Kesari
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
| | | | | | | | - Jaya M. Gill
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
| | - Jose A. Carrillo
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
| | - Naveed Wagle
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
| | - David J. Park
- Providence St. Jude Medical Center, Department of Hematology and Oncology, Fullerton, CA, USA
| | - Minhdan Nguyen
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
| | - Judy Truong
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
| | - Yuki Takasumi
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
- Providence Saint John’s Health Center, Department of Pathology, Santa Monica, CA, USA
| | - Lisa Chaiken
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
- Providence Saint John’s Health Center, Department of Radiology, Santa Monica, CA, USA
| | - Shu-Ching Chang
- Providence St. Vincent Medical Center, Clinical Research Program Services, Portland, OR, USA
| | | | - Daniel F. Kelly
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
| | - Tiffany M. Juarez
- Pacific Neuroscience Institute, Neuro-Oncology, Santa Monica, CA, USA
- Saint John’s Cancer Institute, Translational Neurosciences, Santa Monica, CA, USA
- CureScience Institute, San Diego, CA, USA
| |
Collapse
|
24
|
Nie Y, Schalper KA, Chiang A. Mechanisms of immunotherapy resistance in small cell lung cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:55. [PMID: 39802951 PMCID: PMC11724353 DOI: 10.20517/cdr.2024.154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/05/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025]
Abstract
Small-cell lung cancer (SCLC) is an aggressive neuroendocrine tumor with a poor prognosis. Although the addition of immunotherapy to chemotherapy has modestly improved outcomes, most patients rapidly develop resistance. Resistance to immunotherapy can be broadly categorized into primary resistance and acquired resistance, as proposed by the Society for Immunotherapy of Cancer (SITC) consensus definition. Primary resistance occurs in the setting of failure to respond to immune checkpoint inhibitors (ICIs), while acquired resistance develops after initial response. The mechanisms of acquired and primary resistance to ICI are not well understood in SCLC, denoting an area of critical unmet need. Both intrinsic and extrinsic mechanisms play significant roles in immunotherapy resistance. Intrinsic mechanisms include defects in antigen presentation, mutations in key genes, reduced tumor immunogenicity, and epigenetic alterations. Extrinsic mechanisms involve the tumor microenvironment (TME), which is a complex interplay of both tumor- and immunosuppressive immune cells, vasculature, and microbiome. An understanding of these resistance mechanisms is crucial for developing novel therapeutic strategies to advance effective immunotherapy in patients with SCLC, a critical area of unmet need.
Collapse
Affiliation(s)
- Yunan Nie
- Department of Medical Oncology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Kurt A. Schalper
- Department of Medical Oncology, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT 06510, USA
| | - Anne Chiang
- Department of Medical Oncology, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
25
|
Wang B, Zhang J, Shi Y, Wang Y. Clinical significance of the combined systemic immune-inflammatory index and prognostic nutritional index in predicting the prognosis of patients with extensive-stage small-cell lung cancer receiving immune-combination chemotherapy. BMC Cancer 2024; 24:1574. [PMID: 39719567 DOI: 10.1186/s12885-024-13343-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 12/13/2024] [Indexed: 12/26/2024] Open
Abstract
BACKGROUND The therapeutic efficacy and prognosis of various tumors can be assessed using the systemic immune-inflammatory index (SII) and prognostic nutritional index (PNI). Despite their potential, no studies have investigated the prognostic value of the combined SII-PNI score for outcomes in patients with extensive small cell lung cancer (ES-SCLC) treated with chemotherapy and immune checkpoint inhibitors (ICIs). MATERIALS AND METHODS Our study retrospectively examined 213 ES-SCLC patients treated with chemotherapy and ICIs across two institutions. The patients were divided into three groups based on their SII-PNI scores. Cox regression analysis was employed to identify independent prognostic factors. A nomogram was constructed based on these independent factors. With 1000 repeated samples, the bootstrap method was used to validate the nomogram model internally. The model's performance was assessed using calibration curves, receiver operating characteristic (ROC) curves, and decision curve analysis (DCA). RESULT Before and after chemotherapy with immune checkpoint inhibitors (ICIs), SII was significantly higher in the PD group compared with the PR group (both p < 0.05). In the meantime, PNI was considerably lower in the PD group than in the PR group (both p < 0.01). Kaplan-Meier curves demonstrated that patients with a low SII-PNI had prolonged progression-free survival (PFS) and overall survival (OS) compared to those with a high SII-PNI (all p < 0.01). Multivariate Cox analysis showed that PS = 1, bone metastasis, brain metastasis, and SII-PNI = 1,2 after four treatment cycles were independent risk factors for shorter OS and were included in the nomogram model. The ROC curves, C-index, and DCA curves confirm that the SII-PNI scores-based nomograms have strong predictive accuracy for OS. CONCLUSION There was a significant correlation between pre- and post-treatment SII-PNI and treatment effect in ES-SCLC. The SII-PNI score after four treatment cycles is a useful prognostic indicator for ES-SCLC patients receiving chemotherapy combined with immune checkpoint inhibitors (ICIs).
Collapse
Affiliation(s)
- Bingbing Wang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150000, China
| | - Jingdan Zhang
- Department of Gastroenterology, Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, 028000, China
| | - Yingnan Shi
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150000, China
| | - Yan Wang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150000, China.
| |
Collapse
|
26
|
Hosseininejad-Chafi M, Eftekhari Z, Oghalaie A, Behdani M, Sotoudeh N, Kazemi-Lomedasht F. Nanobodies as innovative immune checkpoint modulators: advancing cancer immunotherapy. Med Oncol 2024; 42:36. [PMID: 39719469 DOI: 10.1007/s12032-024-02588-y] [Citation(s) in RCA: 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] [Accepted: 12/14/2024] [Indexed: 12/26/2024]
Abstract
The immune system relies on a delicate balance between attacking harmful pathogens and preserving the body's own tissues, a balance maintained by immune checkpoints. These checkpoints play a critical role in preventing autoimmune diseases by restraining excessive immune responses while allowing the immune system to recognize and destroy abnormal cells, such as tumors. In recent years, immune checkpoint inhibitors (ICIs) have become central to cancer therapy, enabling the immune system to target and eliminate cancer cells that evade detection. Traditional antibodies, such as IgGs, have been widely used in immune therapies but are limited by their size and complexity. Nanobodies (Nbs), derived from camelid heavy-chain-only antibodies, offer a promising alternative. These small, stable antibody fragments retain the antigen-binding specificity of traditional antibodies but have enhanced solubility and the ability to target otherwise inaccessible epitopes. This review explores the use of Nbs as ICIs, emphasizing their potential in cancer immunotherapy and other immune-related treatments. Their unique structural properties and small size make Nbs highly effective tools for modulating immune responses, representing a novel approach in the evolving landscape of checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Mohammad Hosseininejad-Chafi
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Zohre Eftekhari
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Akbar Oghalaie
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Nazli Sotoudeh
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran.
| |
Collapse
|
27
|
Ge Y, Liu X, Xu Y, Su Y, Li Y, Wang L. Combined systemic immune-inflammatory index and prognostic nutritional index predicts the efficacy and prognosis of ES-SCLC patients receiving PD-L1 inhibitors combined with first-line chemotherapy. Front Oncol 2024; 14:1485849. [PMID: 39697233 PMCID: PMC11652344 DOI: 10.3389/fonc.2024.1485849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 11/12/2024] [Indexed: 12/20/2024] Open
Abstract
Background There is a strong association between inflammation and the formation, progression, and metastasis of malignant tumors, according to earlier studies. Some composite inflammation-nutritional indicators, such as the systemic immune-inflammation index (SII) and the prognostic nutritional index (PNI), have a certain predictive effect on the prognosis of patients with small cell lung cancer (SCLC). However, the relationship between these indicators and the efficacy of immunotherapy in SCLC patients is still not well understood. Therefore, the purpose of this study was to explore how the pre-treatment SII-PNI score can predict the tumor response and prognosis of extensive-stage SCLC patients treated with PD-L1 inhibitors and first-line chemotherapy. Methods This research conducted a retrospective review of 70 ES- SCLC patients from December 2019 to January 2023. According to the SII-PNI score, all patients were categorized into three groups. Overall survival (OS) was assessed by implementing the Kaplan Meier and Cox regression models. In addition, we devised a nomogram and scrutinized its accuracy in prediction through receiver operating characteristic (ROC) curve analysis and visualized it by calibration plots. Subsequently, a risk classification system was established. Results Patients with higher SII-PNI scores exhibited notably poorer survival outcomes compared to their counterpart with low SII-PNI score (p=0.008), as well as poorer short-term curative effects (p=0.004). The results of the multivariate analysis revealed that the SII-PNI score (p=0.036) had an independent association with a less favorable OS. The nomogram has been demonstrated to be a reliable prognostic tool for ES-SCLC patients. A notable difference was identified between the two different levels of risk. Conclusion The baseline SII-PNI score can serve as a reliable prognostic indicator for ES-SCLC patients receiving immunotherapy. Higher SII-PNI scores imply a worse prognosis.
Collapse
Affiliation(s)
- Yi Ge
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoyu Liu
- Department of Oncology, Luohe Central Hospital, Luohe, China
| | - Yishi Xu
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanwei Su
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yixin Li
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liping Wang
- Department of Oncology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
28
|
Hockemeyer KG, Rusthoven CG, Pike LRG. Advances in the Management of Lung Cancer Brain Metastases. Cancers (Basel) 2024; 16:3780. [PMID: 39594735 PMCID: PMC11593022 DOI: 10.3390/cancers16223780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/01/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Lung cancer, both non-small cell and small cell, harbors a high propensity for spreading to the central nervous system. Radiation therapy remains the backbone of the management of brain metastases. Recent advances in stereotactic radiosurgery have expanded its indications and ongoing studies seek to elucidate optimal fractionation and coordination with systemic therapies, especially targeted inhibitors with intracranial efficacy. Efforts in whole-brain radiotherapy aim to preserve neurocognition and to investigate the need for prophylactic cranial irradiation. As novel combinatorial strategies are tested and prognostic/predictive biomarkers are identified and tested, the management of brain metastases in lung cancer will become increasingly personalized to optimally balance intracranial efficacy with preserving neurocognitive function and patient values.
Collapse
Affiliation(s)
- Kathryn G. Hockemeyer
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Chad G. Rusthoven
- Department of Radiation Oncology, University of Colorado, Aurora, CO 80045, USA
| | - Luke R. G. Pike
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
29
|
Lo JW, Schroeder JH, Roberts LB, Mohamed R, Cozzetto D, Beattie G, Omer OS, Ross EM, Heuts F, Jowett GM, Read E, Madgwick M, Neves JF, Korcsmaros T, Jenner RG, Walker LSK, Powell N, Lord GM. CTLA-4 expressing innate lymphoid cells modulate mucosal homeostasis in a microbiota dependent manner. Nat Commun 2024; 15:9520. [PMID: 39496592 PMCID: PMC11535242 DOI: 10.1038/s41467-024-51719-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/10/2024] [Indexed: 11/06/2024] Open
Abstract
The maintenance of intestinal homeostasis is a fundamental process critical for organismal integrity. Sitting at the interface of the gut microbiome and mucosal immunity, adaptive and innate lymphoid populations regulate the balance between commensal micro-organisms and pathogens. Checkpoint inhibitors, particularly those targeting the CTLA-4 pathway, disrupt this fine balance and can lead to inflammatory bowel disease and immune checkpoint colitis. Here, we show that CTLA-4 is expressed by innate lymphoid cells and that its expression is regulated by ILC subset-specific cytokine cues in a microbiota-dependent manner. Genetic deletion or antibody blockade of CTLA-4 in multiple in vivo models of colitis demonstrates that this pathway plays a key role in intestinal homeostasis. Lastly, we have found that this observation is conserved in human IBD. We propose that this population of CTLA-4-positive ILC may serve as an important target for the treatment of idiopathic and iatrogenic intestinal inflammation.
Collapse
Affiliation(s)
- Jonathan W Lo
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, UK
| | | | - Luke B Roberts
- School of Immunology and Microbial Sciences, King's College London, London, UK
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Rami Mohamed
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Domenico Cozzetto
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, UK
| | - Gordon Beattie
- CRUK City of London Centre Single Cell Genomics Facility, UCL Cancer Institute, University College London, London, UK
- Genomics Translational Technology Platform, UCL Cancer Institute, University College London, London, UK
| | - Omer S Omer
- School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Ellen M Ross
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection and Immunity, Royal Free Campus, London, UK
| | - Frank Heuts
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection and Immunity, Royal Free Campus, London, UK
| | - Geraldine M Jowett
- Centre for Host-Microbiome Interactions, King's College London, London, T, UK
- Centre for Craniofacial and Regenerative Biology, King's College London, London, UK
- Centre for Stem Cells & Regenerative Medicine, King's College London, London, UK
| | - Emily Read
- Centre for Host-Microbiome Interactions, King's College London, London, T, UK
| | - Matthew Madgwick
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, UK
- Earlham Institute, Norwich Research Park, Norwich, UK
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Joana F Neves
- Centre for Host-Microbiome Interactions, King's College London, London, T, UK
| | - Tamas Korcsmaros
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, UK
- Earlham Institute, Norwich Research Park, Norwich, UK
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Richard G Jenner
- UCL Cancer Institute and CRUK City of London Centre, University College London, London, UK
| | - Lucy S K Walker
- Institute of Immunity & Transplantation, Pears Building, University College London Division of Infection and Immunity, Royal Free Campus, London, UK
| | - Nick Powell
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London, UK.
| | - Graham M Lord
- School of Immunology and Microbial Sciences, King's College London, London, UK.
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| |
Collapse
|
30
|
Tanaka A, Teranishi S, Kajita Y, Hirose T, Kaneko A, Sairenji Y, Kawashima H, Yumoto K, Tsukahara T, Miura K, Kobayashi N, Yamamoto M, Nishihira R, Kudo M, Miyazawa N, Nishikawa M, Kaneko T. Total baseline tumor size predicts survival among patients with advanced small-cell lung cancer receiving chemotherapy plus programmed death-ligand 1 inhibitor as first-line therapy: a multicenter retrospective observational study. Front Oncol 2024; 14:1400277. [PMID: 39558947 PMCID: PMC11570404 DOI: 10.3389/fonc.2024.1400277] [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: 03/13/2024] [Accepted: 10/18/2024] [Indexed: 11/20/2024] Open
Abstract
Introduction Total baseline tumor size (BTS) is a prognostic factor for programmed death 1 and programmed death-ligand 1 (PD-L1) inhibitor treatments. However, the prognostic value of total BTS for patients with small-cell lung cancer (SCLC) who receive chemotherapy plus PD-L1 inhibitor remains unknown. Thus, in this study, we aimed to determine whether total BTS is associated with prognosis in patients with SCLC who receive chemotherapy plus PD-L1 inhibitor as first-line therapy. Methods This study included patients with extensive-stage SCLC or post-chemoradiotherapy recurrence of limited-stage SCLC who received chemotherapy plus PD-L1 inhibitor as first-line therapy from August 2019 to December 2022. The two lesions with the largest diameter among the measurable lesions in each organ were selected from up to five organs (maximum of 10 lesions), and the sum of all diameters was defined as total BTS. The patients were divided into two groups, large or small, with total BTS using X-tile software. Median survival was analyzed using the Kaplan-Meier method, and the groups were compared using the log-rank test. Univariate and multivariate analyses examined the association between total BTS and prognosis. Results Fifty patients were included; 14% had large total BTS (>183.2 mm) and 86% had small total BTS (≤183.2 mm). The median observation period was 10.5 months. The large total BTS group showed significantly worse overall survival than the small total BTS group (median: 26.8 months vs. 5.7 months, P = 0.0003). The multivariate analysis indicated that large total BTS was an independent negative predictor of overall survival (hazard ratio: 7.14, 95% confidence interval: 1.89-26.96). Discussion Total BTS is a potentially useful prognostic factor for patients with advanced SCLC who receive chemotherapy plus PD-L1 inhibitor as first-line therapy.
Collapse
Affiliation(s)
- Anna Tanaka
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Shuhei Teranishi
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Yukihito Kajita
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Tomofumi Hirose
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Ayami Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yu Sairenji
- Department of Respiratory Medicine, Yokohama Sakae Kyosai Hospital, Yokohama, Japan
| | | | - Kentaro Yumoto
- Department of Respiratory Medicine, Yokohama Minami Kyosai Hospital, Yokohama, Japan
| | - Toshinori Tsukahara
- Department of Respiratory Medicine, Chigasaki Municipal Hospital, Chigasaki, Japan
| | - Kenji Miura
- Department of Respiratory Medicine, Yokohama Sakae Kyosai Hospital, Yokohama, Japan
| | - Nobuaki Kobayashi
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masaki Yamamoto
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Ryuichi Nishihira
- Department of Respiratory Medicine, Kanto Rosai Hospital, Kawasaki, Japan
| | - Makoto Kudo
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Naoki Miyazawa
- Department of Respiratory Medicine, Yokohama Nanbu Hospital, Yokohama, Japan
| | - Masanori Nishikawa
- Department of Respiratory Medicine, Fujisawa Municipal Hospital, Fujisawa, Japan
| | - Takeshi Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
31
|
Tang J, Wang T, Wu H, Bao X, Xu K, Ren T. Efficacy and toxicity of lurbinectedin in subsequent systemic therapy of extensive-stage small cell lung cancer: a meta-analysis. BMC Cancer 2024; 24:1351. [PMID: 39497053 PMCID: PMC11533368 DOI: 10.1186/s12885-024-13104-w] [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: 07/11/2024] [Accepted: 10/24/2024] [Indexed: 11/06/2024] Open
Abstract
OBJECTIVE This study aimed to systematically analyze the efficacy and toxicity of lurbinectedin as a second-line or subsequent treatment for extensive-stage small cell lung cancer (ES-SCLC). METHODS Candidate studies were identified in PubMed, Embase, Cochrane Library, ClinicalTrials.gov, CNKI, and Wanfang databases up to 1 May 2024. Objective remission rate (ORR), disease control rate (DCR), duration of response (DOR), progression-free survival (PFS), overall survival (OS), and adverse events (AEs) were extracted, respectively. The efficacy and toxicity of lurbinectedin in ES-SCLC were analyzed by meta-analysis. RESULTS Six eligible prospective studies were included in this meta-analysis, including 536 patients with ES-SCLC who received second-line or subsequent treatment. In pooled analysis, the ORR of lurbinectedin was 35% (95% confidence interval [CI] 29-41), DCR was 67% (95%CI 58-76), DOR was 5.33 months (95%CI 4.51-6.16), PFS was 3.38 months (95%CI 2.59-4.17), and OS was 7.49 months (95%CI 5.11-9.87). The incidence of AEs and severe adverse events (SAEs) was 92% (95%CI 78-100) and 37% (95%CI 19-57), respectively. The most common AEs were leukopenia, neutropenia, anemia, and thrombocytopenia, with incidences of 81% (68-91), 74% (57-88), 73% (35-98) and 57% (46-68), respectively. CONCLUSION As a promising alternative for second-line treatment for ES-SCLC, lurbinectedin has a certain level of efficacy and a favorable safety profile. The integration of lurbinectedin with other therapeutic modalities presents an emerging area warranting further investigation.
Collapse
Affiliation(s)
- Jiayi Tang
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Tianlei Wang
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Hongwei Wu
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Xinrui Bao
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China
| | - Ke Xu
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China.
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China.
| | - Tao Ren
- Clinical Medical College, Chengdu Medical College, Chengdu, Sichuan, China.
- Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, China.
| |
Collapse
|
32
|
Velut Y, Arqué B, Wislez M, Blons H, Burroni B, Prieto M, Beau S, Fournel L, Birsen G, Cremer I, Alifano M, Damotte D, Mansuet-Lupo A. The tumor immune microenvironment of SCLC is not associated with its molecular subtypes. Eur J Cancer 2024; 212:115067. [PMID: 39413714 DOI: 10.1016/j.ejca.2024.115067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/28/2024] [Accepted: 10/10/2024] [Indexed: 10/18/2024]
Abstract
INTRODUCTION Small-cell lung carcinoma (SCLC) is a high-grade neuroendocrine carcinoma of poor prognosis. Although immune checkpoint blockers have shown promising results in advanced SCLC, the tumor immune microenvironment (TME) remains poorly understood, with no validated prognostic or predictive biomarkers of efficacy. METHODS This retrospective study included surgically samples from 48 SCLC patients between 2009 and 2018. We assessed the TME using two quantitative 7-plex immunofluorescence panels focusing on T and B cells, and compared it to NSCLC (N = 10). Molecular subtypes were determined by assessing the expression of ASCL1, NEUROD1 and YAP1 using immunohistochemistry. RESULTS Immune-hot SCLC were defined as those exhibiting the highest immune cell and immune-related marker densities. They were associated with longer overall survival, significantly more frequently detected at early stages, and exhibited high PD-L1 expression in immune cells, but were not associated with molecular subtypes. Compared to NSCLC, SCLC had significantly lower densities of CD20 + cells and higher density of PD1 + cells, with no significant differences in CD4 + , CD8 + and plasma cell densities. In univariate analysis, the highest OS was significantly associated with early stage (p < 0.001), low expression of NEUROD1 (p = 0.047), high PD1 + cell density (p < 0.001) and high PD-L1 immune cell expression (p = 0.04). Only stage and PD1 + cell density emerged as independent prognostic markers. CONCLUSION SCLC TME is highly heterogeneous. Immune-hot tumors were associated with OS but not with molecular classification. PD1 expression and PD-L1 expression by immune cells may thus serve as a prognostic marker.
Collapse
Affiliation(s)
- Yoan Velut
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France
| | - Basilia Arqué
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France
| | - Marie Wislez
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France; Department of pneumology, Cochin Hospital, AP-HP.centre, Université Paris Cité, Paris, France
| | - Hélène Blons
- Centre de Recherche des Cordeliers, INSERM CNRS SNC 5096, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Paris, France; Department of Biochemistry, Unit of Pharmacogenetic and Molecular Oncology, Georges Pompidou European Hospital, AP-HP.centre, Université Paris Cité, Paris, France
| | - Barbara Burroni
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France; Department of Pathology, Cochin Hospital, AP-HP.centre, Université Paris Cité, Paris, France
| | - Mathilde Prieto
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France; Department of Thoracic Surgery, Cochin Hospital, AP-HP.centre, Université Paris Cité, Paris, France
| | - Siméon Beau
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France
| | - Ludovic Fournel
- Department of Thoracic Surgery, Cochin Hospital, AP-HP.centre, Université Paris Cité, Paris, France
| | - Gary Birsen
- Department of pneumology, Cochin Hospital, AP-HP.centre, Université Paris Cité, Paris, France
| | - Isabelle Cremer
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France
| | - Marco Alifano
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France; Department of Thoracic Surgery, Cochin Hospital, AP-HP.centre, Université Paris Cité, Paris, France
| | - Diane Damotte
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France; Department of Pathology, Cochin Hospital, AP-HP.centre, Université Paris Cité, Paris, France.
| | - Audrey Mansuet-Lupo
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université Paris Cité, Institut du cancer Paris CARPEM, Team Inflammation, Complement and Cancer, Paris, France; Department of Pathology, Cochin Hospital, AP-HP.centre, Université Paris Cité, Paris, France
| |
Collapse
|
33
|
Yin L, Sun P, Guo S, Shuai P, Zhang J. CAR-T cell therapy: Challenge and opportunity for effective treatment of small cell lung cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189228. [PMID: 39615863 DOI: 10.1016/j.bbcan.2024.189228] [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: 11/07/2024] [Accepted: 11/24/2024] [Indexed: 12/08/2024]
Abstract
Small cell lung cancer (SCLC) is a devastating malignancy characterized by rapid metastasis, drug resistance, and frequent recurrence. Owing to the paucity of existing therapeutic options, the prognosis of SCLC remains poor. Recently, the combination of immune checkpoint inhibitors and chemotherapy has resulted in modest improvements in treatment responses. In this review, we characterize the biological signature of SCLC and outline the obstacles to current treatment, including impaired antigen presentation and T cell infiltration. These obstacles may potentially be overcome by chimeric antigen receptor (CAR)-T cell therapy. For the first time, we summarize the available data and discuss the future prospects of CAR-T cell therapy for the treatment of SCLC. Given the high heterogeneity and immunosuppressive tumor microenvironment of SCLC, structural modifications of CAR-T cells and combination therapy may be required to elicit a successful antitumor response. Further research, including clinical trials, is needed to determine the suitability of CAR-T cell therapy as a treatment for SCLC.
Collapse
Affiliation(s)
- Limei Yin
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ping Sun
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Shujin Guo
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ping Shuai
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Junlin Zhang
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
34
|
Li C, Lv P, Yanyan L, Yin M, Li H. Immune checkpoint inhibitor therapy for primary neuroendocrine carcinoma of the gallbladder: A case report and literature review. Medicine (Baltimore) 2024; 103:e40178. [PMID: 39470513 PMCID: PMC11521044 DOI: 10.1097/md.0000000000040178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 10/03/2024] [Indexed: 10/30/2024] Open
Abstract
RATIONALE Gallbladder neuroendocrine carcinoma (GNEC) is indeed a relatively rare malignant tumor of the gallbladder with neuroendocrine differentiation and the ability to produce and secrete a number of neurotransmitters and hormones, characteristics that make its clinical presentation and biological behavior likely to be different from those of other types of gallbladder cancer. Current treatment mostly relies on surgery and adjuvant chemotherapy and radiotherapy. PATIENT CONCERNS We report a 53-year-old middle-aged male patient who underwent radical surgery for gallbladder malignancy after a diagnosis of neuroendocrine carcinoma of the gallbladder. DIAGNOSES Diagnosis of neuroendocrine carcinoma of the gallbladder based on the return of pathologic findings. INTERVENTION After local progression of postoperative chemotherapy with the first-line regimen of etoposide + cisplatin, an immune checkpoint inhibitor (traplizumab) + FOLIFIRI (fluorouracil + calcium folinate + irinotecan) regimen was used. OUTCOMES The patient achieved 20 months of clinical survival and ultimately died of myelosuppression. LESSONS The use of immune checkpoint inhibitors may become an effective tool in the treatment of neuroendocrine carcinoma of the gallbladder.
Collapse
Affiliation(s)
- Chao Li
- Hepatobiliary and Pancreatic Medicine Center, Weifang Peoples Hospital, Weifang, Shandong, P.R. China
| | - Pan Lv
- Hepatobiliary and Pancreatic Medicine Center, Weifang Peoples Hospital, Weifang, Shandong, P.R. China
| | - Liu Yanyan
- Hepatobiliary and Pancreatic Medicine Center, Weifang Peoples Hospital, Weifang, Shandong, P.R. China
| | - Maohui Yin
- Hepatobiliary and Pancreatic Medicine Center, Weifang Peoples Hospital, Weifang, Shandong, P.R. China
| | - Hao Li
- Hepatobiliary and Pancreatic Medicine Center, Weifang Peoples Hospital, Weifang, Shandong, P.R. China
| |
Collapse
|
35
|
Zhao Q, Wang C, Huang W, Song Z, Lang Y, Zhu X. Potential immunologic and prognostic roles of CHRNA6 in SCLC and pan-cancer. Heliyon 2024; 10:e38572. [PMID: 39398083 PMCID: PMC11470509 DOI: 10.1016/j.heliyon.2024.e38572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/22/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024] Open
Abstract
Background Small cell lung cancer (SCLC) is considered the most malignant subtype of lung cancer, and it has a restricted range of therapeutic choices. The emergence of immunotherapy has offered new possibilities for patients with SCLC. However, the scarcity of clinical specimens has hampered the progress of clinical studies and we still face a shortage of dependable indicators to forecast the effectiveness of immunotherapy for SCLC. Methods In our study, we assessed the ImmuneScore and StromalScore of 81 SCLC samples obtained from the cBioPortal database. By comparing gene expression differences between the high and low immune scores groups, we identified 24 differentially expressed genes. Subsequently, an intersection was performed with genes that exhibited differential expression between normal and SCLC tissues, leading us to isolate the gene CHRNA6. To gain a deeper insight into the possible significance of CHRNA6 in SCLC, we singled out 50 genes that showed the most pronounced positive and negative associations with its expression. We then pinpointed hub genes for subsequent functional enrichment analyses by establishing a protein-protein interactions network. We additionally assessed the link between CHRNA6 expression in SCLC and characteristics of the immune microenvironment, along with the efficacy of immunotherapy, using the CIBERSORT, immunophenoscores (IPS), and tumor immune dysfunction and exclusion (TIDE) algorithms. Furthermore, we confirmed the prognostic impact of CHRNA6 expression in SCLC patients undergoing immunotherapy within a clinical cohort. Lastly, we obtained data from The Cancer Genome Atlas (TCGA) to investigate CHRNA6 expression in various tumors and its associations with genetic alterations, DNA methylation, copy number variation, clinicopathological characteristics, biological processes, immune microenvironment, prognosis, and drug sensitivity. Results In SCLC, we found that CHRNA6 function was associated with immune activation pathways such as antigen presentation processing and positive regulation of adaptive immune response, and that CHRNA6 demonstrated a strong correlation with immune cells infiltration. In addition, analysis of the clinical cohort revealed that patients with SCLC who exhibited elevated expression of CHRNA6 experienced better responses to immunotherapy. Our pan-cancer analysis disclosed that the expression of CHRNA6 is dysregulated in a multitude of cancers, potentially due to genetic mutations, copy number gains, and DNA demethylation. The gene set enrichment analysis (GSEA) outcomes indicated that CHRNA6 participates in immune responses and may play a positive immune regulatory role in most cancers. Furthermore, CHRNA6 has been observed to have a notable relationship with immune checkpoints, immunomodulators, immune cell infiltration, patient outcomes, and drug sensitivity across various cancers. Conclusions Our findings indicate that the CHRNA6 may act as a predictive indicator for SCLC patients receiving immunotherapy. The study also uncovers the aberrant expression of CHRNA6 in a range of human cancers and its potential roles in immunology and prognosis, offering novel perspectives for tailored cancer therapies.
Collapse
Affiliation(s)
- Qingqing Zhao
- Department of Respiratory and Critical Care Medicine, Southeast University, Zhongda Hospital, Nanjing, 210009, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Cong Wang
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Wucui Huang
- Department of Respiratory and Critical Care Medicine, Southeast University, Zhongda Hospital, Nanjing, 210009, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Zhongquan Song
- Department of Respiratory and Critical Care Medicine, Southeast University, Zhongda Hospital, Nanjing, 210009, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yang Lang
- Department of Respiratory and Critical Care Medicine, Southeast University, Zhongda Hospital, Nanjing, 210009, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xiaoli Zhu
- Department of Respiratory and Critical Care Medicine, Southeast University, Zhongda Hospital, Nanjing, 210009, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| |
Collapse
|
36
|
Yu T, Lok BH. Strategies to Target Chemoradiotherapy Resistance in Small Cell Lung Cancer. Cancers (Basel) 2024; 16:3438. [PMID: 39456533 PMCID: PMC11506711 DOI: 10.3390/cancers16203438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Small cell lung cancer (SCLC) is a lethal form of lung cancer with few treatment options and a high rate of relapse. While SCLC is initially sensitive to first-line DNA-damaging chemo- and radiotherapy, relapse disease is almost universally therapy-resistant. As a result, there has been interest in understanding the mechanisms of therapeutic resistance in this disease. Conclusions: Progress has been made in elucidating these mechanisms, particularly as they relate to the DNA damage response and SCLC differentiation and transformation, leading to many clinical trials investigating new therapies and combinations. Yet there remain many gaps in our understanding, such as the effect of epigenetics or the tumor microenvironment on treatment response, and no single mechanism has been found to be ubiquitous, suggesting a significant heterogeneity in the mechanisms of acquired resistance. Nevertheless, the advancement of techniques in the laboratory and the clinic will improve our ability to study this disease, especially in patient populations, and identify methods to surmount therapeutic resistance.
Collapse
Affiliation(s)
- Tony Yu
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
| | - Benjamin H. Lok
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, 101 College Street, Toronto, ON M5G 1L7, Canada
- Radiation Medicine Program, Princess Margaret Cancer Centre, 610 University Ave, Toronto, ON M5G 2M9, Canada
- Department of Radiation Oncology, Temerty Faculty of Medicine, University of Toronto, 149 College Street, Toronto, ON M5T 1P5, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, 6 Queen’s Park Crescent, Toronto, ON M5S 3H2, Canada
| |
Collapse
|
37
|
Mo Y, Qin Y, Shangguan J, Wei D, Wu M, Chen D, Yu J. Sex-specific Difference for Small Cell Lung Cancer from Immunotherapy Advancement. Arch Bronconeumol 2024; 60 Suppl 2:S13-S21. [PMID: 38906714 DOI: 10.1016/j.arbres.2024.05.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND The treatment of lung cancer has witnessed significant progress, leading to improved survival rates among patients. It is important to assess the individual contributions of non-small cell lung cancer (NSCLC) and small-cell lung cancer (SCLC) to overall lung-cancer incidence and mortality trends based population, especially sex difference. METHODS We analyzed lung cancer mortality based on subtype, gender, and calendar year. The Joinpoint software was used to identify any changes in incidence and trends in mortality. RESULTS Incidence and incidence-based mortality declined from 2001 to 2019 both NSCLC and SCLC annually. The most significant decrease occurred between 2016 and 2019 with annual percent change of 5.71%. From 2012 to 2016, the incidence-based mortality of SCLC in women changed by 2.7% in tandem with incidence decreased 2.84%. Remarkably, the incidence-based mortality for women declined notably by 5.23% between 2016 and 2019, even as the incidence showed a less extent of decreasing (-2.59%). The survival rate for women was 15.2% in 2001, 19.3% in 2016, it had increased to 21.3% in 2018 but similar trends not in men. The survival curve showed the change in survival outcomes over time among men and women (median overall survival: 13 vs 23months) receiving immunotherapy for SCLC. CONCLUSION Population-level mortality from NSCLC and SCLC in the United States fell sharply from 2016 to 2019 as incidence deceased, and survival improved substantially. Our analysis suggests that approval for and use of immunotherapy may explain the mortality reduction observed during this period, with significant benefits especially for SCLC patient in women.
Collapse
Affiliation(s)
- You Mo
- Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yiwei Qin
- Cheeloo College of Medicine, Shandong University
| | - Jian Shangguan
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Duncan Wei
- Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Meng Wu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Dawei Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| | - Jinming Yu
- Laboratory of Molecular Cardiology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
38
|
Sun M, Ji H, Deng F, Li J, Xu N, Li Y. Clinical outcomes and synergistic effect between radiotherapy and immunotherapy in patients with extensive-stage small cell lung cancer: a real-world study. BMC Cancer 2024; 24:1206. [PMID: 39350057 PMCID: PMC11441094 DOI: 10.1186/s12885-024-12942-y] [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: 06/17/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Patients with extensive-stage small cell lung cancer (ES-SCLC) experience significant therapeutic challenges and limited survival rates. This study aimed to investigate the efficacy of combining immunotherapy (IT) with chemotherapy (CT) for treating ES-SCLC and to explore the synergistic effect between radiotherapy (RT) and IT. METHODS This retrospective analysis examined patients with ES-SCLC who received treatment at three centers. Furthermore, propensity score-matched (PSM) analysis was conducted. The Kaplan‒Meier method and Cox proportional hazards regression were used to compare the survival outcomes. RESULTS A total of 257 eligible patients with ES-SCLC were included in the analysis. Among all patients, the median overall survival (mOS) was 18.0 m in the chemoimmunotherapy (CT + IT) group and 15.7 m in the CT group (p = 0.208). The median real-world progression-free survival (mrwPFS) was 7.7 m and 6.8 m (p = 0.043) in the CT + IT and CT group, respectively. Moreover, the mOS was 22.0 m in the chemoradiotherapy (CT + RT) group and 13.6 m in the CT group (p < 0.001). The mrwPFS was 7.4 m and 6.0 m (p = 0.175) in the CT + RT group and CT group, respectively. The multivariate analyses revealed that sex, liver metastasis and RT were independent prognostic factors for OS (p < 0.05), while liver metastasis and IT were found to be independent predictive factors of real-world progression-free survival (rwPFS) (p < 0.05). After PSM, the mOS was 23.2 m in the CT + IT group and 13.0 m in the CT group (p = 0.008). The mrwPFS was 7.3 m and 6.2 m (p = 0.096) in the CT + IT group and the CT group, respectively. Moreover, the mOS was 21.4 m in the CT + RT group and 12.5 m in the CT group (p < 0.001). The mrwPFS was 7.3 m and 5.2 m (p = 0.220) in the CT + RT group and the CT group, respectively. Additionally, our study revealed that in the PD-1 group, RT significantly improved patient survival (36.0 m vs. 15.8 m, p = 0.041). CONCLUSION An increasing number of treatment options are being explored for ES-SCLC, and CT is the cornerstone of treatment for this disease. Combining CT with IT and RT has demonstrated remarkable efficacy and excellent safety profiles, and such treatments are worthy of further exploration.
Collapse
Affiliation(s)
- Meiling Sun
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107 Wenhua Xilu, Jinan, 250012, Shandong, China
- Department of Respiratory Medicine, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, 70 Heping Road, Weihai, 264200, Shandong, China
| | - Huaijun Ji
- Department of Thoracic Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, 70 Heping Road, Weihai, 264200, Shandong, China.
| | - Fang Deng
- Department of Oncology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, 254300, Shandong, China
| | - Jingyi Li
- Department of Radiation Oncology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, 70 Heping Road, Weihai, 264200, Shandong, China
| | - Ning Xu
- Department of Respiratory Medicine, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, 70 Heping Road, Weihai, 264200, Shandong, China
| | - Yu Li
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107 Wenhua Xilu, Jinan, 250012, Shandong, China.
| |
Collapse
|
39
|
Wang W, Wu G, Luo W, Lin L, Zhou C, Yao G, Chen M, Wu X, Chen Z, Ye J, Yang H, Lv D. Anlotinib plus oral fluoropyrimidine S-1 in refractory or relapsed small-cell lung cancer (SALTER TRIAL): a multicenter, single-arm, phase II trial. BMC Cancer 2024; 24:1182. [PMID: 39333988 PMCID: PMC11437909 DOI: 10.1186/s12885-024-12954-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Patients with small-cell lung cancer (SCLC) have few treatment options and dismal overall survival (OS) after failed platinum-based chemotherapy. METHODS The eligibility criteria of this phase II clinical trial included patients with measurable disease, age of 18 to 75 years, a confirmed diagnosis of disease progression or recurrence after prior platinum-based chemotherapy with a pathologically proven diagnosis of SCLC. Patients were treated with anlotinib at a dosage of 12 mg once daily (QD) and S-1 at 60 mg twice daily (BID) for 2 weeks, followed by a 1-week treatment-free interval. After six cycles of the above treatment, patients continued the maintenance therapy using S-1 monotherapy at 60 mg/ BID for 2 weeks, followed by a 1-week treatment-free interval until disease progression. RESULTS From March 2019 to June 2020, a total of 71 patients were initially assessed for eligibility in this study. Out of these, 52 patients who met the inclusion criteria were enrolled, and 48 patients received at least two doses of the study drug. The median follow-up time was 25.1 months. The ORR was seen in 21 patients (43.8%). The median PFS was 4.5 months (95% CI, 3.5-5.5 months), and the median OS was 5.9 months (95% CI, 4.6-7.3 months). The most common grade 3-4 treatment-related adverse events were thrombocytopenia (16.7%), anemia (14.6%), neutropenia (14.6%), and hypertension (10.4%). No treatment-related death occurred. CONCLUSIONS The combination of anlotinib with oral fluoropyrimidine S-1 demonstrated notable activity in relapsed or refractory SCLC, showing a favorable ORR and an acceptable, manageable safety profile. TRIAL REGISTRATION This trial was registered with ClinicalTrial.gov (NCT03823118) on 3 January 2019.
Collapse
Affiliation(s)
- Wei Wang
- Department of Radiation Oncology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, Zhejiang, 317000, China
- Department of Radiation Oncology, Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, Zhejiang, 317000, China
| | - Guixian Wu
- Department of Respiratory and Critical Care Medicine, Enze Hospital, Taizhou Enze Medical Center (Group), Taizhou, Zhejiang Province, 318050, China
| | - Wujun Luo
- Department of Respiratory and Critical Care Medicine, Sanmen People Hospital, Taizhou, Zhejiang Province, 317100, China
| | - Ling Lin
- Department of Respiratory and Critical Care Medicine, Enze Hospital, Taizhou Enze Medical Center (Group), Taizhou, Zhejiang Province, 318050, China
| | - Chao Zhou
- Department of Radiation Oncology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, Zhejiang, 317000, China
| | - Guifei Yao
- Department of Respiratory and Critical Care Medicine, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, Zhejiang Province, 317000, China
| | - Meifang Chen
- Department of Respiratory and Critical Care Medicine, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, Zhejiang Province, 317000, China
| | - Xiaomai Wu
- Department of Respiratory and Critical Care Medicine, Enze Hospital, Taizhou Enze Medical Center (Group), Taizhou, Zhejiang Province, 318050, China
| | - Ziran Chen
- Department of Radiation Oncology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, Zhejiang, 317000, China
| | - Junhui Ye
- Department of Respiratory and Critical Care Medicine, Sanmen People Hospital, Taizhou, Zhejiang Province, 317100, China.
- Department of Pulmonary Medicine, Sanmen People Hospital, Taizhou, Zhejiang Province, 317100, China.
| | - Haihua Yang
- Department of Radiation Oncology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, Zhejiang, 317000, China.
- Department of Radiation Oncology, Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, Zhejiang, 317000, China.
| | - Dongqing Lv
- Department of Respiratory and Critical Care Medicine, Enze Hospital, Taizhou Enze Medical Center (Group), Taizhou, Zhejiang Province, 318050, China.
| |
Collapse
|
40
|
Yan Y, Shen S, Li J, Su L, Wang B, Zhang J, Lu J, Luo H, Han P, Xu K, Shen X, Huang S. Cross-omics strategies and personalised options for lung cancer immunotherapy. Front Immunol 2024; 15:1471409. [PMID: 39391313 PMCID: PMC11465239 DOI: 10.3389/fimmu.2024.1471409] [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: 07/27/2024] [Accepted: 08/30/2024] [Indexed: 10/12/2024] Open
Abstract
Lung cancer is one of the most common malignant tumours worldwide and its high mortality rate makes it a leading cause of cancer-related deaths. To address this daunting challenge, we need a comprehensive understanding of the pathogenesis and progression of lung cancer in order to adopt more effective therapeutic strategies. In this regard, integrating multi-omics data of the lung provides a highly promising avenue. Multi-omics approaches such as genomics, transcriptomics, proteomics, and metabolomics have become key tools in the study of lung cancer. The application of these methods not only helps to resolve the immunotherapeutic mechanisms of lung cancer, but also provides a theoretical basis for the development of personalised treatment plans. By integrating multi-omics, we have gained a more comprehensive understanding of the process of lung cancer development and progression, and discovered potential immunotherapy targets. This review summarises the studies on multi-omics and immunology in lung cancer, and explores the application of these studies in early diagnosis, treatment selection and prognostic assessment of lung cancer, with the aim of providing more personalised and effective treatment options for lung cancer patients.
Collapse
Affiliation(s)
- Yalan Yan
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Siyi Shen
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Jiamin Li
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Lanqian Su
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Binbin Wang
- Intensive Care Unit, Xichong People’s Hospital, Nanchong, China
| | - Jinghan Zhang
- Department of Anaesthesiology, Southwest Medical University, Luzhou, China
| | - Jiaan Lu
- School of Clinical Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Huiyan Luo
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Ping Han
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Ke Xu
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Xiang Shen
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Shangke Huang
- Department of Oncology, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
41
|
Wang R, Li C, Cheng Z, Li M, Shi J, Zhang Z, Jin S, Ma H. H3K9 lactylation in malignant cells facilitates CD8 + T cell dysfunction and poor immunotherapy response. Cell Rep 2024; 43:114686. [PMID: 39216002 DOI: 10.1016/j.celrep.2024.114686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 07/05/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Histone lysine lactylation (Kla) is a post-translational modification, and its role in tumor immune escape remains unclear. Here, we find that increased histone lactylation is associated with poor response to immunotherapy in head and neck squamous cell carcinoma (HNSCC). H3K9la is identified as a specific modification site in HNSCC. Using cleavage under targets and tagmentation analyses, interleukin-11 (IL-11) is identified as a downstream regulatory gene of H3K9la. IL-11 transcriptionally activates immune checkpoint genes through JAK2/STAT3 signaling in CD8+ T cells. Additionally, IL-11 overexpression promotes tumor progression and CD8+ T cell dysfunction in vivo. Moreover, IL11 knockdown reverses lactate-induced CD8+ T cell exhaustion, and cholesterol-modified siIL11 restores CD8+ T cell killing activity and enhances immunotherapy efficacy. Clinically, H3K9la positively correlates with IL-11 expression and unfavorable immunotherapy responses in patients. This study reveals the crucial role of histone lactylation in immune escape, providing insights into immunotherapy strategies for HNSCC.
Collapse
Affiliation(s)
- Ruijie Wang
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; National Center for Stomatology & National Clinical Research Center for Oral Diseases, Shanghai 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Chuwen Li
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; National Center for Stomatology & National Clinical Research Center for Oral Diseases, Shanghai 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Zhongyi Cheng
- Jingjie PTM Biolab (Hangzhou), Hangzhou, Zhejiang 310018, China
| | - Mingyu Li
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; National Center for Stomatology & National Clinical Research Center for Oral Diseases, Shanghai 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Jianbo Shi
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; National Center for Stomatology & National Clinical Research Center for Oral Diseases, Shanghai 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Zhiyuan Zhang
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; National Center for Stomatology & National Clinical Research Center for Oral Diseases, Shanghai 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China
| | - Shufang Jin
- National Center for Stomatology & National Clinical Research Center for Oral Diseases, Shanghai 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China; Department of Second Dental Center, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine; College of Stomatology, Shanghai Jiao Tong University, Shanghai 200011, China.
| | - Hailong Ma
- Department of Oral Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China; National Center for Stomatology & National Clinical Research Center for Oral Diseases, Shanghai 200011, China; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, Shanghai 200011, China.
| |
Collapse
|
42
|
Jin Y, Wu Y, Reuben A, Zhu L, Gay CM, Wu Q, Zhou X, Mo H, Zheng Q, Ren J, Fang Z, Peng T, Wang N, Ma L, Fan Y, Song H, Zhang J, Chen M. Single-cell and spatial proteo-transcriptomic profiling reveals immune infiltration heterogeneity associated with neuroendocrine features in small cell lung cancer. Cell Discov 2024; 10:93. [PMID: 39231924 PMCID: PMC11375181 DOI: 10.1038/s41421-024-00703-x] [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: 09/27/2023] [Accepted: 06/23/2024] [Indexed: 09/06/2024] Open
Abstract
Small cell lung cancer (SCLC) is an aggressive pulmonary neuroendocrine malignancy featured by cold tumor immune microenvironment (TIME), limited benefit from immunotherapy, and poor survival. The spatial heterogeneity of TIME significantly associated with anti-tumor immunity has not been systemically studied in SCLC. We performed ultra-high-plex Digital Spatial Profiling on 132 tissue microarray cores from 44 treatment-naive limited-stage SCLC tumors. Incorporating single-cell RNA-sequencing data from a local cohort and published SCLC data, we established a spatial proteo-transcriptomic landscape covering over 18,000 genes and 60 key immuno-oncology proteins that participate in signaling pathways affecting tumorigenesis, immune regulation, and cancer metabolism across 3 pathologically defined spatial compartments (pan-CK-positive tumor nest; CD45/CD3-positive tumor stroma; para-tumor). Our study depicted the spatial transcriptomic and proteomic TIME architecture of SCLC, indicating clear intra-tumor heterogeneity dictated via canonical neuroendocrine subtyping markers; revealed the enrichment of innate immune cells and functionally impaired B cells in tumor nest and suggested potentially important immunoregulatory roles of monocytes/macrophages. We identified RE1 silencing factor (REST) as a potential biomarker for SCLC associated with low neuroendocrine features, more active anti-tumor immunity, and prolonged survival.
Collapse
Affiliation(s)
- Ying Jin
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Zhejiang Key Laboratory of Radiation Oncology, Hangzhou, Zhejiang, China
| | - Yuefeng Wu
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE), School of Medicine, Zhejiang University, Haining, Zhejiang, China
| | - Alexandre Reuben
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Liang Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Carl M Gay
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qingzhe Wu
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xintong Zhou
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haomin Mo
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qi Zheng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Junyu Ren
- College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhaoyuan Fang
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE), School of Medicine, Zhejiang University, Haining, Zhejiang, China
| | - Teng Peng
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Nan Wang
- Cosmos Wisdom Biotech Co. Ltd., Hangzhou, Zhejiang, China
| | - Liang Ma
- Department of Cardiovascular Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yun Fan
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
| | - Hai Song
- The MOE Key Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang, China.
- Center for Oncology Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, Zhejiang, China.
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Ming Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China.
- State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.
- United Laboratory of Frontier Radiotherapy Technology of Sun Yat-sen University & Chinese Academy of Sciences Ion Medical Technology Co., Ltd, Guangzhou, Guangdong, China.
| |
Collapse
|
43
|
Murayama T, Mahadevan NR, Meador CB, Ivanova EV, Pan Y, Knelson EH, Tani T, Nakayama J, Ma X, Thai TC, Hung YP, Kim W, Watanabe H, Cai KQ, Hata AN, Paweletz CP, Barbie DA, Cañadas I. Targeting TREX1 Induces Innate Immune Response in Drug-Resistant Small-Cell Lung Cancer. CANCER RESEARCH COMMUNICATIONS 2024; 4:2399-2414. [PMID: 39177280 PMCID: PMC11391691 DOI: 10.1158/2767-9764.crc-24-0360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 07/23/2024] [Accepted: 08/21/2024] [Indexed: 08/24/2024]
Abstract
Small-cell lung cancer (SCLC) is the most lethal type of lung cancer. Paradoxically, this tumor displays an initial exquisite response to chemotherapy; however, at relapse, the tumor is highly resistant to subsequent available therapies. Here, we report that the expression of three prime repair exonuclease 1 (TREX1) is strongly induced in chemoresistant SCLCs. Assay for transposase-accessible chromatin using sequencing and chromatin immunoprecipitation sequencing revealed a significant increase in chromatin accessibility and transcriptional activity of TREX1 gene locus in chemoresistant SCLCs. Analyses of human SCLC tumors and patient-derived xenografts (PDX) also showed an increase in TREX1 expression in postchemotherapy samples. TREX1 depletion caused the activation of cyclic GMP-AMP synthase stimulator of interferon gene pathway due to cytoplasmic accumulation of damage-associated double-stranded DNA, inducing immunogenicity and enhancing the sensitivity of drug-resistant cells to chemotherapy. These findings suggest TREX1 upregulation may partially contribute to the survival of resistant cells, and its inhibition may represent a promising therapeutic strategy to enhance antitumor immunity and potentiate the efficacy of chemotherapy and/or immunotherapy in chemoresistant SCLCs. Significance: In this study, we show that targeting TREX1 induces an innate immune response and resensitizes SCLC cells to chemotherapy, representing a promising novel target for "immunologically" cold tumors, such as SCLC.
Collapse
Affiliation(s)
- Takahiko Murayama
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
- Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
| | - Navin R. Mahadevan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Department of Pathology, Brigham and Women’s Hospital, Boston, Massachusetts.
| | - Catherine B. Meador
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts.
| | - Elena V. Ivanova
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Yuqiao Pan
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
- Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
| | - Erik H. Knelson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Tetsuo Tani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Jun Nakayama
- Department of Oncogenesis and Growth Regulation, Osaka International Cancer Institute, Osaka, Japan.
- Laboratory of Integrative Oncology, National Cancer Center Research Institute, Tokyo, Japan.
| | - Xueying Ma
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
- Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
| | - Tran C. Thai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Yin P. Hung
- Department of Pathology, Massachusetts General Hospital Cancer Center, Boston, Massachusetts.
| | - William Kim
- Moores Cancer Center, UC San Diego, La Jolla, California.
- Center for Novel Therapeutics, UC San Diego, La Jolla, California.
- Department of Medicine, UC San Diego, La Jolla, California.
| | - Hideo Watanabe
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York.
| | - Kathy Q. Cai
- Histopathology Facility, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
| | - Aaron N. Hata
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital Cancer Center, Boston, Massachusetts.
| | - Cloud P. Paweletz
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - David A. Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Israel Cañadas
- Nuclear Dynamics and Cancer Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
- Cancer Epigenetics Institute, Fox Chase Cancer Center, Philadelphia, Pennsylvania.
| |
Collapse
|
44
|
Malhotra J, Chiappori A, Fujioka N, Hanna NH, Feldman LE, Patel M, Moore D, Chen C, Jabbour SK. Phase I/II trial of plinabulin in combination with nivolumab and ipilimumab in patients with recurrent small cell lung cancer (SCLC): Big ten cancer research consortium (BTCRC-LUN17-127) study. Lung Cancer 2024; 195:107932. [PMID: 39173229 DOI: 10.1016/j.lungcan.2024.107932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/06/2024] [Accepted: 08/15/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Plinabulin is a GEF-H1 releasing agent with an immune-enhancing function. We report results from a multicenter Phase I/II study (NCT03575793) assessing plinabulin in combination with nivolumab and ipilimumab for the treatment of recurrent SCLC. METHODS In Phase I, patients were enrolled using a 3 + 3 design to determine dose-limiting toxicities (DLTs) and recommended Phase 2 dose (RP2D). Patients received nivolumab (1 mg/kg), ipilimumab (3 mg/kg), and plinabulin (in escalating doses) on day 1 of each 21-day cycle for 4 cycles followed by maintenance with plinabulin and nivolumab. In phase II, patients with recurrent PD(L)1 inhibitor resistant SCLC were enrolled. The primary objective was median progression-free survival (PFS). RESULTS Between 9/2018 and 2/2023, 39 patients were enrolled, and 36 patients received study treatment and were evaluable for safety (16 in Phase I; 20 in Phase II). In the phase I dose-escalation, there were 2 DLTs; grade 3 altered mental status lasting <24 h and grade 3 infusion reaction. The Plinabulin RP2D was determined to be 30 mg/m2. Common TRAEs were vomiting (44 %), nausea (42 %), and infusion reaction (36 %); 6 % of patients had a ≥grade 3 TRAE. Five patients (14 %) had ≥grade 3 irAEs; there were no cases of immune-related pneumonitis. In the efficacy analysis in 27 patients, the median PFS was 1.6 months (95 % CI 1.2 to 2.7) and the trial did not meet the pre-specified target median PFS of 3.5 months. Four patients treated at 30 mg/m2 had PR (confirmed 1, unconfirmed 3); 5 patients had SD with a CBR of 33 %. Two of 8 patients treated in phase I at the lower 20 mg/m2 dose had confirmed PR, with 1 patient on the drug regimen for >90 cycles. The median OS and follow-up time were 5.5 months and 2.5 months respectively. CONCLUSIONS Plinabulin in combination with nivolumab and ipilimumab was tolerable at the dose of 30 mg/m2. While the clinical responses in PD-1 resistant SCLC were limited, some patients had a long duration of response. The number of ≥grade 3 irAE with the combination were lower than expected.
Collapse
Affiliation(s)
- Jyoti Malhotra
- City of Hope National Medical Center, Duarte, CA, United States; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States.
| | - Alberto Chiappori
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, United States
| | - Naomi Fujioka
- University of Minnesota, Minneapolis, MN, United States
| | - Nasser H Hanna
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, United States
| | - Lawrence E Feldman
- University of Illinois Hospital & Health Sciences System, Chicago, IL, United States
| | - Malini Patel
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Dirk Moore
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Chunxia Chen
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Salma K Jabbour
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
45
|
Andrini E, Ricco G, Zappi A, Aloi S, Giordano M, Altimari A, Gruppioni E, Maloberti T, de Biase D, Campana D, Lamberti G. Challenges and future perspectives for the use of temozolomide in the treatment of SCLC. Cancer Treat Rev 2024; 129:102798. [PMID: 38970838 DOI: 10.1016/j.ctrv.2024.102798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/09/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
Small-cell lung cancer (SCLC), accounting for 10-20 % of all lung tumors, represents the most aggressive high-grade neuroendocrine carcinoma. Most patients are diagnosed with extensive-stage SCLC (ES-SCLC), with brian metastases identified in ∼ 80 % of cases during the disease cours, and the prognosis is dismal, with a 5-year survival rate of less than 5 %. Current available treatments in the second-line setting are limited, and topotecan has long been the only FDA-approved drug in relapsed or refractory ES-SCLC, until the recent approval of lurbinectedin, a selective inhibitor of RNA polymerase II. Temozolomide (TMZ) is an oral alkylating agent, which showed single-agent activity in SCLC, particularly among patients with O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation. Several studies have revealed the synergistic activity of temozolomide with poly-ADP-ribose polymerase (PARP) inhibitors, that prevent repair of TMZ-induced DNA damage. This review focuses on the rationale for the use of TMZ in ES-SCLC and provides an overview of the main trials that have evaluated and are currently investigating its role, both as a single-agent and in combinations, in relapse or refractory disease.
Collapse
Affiliation(s)
- Elisa Andrini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy.
| | - Gianluca Ricco
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy.
| | - Arianna Zappi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy.
| | - Serena Aloi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy.
| | - Mirela Giordano
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy.
| | - Annalisa Altimari
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Elisa Gruppioni
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Thais Maloberti
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Dario de Biase
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Pharmacy and Biotechnology (FaBit), University of Bologna, Bologna, Italy.
| | - Davide Campana
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy.
| | - Giuseppe Lamberti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy.
| |
Collapse
|
46
|
Chen J, Liu S, Ruan Z, Wang K, Xi X, Mao J. Thrombotic events associated with immune checkpoint inhibitors and novel antithrombotic strategies to mitigate bleeding risk. Blood Rev 2024; 67:101220. [PMID: 38876840 DOI: 10.1016/j.blre.2024.101220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/23/2024] [Accepted: 06/05/2024] [Indexed: 06/16/2024]
Abstract
Although immunotherapy is expanding treatment options for cancer patients, the prognosis of advanced cancer remains poor, and these patients must contend with both cancers and cancer-related thrombotic events. In particular, immune checkpoint inhibitors are associated with an increased risk of atherosclerotic thrombotic events. Given the fundamental role of platelets in atherothrombosis, co-administration of antiplatelet agents is always indicated. Platelets are also involved in all steps of cancer progression. Classical antithrombotic drugs can cause inevitable hemorrhagic side effects due to blocking integrin β3 bidirectional signaling, which regulates simultaneously thrombosis and hemostasis. Meanwhile, many promising new targets are emerging with minimal bleeding risk and desirable anti-tumor effects. This review will focus on the issue of thrombosis during immune checkpoint inhibitor treatment and the role of platelet activation in cancer progression as well as explore the mechanisms by which novel antiplatelet therapies may exert both antithrombotic and antitumor effects without excessive bleeding risk.
Collapse
Affiliation(s)
- Jiayi Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shuang Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Zheng Ruan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Kankan Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Sino-French Research Center for Life Sciences and Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Xiaodong Xi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Jianhua Mao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| |
Collapse
|
47
|
Myrda J, Bremm F, Schaft N, Dörrie J. The Role of the Large T Antigen in the Molecular Pathogenesis of Merkel Cell Carcinoma. Genes (Basel) 2024; 15:1127. [PMID: 39336718 PMCID: PMC11431464 DOI: 10.3390/genes15091127] [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: 07/29/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
The large T antigen (LT) of the Merkel cell polyomavirus (MCPyV) is crucial for Merkel cell carcinoma (MCC), a rare but very aggressive form of neuroendocrine skin cancer. The clonal integration of MCPyV DNA into the host genome is a signature event of this malignancy. The resulting expression of oncogenes, including the small T (sT) antigen and a truncated form of the LT (truncLT), directly contribute to carcinogenesis. The truncation of the C-terminus of LT prevents the virus from replicating due to the loss of the origin binding domain (OBD) and the helicase domain. This precludes cytopathic effects that would lead to DNA damage and ultimately cell death. At the same time, the LxCxE motif in the N-terminus is retained, allowing truncLT to bind the retinoblastoma protein (pRb), a cellular tumor suppressor. The continuously inactivated pRb promotes cell proliferation and tumor development. truncLT exerts several classical functions of an oncogene: altering the host cell cycle, suppressing innate immune responses to viral DNA, causing immune escape, and shifting metabolism in favor of cancer cells. Given its central role in MCC, the LT is a major target for therapeutic interventions with novel approaches, such as immune checkpoint inhibition, T cell-based immunotherapy, and cancer vaccines.
Collapse
Affiliation(s)
- Julia Myrda
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Franziska Bremm
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Niels Schaft
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| | - Jan Dörrie
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), 91054 Erlangen, Germany
- Bavarian Cancer Research Center (BZKF), 91054 Erlangen, Germany
| |
Collapse
|
48
|
Thompson JC, Tilsed C, Davis C, Gupta A, Melidosian B, Sun C, Kallen ME, Timmers C, Langer CJ, Albelda SM. Predictive Signatures for Responses to Checkpoint Blockade in Small-Cell Lung Cancer in Second-Line Therapy Do Not Predict Responses in First-Line Patients. Cancers (Basel) 2024; 16:2795. [PMID: 39199568 PMCID: PMC11353197 DOI: 10.3390/cancers16162795] [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: 07/05/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
Although immune checkpoint blockade (ICB) is currently approved for the treatment of extensive-stage small-cell lung cancer (SCLC) in combination with chemotherapy, relatively few patients have demonstrated durable clinical benefit (DCB) to these therapies. Biomarkers predicting responses are needed. Biopsies from 35 SCLC patients treated with ICB were subjected to transcriptomic analysis; gene signatures were assessed for associations with responses. Twenty-one patients were treated with ICB in the first-line setting in combination with platinum-based chemotherapy; fourteen patients were treated in the second-line setting with ICB alone. DCB after ICB in SCLC in the second-line setting (3 of 14 patients) was associated with statistically higher transcriptomic levels of genes associated with inflammation (p = 0.003), antigen presentation machinery (p = 0.03), interferon responses (p < 0.05), and increased CD8 T cells (p = 0.02). In contrast, these gene signatures were not significantly different in the first-line setting. Our data suggest that responses to ICB in SCLC in the second-line setting can be predicted by the baseline inflammatory state of the tumor; however, this strong association with inflammation was not seen in the first-line setting. We postulate that chemotherapy alters the immune milieu allowing a response to ICB. Other biomarkers will be needed to predict responses in first-line therapy patients.
Collapse
Affiliation(s)
- Jeffrey C. Thompson
- Division of Pulmonary, Allergy and Critical Care Medicine, Thoracic Oncology Group, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 228 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA 19104, USA; (J.C.T.); (C.T.); (A.G.)
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (C.D.); (C.J.L.)
| | - Caitlin Tilsed
- Division of Pulmonary, Allergy and Critical Care Medicine, Thoracic Oncology Group, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 228 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA 19104, USA; (J.C.T.); (C.T.); (A.G.)
| | - Christiana Davis
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (C.D.); (C.J.L.)
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Aasha Gupta
- Division of Pulmonary, Allergy and Critical Care Medicine, Thoracic Oncology Group, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 228 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA 19104, USA; (J.C.T.); (C.T.); (A.G.)
| | | | - Chifei Sun
- Incyte, Wilmington, DE 19803, USA; (B.M.); (C.S.); (C.T.)
| | - Michael E. Kallen
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | | | - Corey J. Langer
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (C.D.); (C.J.L.)
- Division of Hematology/Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Steven M. Albelda
- Division of Pulmonary, Allergy and Critical Care Medicine, Thoracic Oncology Group, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, 228 Stemmler Hall, 3450 Hamilton Walk, Philadelphia, PA 19104, USA; (J.C.T.); (C.T.); (A.G.)
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (C.D.); (C.J.L.)
| |
Collapse
|
49
|
Zhang J, Zeng X, Guo Q, Sheng Z, Chen Y, Wan S, Zhang L, Zhang P. Small cell lung cancer: emerging subtypes, signaling pathways, and therapeutic vulnerabilities. Exp Hematol Oncol 2024; 13:78. [PMID: 39103941 DOI: 10.1186/s40164-024-00548-w] [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: 06/26/2024] [Accepted: 07/27/2024] [Indexed: 08/07/2024] Open
Abstract
Small cell lung cancer (SCLC) is a recalcitrant cancer characterized by early metastasis, rapid tumor growth and poor prognosis. In recent decades, the epidemiology, initiation and mutation characteristics of SCLC, as well as abnormal signaling pathways contributing to its progression, have been widely studied. Despite extensive investigation, fewer drugs have been approved for SCLC. Recent advancements in multi-omics studies have revealed diverse classifications of SCLC that are featured by distinct characteristics and therapeutic vulnerabilities. With the accumulation of SCLC samples, different subtypes of SCLC and specific treatments for these subtypes were further explored. The identification of different molecular subtypes has opened up novel avenues for the treatment of SCLC; however, the inconsistent and uncertain classification of SCLC has hindered the translation from basic research to clinical applications. Therefore, a comprehensives review is essential to conclude these emerging subtypes and related drugs targeting specific therapeutic vulnerabilities within abnormal signaling pathways. In this current review, we summarized the epidemiology, risk factors, mutation characteristics of and classification, related molecular pathways and treatments for SCLC. We hope that this review will facilitate the translation of molecular subtyping of SCLC from theory to clinical application.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| | - Xiaoping Zeng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Qiji Guo
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Zhenxin Sheng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Yan Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Shiyue Wan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Lele Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| |
Collapse
|
50
|
He Y, Kong L, Ji X, Zhuo M, An T, Jia B, Chi Y, Wang J, Zhao J, Li J, Yang X, Chen H, Zhai X, Tai Y, Ding L, Wang Z, Wang Y. Women patients with small-cell lung cancer using immunotherapy in a real-world cohort achieved long-term survival. Thorac Cancer 2024; 15:1727-1738. [PMID: 38923348 PMCID: PMC11320081 DOI: 10.1111/1759-7714.15393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Usage of immune checkpoint inhibitors (ICIs) has prolonged the overall survival (OS) of patients with extensive-stage small-cell lung cancer (ES-SCLC). In clinical trials, males accounted for a large proportion, leading to the uncertainty of its efficacy in female patients. We therefore conducted this study to explore the efficacy and safety of using ICIs in female patients with ES-SCLC. METHODS We retrospectively enrolled female SCLC patients and subdivided them into two groups. Group A (n = 40) was defined as ES-SCLC patients who received first-line standard chemotherapy with or without ICIs. Group B (n = 47) included relapsed SCLC patients who were administered with second-line therapies. Kaplan-Meier methodology was used to calculate survival analysis. Chi-squared tests were used to analyze the incidence of adverse events (AEs). RESULTS Median progression-free survival (PFS) and median OS favored the ICI-contained cohorts (Group A PFS: 8.3 vs. 6.1 months; OS: not reached vs. 11.3 months; Group B PFS: 15.1 vs. 3.3 months; OS: 35.3 vs. 8.3 months), especially in those patients who received second-line immunotherapies. Patients who received immunotherapy had a slightly higher incidence rate of grade ≥3 AEs (Group A: 71.4% vs. 46.2%; Group B: 44.5% vs. 13.2%). Those who developed grade ≥3 AEs in first-line ICIs cohort had a more favorable survival (PFS: 8.3 vs. 3.2 months; OS: not reached vs. 5.1 months). CONCLUSIONS Our study suggested that female ES-SCLC patients treated with immunotherapy tended to achieve a relatively longer survival. The incidence of AEs (grade ≥3) was higher in women patients receiving ICIs, which requires monitoring more closely.
Collapse
Affiliation(s)
- Yuling He
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Lingdong Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Xumeng Ji
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Minglei Zhuo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Tongtong An
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Bo Jia
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Yujia Chi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Jingjing Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Jun Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Jianjie Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Xue Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Hanxiao Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Xiaoyu Zhai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Yidi Tai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Lu Ding
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Ziping Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| | - Yuyan Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital & InstituteBeijingChina
| |
Collapse
|