1
|
Mestiri S, Sami A, Sah N, El-Ella DMA, Khatoon S, Shafique K, Raza A, Mathkor DM, Haque S. Cellular plasticity and non-small cell lung cancer: role of T and NK cell immune evasion and acquisition of resistance to immunotherapies. Cancer Metastasis Rev 2025; 44:27. [PMID: 39856479 DOI: 10.1007/s10555-025-10244-8] [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: 11/05/2023] [Accepted: 01/08/2025] [Indexed: 01/27/2025]
Abstract
Lung cancer is a leading global cause of mortality, with non-small cell lung cancer (NSCLC) accounting for a significant portion of cases. Immune checkpoint inhibitors (ICIs) have transformed NSCLC treatment; however, many patients remain unresponsive. ICI resistance in NSCLC and its association with cellular plasticity, epithelial-mesenchymal transition (EMT), enhanced adaptability, invasiveness, and resistance is largely influenced by epigenetic changes, signaling pathways, tumor microenvironment, and associated immune cells, fibroblasts, and cytokines. Immunosuppressive cells, including M2 tumor-associated macrophages, myeloid-derived suppressor cells, and regulatory T cells, contribute to resistance by suppressing the immune response. This cellular plasticity is influenced when B cells, natural killer cells, and T cells are exhausted or inhibited by components of the tumor microenvironment. Conversely, diverse T cell, NK cell, and B cell subsets hold potential as predictive response markers particularly cytotoxic CD8+ T cells, effector memory T cells, activated T cells, tumor infiltrated NK cells, tertiary lymphoid structures, etc. influence treatment response. Identifying specific gene expressions and immunophenotypes within T cells may offer insights into early clinical responses to immunotherapy. ICI resistance in NSCLC is a multifaceted process shaped by tumor plasticity, the complex tumor microenvironment, and dynamic immune cell changes. Comprehensive analysis of these factors may lead to the identification of novel biomarkers and combination therapies to enhance ICI efficacy in NSCLC treatment.
Collapse
Affiliation(s)
- Sarra Mestiri
- Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Ana Sami
- Queen Mary University of London, London, UK
| | - Naresh Sah
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, USA
| | - Dina Moustafa Abo El-Ella
- Translational Cancer Research Facility, National Center for Cancer Care and Research/Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Sabiha Khatoon
- Department of Physiology and Biochemistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Khadija Shafique
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE
| | - Afsheen Raza
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, UAE.
| | - Darin Mansor Mathkor
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia.
- Universidad Espiritu Santo, Samborondon, Ecuador.
| |
Collapse
|
2
|
Chen H, Chen Y, Chung W, Loh Z, Lee K, Hsu H. Circulating CD3 +CD8 + T Lymphocytes as Indicators of Disease Status in Patients With Early Breast Cancer. Cancer Med 2025; 14:e70547. [PMID: 39749673 PMCID: PMC11696249 DOI: 10.1002/cam4.70547] [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: 06/04/2024] [Revised: 12/06/2024] [Accepted: 12/15/2024] [Indexed: 01/04/2025] Open
Abstract
Circulating CD3+CD8+ cell levels were lower in breast cancer patients, elevated posttreatment, and subsequently declining upon recurrence. Elevated plasma chemokine (C-C motif) ligand 2 (CCL2) levels distinguished patients with breast cancer from healthy controls. In summary, circulating CD3+CD8+ CTL and plasma CCL2 levels emerged as promising dual-purpose biomarkers and therapeutic targets in breast cancer management.
Collapse
Affiliation(s)
- Han‐Kun Chen
- Department of SurgeryChi Mei Medical CenterTainanTaiwan
- Department of NursingMeiho UniversityPingtungTaiwan
| | - Yi‐Ling Chen
- Department of Health and NutritionChia Nan University of Pharmacy and ScienceTainanTaiwan
| | - Wei‐Pang Chung
- Department of Oncology, College of MedicineNational Cheng Kung University Hospital, National Cheng Kung UniversityTainanTaiwan
- Center of Applied NanomedicineNational Cheng Kung UniversityTainanTaiwan
| | - Zhu‐Jun Loh
- Department of Surgery, College of MedicineNational Cheng Kung University Hospital, National Cheng Kung UniversityTainanTaiwan
| | - Kuo‐Ting Lee
- Department of Surgery, College of MedicineNational Cheng Kung University Hospital, National Cheng Kung UniversityTainanTaiwan
| | - Hui‐Ping Hsu
- Department of Surgery, College of MedicineNational Cheng Kung University Hospital, National Cheng Kung UniversityTainanTaiwan
- Department of Biochemistry and Molecular BiologyCollege of Medicine, National Cheng Kung UniversityTainanTaiwan
| |
Collapse
|
3
|
Wang Y, Sun C, Liu M, Xu P, Li Y, Zhang Y, Huang J. Dysregulated gene expression of SUMO machinery components induces the resistance to anti-PD-1 immunotherapy in lung cancer by upregulating the death of peripheral blood lymphocytes. Front Immunol 2024; 15:1424393. [PMID: 39211047 PMCID: PMC11357960 DOI: 10.3389/fimmu.2024.1424393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 07/30/2024] [Indexed: 09/04/2024] Open
Abstract
Background The majority of patients with lung cancer exhibit drug resistance after anti-PD-1 immunotherapy, leading to shortened patient survival time. Previous studies have suggested an association between epigenetic abnormalities such as methylation and clinical response to anti-PD-1 immunotherapy, while the role of SUMOylation in resistance to anti-PD-1 antibody immunotherapy is still unclear. Methods Here, the mRNA expression of 15 SUMO machinery components in PBMC from lung cancer patients receiving anti-PD-1 immunotherapy were analyzed using real-time PCR. Base on the percentage change in mRNA levels, the relationship between the expression of SUMO machinery components and outcomes of anti-PD-1 immunotherapy, and the influencing factors of SUMOylation were evaluated. PBMC was treated with different concentrations of 2-D08 (a specific inhibitor of SUMOylation) in vitro, and analyzed the activation and the death rates of lymphocyte subsets by flow cytometry analysis. Results A predictive method, base on the gene expression of three SUMO machinery components (SUMO1, SUMO3 and UBE2I), were developed to distinguish non-responders to PD-1 inhibitors. Furthermore, the number of lymphocytes in peripheral blood significantly reduced in the dysregulated SUMOylation groups (the percentage change >100 or -50 ~ -100 groups). In vitro studies confirmed that lightly low SUMOylation level improved the activation status of T and NK lymphocytes, but extremely low SUMOylation level lead to the increased death rates of lymphocytes. Conclusion Our findings implied that dysregulated gene expression of SUMO machinery components could induce the resistance of anti-PD-1 immunotherapy in lung cancer by upregulating the death of peripheral blood lymphocytes. These data might provide effective circulating biomarkers for predicting the efficacy of anti-PD-1 immunotherapy, and uncovered a novel regulatory mechanism of resistance to anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Ying Wang
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Chao Sun
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Mengmeng Liu
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Panyang Xu
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yanyan Li
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yongsheng Zhang
- Prenatal Diagnosis Center, Reproductive Medicine Center, The First Hospital of Jilin University, Changchun, China
| | - Jing Huang
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
4
|
Zhang X, Chen R, Huo Z, Li W, Jiang M, Su G, Liu Y, Cai Y, Huang W, Xiong Y, Wang S. Blood-based molecular and cellular biomarkers of early response to neoadjuvant PD-1 blockade in patients with non-small cell lung cancer. Cancer Cell Int 2024; 24:225. [PMID: 38951894 PMCID: PMC11218110 DOI: 10.1186/s12935-024-03412-3] [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: 02/23/2024] [Accepted: 06/22/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Despite the improved survival observed in PD-1/PD-L1 blockade therapy, a substantial proportion of cancer patients, including those with non-small cell lung cancer (NSCLC), still lack a response. METHODS Transcriptomic profiling was conducted on a discovery cohort comprising 100 whole blood samples, as collected multiple times from 48 healthy controls (including 43 published data) and 31 NSCLC patients that under treatment with a combination of anti-PD-1 Tislelizumab and chemotherapy. Differentially expressed genes (DEGs), simulated immune cell subsets, and germline DNA mutational markers were identified from patients achieved a pathological complete response during the early treatment cycles. The predictive values of mutational markers were further validated in an independent immunotherapy cohort of 1661 subjects, and then confirmed in genetically matched lung cancer cell lines by a co-culturing model. RESULTS The gene expression of hundreds of DEGs (FDR p < 0.05, fold change < -2 or > 2) distinguished responders from healthy controls, indicating the potential to stratify patients utilizing early on-treatment features from blood. PD-1-mediated cell abundance changes in memory CD4 + and regulatory T cell subset were more significant or exclusively observed in responders. A panel of top-ranked genetic alterations showed significant associations with improved survival (p < 0.05) and heightened responsiveness to anti-PD-1 treatment in patient cohort and co-cultured cell lines. CONCLUSION This study discovered and validated peripheral blood-based biomarkers with evident predictive efficacy for early therapy response and patient stratification before treatment for neoadjuvant PD-1 blockade in NSCLC patients.
Collapse
Affiliation(s)
- Xi Zhang
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China.
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 710069, Shaanxi, Xi'an, China.
| | - Rui Chen
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Zirong Huo
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Wenqing Li
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Mengju Jiang
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Guodong Su
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Yuru Liu
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Yu Cai
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
| | - Wuhao Huang
- Department of Lung Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, 300060, China
| | - Yuyan Xiong
- School of Life Science, Northwest University, Xi'an, Shaanxi, 710069, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, 710069, Shaanxi, Xi'an, China
| | - Shengguang Wang
- Department of Lung Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Lung Cancer Center, Tianjin, 300060, China.
| |
Collapse
|
5
|
Dalle S, Verronese E, N’Kodia A, Bardin C, Rodriguez C, Andrieu T, Eberhardt A, Chemin G, Hasan U, Le-Bouar M, Caramel J, Amini-Adle M, Bendriss-Vermare N, Dubois B, Caux C, Ménétrier-Caux C. Modulation of blood T cell polyfunctionality and HVEM/BTLA expression are critical determinants of clinical outcome in anti-PD1-treated metastatic melanoma patients. Oncoimmunology 2024; 13:2372118. [PMID: 38939518 PMCID: PMC11210932 DOI: 10.1080/2162402x.2024.2372118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 06/20/2024] [Indexed: 06/29/2024] Open
Abstract
The need for reliable biomarkers to predict clinical benefit from anti-PD1 treatment in metastatic melanoma (MM) patients remains unmet. Several parameters have been considered in the tumor environment or the blood, but none has yet achieved sufficient accuracy for routine clinical practice. Whole blood samples from MM patients receiving second-line anti-PD1 treatment (NCT02626065), collected longitudinally, were analyzed by flow cytometry to assess the immune cell subsets absolute numbers, the expression of immune checkpoints or ligands on T cells and the functionality of innate immune cells and T cells. Clinical response was assessed according to Progression-Free Survival (PFS) status at one-year following initiation of anti-PD1 (responders: PFS > 1 year; non-responders: PFS ≤ 1 year). At baseline, several phenotypic and functional alterations in blood immune cells were observed in MM patients compared to healthy donors, but only the proportion of polyfunctional memory CD4+ T cells was associated with response to anti-PD1. Under treatment, a decreased frequency of HVEM on CD4+ and CD8+ T cells after 3 months of treatment identified responding patients, whereas its receptor BTLA was not modulated. Both reduced proportion of CD69-expressing CD4+ and CD8+ T cells and increased number of polyfunctional blood memory T cells after 3 months of treatment were associated with response to anti-PD1. Of upmost importance, the combination of changes of all these markers accurately discriminated between responding and non-responding patients. These results suggest that drugs targeting HVEM/BTLA pathway may be of interest to improve anti-PD1 efficacy.
Collapse
Affiliation(s)
- Stéphane Dalle
- Department of Dermatology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon 1 University, Lyon, France
- Cancer Research Center of Lyon, INSERM 1052 - CNRS 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Estelle Verronese
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| | - Axelle N’Kodia
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| | - Christine Bardin
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| | - Céline Rodriguez
- Cancer Research Center of Lyon, INSERM 1052 - CNRS 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| | - Thibault Andrieu
- Cancer Research Center of Lyon, INSERM 1052 - CNRS 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| | - Anais Eberhardt
- Department of Dermatology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon 1 University, Lyon, France
- Cancer Research Center of Lyon, INSERM 1052 - CNRS 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Gabriel Chemin
- Cancer Research Center of Lyon, INSERM 1052 - CNRS 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Uzma Hasan
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| | - Myrtille Le-Bouar
- Department of Dermatology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon 1 University, Lyon, France
| | - Julie Caramel
- Cancer Research Center of Lyon, INSERM 1052 - CNRS 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Mona Amini-Adle
- Department of Dermatology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, Lyon 1 University, Lyon, France
| | - Nathalie Bendriss-Vermare
- Cancer Research Center of Lyon, INSERM 1052 - CNRS 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| | - Bertrand Dubois
- Cancer Research Center of Lyon, INSERM 1052 - CNRS 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| | - Christophe Caux
- Cancer Research Center of Lyon, INSERM 1052 - CNRS 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| | - Christine Ménétrier-Caux
- Cancer Research Center of Lyon, INSERM 1052 - CNRS 5286, Centre Léon Bérard, Université de Lyon, Université Claude Bernard Lyon 1, Lyon, France
- Lyon Immunotherapy for Cancer Laboratory (LICL), Centre Léon Bérard, Lyon, France
| |
Collapse
|
6
|
Qu J, Wu B, Chen L, Wen Z, Fang L, Zheng J, Shen Q, Heng J, Zhou J, Zhou J. CXCR6-positive circulating mucosal-associated invariant T cells can identify patients with non-small cell lung cancer responding to anti-PD-1 immunotherapy. J Exp Clin Cancer Res 2024; 43:134. [PMID: 38698468 PMCID: PMC11067263 DOI: 10.1186/s13046-024-03046-3] [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: 02/19/2024] [Accepted: 04/13/2024] [Indexed: 05/05/2024] Open
Abstract
BACKGROUND Mucosal-associated invariant T (MAIT) cells have been reported to regulate tumor immunity. However, the immune characteristics of MAIT cells in non-small cell lung cancer (NSCLC) and their correlation with the treatment efficacy of immune checkpoint inhibitors (ICIs) remain unclear. PATIENTS AND METHODS In this study, we performed single-cell RNA sequencing (scRNA-seq), flow cytometry, and multiplex immunofluorescence assays to determine the proportion and characteristics of CD8+MAIT cells in patients with metastatic NSCLC who did and did not respond to anti-PD-1 therapy. Survival analyses were employed to determine the effects of MAIT proportion and C-X-C chemokine receptor 6 (CXCR6) expression on the prognosis of patients with advanced NSCLC. RESULTS The proportion of activated and proliferating CD8+MAIT cells were significantly higher in responders-derived peripheral blood mononuclear cells (PBMCs) and lung tissues before anti-PD-1 therapy, with enhanced expression of cytotoxicity-related genes including CCL4, KLRG1, PRF1, NCR3, NKG7, GZMB, and KLRK1. The responders' peripheral and tumor-infiltrating CD8+MAIT cells showed an upregulated CXCR6 expression. Similarly, CXCR6+CD8+MAIT cells from responders showed higher expression of cytotoxicity-related genes, such as CST7, GNLY, KLRG1, NKG7, and PRF1. Patients with ≥15.1% CD8+MAIT cells to CD8+T cells ratio and ≥35.9% CXCR6+CD8+MAIT cells to CD8+MAIT cells ratio in peripheral blood showed better progression-free survival (PFS) after immunotherapy. The role of CD8+MAIT cells in lung cancer immunotherapy was potentially mediated by classical/non-classical monocytes through the CXCL16-CXCR6 axis. CONCLUSION CD8+MAIT cells are a potential predictive biomarker for patients with NSCLC responding to anti-PD-1 therapy. The correlation between CD8+MAIT cells and immunotherapy sensitivity may be ascribed to high CXCR6 expression.
Collapse
Affiliation(s)
- Jingjing Qu
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| | - Binggen Wu
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| | - Lijun Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P.R. China
| | - Zuoshi Wen
- Department of Cardiology, The First Affiliated Hospital, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
| | - Liangjie Fang
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| | - Jing Zheng
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| | - Qian Shen
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| | - Jianfu Heng
- Department of Clinical Pharmaceutical Research Institution, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, P. R. China.
| | - Jianya Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China.
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China.
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China
- The Clinical Research Center for Respiratory Diseases of Zhejiang Province, Hangzhou, Zhejiang, 310003, P. R. China
| |
Collapse
|
7
|
Tsai YT, Schlom J, Donahue RN. Blood-based biomarkers in patients with non-small cell lung cancer treated with immune checkpoint blockade. J Exp Clin Cancer Res 2024; 43:82. [PMID: 38493133 PMCID: PMC10944611 DOI: 10.1186/s13046-024-02969-1] [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: 12/05/2023] [Accepted: 01/30/2024] [Indexed: 03/18/2024] Open
Abstract
The paradigm of non-small cell lung cancer (NSCLC) treatment has been profoundly influenced by the development of immune checkpoint inhibitors (ICI), but the range of clinical responses observed among patients poses significant challenges. To date, analyses of tumor biopsies are the only parameter used to guide prognosis to ICI therapy. Tumor biopsies, however, are often difficult to obtain and tissue-based biomarkers are limited by intratumoral heterogeneity and temporal variability. In response, there has been a growing emphasis on the development of "liquid biopsy"‒ derived biomarkers, which offer a minimally invasive means to dynamically monitor the immune status of NSCLC patients either before and/or during the course of treatment. Here we review studies in which multiple blood-based biomarkers encompassing circulating soluble analytes, immune cell subsets, circulating tumor DNA, blood-based tumor mutational burden, and circulating tumor cells have shown promising associations with the clinical response of NSCLC patients to ICI therapy. These investigations have unveiled compelling correlations between the peripheral immune status of patients both before and during ICI therapy and patient outcomes, which include response rates, progression-free survival, and overall survival. There is need for rigorous validation and standardization of these blood-based assays for broader clinical application. Integration of multiple blood-based biomarkers into comprehensive panels or algorithms also has the potential to enhance predictive accuracy. Further research aimed at longitudinal monitoring of circulating biomarkers is also crucial to comprehend immune dynamics and resistance mechanisms and should be used alongside tissue-based methods that interrogate the tumor microenvironment to guide treatment decisions and may inform on the development of novel therapeutic strategies. The data reviewed here reinforce the opportunity to refine patient stratification, optimize treatments, and improve outcomes not only in NSCLC but also in the wider spectrum of solid tumors undergoing immunotherapy.
Collapse
Affiliation(s)
- Yo-Ting Tsai
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey Schlom
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Renee N Donahue
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|