1
|
Sposito M, Eccher S, Pasqualin L, Scaglione IM, Avancini A, Tregnago D, Trestini I, Insolda J, Bonato A, Ugel S, Derosa L, Milella M, Pilotto S, Belluomini L. Characterizing the immune tumor microenvironment in ALK fusion-positive lung cancer: state-of-the-art and therapeutical implications. Expert Rev Clin Immunol 2024; 20:959-970. [PMID: 38913940 DOI: 10.1080/1744666x.2024.2372327] [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: 03/11/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
INTRODUCTION Approximately 5% of non-small cell lung cancer (NSCLC), exhibits anaplastic lymphoma kinase (ALK) rearrangements. EML4-ALK fusions account for over 90% of ALK rearrangements in NSCLC. The advent of treatment targeting ALK has significantly improved survival rates in patients with advanced ALK-positive NSCLC. However, the emergence of resistance mechanisms and the subsequent progression disease inevitably occurs. The tumor immune microenvironment (TIME) plays a pivotal role in lung cancer, influencing disease development, patient's outcomes, and response to treatments. AREAS COVERED The aim of this review is to provide a comprehensive characterization of the TIME in ALK rearranged NSCLC and its intrinsic plasticity under treatment pressure. EXPERT OPINION Recognizing the fundamental role of the TIME in cancer progression has shifted the paradigm from a tumor cell-centric perspective to the understanding of a complex tumor ecosystem. Understanding the intricate dynamics of the TIME, its influence on treatment response, and the potential of immunotherapy in patients with ALK-positive NSCLC are currently among the primary research objectives in this patient population.
Collapse
Affiliation(s)
- Marco Sposito
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Serena Eccher
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Luca Pasqualin
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Ilaria Mariangela Scaglione
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Alice Avancini
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Daniela Tregnago
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Ilaria Trestini
- Dietetic Service, Hospital Medical Direction, University and Hospital Trust (AOUI) of Verona, Verona, Italy
| | - Jessica Insolda
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Adele Bonato
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana, Santa Chiara Hospital, Pisa, Italy
| | - Stefano Ugel
- Immunology Section, University Hospital and Department of Medicine, University of Verona, Verona, Italy
| | - Lisa Derosa
- INSERM U1015 Gustave Roussy Cancer Campus, Villejuif Cedex, Villejuif, France
- Faculté de Médicine, Université Paris-Saclay, Le Kremlin-Bicetre, France
| | - Michele Milella
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Sara Pilotto
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Lorenzo Belluomini
- Section of Oncology, Department of Engineering for Innovation Medicine (DIMI), University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| |
Collapse
|
2
|
Amari L, Tomasini P, Dantony E, Rousseau-Bussac G, Ricordel C, Bigay-Game L, Arpin D, Morel H, Veillon R, Justeau G, Huchot E, Fournel P, Vergnenegre A, Bizeux A, Subtil F, Clarisse B, Decroisette C, Chouaid C, Greillier L, Bylicki O. Safety and Patient-Reported outcomes of atezolizumab plus chemotherapy with or without bevacizumab in stage IIIB/IV non-squamous non-small cell lung cancer with EGFR mutation, ALK rearrangement or ROS1 fusion progressing after targeted therapies (GFPC 06-2018 study). Lung Cancer 2024; 193:107843. [PMID: 38830303 DOI: 10.1016/j.lungcan.2024.107843] [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: 04/20/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/05/2024]
Abstract
BACKGROUND In an open-label multicenter non-randomized non-comparative phase II study in patients with stage IIIB/IV non-squamous non-small cell lung cancer (NSCLC), oncogenic addiction (EGFR mutation or ALK/ROS1 fusion), with disease progression after tyrosine-kinase inhibitor and no prior chemotherapy (NCT04042558), atezolizumab, carboplatin, pemetrexed with or without bevacizumab showed some promising result. Beyond the clinical evaluation, we assessed safety and patient-reported outcomes (PROs) to provide additional information on the relative impact of adding atezolizumab to chemotherapy with and without bevacizumab in this population. MATERIALS Patients received platinum-pemetrexed-atezolizumab-bevacizumab (PPAB cohort) or, if not eligible, platinum-pemetrexed-atezolizumab (PPA cohort). The incidence, nature, and severity of adverse events (AEs) were assessed. PROs were evaluated using the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire (EORTC QLQ-Core 30 and EORTC QLQ-Lung Cancer 13). RESULT Overall, 68 (PPAB) and 72 (PPA) patients were evaluable for safety. Grade 3-4 AEs occurred in 83.8% (PPAB) and 63.9% (PPA). Grade 3-4 atezolizumab-related AEs occurred in 29.4% and 19.4%, respectively. Grade 3-4 bevacizumab-related AEs occurred in 36.8% (PPAB). Most frequent grade 3-4 AEs were neutropenia (19.1% in PPAB; 23.6% in PPA) and asthenia (16.2% in PPAB; 9.7% in PPA). In PPAB, we observed a global stability in global health security (GHS) score, fatigue and dyspnea with a constant tendency of improvement, and a significant improvement in cough. In PPA, we observed a significant improvement in GHS score with a significant improvement in fatigue, dyspnea and cough. At week 54, we observed an improvement from baseline in GHS score for 49.2% of patients. In both cohorts, patients reported on average no clinically significant worsening in their overall health or physical functioning scores. CONCLUSION PPAB and PPA combinations seem tolerable and manageable in patients with stage IIIB/IV non-squamous NSCLC with oncogenic addiction (EGFR mutation or ALK/ROS1 fusion) after targeted therapies.
Collapse
Affiliation(s)
- Lyria Amari
- Department of Pneumology, Hôpital d'Instruction des Armées Sainte Anne, Toulon, France
| | - Pascale Tomasini
- Aix Marseille Univ, APHM, INSERM, CNRS, CRCM, Hôpital Nord, Multidisciplinary Oncology and Therapeutic Innovations, Marseille, France
| | - Emmanuelle Dantony
- Service de Biostatistique, Hospices Civils de Lyon, Lyon France; Université Lyon 1, CNRS, Laboratoire de Biométrie et Biologie Évolutive UMR 5558, Villeurbanne, France
| | | | - Charles Ricordel
- Department of Pneumology, CHU Rennes, Univ Rennes 1, INSERM, OSS (Oncogenesis Stress Signaling), UMR_S 1242, CLCC Eugene Marquis, F-35000 Rennes, France
| | - Laurence Bigay-Game
- Department of Pneumology & Thoracic Oncology, CHU Toulouse-Hôpital Larrey, Toulouse, France
| | - Dominique Arpin
- Department of Pneumology, Hôpital Nord-Ouest, Villefranche-sur-Saône, France
| | - Hugues Morel
- Respiratory Medicine Department, Centre Hospitalier Régional d'Orléans Hôpital de La Source, Orléans, France
| | - Remi Veillon
- Oncologie thoracique, CHU de Bordeaux, Bordeaux, France
| | | | - Eric Huchot
- CHU Saint Pierre de La Réunion, Saint-Pierre, La Réunion, France
| | - Pierre Fournel
- Department of Pneumology & Thoracic Oncology, North Hospital, University Hospital of Saint-Etienne, Saint-Etienne, France
| | | | | | - Fabien Subtil
- Service de Biostatistique, Hospices Civils de Lyon, Lyon France; Université Lyon 1, CNRS, Laboratoire de Biométrie et Biologie Évolutive UMR 5558, Villeurbanne, France
| | - Bénédicte Clarisse
- Clinical Research Department, Comprehensive Cancer Centre François Baclesse, Caen, France
| | - Chantal Decroisette
- Department of Pneumology & Thoracic Oncology, CH Annecy-Genevois, 74370, Metz-Tessy, France
| | - Christos Chouaid
- Department of Pneumology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Laurent Greillier
- Aix Marseille Univ, APHM, INSERM, CNRS, CRCM, Hôpital Nord, Multidisciplinary Oncology and Therapeutic Innovations, Marseille, France
| | - Olivier Bylicki
- Department of Pneumology, Hôpital d'Instruction des Armées Sainte Anne, Toulon, France.
| |
Collapse
|
3
|
Hu J, Li M, Xie Z, Chen J. Comparison of the efficacy and safety of domestically produced tislelizumab, camrelizumab, and imported pembrolizumab in the treatment of advanced NSCLC: a real-world retrospective study. Clin Transl Oncol 2024:10.1007/s12094-024-03565-7. [PMID: 38935240 DOI: 10.1007/s12094-024-03565-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Since the imported PD-1 inhibitor pembrolizumab was listed in China in 2018, China has opened up the era of immunotherapy for malignant tumors, with several domestically produced PD-1 inhibitors coming onto the market one after another. To find out whether there are differences in the efficacy and safety of domestic and imported PD-1 inhibitors in patients with advanced non-small cell lung cancer, we conducted this retrospective study in two tertiary hospitals in China. METHODS Patients with advanced NSCLC treated with tislelizumab or camrelizumab or pembrolizumab who met the inclusion criteria were screened through the electronic medical record system. A total of 259 patients were screened, but due to the unbalanced baseline, we performed propensity score matching and finally included 149 patients in three groups: pembrolizumab (n = 38), tislelizumab (n = 38), and camrelizumab (n = 73), which had very balanced baseline characteristics in each group after propensity score matching treatment. RESULTS The results showed that the median progression-free period was 11.3 m vs 10.1 m vs 8.9 m; p = 0.754; and the objective response rate was 63.2% vs 50% vs 57.5%; P = 0.510 for pembrolizumab, tislelizumab, and carrelizumab, respectively. There was no significant difference in median PFS between PD-L1 expression subgroups. In terms of safety, only skin toxicity of any grade of carrelizumab was higher than that of the other two groups (p = 0.034), and the incidence of grade ≥ 3 adverse reactions was not statistically significant among the three groups. CONCLUSION In this real-world study, the efficacy and safety of the domestically produced tislelizumab, camrelizumab, and the imported pembrolizumab were comparable.
Collapse
Affiliation(s)
- Jia Hu
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, No. 19 Nonglinxia Road, Yuexiu District, Guangzhou, 510000, Guangdong, China
| | - MengTing Li
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, No. 19 Nonglinxia Road, Yuexiu District, Guangzhou, 510000, Guangdong, China
| | - ZeYu Xie
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, No. 19 Nonglinxia Road, Yuexiu District, Guangzhou, 510000, Guangdong, China
| | - JiSheng Chen
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, No. 19 Nonglinxia Road, Yuexiu District, Guangzhou, 510000, Guangdong, China.
| |
Collapse
|
4
|
Zhang H, Zhang Z, Yan N, Li X. Association of PD-L1 expression and clinical outcomes in ROS1 - rearranged advanced non-small cell lung cancer treated with crizotinib. Front Oncol 2024; 14:1405683. [PMID: 38835380 PMCID: PMC11148223 DOI: 10.3389/fonc.2024.1405683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
Background Programmed cell death ligand 1 (PD-L1) is more readily expressed in ROS proto-oncogene 1 (ROS1) rearranged non-small cell lung cancer (NSCLC) compared to NSCLC cases lacking driver gene mutations. Prior research has established a link between PD-L1 expression and reduced effectiveness of EGFR or ALK inhibitors in EGFR or ALK-positive NSCLC. Nonetheless, the relationship between initial PD-L1 levels and the clinical impact of first-line crizotinib therapy in ROS1-rearranged NSCLC is still uncertain. Methods From January 2016 to December 2021, a total of 246 patients with ROS1 positive tumors were collected. Out of these, 82 patients with advanced ROS1-rearranged NSCLC, who were treated with crizotinib as their initial therapy, were selected for the study. The study aimed primarily to evaluate the objective response rate (ORR) and progression-free survival (PFS), and secondarily to assess disease control rate (DCR) and overall survival (OS). Results Of the 82 advanced ROS1-rearranged NSCLC patients, 38 exhibited PD-L1 positivity, subdivided into 11 with high and 27 with low expression levels, while the remaining 44 showed no PD-L1 expression. The ORR for all included patients was 80.5%. No statistically significant variance in ORR was observed among ROS1-rearranged NSCLC patients across differing PD-L1 expression statuses. However, there was a statistically significant difference in DCR between PD-L1 negative group (100%) and high expression group (90.9%) (p=0.04). The median PFS spanned 26.4 months for the PD-L1 negative group, 16.6 for the low expression group, and 13.7 for the high expression group (p=0.001). Additionally, a notable statistical disparity was also observed in median PFS between the PD-L1 negative and positive groups (p=0.02). For the entire study population, the median OS was 53.0 months (95% CI 43.8 - 62.2). In the PD-L1-negative group, the median OS reached 57.2 months, compared to 53.0 months in the PD-L1-positive group, a difference lacking statistical significance (p=0.43). Conclusions Our results suggest that for ROS1-positive NSCLC patients receiving crizotinib as first-line therapy, PD-L1 expression may serve as a negative prognostic marker for PFS rather than OS.
Collapse
Affiliation(s)
- Huixian Zhang
- Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ziheng Zhang
- Department of Emergency Medicine, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ningning Yan
- Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xingya Li
- Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Li S, Zhang H, Chen T, Zhang X, Shang G. Current treatment and novel insights regarding ROS1-targeted therapy in malignant tumors. Cancer Med 2024; 13:e7201. [PMID: 38629293 PMCID: PMC11022151 DOI: 10.1002/cam4.7201] [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: 05/20/2023] [Revised: 03/22/2024] [Accepted: 04/06/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND The proto-oncogene ROS1 encodes an intrinsic type I membrane protein of the tyrosine kinase/insulin receptor family. ROS1 facilitates the progression of various malignancies via self-mutations or rearrangements. Studies on ROS1-directed tyrosine kinase inhibitors have been conducted, and some have been approved by the FDA for clinical use. However, the adverse effects and mechanisms of resistance associated with ROS1 inhibitors remain unknown. In addition, next-generation ROS1 inhibitors, which have the advantage of treating central nervous system metastases and alleviating endogenous drug resistance, are still in the clinical trial stage. METHOD In this study, we searched relevant articles reporting the mechanism and clinical application of ROS1 in recent years; systematically reviewed the biological mechanisms, diagnostic methods, and research progress on ROS1 inhibitors; and provided perspectives for the future of ROS1-targeted therapy. RESULTS ROS1 is most expressed in malignant tumours. Only a few ROS1 kinase inhibitors are currently approved for use in NSCLC, the efficacy of other TKIs for NSCLC and other malignancies has not been ascertained. There is no effective standard treatment for adverse events or resistance to ROS1-targeted therapy. Next-generation TKIs appear capable of overcoming resistance and delaying central nervous system metastasis, but with a greater incidence of adverse effects. CONCLUSIONS Further research on next-generation TKIs regarding the localization of ROS1 and its fusion partners, binding sites for targeted drugs, and coadministration with other drugs is required. The correlation between TKIs and chemotherapy or immunotherapy in clinical practice requires further study.
Collapse
Affiliation(s)
- Shizhe Li
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - He Zhang
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Ting Chen
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Xiaowen Zhang
- Medical Research CenterShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Guanning Shang
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| |
Collapse
|
6
|
Tian P, Du D, Yang L, Zhou N, Tao L. SP3-induced Timeless transcription contributes to cell growth of lung adenocarcinoma cells. PLoS One 2024; 19:e0298295. [PMID: 38354174 PMCID: PMC10866488 DOI: 10.1371/journal.pone.0298295] [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: 08/30/2023] [Accepted: 01/23/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND Timeless is well-known for its key role in replication checkpoints. Recent studies reveal the involvement of Timeless and specificity protein (SP) 1 in human malignancies. However, no evidence proved the interaction between SP3 and Timeless in lung adenocarcinoma (LUAD). METHODS The expression and clinical significance of Timeless were analyzed using the LUAD dataset downloaded from the Cancer Genome Atlas (TCGA). Lentivirus-mediated Timeless knockdown in A549 cells was used to examine the role of Timeless in cell proliferation and pemetrexed (PEM) resistance. Transcription factors (TFs) bound to the Timeless promoter were identified by DNA pull-down technology with HPLC-MS/MS analysis and analyzed by the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway. Dual-luciferase reporter assay was used to determine the activity of SP3 in Timeless transcription. RESULTS Timeless was overexpressed in LUAD samples, and it could serve as a potential diagnostic or prognostic biomarker for LUAD patients. shTimeless-mediated knockdown of Timeless reduced cell viability and proliferation and sensitized PEM-resistant A549 cells to PEM. Four fragments (F1: 1-373 bp), (F2: 374-962 bp), (F4: 1274-1645 bp), and (F5: 1646-2000bp) were confirmed as the TF binding profiles of the Timeless promoter. KEGG analysis showed that the TFs bound to the Timeless promoter had relevance to spliceosome, RNA transport, and mRNA surveillance pathways. SP3 promoted the transcription of Timeless via the F2 fragment (374-962 bp) binding motif. CONCLUSION Upregulation of Timeless mediated by SP3 promotes LUAD cell proliferation, providing evidence to support that targeting the SP3/Timeless axis may be a potential therapeutic strategy against LUAD.
Collapse
Affiliation(s)
- Ping Tian
- Medical School, Xinyang Vocational and Technical College, Xinyang, Henan, China
| | - Dajun Du
- Department of Surgical Oncology, Xinyang Central Hospital, Xinyang, Henan, China
| | - Li Yang
- Inspection School, Xinyang Vocational and Technical College, Xinyang, Henan, China
| | - Nan Zhou
- Department of Medical Oncology, Xinyang Central Hospital, Xinyang, Henan, China
| | - Ling Tao
- Inspection School, Xinyang Vocational and Technical College, Xinyang, Henan, China
| |
Collapse
|
7
|
Xu J, Wang Z. Recent advances progress of targeted drugs combined with radiotherapy for advanced non-small cell lung cancer: a review. Front Oncol 2023; 13:1285593. [PMID: 38115908 PMCID: PMC10728551 DOI: 10.3389/fonc.2023.1285593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
Targeted drug therapy plays an important role in the clinical application of non-small cell lung cancer, especially adenocarcinoma. However, for patients with advanced disease, drug resistance after targeted therapy, unclear target, and other reasons that cannot or do not want surgery, the combination of chemotherapy, radiotherapy, immunity, etc. is often used. The synergistic effect of targeted drugs and radiotherapy in non-small cell lung cancer has shown good clinical efficacy. This article reviews the clinical progress of targeted drug therapy combined with radiotherapy in advanced non-small cell lung cancer in recent years, in order to provide new ideas for further clinical research of this treatment mode.
Collapse
Affiliation(s)
- Jiamin Xu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Zhongming Wang
- Department of Oncology and Radiotherapy, Shidong Hospital, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
8
|
Nikmaneshi MR, Baish JW, Zhou H, Padera TP, Munn LL. Transport Barriers Influence the Activation of Anti-Tumor Immunity: A Systems Biology Analysis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304076. [PMID: 37949675 PMCID: PMC10754116 DOI: 10.1002/advs.202304076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 10/07/2023] [Indexed: 11/12/2023]
Abstract
Effective anti-cancer immune responses require activation of one or more naïve T cells. If the correct naïve T cell encounters its cognate antigen presented by an antigen presenting cell, then the T cell can activate and proliferate. Here, mathematical modeling is used to explore the possibility that immune activation in lymph nodes is a rate-limiting step in anti-cancer immunity and can affect response rates to immune checkpoint therapy. The model provides a mechanistic framework for optimizing cancer immunotherapy and developing testable solutions to unleash anti-tumor immune responses for more patients with cancer. The results show that antigen production rate and trafficking of naïve T cells into the lymph nodes are key parameters and that treatments designed to enhance tumor antigen production can improve immune checkpoint therapies. The model underscores the potential of radiation therapy in augmenting tumor immunogenicity and neoantigen production for improved ICB therapy, while emphasizing the need for careful consideration in cases where antigen levels are already sufficient to avoid compromising the immune response.
Collapse
Affiliation(s)
- Mohammad R. Nikmaneshi
- Department of Radiation OncologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - James W. Baish
- Biomedical EngineeringBucknell UniversityLewisburgPA17837USA
| | - Hengbo Zhou
- Department of Radiation OncologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Timothy P. Padera
- Department of Radiation OncologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| | - Lance L. Munn
- Department of Radiation OncologyMassachusetts General Hospital and Harvard Medical SchoolBostonMA02114USA
| |
Collapse
|
9
|
Marchal M, Leroy V, Behal H, Dansin E, Paris N, Bordier S, Humez S, Escande F, Gauvain C, Cortot AB. Histo-Molecular Factors of Response to Combined Chemotherapy and Immunotherapy in Non-Small Cell Lung Cancers. Target Oncol 2023; 18:927-939. [PMID: 37921939 PMCID: PMC10663251 DOI: 10.1007/s11523-023-01009-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 11/05/2023]
Abstract
BACKGROUND Chemo-immunotherapy (CIT) is the standard of care for advanced non-small cell lung cancer (NSCLC), but the impact of routinely available histo-molecular biomarkers on its efficacy has not yet been fully assessed. OBJECTIVE The purpose of this multicenter study was to evaluate the clinical activity of CIT according to oncogenic drivers, STK11 and TP53 mutations, and MET overexpression. PATIENTS AND METHODS Patients receiving CIT for advanced NSCLC with available comprehensive molecular profile were included. The primary endpoint was progression-free survival (PFS), adjusted on main confounding factors, and secondary endpoints were overall survival (OS) and objective response rate. RESULTS Among the 195 patients included between September 2018 and October 2021, 88 (41%) had a KRAS mutation, 16 (8.2%) an EGFR mutation or an ALK, ROS1, or RET rearrangement, 11 (5.6%) a BRAF mutation, 6 (3.1%) a MET exon 14 mutation or MET amplification, and 5 (2.6%) a HER2 mutation. Seventy-seven patients (39.5%) had none of these alterations. The median PFS was 6.4 months (95% CI 5.3-7.3). Per subgroup, the median PFS was 7.1 months (5.4-8.9) for KRAS, 5.5 months (2.5-15.3) for EGFR/ALK/ROS1/RET, 12.9 months (2.6-not reached [NR]) for BRAF, 1.5 months (0.6-NR) for MET, 3.9 months (2.6-NR) for HER2, and 5.6 months (4.7-7.8) for patients without any oncogenic alteration. No difference in PFS was observed between the KRAS, BRAF, EGFR/ALK/ROS1/RET, and no-driver subgroups. STK11 mutations were associated with poor PFS (HR 1.59 [95% CI 1.01-2.51]) whereas TP53 mutations had no impact. MET overexpression was associated with longer PFS (HR 0.59 [95% CI 0.35-0.99]). CONCLUSION This study suggests that the efficacy of combining pembrolizumab with pemetrexed and platinum-based chemotherapy differs according to the histo-molecular biomarkers, which may help to identify patients liable to benefit from CIT.
Collapse
Affiliation(s)
- Marine Marchal
- Department of Thoracic Oncology, Univ. Lille, CHU Lille, Boulevard du Professeur Leclercq, 59000, Lille, France.
| | - Vincent Leroy
- Department of Pneumology, Clinique Tessier, Valenciennes, France
| | - Hélène Behal
- Biostatistics Department, CHU Lille, 59000, Lille, France
| | - Eric Dansin
- Department of Medical Oncology, Centre Oscar Lambret, Lille, France
| | - Nicolas Paris
- Department of Pneumology, Dunkerque Hospital, Dunkerque, France
| | - Soraya Bordier
- Department of Pneumology, Roubaix Hospital, Roubaix, France
| | - Sarah Humez
- Department of Pathology, CHU Lille, Univ. Lille, CHU de Lille, 59000, Lille, France
- CNRS, Inserm, Institut Pasteur de Lille, UMR9020-UMR1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Univ. Lille, CHU Lille, 59000, Lille, France
| | - Fabienne Escande
- Department of Biochemistry and Molecular Biology, CHU Lille, Univ. Lille, CHU de Lille, 59000, Lille, France
| | - Clément Gauvain
- Department of Thoracic Oncology, Univ. Lille, CHU Lille, Boulevard du Professeur Leclercq, 59000, Lille, France
| | - Alexis B Cortot
- Department of Thoracic Oncology, Univ. Lille, CHU Lille, Boulevard du Professeur Leclercq, 59000, Lille, France
- CNRS, Inserm, Institut Pasteur de Lille, UMR9020-UMR1277-Canther-Cancer Heterogeneity, Plasticity and Resistance to Therapies, Univ. Lille, CHU Lille, 59000, Lille, France
| |
Collapse
|
10
|
Ratain MJ, Strohbehn GW. Combining atezolizumab 1200 mg with bevacizumab 15 mg/kg: based on science or just revenues? Eur J Cancer 2023; 194:113349. [PMID: 37806256 DOI: 10.1016/j.ejca.2023.113349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 10/10/2023]
Abstract
Dose optimisation is increasingly important in oncology, as exemplified by the US Food and Drug Administration's Project Optimus initiative, which is aligned with similar initiatives in other countries. In parallel, multiple stakeholders have raised concerns about anticancer drug prices, affordability, and access. This is of particular concern to government payers as well as patients and physicians in low- and middle-income countries. As anticancer drugs have historically been approved at the maximally tolerated dose, it is now highly relevant to question whether lower doses are equally effective and can be delivered at lower doses, resulting in less toxicity for patients, and lower costs for patients and payers. We illustrate this opportunity by discussing the combination of atezolizumab and bevacizumab, approved in multiple countries for both non-small cell lung cancer and hepatocellular cancer. Our conclusion is that the cost of this regimen can be reduced by more than 80%, an opportunity that should be considered by patients, prescribers, payers, and policymakers.
Collapse
Affiliation(s)
- Mark J Ratain
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago, Chicago, Illinois, USA.
| | - Garth W Strohbehn
- Veterans Affairs Center for Clinical Management Research and Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
11
|
Sicard G, Protzenko D, Giacometti S, Barlési F, Ciccolini J, Fanciullino R. Harnessing tumor immunity with cytotoxics: T cells monitoring in mice bearing lung tumors treated with anti-VEGF and pemetrexed-cisplatin doublet. Br J Cancer 2023; 129:1373-1382. [PMID: 37524968 PMCID: PMC10628115 DOI: 10.1038/s41416-023-02350-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/19/2023] [Accepted: 06/27/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Successful immunotherapy is restricted to some cancers only, and combinatorial strategies with other drugs could help to improve their efficacy. Here, we monitor T cells in NSCLC model after treatment with cytotoxics (CT) and anti-VEGF drugs, to understand when immune checkpoint inhibitors should be best associated next. METHODS In vivo study was performed on BALB/c mice grafted with KLN205 cells. Eight treatments were tested including control, cisplatin and pemetrexed as low (LD CT) and full (MTD CT) dose as single agents, flat dose anti-VEGF and the association anti-VEGF + CT. Full immunomonitoring was performed by flow cytometry on tumor, spleen and blood over 3 weeks. RESULTS Immunomodulatory effect was dependent upon both treatments and time. In tumors, combination groups shown numerical lower Treg cells on Day 21. In spleen, anti-VEGF and LD CT group shown higher CD8/Treg ratio on Day 7; on Day 14, higher T CD4 were observed in both combination groups. Finally, in blood, Tregs were lower and CD8/Treg ratio higher, on Day 14 in both combination groups. On Day 21, CD4 and CD8 T cells were higher in the anti-VEGF + MTD CT group. CONCLUSIONS Anti-VEGF associated to CT triggers notable increase in CD8/Tregs ratio. Regarding the scheduling, a two-week delay after using anti-VEGF and CT could be the best sequence to optimize antitumor efficacy.
Collapse
Affiliation(s)
- G Sicard
- SMARTc & COMPO Team, CRCM Inserm U1068, Aix Marseille University, 13007, Marseille, France
| | - D Protzenko
- SMARTc & COMPO Team, CRCM Inserm U1068, Aix Marseille University, 13007, Marseille, France
| | - S Giacometti
- SMARTc & COMPO Team, CRCM Inserm U1068, Aix Marseille University, 13007, Marseille, France
| | - F Barlési
- School of Medicine, Aix Marseille University, 13007, Marseille, France
- Gustave Roussy Institute, 94800, Villejuif, France
| | - J Ciccolini
- SMARTc & COMPO Team, CRCM Inserm U1068, Aix Marseille University, 13007, Marseille, France.
| | - R Fanciullino
- SMARTc & COMPO Team, CRCM Inserm U1068, Aix Marseille University, 13007, Marseille, France
| |
Collapse
|
12
|
Wang Z, Zhou F, Xu S, Wang K, Ding H. The efficacy and safety of immune checkpoint inhibitors for patients with EGFR-mutated non-small cell lung cancer who progressed on EGFR tyrosine-kinase inhibitor therapy: A systematic review and network meta-analysis. Cancer Med 2023; 12:18516-18530. [PMID: 37584242 PMCID: PMC10557893 DOI: 10.1002/cam4.6453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/22/2023] [Accepted: 08/04/2023] [Indexed: 08/17/2023] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) patients harboring epidermal growth factor receptor (EGFR)-mutated who progressed on EGFR tyrosine-kinase inhibitor (EGFR-TKI) therapy have limited therapeutic options. There is still no consensus on the role of immune checkpoint inhibitors (ICIs) in NSCLC with EGFR mutations. METHODS We did a network meta-analysis (NMA) with a systematic literature search on PubMed, Embase, Web of Science, and The Cochrane Library. We included all phase II and III randomized controlled trials (RCTs), non-randomized controlled trials (Non-RCTs), and retrospective studies. Progression-free survival (PFS) and overall survival (OS) were assessed through hazard ratios (HR). Objective response rate (ORR) and adverse events (AEs) were assessed through odds ratio (OR) and relative risk (RR), respectively. R software was used to compare the outcomes of different treatments by Bayesian NMA. FINDINGS We identified 1835 published results and 17 studies were included ultimately. A total of 2085 patients were included and accepted the following six treatments: ICIs plus chemotherapy (ICIs+Chemo), chemotherapy (Chemo), ICIs monotherapy (ICIs), ICIs plus chemotherapy and antiangiogenic therapy (ICIs+Chemo+Antiangio), antiangiogenic therapy plus chemotherapy (Antiangio+Chemo), ICIs plus antiangiogenic therapy (ICIs+Antiangio). ICIs+Chemo+Antiangio was associated with longer PFS and OS, as well as higher ORR (surface under the cumulative ranking curve [SUCRA], 96%, 90%, 91%). ICIs conferred the safety profile in terms of any-grade AEs, grade greater than or equal to 3 AEs and any grade leading to treatment discontinuation occurred AEs (SUCRA, 99%, 68%, 94%). INTERPRETATION ICIs+Chemo+Antiangio brings the greatest survival benefit in NSCLC patients with EGFR mutations who progressed on EGFR-TKI therapy, even for whom with baseline brain metastases. Compared with chemotherapy, ICIs has a low incidence of AEs and a benefit in OS.
Collapse
Affiliation(s)
- Zhen Wang
- Department of RadiotherapyThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Fang Zhou
- Department of RadiotherapyThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Shan Xu
- Department of OncologyZaozhuang Municipal HospitalZao ZhuangChina
| | - Kang Wang
- Department of OncologyZaozhuang Municipal HospitalZao ZhuangChina
| | - Huan Ding
- Department of OncologyZaozhuang Municipal HospitalZao ZhuangChina
| |
Collapse
|