1
|
Song Q, Yu Z, Lu W, Zhuo Z, Chang L, Mei H, Cui Y, Zhang D. PD-1/PD-L1 inhibitors related adverse events: A bibliometric analysis from 2014 to 2024. Hum Vaccin Immunother 2025; 21:2424611. [PMID: 39757956 DOI: 10.1080/21645515.2024.2424611] [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: 07/31/2024] [Revised: 10/14/2024] [Accepted: 10/29/2024] [Indexed: 01/07/2025] Open
Abstract
Programmed cell death-1 (PD-1) inhibitors and programmed cell death ligand 1 (PD-L1) inhibitors are considered effective alternatives for the primary treatment of recurrent metastatic cancers. However, they can induce various adverse events affecting multiple organ systems, potentially diminishing patients' quality of life, and even leading to treatment interruptions. Adverse events related to PD-1/PD-L1 inhibitors differ from those associated with CTLA-4 inhibitors and are more commonly observed in the treatment of solid tumors. This study aimed to address the knowledge gap regarding adverse events related to PD-1/PD-L1 inhibitors. A visual bibliometric network was constructed using VOSviewer, CiteSpace, R software, and the Web of Science Core Collection (WoSCC) to quantitatively analyze this research field. Future research directions were also explored. The USA ranked first in publication count and total citations. Over time, publication types transitioned from case reports to clinical trials. Research on for nivolumab was the most prevalent. The spectrum of cancers treated by PD-1/PD-L1 inhibitors expanded beyond melanoma and lung cancer to include renal cell carcinoma, esophageal cancer, and others. Common adverse events included pneumonitis, myasthenia gravis, and vitiligo. There was a significant increase in multi-phase clinical trials and studies related to biomarkers. This study offers valuable insights for potential collaborators and institutions, highlighting trends in the study of adverse events related to PD-1/PD-L1 inhibitors. The management of these adverse events has become more refined and standardized. Biomarker research and multi-phase clinical trials are likely to be key areas of focus in future studies.
Collapse
Affiliation(s)
- Qingya Song
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zongliang Yu
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Wenping Lu
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhili Zhuo
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lei Chang
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Heting Mei
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Yongjia Cui
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Dongni Zhang
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
2
|
Le J, Sun Y, Deng G, Dian Y, Xie Y, Zeng F. Immune checkpoint inhibitors in cancer patients with autoimmune disease: Safety and efficacy. Hum Vaccin Immunother 2025; 21:2458948. [PMID: 39894761 PMCID: PMC11792813 DOI: 10.1080/21645515.2025.2458948] [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: 11/10/2024] [Revised: 01/09/2025] [Accepted: 01/23/2025] [Indexed: 02/04/2025] Open
Abstract
The utilization of immune-checkpoint inhibitors (ICIs) in cancer immunotherapy frequently leads to the occurrence of immune-related adverse events (irAEs), making it generally not recommended for patients with preexisting autoimmune diseases. Hence, we conducted a meta-analysis on safety and efficacy of ICIs in cancer patients with preexisting autoimmune diseases to provide further insights. PubMed, EMBASE, and Cochrane Library were systematically searched until December 20, 2024. The main summary measures used were pooled rate and risk ratio (RR) with 95% confidential interval (CI), which were analyzed using R statistic software. A total of 52 articles were included in the study. When cancer patients with preexisting autoimmune diseases received ICIs treatment, the overall incidence was 0.610 (95% CI: 0.531-0.686) for any grade irAEs, 0.295 (95% CI: 0.248-0.343) for flares, 0.325 (95% CI: 0.258-0.396) for de novo irAEs, 0.238 (95% CI: 0.174-0.309) for grade ≥3 irAEs, and 0.143 (95% CI: 0.109-0.180) for discontinuation due to immunotoxicity. Compared with those without autoimmune diseases, cancer patients with autoimmune diseases experienced a higher risk of any-grade irAEs (RR: 1.23, 95% CI: 1.12-1.35) and discontinuation due to immunotoxicity (1.40, 95% CI: 1.11-1.78). However, no statistically significant differences were observed in the incidence of grade ≥3 irAEs, objective response rate (ORR), disease control rate (DCR), overall survival (OS), and progression-free survival (PFS) between the two groups. During ICIs treatment, irAEs are common among cancer patients with autoimmune diseases, but severe irAEs is relatively low. ICIs are effective in this population, but should be strictly monitored when used to avoid immunotoxicity.
Collapse
Affiliation(s)
- Jiayuan Le
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- Furong Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuming Sun
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- Furong Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- Furong Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China
- Furong Laboratory, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yanli Xie
- Department of Rheumatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
3
|
Paz-Ares L, Borghaei H, Liu SV, Peters S, Herbst RS, Stencel K, Majem M, Şendur MAN, Czyżewicz G, Caro RB, Lee KH, Johnson ML, Karadurmuş N, Grohé C, Baka S, Csőszi T, Ahn JS, Califano R, Yang TY, Kemal Y, Ballinger M, Cuchelkar V, Graupner V, Lin YC, Chakrabarti D, Bhatt K, Cai G, Iannone R, Reck M, IMforte investigators. Efficacy and safety of first-line maintenance therapy with lurbinectedin plus atezolizumab in extensive-stage small-cell lung cancer (IMforte): a randomised, multicentre, open-label, phase 3 trial. Lancet 2025; 405:2129-2143. [PMID: 40473449 DOI: 10.1016/s0140-6736(25)01011-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 05/09/2025] [Accepted: 05/11/2025] [Indexed: 06/16/2025]
Abstract
BACKGROUND Despite improved efficacy with first-line immune checkpoint inhibitors plus platinum-based chemotherapy for extensive-stage small-cell lung cancer (ES-SCLC), survival remains poor. In this study, we aimed to compare lurbinectedin plus atezolizumab and atezolizumab alone as maintenance therapies in patients with ES-SCLC without progression after induction therapy with atezolizumab, carboplatin, and etoposide. METHODS IMforte was a randomised, open-label, phase 3 trial done at 96 hospitals and medical centres in 13 countries (Belgium, Germany, Greece, Hungary, Italy, Mexico, Poland, South Korea, Spain, Taiwan, Türkiye, the UK, and the USA). Eligible patients were aged 18 years or older with treatment-naive ES-SCLC. Patients received four 21-day cycles of induction treatment (atezolizumab, carboplatin, and etoposide). After completing induction treatment, eligible patients without disease progression were randomly assigned (1:1) using permuted blocks (Interactive Voice/Web Response System) to receive maintenance treatment intravenously every 3 weeks with lurbinectedin (3·2 mg/m2; with granulocyte colony-stimulating factor prophylaxis) plus atezolizumab (1200 mg) or atezolizumab (1200 mg). The two primary endpoints were independent review facility-assessed (IRF) progression-free survival and overall survival, measured from randomisation into the maintenance phase. Efficacy endpoints were assessed in the full analysis set, which included all patients who were randomly assigned to maintenance phase treatment, regardless of whether they received their assigned study treatment. Safety was assessed in all patients who received at least one dose of lurbinectedin or atezolizumab, and was analysed according to the treatment received. This study is registered with ClinicalTrials.gov, NCT05091567, and is closed for recruitment. FINDINGS Between Nov 17, 2021, and Jan 11, 2024, 895 patients were screened for enrolment, of whom 660 (74%) were enrolled into the induction phase. Between May 24, 2022, and April 30, 2024, 483 (73%) of 660 patients entered the maintenance phase and were randomly assigned to lurbinectedin plus atezolizumab (n=242) or atezolizumab (n=241). At the data cutoff (July 29, 2024), IRF progression-free survival was longer in the lurbinectedin plus atezolizumab group than the atezolizumab group (stratified hazard ratio [HR] 0·54 [95% CI 0·43-0·67]; p<0·0001), as was overall survival (stratified HR 0·73 [0·57-0·95]; p=0·017). 92 (38%) of 242 patients in the lurbinectedin plus atezolizumab group and 53 (22%) of 240 patients in the atezolizumab group had grade 3-4 adverse events. The most common grade 3-4 events in the lurbinectedin plus atezolizumab group were anaemia (20 [8%] of 242 patients), decreased neutrophil count (18 [7%] patients), and decreased platelet count (18 [7%] patients) and the most common events in the atezolizumab group were hyponatremia (five [2%] of 240 patients), dyspnoea (four [2%] patients), and pneumonia (four [2%] patients). Grade 5 adverse events occurred in 12 (5%) of 242 patients in the lurbinectedin plus atezolizumab group and six (3%) of 240 patients in the atezolizumab group. The incidence of myelosuppressive toxicities (eg, neutropenia and leukopenia) was higher in the lurbinectedin plus atezolizumab group than the atezolizumab group. INTERPRETATION IRF progression-free survival and overall survival were longer in the lurbinectedin plus atezolizumab group than the atezolizumab group for patients with ES-SCLC, albeit with a higher incidence of adverse events. Lurbinectedin plus atezolizumab represents a novel therapeutic option for first-line maintenance treatment in this setting. FUNDING F Hoffmann-La Roche and Jazz Pharmaceuticals.
Collapse
Affiliation(s)
- Luis Paz-Ares
- Department of Medical Oncology, Hospital Universitario 12 de Octubre, H12O-CNIO Lung Cancer Unit, Universidad Complutense and Ciberonc, Madrid, Spain.
| | - Hossein Borghaei
- Department of Hematology and Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Stephen V Liu
- Department of Medical Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC, USA
| | - Solange Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Roy S Herbst
- Department of Medical Oncology and Hematology, Yale School of Medicine, New Haven, CT, USA
| | - Katarzyna Stencel
- Department of Clinical Oncology, Wielkopolska Center of Pulmonology and Thoracic Surgery of Eugenia and Janusz Zeyland, Poznan, Poland
| | - Margarita Majem
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Mehmet Ali Nahit Şendur
- Department of Medical Oncology, Ankara Yıldırım Beyazıt University and Ankara Bilkent City Hospital, Ankara, Türkiye
| | - Grzegorz Czyżewicz
- Department of Clinical Oncology, John Paul II Specialist Hospital, Kraków, Poland
| | - Reyes Bernabé Caro
- Department of Medical Oncology, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Ki Hyeong Lee
- Department of Medicine, Chungbuk National University Hospital, Cheongju, South Korea
| | - Melissa L Johnson
- Department of Medical Oncology, Sarah Cannon Research Institute, Tennessee Oncology, Nashville, TN, USA
| | - Nuri Karadurmuş
- Department of Medical Oncology, University of Health Sciences, Gülhane Training and Research Hospital, Ankara, Türkiye
| | - Christian Grohé
- Department of Pulmonology and Pulmonary Oncology, Klinik für Pneumologie, Evangelische Lungenklinik Berlin, Berlin, Germany
| | - Sofia Baka
- Department of Oncology, Interbalkan European Medical Center, Thessaloniki, Greece
| | - Tibor Csőszi
- Department of Oncology, Hetenyi Geza Korhaz Onkologiai Kozpont, Szolnok, Hungary
| | - Jin Seok Ahn
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Raffaele Califano
- Department of Medical Oncology, The Christie NHS Foundation Trust and Division of Cancer Sciences, University of Manchester, Manchester, UK
| | - Tsung-Ying Yang
- Department of Chest Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Yasemin Kemal
- Department of Medical Oncology, Altınbaş University, İstanbul, Türkiye
| | | | | | | | | | | | | | | | | | - Martin Reck
- Department of Thoracic Oncology, Lung Clinic Grosshansdorf, Airway Research Center North, German Center of Lung Research, Grosshansdorf, Germany
| | | |
Collapse
|
4
|
Choudhury NJ, Garassino MC. Lurbinectedin with atezolizumab maintenance therapy in extensive-stage small-cell lung cancer. Lancet 2025; 405:2104-2106. [PMID: 40516996 DOI: 10.1016/s0140-6736(25)01145-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2025] [Accepted: 05/28/2025] [Indexed: 06/16/2025]
Affiliation(s)
- Noura J Choudhury
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | | |
Collapse
|
5
|
Sahin TK, Guven DC. Letter Re: Correlation between irAEs and survival outcomes in patients with ES-SCLC treated with first-line chemoimmunotherapy. Eur J Cancer 2025:115562. [PMID: 40514272 DOI: 10.1016/j.ejca.2025.115562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2025] [Accepted: 05/28/2025] [Indexed: 06/16/2025]
|
6
|
Chen TF, Li ZJ, Zhao HS, Yang R. Feasibility and Safety of Anlotinib Combined with Immune Checkpoint Inhibitors in Patients with Previously Immunotherapy-Treated Extensive-Stage Small Cell Lung Cancer: A Retrospective Exploratory Study. Drug Des Devel Ther 2025; 19:4991-5005. [PMID: 40521010 PMCID: PMC12164881 DOI: 10.2147/dddt.s525481] [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: 02/28/2025] [Accepted: 05/22/2025] [Indexed: 06/18/2025] Open
Abstract
Objective This study aims to evaluate the efficacy and safety of anlotinib combined with immune checkpoint inhibitors (ICIs) in patients with previously immunotherapy-treated extensive-stage small cell lung cancer (ES-SCLC). Methods This retrospective study screened ES-SCLC patients who experienced failure after prior ICIs-based treatment and received anlotinib combined with ICIs therapy clinically. Anlotinib was administered at a standard dosage, ICIs regimens included PD-1 inhibitors (Tislelizumab, 200 mg; serplulimab, 4.5 mg/kg) and PD-L1 inhibitors. Efficacy and safety data during treatment were retrospectively collected, and patients were followed up regularly to obtain long-term survival data. Subgroup analysis was conducted to identify the differences in treatment outcomes in various baseline characteristics. Results Of the 68 ES-SCLC patients included, no complete response, 22 patients achieved partial response, 28 had stable disease, 14 experienced disease progression and 4 patients were not available for efficacy. Objective response rate (ORR) was 32.4% (95% CI: 21.5-44.8%), and disease control rate (DCR) was 73.5% (95% CI: 61.4-83.5%). The median progression-free survival (PFS) was 5.6 months (95% CI: 3.87-7.33), the median duration of response (DoR) was 6.8 months (95% CI: 0.80-12.83). At the data cutoff date, the median follow-up duration was 12.5 months (range: 1.1-31.5 months), yielding a median overall survival (OS) of 13.2 months (95% CI: 7.09-19.31). Subgroup analysis highlighted that patients who were previously intolerant to immunotherapy had a longer OS (median OS: 15.5 vs 10.9 months, P = 0.031). Safety analysis showed that 61 patients (89.7%) experienced treatment-related adverse events (TRAEs) of varying severity with 37 patients (54.4%) developing grade 3 or higher. The most common TRAEs included fatigue, nausea and vomiting, hypertension, hematologic toxicity, and liver function abnormalities. Conclusion Anlotinib combined with ICIs demonstrated that immunotherapy rechallenge in previously ICIs-treated ES-SCLC was feasible and of clinical significance preliminarily. However, larger-scale clinical studies were required to validate these findings subsequently.
Collapse
Affiliation(s)
- Teng-Fei Chen
- Department of Respiratory Medicine Ward 2, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Zhan-Jiang Li
- Department of Respiratory Medicine Ward 1, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Hua-Si Zhao
- Department of Respiratory Medicine Ward 2, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Rui Yang
- Department of Respiratory Medicine Ward 1, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| |
Collapse
|
7
|
Wang Y, Su X, Jia J, Zhou T, Lu Y, Zhao L, Yang Z, Fu X, Zeng Y, Cai X. Early radiotherapy improved survival of patients with extensive-stage small cell lung cancer treated with first-line chemo-immunotherapy. BMC Cancer 2025; 25:1012. [PMID: 40481435 PMCID: PMC12142918 DOI: 10.1186/s12885-025-14417-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 05/30/2025] [Indexed: 06/11/2025] Open
Abstract
BACKGROUND AND PURPOSE This real-world study aimed to investigate the efficacy of early radiotherapy (RT) in ES-SCLC patients treated with first-line chemo-immunotherapy. MATERIALS AND METHODS ES-SCLC patients were enrolled from August 2018 to October 2023. Patients who received early radiotherapy before disease progression were defined as Early RT group, while the others, named Salvage and Non-RT (S&N RT) group. Propensity score matching (PSM) with a 1:1 ratio was performed to balance the baseline characteristics. RESULTS In this study, 375 patients with ES-SCLC treated with first-line chemo-immunotherapy were enrolled. The median PFS was 11.4 months of the Early RT group compared to 6.1 months of the S&N RT group (HR = 0.59, 95%CI 0.45-0.77; p < 0.001). The median OS was 23.8 months of the Early RT group versus 18.0 months of the S&N RT group (HR = 0.50, 95%CI 0.34-0.73; p = 0.004). The survival benefit persisted in the PSM cohort. Furthermore, survival was significantly improved in Early RT group compared to Salvage RT group (p = 0.028), while Salvage RT group had a similar survival with Non-RT group (p = 0.868). The risk of adverse events was tolerable. The multivariate analysis also demonstrated that early radiotherapy was an independently positive predictor for PFS and OS. CONCLUSIONS Administering early radiotherapy significantly improved both PFS and OS in patients with ES-SCLC treated with first-line chemo-immunotherapy with tolerable adverse events, while salvage radiotherapy did not improve survival. This finding warrants further validation through prospective randomized studies.
Collapse
Affiliation(s)
- Yunfeng Wang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Xi Su
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Jingyi Jia
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Tongfang Zhou
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Yifei Lu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Lei Zhao
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Zhangru Yang
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Xiaolong Fu
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China
| | - Ya Zeng
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| | - Xuwei Cai
- Department of Radiation Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200030, China.
| |
Collapse
|
8
|
Li K, Liu C, Sui X, Li C, Zhang T, Zhao T, Zhang D, Wu H, Liu Y, Wang S, Yang Y, Lin B, Wang W, Yang F, Chen X, Liu P. An organoid co-culture model for probing systemic anti-tumor immunity in lung cancer. Cell Stem Cell 2025:S1934-5909(25)00191-2. [PMID: 40513558 DOI: 10.1016/j.stem.2025.05.011] [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: 07/10/2024] [Revised: 04/21/2025] [Accepted: 05/19/2025] [Indexed: 06/16/2025]
Abstract
Deciphering interactions between tumor micro- and systemic immune macroenvironments is essential for developing more effective cancer diagnosis and therapeutic strategies. Here, we established a gel-liquid interface (GLI) co-culture model of lung cancer organoids (LCOs) and paired peripheral-blood mononuclear cells (PBMCs), featuring enhanced interactions between immune cells and tumor organoids for optimized simulation of in vivo systemic anti-tumor immunity. By constructing a cohort of lung cancer patients, we demonstrated that the responses of GLI models under αPD1 treatment reflected the immunotherapy outcomes of the corresponding patients precisely. Furthermore, we dissected the various tumor immune processes mediated by PBMC-derived T cells within GLI models through functional multi-omics analyses, along with the characterization of circulating tumor-reactive T cells (GNLY+CD44+CD9+) with effector memory-like phenotypes as a potential indicator of immunotherapy efficacy. Our findings indicate that the GLI co-culture model can be used to develop diagnostic strategies for precision immunotherapies, as well as understanding the underlying mechanisms.
Collapse
Affiliation(s)
- Kaiyi Li
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Chang Liu
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Xizhao Sui
- Department of Thoracic Surgery, People's Hospital, Peking University, Beijing 100044, China
| | - Chao Li
- Department of Thoracic Surgery, People's Hospital, Peking University, Beijing 100044, China
| | - Ting Zhang
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Tian Zhao
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Dong Zhang
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Hainan Wu
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China
| | - Yuhan Liu
- Tanwei College, Tsinghua University, Beijing 100084, China
| | - Shuai Wang
- Department of Thoracic Surgery, Beijing Haidian Hospital (HaidianSection of Peking University Third Hospital), Beijing 100080, China
| | - Yingshun Yang
- Department of Thoracic Surgery, Beijing Haidian Hospital (HaidianSection of Peking University Third Hospital), Beijing 100080, China
| | - Baobao Lin
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China
| | - Wenyan Wang
- State Key Laboratory of Molecular Oncology, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Fan Yang
- Department of Thoracic Surgery, People's Hospital, Peking University, Beijing 100044, China.
| | - Xiaofang Chen
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China.
| | - Peng Liu
- School of Biomedical Engineering, Tsinghua University, Beijing 100084, China; Changping Laboratory, Beijing 102206, China.
| |
Collapse
|
9
|
Chu G, Zhu X, Wu J, Tang Y, Luu J, He C, Huang SP, Liu L, Hsieh HJ. Assessing Correlation between Surrogate Endpoints and Overall Survival for Oncology Clinical Trials. Clin Pharmacol Ther 2025; 117:1706-1717. [PMID: 40013399 DOI: 10.1002/cpt.3613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/28/2025] [Indexed: 02/28/2025]
Abstract
Surrogate endpoints, such as progression-free survival (PFS) and objective response rate (ORR), are increasingly used in oncology trials to expedite drug development and decision making. This paper evaluates the effectiveness of these surrogate endpoints by analyzing their correlations with overall survival (OS) across different cancer types, treatments, and therapy lines at both the patient and trial levels using an integrated dataset from Bristol Myers Squibb. At the patient level, correlation between OS and PFS was consistently stronger than those between OS and best overall response (BOR), suggesting that PFS may serve as a more reliable surrogate for OS. Melanoma patients exhibited the highest correlation between OS and BOR, and immune-oncology (IO) therapy patients showed stronger correlations than those treated with chemotherapy. First-line therapy patients demonstrated stronger correlations between BOR, PFS, and OS compared with second-line or third-line patients. At the trial level, the correlation between PFS hazard ratio (HR) and difference in ORR (∆ORR) was stronger than that between other endpoints. Melanoma studies exhibited strong correlations with significant P-values. IO therapy studies exhibited more consistent correlations.
Collapse
Affiliation(s)
- Guotao Chu
- Global Biometrics & Data Sciences, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Xiaochen Zhu
- Global Biometrics & Data Sciences, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Jiaju Wu
- Global Biometrics & Data Sciences, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Yike Tang
- Global Biometrics & Data Sciences, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Jonathan Luu
- Graduate School of Arts and Sciences, Harvard University, Cambridge, Massachusetts, USA
| | - Chunsheng He
- Global Biometrics & Data Sciences, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Shu-Pang Huang
- Global Biometrics & Data Sciences, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Liangang Liu
- Global Biometrics & Data Sciences, Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Hsin-Ju Hsieh
- Global Biometrics & Data Sciences, Bristol Myers Squibb, Princeton, New Jersey, USA
| |
Collapse
|
10
|
Uzel Şener M, Akın Kabalak P, Kavurgacı S, Yılmaz Demirci N, Kızılgöz D, Yanık F, Ermin S, Söyler Y, Karamustafaoğlu YA, Türkay Pakna D, Dumanlı A, Yılmaz Ü. Different approach to M descriptor for future staging of oligometastatic disease in SCLC: A cross-sectional survival analysis. Clin Transl Oncol 2025; 27:2750-2760. [PMID: 39496913 DOI: 10.1007/s12094-024-03778-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 10/22/2024] [Indexed: 11/06/2024]
Abstract
PURPOSE This study aimed to investigate the impact of oligometastasis and the M descriptor on survival in small cell lung cancer (SCLC). METHODS This multicenter, retrospective study included patients with newly diagnosed extensive-stage SCLC(ES-SCLC) from 2010 to 2020. Subgroups: Group 1: single metastasis in a single organ, Group 2: 2-5 metastases in a single organ, Group 3: 6 or more metastases in a single organ, and Group 4: metastases in two or more organs. This classification was based on the 9th Staging-M descriptor. Three-year progression-free survival (PFS) and overall survival (OS) analyses were conducted. RESULTS The mean age of the 439 patients was 62 ± 10 years, and 89.5% of them were male. The mean PFS for Groups 1, 2, 3, 4 was 10.7 months (95% CI 8.9-12.5), 7.5 months (95% CI 5.6-9.4), 4.3 months (95% CI 2.9-5.7), and 5.4 months (95% CI 4.7-6.1), respectively. PFS in Group 2 was significantly higher. The mean OS for Groups 1, 2, 3, 4 was 13.3 months (95% CI 11.2-15.3), 9.5 months (95% CI 7.1-11.9), 7.1 months (95% CI 4.5-9.7), and 6.9 months (95% CI 6.0-7.9), respectively. OS in Group 1 was significantly higher. OS and PFS in the M1b group were significantly higher than in the M1c1 and M1c2 groups (p < 0.05) with no statistical difference between the M1c1 and M1c2 groups. CONCLUSION There is no significant difference in survival between the M1c1 and M1c2 groups. In ES-SCLC, the number of metastases may be a more predictive factor for prognosis than the number of metastatic organs.
Collapse
Affiliation(s)
- Melahat Uzel Şener
- Department of Chest Diseases, University of Health Sciences, Ankara Atatürk Sanatorium Training and Research Hospital, Keçiören, Ankara, Turkey.
| | - Pınar Akın Kabalak
- Department of Chest Diseases, University of Health Sciences, Ankara Atatürk Sanatorium Training and Research Hospital, Keçiören, Ankara, Turkey
| | - Suna Kavurgacı
- Department of Chest Diseases, University of Health Sciences, Ankara Atatürk Sanatorium Training and Research Hospital, Keçiören, Ankara, Turkey
| | | | - Derya Kızılgöz
- Department of Chest Diseases, University of Health Sciences, Ankara Atatürk Sanatorium Training and Research Hospital, Keçiören, Ankara, Turkey
| | - Fazlı Yanık
- Department of Thoracic Surgery, Faculty of Medicine, Trakya University, Edirne, Turkey
| | - Sinem Ermin
- Department of Chest Diseases, Dr. Suat Seren Chest Diseases and Surgery Training and Research Hospital, University of Health Sciences, Izmir, Turkey
| | - Yasemin Söyler
- Department of Chest Diseases, University of Health Sciences, Ankara Atatürk Sanatorium Training and Research Hospital, Keçiören, Ankara, Turkey
| | | | - Demet Türkay Pakna
- Department of Chest Diseases, University of Health Sciences, Ankara Atatürk Sanatorium Training and Research Hospital, Keçiören, Ankara, Turkey
| | - Ahmet Dumanlı
- Department of Thoracic Surgery, Faculty of Medicine, Afyon Health Sciences University, Afyonkarahisar, Turkey
| | - Ülkü Yılmaz
- Department of Chest Diseases, University of Health Sciences, Ankara Atatürk Sanatorium Training and Research Hospital, Keçiören, Ankara, Turkey
| |
Collapse
|
11
|
Kulkarni R, Zeine E, Potugari B, Gadgeel S, Montecalvo J, Rous FA. Small Cell Lung Cancer With de novo BRAF V600E Mutation and Durable Response to Targeted Therapy: A Case Report. Clin Lung Cancer 2025; 26:e306-e310. [PMID: 40133188 DOI: 10.1016/j.cllc.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/27/2025]
Affiliation(s)
- Radhika Kulkarni
- Division of Hematology/Oncology, Department of Internal Medicine, Henry Ford Cancer Institute/Henry Ford Health, Detroit, MI
| | - Elias Zeine
- Division of Hematology/Oncology, Karmanos Cancer Institute at McLaren Central Michigan, Morey Cancer Center, Mount Pleasant, MI
| | - Bindu Potugari
- Division of Hematology/Oncology, Department of Internal Medicine, Henry Ford Cancer Institute/Henry Ford Health, Detroit, MI
| | - Shirish Gadgeel
- Division of Hematology/Oncology, Department of Internal Medicine, Henry Ford Cancer Institute/Henry Ford Health, Detroit, MI
| | | | - Fawzi Abu Rous
- Division of Hematology/Oncology, Department of Internal Medicine, Henry Ford Cancer Institute/Henry Ford Health, Detroit, MI.
| |
Collapse
|
12
|
Simpson KL, Rothwell DG, Blackhall F, Dive C. Challenges of small cell lung cancer heterogeneity and phenotypic plasticity. Nat Rev Cancer 2025; 25:447-462. [PMID: 40211072 DOI: 10.1038/s41568-025-00803-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/19/2025] [Indexed: 04/12/2025]
Abstract
Small cell lung cancer (SCLC) is an aggressive neuroendocrine malignancy with ~7% 5-year overall survival reflecting early metastasis and rapid acquired chemoresistance. Immunotherapy briefly extends overall survival in ~15% cases, yet predictive biomarkers are lacking. Targeted therapies are beginning to show promise, with a recently approved delta-like ligand 3 (DLL3)-targeted therapy impacting the treatment landscape. The increased availability of patient-faithful models, accumulating human tumour biobanks and numerous comprehensive molecular profiling studies have collectively facilitated the mapping and understanding of substantial intertumoural and intratumoural heterogeneity. Beyond the almost ubiquitous loss of wild-type p53 and RB1, SCLC is characterized by heterogeneously mis-regulated expression of MYC family members, yes-associated protein 1 (YAP1), NOTCH pathway signalling, anti-apoptotic BCL2 and epigenetic regulators. Molecular subtypes are based on the neurogenic transcription factors achaete-scute homologue 1 (ASCL1) and neurogenic differentiation factor 1 (NEUROD1), the rarer non-neuroendocrine transcription factor POU class 2 homeobox 3 (POU2F3), and immune- and inflammation-related signatures. Furthermore, SCLC shows phenotypic plasticity, including neuroendocrine-to-non-neuroendocrine transition driven by NOTCH signalling, which is associated with disease progression, chemoresistance and immune modulation and, in mouse models, with metastasis. Although these features pose substantial challenges, understanding the molecular vulnerabilities of transcription factor subtypes, the functional relevance of plasticity and cell cooperation offer opportunities for personalized therapies informed by liquid and tissue biomarkers.
Collapse
Affiliation(s)
- Kathryn L Simpson
- SCLC Biology Group, Cancer Research UK Manchester Institute, Manchester, UK
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
| | - Dominic G Rothwell
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
| | - Fiona Blackhall
- CRUK Lung Cancer Centre of Excellence, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Medical Oncology, Christie Hospital National Health Service, Foundation Trust, Manchester, UK
| | - Caroline Dive
- SCLC Biology Group, Cancer Research UK Manchester Institute, Manchester, UK.
- CRUK National Biomarker Centre, University of Manchester, Manchester, UK.
- CRUK Lung Cancer Centre of Excellence, Manchester, UK.
| |
Collapse
|
13
|
Corica DA, Bell SD, Zhao L, Lawler NJ, Poirier MA, Miller PJ, Wakefield MR, Fang Y. The Era of Precision Medicine: Advancing Treatment Paradigms for Small Cell Lung Cancer. Cancers (Basel) 2025; 17:1847. [PMID: 40507328 PMCID: PMC12153792 DOI: 10.3390/cancers17111847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/29/2025] [Accepted: 05/29/2025] [Indexed: 06/16/2025] Open
Abstract
Small cell lung cancer (SCLC) remains a challenge prognostically. A clinically silent early stage and predilection for early metastasis leads to over half of patients presenting with metastatic disease at the time of diagnosis. Akin to many other cancers, once SCLC metastasizes, current therapies begin to lose their effectiveness. The future of SCLC rests in innovative treatments aimed at improving patient outcomes. Chemotherapy and radiation remain the backbone treatment for SCLC. Most patients diagnosed with SCLC begin treatment with combination chemotherapy consisting of a platinum analog and topoisomerase inhibitor with or without concurrent radiation. Disease progression or recurrence warrants new treatment approaches. New chemotherapy combinations and advances in radiation precision offer patients novel approaches using the same backbone of treatment used in many other cancers. The introduction of newer therapeutic approaches, such as immune checkpoint inhibitors, small molecule targeted therapies, bispecific antibodies, and antibody-drug conjugates offer a bright future for patients with SCLC who fail first-line therapy. This review will focus on advancing treatment paradigms for SCLC in the era of precision medicine. Such a study might be helpful for pulmonologists and oncologists to manage precisely patients with SCLC.
Collapse
Affiliation(s)
- Derek A. Corica
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
| | - Scott D. Bell
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
| | - Lei Zhao
- The Department of Respiratory Medicine, the 2nd People’s Hospital of Hefei and Hefei Hospital Affiliated to Anhui Medical University, Hefei 230002, China;
| | - Nicholas J. Lawler
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
| | - McKade A. Poirier
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
| | - Peyton J. Miller
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
| | - Mark R. Wakefield
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA;
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| | - Yujiang Fang
- Department of Microbiology, Immunology & Pathology, Des Moines University, West Des Moines, IA 50266, USA; (D.A.C.); (S.D.B.); (N.J.L.); (M.A.P.); (P.J.M.)
- Department of Surgery, University of Missouri School of Medicine, Columbia, MO 65212, USA;
- Ellis Fischel Cancer Center, University of Missouri School of Medicine, Columbia, MO 65212, USA
| |
Collapse
|
14
|
Chen F, Feng X, Xiao LL, Tang HJ, Qin SX, Peng JP, Bai J. Cost-effectiveness analysis of first-line tislelizumab plus chemotherapy for extensive-stage small cell lung cancer from the perspective of the healthcare system in China. Front Public Health 2025; 13:1552734. [PMID: 40520302 PMCID: PMC12162963 DOI: 10.3389/fpubh.2025.1552734] [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: 12/29/2024] [Accepted: 05/13/2025] [Indexed: 06/18/2025] Open
Abstract
Background Extensive-stage small-cell lung cancer (ES-SCLC) poses a formidable challenge due to its aggressive nature and poor prognosis. While immune checkpoint inhibitors have shown promise as part of first-line therapy, their cost-effectiveness and survival benefits in the Chinese healthcare system are not well understood. This study evaluates the cost-effectiveness of first-line tislelizumab combined with chemotherapy versus chemotherapy alone for ES-SCLC. Methods We conducted a cost-effectiveness analysis using a partitioned survival mode (PSM) to compare tislelizumab plus chemotherapy versus chemotherapy alone for the first-line treatment of ES-SCLC. The model integrated survival estimates from the RATIONALE-312 Phase III clinical trial, direct medical costs, and quality-adjusted life year (QALY) sourced from the literature. We calculated 10-year cost per QALY gained from Chinese healthcare system perspective. The analysis of cost-effectiveness was benchmarked against a willingness-to-pay threshold three times of GDP per capita in China. Sensitivity analyses were conducted to evaluate parametric uncertainty and model robustness. Results Compared to the chemotherapy alone group, the tislelizumab plus chemotherapy group resulted in an incremental cost-effectiveness ratio (ICER) of US$31,072.79 per quality-adjusted life-year (QALY), which is below the threshold of US$37,765 per QALY. Sensitivity analyses indicated that the utility value of progression-free survival (PFS) is a principal determinant of the ICER, with the ratio fluctuating between $27,246 and $36,417 per QALY, well below the willingness-to-pay threshold. In scenario analyses, tislelizumab plus chemotherapy resulted in an ICER of US$ 38,665.59/QALY with PET-CT imaging (exceeding the $37,765/QALY threshold) but was cost-effective at US$ 30,076.37/QALY when imported topotecan was used as second-line treatment. Conclusion Tislelizumab plus chemotherapy demonstrates cost-effectiveness in the first-line treatment of ES-SCLC in China. This study provides preliminary evidence for the economic value of tislelizumab in the treatment of ES-SCLC, supporting its consideration as a first-line therapeutic option.
Collapse
Affiliation(s)
- Feng Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xue Feng
- Department of Health Economics, School of Information and Management, Guangxi Medical University, Nanning, Guangxi, China
| | - Lin Lin Xiao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Hai Juan Tang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shu Xia Qin
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jie Ping Peng
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jing Bai
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
15
|
Guo Y, Han S, Guo Q, Zhai J, Ren X, Li S, Duan J. Real-world analysis of immunochemotherapy in recurrent small-cell lung cancer: opportunities for second-line approaches. Front Pharmacol 2025; 16:1591643. [PMID: 40520158 PMCID: PMC12162476 DOI: 10.3389/fphar.2025.1591643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 05/16/2025] [Indexed: 06/18/2025] Open
Abstract
Background Small-cell lung cancer (SCLC) has limited therapeutic options beyond first-line treatment, and the efficacy of PD-1/PD-L1-based immunotherapy in this setting remains uncertain. This study evaluates the efficacy and safety of serplulimab-based immunochemotherapy as a second- or later-line treatment for SCLC. Methods This retrospective, real-world study included 39 SCLC patients treated with post-initial serplulimab-based immunochemotherapy at Shanxi Provincial Cancer Hospital between May 2022 and November 2023. Primary and secondary endpoints were overall survival (OS) and progression-free survival (PFS), respectively. Cox analyses were conducted to explore factors associated with survival outcomes. Results The median follow-up duration was 13.7 months. The OS was 12.00 months (95% CI: 6.87-not reached), and the median PFS was 4.07 months (95% CI: 3.07-7.17), with an objective response rate of 20.51%. Patients who underwent immunotherapy re-challenge showed numerically higher median OS (12.77 vs. 9.17 months) and PFS (5.93 vs. 3.87 months) than those without prior immunotherapy. Patients with an objective response to front-line therapy exhibited a trend toward improved median OS (not reached vs. 6.47 months) and PFS (5.93 vs. 3.17 months). Cox analysis identified ECOG PS of 2, elevated LDH, ProGrp, and NSE, and liver metastasis were associated with worse OS. The most common adverse events were thrombocytopenia, elevated ALT, and hypothyroidism, with a manageable safety profile. Conclusion Second- or later-line serplulimab-based immunochemotherapy shows promising antitumor activity and survival benefits for SCLC, regardless of prior immunotherapy exposure. Although limited by sample size and retrospective design, these findings highlight the potential of immunotherapy combinations beyond first-line therapy.
Collapse
Affiliation(s)
- Yanrong Guo
- Department of Respiratory Medicine, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Songyan Han
- Department of Respiratory Medicine, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Qinxiang Guo
- Department of Respiratory Medicine, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Jinfang Zhai
- Department of Respiratory Medicine, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Xiaohui Ren
- Department of Respiratory Medicine, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Shengshu Li
- Department of Respiratory Medicine, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, China
| | - Jianchun Duan
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
Andrade-Perez V, Raynal NJM. Interplay Between the Epigenome, the Microenvironment, and the Immune System in Neuroblastoma. Cancers (Basel) 2025; 17:1812. [PMID: 40507292 PMCID: PMC12153772 DOI: 10.3390/cancers17111812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 05/10/2025] [Accepted: 05/20/2025] [Indexed: 06/16/2025] Open
Abstract
Neuroblastoma (NB) is the most prevalent extracranial childhood tumor and the third leading cause of death from cancer in children. Despite having a high overall survival rate for low- and intermediate-risk patients, survival rates for high-risk cases remain unsatisfactory. The current standard treatment for high-risk NB involves surgery, chemotherapy, radiotherapy, autologous stem cell transplantation, immunotherapy with anti-ganglioside GD2, and differentiation therapy with isotretinoin. Besides not being enough to achieve a high survival rate in high-risk patients, these treatments are associated with significant side effects. With next-generation sequencing technologies, a better understanding of the genetic and epigenetic landscapes of NB has been achieved. This has led to the study of novel treatments to improve the overall survival rate of high-risk NB and reduce the toxicity of conventional treatments. Current research is focusing on the development of targeted drugs for genetic and epigenetic alterations, and protein degraders. Moreover, immunotherapy to enhance anticancer immune responses and by using cell-engineering techniques with chimeric antigen receptor (CAR) T and NK cells are being explored to target NB cells. Here, we review promising novel treatment strategies for NB, which target genetics, epigenetics, the tumor microenvironment, and the immune landscape, highlighting preclinical studies and ongoing clinical trials.
Collapse
Affiliation(s)
- Valentina Andrade-Perez
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1C5, Canada
- Centre de Recherche de l’Hôpital Sainte-Justine, 3175, Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| | - Noël J.-M. Raynal
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montréal, QC H3T 1C5, Canada
- Centre de Recherche de l’Hôpital Sainte-Justine, 3175, Chemin de la Côte-Sainte-Catherine, Montréal, QC H3T 1C5, Canada
| |
Collapse
|
17
|
Sun H, Zhang CY, Zhang XH, Tai ZX, Su JW, Lin XC, Zhang SL, Li YF, Zhang C, Cai M, Zhang XC, Chen HJ, Zhou Q, Wu YL, Feng WN, Yang JJ. Transcriptomic analysis of transformed small-cell lung cancer from EGFR-mutated lung adenocarcinoma reveals distinct subgroups and precision therapy opportunities. Biomark Res 2025; 13:79. [PMID: 40437627 PMCID: PMC12121208 DOI: 10.1186/s40364-025-00789-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 05/14/2025] [Indexed: 06/01/2025] Open
Abstract
BACKGROUND Small-cell lung cancer (SCLC) transformation is one of the major mechanisms of resistance to Epidermal Growth Factor Receptor tyrosine kinase inhibitors (EGFR-TKIs). Chemotherapy is typically the recommended treatment for transformed SCLC (T-SCLC), similar to primary SCLC. However, the benefits of chemotherapy alone are minimal. Prior research highlights differences between the biological traits of T-SCLC and primary SCLC or EGFR-mutated lung adenocarcinoma (LUAD). This study aims to elucidate the molecular characteristics of T-SCLC and identify potential treatment modalities. METHODS We retrospectively collected tissue samples from LUAD, T-SCLC post-EGFR-TKI resistance, and primary SCLC. Genomics, transcriptomics, and proteomics were performed to clarify the differences between T-SCLC, LUAD, and primary SCLC. Hierarchical clustering analysis was then used to categorize the molecular subtype of T-SCLC, followed by a survival analysis based on these subtypes. RESULTS A study involving 61 patients investigated differences between LUAD, SCLC, and primary SCLC. RNA sequencing revealed distinct gene expression variations, particularly up-regulation in PPM1E, INSM1, and KCNC1 genes in T-SCLC. Pathway analysis linked T-SCLC to the cell cycle and neural differentiation. By conducting Hierarchical clustering analysis on RNA-seq data, the entire population can be categorized into two distinct groups. While certain T-SCLC showed similarities and differences compared to SCLC, with subtypes: LUAD-like and Non-LUAD-like. Notably, the LUAD-like subtype had significantly higher NKX2-1 expression (mean 371.8 vs. 41.8, P < 0.0001). T-SCLC treatment approaches were categorized into matched and unmatched groups, delineated by the alignment of specific therapies with corresponding pathologies. The matched group (13 cases) exhibited a significantly prolonged median progression-free survival compared to the unmatched group (10 cases) (5.4 months vs. 3.6 months, P = 0.02). CONCLUSIONS T-SCLC exhibits marked molecular distinctiveness, setting it apart not only from LUAD but also from classical SCLC. This distinction extends to its classification into two discernible molecular subtypes: LUAD-like and Non-LUAD-like. Customizing therapeutic protocols to align with these specific subtypes have the potential to identify the most appropriate treatment for T-SCLC.
Collapse
Affiliation(s)
- Hao Sun
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No 106, Zhongshan Second Road, Guangzhou, 510080, China
| | - Chan-Yuan Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No 106, Zhongshan Second Road, Guangzhou, 510080, China
- Department of Pulmonary Oncology, The First People's Hospital of Foshan, Foshan, Guangdong, 528000, China
| | - Xiu-Hao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No 106, Zhongshan Second Road, Guangzhou, 510080, China
| | | | - Jun-Wei Su
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No 106, Zhongshan Second Road, Guangzhou, 510080, China
| | - Xiao-Cheng Lin
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No 106, Zhongshan Second Road, Guangzhou, 510080, China
| | - Shi-Ling Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No 106, Zhongshan Second Road, Guangzhou, 510080, China
| | - Yu-Fa Li
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No 106, Zhongshan Second Road, Guangzhou, 510080, China
| | | | - Miao Cai
- Geneplus-Beijing, Beijing, China
| | - Xu-Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No 106, Zhongshan Second Road, Guangzhou, 510080, China
| | - Hua-Jun Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No 106, Zhongshan Second Road, Guangzhou, 510080, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No 106, Zhongshan Second Road, Guangzhou, 510080, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No 106, Zhongshan Second Road, Guangzhou, 510080, China.
| | - Wei-Neng Feng
- Department of Pulmonary Oncology, The First People's Hospital of Foshan, Foshan, Guangdong, 528000, China.
| | - Jin-Ji Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, No 106, Zhongshan Second Road, Guangzhou, 510080, China.
| |
Collapse
|
18
|
Wespiser M, Gille R, Perol M. Clinical progress of B7-H3 targeted antibody drug conjugate ifinatamab deruxtecan for small-cell lung cancer. Expert Opin Investig Drugs 2025. [PMID: 40418751 DOI: 10.1080/13543784.2025.2512566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 05/19/2025] [Accepted: 05/25/2025] [Indexed: 05/28/2025]
Abstract
INTRODUCTION Small cell lung cancer is an aggressive malignancy with limited treatment options and poor prognosis, particularly at extensive stage. While first-line platinum-etoposide chemotherapy combined with anti-PD-L1 therapy improves survival, most patients relapse, highlighting the need for novel therapies. AREAS COVERED B7-Homolog3 (B7-H3), an immune checkpoint molecule overexpressed in SCLC, has emerged as a promising therapeutic target. Ifinatamab deruxtecan (I-DXd) is an antibody-drug conjugate targeting B7-H3, delivering a topoisomerase I inhibitor. Early clinical trials (IDeate-PanTumor01 and IDeate-Lung01) have demonstrated encouraging results in pretreated ES-SCLC. In the 12 mg/kg cohort of IDeate-Lung01, I-DXd achieved an objective response rate of 54.8%, median progression-free survival of 5.5 months, and median overall survival of 11.8 months. Notably, it showed intracranial activity with a central nervous system-confirmed response rate of 37.8%. EXPERT OPINION I-DXd is currently being evaluated in the phase III IDeate-Lung02 trial. Its promising efficacy, manageable safety profile, and potential in combination strategies position it as a key candidate for future SCLC treatment.
Collapse
Affiliation(s)
- Mylène Wespiser
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Romane Gille
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Maurice Perol
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| |
Collapse
|
19
|
Zhang X, Zhang Y, Zhang J, Yuan J, Zhu S, Li X. Immunotherapy of small cell lung cancer based on prognostic nutritional index. Front Immunol 2025; 16:1560241. [PMID: 40491917 PMCID: PMC12146305 DOI: 10.3389/fimmu.2025.1560241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 05/08/2025] [Indexed: 06/11/2025] Open
Abstract
Platinum-based first-line chemotherapy for small lung cancers has been a mainstream therapy for the past several decades. However, its efficacy has been suboptimal, and the research is now focused on improving the treatment and prognosis of competitive nutrition and multidrug combination techniques. Small cell lung cancer (SCLC) is not only affected by smoking, age, sex and other external factors, but also the tumor micro-environment and the nutritional status of patients themselves are of great significance for the prevention and treatment of SCLC, a malignant tumor. According to past research, malnutrition is related to the intolerance to immunotherapy, decline in quality of life, psychological disturbances, and low survival rates and prognosis. Numerous studies have shown that a low Prognostic Nutritional Index (PNI) serves as an independent prognostic factor linked to reduced overall survival across various cancer types. Additionally, PNI has been associated with disease-free survival and progression-free survival in certain cancers, such as lung cancer (LC). Recent research has indicated that the PNI can serve as an independent predictor of both long-term outcomes and short-term complications in SCLC patients. However, a systematic consensus on this matter has yet to be established. This paper focuses on the role and influence of PNI in the immunotherapy of SCLC, and proposes the possibility of dietary therapy for SCLC patients under the guidance of PNI. Finally, the authors pointed out that PNI will become a new strategy for comprehensive immunotherapy of SCLC.
Collapse
Affiliation(s)
- Xinling Zhang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Yaxuan Zhang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jiaqi Zhang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Jinghua Yuan
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Shuying Zhu
- Department of Microbiology Laboratory, Yiwu Center for Disease Prevention and Control, Yiwu, China
| | - Xiaoping Li
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| |
Collapse
|
20
|
Sun RZ, Zong D, Chen X, Ge YZ, Jiang N, Zhao LJ, Song X, He X, Zhu XZ. The effect and toxicity profile of consolidative or salvage thoracic radiotherapy following chemoimmunotherapy in patients with extensive stage small cell lung cancer. J Biomed Res 2025; 39:1-11. [PMID: 40420608 DOI: 10.7555/jbr.39.20250067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/28/2025] Open
Abstract
This study evaluated the efficacy and safety of thoracic radiotherapy (TRT) after first-line chemotherapy or chemoimmunotherapy in patients with extensive-stage small cell lung cancer (ES-SCLC), focusing on the impact of different TRT timing strategies (consolidative vs. salvage) on survival. A total of 54 ES-SCLC patients treated between January 2019 and July 2022 were retrospectively analyzed. Patients receiving consolidative TRT (cTRT) within 3 months after first-line treatment completion were compared to those receiving salvage TRT (sTRT) following disease progression. Primary endpoints included overall survival (OS), progression-free survival (PFS), locoregional-free survival (LRFS), and distant metastasis-free survival (DMFS); safety was a secondary endpoint. The cTRT group (n=41) showed significantly longer median OS (26.6 vs. 14.8 months, P=0.048), PFS (12.9 vs. 3.5 months, P< 0.0001), and DMFS (10.7 vs. 3.4 months, P= 0.0044) than the sTRT group (n=13). Multivariate analysis identified cTRT as an independent favorable prognostic factor. No significant differences in OS or LRFS were found between high-dose (≥50 Gy) and low-dose (<50 Gy) TRT. Hematologic and respiratory toxicities were the most common adverse events, with acceptable tolerability. In conclusion, consolidative TRT after chemoimmunotherapy significantly improves survival outcomes in ES-SCLC patients, and low-dose TRT may be a suitable option.
Collapse
Affiliation(s)
- Ruo-Zhou Sun
- Department of Radiation Oncology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Dan Zong
- Department of Radiation Oncology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xin Chen
- Department of Radiation Oncology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yi-Zhi Ge
- Department of Radiation Oncology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Ning Jiang
- Department of Radiation Oncology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Li-Jun Zhao
- Department of Radiation Oncology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Xue Song
- Department of Radiation Oncology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| | - Xia He
- Department of Radiation Oncology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiang-Zhi Zhu
- Department of Radiation Oncology, Nanjing Medical University Affiliated Cancer Hospital, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, Jiangsu 210009, China
| |
Collapse
|
21
|
Orlandi R. Neuroendocrine neoplasms of the lung: The latest updates. World J Clin Oncol 2025; 16:106630. [DOI: 10.5306/wjco.v16.i5.106630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/21/2025] [Accepted: 04/01/2025] [Indexed: 05/19/2025] Open
Abstract
Neuroendocrine neoplasms are a group of tumors with heterogenous malignancy that evolve from neuroendocrine cells, most frequently in the gastrointestinal tract and in the lung. The latest 2021 World Health Organization (WHO) classification of lung tumors defines neuroendocrine neoplasms of the lung as an independent group of tumors, including typical and atypical neuroendocrine tumors and small cell and large cell neuroendocrine carcinomas. Although the overall nomenclature is essentially unchanged from the fourth WHO classification, there are several clinically relevant updates. In this review article, we discuss the epidemiological, clinical, diagnostic, therapeutic and prognostic features of these fascinating neoplasms, including the latest insights, current challenges and future perspectives.
Collapse
Affiliation(s)
- Riccardo Orlandi
- Department of Thoracic Surgery, University of Milan, Milan 20122, Italy
| |
Collapse
|
22
|
Xie M, Bao M, Dong X, Wu L, Liu L, Zhao J, Chu X, Wu Y, Ji X, Fang Y, Yu X, Zhang S, Wang Q, Hu T, Wang J, Zhu C, Su C. Harnessing local and system immune profiling delineating differential responders to first-line sintilimab (anti-PD-1 antibody) combined with chemotherapy in extensive-stage small cell lung cancer: an exploratory biomarker analysis of a phase II study. Signal Transduct Target Ther 2025; 10:168. [PMID: 40404633 PMCID: PMC12098833 DOI: 10.1038/s41392-025-02252-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 04/04/2025] [Accepted: 04/28/2025] [Indexed: 05/24/2025] Open
Abstract
While chemo-immunotherapy has been established as the frontline therapeutic regimen for extensive-stage small cell lung cancer (ES-SCLC), durable responses persist predominantly in a minor population of patients. This underscores critical need to elucidate underlying local tumor microenvironment and systematic immune profiles for biomarker discovery. In this phase II trial (ChiCTR2000038354), the efficacy, safety, and immune-genomic signatures of sintilimab (anti-PD-1 antibody) synergized with chemotherapy as first-line regimen for ES-SCLC were evaluated. The regimen demonstrated a median progression-free survival (PFS) of 6.9 months and median overall survival of 17.1 months, accompanied by a 12-month PFS rate of 16.9%, fulfilling the primary endpoint. Manageable grade 3 or 4 treatment-related adverse events developed in 27.3% (12/44) of patients. The exploratory study indicated a higher infiltration of CD4+/CD8+ CXCR5+ T follicle helper cells, CD8+CD103+ tissue-resident memory T cells, and B cells in tumor tissue, associated with better response and prognosis. The study also indicated the presence of tumor macrophages (CD68+CD163+CSF1R+SIGLEC5+) associated with immunotherapy resistance. Higher levels of monocyte-dendritic cells in pre-treatment peripheral blood mononuclear cells were found in durable clinical benefit group. Also, higher CD83, CD244, IL-12, and CD70, which are hallmarks of dendritic cells and activated T cells, were discovered by plasma proteomics to be connected with enhanced outcomes, while chemoattractant of macrophage, CSF-1, CCL3, CCL4, and IL-8, were found to predict a worse prognosis. Furthermore, a multimodal model was constructed and validated for stratifying ES-SCLC into high or low risk to predict the immunotherapy efficacy. This study sheds light on harnessing local and systematic immune profiles to better stratify patients with ES-SCLC for immunotherapy and putative combinational treatment.
Collapse
Affiliation(s)
- Mengqing Xie
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Minwei Bao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Xiaorong Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Wu
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Changsha, China
| | - Li Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Jing Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Xiangling Chu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Yan Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Xianxiu Ji
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Yujia Fang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Xin Yu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Shiji Zhang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Qi Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China
| | - Tao Hu
- Amoy Diagnostics Co., Ltd., Xiamen, Fujian, China
| | - Jin Wang
- Amoy Diagnostics Co., Ltd., Xiamen, Fujian, China
| | - Changbin Zhu
- Amoy Diagnostics Co., Ltd., Xiamen, Fujian, China
| | - Chunxia Su
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
23
|
Zhou S, Zhai W, Zhang Q, Li H, Fan Y. Impact of prophylactic cranial irradiation on survival in extensive-stage small cell lung cancer receiving first-line chemoimmunotherapy: a propensity score-matched study. Ther Adv Med Oncol 2025; 17:17588359251341158. [PMID: 40415872 PMCID: PMC12102569 DOI: 10.1177/17588359251341158] [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: 02/05/2025] [Accepted: 04/23/2025] [Indexed: 05/27/2025] Open
Abstract
Background Chemoimmunotherapy has emerged as the standard first-line treatment for extensive-stage small cell lung cancer (ES-SCLC), improving survival outcomes. However, the role of prophylactic cranial irradiation (PCI) in the context of chemoimmunotherapy remains undefined. Objectives This study aimed to evaluate the impact of PCI on overall survival (OS) in patients with ES-SCLC after chemoimmunotherapy administration. Design Retrospective study. Methods This retrospective analysis included 261 patients with ES-SCLC treated with first-line chemoimmunotherapy between January 2019 and December 2023. All patients underwent MRI scans to confirm the absence of brain metastases. After 1:2 propensity score matching (PSM), 46 and 81 patients were assigned to the PCI and observation groups, respectively. The primary endpoint was OS, with additional exploration of progression-free survival (PFS), the cumulative incidence of intracranial metastases, and intracranial progression-free survival (iPFS). Results After PSM, the two groups were well-balanced in baseline characteristics. Survival analysis showed a median OS of 19.9 months (95% confidence interval (CI): 11.8-28.0) in the PCI group and 15.6 months (12.3-18.9) in the observation group, without a significant difference (hazard ratio (HR) = 0.763 (95% CI: 0.484-1.206), log-rank p = 0.265). PCI significantly reduced the risk of brain metastasis (Fine-Gray p = 0.002), with 1-year cumulative incidence rates of 13.8% (3.4%-24.2%) in the PCI group and 53.4% (41.3%-65.6%) in the observation group. Subgroup analysis showed that for ES-SCLC patients achieving a partial response to initial chemoimmunotherapy, the PCI group had longer median OS (25.7 months (95% CI: 15.4-36.1) vs 19.4 months (15.4-23.4); HR = 0.502 (0.284-0.886); log-rank p = 0.021). Conclusion PCI did not improve OS in ES-SCLC patients receiving first-line chemoimmunotherapy, while it may confer a survival benefit for patients who achieve remission following chemoimmunotherapy. In addition, PCI significantly reduced the incidence of brain metastases. These findings warrant further randomized studies for verification.
Collapse
Affiliation(s)
- Shichao Zhou
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Wanchen Zhai
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Department of Oncology, The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Qian Zhang
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Department of Oncology, The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Hui Li
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang 310022, China
| | - Yun Fan
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine, Chinese Academy of Sciences, No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
24
|
Zhang Q, Wang G, Yan W, Wang D, Yin J, Song Y, Ye M, Lv T. Molecular subtyping dictates therapeutic response to anti-PD-L1 immunotherapy in ES-SCLC. Cancer Immunol Immunother 2025; 74:213. [PMID: 40402312 PMCID: PMC12098246 DOI: 10.1007/s00262-025-04068-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 04/25/2025] [Indexed: 05/23/2025]
Abstract
Anti-PD-L1 immunotherapy is recommended as standard of care for patients with extensive stage small cell lung cancer (ES-SCLC); however, there are no reliable biomarkers guiding patient selection and the survival benefit of PD-L1 inhibitors in the overall population is limited. In this study, we retrospectively analyzed a total number of 61 cases of ES-SCLC who underwent anti-PD-L1 immunotherapy. Patient demographic characteristics and laboratory findings were processed for univariate and multivariate analysis. Subgrouping of SCLC was performed on IHC platform using antibodies against ASCL1, NEUROD1 and POU2F3. The tumor microenvironment (TME) of ES-SCLC was evaluated by CD8 + T cell infiltration, granzyme B production and PD-L1 expression. We found limited efficacy of defined variable factors conferring therapeutic outcomes of anti-PD-L1 immunotherapy in patients with ES-SCLC. Intriguingly, there was a profound difference in TME and response to anti-PD-L1 immunotherapy when classifying SCLC into A/N/P/I subgroups. Although accounted for a small proportion of SCLC, the SCLC-P and SCLC-I subtypes manifested as T cell-enriched "hot" tumor and elicited more favorable response to immunotherapy, whereas the SCLC-A and SCLC-N subgroups were T cell-absent "cold" tumor. There was also a significant difference in progression free survival and overall survival across these subsets. Moreover, we found the SCLC-P and SCLC-I tumors revealed features of low neuroendocrine (NE) differentiation and showed clinicopathologic features overlapping with the SCLC non-NE lineage. These findings may aid clinicians to select ES-SCLC patients who were more likely to gain higher response rate and longer survival to anti-PD-L1 immunotherapy. Revisiting SCLC according to A/N/P/I subtyping and NE/non-NE differentiation is a reliable approach to guide therapeutic strategy in patients with ES-SCLC.
Collapse
Affiliation(s)
- Qianqian Zhang
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, #305 East Zhongshan Road, Nanjing, 210002, China
| | - Guoxin Wang
- Department of Respiratory Medicine, Affiliated Hospital to Medical School, Nanjing University, Nanjing, 210002, China
| | - Wenjie Yan
- Department of Respiratory Medicine, Affiliated Hospital to Medical School, Nanjing University, Nanjing, 210002, China
| | - Dong Wang
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, #305 East Zhongshan Road, Nanjing, 210002, China
- Department of Respiratory Medicine, Affiliated Hospital to Medical School, Nanjing University, Nanjing, 210002, China
| | - Jie Yin
- Department of Respiratory Medicine, Affiliated Hospital to Medical School, Nanjing University, Nanjing, 210002, China
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, #305 East Zhongshan Road, Nanjing, 210002, China.
- Department of Respiratory Medicine, Affiliated Hospital to Medical School, Nanjing University, Nanjing, 210002, China.
| | - Mingxiang Ye
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, #305 East Zhongshan Road, Nanjing, 210002, China.
- Department of Respiratory Medicine, Affiliated Hospital to Medical School, Nanjing University, Nanjing, 210002, China.
| | - Tangfeng Lv
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, #305 East Zhongshan Road, Nanjing, 210002, China.
- Department of Respiratory Medicine, Affiliated Hospital to Medical School, Nanjing University, Nanjing, 210002, China.
| |
Collapse
|
25
|
Wu Y, Deng L, Wang J, Zhang T, Cao J, Zhou X, Duan J, Bi N. Single-arm phase II study of consolidation serplulimab following hypofractionated radiotherapy with concurrent chemotherapy for patients with limited stage small-cell lung cancer: ASTRUM-LC01 study protocol. BMJ Open 2025; 15:e085552. [PMID: 40398957 PMCID: PMC12096970 DOI: 10.1136/bmjopen-2024-085552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/30/2025] [Indexed: 05/23/2025] Open
Abstract
INTRODUCTION With the inspiring results of the PACIFIC trial in non-small-cell lung cancer (NSCLC), and the CAPIAN and IMpower133 trials in extensive-stage small-cell lung cancer (SCLC), immunotherapy has increasingly gained attention. Serplulimab, a PD-1 inhibitor, showed great antitumour activity in the ASTRUM-005 trial and has been recommended as first-line therapy in extensive-stage SCLC. Whether serplulimab following hypofractionation radiotherapy and chemotherapy could bring better outcomes in limited-stage SCLC remains to be answered. METHODS AND ANALYSIS We designed a prospective multicentre single-arm phase II clinical trial to evaluate both the efficacy and safety of chemoradiotherapy and consolidation by serplulimab in limited-stage SCLC. Eligible patients will receive standard chemotherapy for four cycles and concurrent thoracic radiotherapy with a total dose of 45 Gy in 3 weeks and a 3 Gy dose per fraction. Prophylactic cranial irradiation is recommended for responding patients. Serplulimab will be delivered afterwards every 3 weeks for up to 1 year. Based on sample size estimation, 55 patients will be enrolled in total. ETHICS AND DISSEMINATION Ethics approval was obtained from the Independent Ethics Committee of National Cancer Centre/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College (22/236-3438). TRIAL REGISTRATION NUMBER NCT05443646.
Collapse
Affiliation(s)
- Yuqi Wu
- Department of Radiation Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Lei Deng
- Department of Radiation Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Jianyang Wang
- Department of Radiation Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Tao Zhang
- Department of Radiation Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Jianzhong Cao
- Department of Radiation Oncology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Taiyuan, People's Republic of China
| | - Xiaohong Zhou
- Department of Radiation Oncology, Jiamusi Medical College, Jiamusi, Heilongjiang, China
| | - Jianchun Duan
- Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| | - Nan Bi
- Department of Radiation Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
26
|
Kejamurthy P, Mk J, Kt RD. A novel anti-PD-L1 DNA aptamer, Apta35 enhances non-small cell lung cancer cell cytotoxicity and apoptosis through lung cancer-activated T lymphocytes. Int Immunopharmacol 2025; 155:114621. [PMID: 40209314 DOI: 10.1016/j.intimp.2025.114621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 01/31/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
The prevalence of Programmed death ligand 1 (PD-L1) expression in the population of NSCLC patients and blocking the PD1/PD-L1 pathway by inhibiting the PD-1 receptor on immune cells or the PD-L1 ligand on tumour and/or immune cells can inhibit tumour growth. EFBALite algorithm that enables efficient and cost-effective selection of aptamers, expediting the process. Here, we present the development, computational validation, and in vitro analysis of NSCLC of DNA aptamers targeting PD-L1. The Gibbs free energy of two anti-PD-L1 aptamers, Apta35 and Apta90 with -3.06 and - 2.4 kcal/mol respectively. The docking score for Apta35 was -272.3 and 1171.765 for HDOCK and ZDOCK respectively. Further, the Apta35 was taken for the in vitro studies as it was more stable and incubated with NCI-H460. Initially, we confirmed the binding of the TAMRA-labelled Apta35 to the NCI-H460 cell surface through microscopic imaging and further confirmed through FACS analysis. Further experimental results showed that the Apta35 treated along with the act-T cells group reduced the percentage of viability (28 ± 3.5), increased toxicity, and reduced count of NCI-H460 cells when compared with the cells treated only with the act-T cells concerning the treatment to 50 nM concentration. In summary, targeting PD-L1 with a specific aptamer provides an innovative strategy for targeting NSCLC. Apta35 aptamer showed no significant toxicity in the BALB/c nude mice while it was injected every 2 days for a total of 12 days of treatment.
Collapse
Affiliation(s)
- Priyatharcini Kejamurthy
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Jaganathan Mk
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Ramya Devi Kt
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India.
| |
Collapse
|
27
|
Monaca F, Gomez-Randulfe I, Parreira AS, Longo V, Galetta D, Pilotto S, Polidori S, Cantale O, Stefani A, Vita E, Taylor P, Gomes F, Cove-Smith L, Summers Y, Tortora G, Blackhall F, Novello S, Bria E, Califano R. Correlation between irAEs and survival outcomes in patients with ES-SCLC treated with first-line chemoimmunotherapy. Eur J Cancer 2025; 221:115435. [PMID: 40250285 DOI: 10.1016/j.ejca.2025.115435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 04/04/2025] [Accepted: 04/13/2025] [Indexed: 04/20/2025]
Abstract
INTRODUCTION Chemo-immunotherapy (CT-IO) has improved median overall survival (mOS) for patients with extensive-stage small cell lung cancer (ES-SCLC), but its association with immune-related adverse events (irAEs) remains unclear. While irAEs are often linked to better outcomes in other cancers, their prognostic value in ES-SCLC is not well understood. METHODS We conducted a retrospective analysis of 399 consecutive ES-SCLC patients treated with first-line CT-IO between January 2020 and September 2024 across five European centres. Demographic and clinical data were collected. The impact of irAEs on progression-free survival (PFS) and overall survival (OS) was assessed using time-dependent Cox regression. RESULTS The median follow-up was 15.0 months. The overall response rate was 80.3 %, with a median PFS of 6.0 months (95 % CI 5.7-6.3) and mOS of 10.4 months (95 % CI 9.2-11.6). IrAEs occurred in 30.6 % of patients, most commonly affecting the skin (11.0 %). The median time to onset of irAEs was 171 days. Patients with irAEs had significantly longer mPFS (10.8 vs. 5.3 months, p < 0.001) and mOS (18.8 vs. 7.6 months, p < 0.001) compared to those without. No significant difference was found between patients with grade ≥ 3 (n = 46) and < 3 irAEs (n = 76). Multivariate analysis confirmed that irAEs were associated with improved OS (HR 0.64; 95 % CI 0.51-0.80, p < 0.001) and showed a trend towards longer PFS (p = 0.028). CONCLUSION This is the largest retrospective study to demonstrate that irAEs are associated with improved clinical outcomes in ES-SCLC pts receiving 1 L CT-IO.
Collapse
Affiliation(s)
- Federico Monaca
- Università Cattolica del Sacro Cuore, Rome, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Igor Gomez-Randulfe
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Ana Sofia Parreira
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Vito Longo
- Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Domenico Galetta
- Thoracic Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Sara Pilotto
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Sara Polidori
- Università Cattolica del Sacro Cuore, Rome, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ornella Cantale
- Department of Oncology, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Torino, Italy
| | - Alessio Stefani
- Università Cattolica del Sacro Cuore, Rome, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Emanuele Vita
- Università Cattolica del Sacro Cuore, Rome, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Paul Taylor
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Fabio Gomes
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Laura Cove-Smith
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Yvonne Summers
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Giampaolo Tortora
- Università Cattolica del Sacro Cuore, Rome, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Fiona Blackhall
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Silvia Novello
- Department of Oncology, University of Turin, San Luigi Gonzaga Hospital, Orbassano, Torino, Italy
| | - Emilio Bria
- Università Cattolica del Sacro Cuore, Rome, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy; Ospedale Isola Tiberina - Gemelli Isola, Rome, Italy
| | - Raffaele Califano
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK; Division of Cancer Sciences, The University of Manchester, Manchester, UK.
| |
Collapse
|
28
|
Zhang Y, Xu Y, Zhong W, Zhao J, Liu X, Gao X, Chen M, Wang M. Vitamin D and Immune Checkpoint Inhibitors in Lung Cancer: A Synergistic Approach to Enhancing Treatment Efficacy. Int J Mol Sci 2025; 26:4511. [PMID: 40429656 PMCID: PMC12111780 DOI: 10.3390/ijms26104511] [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: 03/15/2025] [Revised: 04/27/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Lung cancer, a malignant neoplasm that is globally prevalent and characterized by high incidence and mortality rates, has seen the rise of immune checkpoint inhibitors (ICIs) as a crucial systemic treatment. However, a subset of patients exhibits suboptimal responses to ICIs. Recently, studies revealed the role of vitamin D in inflammation modulation, cell differentiation, and cancer prevention. Vitamin D precisely modulates immune responses and inflammatory states within the tumor microenvironment (TME) by targeting both innate and adaptive immunity. These effects may reduce immune tolerance to ICIs and synergistically enhance their therapeutic efficacy. Here, we review vitamin D metabolism in lung cancer patients, as well as its anti-tumor mechanisms, immune regulation, and the significant promise of vitamin D in lung cancer immunotherapy and adjuvant therapeutic strategies. Further research is imperative to surmount these challenges and fully realize vitamin D's potential in improving lung cancer immunotherapy outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Minjiang Chen
- Department of Respiratory & Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100730, China; (Y.Z.); (Y.X.); (W.Z.); (J.Z.); (X.L.); (X.G.); (M.W.)
| | | |
Collapse
|
29
|
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
|
30
|
Lamberti G, Rihawi K, Mazzoni F, Riccardi F, Follador A, Tiseo M, Frassoldati A, Colantonio I, Bonetti A, Genova C, Giardina D, Bertolini F, Cinieri S, Pasello G, Brighenti M, Andrini E, Tognetto M, Boni L, Ardizzoni A. Carboplatin, etoposide, atezolizumab, and bevacizumab in the first-line treatment of patients with extensive stage small-cell lung cancer: the GOIRC-01-2019 CeLEBrATE study. J Immunother Cancer 2025; 13:e010694. [PMID: 40341031 PMCID: PMC12067786 DOI: 10.1136/jitc-2024-010694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 04/18/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND The addition of a programmed death-ligand 1 (PD-L1) inhibitor, either atezolizumab or durvalumab, to platinum-etoposide prolonged survival in a limited subset of patients with extensive-stage small-cell lung cancer (ES-SCLC). Preclinical studies demonstrated synergistic antitumor activity of combined vascular endothelial growth factor receptor and PD-L1 inhibition in SCLC. Since bevacizumab added to platinum-etoposide was safe and active in ES-SCLC, we investigated the efficacy of atezolizumab, bevacizumab, carboplatin, and etoposide as first-line treatment of ES-SCLC. METHODS The CeLEBrATE study is an Italian multicentric single-arm phase II trial of carboplatin (area under the curve 5 ml/min), etoposide (100 mg/sqm), bevacizumab (7.5 mg/kg), and atezolizumab (1,200 mg) every 3 weeks (q3w) for four to six courses, followed by bevacizumab and atezolizumab maintenance q3w in patients with ES-SCLC and no contraindications to immunotherapy or antiangiogenic therapy. Patients with asymptomatic brain metastases were eligible. Prophylactic cranial irradiation and consolidation thoracic external radiotherapy were not permitted while on study treatment. Primary endpoint was overall survival (OS) rate at 1 year. RESULTS 53 patients were enrolled (45.3% women, median age 65 years) and received at least one dose of study treatment. At a median follow-up time of 23.4 months (95% CI: 21.1 to 26.0), the 1-year OS rate was 61.8% (90% CI: 50.7% to 72.8%; p=0.04), with a median OS of 12.9 months (95% CI: 11.6 to 17.5). Median progression-free survival was 6.2 months (95% CI: 5.4 to 6.6) and objective response rate was 83.3% (95% CI: 69.8% to 92.5%). Grade 3-4 adverse events were reported in 34 patients (64.2%) leading to dose reductions in 24 (45.3%), and dose delays in 39 (73.9%) and 32 (69.6%) during the induction and maintenance phase, respectively. 19 (35.8%) treatment-related serious adverse events were reported. CONCLUSION The CeLEBrATE study met its primary objective demonstrating a signal of efficacy of bevacizumab, atezolizumab, carboplatin, and etoposide in the first-line treatment of patients with ES-SCLC. TRIAL REGISTRATION NUMBER GOIRC-01-2019 ML41241, Eudract Number: 2019-003798-2.
Collapse
Affiliation(s)
- Giuseppe Lamberti
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
| | - Karim Rihawi
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
| | - Francesca Mazzoni
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Medical Oncology Unit, Department of Oncology, Careggi University Hospital, Firenze, Italy
| | - Ferdinando Riccardi
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Medical Oncology Unit, Azienda Ospedaliera Cardarelli, Napoli, Italy
| | - Alessandro Follador
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Department of Oncology, University Hospital Santa Maria Della Misericordia, Udine, Italy
| | - Marcello Tiseo
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Department of Medicine and Surgery, University of Parma and Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Antonio Frassoldati
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Department of Oncology, Azienda Ospedaliero Universitaria di Ferrara-Arcispedale Sant'Anna, Ferrara, Italy
| | - Ida Colantonio
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Medical Oncology Unit, S. Croce e Carle General Hospital, Cuneo, Italy
| | - Andrea Bonetti
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Department of Oncology, "Mater Salutis" Hospital, Legnago, Italy
| | - Carlo Genova
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- AcademicOncology Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine and Medical Specialties (DiMI), Università degli Studi di Genova, Genova, Italy
| | - Donatella Giardina
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Medical Oncology, Ospedale Ramazzini di Carpi and Ospedale di Mirandola, Azienda Usl Modena, Carpi, Italy
| | - Federica Bertolini
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Division of Medical Oncology, Azienda Ospedaliero-Universitaria Policlinico, Modena, Italy
| | - Saverio Cinieri
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Medical Oncology Unit, Hospital of Brindisi, Brindisi, Italy
| | - Giulia Pasello
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Matteo Brighenti
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Medical Oncology Department, ASST Cremona, Cremona, Italy
| | - Elisa Andrini
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Michele Tognetto
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
| | - Luca Boni
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
- Clinical Epidemiology Unit, IRCSS Ospedale Policlinico San Martino, Genova, Italy
| | - Andrea Ardizzoni
- Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum University of Bologna, Bologna, Italy
- Medical Oncology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Gruppo Oncologico Italiano di Ricerca Clinica (GOIRC), Parma, Italy
| |
Collapse
|
31
|
Kim H, Hwang J, Kim SM, Yang DS. Preliminary Results of Clinical Experience with Consolidative High-Dose Thoracic Radiotherapy for Patients with Extensive-Stage Small Cell Lung Cancer. Tomography 2025; 11:55. [PMID: 40423257 DOI: 10.3390/tomography11050055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/09/2025] [Accepted: 05/06/2025] [Indexed: 05/28/2025] Open
Abstract
OBJECTIVES Extensive-stage small-cell lung cancer (SCLC) has a poor prognosis, but recently, the combination of immunotherapy and chemotherapy has improved treatment outcomes in some patients, and treatment plans may vary depending on the individual's general condition and tumor response. In addition, intrathoracic tumor control remains a major challenge for this disease. In the current study, we aim to share our clinical experience and demonstrate that consolidative high-dose thoracic radiotherapy effectively reduces intrathoracic tumor recurrence while maintaining acceptable treatment-related toxicities. MATERIALS AND METHODS The medical records of 81 SCLC patients treated at Korea University Guro Hospital from January 2019 to December 2023 were reviewed retrospectively. Among them, 22 patients with extensive-stage SCLC who had a favorable tumor response after systemic therapy, including those with oligo-progressive disease limited to the thoracic region and suitable for curative local therapy, received consolidative radiotherapy. A total dose of 52.5 Gy in 25 fractions was administered over 5 weeks to all patients with extensive-stage SCLC. RESULTS AND CONCLUSIONS The median follow-up time was 22 months (range: 8-59 months), with the median follow-up period after completing consolidative radiotherapy being 13 months (range: 4-35 months). In-field local recurrence occurred in only one patient after consolidative thoracic radiotherapy. Most importantly, 10 patients with oligo-progressive disease at the thoracic site, at the time of tumor response, remained stable without further intrathoracic in-field recurrence. Additionally, no severe cases of radiation pneumonitis or esophagitis were observed. Based on our institution's experience, consolidative high-dose thoracic radiotherapy is well-tolerated and associated with fewer intrathoracic recurrences, leading to improved long-term survival in carefully selected patients with extensive-stage SCLC. Given these findings, we believe consolidative radiotherapy should be considered more proactively in clinical practice. Furthermore, these results may help guide the design of future clinical trials.
Collapse
Affiliation(s)
- Hakyoung Kim
- Departments of Radiation Oncology, Korea University Guro Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Jeongeun Hwang
- Department of Medical IT Engineering, College of Software Convergence, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Sun Myung Kim
- Departments of Radiation Oncology, Korea University Guro Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Dae Sik Yang
- Departments of Radiation Oncology, Korea University Guro Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea
| |
Collapse
|
32
|
Yang N, Ma ZX, Wang X, Xiao L, Jin L, Li M. Development and validation of a CT-based radiomics nomogram for predicting progression-free survival in patients with small cell lung cancer. BMC Med Imaging 2025; 25:154. [PMID: 40329257 PMCID: PMC12057258 DOI: 10.1186/s12880-025-01691-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 04/25/2025] [Indexed: 05/08/2025] Open
Abstract
PURPOSE Small cell lung cancer (SCLC) is a highly aggressive form of lung cancer, representing about 15% of cases worldwide. Despite advances in imaging, such as low-dose CT, which have increased diagnostic rates, survival outcomes for SCLC patients have remained stagnant. Recent studies have only focused on radiomics, which extracts detailed quantitative features from imaging, with clinical risk factors to improve prognostic models. Therefore, this study aimed to develop a clinical-radiomics fusion nomogram based on computed tomography (CT) to estimate progression-free survival (PFS) in patients diagnosed with SCLC. By integrating radiomics features extracted from CT with clinical data, this model provides personalized prognostic assessment for clinicians. Its clinical utility lies in aiding treatment decision-making by offering more accurate prognostic evaluation, optimizing therapeutic strategies, and identifying high-risk patients at an early stage, ultimately improving overall survival and quality of life. METHODS To develop the nomogram model, 95 patients diagnosed with pathologically confirmed SCLC between January 1, 2013, and December 31, 2023, were included in the study cohort. Participants were randomly divided into training and validation cohorts in a 7:3 ratio. Radiomics features associated with PFS were generated using the least absolute shrinkage and selection operator (LASSO) along with univariate and multivariate analyses. Additionally, in the training cohort, both univariate and multivariate analyses using Cox regression were conducted to identify the significant clinical risk factors influencing PFS. The predictive performance of the clinical and clinical-radiomics fusion nomogram were evaluated using the concordance index, calibration plots, and decision curve analysis (DCA). RESULTS Five radiomics features were selected and used to calculate the radiomics score (Rad-score). The radiomics features were significantly associated with PFS (hazard ratio: 0.5765, 95% confidence interval: 0.3641-0.9128, p < 0.05). Three clinical risk factors significantly associated with PFS were identified: neuron-specific enolase (NSE), carbohydrate antigen 125 levels (CA125), and treatment type, such as surgery. The clinical-radiomics fusion nomogram model (C-index:0.744) demonstrated superior performance compared to the clinical nomogram model (C-index: 0.718) in the training cohort. DCA indicated that the clinical-radiomics fusion nomogram outperformed the clinical nomogram in terms of clinical usefulness. CONCLUSIONS A CT-based clinical-radiomics fusion nomogram was developed to predict PFS in patients with SCLC, which is useful in providing individualized information. ADVANCES IN KNOWLEDGE A clinical-radiomics fusion nomogram was constructed to estimate the probability of PFS based on clinical risk factors and the rad-score.
Collapse
Affiliation(s)
- Nan Yang
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, 200040, China
- Zhang Guozhen Small Pulmonary Nodules Diagnosis and Treatment Center, Shanghai, 200040, China
| | - Zhuang Xuan Ma
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, 200040, China
- Zhang Guozhen Small Pulmonary Nodules Diagnosis and Treatment Center, Shanghai, 200040, China
| | - Xin Wang
- Department of Pathology, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Li Xiao
- Department of Pathology, Huadong Hospital, Fudan University, Shanghai, 200040, China
| | - Liang Jin
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, 200040, China.
- Zhang Guozhen Small Pulmonary Nodules Diagnosis and Treatment Center, Shanghai, 200040, China.
| | - Ming Li
- Department of Radiology, Huadong Hospital, Fudan University, Shanghai, 200040, China.
- Zhang Guozhen Small Pulmonary Nodules Diagnosis and Treatment Center, Shanghai, 200040, China.
| |
Collapse
|
33
|
Vitale E, Rizzo A, Maistrello L, Guven DC, Cauli O, Galetta D, Longo V. Treatment-Related Adverse Events in Extended Stage Small Cell Lung Cancer Patients Receiving First-Line Chemoimmunotherapy Versus Chemotherapy Alone: A Systematic Review and Meta-Analysis. Cancers (Basel) 2025; 17:1571. [PMID: 40361497 PMCID: PMC12072015 DOI: 10.3390/cancers17091571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/28/2025] [Accepted: 05/03/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction: Nowadays the prognosis of extended stage (ES) small cell lung cancer (SCLC) patients is poor. However, a high response rate to first-line chemotherapy (CT) and the addition of immune checkpoint inhibitors (ICIs) have notably ameliorated the outcome of these patients. The aim of our study is to compare treatment-related adverse events (TRAEs) between ES- SCLC patients receiving first-line ICIs adding CT and those receiving only CT. Methods: All phase III clinical trials published between 15 June 2008, and 30 June 2024, likenessing ICIs adding systemic CT and only CT in treatment-naïve ES-SCLC patients were retrieved. Results: Twenty-six types of adverse events were included, grouped into ten categories, for a total of 43,391 observations (observations in immune group n = 22,643 and in placebo group n = 20,748) and 9831 events. Our analysis suggested a statistically significant increase in hematological events in patients receiving ICIs plus CT compared with CT alone. Conversely, blood pressure alterations such as hypertension were more frequent in patients treated with CT alone. Conclusions: Despite our analysis confirming the manageable safety profile of chemoimmunotherapy, this remains an issue to be further investigated.
Collapse
Affiliation(s)
- Elsa Vitale
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (E.V.); (D.G.)
| | - Alessandro Rizzo
- Struttura S.S.D.C.O.r.O., Bed Management Presa in Carico, TDM, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
| | | | - Deniz Can Guven
- Medical Oncology Clinic, Health Sciences University, Elazig City Hospital, Elazig 23280, Turkey;
| | - Omar Cauli
- Nursing Department, Faculty of Nursing and Podiatrics, Universitat de València, 46010 Valencia, Spain;
| | - Domenico Galetta
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (E.V.); (D.G.)
| | - Vito Longo
- Medical Thoracic Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (E.V.); (D.G.)
| |
Collapse
|
34
|
Morinaga D, Sakakibara-Konishi J, Kawai Y, Morinaga Y, Mizobuchi S, Okamoto Y, Yamanaka Y, Takahashi K, Kikuchi H, Sukoh N, Takashina T, Kitai H, Konno S. Efficacy of second line and subsequent treatments of small cell lung cancer with and without immune checkpoint inhibitor combination therapy. Respir Investig 2025; 63:423-430. [PMID: 40120158 DOI: 10.1016/j.resinv.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/19/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) combined with platinum-doublet chemotherapy (ICI-chemo) have become the standard of care for extensive-stage small cell lung cancer (ES-SCLC). However, the effect of ICI-chemo on the efficacy of subsequent chemotherapy remains unknown. This study aimed to investigate the efficacy of second and subsequent treatments of SCLC with and without ICI combination therapy. METHODS We performed an analysis of patients with ES-SCLC between January 2015 and June 2023. The ICI-chemo groups were defined as patients who received ICI-chemo as first-line therapy between September 2019 and June 2023, after ICI-chemo was reimbursed in Japan. The non-ICI-chemo groups were defined as patients who received platinum-doublet therapy between January 2015 and August 2019 and were considered eligible for ICI-chemo. RESULTS In total, 224 patients were included (91 and 133 patients who received ICI-chemo and non-ICI-chemo, respectively). There were no significant differences in patient characteristics between the groups. There was no significant difference in progression-free survival (PFS) and overall survival (OS) for first-line treatment between the two groups. The median PFS and OS periods for second-line treatment were 3.9 and 3.9 months and 10.3 and 10.7 months in the ICI-chemo and non-ICI-chemo groups, respectively, without significant difference. Most patients in both groups received amrubicin as the second-line treatment. Moreover, the PFS and OS periods for third-line treatment were not significantly different between the ICI-chemo and non-ICI-chemo groups. CONCLUSIONS In ES-SCLC, there is no significant additive effect on PFS and OS of second- and subsequent line treatments following ICI-chemo at first-line treatment.
Collapse
Affiliation(s)
- Daisuke Morinaga
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, North 15, West 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Jun Sakakibara-Konishi
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, North 15, West 7, Kita-ku, Sapporo, 060-8638, Japan; Medical Network and Welfare Center, Hokkaido University Hospital, North 15, West 7, Kita-ku, Sapporo, 060-8638, Japan.
| | - Yasutaka Kawai
- Department of Respiratory Medicine, Oji General Hospital, 3-4-8, Wakakusa-cho, Tomakomai, 053-0021, Japan
| | - Yumi Morinaga
- Department of Respiratory Medicine, Obihiro-Kosei General Hospital, West 15, South 10, Obihiro, 080-0024, Japan
| | - Shohei Mizobuchi
- Department of Respiratory Medicine, NHO Hokkaido Medical Center, 5-7-1-1, Nishi-ku, Sapporo, 063-0005, Japan
| | - Yoshihiro Okamoto
- Department of Respiratory Medicine, NHO Hokkaido Medical Center, 5-7-1-1, Nishi-ku, Sapporo, 063-0005, Japan
| | - Yasunari Yamanaka
- Department of Respiratory Medicine, Iwamizawa Municipal General Hospital, 9jo, West 7, Iwamizawa, 068-8555, Japan
| | - Kei Takahashi
- Department of Respiratory Medicine, Iwamizawa Municipal General Hospital, 9jo, West 7, Iwamizawa, 068-8555, Japan
| | - Hajime Kikuchi
- Department of Respiratory Medicine, Obihiro-Kosei General Hospital, West 15, South 10, Obihiro, 080-0024, Japan
| | - Noriaki Sukoh
- Department of Respiratory Medicine, NHO Hokkaido Medical Center, 5-7-1-1, Nishi-ku, Sapporo, 063-0005, Japan
| | - Taichi Takashina
- Department of Respiratory Medicine, Iwamizawa Municipal General Hospital, 9jo, West 7, Iwamizawa, 068-8555, Japan
| | - Hidenori Kitai
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, North 15, West 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Satoshi Konno
- Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, North 15, West 7, Kita-ku, Sapporo, 060-8638, Japan
| |
Collapse
|
35
|
Tulsian K, Thakker D, Vyas VK. Overcoming chimeric antigen receptor-T (CAR-T) resistance with checkpoint inhibitors: Existing methods, challenges, clinical success, and future prospects : A comprehensive review. Int J Biol Macromol 2025; 306:141364. [PMID: 39988153 DOI: 10.1016/j.ijbiomac.2025.141364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/20/2024] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Immune checkpoint blockade is, as of today, the most successful form of cancer immunotherapy, with more than 43 % of cancer patients in the US eligible to receive it; however, only up to 12.5 % of patients respond to it. Similarly, adoptive cell therapy using bioengineered chimeric antigen receptorT (CAR-T) cells and T-cell receptor (TCR) cells has provided excellent responses against liquid tumours, but both forms of immunotherapy have encountered challenges within a tumour microenvironment that is both lacking in tumour-specific T-cells and is strongly immunosuppressive toward externally administered CAR-T and TCR cells. This review focuses on understanding approved checkpoint blockade and adoptive cell therapy at both biological and clinical levels before delving into how and why their combination holds significant promise in overcoming their individual shortcomings. The advent of next-generation checkpoint inhibitors has further strengthened the immune checkpoint field, and a special section explores how these inhibitors can address existing hurdles in combining checkpoint blockade with adoptive cell therapy and homing in on our cancer target for long-term immunity.
Collapse
Affiliation(s)
- Kartik Tulsian
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Dhinal Thakker
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India.
| |
Collapse
|
36
|
Al Maqrashi ZAA, Chan SWS, Siddiqui Z, Dotan E. Oncology: What You May Have Missed in 2024. Ann Intern Med 2025; 178:S89-S109. [PMID: 40163865 DOI: 10.7326/annals-25-00963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/02/2025] Open
Abstract
Over the past 5 decades, substantial advances in oncology have reshaped cancer care, reflecting the dynamic role of internal medicine physicians in patients' journey from screening to diagnosis, treatment, and surveillance. This review highlights 10 landmark studies from 2024 that address emerging therapies and evolving clinical standards. Immunotherapy remains a central focus, with checkpoint inhibitors redefining the management of solid tumors and showing expanded applications across disease sites and earlier stages of disease. Targeted therapies and antibody-drug conjugates, including trastuzumab deruxtecan and enfortumab vedotin, are enhancing precision treatment options in metastatic cancer. Meanwhile, advances in supportive care, such as magnetic resonance imaging-guided prostate cancer screening, ponsegromab for cachexia, and celiac plexus radiosurgery for pain, show enhanced symptom management and quality of life for patients. These innovations highlight the critical role of multidisciplinary approaches, where internal medicine physicians contribute to co-management and toxicity monitoring, ultimately optimizing patient care. By staying current with these developments, internal medicine physicians are positioned to navigate complex oncologic care, ensuring that the benefits of novel therapies are maximized while mitigating their challenges.
Collapse
Affiliation(s)
- Zainab Ali Amer Al Maqrashi
- Department of Oncology, McMaster University, and Juravinski Cancer Centre, Hamilton, Ontario, Canada (Z.A.A.A.M., S.W.S.C., Z.S.)
| | - Sze Wah Samuel Chan
- Department of Oncology, McMaster University, and Juravinski Cancer Centre, Hamilton, Ontario, Canada (Z.A.A.A.M., S.W.S.C., Z.S.)
| | - Zeba Siddiqui
- Department of Oncology, McMaster University, and Juravinski Cancer Centre, Hamilton, Ontario, Canada (Z.A.A.A.M., S.W.S.C., Z.S.)
| | - Efrat Dotan
- Penn Medicine, Ann B. Barshinger Cancer Institute, Lancaster General Health, Lancaster, Pennsylvania (E.D.)
| |
Collapse
|
37
|
Wang G, Yan W, Cai J, Zhang F, Lv T, Ye M. Durvalumab-induced organizing pneumonia in extensive-stage small cell lung cancer: A case report and literature review. Radiol Case Rep 2025; 20:2253-2257. [PMID: 40129823 PMCID: PMC11930415 DOI: 10.1016/j.radcr.2025.01.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/04/2025] [Accepted: 01/23/2025] [Indexed: 03/26/2025] Open
Abstract
The etoposide-carboplatin and anti-PD-L1 combination has become the standard-of-care for patients with extensive-stage small cell lung cancer (ES-SCLC). This combinational strategy is well tolerated with manageable immune-related adverse effects (irAEs). In this report, we presented a rare immediate irAE after one course of anti-tumor treatment. The patient with ES-SCLC was treated with first-line etoposide-carboplatin chemotherapy and Durvalumab immunotherapy. After one cycle of indicated treatment, the patient developed persistent high-grade fever with extensive consolidation, surrounding by ground glass opacifications. The lesions did not respond to empirical antibiotics and the results for pathogen testing were negative. Histological analysis of biopsy sample yielded organizing pneumonia that was very likely to associate with Durvalumab treatment. The patient was therefore treated with prednisolone that resulted in a rapid radiological improvement. The reporting of this case is imperative for informing acute onset of irAE in patients with ES-SCLC treated with anti-PD-L1 immunotherapy. Differential diagnosis of infection, tumor progression and exacerbation of underlying illness should be considered before the initiation of prednisolone therapy.
Collapse
Affiliation(s)
- Guoxin Wang
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wenjie Yan
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jun Cai
- Department of Radiology, Medical Imaging Center, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Fang Zhang
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tangfeng Lv
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Mingxiang Ye
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
38
|
Paz-Ares L, Gupta B, Baena J, Liu SV. Unmet Needs in Maintenance Therapy for Extensive Stage Small Cell Lung Cancer. Clin Lung Cancer 2025; 26:168-178. [PMID: 40155220 DOI: 10.1016/j.cllc.2025.02.015] [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: 03/26/2024] [Revised: 11/19/2024] [Accepted: 02/27/2025] [Indexed: 04/01/2025]
Abstract
Small cell lung cancer (SCLC) is a highly aggressive malignancy and an exceptionally lethal disease; most patients present with extensive stage (ES) disease at diagnosis. Very little had changed in the treatment of ES-SCLC for decades until immune checkpoint inhibitor (ICI) therapy combined with chemotherapy followed by ICI maintenance monotherapy was added to standard treatment paradigms in 2019. Despite this important advance, high rates of relapse are still observed in patients with ES-SCLC and long-term survival rates remain low, with approximately 40% of patients proceeding to receive second-line treatment. There is an urgent need for novel treatment strategies to improve patient outcomes. In this review, we describe the rationale for maintenance therapy approaches in ES-SCLC and summarize the existing data on chemotherapy, ICIs, and other agents in the first-line maintenance setting. Predictive biomarkers, SCLC subtypes, and new therapeutics in development are discussed including lurbinectedin, antibody-drug conjugates, and T-cell engager molecules.
Collapse
Affiliation(s)
- Luis Paz-Ares
- Hospital Universitario 12 de Octubre, H120H120-CNIO Lung Cancer Clinical Research Unit, Universidad Complutense & Ciberonc, Madrid, Spain
| | - Brinda Gupta
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Javier Baena
- Hospital Universitario 12 de Octubre, H120H120-CNIO Lung Cancer Clinical Research Unit, Universidad Complutense & Ciberonc, Madrid, Spain
| | - Stephen V Liu
- Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC.
| |
Collapse
|
39
|
Wang J, Chen Q, Shan Q, Liang T, Forde P, Zheng L. Clinical development of immuno-oncology therapeutics. Cancer Lett 2025; 617:217616. [PMID: 40054657 PMCID: PMC11930610 DOI: 10.1016/j.canlet.2025.217616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
Immuno-oncology (IO) is one of the fastest growing therapeutic areas within oncology. IO agents work indirectly via the host's adaptive and innate immune system to recognize and eradicate tumor cells. Despite checkpoint inhibitors being only introduced to the market since 2011, they have become the second most approved product category. Current Food and Drug Administration (FDA)-approved classes of IO agents include: immune checkpoint inhibitors (ICIs), chimeric antigen receptor T-cell therapy (CAR-T), bi-specific T-cell engager (BiTE) antibody therapy, T-cell receptor (TCR) engineered T cell therapy, tumor-infiltrating lymphocyte (TIL) therapy, cytokine therapy, cancer vaccine therapy, and oncolytic virus therapy. Cancer immunotherapy has made progress in multiple cancer types including melanoma, non-small cell lung cancer (NSCLC), renal cell carcinoma (RCC), and urothelial carcinoma; however, several cancers remain refractory to immunotherapy. Future directions of IO include exploration in the neoadjuvant/perioperative setting, combination strategies, and optimizing patient selection through improved biomarkers.
Collapse
Affiliation(s)
- Jianxin Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Qi Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Qiang Shan
- Department of General Surgery, Haining People's Hospital, Haining, 314400, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, 310003, China; The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province, Hangzhou, 310003, China
| | - Patrick Forde
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA; The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Mays Cancer Center at the University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Lei Zheng
- Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans St, Baltimore, MD, 21287, USA; The Pancreatic Cancer Precision Medicine Center of Excellence Program, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; The Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA; Mays Cancer Center at the University of Texas Health San Antonio, San Antonio, TX, 78229, USA.
| |
Collapse
|
40
|
Özgüroğlu M, Goldman JW, Chen Y, Garassino MC, Medic N, Mann H, Chugh P, Dalvi T, Paz-Ares L. Adverse events self-reported by patients with extensive-stage small-cell lung cancer in the phase III CASPIAN study. Future Oncol 2025; 21:1511-1523. [PMID: 40331625 PMCID: PMC12077470 DOI: 10.1080/14796694.2025.2491297] [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: 02/26/2024] [Accepted: 04/07/2025] [Indexed: 05/08/2025] Open
Abstract
AIM In the phase III CASPIAN study, first-line durvalumab plus platinum-etoposide (EP) improved survival compared with EP in patients with extensive-stage small-cell lung cancer. We report an exploratory analysis of patient-reported adverse events (AEs). METHODS Of 537 patients randomized to durvalumab + EP or EP arms, 164 were asked to complete the Patient-Reported Outcomes version of the Common Terminology Criteria (PRO-CTCAE) for AEs at baseline, every 3 weeks (q3w) during EP, then q4w until disease progression, then post-progression on day 28, 2 months, and q8w until second progression/death. Presence/absence, frequency, or severity of 11 selected AEs were examined during the first 24 weeks of treatment, alongside interference with usual/daily activities for five AEs. RESULTS AND CONCLUSIONS A minority of patients reported the examined AEs before starting treatment, from 3-5% who reported hand-foot syndrome, up to 34-41% for dry mouth. AE rates were generally comparable with baseline and the patterns of AEs reported by patients over time were similar in both treatment arms. Most patients indicated that reported AEs occurred rarely/occasionally and were mild/moderate in severity. These PRO-CTCAE data complement the clinician-reported AEs and give insight into patients' experience of treatment. CLINICAL TRIAL REGISTRATION www.clinicaltrials.gov identifier is NCT03043872.
Collapse
Affiliation(s)
- Mustafa Özgüroğlu
- Cerrahpaşa Faculty of Medicine, Istanbul University−Cerrahpaşa, Istanbul, Turkey
| | - Jonathan W. Goldman
- Hematology and Oncology Division, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yuanbin Chen
- Medical Oncology, Cancer & Hematology Centers of Western Michigan, Grand Rapids, MI, USA
| | | | | | - Helen Mann
- Oncology Biometrics, AstraZeneca, Cambridge, UK
| | - Priti Chugh
- Late Development Oncology, AstraZeneca, Waltham, MA, USA
| | - Tapashi Dalvi
- Late Development Oncology, AstraZeneca, Gaithersburg, MD, USA
| | - Luis Paz-Ares
- Hospital Universitario 12 de Octubre, H12O-CNIO Lung Cancer Unit, Universidad Complutense and Ciberonc, Madrid, Spain
| |
Collapse
|
41
|
Morinaga D, Hashimoto K, Asahina H, Tanaka H, Honjo O, Harada T, Yokouchi H, Kikuchi H, Shigaki R, Takashina T, Nakamura K, Kawai Y, Takahashi M, Kida R, Sukoh N, Ito K, Takahashi A, Hommura F, Ohhara Y, Furuta M, Konno S, Hosomi Y, Oizumi S. Prognostic impact of oligometastasis in older patients with extensive-stage small cell lung cancer. Respir Investig 2025; 63:373-382. [PMID: 40107221 DOI: 10.1016/j.resinv.2025.03.008] [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: 01/14/2025] [Revised: 03/02/2025] [Accepted: 03/09/2025] [Indexed: 03/22/2025]
Abstract
BACKGROUND Immune checkpoint inhibitor plus chemotherapy (ICT) is the standard treatment for extensive-stage small cell lung cancer (ES-SCLC). We previously reported that oligometastasis (OM) is a predictor of ICT efficacy, however, the relationship between ICT efficacy and OM in older patients remains unknown. Therefore, this study examined the efficacy of ICT in the older patients including the influence of OM. METHODS We enrolled patients with ES-SCLC who received ICT as first-line treatment between September 2019 and June 2022. Patient characteristics and treatment efficacy were compared between older (≥75 years) and non-older (<75 years) patients. RESULTS We enrolled 228 patients, including 42 older patients. The prevalence of synchronous oligometastasis (SOM) at the start of first-line treatment was 21.0 % and 21.4 % (p = 1.0) in the older and non-older groups, respectively. The progression-free survival (PFS) with first-line therapy was 5.4 and 4.5 months (p = 0.55) and overall survival (OS) was 11.5 and 12.6 months (p = 0.74) for the SOM and non-SOM subgroups in the older group, respectively. For second-line treatment, PFS was 4.5 and 6.3 months (p = 0.79), and OS after second-line initiation was 16.0 and 13.2 months (p = 0.55) in oligoprogression (OP) and non-OP patients in the older group, respectively. CONCLUSIONS The frequencies of SOM and OP were not significantly different between older and non-older patients. Although the small number of older patients in this study makes it impossible to conclude definitively, we did not observe a significant prognostic prolongation in older patients with OM as in non-older patients.
Collapse
Affiliation(s)
- Daisuke Morinaga
- Department of Respiratory Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, North 15, West 7, Kita-ku, Sapporo, 060-8638, Japan.
| | - Kana Hashimoto
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Hajime Asahina
- Department of Respiratory Medicine, NHO Hokkaido Cancer Center, 2-3-54 Kikusui 4, Shiroishi-ku, Sapporo, 003-0804, Japan.
| | - Hisashi Tanaka
- Department of Respiratory Medicine, Hirosaki University, Graduate School of Medicine, 5 Zaifucho, Hirosaki, 036-8562, Japan
| | - Osamu Honjo
- Department of Respiratory Medicine, Sapporo Minami-Sanjo Hospital, 4-2, South 6, West 3, Chuou-ku, Sapporo, 060-0063, Japan
| | - Toshiyuki Harada
- Department of Respiratory Medicine, JCHO Hokkaido Hospital, 1-8-3-18, Nakanoshima, Toyohira-ku, Sapporo, 062-0921, Japan
| | - Hiroshi Yokouchi
- Department of Respiratory Medicine, NHO Hokkaido Cancer Center, 2-3-54 Kikusui 4, Shiroishi-ku, Sapporo, 003-0804, Japan
| | - Hajime Kikuchi
- Department of Respiratory Medicine, Obihiro-Kosei General Hospital, West 15, South 10, Obihiro, 080-0024, Japan
| | - Ryota Shigaki
- Department of Internal Medicine, Division of Respiratory Medicine and Neurology, Asahikawa Medical University, 2-1-1-1, Midorigaoka-Highashi, Asahikawa, 078-8510, Japan
| | - Taichi Takashina
- Department of Respiratory Medicine, Iwamizawa Municipal General Hospital, 9jo, West 7, Iwamizawa, 068-8555, Japan
| | - Keiichi Nakamura
- Department of Respiratory Medicine, NHO Asahikawa Medical Center, 7-4048, Hanasaki-cho, Asahikawa, 070-8644, Japan
| | - Yasutaka Kawai
- Department of Respiratory Medicine, Oji General Hospital, 3-4-8, Wakakusa-cho, Tomakomai, 053-0021, Japan
| | - Mamoru Takahashi
- Department of Respiratory Medicine and Allergology, Sapporo Medical University School of Medicine, West 16, South 1, Chuou-ku, Sapporo, 060-8543, Japan
| | - Ryotaro Kida
- Department of Respiratory Medicine, Hokkaido Prefectural Kitami Hospital, 2-1, East 2, North 7, Kitami, 090-0027, Japan
| | - Noriaki Sukoh
- Department of Respiratory Medicine, NHO Hokkaido Medical Center, 5-7-1-1, Nishi-ku, Sapporo, 063-0005, Japan
| | - Kenichiro Ito
- Department of Respiratory Medicine, KKR Sapporo Medical Center, 1-6-3-40, Hiragishi, Toyohira-ku, Sapporo, 062-0931, Japan
| | - Ayumu Takahashi
- Department of Medicine, Hokkaido Chuo Rosai Hospital, Japan Organization of Occupational Health and Safety, 4-16-5, Iwamizawa, 068-0004, Japan
| | - Fumihiro Hommura
- Department of Respiratory Medicine, Sapporo City General Hospital, 1-1, West 13, North 11, Chuou-ku, Sapporo, 060-8604, Japan
| | - Yoshihito Ohhara
- Department of Medical Oncology, Hokkaido University Hospital, North 15, West 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Megumi Furuta
- Department of Respiratory Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, North 15, West 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Satoshi Konno
- Department of Respiratory Medicine, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, North 15, West 7, Kita-ku, Sapporo, 060-8638, Japan
| | - Yukio Hosomi
- Department of Thoracic Oncology and Respiratory Medicine, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, 3-18-22, Bunkyo-ku, Tokyo, 113-8677, Japan
| | - Satoshi Oizumi
- Department of Respiratory Medicine, NHO Hokkaido Cancer Center, 2-3-54 Kikusui 4, Shiroishi-ku, Sapporo, 003-0804, Japan
| |
Collapse
|
42
|
Li Y, Lin X, Peng Y, Tang J, Li X. Anlotinib as a Tailored Treatment for Extensive-Stage Small Cell Lung Cancer with Stable Disease after Two Cycles of First Chemotherapy Plus Immunotherapy: A Retrospective Study. Cancer Biother Radiopharm 2025; 40:277-284. [PMID: 40260508 DOI: 10.1089/cbr.2024.0220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025] Open
Abstract
Objective: Optimize the treatment of extensive-stage small cell lung cancer (ES-SCLC). Materials and Methods: We collected data on patients with ES-SCLC who had stable disease (SD) after two cycles of first-line chemotherapy combined with immunotherapy (CIO) and subsequently received tailored treatment with anlotinib. The primary endpoints of the study were progression-free survival and overall survival (OS), while secondary endpoints included overall response rate (ORR), disease control rate (DCR), and adverse events (AEs). Results: A total of 45 patients were ultimately enrolled in the study. Preliminary analysis indicated that the integration of anlotinib provides promising efficacy for patients with ES-SCLC who had SD after two cycles of CIO. ORR and DCR were 26.67% and 62.22%, respectively, with median progression-free survival and median OS were 6.0 months and 10.0 months. Furthermore, subgroup analysis results showed that patients who experienced hypertension, proteinuria, and hand-foot syndrome during treatment had better efficacy. In addition, mechanistic analysis suggested that this regimen may activate the immune system by depleting immune suppression, thereby exerting a synergistic antitumor effect. Beyond the promising efficacy, the overall AEs of this regimen were manageable, indicating a potential positive outlook for this treatment model in this patient population. Conclusion: The adaptive treatment with anlotinib can significantly improve outcomes for these patients, with manageable toxicities, suggesting that this treatment model has the potential to become an important option for first-line therapy in ES-SCLC. However, its true clinical value requires further research for validation.
Collapse
Affiliation(s)
- Yonghong Li
- Department of Oncology, The First People's Hospital of Tianmen, Tianmen, China
| | - Xi Lin
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Peng
- Department of Radiotherapy, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jing Tang
- Department of Lymphoma, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaobing Li
- Department of Thoracic Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
43
|
Liu M, Guan W, Xie X, Li Z, Qiu G, Lin X, Xie Z, Zhang J, Qin Y, Huang Z, Xu X, Zhou C. Phase I Clinical Trial of Autologous Hematopoietic Stem Cell Transplantation-Supported Dose-Intensified Chemotherapy With Adebrelimab as First-Line Treatment for Extensive-Stage Small Cell Lung Cancer. Clin Lung Cancer 2025; 26:e236-e241. [PMID: 39848827 DOI: 10.1016/j.cllc.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 12/26/2024] [Accepted: 12/26/2024] [Indexed: 01/25/2025]
Abstract
BACKGROUND Small cell lung cancer (SCLC) is initially highly sensitive to chemotherapy, which often leads to significant tumor reduction. However, the majority of patients eventually develop resistance, and the disease is further complicated by its "cold" tumor microenvironment, characterized by low tumor immunogenicity and limited CD8+ T cell infiltration. These factors contribute to the poor response to immunotherapy in many cases of extensive-stage SCLC (ES-SCLC). High-dose chemotherapy has shown potential in enhancing tumor cytoreduction, but its use is often limited by severe hematologic toxicity. Combining chemotherapy with immune checkpoint inhibitors (ICIs) can create a synergistic effect by promoting immunogenic cell death and enhancing immune activation. Autologous hematopoietic stem cell transplantation (auto-HSCT) provides a means to support hematopoietic recovery, mitigate chemotherapy-induced myelosuppression, and contribute to immune reconstitution. In this context, the integration of auto-HSCT with dose-intensified chemotherapy and ICIs aims to both protect the bone marrow and enhance antitumor immune responses, potentially overcoming resistance to immunotherapy in ES-SCLC. METHODS A phase I, single-center, single-arm trial was designed to evaluate the safety and efficacy of auto-HSCT-supported dose-intensified chemotherapy combined with adebrelimab in treatment-naive ES-SCLC patients. Participants will receive induction chemotherapy followed by stem cell mobilization, apheresis, and cryopreservation. After successful mobilization, consolidation chemotherapy with stem cell reinfusion and granulocyte colony-stimulating factor (G-CSF) support will be performed. Maintenance therapy with adebrelimab continues until disease progression or unacceptable toxicity. Safety and efficacy data, including adverse events, objective response rate (ORR), progression-free survival (PFS), and overall survival (OS), will be analyzed. RESULTS The study aims to enhance treatment outcomes by overcoming resistance to immuno-chemotherapy and promoting immune reconstitution. The trial is ongoing at the First Affiliated Hospital of Guangzhou Medical University. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT06597513.
Collapse
Affiliation(s)
- Ming Liu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Wenhui Guan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaohong Xie
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zekun Li
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Guihuan Qiu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xinqing Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhanhong Xie
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiexia Zhang
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yinyin Qin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhenqian Huang
- Department of Hematology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Xin Xu
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Organ Transplantation, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| | - Chengzhi Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
44
|
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
|
45
|
Fukushima T, Togasaki K, Hamamoto J, Emoto K, Ebisudani T, Mitsuishi A, Sugihara K, Shinozaki T, Okada M, Saito A, Takaoka H, Ito F, Shigematsu L, Ohta Y, Takahashi S, Matano M, Kurebayashi Y, Ohgino K, Sato T, Kawada I, Asakura K, Hishida T, Asamura H, Ikemura S, Terai H, Soejima K, Oda M, Fujii M, Fukunaga K, Yasuda H, Sato T. An organoid library unveils subtype-specific IGF-1 dependency via a YAP-AP1 axis in human small cell lung cancer. NATURE CANCER 2025:10.1038/s43018-025-00945-y. [PMID: 40307487 DOI: 10.1038/s43018-025-00945-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 03/07/2025] [Indexed: 05/02/2025]
Abstract
Small cell lung cancer (SCLC) is a devastating disease with limited therapeutic advancements. Although SCLC has recently been classified into four molecular subtypes, subtype-specific therapies are still lacking. Here, we established 40 patient-derived SCLC organoid lines with predominant TP53 and RB1 alterations and rare targetable genetic lesions. Transcriptome profiling divided the SCLC organoids into neuroendocrine (NE)-type SCLC and non-NE-type SCLC, with the latter characterized by YAP1 or POU2F3 expression. NE-type SCLC organoids grew independent of alveolar niche factors, whereas non-NE-type SCLC organoids relied on insulin-like growth factor (IGF)-1-driven YAP1 and AP1 activation. Therapeutic targeting of IGF-1, YAP1 and AP1 effectively suppressed the growth of non-NE-type organoids. Co-knockout of TP53 and RB1 in human alveolar cells altered their lineage toward the airway epithelium-like fate and conferred IGF-1 dependency, validating the subtype-phenotype connection. Our SCLC organoid library represents a valuable resource for developing biology-based therapies and has the potential to reshape the drug discovery landscape.
Collapse
Affiliation(s)
- Takahiro Fukushima
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Kazuhiro Togasaki
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Integrative Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Division of Gastroenterology Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Junko Hamamoto
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Katsura Emoto
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Toshiki Ebisudani
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Akifumi Mitsuishi
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Kai Sugihara
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Taro Shinozaki
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Masahiko Okada
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Ayaka Saito
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Hatsuyo Takaoka
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Fumimaro Ito
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Lisa Shigematsu
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Yuki Ohta
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Integrative Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Sirirat Takahashi
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Integrative Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Mami Matano
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Integrative Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Yutaka Kurebayashi
- Department of Pathology, Keio University School of Medicine, Tokyo, Japan
| | - Keiko Ohgino
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Takashi Sato
- Department of Respiratory Medicine, Kitasato University School of Medicine, Kanagawa, Japan
| | - Ichiro Kawada
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Keisuke Asakura
- Division of Thoracic Surgery, Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Tomoyuki Hishida
- Division of Thoracic Surgery, Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hisao Asamura
- Division of Thoracic Surgery, Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Shinnosuke Ikemura
- Department of Pulmonary Medicine, Faculty of Medicine University of Yamanashi, Yamanashi, Japan
| | - Hideki Terai
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Kenzo Soejima
- Department of Pulmonary Medicine, Faculty of Medicine University of Yamanashi, Yamanashi, Japan
| | - Mayumi Oda
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Integrative Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Masayuki Fujii
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Integrative Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan
| | - Hiroyuki Yasuda
- Division of Pulmonary Medicine, Department of Medicine, Keio University, School of Medicine, Tokyo, Japan.
| | - Toshiro Sato
- Department of Organoid Medicine, Keio University School of Medicine, Tokyo, Japan.
- Department of Integrative Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
46
|
Muscolino P, Omero F, Speranza D, Infurna C, Parisi S, Cianci V, Berretta M, Russo A, Santarpia M. ADCs and TCE in SCLC Therapy: The Beginning of a New Era? Curr Oncol 2025; 32:261. [PMID: 40422520 DOI: 10.3390/curroncol32050261] [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: 03/22/2025] [Revised: 04/27/2025] [Accepted: 04/28/2025] [Indexed: 05/28/2025] Open
Abstract
The therapeutic landscape for small cell lung cancer (SCLC) has remained stationary for decades, with chemotherapy representing the sole treatment strategy, with a modest survival benefit. The addition of immune checkpoint inhibitors (ICIs) to standard first-line chemotherapy for SCLC was a considerable milestone. However, despite high overall response rates, this strategy failed to deliver long-term benefits for most patients, who continue to face a poor prognosis. Over the last few years, a deeper knowledge of the molecular biology of SCLC and the impressive advancements in drug development, have led to the generation of novel classes of systemic therapies that promise to revolutionize the current therapeutic scenario. Among the various therapeutic approaches in development, T-cell Engagers (TCE) and antibody-drug conjugates (ADCs) stand out due to their unique structural characteristics and mechanisms of action. These therapies represent a paradigm shift from traditional monoclonal antibody (mAb) and chemotherapy regimens, allowing direct engagement of multiple targets associated with tumor progression. In this review, we provide an overview of current drug development in SCLC, specifically focusing on these new agents, summarizing available evidence, and tracking future directions.
Collapse
Affiliation(s)
- Paola Muscolino
- School of Specialization in Medical Oncology, Department of Human Pathology "G. Barresi", University of Messina, 98125 Messina, Italy
| | - Fausto Omero
- School of Specialization in Medical Oncology, Department of Human Pathology "G. Barresi", University of Messina, 98125 Messina, Italy
| | - Desirèe Speranza
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, 98125 Messina, Italy
| | - Carla Infurna
- School of Specialization in Medical Oncology, Department of Human Pathology "G. Barresi", University of Messina, 98125 Messina, Italy
| | - Silvana Parisi
- Radiation Oncology Unit, Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98125 Messina, Italy
| | - Vincenzo Cianci
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy
| | - Massimiliano Berretta
- Division of Medical Oncology, AOU "G. Martino" Hospital, University of Messina, 98124 Messina, Italy
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Alessandro Russo
- Department of Medical Oncology, Humanitas Istituto Clinico Catanese, 95045 Misterbianco, Italy
- Department of Biomedical Sciences, Humanitas University, 20072 Pieve Emanuele, Italy
| | - Mariacarmela Santarpia
- Division of Medical Oncology, AOU "G. Martino" Hospital, University of Messina, 98124 Messina, Italy
- Department of Human Pathology "G. Barresi", University of Messina, 98125 Messina, Italy
| |
Collapse
|
47
|
Azmal M, Miah MM, Prima FS, Paul JK, Haque ASNB, Ghosh A. Advances and challenges in cancer immunotherapy: Strategies for personalized treatment. Semin Oncol 2025; 52:152345. [PMID: 40305928 DOI: 10.1016/j.seminoncol.2025.152345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 05/02/2025]
Abstract
Cancer immunotherapy has transformed oncology by harnessing the immune system to specifically target cancer cells, offering reduced systemic toxicity compared to traditional therapies. This review highlights key strategies, including adoptive cell transfer (ACT), immune checkpoint inhibitors, oncolytic viral (OV) therapy, monoclonal antibodies (mAbs), and mRNA-based vaccines. ACT reinfuses enhanced immune cells like tumor-infiltrating lymphocytes (TILs) to combat refractory cancers, while checkpoint inhibitors (eg, PD-1 and CTLA-4 blockers) restore T-cell activity. OV therapy uses engineered viruses (eg, T-VEC) to selectively lyse cancer cells, and advanced mAbs improve targeting precision. mRNA vaccines introduce tumor-specific antigens to trigger robust immune responses. Despite significant progress, challenges like immune-related side effects, high costs, and immunosuppressive tumor microenvironments persist. This review underscores the need for combination strategies and precision medicine to overcome these barriers and maximize the potential of immunotherapy in personalized cancer treatment.
Collapse
Affiliation(s)
- Mahir Azmal
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Md Munna Miah
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Fatema Sultana Prima
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Jibon Kumar Paul
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Anm Shah Newaz Been Haque
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh
| | - Ajit Ghosh
- Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet, Bangladesh.
| |
Collapse
|
48
|
Barba A, López-Vilaró L, Ferre M, Martinez-Recio S, Majem M, Sullivan I, Salazar J. CD274 ( PD-L1) Polymorphisms as Predictors of Efficacy in First-Line Platinum-Based Chemotherapy for Extensive-Stage Small Cell Lung Cancer. Int J Mol Sci 2025; 26:4245. [PMID: 40362483 PMCID: PMC12072405 DOI: 10.3390/ijms26094245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 04/22/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
The cornerstone of first-line treatment in extensive-stage small cell lung cancer (ES-SCLC) is platinum- and etoposide-based chemotherapy. Platinum compounds could immunomodulate the tumor microenvironment in addition to their cytotoxic effect. Genetic variation in immune checkpoint (IC) pathways may predict chemotherapy efficacy. Polymorphisms in the IC genes were determined, and their association with survival was analyzed in 78 patients with ES-SCLC treated with chemotherapy. PD-L1 protein expression in tumor tissue was determined. Three variants in CD274 were associated with better median progression-free survival (mPFS): rs2297136 (hazard ratio [HR] 0.52, 95% CI 0.29-0.93; p = 0.03), rs2282055 (HR 0.23, 95% CI 0.09-0.64; p = 0.005), and rs822336 (HR 0.41, 95% CI 0.23-0.73; p = 0.002). CTLA4 rs231775 was also associated with mPFS (HR 0.30, 95% CI 0.14-0.63; p = 0.002). The variants CD274 rs2297136 and CD274 rs822336 were associated with platinum sensitivity (odds ratio [OR] 0.13, 95% CI 0.02-0.70; p = 0.02, and OR 0.08, 95% CI 0.01-0.46; p = 0.005, respectively). CD274 rs2297136 was also associated with better overall survival (p = 0.02), but not after adjustment for covariates. No association was found between CD274 germline variants and PD-L1 tumor expression. Our results suggest that CD274 and CTLA4 variants may be predictive biomarkers for platinum plus etoposide treatment in ES-SCLC.
Collapse
Affiliation(s)
- Andrés Barba
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (A.B.); (S.M.-R.); (M.M.); (I.S.)
- Department of Medicine, Faculty of Medicine, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
- Translational Medical Oncology Laboratory, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain
| | - Laura López-Vilaró
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (L.L.-V.); (M.F.)
| | - Malena Ferre
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (L.L.-V.); (M.F.)
| | - Sergio Martinez-Recio
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (A.B.); (S.M.-R.); (M.M.); (I.S.)
- Translational Medical Oncology Laboratory, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain
| | - Margarita Majem
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (A.B.); (S.M.-R.); (M.M.); (I.S.)
- Translational Medical Oncology Laboratory, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain
| | - Ivana Sullivan
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (A.B.); (S.M.-R.); (M.M.); (I.S.)
- Translational Medical Oncology Laboratory, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain
| | - Juliana Salazar
- Translational Medical Oncology Laboratory, Institut de Recerca Sant Pau (IR Sant Pau), 08041 Barcelona, Spain
| |
Collapse
|
49
|
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
|
50
|
Brumage L, Best S, Hippe DS, Grunblatt E, Chanana P, Wu F, Lee MC, Ying Z, Ibrahim A, Chung JH, Vigil A, Fatherree J, Beronja S, Paddison P, Sullivan L, Nabet B, MacPherson D. In vivo functional screens reveal KEAP1 loss as a driver of chemoresistance in small cell lung cancer. SCIENCE ADVANCES 2025; 11:eadq7084. [PMID: 40267200 PMCID: PMC12017300 DOI: 10.1126/sciadv.adq7084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 03/18/2025] [Indexed: 04/25/2025]
Abstract
Exquisitely chemosensitive initially, small cell lung cancer (SCLC) exhibits dismal outcomes owing to rapid transition to chemoresistance. Elucidating the genetic underpinnings has been challenging owing to limitations with cellular models. As SCLC patient-derived xenograft (PDX) models mimic therapeutic responses, we perform genetic screens in chemosensitive PDX models to identify drivers of chemoresistance. cDNA overexpression screens identify MYC, MYCN, and MYCL, while CRISPR deletion screens identify KEAP1 loss as driving chemoresistance. Deletion of KEAP1 switched a chemosensitive SCLC PDX model to become chemoresistant and resulted in sensitivity to inhibition of glutamine metabolism. Data from the IMpower133 clinical trial revealed ~6% of patients with extensive-stage SCLC exhibit KEAP1 genetic alterations, with activation of a KEAP1/NRF2 transcriptional signature associated with reduced survival upon chemotherapy treatment. While roles for KEAP1/NRF2 have been unappreciated in SCLC, our genetic screens revealed KEAP1 loss as a driver of chemoresistance, while patient genomic analyses demonstrate clinical importance.
Collapse
Affiliation(s)
- Lauren Brumage
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington Seattle, Seattle, WA, USA
| | - Scott Best
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Molecular and Cellular Biology Program, University of Washington Seattle, Seattle, WA, USA
| | - Daniel S. Hippe
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Eli Grunblatt
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Pritha Chanana
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Feinan Wu
- Genomics and Bioinformatics Shared Resource, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Zhe Ying
- Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ali Ibrahim
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jae Heun Chung
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Anna Vigil
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Jackson Fatherree
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Slobodan Beronja
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Patrick Paddison
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Lucas Sullivan
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - David MacPherson
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| |
Collapse
|