51
|
Fan L, Lin Y, Fu Y, Wang J. Small cell lung cancer with liver metastases: from underlying mechanisms to treatment strategies. Cancer Metastasis Rev 2024; 44:5. [PMID: 39585433 DOI: 10.1007/s10555-024-10220-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/06/2024] [Indexed: 11/26/2024]
Abstract
Small cell lung cancer (SCLC) represents an aggressive neuroendocrine (NE) tumor within the pulmonary region, characterized by very poor prognoses. Druggable targets for SCLC remain limited, thereby constraining treatment options available to patients. Immuno-chemotherapy has emerged as a pivotal therapeutic strategy for extensive-stage SCLC (ES-SCLC), yet it fails to confer significant efficacy in cases involving liver metastases (LMs) originating from SCLC. Therefore, our attention is directed towards the challenging subset of SCLC patients with LMs. Disease progression of LM-SCLC patients is affected by various factors in the tumor microenvironment (TME), including immune cells, blood vessels, inflammatory mediators, metabolites, and NE substances. Beyond standard immuno-chemotherapy, ongoing efforts to manage LMs in SCLC encompass anti-angiogenic therapy, radiotherapy, microwave ablation (MWA) / radiofrequency ablation (RFA), trans-arterial chemoembolization (TACE), and systemic therapies in conjunction with local interventions. Prospective experimental and clinical investigations into SCLC should prioritize precise and individualized approaches to enhance the prognosis across distinct patient cohorts.
Collapse
Affiliation(s)
- Linjie Fan
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yiwen Lin
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yunjie Fu
- School of Basic Medical Sciences, Peking University, Beijing, China
| | - Jie Wang
- CAMS Key Laboratory of Translational Research on Lung Cancer, State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
52
|
Morgensztern D, Ready N, Johnson ML, Dowlati A, Choudhury N, Carbone DP, Schaefer E, Arnold SM, Puri S, Piotrowska Z, Hegde A, Chiang AC, Iams W, Tolcher A, Nosaki K, Kozuki T, Li T, Santana-Davila R, Akamatsu H, Murakami H, Yokouchi H, Wang S, Zha J, Li R, Robinson RR, Hingorani P, Jeng EE, Furqan M. A Phase I First-in-Human Study of ABBV-011, a Seizure-Related Homolog Protein 6-Targeting Antibody-Drug Conjugate, in Patients with Small Cell Lung Cancer. Clin Cancer Res 2024; 30:5042-5052. [PMID: 39287821 PMCID: PMC11565168 DOI: 10.1158/1078-0432.ccr-24-1547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/17/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
PURPOSE Seizure-related homolog protein 6 (SEZ6) is a novel target expressed in small cell lung cancer (SCLC). ABBV-011, a SEZ6-targeted antibody conjugated to calicheamicin, was evaluated in a phase I study (NCT03639194) in patients with relapsed/refractory SCLC. We report initial outcomes of ABBV-011 monotherapy. PATIENTS AND METHODS ABBV-011 was administered intravenously once every 3 weeks during dose escalation (0.3-2 mg/kg) and expansion. Patients with SEZ6-positive tumors (≥25% of tumor cells with ≥1+ staining intensity by IHC) were preselected for expansion. Safety, tolerability, antitumor activity, and pharmacokinetics were evaluated. RESULTS As of August 2022, 99 patients received ABBV-011 monotherapy [dose escalation, n = 36; Japanese dose evaluation, n = 3; dose expansion, n = 60 (1 mg/kg, n = 40)]; the median age was 63 years (range, 41-79 years). Also, 32%, 41%, and 26% of patients received 1, 2, and ≥3 prior therapies, respectively. The maximum tolerated dose was not reached through 2.0 mg/kg. The most common treatment-emergent adverse events were fatigue (50%), nausea (42%), and thrombocytopenia (41%). The most common hepatic treatment-emergent adverse events were increased aspartate aminotransferase (22%), increased γ-glutamyltransferase (21%), and hyperbilirubinemia (17%); two patients experienced veno-occlusive liver disease. The objective response rate was 19% (19/98). In the 1-mg/kg dose-expansion cohort (n = 40), the objective response rate was 25%; the median response duration was 4.2 months (95% confidence interval, 2.6-6.7); and the median progression-free survival was 3.5 months (95% confidence interval, 1.5-4.2). CONCLUSIONS ABBV-011 1.0 mg/kg every 3 weeks monotherapy was well tolerated and demonstrated encouraging antitumor activity in heavily pretreated patients with relapsed/refractory SCLC. SEZ6 is a promising novel SCLC target and warrants further investigation.
Collapse
Affiliation(s)
| | - Neal Ready
- Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Melissa L. Johnson
- Sarah Cannon Research Institute, Tennessee Oncology, PLLC, Nashville, Tennessee
| | - Afshin Dowlati
- University Hospitals Seidman Cancer Center, Cleveland, Ohio
- Case Western Reserve University, Cleveland, Ohio
| | | | - David P. Carbone
- The Ohio State University James Cancer Center and the Pelotonia Institute for Immuno-Oncology, Columbus, Ohio
| | | | | | - Sonam Puri
- Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah
| | - Zofia Piotrowska
- Massachusetts General Hospital Cancer Center/Harvard Medical School, Boston, Massachusetts
| | - Aparna Hegde
- University of Alabama at Birmingham School of Medicine, Birmingham, Alabama
| | | | - Wade Iams
- Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Kaname Nosaki
- National Cancer Center Hospital East, Kashiwa, Japan
| | - Toshiyuki Kozuki
- National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Tianhong Li
- University of California, Davis Comprehensive Cancer Center, Sacramento, California
| | | | | | | | - Hiroshi Yokouchi
- National Hospital Organization Hokkaido Cancer Center, Sapporo, Japan
| | - Song Wang
- AbbVie, Inc., North Chicago, Illinois
| | | | - Rui Li
- AbbVie, Inc., North Chicago, Illinois
| | | | | | | | - Muhammad Furqan
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa
| |
Collapse
|
53
|
Li S, Yuan T, Yuan J, Zhu B, Chen D. Opportunities and challenges of using circulating tumor DNA to predict lung cancer immunotherapy efficacy. J Cancer Res Clin Oncol 2024; 150:501. [PMID: 39545998 PMCID: PMC11568038 DOI: 10.1007/s00432-024-06030-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Immune checkpoint inhibitors (ICIs), particularly anti-programmed death 1 (PD-1)/ programmed death ligand 1 (PD-L1) antibodies, have led to significant progress in lung cancer treatment. However, only a minority of patients have responses to these therapies. Detecting peripheral blood of circulating tumor DNA (ctDNA) allows minimally invasive diagnosis, characterization, and monitoring of lung cancer. ctDNA has potential to be a prognostic biomarker and a predictor of the response to ICI therapy since it can indicate the genomic status and tumor burden. Recent studies on lung cancer have shown that pretreatment ctDNA analysis can detect residual proliferative disease in the adjuvant immunotherapy setting and evaluate tumor burden in patients with metastatic disease. Early ctDNA dynamics can not only predict the clinical outcome of ICI therapy but also help distinguish between pseudoprogression and real progression. Furthermore, in addition to quantitative assessment, ctDNA can also detect genetic predictors of response to ICI therapy. However, barriers still exist in the application of ctDNA analysis in clinical lung cancer treatment. The predictive value of ctDNA in lung cancer immunotherapy requires further identification and resolution of these challenges. This review aims to summarize the existing data of ctDNA analysis in patients receiving immunotherapy for lung cancer, understand the limitations of clinical treatment, and discuss future research directions.
Collapse
Affiliation(s)
- Shanshan Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Ting Yuan
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Jing Yuan
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Degao Chen
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
54
|
Jiang Y, Xie J, Cheng Q, Cai Z, Xu K, Lu W, Wang F, Wu X, Song Y, Lv T, Zhan P. Comprehensive genomic and spatial immune infiltration analysis of survival outliers in extensive-stage small cell lung cancer receiving first-line chemoimmunotherapy. Int Immunopharmacol 2024; 141:112901. [PMID: 39151386 DOI: 10.1016/j.intimp.2024.112901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/20/2024] [Accepted: 08/05/2024] [Indexed: 08/19/2024]
Abstract
BACKGROUND A minority of patients with extensive-stage small cell lung cancer (ES-SCLC) exhibit prolonged survival following first-line chemoimmunotherapy, which warrants the use of reliable biomarkers. Here, we investigated the disparities in genomics and immune cell spatial distribution between long- and short-term survival of patients with ES-SCLC. METHODS We retrospectively recruited 11 long-term (>2 years) and 13 short-term (<9 months) ES-SCLC survivors receiving first-line chemoimmunotherapy. The samples were processed using targeted next-generation sequencing (tNGS), programmed death ligand-1 staining, multiplex immunohistochemical staining for immune cells (mIHC), tumor mutation burden (TMB), and chromosomal instability score measurements. The expression of putative genes in SCLC at the bulk and single-cell RNA-sequencing levels, as well as the role of putative genes in pan-cancer immunotherapy cohorts, were analyzed. RESULTS At the genomic level, a greater proportion of the smoking signature and higher TMB (>3.1) were associated with favorable survival. At the single-gene and pathway levels, tNGS revealed that MCL1 and STMN1 amplification and alterations in the apoptosis pathway were more common in short-term survivors, whereas alterations in the DLL3, KMT2B, HGF, EPHA3, ADGRB3, lysine deprivation, and HGF-cMET pathways were observed more frequently in long-term survivors. mIHC analysis of immune cells with different spatial distributions revealed that long-term survivors presented increased numbers of M1-like macrophages in all locations and decreased numbers of CD8+ T cells in the tumor stroma. Bulk transcriptomic analysis demonstrated that high levels of STMN1 and DLL3 represented an immune-suppressive tumor immune microenvironment (TIME), whereas HGF indicated an immune-responsive TIME. The expression levels of our putative genes were comparative in both TP53/RB1 mutant-type and TP53/RB1 wild-type. At the single-cell level, STMN1, MCL1, and DLL3 were highly expressed among all molecular subtypes (SCLC-A, SCLC-N, and SCLC-P), with STMN1 being enriched in cell division and G2M checkpoint pathways. CONCLUSIONS For ES-SCLC patients receiving first-line chemoimmunotherapy, alterations in DLL3, KMT2B, HGF, EPHA3, and ADGRB3 and a greater proportion of M1-like macrophages infiltration in all locations were predictors of favorable survival, while MCL1 and STMN1 amplification, as well as a greater proportion of CD8+ T cells infiltrating the tumor stroma, predicted worse survival.
Collapse
Affiliation(s)
- Yuxin Jiang
- School of Medicine, Southeast University, Nanjing 210000, China
| | - Jingyuan Xie
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Qinpei Cheng
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Zijing Cai
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China
| | - Ke Xu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Wanjun Lu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China
| | - Fufeng Wang
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Xiaoying Wu
- Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Yong Song
- School of Medicine, Southeast University, Nanjing 210000, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Southeast University, Nanjing 210002, China.
| | - Tangfeng Lv
- School of Medicine, Southeast University, Nanjing 210000, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Southeast University, Nanjing 210002, China.
| | - Ping Zhan
- School of Medicine, Southeast University, Nanjing 210000, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Nanjing Medical School, Nanjing 210002, China; Department of Respiratory and Critical Care Medicine, Jinling Hospital, School of Medicine, Southeast University, Nanjing 210002, China.
| |
Collapse
|
55
|
Ren Z, Shang S, Chen D. Recent advances in immunotherapy for small cell lung cancer. Curr Opin Oncol 2024:00001622-990000000-00220. [PMID: 39526685 DOI: 10.1097/cco.0000000000001105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
PURPOSE OF REVIEW This review aims to provide an overview of recent advances in immunotherapy for small cell lung cancer (SCLC), with a focus on the current status of immune checkpoint inhibitors (ICIs), novel combination strategies, and key biomarkers. RECENT FINDINGS The integration of ICIs into standard chemotherapy has established them as the first-line treatment for extensive-stage SCLC (ES-SCLC). The ADRIATIC trial further demonstrated the efficacy of ICI maintenance therapy in limited-stage SCLC. Additionally, combining radiotherapy with ICIs has shown promising synergistic effects, including the abscopal and radscopal effects. Ongoing investigations into the combination of ICIs with targeted therapies, such as antiangiogenic agents and DNA damage response inhibitors, have yielded encouraging preliminary results. Notably, the novel therapeutic agent tarlatamab, the first bispecific DLL3-directed CD3 T-cell engager, has recently received FDA approval for second-line treatment of ES-SCLC. Advances in omics technologies have shed light on the intra-tumor and inter-tumor heterogeneity of SCLC, leading to the identification of new molecular subtypes and biomarkers, thereby paving the way for precision medicine. SUMMARY Despite the improved outcomes associated with immunotherapy in SCLC, the overall clinical benefit remains modest. Further preclinical and clinical studies are essential to identify optimal treatment regimens and enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Ziyuan Ren
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | | | | |
Collapse
|
56
|
Tanaka A, Teranishi S, Kajita Y, Hirose T, Kaneko A, Sairenji Y, Kawashima H, Yumoto K, Tsukahara T, Miura K, Kobayashi N, Yamamoto M, Nishihira R, Kudo M, Miyazawa N, Nishikawa M, Kaneko T. Total baseline tumor size predicts survival among patients with advanced small-cell lung cancer receiving chemotherapy plus programmed death-ligand 1 inhibitor as first-line therapy: a multicenter retrospective observational study. Front Oncol 2024; 14:1400277. [PMID: 39558947 PMCID: PMC11570404 DOI: 10.3389/fonc.2024.1400277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 10/18/2024] [Indexed: 11/20/2024] Open
Abstract
Introduction Total baseline tumor size (BTS) is a prognostic factor for programmed death 1 and programmed death-ligand 1 (PD-L1) inhibitor treatments. However, the prognostic value of total BTS for patients with small-cell lung cancer (SCLC) who receive chemotherapy plus PD-L1 inhibitor remains unknown. Thus, in this study, we aimed to determine whether total BTS is associated with prognosis in patients with SCLC who receive chemotherapy plus PD-L1 inhibitor as first-line therapy. Methods This study included patients with extensive-stage SCLC or post-chemoradiotherapy recurrence of limited-stage SCLC who received chemotherapy plus PD-L1 inhibitor as first-line therapy from August 2019 to December 2022. The two lesions with the largest diameter among the measurable lesions in each organ were selected from up to five organs (maximum of 10 lesions), and the sum of all diameters was defined as total BTS. The patients were divided into two groups, large or small, with total BTS using X-tile software. Median survival was analyzed using the Kaplan-Meier method, and the groups were compared using the log-rank test. Univariate and multivariate analyses examined the association between total BTS and prognosis. Results Fifty patients were included; 14% had large total BTS (>183.2 mm) and 86% had small total BTS (≤183.2 mm). The median observation period was 10.5 months. The large total BTS group showed significantly worse overall survival than the small total BTS group (median: 26.8 months vs. 5.7 months, P = 0.0003). The multivariate analysis indicated that large total BTS was an independent negative predictor of overall survival (hazard ratio: 7.14, 95% confidence interval: 1.89-26.96). Discussion Total BTS is a potentially useful prognostic factor for patients with advanced SCLC who receive chemotherapy plus PD-L1 inhibitor as first-line therapy.
Collapse
Affiliation(s)
- Anna Tanaka
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Shuhei Teranishi
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Yukihito Kajita
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Tomofumi Hirose
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Ayami Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yu Sairenji
- Department of Respiratory Medicine, Yokohama Sakae Kyosai Hospital, Yokohama, Japan
| | | | - Kentaro Yumoto
- Department of Respiratory Medicine, Yokohama Minami Kyosai Hospital, Yokohama, Japan
| | - Toshinori Tsukahara
- Department of Respiratory Medicine, Chigasaki Municipal Hospital, Chigasaki, Japan
| | - Kenji Miura
- Department of Respiratory Medicine, Yokohama Sakae Kyosai Hospital, Yokohama, Japan
| | - Nobuaki Kobayashi
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Masaki Yamamoto
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Ryuichi Nishihira
- Department of Respiratory Medicine, Kanto Rosai Hospital, Kawasaki, Japan
| | - Makoto Kudo
- Respiratory Disease Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Naoki Miyazawa
- Department of Respiratory Medicine, Yokohama Nanbu Hospital, Yokohama, Japan
| | - Masanori Nishikawa
- Department of Respiratory Medicine, Fujisawa Municipal Hospital, Fujisawa, Japan
| | - Takeshi Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
57
|
Moro-Sibilot D, Falchero L, Ardin C, Zouak A, Molinier O, Romand P, Leleu O, Amrane K, Berndt C, Langlais A, Morin F, Westeel V. Study design and rationale for IFCT- 2203 TAXIO: A study that aims to evaluate the effectiveness of a first-line chemotherapy regimen without etoposide, combined with durvalumab, for patients with extensive disease small cell lung cancer. Respir Med Res 2024; 86:101113. [PMID: 38843598 DOI: 10.1016/j.resmer.2024.101113] [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: 03/28/2024] [Accepted: 04/25/2024] [Indexed: 12/18/2024]
Abstract
BACKGROUND Studies have shown improvement in overall survival with anti-PD1/PD-L1 molecules in combination with cisplatin/carboplatin and etoposide as a first-line treatment for Small Cell Lung Cancer (SCLC). However, first-line efficacy remains limited and well below that observed in Non-Small Cell Lung Cancer (NSCLC). Etoposide may have a detrimental effect on lymphocyte activation, which could explain the limited benefit of immunotherapy in the first line and the lack of benefit in the second line for patients previously exposed to high levels of etoposide. METHODS We initiated a multicenter, single-arm, open-label phase II study of a chemotherapy regimen with durvalumab, combined with carboplatin and paclitaxel for extensive disease SCLC. Eligible patients will receive durvalumab plus carboplatin and paclitaxel every 3 weeks for up to 4 cycles, followed by durvalumab every 4 weeks until progression or unacceptable toxicity. A total of 67 patients will be enrolled in this study, with a 12-month enrollment period and 36-month follow-up. The primary endpoint is Overall Survival (OS) rate at 12 months. Secondary endpoints are best response rate, OS, OS at 24- and 36 months, progression free survival (PFS), duration of response, quality of life and safety. RESULTS This study aims to establish the efficacy of durvalumab combined with carboplatin and paclitaxel in patients with extensive disease Small Cell Lung Cancer. CLINICAL TRIAL REGISTRATION EU CT: 2023-504670-38-00.
Collapse
Affiliation(s)
- Denis Moro-Sibilot
- Thoracic Oncology Unit, CS10217 cedex CHU-Grenoble-Alpes, 38043 Grenoble, France.
| | - Lionel Falchero
- Pneumology Department and Thoracic Oncology, Hôpital Nord-Ouest, Plateau d'Ouilly - BP 80436, 69655 Villefranche-sur-Saône, France
| | - Camille Ardin
- Thoracic Oncology Unit, CS10217 cedex CHU-Grenoble-Alpes, 38043 Grenoble, France
| | - Ayoube Zouak
- Department of Respiratory Diseases, CHU Dijon Bourgogne,1 Boulevard Jeanne D'Arc BP77908, 21079 Dijon, Cedex, France
| | - Olivier Molinier
- Respiratory Medicine Department, CH Le Mans 194 avenue Rubillard 72037 Le MANS Cedex 9, France
| | - Philippe Romand
- Pneumology Department, Centre Hospitalier Alpes-Leman, 558 route de Findrol,74130 Contamine sur Arve, France
| | - Olivier Leleu
- Pneumology Department, Centre Hospitalier d'Abbeville 43 rue de l'ISLE, 80 142 Abbeville, Cedex, France
| | - Karim Amrane
- Oncology Department, Centre Hospitalier des Pays de Morlaix, 15 rue de Kersaint-Gilly, BP 97327, 29672 Morlaix, Cedex, France
| | - Célia Berndt
- Clinical Research Unit, Intergroupe Francophone de Cancérologie Thoracique, 10 rue de la grange batelière, 75009 Paris, France
| | - Alexandra Langlais
- Clinical Research Unit, Intergroupe Francophone de Cancérologie Thoracique, 10 rue de la grange batelière, 75009 Paris, France
| | - Franck Morin
- Clinical Research Unit, Intergroupe Francophone de Cancérologie Thoracique, 10 rue de la grange batelière, 75009 Paris, France
| | - Virginie Westeel
- Clinical Research Unit, Intergroupe Francophone de Cancérologie Thoracique, 10 rue de la grange batelière, 75009 Paris, France; Pneumology Department, CHU Besançon, 3 boulevard A.Fleming, 25030 Besançon, Cedex, France
| |
Collapse
|
58
|
Bruni A, Scotti V, Zerella MA, Bertolini F, Imbrescia J, Olmetto E, Bennati C, Cuccia F, Miele M, Giaj-Levra N, Tiseo M, Ciammella P, Vagge S, Galaverni M, Pontoriero A, Badellino S, Spoto R, Alì E, Borghetti P. Current Radiotherapy Management of Extensive-Stage Small-Cell Lung Cancer in the Immunotherapy Era: An Italian National Survey on Behalf of the Italian Association of Radiotherapy and Clinical Oncology (AIRO). Curr Oncol 2024; 31:6791-6802. [PMID: 39590132 PMCID: PMC11592792 DOI: 10.3390/curroncol31110501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/29/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Extensive-stage small-cell lung cancer (ES-SCLC) treatment has recently been revolutionized by the advent of immune checkpoint inhibitors. This survey was conducted to evaluate the current pattern of care among Italian clinicians, in particular about the integration with radiation therapy (RT). METHODS In June 2023, 225 Italian cancer care professionals were invited to complete a 21-question web-based survey about ES-SCLC management through personal contacts and the Italian Association for Radiotherapy and Clinical Oncology (AIRO) network. RESULTS We received 90 responses; the majority were radiation oncologists (89%) with more than 10 years of experience (51%). The preferred management of ES-SCLC in patients with a good performance status was concomitant chemo-immunotherapy (84%). Almost all respondents recommended prophylactic cranial irradiation (PCI) (85%), taking into account age and thoracic response; PCI was performed mainly between the end of chemotherapy and before starting immunotherapy (37%), with a three-dimensional conformal technique (46%). Furthermore, 83% of respondents choose to deliver thoracic RT in the case of both an intrathoracic and extrathoracic response, with an RT schedule of 30 Gy/10 fractions. Stereotactic RT is increasingly being used in oligoprogressions. CONCLUSIONS Our analysis showed the variability of real-world management of ES-SCLC. Future clinical trials and developments are needed to improve the multidisciplinary treatment of these patients.
Collapse
Affiliation(s)
- Alessio Bruni
- Radiation Oncology Unit, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (A.B.)
| | - Vieri Scotti
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, 50134 Firenze, Italy
| | - Maria Alessia Zerella
- Division of Radiotherapy, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, 20141 Milano, Italy
| | - Federica Bertolini
- Division of Oncology, Department of Oncology and Hematology, Modena University Hospital, 41124 Modena, Italy
| | - Jessica Imbrescia
- Radiation Oncology Unit, Department of Oncology and Hematology, University Hospital of Modena, 41124 Modena, Italy; (A.B.)
| | - Emanuela Olmetto
- Radiation Oncology Unit, Oncology Department, Azienda Ospedaliero Universitaria Careggi, 50134 Firenze, Italy
| | - Chiara Bennati
- Department of Onco-Hematology, AUSL della Romagna, 48121 Ravenna, Italy
| | | | - Marianna Miele
- Operative Research Unit of Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
| | - Niccolò Giaj-Levra
- Advanced Radiation Oncology Department, Cancer Care Center, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Negrar, Italy
| | - Marcello Tiseo
- Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
- Medical Oncology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Patrizia Ciammella
- Radiation Oncology Unit, Azienda USL-Istituto di Ricovero e Cura a Carattere Scientifico Di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Stefano Vagge
- Radiotherapy Department, E.O. Ospedali Galliera, 16128 Genova, Italy
| | - Marco Galaverni
- Radiation Oncology Unit, University Hospital of Parma, 43126 Parma, Italy
| | - Antonio Pontoriero
- Radiation Oncology Unit, Department of Biomedical, Dental and Morphological and Functional Imaging Sciences, University of Messina, 98122 Messina, Italy
| | - Serena Badellino
- Radiation Oncology, Department of Oncology, University of Turin, 10125 Turin, Italy
| | - Ruggero Spoto
- Department of Radiotherapy and Radiosurgery, Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, 20089 Milan, Italy
| | - Emanuele Alì
- Radiation Oncology Unit, Azienda USL-Istituto di Ricovero e Cura a Carattere Scientifico Di Reggio Emilia, 42123 Reggio Emilia, Italy
| | - Paolo Borghetti
- Radiation Oncology Department, ASST Spedali Civili and University of Brescia, P.le Spedali Civili, 1, 25123 Brescia, Italy;
| |
Collapse
|
59
|
Brune MM, Savic Prince S, Vlajnic T, Chijioke O, Roma L, König D, Bubendorf L. MTAP as an emerging biomarker in thoracic malignancies. Lung Cancer 2024; 197:107963. [PMID: 39357262 DOI: 10.1016/j.lungcan.2024.107963] [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: 07/29/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024]
Abstract
S-methyl-5'-thioadenosine phosphorylase (MTAP) deficiency is an emerging biomarker in non-small cell lung cancer (NSCLC) and beyond. The MTAP gene is located in the chromosomal region 9p21.3, which shows one of the most common homozygous deletions across all human cancers (9p21 loss). Loss of 9p21 is found in the majority of pleural mesotheliomas, where it serves as an established diagnostic marker. Until recently, fluorescence in situ hybridization (FISH) was the gold standard for the detection of 9p21 losses, but loss of MTAP expression by immunohistochemistry (IHC) gains increasing importance as an easy to apply and cost-effective diagnostic surrogate marker. Besides, MTAP loss, which has been reported in 13% of NSCLC, is becoming an emerging predictive biomarker in two different scenarios in NSCLC and other cancer types: 1) MTAP loss seems to negatively predict the response to immune checkpoint inhibitor (ICI) treatment via silencing of the tumor microenvironment, and 2) MTAP loss serves as a predictive biomarker for novel targeted treatment strategies. MTAP deficiency leads to an impaired function of the protein arginine methyltransferase 5 (PRMT5) due to its partial inhibition by MTAP's accumulating substrate methylthioadenosine (MTA). This process leaves MTAP deficient tumor cells heavily dependent on the remaining function of PRMT5, making it a perfect target for synthetic lethality. Indeed, MTA-cooperative PRMT5-inhibitors are now tested in several clinical trials with promising early results in solid malignancies. With its emergence as a predictive biomarker, the implementation of MTAP IHC into diagnostic routine for NSCLC and other tumors is likely to take place soon. In this review article, we summarize the current literature on the role of MTAP in thoracic tumors and evaluate different testing methods, including IHC, FISH and next generation sequencing.
Collapse
Affiliation(s)
- Magdalena M Brune
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Schönbeinstrasse 40, CH-4031 Basel, Switzerland
| | - Spasenija Savic Prince
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Schönbeinstrasse 40, CH-4031 Basel, Switzerland
| | - Tatjana Vlajnic
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Schönbeinstrasse 40, CH-4031 Basel, Switzerland
| | - Obinna Chijioke
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Schönbeinstrasse 40, CH-4031 Basel, Switzerland
| | - Luca Roma
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Schönbeinstrasse 40, CH-4031 Basel, Switzerland
| | - David König
- Division of Medical Oncology, University Hospital Basel, Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Lukas Bubendorf
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Schönbeinstrasse 40, CH-4031 Basel, Switzerland.
| |
Collapse
|
60
|
Yin L, Sun P, Guo S, Shuai P, Zhang J. CAR-T cell therapy: Challenge and opportunity for effective treatment of small cell lung cancer. Biochim Biophys Acta Rev Cancer 2024; 1879:189228. [PMID: 39615863 DOI: 10.1016/j.bbcan.2024.189228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/07/2024] [Accepted: 11/24/2024] [Indexed: 12/08/2024]
Abstract
Small cell lung cancer (SCLC) is a devastating malignancy characterized by rapid metastasis, drug resistance, and frequent recurrence. Owing to the paucity of existing therapeutic options, the prognosis of SCLC remains poor. Recently, the combination of immune checkpoint inhibitors and chemotherapy has resulted in modest improvements in treatment responses. In this review, we characterize the biological signature of SCLC and outline the obstacles to current treatment, including impaired antigen presentation and T cell infiltration. These obstacles may potentially be overcome by chimeric antigen receptor (CAR)-T cell therapy. For the first time, we summarize the available data and discuss the future prospects of CAR-T cell therapy for the treatment of SCLC. Given the high heterogeneity and immunosuppressive tumor microenvironment of SCLC, structural modifications of CAR-T cells and combination therapy may be required to elicit a successful antitumor response. Further research, including clinical trials, is needed to determine the suitability of CAR-T cell therapy as a treatment for SCLC.
Collapse
Affiliation(s)
- Limei Yin
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ping Sun
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Shujin Guo
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Ping Shuai
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Junlin Zhang
- Department of Health Management Center & Institute of Health Management, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
61
|
Chen Y, Liu H, Bai S, Han X, Jin F, Cui B. Clinical Benefits of new Systemic Therapy for Small-Cell Lung Cancer Over Two Decades: A Cross-Sectional Study. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e70032. [PMID: 39476816 PMCID: PMC11524636 DOI: 10.1111/crj.70032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 09/25/2024] [Accepted: 10/15/2024] [Indexed: 11/03/2024]
Abstract
INTRODUCTION Small cell lung cancer (SCLC) is one of the most lethal malignancies worldwide. This study aimed to examine the clinical benefits of new systemic therapies derived from randomized controlled trials (RCTs) published from 2002 to 2023 based on the magnitude of clinical benefit scale developed by the European Society for Medical Oncology (ESMO-MCBS). METHODS We searched PubMed for Phase 3 RCTs on systemic therapy for SCLC published between January 2002 and December 2023. Therapeutic benefit was graded from 5 to 1 according to the ESMO-MCBS framework, with a score of 4 or 5 representing a meaningful clinical benefit. The statistical power of the trial design was also assessed using ESMO-MCBS. RESULTS Sixty-four RCTs with 23 683 participants were eligible for inclusion. The number of RCTs related to molecular targeted therapy or immunotherapy has increased over the years. Among the 62 RCTs for which statistical power could be evaluated, 38 (61.3%) were designed to identify an effect size that would meet the ESMO-MCBS benefit threshold and were less likely to investigate second- or subsequent-line treatment (15.8% vs. 50.0%, p = 0.004), have noninferiority design (0% vs. 25.0%, p = 0.002) and set PFS (0% vs. 16.7%) or response rate (0% vs. 16.7%) as the only primary endpoint (p = 0.002). The ESMO-MCBS framework was applied in 29 RCTs reporting positive results, and only 8 (27.6%) met the threshold for a clinical benefit. The RCTs designed to detect differences that would meet the thresholds were more likely to demonstrate meaningful clinical benefit (87.5% vs. 50.0%, p = 0.099). CONCLUSION Most positive SCLC-RCTs did not meet the ESMO-MCBS threshold for meaningful clinical benefits. Strict power calculations should be adopted in the design of future RCTs.
Collapse
Affiliation(s)
- Yuejing Chen
- Department of Pulmonary and Critical Care MedicineXingtai Third HospitalXingtaiHebeiChina
| | - Honghong Liu
- Department of Pulmonary and Critical Care MedicineXingtai Third HospitalXingtaiHebeiChina
| | - Shaohua Bai
- Department of Pulmonary and Critical Care MedicineXingtai Third HospitalXingtaiHebeiChina
| | - Xuejiao Han
- Department of Pulmonary and Critical Care MedicineXingtai Third HospitalXingtaiHebeiChina
| | - Fei Jin
- Department of Pulmonary and Critical Care MedicineXingtai Third HospitalXingtaiHebeiChina
| | - Bo Cui
- Department of Pulmonary and Critical Care MedicineXingtai Third HospitalXingtaiHebeiChina
| |
Collapse
|
62
|
Jiang MJ, Cui HP, Li TT, Yang XM, Lu XL, Liu AQ. A novel anti-CTLA-4 nanobody-IL12 fusion protein in combination with a dendritic cell/tumour fusion cell vaccine enhances the antitumour activity of CD8 + T cells in solid tumours. J Nanobiotechnology 2024; 22:645. [PMID: 39427185 PMCID: PMC11490160 DOI: 10.1186/s12951-024-02914-6] [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: 11/09/2023] [Accepted: 10/07/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND We previously developed a nanobody targeting CTLA-4 and demonstrated that it can boost antitumour T-cell responses in vitro; however, the resulting responses after the injection of T cells into cancer models are usually weak and transient. Here, we explored whether fusing our nanobody to IL-12 would enable it to induce stronger, longer-lasting T-cell immune responses after exposure to immature dendritic cell and tumour cell fusions. RESULTS The fusion protein enhanced the response of CD8+ T cells to tumour antigens in vitro and led to stronger, more persistent immune responses after the T cells were injected into mice bearing different types of xenografts. CONCLUSION Our in vitro and in vivo results suggest the anticancer potential of our nanobody-interleukin fusion system and support the clinical application of this fusion approach for various nanobodies.
Collapse
Affiliation(s)
- Meng-Jie Jiang
- Department of Gastroenterology and Respiratory Medicine & Endoscopy Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, China
| | - Hao-Peng Cui
- Department of Gastroenterology and Respiratory Medicine & Endoscopy Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, China
| | - Ting-Ting Li
- Department of Gastroenterology and Respiratory Medicine & Endoscopy Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, China
| | - Xiao-Mei Yang
- Guangxi Key Laboratory of Nanobody Research and Guangxi Nanobody Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Xiao-Ling Lu
- Guangxi Key Laboratory of Nanobody Research and Guangxi Nanobody Engineering Research Center, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Ai-Qun Liu
- Department of Gastroenterology and Respiratory Medicine & Endoscopy Center, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, China.
| |
Collapse
|
63
|
Zhao Q, Wang C, Huang W, Song Z, Lang Y, Zhu X. Potential immunologic and prognostic roles of CHRNA6 in SCLC and pan-cancer. Heliyon 2024; 10:e38572. [PMID: 39398083 PMCID: PMC11470509 DOI: 10.1016/j.heliyon.2024.e38572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/22/2024] [Accepted: 09/26/2024] [Indexed: 10/15/2024] Open
Abstract
Background Small cell lung cancer (SCLC) is considered the most malignant subtype of lung cancer, and it has a restricted range of therapeutic choices. The emergence of immunotherapy has offered new possibilities for patients with SCLC. However, the scarcity of clinical specimens has hampered the progress of clinical studies and we still face a shortage of dependable indicators to forecast the effectiveness of immunotherapy for SCLC. Methods In our study, we assessed the ImmuneScore and StromalScore of 81 SCLC samples obtained from the cBioPortal database. By comparing gene expression differences between the high and low immune scores groups, we identified 24 differentially expressed genes. Subsequently, an intersection was performed with genes that exhibited differential expression between normal and SCLC tissues, leading us to isolate the gene CHRNA6. To gain a deeper insight into the possible significance of CHRNA6 in SCLC, we singled out 50 genes that showed the most pronounced positive and negative associations with its expression. We then pinpointed hub genes for subsequent functional enrichment analyses by establishing a protein-protein interactions network. We additionally assessed the link between CHRNA6 expression in SCLC and characteristics of the immune microenvironment, along with the efficacy of immunotherapy, using the CIBERSORT, immunophenoscores (IPS), and tumor immune dysfunction and exclusion (TIDE) algorithms. Furthermore, we confirmed the prognostic impact of CHRNA6 expression in SCLC patients undergoing immunotherapy within a clinical cohort. Lastly, we obtained data from The Cancer Genome Atlas (TCGA) to investigate CHRNA6 expression in various tumors and its associations with genetic alterations, DNA methylation, copy number variation, clinicopathological characteristics, biological processes, immune microenvironment, prognosis, and drug sensitivity. Results In SCLC, we found that CHRNA6 function was associated with immune activation pathways such as antigen presentation processing and positive regulation of adaptive immune response, and that CHRNA6 demonstrated a strong correlation with immune cells infiltration. In addition, analysis of the clinical cohort revealed that patients with SCLC who exhibited elevated expression of CHRNA6 experienced better responses to immunotherapy. Our pan-cancer analysis disclosed that the expression of CHRNA6 is dysregulated in a multitude of cancers, potentially due to genetic mutations, copy number gains, and DNA demethylation. The gene set enrichment analysis (GSEA) outcomes indicated that CHRNA6 participates in immune responses and may play a positive immune regulatory role in most cancers. Furthermore, CHRNA6 has been observed to have a notable relationship with immune checkpoints, immunomodulators, immune cell infiltration, patient outcomes, and drug sensitivity across various cancers. Conclusions Our findings indicate that the CHRNA6 may act as a predictive indicator for SCLC patients receiving immunotherapy. The study also uncovers the aberrant expression of CHRNA6 in a range of human cancers and its potential roles in immunology and prognosis, offering novel perspectives for tailored cancer therapies.
Collapse
Affiliation(s)
- Qingqing Zhao
- Department of Respiratory and Critical Care Medicine, Southeast University, Zhongda Hospital, Nanjing, 210009, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Cong Wang
- Department of Pathology, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Wucui Huang
- Department of Respiratory and Critical Care Medicine, Southeast University, Zhongda Hospital, Nanjing, 210009, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Zhongquan Song
- Department of Respiratory and Critical Care Medicine, Southeast University, Zhongda Hospital, Nanjing, 210009, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yang Lang
- Department of Respiratory and Critical Care Medicine, Southeast University, Zhongda Hospital, Nanjing, 210009, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Xiaoli Zhu
- Department of Respiratory and Critical Care Medicine, Southeast University, Zhongda Hospital, Nanjing, 210009, China
- School of Medicine, Southeast University, Nanjing, 210009, China
| |
Collapse
|
64
|
Oh S, Koh J, Kim TM, Kim S, Youk J, Kim M, Keam B, Jeon YK, Ku JL, Kim DW, Chung DH, Heo DS. Transcriptomic Heterogeneity of EGFR-Mutant Non-Small Cell Lung Cancer Evolution Toward Small-Cell Lung Cancer. Clin Cancer Res 2024; 30:4729-4742. [PMID: 39150541 DOI: 10.1158/1078-0432.ccr-24-0160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 06/25/2024] [Accepted: 08/14/2024] [Indexed: 08/17/2024]
Abstract
PURPOSE Histologic transformation from EGFR-mutant non-small cell lung cancer (NSCLC) to small-cell lung cancer (SCLC) is a key mechanism of resistance to EGFR tyrosine kinase inhibitors (TKI). However, transcriptomic changes between NSCLC and transformed SCLC (t-SCLC) remain unexplored. EXPERIMENTAL DESIGN We conducted whole-transcriptome analysis of 59 regions of interest through the spatial profiling of formalin-fixed, paraffin-embedded tissues obtained from 10 patients (lung adenocarcinoma, 22; combined SCLC/NSCLC, 7; and t-SCLC, 30 regions of interests). Transcriptomic profiles and differentially expressed genes were compared between pre- and post-transformed tumors. RESULTS Following EGFR-TKI treatment, 93.7% (15/16) of t-SCLC components evolved into neuroendocrine-high subtypes (SCLC-A or SCLC-N). The transition to t-SCLC occurred regardless of EGFR-TKI treatment and EGFR mutational status, with a notable decrease in EGFR expression (P < 0.001) at both mRNA and protein levels. Pathway analysis revealed that gene overexpression was related to epigenetic alterations in t-SCLC. Interestingly, histone deacetylase inhibitors restored EGFR expression in SNU-2962A cells and their organoid model. The synergistic effects of third-generation EGFR-TKI osimertinib and the histone deacetylase inhibitor fimepinostat were validated in both in vitro and in vivo models. CONCLUSIONS Our study demonstrated that most t-SCLC cases showed neuronal subtypes with low EGFR expression. Differentially expressed gene analysis and t-SCLC preclinical models identified an epigenetic modifier as a promising treatment strategy for t-SCLC.
Collapse
Affiliation(s)
- Songji Oh
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, South Korea
| | - Jaemoon Koh
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Tae Min Kim
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Soyeon Kim
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, South Korea
| | - Jeonghwan Youk
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Miso Kim
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Bhumsuk Keam
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Yoon Kyung Jeon
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, South Korea
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Ja-Lok Ku
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea
| | - Dong-Wan Kim
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University College of Medicine, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Doo Hyun Chung
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Dae Seog Heo
- Cancer Research Institute, Seoul National University, Seoul, South Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| |
Collapse
|
65
|
Zhai W, Yu Y, Wu H, Zhang Q, Chen Y, Yang Y, Fan Y. Real-world efficacy of PD-1/PD-L1 inhibitors in patients with advanced pulmonary neuroendocrine carcinoma: a single-center analysis. Ther Adv Med Oncol 2024; 16:17588359241288130. [PMID: 39410959 PMCID: PMC11475206 DOI: 10.1177/17588359241288130] [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: 04/29/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
Background Immunotherapy blocking programmed death-1 (PD-1)/programmed death ligand-1 (PD-L1) has revolutionized the treatment of extensive-stage small-cell lung cancer (SCLC), but only with limited real-world efficacy data; evidence from immunotherapy for other pulmonary neuroendocrine carcinoma (PNEC) is scarce. Objective The purpose of this study is to evaluate the efficacy of receiving PD-1/PD-L1 inhibitors in patients with advanced PNEC and explore factors related to survival prognosis, providing clues for treatment for patients with advanced PNEC. Methods In all, 203 patients with advanced PNEC who received PD-1/PD-L1 inhibitors between January 2019 and December 2021 were retrospectively analyzed. Kaplan-Meier curves were constructed for progression-free survival (PFS) and overall survival (OS). Results For the 203 patients, the objective response rate (ORR) was 48.3%, the disease control rate (DCR) was 83.3%, the median PFS (mPFS) was 6.0 months, and the median OS (mOS) was 13.1 months. Among them, the histology was 166 SCLC, 13 large-cell neuroendocrine carcinoma, and 24 other unspecified PNEC. Histologically, no significant difference was observed in PFS (p = 0.240) or OS (p = 0.845). In first-line (1L) treatment (N = 125), patients received chemoimmunotherapy and had an ORR of 64.8%, DCR of 92.0%, mPFS of 6.6 months, and mOS of 14.9 months. In second-line (2L) or later-line setting, the ORR, DCR, mPFS, and mOS were 21.8%, 69.2%, 4.4, and 9.4 months; immunotherapy plus small-molecule antiangiogenic agents showed significantly greater PFS than immunotherapy monotherapy or chemoimmunotherapy (6.4 vs 1.4 vs 3.7 months, p = 0.041). Patients without liver metastasis had superior PFS (7.0 vs 5.1 months, p < 0.001) and OS (19.2 vs 9.6 months, p < 0.001) than those with liver metastasis. Conclusion In clinical practice, PD-1/PD-L1 inhibitors are effective in patients with advanced PNEC, regardless of the pathological histology. The efficacy of 1L immunochemotherapy is worthy of recognition, and the addition of small-molecule antiangiogenic agents to immunotherapy in 2L or later-line treatment provides a better survival trend. Design Retrospective study.
Collapse
Affiliation(s)
- Wanchen Zhai
- Department of Oncology, The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Ying Yu
- Department of Oncology, The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Haicheng Wu
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, Wenzhou, China
| | - Qian Zhang
- Department of Oncology, The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Yunfei Chen
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, Wenzhou, China
| | - Yehao Yang
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, Wenzhou, China
| | - Yun Fan
- Department of Thoracic Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, No. 1 East Banshan Road, Gongshu District, Hangzhou, Zhejiang 310022, China
| |
Collapse
|
66
|
Cui Y, Chen Y, Zhao P, Li S, Cheng Y, Ren X. Peripheral NK cells identified as the predictor of response in extensive-stage small cell lung cancer patients treated with first-line immunotherapy plus chemotherapy. Clin Transl Oncol 2024; 26:2522-2530. [PMID: 38662170 DOI: 10.1007/s12094-024-03479-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 03/23/2024] [Indexed: 04/26/2024]
Abstract
PURPOSE Although immunotherapy improves outcomes in extensive-stage small-cell lung cancer (ES-SCLC), the search for biomarkers predicting treatment success is crucial. Natural killer (NK) cells are potential indicators in various cancers, however, their precise role in ES-SCLC prognosis remains unclear. METHODS In this retrospective study, 33 patients with ES-SCLC treated with first-line immuno-chemotherapy were enrolled. The peripheral NK cell percentage and its longitudinal dynamics were analyzed using flow cytometry. Progression-free survival (PFS) and overall survival (OS) were calculated as hazard ratio (HR) and compared statistically. RESULTS The median PFS was better in the group with normal baseline NK cell levels than the low group (7.0 vs. 4.6 months; HR = 0.17; 95% CI 0.07-0.41; P < 0.0001), but there was no association with OS (14.9 vs. 10.3 months; HR = 0.55; 95% CI 0.23-1.31; P = 0.171). Furthermore, the NK cell% for 95.0% of patients increased after immunochemotherapy in the clinical response group (P = 0.0047), which led to a better median PFS (6.3 vs. 2.1 months; HR = 0.23; 95% CI 0.05-0.98; P < 0.0001) and OS (14.9 vs. 5.9 months; HR = 0.20; 95% CI 0.04-1.02; P < 0.0001). Similar trends were observed with NK cell% changes up to disease progression, improving PFS (6.5 vs. 4.3; HR = 0.41; 95% CI 0.12-0.92; P = 0.0049) and OS (17.4 vs. 9.7; HR = 0.42; 95% CI 0.17-1.02; P < 0.0001). CONCLUSION In patients with ES-SCLC, the percentage and changes in peripheral NK cells can predict the response to combined immunotherapy and chemotherapy.
Collapse
Affiliation(s)
- Yanan Cui
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
- Division of Thoracic Oncology, Jilin Cancer Hospital, Changchun, China
| | - Yanping Chen
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China
- Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Peiyan Zhao
- Translational Oncology Research Lab, Jilin Cancer Hospital, Changchun, China
| | - Shuang Li
- Big Data Center of Clinical, Jilin Cancer Hospital, Changchun, China
| | - Ying Cheng
- Division of Thoracic Oncology, Jilin Cancer Hospital, Changchun, China.
| | - Xiubao Ren
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
- Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, China.
| |
Collapse
|
67
|
Sun M, Ji H, Deng F, Li J, Xu N, Li Y. Clinical outcomes and synergistic effect between radiotherapy and immunotherapy in patients with extensive-stage small cell lung cancer: a real-world study. BMC Cancer 2024; 24:1206. [PMID: 39350057 PMCID: PMC11441094 DOI: 10.1186/s12885-024-12942-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Patients with extensive-stage small cell lung cancer (ES-SCLC) experience significant therapeutic challenges and limited survival rates. This study aimed to investigate the efficacy of combining immunotherapy (IT) with chemotherapy (CT) for treating ES-SCLC and to explore the synergistic effect between radiotherapy (RT) and IT. METHODS This retrospective analysis examined patients with ES-SCLC who received treatment at three centers. Furthermore, propensity score-matched (PSM) analysis was conducted. The Kaplan‒Meier method and Cox proportional hazards regression were used to compare the survival outcomes. RESULTS A total of 257 eligible patients with ES-SCLC were included in the analysis. Among all patients, the median overall survival (mOS) was 18.0 m in the chemoimmunotherapy (CT + IT) group and 15.7 m in the CT group (p = 0.208). The median real-world progression-free survival (mrwPFS) was 7.7 m and 6.8 m (p = 0.043) in the CT + IT and CT group, respectively. Moreover, the mOS was 22.0 m in the chemoradiotherapy (CT + RT) group and 13.6 m in the CT group (p < 0.001). The mrwPFS was 7.4 m and 6.0 m (p = 0.175) in the CT + RT group and CT group, respectively. The multivariate analyses revealed that sex, liver metastasis and RT were independent prognostic factors for OS (p < 0.05), while liver metastasis and IT were found to be independent predictive factors of real-world progression-free survival (rwPFS) (p < 0.05). After PSM, the mOS was 23.2 m in the CT + IT group and 13.0 m in the CT group (p = 0.008). The mrwPFS was 7.3 m and 6.2 m (p = 0.096) in the CT + IT group and the CT group, respectively. Moreover, the mOS was 21.4 m in the CT + RT group and 12.5 m in the CT group (p < 0.001). The mrwPFS was 7.3 m and 5.2 m (p = 0.220) in the CT + RT group and the CT group, respectively. Additionally, our study revealed that in the PD-1 group, RT significantly improved patient survival (36.0 m vs. 15.8 m, p = 0.041). CONCLUSION An increasing number of treatment options are being explored for ES-SCLC, and CT is the cornerstone of treatment for this disease. Combining CT with IT and RT has demonstrated remarkable efficacy and excellent safety profiles, and such treatments are worthy of further exploration.
Collapse
Affiliation(s)
- Meiling Sun
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107 Wenhua Xilu, Jinan, 250012, Shandong, China
- Department of Respiratory Medicine, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, 70 Heping Road, Weihai, 264200, Shandong, China
| | - Huaijun Ji
- Department of Thoracic Surgery, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, 70 Heping Road, Weihai, 264200, Shandong, China.
| | - Fang Deng
- Department of Oncology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, 254300, Shandong, China
| | - Jingyi Li
- Department of Radiation Oncology, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, 70 Heping Road, Weihai, 264200, Shandong, China
| | - Ning Xu
- Department of Respiratory Medicine, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, 70 Heping Road, Weihai, 264200, Shandong, China
| | - Yu Li
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, No. 107 Wenhua Xilu, Jinan, 250012, Shandong, China.
| |
Collapse
|
68
|
Taniguchi H, Chakraborty S, Takahashi N, Banerjee A, Caeser R, Zhan YA, Tischfield SE, Chow A, Nguyen EM, Villalonga ÁQ, Manoj P, Shah NS, Rosario S, Hayatt O, Qu R, de Stanchina E, Chan J, Mukae H, Thomas A, Rudin CM, Sen T. ATR inhibition activates cancer cell cGAS/STING-interferon signaling and promotes antitumor immunity in small-cell lung cancer. SCIENCE ADVANCES 2024; 10:eado4618. [PMID: 39331709 PMCID: PMC11430494 DOI: 10.1126/sciadv.ado4618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 08/23/2024] [Indexed: 09/29/2024]
Abstract
Patients with small-cell lung cancer (SCLC) have poor prognosis and typically experience only transient benefits from combined immune checkpoint blockade (ICB) and chemotherapy. Here, we show that inhibition of ataxia telangiectasia and rad3 related (ATR), the primary replication stress response activator, induces DNA damage-mediated micronuclei formation in SCLC models. ATR inhibition in SCLC activates the stimulator of interferon genes (STING)-mediated interferon signaling, recruits T cells, and augments the antitumor immune response of programmed death-ligand 1 (PD-L1) blockade in mouse models. We demonstrate that combined ATR and PD-L1 inhibition causes improved antitumor response than PD-L1 alone as the second-line treatment in SCLC. This study shows that targeting ATR up-regulates major histocompatibility class I expression in preclinical models and SCLC clinical samples collected from a first-in-class clinical trial of ATR inhibitor, berzosertib, with topotecan in patients with relapsed SCLC. Targeting ATR represents a transformative vulnerability of SCLC and is a complementary strategy to induce STING-interferon signaling-mediated immunogenicity in SCLC.
Collapse
Affiliation(s)
- Hirokazu Taniguchi
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Subhamoy Chakraborty
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nobuyuki Takahashi
- Department of Medical Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Avisek Banerjee
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rebecca Caeser
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yingqian A. Zhan
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sam E. Tischfield
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew Chow
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Evelyn M. Nguyen
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Álvaro Quintanal Villalonga
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Parvathy Manoj
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nisargbhai S. Shah
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Samantha Rosario
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar Hayatt
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rui Qu
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Elisa de Stanchina
- Antitumor Assessment Core, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Joseph Chan
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Program for Computational and Systems Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Hiroshi Mukae
- Department of Respiratory Medicine, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Anish Thomas
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, Bethesda, USA
| | - Charles M. Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Triparna Sen
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
69
|
Gomez-Randulfe I, Silva Díaz S, Escriu C, Mohammed S, Shah R, Benitez Fuentes JD, Cox S, Monaca F, Bria E, García-Campelo MR, Crook B, Talbot T, Leporati R, Balachandran K, Newsom-Davis T, Hughes S, Cove-Smith L, Taylor P, Blackhall F, Califano R. Efficacy of carboplatin-etoposide rechallenge after first-line chemo-immunotherapy in ES-SCLC: an international multicentric analysis. Ther Adv Med Oncol 2024; 16:17588359241272957. [PMID: 39355343 PMCID: PMC11443580 DOI: 10.1177/17588359241272957] [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: 05/20/2024] [Accepted: 07/17/2024] [Indexed: 10/03/2024] Open
Abstract
Background and objectives Second-line treatment for small-cell lung cancer (SCLC) is primarily guided by the time elapsed since the last platinum dose. Rechallenge with carboplatin and etoposide has demonstrated superior outcomes compared to topotecan if the platinum-free interval (PFI) is longer than 90 days and is considered the standard of care. However, these findings predate the chemo-immunotherapy era. This study investigates the effectiveness of the rechallenge strategy after chemo-immunotherapy in a real-world setting. Design and methods We retrospectively reviewed patients with the extensive stage (ES)-SCLC who received rechallenge with carboplatin and etoposide after first-line chemoimmunotherapy between September 2020 and August 2023 in nine European centres. Demographic and clinical data were collected and analysed. Results A total of 93 patients were included. Sixty-six (71%) patients had a PFI between 3 and 6 months. Consolidation thoracic radiotherapy and prophylactic cranial irradiation had been administered in 31 (33.3%) patients and 20 (21.5%) patients, respectively. Overall response rate was 59.1%. Median progression-free survival (PFS) was 5 months (95% confidence interval (CI) 4.3-5.7) and median overall survival (OS) was 7 months (95% CI 5.7-8.3). Notably, PFS and OS were not different according to PFI (3-6 m vs > 6 m). Conclusion Rechallenge with carboplatin and etoposide is a valid second-line option in patients with ES-SCLC whose disease progresses after first-line chemoimmunotherapy. Our analysis shows similar results to previous studies. Furthermore, outcomes were consistent across patients with different PFIs, confirming its efficacy in patients with a PFI longer than 3 months.
Collapse
Affiliation(s)
- Igor Gomez-Randulfe
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Sofía Silva Díaz
- Medical Oncology, Complejo Hospitalario Universitario A Coruña, Coruña, Spain
| | - Carles Escriu
- Medical Oncology, The Clatterbridge Cancer Centre, Liverpool, UK
| | | | - Riyaz Shah
- Oncology Department, Maidstone Hospital, Maidstone, UK
| | | | - Samantha Cox
- Clinical Oncology, Velindre Cancer Centre NHS Wales, Cardiff, UK
| | - Federico Monaca
- Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Emilio Bria
- Medical Oncology Department, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | | | | | | | - Rita Leporati
- Oncology Department, Istituto Nazionale dei Tumori di Milano – Fondazione IRCCS, Milan, Italy
| | | | - Tom Newsom-Davis
- Oncology Department, Chelsea and Westminster Hospital NHS Trust, London, UK
| | - Sarah Hughes
- 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
| | - Paul Taylor
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| | - Fiona Blackhall
- Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | - Raffaele Califano
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK Division of Cancer Sciences, The University of Manchester, Manchester, UK
| |
Collapse
|
70
|
Reinmuth N, Juan-Vidal O, Kowalski D, Bryl M, Kryzhanivska A, Vicente D, Horváth Z, Gálffy G, Csánky E, Pápai Székely Z, Vynnychenko I, Armstrong J, Dalvi T, Xie M, Iyer S, Shrestha Y, Jiang H, Bondarenko I. Novel Combinations of Immunotherapies or DNA Damage Repair Inhibitors in Platinum-Refractory Extensive-Stage Small Cell Lung Cancer: The Phase II BALTIC Study. Clin Cancer Res 2024; 30:4055-4067. [PMID: 39017667 PMCID: PMC11393542 DOI: 10.1158/1078-0432.ccr-24-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/12/2024] [Accepted: 07/16/2024] [Indexed: 07/18/2024]
Abstract
PURPOSE The phase II, multiarm, signal-searching BALTIC study (NCT02937818) assessed novel treatment combinations for platinum-refractory/resistant extensive-stage small cell lung cancer (ES-SCLC). PATIENTS AND METHODS Patients with ES-SCLC with progressive disease during or within 90 days of completing first-line platinum-based chemotherapy received one of three regimens: durvalumab plus tremelimumab followed by durvalumab monotherapy (arm A), adavosertib plus carboplatin (arm B), or ceralasertib plus olaparib (arm C). The primary endpoint was the objective response rate. Prespecified exploratory biomarker analyses were conducted in arms A and C. RESULTS In arm A (n = 41), arm B (n = 10), and arm C (n = 21), the confirmed objective response rates were 7.3%, 0%, and 4.8%, respectively. Safety profiles in all arms were consistent with those of the individual drugs. In arm A, patients with PD-L1 expression (tumor cells or immune cells) ≥1% seemed to have a greater likelihood of achieving disease control with durvalumab plus tremelimumab than those with PD-L1 (tumor cells and immune cells) <1%, and lower baseline ctDNA and reduction in the on-treatment ctDNA level were both associated with longer overall survival. Among patients treated with ceralasertib plus olaparib in arm C, specific immune response-relevant circulating chemokines and cytokines were identified as early biomarkers of survival and pharmacodynamic biomarkers. CONCLUSIONS In BALTIC, all combination regimens demonstrated tolerable safety profiles, but antitumor activity was limited in refractory/resistant ES-SCLC. Among patients treated with durvalumab plus tremelimumab, an association of on-treatment reduction in ctDNA with longer overall survival suggests the potential use of ctDNA as a surrogate of treatment response, warranting further investigation.
Collapse
Affiliation(s)
- Niels Reinmuth
- Department of Thoracic Oncology, Asklepios Lung Clinic Munich-Gauting, Gauting, Germany.
- German Center of Lung Research, LMU Munich, Munich, Germany.
| | - Oscar Juan-Vidal
- Department of Medical Oncology, La Fe University Hospital, Valencia, Spain.
| | - Dariusz Kowalski
- Department of Lung Cancer and Thoracic Tumours, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland.
| | - Maciej Bryl
- Oncology Department, E.J. Zeyland Wielkopolska Center of Pulmonology and Thoracic Surgery, Poznan, Poland.
| | - Anna Kryzhanivska
- Ivano-Frankivsk National Medical University, Ivano-Frankivsk, Ukraine.
| | - David Vicente
- Hospital Universitario Virgen Macarena, Seville, Spain.
| | - Zsolt Horváth
- Bacs-Kiskun County Teaching Hospital, Kecskemét, Hungary.
| | | | - Eszter Csánky
- Department of Pulmonology, Semmelweis Hospital, Miskolc, Hungary.
| | | | | | | | | | | | | | | | | | - Igor Bondarenko
- Dnipropetrovsk State Medical Academy, Dnipropetrovsk, Ukraine.
| |
Collapse
|
71
|
Li X, Tong L, Wang S, Yu J, Lu B, Wang Q, Hu M, Wu J, Yu J, Li B, Zhang T. Integration of clinical and blood parameters in risk prognostication for patients receiving immunochemotherapy for extensive stage small cell lung cancer: real-world data from two centers. BMC Med 2024; 22:381. [PMID: 39256789 PMCID: PMC11389556 DOI: 10.1186/s12916-024-03612-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 09/04/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) had modest advances in the treatment of extensive-stage small cell lung cancer (ES-SCLC) in clinical trials, but there is a lack of biomarkers for prognosis in clinical practice. METHODS We retrospectively collected data from ES-SCLC patients who received ICIs combined chemotherapy from two centers in China, integrated clinical and blood parameters, and constructed risk prognostication for immunochemotherapy. The population was divided into high- and low-risk groups, and the performance of the model was assessed separately in the training and validation cohorts. RESULTS Two hundred and twenty and 43 patients were included in the training and validation groups, respectively. The important predictors were screened including body mass index, liver metastases, coefficient variation of red blood cell distribution width, lactate dehydrogenase, albumin, and C-reactive protein. Predicting 1-year overall survival (OS), the AUC values under ROC for the model under training, internal validation, and external validation were 0.760, 0.732, and 0.722, respectively, and the calibration curve and clinical decision curve performed well. Applied the model to divide patients into low-risk and high-risk groups, and the median OS was 23.7 months and 9.1 months, and the median progression-free survival was 8.2 months and 4.8 months, respectively; furthermore, this ability to discriminate survival was also observed in the validation cohort. CONCLUSIONS We constructed a novel prognostic model for ES-SCLC to predict survival employing baseline tumor burden, nutritional and inflammatory parameters, it is easily measured to screen high-risk patient populations.
Collapse
Affiliation(s)
- Xiaomi Li
- Department of Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
- Department of Oncology, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Li Tong
- Department of Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Shan Wang
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Jiaqi Yu
- First Clinical Medical College, Shaanxi University of Traditional Chinese Medicine, Xianyang, 712000, Shaanxi, China
| | - Baohua Lu
- Department of Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Qunhui Wang
- Department of Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Mingming Hu
- Department of Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China
| | - Jinxiang Wu
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Jing Yu
- Cancer Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China.
| | - Baolan Li
- Department of Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
| | - Tongmei Zhang
- Department of Oncology, Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, 101149, China.
- Laboratory for Clinical Medicine, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
72
|
Andrini E, Ricco G, Zappi A, Aloi S, Giordano M, Altimari A, Gruppioni E, Maloberti T, de Biase D, Campana D, Lamberti G. Challenges and future perspectives for the use of temozolomide in the treatment of SCLC. Cancer Treat Rev 2024; 129:102798. [PMID: 38970838 DOI: 10.1016/j.ctrv.2024.102798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 06/09/2024] [Accepted: 07/02/2024] [Indexed: 07/08/2024]
Abstract
Small-cell lung cancer (SCLC), accounting for 10-20 % of all lung tumors, represents the most aggressive high-grade neuroendocrine carcinoma. Most patients are diagnosed with extensive-stage SCLC (ES-SCLC), with brian metastases identified in ∼ 80 % of cases during the disease cours, and the prognosis is dismal, with a 5-year survival rate of less than 5 %. Current available treatments in the second-line setting are limited, and topotecan has long been the only FDA-approved drug in relapsed or refractory ES-SCLC, until the recent approval of lurbinectedin, a selective inhibitor of RNA polymerase II. Temozolomide (TMZ) is an oral alkylating agent, which showed single-agent activity in SCLC, particularly among patients with O6-methylguanine-DNA methyltransferase (MGMT) promoter methylation. Several studies have revealed the synergistic activity of temozolomide with poly-ADP-ribose polymerase (PARP) inhibitors, that prevent repair of TMZ-induced DNA damage. This review focuses on the rationale for the use of TMZ in ES-SCLC and provides an overview of the main trials that have evaluated and are currently investigating its role, both as a single-agent and in combinations, in relapse or refractory disease.
Collapse
Affiliation(s)
- Elisa Andrini
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy.
| | - Gianluca Ricco
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy.
| | - Arianna Zappi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy.
| | - Serena Aloi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy.
| | - Mirela Giordano
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy.
| | - Annalisa Altimari
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Elisa Gruppioni
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Thais Maloberti
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
| | - Dario de Biase
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Pharmacy and Biotechnology (FaBit), University of Bologna, Bologna, Italy.
| | - Davide Campana
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy.
| | - Giuseppe Lamberti
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; Department of Medical and Surgical Sciences (DIMEC), Alma Mater Studiorum, University of Bologna, 40126 Bologna, Italy.
| |
Collapse
|
73
|
Mu X, Zhou Y, Liu Q, Wang J, Xu F, Luo F, Wang K, Li L, Tian P, Li Y, Liu J, Zhang Y, Liu J, Li Y. Impact of thoracic radiotherapy on first-line treatment outcomes in ES-SCLC patients. Cancer Med 2024; 13:e70175. [PMID: 39240166 PMCID: PMC11378358 DOI: 10.1002/cam4.70175] [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: 11/27/2023] [Revised: 07/17/2024] [Accepted: 08/20/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND The therapeutic advantage of thoracic radiotherapy (tRT) as an adjunct to first-line immunotherapy and chemotherapy in patients with extensive-stage small cell lung cancer (ES-SCLC) remains unclear. We sought to elucidate this in a retrospective cohort study comparing the effectiveness and safety of tRT in combination with first-line immunotherapy and chemotherapy. METHODS Our retrospective study included patients with ES-SCLC, treated at the West China Hospital between January 2019 and December 2022. They received first-line immunotherapy and chemotherapy and were categorized into two cohorts based on the administration of tRT. The primary outcomes were overall survival (OS) and progression-free survival (PFS). Cox regression analysis was utilized to identify potential independent predictors of prognosis and to compare the treatment outcomes across various patient subgroups. Treatment-related toxicities across both cohorts were compared using the Chi-squared test. RESULTS A total of 99patients were eligible for the study, out of which 55 received tRT. The medianduration of follow-up was 39 months. Remarkably, patients who received tRTdemonstrated superior OS and PFS in comparison to those who did not (P < 0.05). Subgroup analysis further confirmed these findings. Multivariate analysisidentified treatment group and liver metastasis as independent prognosticfactors (P < 0.05). The incidence of grade 3-4 adverse events showed nostatistically significant difference between the two cohorts. CONCLUSIONS Thus, weconfirmed that the addition of tRT to the conventional regimen of first-linechemotherapy and immunotherapy yields better survival outcomes without asignificant increase in toxicity.
Collapse
Affiliation(s)
- Xiaoli Mu
- The Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yixin Zhou
- The Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qing Liu
- Lung cancer center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiantao Wang
- Lung cancer center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Xu
- Lung cancer center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Luo
- Lung cancer center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ke Wang
- Lung cancer center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lu Li
- Lung cancer center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Panwen Tian
- Lung cancer center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yalun Li
- Lung cancer center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiewei Liu
- Lung cancer center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Zhang
- Lung cancer center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiyan Liu
- The Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Li
- Lung cancer center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
74
|
Chen D, Zou B, Li B, Gao A, Huang W, Shao Q, Meng X, Zhang P, Tang X, Hu X, Zhang Y, Guo J, Zhao C, Yuan J, Li Q, Zhu C, Yu J, Wang L. Adebrelimab plus chemotherapy and sequential thoracic radiotherapy as first-line therapy for extensive-stage small-cell lung cancer (ES-SCLC): a phase II trial. EClinicalMedicine 2024; 75:102795. [PMID: 39252865 PMCID: PMC11381814 DOI: 10.1016/j.eclinm.2024.102795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
Background This phase II prospective trial aimed to investigate the efficacy and safety of adebrelimab (PD-L1 antibody) plus first-line chemotherapy followed by sequential thoracic radiotherapy (TRT) combined with adebrelimab in extensive-stage small-cell lung cancer (ES-SCLC). Biomarkers associated with potential therapeutic effects were also explored. Methods Patients with previously untreated ES-SCLC were enrolled at Shandong Cancer Hospital and Institute (Jinan, China). Patients received 4-6 cycles of adebrelimab (20 mg/kg, D1, Q3W) combined with EP/EC (etoposide, 100 mg/m2, D1-3, Q3W and cisplatin, 75 mg/m2, D1, Q3W or carboplatin, AUC = 5, D1, Q3W). Then patients with response sequentially underwent consolidative TRT (≥30 Gy in 10 fractions or ≥50 Gy in 25 fractions, involved-field irradiation), and maintenance adebrelimab until disease progression or intolerable adverse events (AEs). The primary endpoint was overall survival (OS). Genomic and circulating tumour DNA (ctDNA) profiling were also analyzed with tumour tissues and peripheral blood. This trial was registered with ClinicalTrials.gov, NCT04562337. Findings From October 2020 to April 2023, 67 patients diagnosed with ES-SCLC were enrolled and received at least one dose of study treatment. All patients were included in the efficacy and safety analyses. 45 patients received sequential TRT as planned. The median OS and progression-free survival (PFS) was 21.4 months (95% CI: 17.2-not reached months) and 10.1 months (95% CI: 6.9-15.5 months), respectively. The confirmed objective response rate was 71.6% (48/67, 95% CI: 59.3-82.0%) and disease control rate was 89.6% (60/67, 95% CI: 79.7-95.7%). There were no treatment-related deaths. The most common grade 3 or higher treatment-related adverse events (TRAEs) were hematological toxicities. The incidence of any grade and G3+ pneumonitis was 25% (17/67) and 6% (4/67), respectively. No unexpected adverse events were observed. Patients without co-mutations of TP53/RB1 in both tissue and peripheral blood displayed longer PFS (tissue, P = 0.071; ctDNA, P = 0.060) and OS (tissue, P = 0.032; ctDNA, P = 0.031). Interpretation Adebrelimab plus chemotherapy and sequential TRT as first-line therapy for ES-SCLC showed promising efficacy and acceptable safety. Funding This study was funded by the National Natural Science Foundation of China (82172865), Jiangsu Hengrui Pharmaceuticals Co., Ltd. and Amoy Diagnostics Co., Ltd.
Collapse
Affiliation(s)
- Dawei Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Bing Zou
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Butuo Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Aiqin Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wei Huang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Qian Shao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Pinliang Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiaoyong Tang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xudong Hu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yan Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jun Guo
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Changhong Zhao
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Shanghai, China
| | - Jiajia Yuan
- Jiangsu Hengrui Pharmaceuticals Co., Ltd., Shanghai, China
| | - Qian Li
- Amoy Diagnostics Co., Ltd., Xiamen, Fujian, China
| | - Changbin Zhu
- Amoy Diagnostics Co., Ltd., Xiamen, Fujian, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Linlin Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
75
|
Tian L, Huang JX, Wan R, Zhang J, Zhang X, Li N, Li N, Liu XQ, Song CX, Wang XY, Yu L, Wang SM, Wang ZJ, Cong MH. Association Between Sarcopenia, Clinical Outcomes, and Survival in Patients with Extensive-Stage Small Cell Lung Cancer Treated with First-Line Immunochemotherapy: A Prospective Cohort Study. Nutr Cancer 2024; 77:62-69. [PMID: 39160664 DOI: 10.1080/01635581.2024.2392297] [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: 05/29/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/21/2024]
Abstract
OBJECTIVE To investigate the association between sarcopenia, short-term efficacy, and long-term survival in patients with extensive small-cell lung cancer (SCLC) treated with standard first-line immunochemotherapy. METHODS A total of 63 patients initially diagnosed with extensive-stage small cell lung cancer were enrolled in the prospective study from December 1, 2020 to December 31, 2022. The clinical characteristics, body composition, blood test results, and image data were obtained before treatment. Patients were divided into sarcopenia and non-sarcopenia groups according to the diagnostic criteria of the Asian Sarcopenia Working Group 2019. The primary outcome was overall survival (OS) and comprehensive survival analyses were performed. Secondary outcomes included short-term efficacy and adverse events associated with first-line immunochemotherapy. RESULTS The median age of the 63 patients enrolled in our study was 63.0 years (40-80 years). The incidence of sarcopenia was 19.0% (12/63) in patients with extensive SCLC. Compared with non-sarcopenia patients, extensive-stage SCLC patients with sarcopenia were significantly older (69.0 vs. 62.0, P = 0.017), and had lower body mass index (BMI) (20.29 vs. 24.27, P < 0.001), hand grip strength (HGS) (20.42 vs. 30.75, P < 0.001), and albumin (35.9 vs. 41.40, P < 0.001). The objective response rate after two cycles of standard first-line immunochemotherapy in the sarcopenia group was lower than in the non-sarcopenia group (30.0 vs. 78.9%, P = 0.012). There was no significant difference in chemotherapy-related hematological toxicity between the two groups. During a median follow-up of 15 months (3-33 months), patients with extensive SCLC had a median OS of 24 months, with 1-year survival of 75% and 2-year survival of 52%, respectively. Compared to non-sarcopenia patients, the median OS in the sarcopenia group was significantly shorter (9 vs. 24 months, P = 0.0014). Multivariate Cox analysis showed that sarcopenia was an independent risk factor for OS in patients with extensive SCLC (HR = 4.993, 95%CI = 1.106-22.538, P = 0.037). CONCLUSIONS Patients with Extensive SCLC and sarcopenia had worse clinical outcomes and shorter OS. Sarcopenia is a prognostic factor affecting first-line treatment efficacy and long-term survival of patients with SCLC in the era of immunotherapy.
Collapse
Affiliation(s)
- Le Tian
- Department of Comprehensive Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia-Xin Huang
- Department of Comprehensive Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui Wan
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jie Zhang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xi Zhang
- Department of Comprehensive Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning Li
- Department of Comprehensive Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na Li
- Department of Comprehensive Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin-Qi Liu
- Department of Traditional Chinese Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Chen-Xin Song
- Department of Comprehensive Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin-Yi Wang
- Department of Medicine, Gesi (Hebei) Medical Research Institute, Langfang, China
| | - Lei Yu
- Department of Comprehensive Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shao-Ming Wang
- National Central Cancer Registry, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhi-Jie Wang
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Ming-Hua Cong
- Department of Comprehensive Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
76
|
Qiang M, Liu H, Yang L, Wang H, Guo R. Immunotherapy for small cell lung cancer: the current state and future trajectories. Discov Oncol 2024; 15:355. [PMID: 39152301 PMCID: PMC11329494 DOI: 10.1007/s12672-024-01119-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/21/2024] [Indexed: 08/19/2024] Open
Abstract
Small cell lung cancer (SCLC) constitutes approximately 10% to 15% of all lung cancer diagnoses and represents a pressing global public health challenge due to its high mortality rates. The efficacy of conventional treatments for SCLC is suboptimal, characterized by limited anti-tumoral effects and frequent relapses. In this context, emerging research has pivoted towards immunotherapy combined with chemotherapy, a rapidly advancing field that has shown promise in ameliorating the clinical outcomes of SCLC patients. Through originally developed for non-small cell lung cancer (NSCLC), these therapies have extended new treatment avenues for SCLC. Currently, a nexus of emerging hot-spot treatments has demonstrated significant therapeutic efficacy. Based on the amalgamation of chemotherapy and immunotherapy, and the development of new immunotherapy agents, the treatment of SCLC has seen the hoping future. Progress has been achieved in enhancing the tumor immune microenvironment through the concomitant use of chemotherapy, immunotherapy, and tyrosine kinase inhibitors (TKI), as evinced by emerging clinical trial data. Moreover, a tripartite approach involving immunotherapy, targeted therapy, and chemotherapy appears auspicious for future clinical applications. Overcoming resistance to post-immunotherapy regimens remains an urgent area of exploration. Finally, bispecific antibodies, adoptive cell transfer (ACT), oncolytic virus, monotherapy, including Delta-like ligand 3 (DLL3) and T cell immunoreceptor with Ig and ITIM domains (TIGIT), as well as precision medicine, may present a prospective route towards achieving curative outcomes in SCLC. This review aims to synthesize extant literature and highlight future directions in SCLC treatment, acknowledging the persistent challenges in the field. Furthermore, the continual development of novel therapeutic agents and technologies renders the future of SCLC treatment increasingly optimistic.
Collapse
Affiliation(s)
- Min Qiang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Hongyang Liu
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Lei Yang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Hong Wang
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Rui Guo
- Clinical Laboratory, The First Hospital of Jilin University, Jilin University, Changchun, China.
| |
Collapse
|
77
|
Lin S, Zhang Y, Yao J, Yang J, Qiu Y, Zhu Z, Hua H. DB-1314, a novel DLL3-targeting ADC with DNA topoisomerase I inhibitor, exhibits promising safety profile and therapeutic efficacy in preclinical small cell lung cancer models. J Transl Med 2024; 22:766. [PMID: 39143619 PMCID: PMC11323672 DOI: 10.1186/s12967-024-05568-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/03/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Delta-like ligand 3 (DLL3) is highly expressed on the cell surface of small cell lung cancer (SCLC), one of the most lethal malignancies, but minimally or not in normal tissues, making it an attractive target for SCLC. However, none of the DLL3-targeting antibody-drug conjugates (ADCs) have been approved for SCLC therapy yet. We developed DB-1314, the new anti-DLL3 ADC composed of a novel humanized anti-DLL3 monoclonal antibody (DB131401) conjugated with eight molecules of P1021 (topoisomerase I inhibitor), and described its preclinical profiles. METHODS The binding epitope for DB131401 and Rovalpituzumab was tested by biolayer interferometry. The binding affinity and specificity of DB-1314 to DLL3 and other homologous proteins were respectively measured by surface plasmon resonance and enzyme-linked immunosorbent assay. Internalization, bystander effects, and antibody-dependent cell-mediated cytotoxicity (ADCC) were assessed by respective assay. DLL3 was quantified by antibodies bound per cell assay and immunohistochemistry. In vitro and in vivo growth inhibition studies were evaluated in SCLC cell lines, and cell line/patient-derived xenograft models. The safety profile was measured in cynomolgus monkeys. RESULTS DB-1314 induces potent, durable, and dose-dependent antitumor effects in cells in vitro and in cell/patient-derived xenograft models in vivo. The killing activity of DB-1314 mechanically arises from P1021-induced DNA damage, whereby P1021 is delivered and released within tumor cells through DLL3-specific binding and efficient internalization. Bystander effects and ADCC also contribute to the antitumor activity of DB-1314. DB-1314 displays favorable pharmacokinetic and toxicokinetic profiles in rats and cynomolgus monkeys; besides, DB-1314 is well-tolerated at a dose of up to 60 mg/kg in monkeys. CONCLUSIONS These results suggest that DB-1314 may be a candidate ADC targeting DLL3 for the treatment of DLL3-positive SCLC, supporting further evaluation in the clinical setting.
Collapse
Affiliation(s)
- Shengchao Lin
- Department of Research and Development, Duality Biologics, LTD, Unite 1106 868 Yinghua Road, Unite, 1106, 201204, Shanghai, P.R. China.
| | - Yu Zhang
- Department of Research and Development, Duality Biologics, LTD, Unite 1106 868 Yinghua Road, Unite, 1106, 201204, Shanghai, P.R. China
| | - Jun Yao
- Department of Research and Development, Duality Biologics, LTD, Unite 1106 868 Yinghua Road, Unite, 1106, 201204, Shanghai, P.R. China
| | - Junjie Yang
- Department of Research and Development, Duality Biologics, LTD, Unite 1106 868 Yinghua Road, Unite, 1106, 201204, Shanghai, P.R. China
| | - Yang Qiu
- Department of Research and Development, Duality Biologics, LTD, Unite 1106 868 Yinghua Road, Unite, 1106, 201204, Shanghai, P.R. China
| | - Zhongyuan Zhu
- Department of Research and Development, Duality Biologics, LTD, Unite 1106 868 Yinghua Road, Unite, 1106, 201204, Shanghai, P.R. China
| | - Haiqing Hua
- Department of Research and Development, Duality Biologics, LTD, Unite 1106 868 Yinghua Road, Unite, 1106, 201204, Shanghai, P.R. China.
| |
Collapse
|
78
|
Fei X, Xue JW, Wu JZ, Yang CY, Wang KJ, Ma Q. Promising therapy for neuroendocrine prostate cancer: current status and future directions. Ther Adv Med Oncol 2024; 16:17588359241269676. [PMID: 39131727 PMCID: PMC11311189 DOI: 10.1177/17588359241269676] [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: 04/09/2024] [Accepted: 06/24/2024] [Indexed: 08/13/2024] Open
Abstract
Neuroendocrine prostate cancer (NEPC) is a highly aggressive variant of castration-resistant prostate cancer. It is characterized by low or no expression of the androgen receptor (AR), activation of AR-independent signaling, and increased neuroendocrine phenotype. Most of NEPC is induced by treatment of androgen deprivation therapy and androgen receptor pathway inhibitors (ARPIs). Currently, the treatment of NEPC follows the treatment strategy for small-cell lung cancer, lacking effective drugs and specific treatment options. This review summarizes potential novel targets and therapies for NEPC treatment, including epigenetic regulators (zeste homolog 2 inhibitors, lysine-specific demethylase 1 inhibitors), aurora kinase A inhibitors, poly-ADP-ribose polymerase inhibitors, delta-like ligand 3 targeted therapies, a combination of immunotherapies, etc. Other promising targets and future directions are also discussed in this review. These novel targets and therapies may provide new opportunities for the treatment of NEPC.
Collapse
Affiliation(s)
- Xin Fei
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
| | - Jia-Wei Xue
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- Department of Urology, The First Hospital of Ninghai, Ningbo, China
| | - Ji-zhongrong Wu
- Health Science Center, Ningbo University, Ningbo, Zhejiang, China
- Department of Urology, Shengzhou People’s Hospital, Shaoxing, China
| | - Chong-Yi Yang
- Department of Urology, The First Hospital of Ninghai, 142 Taoyuan Middle Road, Yuelong Street, Ninghai county, Ningbo, Zhejiang 315699, China
| | - Ke-Jie Wang
- Comprehensive Genitourinary Cancer Center, The First Affiliated Hospital of Ningbo University, 52, Liuting Street, Haishu District, Ningbo, Zhejiang 315010, China
| | - Qi Ma
- Department of Urology, the First Affiliated Hospital of Ningbo University, 52, Liuting Street, Haishu District,Ningbo, Zhejiang 315010, China
- Comprehensive Genitourinary Cancer Center, The First Affiliated Hospital of Ningbo University, 52, Liuting Street, Haishu District, Ningbo, Zhejiang 315010, China
- Yi-Huan Genitourinary Cancer Group, 52, Liuting Street, Haishu District, Ningbo,Zhejiang 315010, China
| |
Collapse
|
79
|
Zhang J, Zeng X, Guo Q, Sheng Z, Chen Y, Wan S, Zhang L, Zhang P. Small cell lung cancer: emerging subtypes, signaling pathways, and therapeutic vulnerabilities. Exp Hematol Oncol 2024; 13:78. [PMID: 39103941 DOI: 10.1186/s40164-024-00548-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 07/27/2024] [Indexed: 08/07/2024] Open
Abstract
Small cell lung cancer (SCLC) is a recalcitrant cancer characterized by early metastasis, rapid tumor growth and poor prognosis. In recent decades, the epidemiology, initiation and mutation characteristics of SCLC, as well as abnormal signaling pathways contributing to its progression, have been widely studied. Despite extensive investigation, fewer drugs have been approved for SCLC. Recent advancements in multi-omics studies have revealed diverse classifications of SCLC that are featured by distinct characteristics and therapeutic vulnerabilities. With the accumulation of SCLC samples, different subtypes of SCLC and specific treatments for these subtypes were further explored. The identification of different molecular subtypes has opened up novel avenues for the treatment of SCLC; however, the inconsistent and uncertain classification of SCLC has hindered the translation from basic research to clinical applications. Therefore, a comprehensives review is essential to conclude these emerging subtypes and related drugs targeting specific therapeutic vulnerabilities within abnormal signaling pathways. In this current review, we summarized the epidemiology, risk factors, mutation characteristics of and classification, related molecular pathways and treatments for SCLC. We hope that this review will facilitate the translation of molecular subtyping of SCLC from theory to clinical application.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| | - Xiaoping Zeng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Qiji Guo
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Zhenxin Sheng
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Yan Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Shiyue Wan
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Lele Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China
| | - Peng Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, 200433, China.
| |
Collapse
|
80
|
Luo Z, Li Y, Xu B, Yu T, Luo M, You P, Niu X, Li J. Overexpression of ESYT3 improves radioimmune responses through activating cGAS-STING pathway in lung adenocarcinoma. Exp Hematol Oncol 2024; 13:77. [PMID: 39103908 DOI: 10.1186/s40164-024-00546-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 07/26/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Radiotherapy can modulate systemic antitumor immunity, while immune status in the tumor microenvironment also influences the efficacy of radiotherapy, but relevant molecular mechanisms are poorly understood in lung adenocarcinoma (LUAD). METHODS In this study, we innovatively proposed a radiotherapy response classification for LUAD, and discovered ESYT3 served as a tumor suppressor and radioimmune response sensitizer. ESYT3 expression was measured both in radioresistant and radiosensitive LUAD tissues and cells. The influence of ESYT3 on radiotherapy sensitivity and resistance was then investigated. Interaction between ESYT3 and STING was evaluated through multiple immunofluorescent staining and coimmunoprecipitation, and downstream molecules were further analyzed. In vivo models were constructed to assess the combination treatment efficacy of ESYT3 overexpression with radiotherapy. RESULTS We found that radioresistant subtype presented immunosuppressive state and activation of DNA damage repair pathways than radiosensitive subtype. ESYT3 expression was remarkably attenuated both in radioresistant LUAD tissues and cells. Clinically, low ESYT3 expression was linked with radioresistance. Overexpression of ESYT3 enabled to alleviate radioresistance, and sensitize LUAD cells to DNA damage induced by irradiation. Mechanically, ESYT3 directly interacted with STING, and activated cGAS-STING signaling, subsequently increasing the generation of type I IFNs as well as downstream chemokines CCL5 and CXCL10, thus improving radioimmune responses. The combination treatment of ESYT3 overexpression with radiotherapy had a synergistic anticancer effect in vitro and in vivo. CONCLUSIONS In summary, low ESYT3 expression confers resistance to radiotherapy in LUAD, and its overexpression can improve radioimmune responses through activating cGAS-STING-dependent pathway, thus providing an alternative combination therapeutic strategy for LUAD patients.
Collapse
Affiliation(s)
- Zan Luo
- Department of Radiation Oncology, The Second Affiliated Hospital of Nanchang Medical College), Jiangxi Cancer Hospital, Nanchang, Jiangxi, 330029, China
- Jiangxi Key Laboratory of Oncology, The Second Affiliated Hospital of Nanchang Medical College), Jiangxi Cancer Hospital, Nanchang, Jiangxi, 330029, China
| | - Ying Li
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430079, Hubei, China
| | - Bin Xu
- Laboratory of Tumor Metastasis, The Second Affiliated Hospital of Nanchang Medical College), Jiangxi Health Committee Key (JHCK), Jiangxi Cancer Hospital, Nanchang, Jiangxi, 330029, China.
| | - Tenghua Yu
- Department of Breast Surgery, The Second Affiliated Hospital of Nanchang Medical College), Jiangxi Cancer Hospital, Nanchang, Jiangxi, 330029, China
| | - Mingming Luo
- Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College), Jiangxi Cancer Hospital, Nanchang, Jiangxi, 330029, China
| | - PeiMeng You
- Department of Radiation Oncology, Jiangxi Key Laboratory of Translational Cancer Research, Cancer Hospital of Nanchang University, Jiangxi Cancer Hospital of Nanchang University), Nanchang, Jiangxi, 330029, China
| | - Xing Niu
- Experimental Center of BIOQGene, YuanDong International Academy Of Life Sciences, Hong Kong, Hong Kong, 999077, China.
- China Medical University, Shenyang, Liaoning, 110122, China.
| | - Junyu Li
- Department of Radiation Oncology, The Second Affiliated Hospital of Nanchang Medical College), Jiangxi Cancer Hospital, Nanchang, Jiangxi, 330029, China.
- Jiangxi Key Laboratory of Oncology, The Second Affiliated Hospital of Nanchang Medical College), Jiangxi Cancer Hospital, Nanchang, Jiangxi, 330029, China.
| |
Collapse
|
81
|
Ilhan Y, Ucar G, Baser MN, Guzel HG, Efil SC, Demir B, Ercan Uzundal D, Karacelik T, Sever N, Balcik OY, Arvas H, Karadag I, Kadioglu A, Ekinci ÖB, Karacin C, Urakci Z, Kostek O, Karakurt Eryilmaz M, Yazici O, Sendur MAN, Ozturk B, Uncu D, Ergun Y. Efficacy and safety of G-CSF prophylaxis in patients with extensive-stage small cell lung cancer receiving chemoimmunotherapy. Expert Opin Pharmacother 2024; 25:1555-1563. [PMID: 39115275 DOI: 10.1080/14656566.2024.2391007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 08/07/2024] [Indexed: 08/13/2024]
Abstract
OBJECTIVES We aimed to evaluate the efficacy and safety of granulocyte-colony stimulating factor (G-CSF) prophylaxis during chemoimmunotherapy with carboplatin plus etoposide and atezolizumab in extensive-stage small cell lung cancer (ES-SCLC). METHODS This retrospective, multicenter study enrolled ES-SCLC patients receiving carboplatin plus etoposide and atezolizumab, categorized into G-CSF and non-G-CSF groups. Demographic and disease-related data were collected. Response rates, progression-free survival (PFS), overall survival (OS), and toxicity were analyzed. RESULTS Of 119 patients (median age: 63 years), the overall response rate (ORR) and disease control rate (DCR) were 72.3% and 81.5%, respectively. In the G-CSF group, the ORR was 76.4% compared to 60.0% in the non-G-CSF group (p = 0.33), and the DCR was 85.4% versus 70.0%, respectively (p = 0.46). Median PFS was 8.3 months (95% CI, 6.8-9.8) in the G-CSF group and 6.8 months (95% CI, 6.2-7.5) in the non-G-CSF group (p = 0.24). Median OS was 13.8 months (95% CI, 9.6-18.1) for the G-CSF group and 10.6 months (95% CI, 7.9-13.3) for the non-G-CSF group (p = 0.47). Grade 3 ≥ adverse events were similar between groups (49.4% vs. 33.3%, respectively, p = 0.12). CONCLUSION G-CSF prophylaxis can be safely used in ES-SCLC patients undergoing carboplatin plus etoposide and atezolizumab regimen without significantly altering efficacy or increasing toxicity.
Collapse
Affiliation(s)
- Yusuf Ilhan
- Department of Medical Oncology, Antalya City Hospital, Antalya, Turkey
| | - Gokhan Ucar
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Mehmet Nuri Baser
- Department of Medical Oncology, Adnan Menderes University, Aydın, Turkey
| | - Halil Goksel Guzel
- Antalya Education and Research Hospital, Department of Medical Oncology, Health Sciences University, Antalya, Turkey
| | - Safa Can Efil
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Bilgin Demir
- Department of Medical Oncology, Adnan Menderes University, Aydın, Turkey
| | - Duygu Ercan Uzundal
- Faculty of Medicine, Department of Medical Oncology, Gazi University, Ankara, Turkey
| | - Tuba Karacelik
- Faculty of Medicine, Department of Medical Oncology, Necmettin Erbakan University, Konya, Turkey
| | - Nadiye Sever
- Faculty of Medicine, Department of Medical Oncology, Marmara University, Istanbul, Turkey
| | - Onur Yazdan Balcik
- Department of Medical Oncology, Mardin Training and Research Hospital, Mardin, Turkey
| | - Hayati Arvas
- Faculty of Medicine, Department of Medical Oncology, Dicle University, Diyarbakır, Turkey
| | - Ibrahim Karadag
- Erol Olcok Education and Research Hospital, Department of Medical Oncology, Hitit University, Corum, Turkey
| | - Ahmet Kadioglu
- Department of Medical Oncology, University of Health Sciences, Dr. Abdurrahman Yurtaslan Oncology Training and Research Hospital, Ankara, Turkey
| | - Ömer Burak Ekinci
- Department of Medical Oncology, Cemil Tascioglu City Hospital, Istanbul, Turkey
| | - Cengiz Karacin
- Department of Medical Oncology, University of Health Sciences, Dr. Abdurrahman Yurtaslan Oncology Training and Research Hospital, Ankara, Turkey
| | - Zuhat Urakci
- Faculty of Medicine, Department of Medical Oncology, Dicle University, Diyarbakır, Turkey
| | - Osman Kostek
- Faculty of Medicine, Department of Medical Oncology, Marmara University, Istanbul, Turkey
| | - Melek Karakurt Eryilmaz
- Faculty of Medicine, Department of Medical Oncology, Necmettin Erbakan University, Konya, Turkey
| | - Ozan Yazici
- Faculty of Medicine, Department of Medical Oncology, Gazi University, Ankara, Turkey
| | | | - Banu Ozturk
- Antalya Education and Research Hospital, Department of Medical Oncology, Health Sciences University, Antalya, Turkey
| | - Dogan Uncu
- Department of Medical Oncology, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Yakup Ergun
- Department of Medical Oncology, Antalya City Hospital, Antalya, Turkey
| |
Collapse
|
82
|
Hayne D, Ong K, Swarbrick N, McCombie SP, Moe A, Hawks C, Viswambaram P, Conduit C, Liow E, Spalding L, Lim J, Ferguson T, Meehan K, Davis ID, Redfern AD. The SUB-urothelial DUrvalumab InjEction-1 (SUBDUE-1) trial: first-in-human trial in patients with bladder cancer. BJU Int 2024; 134:283-290. [PMID: 38469652 DOI: 10.1111/bju.16325] [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] [Indexed: 03/13/2024]
Abstract
OBJECTIVES To assess the safety of sub-urothelial injection of durvalumab and examine the impact on tissue and circulating immune cell populations. PATIENTS AND METHODS The patients were chemotherapy and immunotherapy naïve (bacille Calmette-Guérin allowed) with non-metastatic muscle-invasive bladder cancer or non-muscle-invasive bladder cancer planned for radical cystectomy (RC). The study was a Phase Ib 3 + 3 dose-escalation design with sub-urothelial injection of durvalumab at three pre-determined doses (25, 75, 150 mg) diluted in 25 mL normal saline, injected at 25 locations (25 × 1 mL injections), at least 2 weeks before RC. RESULTS A total of 11 patients were recruited (10 male, one female). No significant changes were reported on American Urological Association Symptom Score or O'Leary Interstitial Cystitis Scale. In all, 14 adverse events (AEs) were reported (10 Grade 1, three Grade 2, one Grade 3), none considered immune-related. No Grade 4 or 5 AEs were recorded. All the patients underwent RC. Tissue immune populations changed following durvalumab injection (P = 0.012), with a statistically significant increase in M2-macrophage (CD163) when comparing the 25-150 mg dose (P = 0.021). Basal/mixed cancers showed a larger CD163 increase than luminal cancers (P = 0.033). CONCLUSION Sub-urothelial injection of durvalumab is feasible and safe without immune-related AEs and shows local immunological effects.
Collapse
Affiliation(s)
- Dickon Hayne
- UWA Medical School, University of Western Australia, Crawley, Western Australia, Australia
- South Metropolitan Health Service, Murdoch, Western Australia, Australia
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, New South Wales, Australia
| | - Katherine Ong
- South Metropolitan Health Service, Murdoch, Western Australia, Australia
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, New South Wales, Australia
| | - Nicole Swarbrick
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia, Australia
- Department of Anatomical Pathology, PathWest Laboratory Medicine, Perth, Western Australia, Australia
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, New South Wales, Australia
| | - Steve P McCombie
- UWA Medical School, University of Western Australia, Crawley, Western Australia, Australia
- South Metropolitan Health Service, Murdoch, Western Australia, Australia
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, New South Wales, Australia
| | - Andrew Moe
- UWA Medical School, University of Western Australia, Crawley, Western Australia, Australia
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, New South Wales, Australia
| | - Cynthia Hawks
- UWA Medical School, University of Western Australia, Crawley, Western Australia, Australia
- South Metropolitan Health Service, Murdoch, Western Australia, Australia
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, New South Wales, Australia
| | - Pravin Viswambaram
- UWA Medical School, University of Western Australia, Crawley, Western Australia, Australia
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, New South Wales, Australia
| | - Ciara Conduit
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, New South Wales, Australia
- Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Melbourne, Victoria, Australia
- Personalised Oncology, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Elizabeth Liow
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, New South Wales, Australia
- Monash Health, Clayton, Victoria, Australia
| | - Lisa Spalding
- Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
- University of Western Australia Centre for Medical Research, Nedlands, Western Australia, Australia
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, New South Wales, Australia
| | - Jayne Lim
- UWA Medical School, University of Western Australia, Crawley, Western Australia, Australia
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, New South Wales, Australia
| | - Thomas Ferguson
- South Metropolitan Health Service, Murdoch, Western Australia, Australia
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, New South Wales, Australia
| | - Katie Meehan
- UWA Medical School, University of Western Australia, Crawley, Western Australia, Australia
- Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, New South Wales, Australia
| | - Ian D Davis
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, New South Wales, Australia
- Monash University, Melbourne, Victoria, Australia
- Eastern Health, Melbourne, Victoria, Australia
| | - Andrew D Redfern
- UWA Medical School, University of Western Australia, Crawley, Western Australia, Australia
- South Metropolitan Health Service, Murdoch, Western Australia, Australia
- Australian and New Zealand Urogenital and Prostate (ANZUP) Cancer Trials Group, Camperdown, New South Wales, Australia
| |
Collapse
|
83
|
Ciesiołkiewicz A, Lizandra Perez J, Skalniak L, Noceń P, Berlicki Ł. Miniprotein engineering for inhibition of PD-1/PD-L1 interaction. Protein Sci 2024; 33:e5106. [PMID: 39012010 PMCID: PMC11250529 DOI: 10.1002/pro.5106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 06/07/2024] [Accepted: 06/23/2024] [Indexed: 07/17/2024]
Abstract
Miniproteins constitute an excellent basis for the development of structurally demanding functional molecules. The engrailed homeodomain, a three-helix-containing miniprotein, was applied as a scaffold for constructing programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) interaction inhibitors. PD-L1 binders were initially designed using the computer-aided approach and subsequently optimized iteratively. The conformational stability was assessed for each obtained miniprotein using circular dichroism spectroscopy, indicating that numerous mutations could be introduced. The formation of a sizable hydrophobic surface at the inhibitor that fits the molecular target imposed the necessity for the incorporation of additional charged amino acid residues to retain its appropriate solubility. Finally, the miniprotein effectively binding to PD-L1 (KD = 51.4 nM) that inhibits PD-1/PD-L1 interaction in cell-based studies with EC50 = 3.9 μM, was discovered.
Collapse
Affiliation(s)
| | - Juan Lizandra Perez
- Department of Bioorganic ChemistryWrocław University of Science and TechnologyWrocławPoland
| | | | - Paweł Noceń
- Department of Bioorganic ChemistryWrocław University of Science and TechnologyWrocławPoland
| | - Łukasz Berlicki
- Department of Bioorganic ChemistryWrocław University of Science and TechnologyWrocławPoland
| |
Collapse
|
84
|
Pons-Tostivint E, Ezzedine R, Goronflot T, Crequit P, Chatellier T, Raimbourg J, Bennouna J, Giroux Leprieur E, Porte M. Second-line treatment outcomes after first-line chemotherapy plus immunotherapy in Extensive-Stage small cell lung cancer (ES-SCLC) patients: A large French multicenter study. Lung Cancer 2024; 194:107887. [PMID: 38991282 DOI: 10.1016/j.lungcan.2024.107887] [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: 04/07/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/13/2024]
Abstract
INTRODUCTION Chemotherapy combined with immunotherapy (CT-IO) is the standard treatment for patients with Extensive-Stage Small Cell Lung Cancer (ES-SCLC). This study evaluates the effectiveness of second-line (2L) following CT-IO. PATIENTS AND METHODS All patients from 10 centers who received a 2L after a first-line CT-IO were included. They were divided into 3 groups: platinum-based, lurbinectedin or others (topotecan, CAV, taxanes). We assessed overall survival (OS) and 2L progression-free survival (2L-PFS) according to treatment and platinum free-interval (PFI) < or ≥ 90 days. RESULTS Among 82 patients included, median age was 67.0 years, 29.3 % had a Performans Status ≥ 2, 36.6 % had brain progression, 69.5 % were considered "platine-sensitive" and 30.5 % "platine-resistant" (PFI ≥ or < 90 days, respectively). As 2L, 37/82 patients (45.1 %) received platinum-doublet, 21/82 (25.6 %) lurbinectedin and 24/82 (29.3 %) others. Patients with a PFI ≥ 90 days received mainly platinum-based rechallenge (34/57, 59.6 %). With a median follow-up of 18.5 months, the median OS was 5.0 months (95 %CI, 1.5-7.9) / 6.8 months (95 %CI, 5.5-8.7) for platinum-resistant / sensitive, respectively (log rank p = 0.017). The median 2L-PFS was 1.9 months (95 %CI, 1.2-4.7) / 3.9 months (95 %CI, 2.9-6.0) for platinum-resistant / sensitive, respectively. Median OS was 8.1 (95 %CI, 6.3-12.9) / 4.9 (95 %CI, 3.7-6.8) / 5.1 months (95 %CI, 2.5-7.8) with platinum rechallenge / lurbinectedin / others, respectively (p = 0.017). Median 2L-PFS was 4.6 (95 %CI, 3.9-7.2) / 2.7 (95 %CI, 1.6-3.9) / 2.2 months (95 %CI, 1.5-4.1) with platinum rechallenge / lurbinectedin / others, respectively (p = 0.025). DISCUSSION Platinum-based rechallenge after a first-line CT-IO showed promising results despite particularly unfavorable characteristics within our real-word population. Lurbinectedin when used after IO demonstrated as low efficacy as other platinum-free regimens.
Collapse
Affiliation(s)
- Elvire Pons-Tostivint
- Nantes Université, Centre Hospitalier Universitaire Nantes, Medical oncology, F-44000 Nantes, France; Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d'Angers, CRCI2NA Nantes, France.
| | - Remy Ezzedine
- Service de pneumologie et oncologie thoracique, CHU d'Ambroise-Paré, Boulogne-Billancourt, France
| | - Thomas Goronflot
- Nantes Université, CHU Nantes, Pôle Hospitalo-Universitaire 11 : Santé Publique, Clinique des données, INSERM, CIC 1413, F-44000 Nantes, France
| | - Perrine Crequit
- Department of Medical Oncology, Hospital Foch, Suresnes, France
| | - Thierry Chatellier
- Medical Oncology Unit, Clinique Mutualiste de l'Estuaire, Saint-Nazaire, France
| | - Judith Raimbourg
- Department of Medical Oncology, Comprehensive Cancer Center, Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - Jaafar Bennouna
- Department of Medical Oncology, Hospital Foch, Suresnes, France
| | - Etienne Giroux Leprieur
- Service de pneumologie et oncologie thoracique, CHU d'Ambroise-Paré, Boulogne-Billancourt, France
| | - Marie Porte
- Nantes Université, Centre Hospitalier Universitaire Nantes, Medical oncology, F-44000 Nantes, France
| |
Collapse
|
85
|
Tang L, Tian G, Li N. Current dilemma and future directions over prophylactic cranial irradiation in SCLC: a systematic review in MRI and immunotherapy era. Front Oncol 2024; 14:1382220. [PMID: 39139283 PMCID: PMC11319250 DOI: 10.3389/fonc.2024.1382220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Small cell lung cancer (SCLC) is the most malignant pathological type of lung cancer with the highest mortality, and the incidence of brain metastasis (BM) is in high frequency. So far, prophylactic cranial irradiation (PCI) has been suggested as an effective treatment for preventing brain metastasis of SCLC. PCI has long been applied to limited-stage SCLC (LS-SCLC) patients who have achieved complete remission after radiotherapy and chemotherapy as a standard treatment. However, the neurocognitive decline is a major concern surrounding PCI. New therapeutic approaches targeting PCI-induced neurotoxicity, including hippocampal protection or memantine, have been increasingly incorporated into the therapeutic interventions of PCI. Helical tomotherapy, RapidArc, and Volumetric-modulated arc therapy (VMAT) with a head-tilting baseplate are recommended for hippocampal protection. Besides, in the MRI and immunotherapy era, the significance of PCI in SCLC patients is controversial. SCLC patients with PCI should be recruited in clinical trials since this is the only way to improve the existing standard of care. This review summarizes the current therapeutic strategy and dilemma over PCI for SCLC, providing a theoretical basis for clinical decision-making and suggestions for PCI practice in clinical.
Collapse
Affiliation(s)
| | | | - Nan Li
- Department of Radiation Oncology, the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
86
|
Liu Y, Zhu J, Du TY, Liu XH, Xin Y, Wang Y, Wang YP, Xu JH, Chen Y, Wei HF, Cheng Y. Navigating first-line therapies for extensive-stage small-cell lung cancer: a frequentist network meta-analysis and systematic review. Future Oncol 2024; 20:2109-2122. [PMID: 39072397 PMCID: PMC11497961 DOI: 10.1080/14796694.2024.2376514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024] Open
Abstract
Aim: To identify the optimal first-line treatment for patients with extensive-stage small-cell lung cancer (ES-SCLC).Materials & methods: We conducted a network meta-analysis (CRD42023486863) to systematically evaluate the efficacy and safety of eight first-line treatment regimens for ES-SCLC, including 15 clinical trials.Results: Our analysis showed that the PD-1/PD-L1 + etoposide combined with platinum (EP) and PD-L1 + vascular endothelial growth factor (VEGF) + EP regimens significantly enhanced overall survival and progression-free survival, with subgroup analysis revealing that serplulimab ranked as the most promising option for improving overall survival. Integrating anti-angiogenesis drugs into immunochemotherapy presents potential benefits, with an increased incidence of adverse events necessitating further investigation.Conclusion: Our findings offer valuable insights for future research and for developing more effective treatment strategies for ES-SCLC, underscoring the critical need for continued innovation in this therapeutic area.
Collapse
Affiliation(s)
- Ying Liu
- Department of Oncology, Jilin Cancer Hospital, 1066 Jinhu Rd, Jilin, Changchun, 130000, China
| | - Jing Zhu
- Department of Oncology, Jilin Cancer Hospital, 1066 Jinhu Rd, Jilin, Changchun, 130000, China
| | - Tian-Ying Du
- Department of Oncology, Jilin Cancer Hospital, 1066 Jinhu Rd, Jilin, Changchun, 130000, China
| | - Xian-Hong Liu
- Department of Oncology, Jilin Cancer Hospital, 1066 Jinhu Rd, Jilin, Changchun, 130000, China
| | - Ying Xin
- Department of Oncology, Jilin Cancer Hospital, 1066 Jinhu Rd, Jilin, Changchun, 130000, China
| | - Ying Wang
- Department of Oncology, Jilin Cancer Hospital, 1066 Jinhu Rd, Jilin, Changchun, 130000, China
| | - Yan-Ping Wang
- Department of Oncology, Jilin Cancer Hospital, 1066 Jinhu Rd, Jilin, Changchun, 130000, China
| | - Jin-Hua Xu
- Department of Oncology, Jilin Cancer Hospital, 1066 Jinhu Rd, Jilin, Changchun, 130000, China
| | - Yan Chen
- Department of Oncology, Jilin Cancer Hospital, 1066 Jinhu Rd, Jilin, Changchun, 130000, China
| | - Hua-Fang Wei
- Department of Oncology, Jilin Cancer Hospital, 1066 Jinhu Rd, Jilin, Changchun, 130000, China
| | - Ying Cheng
- Department of Oncology, Jilin Cancer Hospital, 1066 Jinhu Rd, Jilin, Changchun, 130000, China
| |
Collapse
|
87
|
Zugazagoitia J, Osma H, Baena J, Ucero AC, Paz-Ares L. Facts and Hopes on Cancer Immunotherapy for Small Cell Lung Cancer. Clin Cancer Res 2024; 30:2872-2883. [PMID: 38630789 DOI: 10.1158/1078-0432.ccr-23-1159] [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: 10/09/2023] [Revised: 12/08/2023] [Accepted: 02/01/2024] [Indexed: 04/19/2024]
Abstract
Platinum-based chemotherapy plus PD1 axis blockade is the standard of care in the first-line treatment of extensive-stage small cell lung cancer (SCLC). Despite the robust and consistent increase in long-term survival with PD1 axis inhibition, the magnitude of the benefit from immunotherapy seems lower than that for other solid tumors. Several immune evasive mechanisms have been shown to be prominently altered in human SCLC, including T-cell exclusion, downregulation of components of the MHC class I antigen processing and presentation machinery, or upregulation of macrophage inhibitory checkpoints, among others. New immunotherapies aiming to target some of these dominant immune suppressive features are being intensively evaluated preclinically and clinically in SCLC. They include strategies to enhance the efficacy and/or reverse features that promote intrinsic resistance to PD1 axis inhibition (e.g., restoring MHC class I deficiency and targeting DNA damage response) and novel immunomodulatory agents beyond T-cell checkpoint blockers (e.g., T cell-redirecting strategies, antibody-drug conjugates, or macrophage checkpoint blockers). Among them, delta-like ligand 3-targeted bispecific T-cell engagers have shown the most compelling preliminary evidence of clinical efficacy and hold promise as therapies that might contribute to further improve patient outcomes in this disease. In this study, we first provide a brief overview of key tumor microenvironment features of human SCLC. Then, we update the current clinical evidence with immune checkpoint blockade and review other emerging immunotherapy strategies that are gaining increasing attention in SCLC. We finally summarize our future perspective on immunotherapy and precision oncology for this disease.
Collapse
Affiliation(s)
- Jon Zugazagoitia
- Department of Medical Oncology, 12 de Octubre Hospital, Madrid, Spain
- Lung Cancer Clinical Research Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- CIBERONC, Carlos III Research Institute, Madrid, Spain
| | - Handerson Osma
- Department of Medical Oncology, Clinica Vida and Hospital Alma Mater de Antioquia, Medellín, Colombia
- Asociación Colombiana de Hematología y Oncología (ACHO), Bogotá DC, Colombia
| | - Javier Baena
- Department of Medical Oncology, 12 de Octubre Hospital, Madrid, Spain
- Lung Cancer Clinical Research Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Alvaro C Ucero
- Department of Medical Oncology, 12 de Octubre Hospital, Madrid, Spain
- Lung Cancer Clinical Research Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- CIBERONC, Carlos III Research Institute, Madrid, Spain
- Department of Medicine, Complutense University, Madrid, Spain
- Department of Physiology, Complutense University, Madrid, Spain
| | - Luis Paz-Ares
- Department of Medical Oncology, 12 de Octubre Hospital, Madrid, Spain
- Lung Cancer Clinical Research Group, Spanish National Cancer Research Center (CNIO), Madrid, Spain
- CIBERONC, Carlos III Research Institute, Madrid, Spain
- Department of Medicine, Complutense University, Madrid, Spain
- Department of Physiology, Complutense University, Madrid, Spain
| |
Collapse
|
88
|
Yi L, Zhou Z, Zeng X, Tan C, Liu Q. First-line treatments for extensive-stage small-cell lung cancer with immune checkpoint inhibitors plus chemotherapy: a China-based cost-effectiveness analysis. Front Immunol 2024; 15:1408928. [PMID: 39035009 PMCID: PMC11257869 DOI: 10.3389/fimmu.2024.1408928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/18/2024] [Indexed: 07/23/2024] Open
Abstract
Objective To determine the cost-effectiveness of imported immune checkpoint inhibitors (ICIs) such as atezolizumab and durvalumab, and domestic ICIs like serplulimab and adebrelimab, in combination with chemotherapy for extensive-stage small cell lung cancer (ES-SCLC) in China. Methods Using a 21-day cycle length and a 20-year time horizon, a Markov model was established to compare the clinical and economic outcomes of five first-line ICIs plus chemotherapy versus chemotherapy alone, as well as against each other, from the perspective of the Chinese healthcare system. Transition probabilities were estimated by combining the results of the CAPSTONE-1 trial and a published network meta-analysis. Cost and health state utilities were collected from multiple sources. Both cost and effectiveness outcomes were discounted at a rate of 5% annually. The primary model output was incremental cost-effectiveness ratios (ICERs). A series of sensitivity analyses were preformed to assess the robustness of the model. Results In the base-case analysis, the addition of first-line ICIs to chemotherapy resulted in the ICERs ranged from $80,425.31/QALY to $812,415.46/QALY, which exceeded the willing-to-pay threshold set for the model. When comparing these first-line immunochemotherapy strategies, serplulimab plus chemotherapy had the highest QALYs of 1.51286 and the second lowest costs of $60,519.52, making it is the most cost-effective strategy. Our subgroup-level analysis yielded results that are consistent with the base-case analysis. The sensitivity analysis results confirmed the validity and reliability of the model. Conclusion In China, the combination of fist-line ICIs plus chemotherapy were not considered cost-effective when compared to chemotherapy alone. However, when these fist-line immunochemotherapy strategies were compared with each other, first-line serplulimab plus chemotherapy consistently demonstrated superiority in terms of cost-effectiveness. Reducing the cost of serplulimab per 4.5 mg/kg would be a realistic step towards making first-line serplulimab plus chemotherapy more accessible and cost-effective.
Collapse
Affiliation(s)
- Lidan Yi
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhen Zhou
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Xiaohui Zeng
- Positron Emission Tomography - Computed Tomography (PET-CT) Center, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Chongqing Tan
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Qiao Liu
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
89
|
Zhang S, Li S, Cheng Y. The efficacy and safety of immunotherapy as first-line treatment for extensive-stage small cell lung cancer: evaluating based on reconstructed individual patient data. Front Oncol 2024; 14:1371313. [PMID: 39026980 PMCID: PMC11254656 DOI: 10.3389/fonc.2024.1371313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Objective Selecting between programmed cell death ligand 1 (PD-L1) inhibitor or programmed cell death 1 (PD-1) inhibitor plus chemotherapy as first-line treatment for extensive-stage small cell lung cancer (ES-SCLC) patients urgently needs to be answered. Methods Eligible phase 3 randomized clinical trials evaluating regimens based on PD-1/PD-L1 inhibitors as first-line treatment in ES-SCLC patients were systematically searched on the PubMed and Cochrane Library databases and major international conferences from 01/01/2018 to 18/09/2023. The individual patient data (IPD) were recuperated from the Kaplan-Meier curves of the overall survival (OS) and progression-free survival (PFS) of the included studies using the IPDfromKM method. The reconstructed data were pooled into unified arms, including the PD-L1 inhibitor plus chemotherapy group (PD-L1 group), PD-1 inhibitor plus chemotherapy group (PD-1 group), and PD-1 (L1) inhibitor and chemotherapy plus other (anlotinib group, tiragolumab group, and tremelimumab group). Subsequently, the PD-L1 group was indirectly compared with the other groups. A standard statistical analysis was conducted using the "survival" package for the time-to-event endpoint. The primary outcomes were the OS and PFS of the PD-L1 group and the PD-1 inhibitor group. The secondary outcomes included safety and the 12- and 24-month restricted mean survival time (RMST) of the PD-L1 group and PD-1 group. Results A total of 9 studies including 11 immunotherapy cohorts were included. No significant difference in PFS (hazard ratio [HR]: 0.96, 95% confidence interval [CI]: 0.86-1.06), OS (HR: 0.94, 95% CI: 0.84-1.05), and 12-month and 24-month RMST for OS (P = 0.198 and P = 0.216, respectively) was observed between the PD-L1 group and the PD-1 group. In contrast, the anlotinib group showed significantly better OS (HR: 0.70, 95% CI: 0.55-0.89), PFS (HR: 0.69, 95% CI: 0.58-0.83), and RMST for OS compared to the PD-L1 group. The tiragolumab group showed similar efficacy to the PD-L1 group. However, the tremelimumab group exhibited inferior efficacy than the PD-L1 group. The incidence of ≥grade 3 treatment-emergent adverse events (TEAEs) was significantly higher in the PD-1 group compared to the PD-L1 group (85.4% vs. 69.6%, P <.001), whereas the incidence of irAEs was similar between the two groups. Conclusion This reconstructed IPD analysis revealed that PD-1 inhibitors plus chemotherapy achieved similar efficacy to PD-L1 inhibitors plus chemotherapy as first-line treatment in ES-SCLC patients, whereas PD-L1 inhibitors plus chemotherapy had a better safety profile.
Collapse
Affiliation(s)
- Shuang Zhang
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun, China
- Clinical Research Big Data Center, Jilin Cancer Hospital, Changchun, China
| | - Shuang Li
- Clinical Research Big Data Center, Jilin Cancer Hospital, Changchun, China
| | - Ying Cheng
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun, China
| |
Collapse
|
90
|
Shrestha P, Kao S, Cheung VK, Cooper WA, van Zandwijk N, Rasko JEJ, Yeo D. Circulating tumor cells: advancing personalized therapy in small cell lung cancer patients. Mol Oncol 2024. [PMID: 38956984 DOI: 10.1002/1878-0261.13696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 03/27/2024] [Accepted: 06/20/2024] [Indexed: 07/04/2024] Open
Abstract
Small cell lung cancer (SCLC) is a highly aggressive cancer with a dismal 5-year survival of < 7%, despite the addition of immunotherapy to first-line chemotherapy. Specific tumor biomarkers, such as delta-like ligand 3 (DLL3) and schlafen11 (SLFN11), may enable the selection of more efficacious, novel immunomodulating targeted treatments like bispecific T-cell engaging monoclonal antibodies (tarlatamab) and chemotherapy with PARP inhibitors. However, obtaining a tissue biopsy sample can be challenging in SCLC. Circulating tumor cells (CTCs) have the potential to provide molecular insights into a patient's cancer through a "simple" blood test. CTCs have been studied for their prognostic ability in SCLC; however, their value in guiding treatment decisions is yet to be elucidated. This review explores novel and promising targeted therapies in SCLC, summarizes current knowledge of CTCs in SCLC, and discusses how CTCs can be utilized for precision medicine.
Collapse
Affiliation(s)
- Prajwol Shrestha
- Li Ka Shing Cell and Gene Therapy Program, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
- Precision Oncology Program, Gene and Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, Australia
- Medical Oncology, Calvary Mater Newcastle, Waratah, Australia
| | - Steven Kao
- Faculty of Medicine and Health, University of Sydney, Australia
- Medical Oncology, Chris O'Brien Lifehouse, Camperdown, Australia
| | - Veronica K Cheung
- Faculty of Medicine and Health, University of Sydney, Australia
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Camperdown, Australia
| | - Wendy A Cooper
- Faculty of Medicine and Health, University of Sydney, Australia
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Camperdown, Australia
- School of Medicine, University of Western Sydney, Australia
| | - Nico van Zandwijk
- Faculty of Medicine and Health, University of Sydney, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
- Concord Repatriation General Hospital, Sydney Local Health District, Concord, Australia
| | - John E J Rasko
- Li Ka Shing Cell and Gene Therapy Program, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
- Precision Oncology Program, Gene and Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
| | - Dannel Yeo
- Li Ka Shing Cell and Gene Therapy Program, Faculty of Medicine and Health, University of Sydney, Camperdown, Australia
- Precision Oncology Program, Gene and Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, Australia
- Cell and Molecular Therapies, Royal Prince Alfred Hospital, Sydney Local Health District, Camperdown, Australia
| |
Collapse
|
91
|
Chen H, Ma X, Liu J, Yang Y, He Y, Fang Y, Wang L, Fang J, Zhao J, Zhuo M. Real-world evaluation of first-line treatment of extensive-stage small-cell lung cancer with atezolizumab plus platinum/etoposide: a focus on patients with brain metastasis. Clin Transl Oncol 2024; 26:1664-1673. [PMID: 38329610 DOI: 10.1007/s12094-024-03387-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 01/06/2024] [Indexed: 02/09/2024]
Abstract
PURPOSE A previous real-world study conducted in China confirmed that first-line atezolizumab, in combination with etoposide/platinum (EP), leads to significantly longer progression-free survival (PFS) compared to EP alone in patients with extensive-stage small-cell lung cancer (ES-SCLC). The present study aimed to provide updated survival outcome data and evaluate the clinical efficacy of atezolizumab plus chemotherapy in ES-SCLC patients with brain metastasis (BM). METHODS This retrospective study included 225 patients with ES-SCLC who were treated with EP alone (EP group) or a combination of EP + atezolizumab (atezolizumab group). Survival outcomes for the total study sample and patients in the BM subgroup were estimated using the Kaplan-Meier method. RESULTS The atezolizumab group continued to demonstrate significantly longer PFS than the EP group (hazard ratio [HR], 0.68). The median overall survival (OS) was 26.2 months in the atezolizumab group vs. 14.8 months in the EP group (HR, 0.63). Additionally, among the BM patients in our study, the median PFS was found to be longer in the atezolizumab group (7.0 months) than in the EP group (4.1 months) (HR, 0.46). The OS of the BM patients did not differ significantly between the two treatment groups. CONCLUSIONS The addition of atezolizumab to EP as a first-line treatment for ES-SCLC was found to improve survival outcomes. This treatment combination may also prolong PFS in patients with BM, regardless of the administration of cranial irradiation. However, among the BM patients in our study, there was no significant difference in OS between the two treatment groups.
Collapse
Affiliation(s)
- Hanxiao Chen
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiangjuan Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department II of Thoracic Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jie Liu
- Cancer Center, Shandong Public Health Clinical Center, Public Health Clinical Center Affiliated to Shandong University, Shandong University, Jinan, China
| | - Yu Yang
- Department of Oncology, The 2nd Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yanhui He
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yong Fang
- Department of Oncology, Sir Run Run Shaw Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Liping Wang
- Department of Oncology, Baotou Cancer Hospital, Baotou, China
| | - Jian Fang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department II of Thoracic Oncology, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Jun Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic Oncology, Peking University Cancer Hospital & Institute, Beijing, China.
| | - Minglei Zhuo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department I of Thoracic Oncology, Peking University Cancer Hospital & Institute, Beijing, China.
| |
Collapse
|
92
|
Tosi A, Lorenzi M, Del Bianco P, Roma A, Pavan A, Scapinello A, Resi MV, Bonanno L, Frega S, Calabrese F, Guarneri V, Rosato A, Pasello G. Extensive-stage small-cell lung cancer in patients receiving atezolizumab plus carboplatin-etoposide: stratification of outcome based on a composite score that combines gene expression profiling and immune characterization of microenvironment. J Immunother Cancer 2024; 12:e008974. [PMID: 38955418 PMCID: PMC11218000 DOI: 10.1136/jitc-2024-008974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2024] [Indexed: 07/04/2024] Open
Abstract
PURPOSE Small-cell lung cancer (SCLC) is an aggressive disease with a dismal prognosis. The addition of immune checkpoints inhibitors to standard platinum-based chemotherapy in first-line setting achieves a durable benefit only in a patient subgroup. Thus, the identification of predictive biomarkers is an urgent unmet medical need. EXPERIMENTAL DESIGN Tumor samples from naive extensive-stage (ES) SCLC patients receiving atezolizumab plus carboplatin-etoposide were analyzed by gene expression profiling and two 9-color multiplex immunofluorescence panels, to characterize the immune infiltrate and SCLC subtypes. Associations of tissue biomarkers with time-to-treatment failure (TTF), progression-free survival (PFS) and overall survival (OS), were assessed. RESULTS 42 patients were included. Higher expression of exhausted CD8-related genes was independently associated with a longer TTF and PFS while increased density of B lymphocytes correlated with longer TTF and OS. Higher percentage of M2-like macrophages close to tumor cells and of CD8+T cells close to CD4+T lymphocytes correlated with increased risk of TF and longer survival, respectively. A lower risk of TF, disease progression and death was associated with a higher density of ASCL1+tumor cells while the expression of POU2F3 correlated with a shorter survival. A composite score combining the expression of exhausted CD8-related genes, B lymphocyte density, ASCL1 tumor expression and quantification of CD163+macrophages close to tumor cells, was able to stratify patients into high-risk and low-risk groups. CONCLUSIONS In conclusion, we identified tissue biomarkers and a combined score that can predict a higher benefit from chemoimmunotherapy in ES-SCLC patients.
Collapse
Affiliation(s)
- Anna Tosi
- Immunology and Molecular Oncology Diagnostics, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Martina Lorenzi
- Department of Medical Oncology, Santa Chiara Hospital, Trento, Italy
| | - Paola Del Bianco
- Clinical Trials and Biostatistics, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Anna Roma
- Medical Oncology 3, Istituto Oncologico Veneto IOV-IRCCS, Castelfranco Veneto, Italy
| | - Alberto Pavan
- Department of Medical Oncology, AULSS 3 Serenissima, Venezia, Italy
| | | | - Maria Vittoria Resi
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Laura Bonanno
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Stefano Frega
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Fiorella Calabrese
- Department of CardioThoracic Vascular Sciences and Public Health, Università degli Studi di Padova, Padova, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Diagnostics, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Giulia Pasello
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
- Medical Oncology 2, Istituto Oncologico Veneto IOV-IRCCS, Padova, Italy
| |
Collapse
|
93
|
Ceresoli GL, Rossi G, Agustoni F, Bonomi L, Borghetti P, Bulotta A, Casartelli C, Cerea G, Colonese F, Del Signore E, Finocchiaro G, Gianoncelli L, Grisanti S, Maiolani M, Pagni F, Proto C, Rijavec E, Vittimberga I, Arcangeli S, Filippi AR. Management of patients with extensive small-cell lung cancer in the immunotherapy era: An Italian consensus through a Delphi approach. Crit Rev Oncol Hematol 2024; 199:104247. [PMID: 38307393 DOI: 10.1016/j.critrevonc.2023.104247] [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: 01/30/2023] [Revised: 12/06/2023] [Accepted: 12/19/2023] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Immunotherapy represented a turning point for treating extensive small-cell lung cancer (ES-SCLC). Although, many issues remain debated. METHODS A group of Italian medical and radiation oncologists with expertise in managing patients with ES-SCLC developed a list of statements divided in six areas of interest. The Delphi method was used to assess the consensus on the defined list of statements. RESULTS 32 statements were included in the final list to be voted by the Delphi panel, and 26 reached a consensus on the agreement. A prompt involvement of a multidisciplinary team is a priority to provide an integrated treatment strategy. First-line recommended treatment is immunotherapy in combination with platinum-based chemotherapy and etoposide for four cycles followed by maintenance immunotherapy. CONCLUSIONS While awaiting new data from clinical trials and real-world studies, these recommendations can represent a useful tool to guide the management of ES-SCLC patients in daily practice.
Collapse
Affiliation(s)
| | - Giulio Rossi
- Pathology Unit, Hospital Institute Fondazione Poliambulanza, Via Bissolati 57, 25124 Brescia, Italy
| | - Francesco Agustoni
- Department of Internal Medicine and Medical Therapy, University of Pavia, Pavia, Italy; Medical Oncology Unit, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Lucia Bonomi
- Unit of Oncology, ASST Papa Giovanni XXIII Hospital, Bergamo, Italy
| | - Paolo Borghetti
- Radiation Oncology Department, ASST Spedali Civili and University of Brescia, Brescia, Italy
| | - Alessandra Bulotta
- Department of Oncology, IRCCS San Raffaele, via Olgettina 60, Milan, Italy
| | | | - Giulio Cerea
- Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Milano, Italy
| | | | - Ester Del Signore
- Division of Thoracic Oncology, European Institute of Oncology, IEO, Milan, Italy
| | - Giovanna Finocchiaro
- Medical Oncology and Hematologic Unit, Humanitas Cancer Center, Istituto Clinico Humanitas-IRCCS, Rozzano, Italy
| | - Letizia Gianoncelli
- Medical Oncology Unit, San Paolo Hospital, ASST Santi Paolo e Carlo, Milan, Italy
| | - Salvatore Grisanti
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Martina Maiolani
- U.O.C Oncologia Medica ASST Valtellina e Alto Lario, Sondrio, Italy
| | - Fabio Pagni
- Pathology, Department of Medicine and Surgery, University Milan Bicocca, Via Cadore 48, 20900 Monza, Italy
| | - Claudia Proto
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale Dei Tumori, Milan, Italy
| | - Erika Rijavec
- Unit of Medical Oncology, Ospedale di Circolo e Fondazione Macchi, ASST Sette Laghi, Varese, Italy
| | | | - Stefano Arcangeli
- Department of Radiation Oncology, University of Milan Bicocca, Milan, Italy
| | - Andrea Riccardo Filippi
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy; Radiation Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| |
Collapse
|
94
|
Dehar N, Meem M, Aggarwal I, Hopman W, Gaudreau PO, Robinson A, Fung AS. Brief Report: Real-World Eligibility for Clinical Trials in Patients With Extensive-Stage SCLC at a Tertiary Care Center. JTO Clin Res Rep 2024; 5:100696. [PMID: 39091596 PMCID: PMC11293570 DOI: 10.1016/j.jtocrr.2024.100696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/22/2024] [Accepted: 06/02/2024] [Indexed: 08/04/2024] Open
Abstract
Introduction The CASPIAN and IMpower133 trials revealed a significant survival benefit of chemotherapy plus immunotherapy in patients with extensive-stage SCLC. The current study characterizes the proportion of real-world patients who would have met eligibility for these trials and highlights factors influencing eligibility in the real-world setting. Methods A retrospective analysis of patient data was conducted for stage IV patients with SCLC treated at the Cancer Centre of Southeastern Ontario, Canada. Trial eligibility was based on criteria used in the IMpower133 and CASPIAN trials. Data were summarized using descriptive statistics. Overall survival was assessed using the Kaplan-Meier method. Results Of the 116 patients included, only 12.1% met the overall eligibility criteria for the IMpower133 trial, and 14.7% for the CASPIAN trial. The most common reasons for ineligibility included: Eastern Cooperative Oncology Group (ECOG) 2 or greater (77.5%), inadequate organ function (48%), and the presence of brain metastases at diagnosis (37.3%). Sixty-one patients (59.8%) met two or more major ineligibility criteria. If trial eligibility was expanded to include ECOG 2 patients, an additional 10.3% would have met eligibility. The median overall survival for all-comers was 6.5 months. Conclusions Only a small minority of real-world patients with extensive-stage SCLC would have met eligibility for the IMpower133 and CASPIAN trials, with ECOG greater than or equal to 2, inadequate organ function, and brain metastases comprising the most common reasons for trial ineligibility. Future clinical trials should expand the inclusion criteria to better represent real-world patient populations.
Collapse
Affiliation(s)
- Navdeep Dehar
- Department of Oncology, Queen’s School of Medicine, Queen’s University, Kingston, Ontario, Canada
- Cancer Centre of Southeastern Ontario, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | - Mahbuba Meem
- Department of Oncology, Queen’s School of Medicine, Queen’s University, Kingston, Ontario, Canada
- Cancer Centre of Southeastern Ontario, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | - Ishita Aggarwal
- Department of Oncology, Queen’s School of Medicine, Queen’s University, Kingston, Ontario, Canada
- Cancer Centre of Southeastern Ontario, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | - Wilma Hopman
- Department of Oncology, Queen’s School of Medicine, Queen’s University, Kingston, Ontario, Canada
| | - Pierre-Olivier Gaudreau
- Department of Oncology, Queen’s School of Medicine, Queen’s University, Kingston, Ontario, Canada
- Cancer Centre of Southeastern Ontario, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | - Andrew Robinson
- Department of Oncology, Queen’s School of Medicine, Queen’s University, Kingston, Ontario, Canada
- Cancer Centre of Southeastern Ontario, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| | - Andrea S. Fung
- Department of Oncology, Queen’s School of Medicine, Queen’s University, Kingston, Ontario, Canada
- Cancer Centre of Southeastern Ontario, Kingston Health Sciences Centre, Kingston, Ontario, Canada
| |
Collapse
|
95
|
Gan Y, Shi F, Zhu H, Li H, Han S, Li D. Cost-effectiveness of durvalumab plus tremelimumab in combination with chemotherapy for the treatment of metastatic non-small-cell lung cancer from the US healthcare sector's and societal perspectives. Front Pharmacol 2024; 15:1256992. [PMID: 38915475 PMCID: PMC11194367 DOI: 10.3389/fphar.2024.1256992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 05/27/2024] [Indexed: 06/26/2024] Open
Abstract
Purpose Metastatic non-small cell lung cancer (mNSCLC) has a high incidence rate, and economic burdens to patients, healthcare systems, and societies. Durvalumab plus tremelimumab and chemotherapy (T+D+CT) is a novel therapeutic strategy for mNSCLC, which demonstrated promising efficacy in a phase-3 randomized clinical trial, but its economic value remains unclear. Methods This economic evaluation used a hypothetical cohort of patients with mNSCLC, with characteristics mirroring those of the participants in the POSEIDON trial. Several partitioned survival models were constructed to estimate 15-year costs and health outcomes associated with the T+D+CT, durvalumab plus chemotherapy (D+CT) and chemotherapy alone (CT) strategies, discounting costs and effectiveness at 3% annually. Costs were in 2023 US dollars. Data were derived from the POSEIDON trial and published literature. Deterministic and probabilistic sensitivity analyses were performed to assess the uncertainty of input parameters and study generalizability. The analysis was designed and conducted from September 2022 to March 2023. To evaluate the cost-effectiveness of T+D+CT, compared with CT and D+CT, for mNSCLC from the perspectives of the US healthcare sector and society. Findings From the healthcare sector's perspective, the T+D+CT yielded an additional 0.09 QALYs at an increased cost of $7,108 compared with CT, which resulted in an ICER of $82,501/QALY. The T+D+CT strategy yielded an additional 0.02 QALYs at an increased cost of $27,779 compared with the D+CT, which resulted in an ICER of $1,243,868/QALY. The economic results of T+D+CT vs. CT were most sensitive to the annual discount rate, subsequent immunotherapy cost, tremelimumab cost, palliative care and death cost, pemetrexed cost, and durvalumab cost. The T+D+CT strategy was considered cost-effective relative to CT in 59%-82% of model iterations against willingness-to-pay. thresholds of $100,000/QALY gained to $150,000/QALY gained. From the societal perspective, the T+D+CT can be considered as cost-effective as compared with CT or D+CT, independent of histology. Implications In this cost-effectiveness analysis, the T+D+CT strategy represented good value compared with CT for patients with mNSCLC from the perspectives of the healthcare sector and the society. This treatment strategy may be prioritized for mNSCLC patients at high risks of disease progression.
Collapse
Affiliation(s)
- Yena Gan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Fenghao Shi
- International Research Center for Medicinal Administration, Peking University, Beijing, China
- School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - He Zhu
- International Research Center for Medicinal Administration, Peking University, Beijing, China
- School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Huangqianyu Li
- International Research Center for Medicinal Administration, Peking University, Beijing, China
- School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Sheng Han
- International Research Center for Medicinal Administration, Peking University, Beijing, China
- School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Duoduo Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
96
|
Chen Y, Han H, Cheng J, Cheng Q, Zhu S, Zhan P, Liu H, Song Y, Lv T. Efficacy and safety of anti-PD-1/PD-L1-based dual immunotherapies versus PD-1/PD-L1 inhibitor alone in patients with advanced solid tumor: a systematic review and meta-analysis. Cancer Immunol Immunother 2024; 73:155. [PMID: 38834888 PMCID: PMC11150353 DOI: 10.1007/s00262-024-03734-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 05/13/2024] [Indexed: 06/06/2024]
Abstract
INTRODUCTION Numerous randomized controlled trials (RCTs) have investigated PD-1/PD-L1 inhibitor-based combination therapies. The debate surrounding the potential additive clinical benefits of combination of two immune-oncology (IO) therapies for cancer patients persists. METHODS Both published and grey sources of randomized clinical trials that compared anti-PD-1/PD-L1-based immunotherapy combinations with monotherapy in patients with advanced or metastatic solid tumors were encompassed. The primary outcome was progression-free survival (PFS), and secondary outcomes included objective response rate (ORR), overall survival (OS) and treatment-related adverse events (TRAEs). RESULTS Our analysis encompassed 31 studies comprising 10,341 patients, which covered 12 distinct immune-oncology combination regimens. Across all patients, the immunotherapy combinations exhibited the capability to enhance the ORR (OR = 1.23 [95% CI 1.13-1.34]) and extend PFS (HR = 0.91 [95% CI 0.87-0.95]). However, the observed enhancement in OS (HR = 0.96 [95% CI 0.91-1.01]) was of no significance. Greater benefits in terms of PFS (HR = 0.82 [95% CI 0.72 to 0.93]) and OS (HR = 0.85 [95% CI 0.73 to 0.99]) may be particularly pronounced in cases where PD-L1 expression is negative. Notably, despite a heightened risk of any-grade TRAEs (OR = 1.72 [95% CI 1.40-2.11]) and grade greater than or equal to 3 TRAEs (OR = 2.01 [95% CI 1.67-2.43]), toxicity was generally manageable. CONCLUSIONS This study suggests that incorporating an additional immunotherapy agent with PD-1/PD-L1 inhibitors can elevate the response rate and reduce the risk of disease progression, all while maintaining manageable toxicity. However, there remains a challenge in translating these primary clinical benefits into extended overall survival.
Collapse
Affiliation(s)
- Yueying Chen
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hedong Han
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jing Cheng
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qinpei Cheng
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Suhua Zhu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Ping Zhan
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hongbing Liu
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yong Song
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tangfeng Lv
- Department of Respiratory and Critical Care Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
97
|
Liu H, Fu L, Jin S, Ye X, Chen Y, Pu S, Xue Y. Cardiovascular toxicity with CTLA-4 inhibitors in cancer patients: A meta-analysis. CANCER INNOVATION 2024; 3:e116. [PMID: 38947758 PMCID: PMC11212283 DOI: 10.1002/cai2.116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/04/2023] [Accepted: 12/25/2023] [Indexed: 07/02/2024]
Abstract
Background With the emergence of cytotoxic T lymphocyte-associated protein-4 (CTLA-4) inhibitors, the outcomes of patients with malignant tumors have improved significantly. However, the incidence of cardiovascular adverse events has also increased, which can affect tumor treatment. In this study, we evaluated the incidence and severity of adverse cardiovascular events caused by CTLA-4 inhibitors by analyzing reported trials that involved CTLA-4 inhibitor therapy. Methods Randomized clinical trials published in English from January 1, 2013, to November 30, 2022, were searched using the Cochrane Library and PubMed databases. All included trials examined all grade and grades 3-5 cardiac and vascular adverse events. These involved comparisons of CTLA-4 inhibitors to placebo, CTLA-4 inhibitors plus chemotherapy to chemotherapy alone, CTLA-4 inhibitors combined with PD-1/PD-L1 inhibitors to PD-1/PD-L1 inhibitors alone, and CTLA-4 inhibitors plus target agent to PD-1/PD-L1 inhibitors plus target agent. The odds ratio (OR) and corresponding 95% confidence intervals (CIs) were calculated using the Mantel-Haenszel method. Results Overall, 20 trials were included. CTLA-4 inhibitors significantly increased the incidence of all-grade cardiovascular toxicity (OR = 1.33, 95% CI: 1.00-1.75, p = 0.05). The incidence of all-grade cardiovascular toxicity increased in malignant tumor patients who received single-agent CTLA-4 inhibitors (OR = 1.73, 95% CI: 1.13-2.65, p = 0.01), as well as the incidence rate of grades 3-5 cardiovascular adverse events (OR = 2.00, 95% CI: 1.08-3.70, p = 0.03). Compared with the non-CTLA-4 inhibitor group, CTLA-4 inhibitors plus chemotherapy, PD-1/PD-L1 inhibitors, or target agent did not significantly affect the incidence of cardiac and vascular toxicity. The incidence of grades 3-5 cardiac failure, hypertension, pericardial effusion, myocarditis, and atrial fibrillation were much higher among patients exposed to CTLA-4 inhibitor, but the data were not statistically significant. Conclusion Our findings suggest that the incidence rate of all cardiovascular toxicity and severe cardiovascular toxicity increased in patients who were administered CTLA-4 inhibitors. In addition, the risk of serious cardiovascular toxic events was independent of the type of adverse event. From these results, physicians should assess the benefits and risks of CTLA-4 inhibitors when treating malignancies.
Collapse
Affiliation(s)
- Huiyi Liu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouGuangdongChina
| | - Lu Fu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouGuangdongChina
| | - Shuyu Jin
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouGuangdongChina
- The Second School of Clinical MedicineSouthern Medical UniversityGuangzhouGuangdongChina
| | - Xingdong Ye
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouGuangdongChina
| | - Yanlin Chen
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouGuangdongChina
| | - Sijia Pu
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouGuangdongChina
- School of MedicineSouth China University of TechnologyGuangzhouGuangdongChina
| | - Yumei Xue
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouGuangdongChina
- Guangdong Provincial Key Laboratory of Clinical Pharmacology, Research Center of Medical Sciences, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouGuangdongChina
| |
Collapse
|
98
|
Uccella S. Molecular Classification of Gastrointestinal and Pancreatic Neuroendocrine Neoplasms: Are We Ready for That? Endocr Pathol 2024; 35:91-106. [PMID: 38470548 PMCID: PMC11176254 DOI: 10.1007/s12022-024-09807-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/29/2024] [Indexed: 03/14/2024]
Abstract
In the last two decades, the increasing availability of technologies for molecular analyses has allowed an insight in the genomic alterations of neuroendocrine neoplasms (NEN) of the gastrointestinal tract and pancreas. This knowledge has confirmed, supported, and informed the pathological classification of NEN, clarifying the differences between neuroendocrine carcinomas (NEC) and neuroendocrine tumors (NET) and helping to define the G3 NET category. At the same time, the identification genomic alterations, in terms of gene mutation, structural abnormalities, and epigenetic changes differentially involved in the pathogenesis of NEC and NET has identified potential molecular targets for precision therapy. This review critically recapitulates the available molecular features of digestive NEC and NET, highlighting their correlates with pathological aspects and clinical characteristics of these neoplasms and revising their role as predictive biomarkers for targeted therapy. In this context, the feasibility and applicability of a molecular classification of gastrointestinal and pancreatic NEN will be explored.
Collapse
Affiliation(s)
- Silvia Uccella
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072, Pieve Emanuele, Milan, Italy.
- Pathology Service IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089, Rozzano, Milan, Italy.
| |
Collapse
|
99
|
Konen JM, Wu H, Gibbons DL. Immune checkpoint blockade resistance in lung cancer: emerging mechanisms and therapeutic opportunities. Trends Pharmacol Sci 2024; 45:520-536. [PMID: 38744552 PMCID: PMC11189143 DOI: 10.1016/j.tips.2024.04.006] [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: 02/13/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
Immune checkpoint blockade (ICB) therapy works by inhibiting suppressive checkpoints that become upregulated after T cell activation, like PD-1/PD-L1 and CTLA-4. While the initial FDA approvals of ICB have revolutionized cancer therapies and fueled a burgeoning immuno-oncology field, more recent clinical development of new agents has been slow. Here, focusing on lung cancer, we review the latest research uncovering tumor cell intrinsic and extrinsic ICB resistance mechanisms as major hurdles to treatment efficacy and clinical progress. These include genomic and non-genomic tumor cell alterations, along with host and microenvironmental factors like the microbiome, metabolite accumulation, and hypoxia. Together, these factors can cooperate to promote immunosuppression and ICB resistance. Opportunities to prevent resistance are constantly evolving in this rapidly expanding field, with the goal of moving toward personalized immunotherapeutic regimens.
Collapse
Affiliation(s)
- Jessica M Konen
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.
| | - Haoyi Wu
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
100
|
Wang X, Chiang AC. Big Decisions on Small Cell Lung Cancer: A Focus on Clinical Care Updates and Patient Perspectives. Am Soc Clin Oncol Educ Book 2024; 44:e432520. [PMID: 38830134 DOI: 10.1200/edbk_432520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
Small cell lung cancer (SCLC) is an uncommon, aggressive high-grade neuroendocrine carcinoma, associated with tobacco use. It is a highly chemosensitive disease that initially responds quickly to systemic therapy, although patients with SCLC tend to develop relapse. Although the landscape of SCLC treatment has remained stagnant for many decades, the field has seen notable advances in the past few years, including the use of immunotherapy, the development of further lines of systemic therapy, the refinement of thoracic and intracranial radiotherapy, and-most recently-the promise of more targeted therapies. Patients with SCLC also must face unique psychosocial burdens in their experience with their cancer, distinct from patients with other lung cancer. In this article, we review the latest literature and future directions in the management and investigation of SCLC, as well as the critical decisions that providers and patients must navigate in the current landscape. We also present the perspectives of several patients with SCLC in conjunction with this summary, to spotlight their individual journeys in the context of this challenging disease.
Collapse
Affiliation(s)
- Xiao Wang
- Yale School of Medicine, New Haven, CT
| | | |
Collapse
|