1
|
Zang T, Luo X, Mo Y, Lin J, Lu W, Li Z, Zhou Y, Chen S. A novel model for predicting immunotherapy response and prognosis in NSCLC patients. Cancer Cell Int 2025; 25:178. [PMID: 40375214 PMCID: PMC12083170 DOI: 10.1186/s12935-025-03800-3] [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: 01/20/2025] [Accepted: 04/24/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND How to screen beneficiary populations has always been a clinical challenge in the treatment of non-small-cell lung cancer (NSCLC) with immune checkpoint inhibitors (ICIs). Routine blood tests, due to their advantages of being minimally invasive, convenient, and capable of reflecting tumor dynamic changes, have potential value in predicting the efficacy of ICIs treatment. However, there are few models based on routine blood tests to predict the efficacy and prognosis of immunotherapy. METHODS Patients were randomly divided into training cohort and validation cohort at a ratio of 2:1. The random forest algorithm was applied to select important variables based on routine blood tests, and a random forest (RF) model was constructed to predict the efficacy and prognosis of ICIs treatment. For efficacy prediction, we assessed receiver operating characteristic (ROC) curves, decision curve analysis (DCA) curves, clinical impact curve (CIC), integrated discrimination improvement (IDI) and net reclassification improvement (NRI) compared with the Nomogram model. For prognostic evaluation, we utilized the C-index and time-dependent C-index compared with the Nomogram model, Lung Immune Prognostic Index (LIPI) and Systemic Inflammatory Score (SIS). Patients were classified into high-risk and low-risk groups based on RF model, then the Kaplan-Meier (K-M) curve was used to analyze the differences in progression-free survival (PFS) and overall survival (OS) of patients between the two groups. RESULTS The RF model incorporated RDW-SD, MCV, PDW, CD3+CD8+, APTT, P-LCR, Ca, MPV, CD4+/CD8+ ratio, and AST. In the training and validation cohorts, the RF model exhibited an AUC of 1.000 and 0.864, and sensitivity/specificity of (100.0%, 100.0%) and (70.3%, 93.5%), respectively, which had superior performance compared to the Nomogram model (training cohort: AUC = 0.531, validation cohort: AUC = 0.552). The C-index of the RF model was 0.803 in the training cohort and 0.712 in the validation cohort, which was significantly higher than Nomogram model, LIPI and SIS. K-M survival curves revealed that patients in the high-risk group had significantly shorter PFS/OS than those in the low-risk group. CONCLUSIONS In this study, we developed a novel model (RF model) to predict the response to immunotherapy and prognosis in NSCLC patients. The RF model demonstrated better predictive performance for immunotherapy responses than the Nomogram model. Moreover, when predicting the prognosis of immunotherapy, it outperformed the Nomogram model, LIPI, and SIS.
Collapse
Affiliation(s)
- Ting Zang
- The First Clinical Medical College and the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
| | - Xiaorong Luo
- The First Clinical Medical College and the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
| | - Yangyu Mo
- The First Clinical Medical College and the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
| | - Jietao Lin
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
- Baiyun Hospital of The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510470, Guangdong, People's Republic of China
| | - Weiguo Lu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China
| | - Zhiling Li
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, Guangdong, People's Republic of China.
| | - Yingchun Zhou
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, Guangdong, People's Republic of China.
- Baiyun Hospital of The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510470, Guangdong, People's Republic of China.
| | - Shulin Chen
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, Guangdong, People's Republic of China.
| |
Collapse
|
2
|
Huang P, Zhao M, Xia J, Li H, Sun J, Li X, Yang C, Gao G, Zhou W, Zhong M, Yong H. IL-6 is a prognostic biomarker in patients with advanced esophageal squamous cell carcinoma received with PD-1 inhibitors. Front Immunol 2025; 16:1569042. [PMID: 40433391 PMCID: PMC12106429 DOI: 10.3389/fimmu.2025.1569042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 04/23/2025] [Indexed: 05/29/2025] Open
Abstract
Background Due to the low efficacy rates, effective biomarkers are desperately needed to determine populations of advanced esophageal squamous cell carcinoma (ESCC) that may benefit from immune checkpoint inhibitor (ICI) treatment. Objectives To explore the relationship between IL-6 and the esophageal cancer tumor immune microenvironment using online databases and esophageal cancer tissue microarrays and to investigate the predictive role of IL-6 for immunotherapy in esophageal squamous carcinoma patients through clinical study data. Methods RNA-seq datasets of ESCC patients were obtained from TCGA, and the relationship between IL-6 and immune cells was discovered using TIMER 2.0 databases. CD8, IL-6, and PD-L1 expression in ESCC tissue microarrays were measured using immunohistochemistry, and then the tumor microenvironment was classified. Furthermore, blood specimens were collected from advanced ESCC patients before they received PD-1 inhibitors, and follow-up was conducted to gather clinical survival data. Based on IL-6 levels. We divided the population into the high and low IL-6 groups, comparing the efficacy and survival of the two groups. Results IL-6 positively correlated with mRNA levels of PD-L1, negatively correlated with immune cells, and positively correlated with immunosuppressive cells. High IL-6 expression in tissues might make PD-1/L1 blockade therapy less effective. Individuals with higher baseline plasma IL-6 levels had significantly lower objective remission rates and inferior PFS and OS. Elevated baseline IL-6 was demonstrated to be an independent risk factor for the prognosis of advanced ESCC patients using PD-1 inhibitors, according to COX regression analysis. Conclusion IL-6 overexpression correlates with the immunosuppressive tumor microenvironment in ESCC, and it can be a predictive biomarker in ESCC patients received with PD-1 inhibitors.
Collapse
Affiliation(s)
- Ping Huang
- Department of Medical Oncology, Huai’an Hospital Affiliated to Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Manyi Zhao
- Department of Medical Oncology, Huai’an Hospital Affiliated to Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Jianhong Xia
- Department of Radiation Oncology, Huai’an Hospital Affiliated to Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Hongliang Li
- Department of Radiation Oncology, Huai’an Hospital Affiliated to Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Junxia Sun
- Department of Radiation Oncology, Huai’an Hospital Affiliated to Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Xin Li
- Department of Radiation Oncology, Huai’an Hospital Affiliated to Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Chunsheng Yang
- Department of Dermatology, Huai’an Hospital Affiliated to Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Guangyi Gao
- Department of Medical Oncology, Huai’an Hospital Affiliated to Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Wenhang Zhou
- Department of Medical Oncology, Huai’an Hospital Affiliated to Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Meifeng Zhong
- Department of Pediatrics, Huai’an Hospital Affiliated to Xuzhou Medical University, Huai’an, Jiangsu, China
| | - Hongmei Yong
- Department of Medical Oncology, Huai’an Hospital Affiliated to Xuzhou Medical University, Huai’an, Jiangsu, China
| |
Collapse
|
3
|
Dai L, Huang L, Li L, Tang L, Yao J, Shi Y, Han X. Pretreatment plasma sCD14 as a prognostic indicator in advanced non-small cell lung cancer patients undergoing immunotherapy. BMC Cancer 2025; 25:763. [PMID: 40269765 PMCID: PMC12016321 DOI: 10.1186/s12885-025-14148-2] [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: 02/11/2025] [Accepted: 04/14/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND This study aims to evaluate cytokines as a prognostic biomarker in patients with advanced non-small cell lung cancer (aNSCLC) undergoing immunotherapy. METHODS A comprehensive analysis was conducted to assess the prognostic significance of sCD14 and other cytokines in aNSCLC patients receiving immune checkpoint inhibitors (ICIs) using flow fluorescence. A discovery cohort (n = 42) was used to evaluate the differential expression of 41 cytokines between durable clinical benefit (DCB) and no durable benefit (NDB) groups in Cancer Hospital, Chinese Academy of Medical Sciences (CHCAMS). The prognostic value was further validated in multiple independent cohorts, including plasma protein measurements (n = 109), multiplex immunofluorescence (mIF) (n = 22), and messenger RNA datasets (n = 403) of NSCLC in CHCAMS. RESULTS In the discovery cohort, 7 cytokines (CD14, CCL27, IL-17 A, EGF, TNFR1, GFAP, CHI3L1) exhibited differential expression between the DCB and NDB groups. Among these, CD14, CCL27, IL-17 A, and TNFR1 were significantly elevated in the DCB group, while EGF, CHI3L1, and CCL5 were higher in the NDB group. CD14 showed a high area under the curve (AUC = 0.84) for predicting clinical benefit. Functional enrichment analysis indicated that these cytokines are involved in key immune pathways, including the inflammatory response and MAPK signaling. Univariate COX for progression-free survival (PFS) analysis demonstrated prognostic value for CD14 (p < 0.001, HR = 0.054 [0.014-0.219]), CCL27 (p < 0.001, HR = 0.054 [0.015-0.196]), IL-17 A (p < 0.001, HR = 0.110 [0.041-0.298]), and CCL5 (p < 0.05, HR = 2.387 [1.023-5.570]). Validation in the CHCAMS cohort confirmed that CD14 expression, measured via mIF, was a predictor of PFS (p < 0.05). Furthermore, high CD14 expression was consistently associated with superior PFS across multiple external datasets (GSE126044, GSE135222, GSE136961, and GSE218989). CD14 expression was found to be elevated in various normal tissue types, particularly in lung adenocarcinoma and lung squamous cell carcinoma, compared to tumors, indicating its potential role in immune surveillance. CONCLUSION sCD14 is a promising prognostic biomarker for aNSCLC patients undergoing immunotherapy. Elevated plasma sCD14 levels are associated with improved PFS and a favorable immune response.
Collapse
Affiliation(s)
- Liyuan Dai
- Department of Medical Oncology, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases;National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Liling Huang
- Department of Medical Oncology, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases;National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Lin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Le Tang
- Department of Medical Oncology, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases;National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Jiarui Yao
- Department of Medical Oncology, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases;National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yuankai Shi
- Department of Medical Oncology, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases;National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital,, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| | - Xiaohong Han
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Key Technologies for Early Clinical Trial Evaluation of Innovative Drugs for Major Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, No.1, Shuaifuyuan, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
4
|
Toney NJ, Lynch MT, Lynce F, Mainor C, Isaacs C, Schlom J, Donahue RN. Serum analytes as predictors of disease recurrence and the duration of invasive disease-free survival in patients with triple negative breast cancer enrolled in the OXEL trial treated with immunotherapy, chemotherapy, or chemoimmunotherapy. J Immunother Cancer 2025; 13:e011379. [PMID: 40274284 PMCID: PMC12020768 DOI: 10.1136/jitc-2024-011379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/07/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND The OXEL study (NCT03487666) was a phase II trial of patients with triple negative breast cancer (TNBC) with residual disease following neoadjuvant chemotherapy, randomized to receive immunotherapy (anti-programmed cell death protein 1, nivolumab), chemotherapy (capecitabine), or chemoimmunotherapy. We previously reported on the primary endpoint of the OXEL trial, demonstrating that a peripheral immunoscore based on circulating immune cells reflecting immune activation was increased in patients treated with immunotherapy. However, compared with cell-based immune assays, sera assays are more cost-effective, less labor-intensive, and samples easier to obtain. Here, we report on differences in serum analytes between treatment arms and associations with clinical response. METHODS Patients (n=38) were assayed for 97 serum analytes before and after 6 and 12 weeks of therapy. Serum analytes were assessed for changes with therapy, and as predictors of disease recurrence and the duration of invasive disease-free survival (iDFS) in both single analyte analyses and machine learning models. RESULTS Levels of specific analytes at baseline and changes in levels at early time points on treatment preceding recurrence were associated with eventual development of disease recurrence and/or the duration of iDFS. These associations varied based on the therapy patients received. Immunotherapy led to enrichment in pro-inflammatory analytes following treatment, whereas chemotherapy resulted in overall decreases. Changes seen in patients receiving chemoimmunotherapy more closely resembled those observed in patients receiving immunotherapy alone as opposed to chemotherapy alone. Furthermore, logistic regression and Cox proportional hazard models, developed using machine learning methods, demonstrated that combinations of serum analytes were more predictive of disease recurrence and iDFS duration than analyses of single serum analytes. Notably, the multivariable models that predicted patient outcomes were highly specific to the class of treatment patients received. CONCLUSIONS In patients with TNBC with residual disease after neoadjuvant chemotherapy, treatment with immunotherapy alone or chemoimmunotherapy resulted in enhanced immune activation compared with chemotherapy alone as measured by changes in serum analyte levels. Distinct serum analytes, both at baseline and as changes after therapy, predicted clinical outcomes for patients receiving immunotherapy alone, chemotherapy alone, or chemoimmunotherapy. TRIAL REGISTRATION NUMBER NCT03487666.
Collapse
Affiliation(s)
- Nicole J Toney
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Megan T Lynch
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Filipa Lynce
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Candace Mainor
- MedStar Georgetown University Hospital, Washington, District of Columbia, USA
| | - Claudine Isaacs
- Georgetown University, Washington, District of Columbia, USA
| | - Jeffrey Schlom
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Renee N Donahue
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
5
|
Jeong H, Koh J, Kim S, Yim J, Song SG, Kim H, Li Y, Lee SH, Chung YK, Kim H, Lee CH, Kim HY, Keam B, Lee SH, Chung DH, Jeon YK. Cell-intrinsic PD-L1 signaling drives immunosuppression by myeloid-derived suppressor cells through IL-6/Jak/Stat3 in PD-L1-high lung cancer. J Immunother Cancer 2025; 13:e010612. [PMID: 40050048 PMCID: PMC11887297 DOI: 10.1136/jitc-2024-010612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 02/24/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Some patients with non-small-cell lung cancer (NSCLC) benefit from immune checkpoint inhibitors (ICIs) despite programmed death-ligand 1 (PD-L1) expression. To address the mechanism of ICI resistance in PD-L1-positive NSCLC, we investigated the role of tumor-cell-intrinsic function of PD-L1 in interleukin (IL)-6-mediated immunosuppression. METHODS Cohorts of NSCLC patients treated with ICI and public datasets were analyzed. PD-L1-overexpressing and PD-L1-knockdown NSCLC cells were submitted to RNA-seq, in vitro analyses, chromatin immunoprecipitation-qPCR, CUT&Tag, and biochemical assays. Human myeloid-derived suppressor cells (MDSCs) sorted from peripheral blood mononuclear cells were co-cultured with NSCLC cells and then assessed for their immunosuppressive activity on T-cells. Mouse Lewis lung carcinoma (LLC) cells with PD-L1 overexpression or knockdown were subcutaneously injected into wild-type or PD-1-knockout C57BL/6 mice in the presence of IL-6 and/or PD-1 blockade. RESULTS In the ICI cohort with RNA-seq data, the IL-6/Jak/Stat3 pathway was enriched, and IL-6 expression was higher in patients with PD-L1-high NSCLCs who did not respond to ICIs. In another cohort, a higher baseline serum IL-6 level was associated with poor clinical outcomes after ICI therapy. IL-6 expression and the IL-6/Jak/Stat3 pathway were enhanced in PD-L1-high NSCLCs in the ICI cohorts and The Cancer Genome Atlas analysis. IL-6 expression correlated positively with tumor-infiltrating MDSCs in NSCLCs. In NSCLC cells, PD-L1 activated Jak2/Stat3 signaling by binding to and inhibiting protein tyrosine phosphatase 1B. PD-L1 also bound to p-Stat3 in the nucleus, thus promoting the activity of p-Stat3 in the transcription of several cytokines (IL-6, TGF-β, TNF-α, IL-1β) and chemokines. PD-L1-overexpressing NSCLC cells enhanced the migration and immunosuppressive activity of human MDSCs in vitro, mediated by IL-6 and CXCL1. In both wild-type and PD-1-knockout mice, PD-L1-overexpressing LLC tumors were infiltrated by increased MDSCs with high immunosuppressive function, increased Tregs, and decreased granzyme B+ or IFNγ+ CD8 T-cells. These responses were mediated by IL-6 secreted from PD-L1-overexpressing tumor cells. Combined blockade of PD-1 and IL-6 was effective in tumor control and decreased MDSCs while increasing granzyme B+ or IFNγ+ CD8 T-cells. CONCLUSIONS The tumor-cell-intrinsic function of PD-L1 drives immunosuppression and tumor progression through the PD-L1/Jak/Stat3/IL-6/MDSC axis. This pathway represents a potential therapeutic target to improve ICI efficacy in PD-L1-high NSCLC.
Collapse
Affiliation(s)
- Hyein Jeong
- Cancer Research Institute, Seoul National University, Seoul, Korea (the Republic of)
- Interdiscipilinary Program of Cancer Biology, Seoul National University Graduate School, Seoul, Korea (the Republic of)
| | - Jaemoon Koh
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Sehui Kim
- Department of Pathology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea (the Republic of)
| | - Jeemin Yim
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- Department of Pathology, Boramae Medical Center, Seoul National University, Seoul, Korea (the Republic of)
| | - Seung Geun Song
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Hanbyeol Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea (the Republic of)
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Yingying Li
- Cancer Research Institute, Seoul National University, Seoul, Korea (the Republic of)
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Soo Hyun Lee
- Cancer Research Institute, Seoul National University, Seoul, Korea (the Republic of)
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Yeon Kyu Chung
- Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Hongsoon Kim
- Cancer Research Institute, Seoul National University, Seoul, Korea (the Republic of)
- Interdiscipilinary Program of Cancer Biology, Seoul National University Graduate School, Seoul, Korea (the Republic of)
| | - Chul-Hwan Lee
- Cancer Research Institute, Seoul National University, Seoul, Korea (the Republic of)
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- Department of Pharmacology, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Hye Young Kim
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Korea (the Republic of)
| | - Bhumsuk Keam
- Cancer Research Institute, Seoul National University, Seoul, Korea (the Republic of)
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Gangnam-gu, Korea (the Republic of)
- Department of Health Sciences and Technology, Samsung Advanced Institute of Health Sciences and Technology, Sungkyunkwan University, Seoul, Korea (the Republic of)
| | - Doo Hyun Chung
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- BK21 FOUR Biomedical Science Project, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| | - Yoon Kyung Jeon
- Cancer Research Institute, Seoul National University, Seoul, Korea (the Republic of)
- Interdiscipilinary Program of Cancer Biology, Seoul National University Graduate School, Seoul, Korea (the Republic of)
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
- BK21 FOUR Smart Healthcare, Seoul National University College of Medicine, Seoul, Korea (the Republic of)
| |
Collapse
|
6
|
Fomin V, So WV, Barbieri RA, Hiller-Bittrolff K, Koletou E, Tu T, Gomes B, Cai J, Charo J. Machine learning identifies clinical tumor mutation landscape pathways of resistance to checkpoint inhibitor therapy in NSCLC. J Immunother Cancer 2025; 13:e009092. [PMID: 40032600 PMCID: PMC11877243 DOI: 10.1136/jitc-2024-009092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (CPIs) have revolutionized cancer therapy for several tumor indications. However, a substantial fraction of patients treated with CPIs derive no benefit or have short-lived responses to CPI therapy. Identifying patients who are most likely to benefit from CPIs and deciphering resistance mechanisms is therefore essential for developing adjunct treatments that can abrogate tumor resistance. PATIENTS AND METHODS In this study, we used a machine learning approach that used the US-based nationwide de-identified Flatiron Health and Foundation Medicine non-small cell lung carcinoma (NSCLC) clinico-genomic database to identify genomic markers that predict clinical responses to CPI therapy. In total, we analyzed data from 4,433 patients with NSCLC. RESULTS Analysis of pretreatment genomic data from 1,511 patients with NSCLC identified. Of the 36 genomic signatures identified, 33 exhibited strong predictive capacity for CPI response (n=1150) compared with chemotherapy response (n=361), while three signatures were prognostic. These 36 genetic signatures had in common a core set of four genes (BRAF, BRIP1, FGF10, and FLT1). Interestingly, we observed that some (n=19) of the genes in the signatures (eg, TP53, EZH2, KEAP1 and FGFR2) had alternative mutations with contrasting clinical outcomes to CPI therapy. Finally, the genetic signatures revealed multiple biological pathways involved in CPI response, including MAPK, PDGF, IL-6 and EGFR signaling. CONCLUSIONS In summary, we found several genomic markers and pathways that provide insight into biological mechanisms affecting response to CPI therapy. The analyses identified novel targets and biomarkers that have the potential to provide candidates for combination therapies or patient enrichment strategies, which could increase response rates to CPI therapy in patients with NSCLC.
Collapse
Affiliation(s)
- Vitalay Fomin
- Roche Pharmaceutical Research and Early Development, Data & Analytics, Roche Innovation Center New York, Little Falls, New Jersey, USA
- Numenos, New York, NY, USA
| | - WeiQing Venus So
- Roche Pharmaceutical Research and Early Development, Data & Analytics, Roche Innovation Center New York, Little Falls, New Jersey, USA
| | | | | | - Elina Koletou
- Roche Pharmaceutical Research and Early Development, Data and Analytics, Roche Innovation Center Basel, Basel, Switzerland
| | - Tiffany Tu
- Roche Pharmaceutical Research and Early Development, Data & Analytics, Roche Innovation Center New York, Little Falls, New Jersey, USA
| | - Bruno Gomes
- Roche Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Basel, Basel, Switzerland
| | - James Cai
- Roche Pharmaceutical Research and Early Development, Data & Analytics, Roche Innovation Center New York, Little Falls, New Jersey, USA
| | - Jehad Charo
- Roche Pharmaceutical Research and Early Development Oncology, Roche Innovation Center Zurich, Schlieren, Switzerland
| |
Collapse
|
7
|
Liu Q, Shaibu Z, Xu A, Yang F, Cao R, Yang F. Predictive value of serum cytokines in patients with non-small-cell lung cancer receiving anti-PD-1 blockade therapy: a meta-analysis. Clin Exp Med 2025; 25:59. [PMID: 39955679 PMCID: PMC11830645 DOI: 10.1007/s10238-025-01587-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/03/2025] [Indexed: 02/17/2025]
Abstract
Non-small cell lung cancer (NSCLC) is a leading cause of cancer-related deaths worldwide. Immunotherapy, particularly PD-1 inhibitors, has revolutionized the treatment landscape for NSCLC. However, the predictive biomarkers for PD-1 inhibitor therapy are still limited. Serum cytokines have emerged as potential biomarkers for predicting treatment outcomes. This meta-analysis aims to investigate the predictive value of serum cytokines in PD-1 inhibitor therapy for NSCLC. We conducted a comprehensive literature search in major databases, including PubMed, Google scholar, Embase, and Cochrane database, with a focus on literature published up until October 22, 2024. Studies investigating the association between serum cytokine levels and treatment outcomes in NSCLC patients receiving PD-1 inhibitor therapy were included. The primary outcomes were progression-free survival (PFS) and overall survival (OS). The meta-analysis revealed that elevated IL-6 levels were significantly associated with poorer PFS in NSCLC patients (HR = 2.30, 95% CI [1.39-3.80], P = 0.001). Additionally, high IL-10 expression was related to poorer PFS in NSCLC after therapy (HR = 2.45, 95% CI [1.26-4.76], P = 0.009). In contrast, no significant associations were found between OS and the expression of various cytokines, including IL-4, IL-5, IL-6, IL-8, IL-10, IFN-γ, IL-1β, TNF-α, and IL-12p70. This meta-analysis demonstrates that elevated IL-6 and IL-10 levels are significantly associated with poorer PFS in NSCLC patients receiving PD-1 inhibitor therapy. These findings suggest that serum cytokine levels may serve as predictive biomarkers for treatment outcomes. Further studies are needed to validate these results and explore the underlying mechanisms.
Collapse
Affiliation(s)
- Qian Liu
- Department of Laboratory Medicine, Lianyungang Clinical College Jiangsu University & the Second People'S Hospital of Lianyungang, 41 East Hailian Road, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College Xuzhou Medical University & the Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, The Second People'S Hospital of Lianyungang Affiliated with Kangda College of Nanjing Medical University, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College, Bengbu Medical University & the Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China
| | - Zakari Shaibu
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Aiguo Xu
- Department of Oncology, The Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China
| | - Fang Yang
- Department of Laboratory Medicine, Lianyungang Clinical College Jiangsu University & the Second People'S Hospital of Lianyungang, 41 East Hailian Road, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College Xuzhou Medical University & the Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, The Second People'S Hospital of Lianyungang Affiliated with Kangda College of Nanjing Medical University, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College, Bengbu Medical University & the Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China
| | - Ruoxue Cao
- Department of Laboratory Medicine, Lianyungang Clinical College Jiangsu University & the Second People'S Hospital of Lianyungang, 41 East Hailian Road, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College Xuzhou Medical University & the Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, The Second People'S Hospital of Lianyungang Affiliated with Kangda College of Nanjing Medical University, Lianyungang, 222006, Jiangsu, China
- Department of Laboratory Medicine, Lianyungang Clinical College, Bengbu Medical University & the Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China
| | - Fumeng Yang
- Department of Laboratory Medicine, Lianyungang Clinical College Jiangsu University & the Second People'S Hospital of Lianyungang, 41 East Hailian Road, Lianyungang, 222006, Jiangsu, China.
- Department of Laboratory Medicine, Lianyungang Clinical College Xuzhou Medical University & the Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China.
- Department of Laboratory Medicine, The Second People'S Hospital of Lianyungang Affiliated with Kangda College of Nanjing Medical University, Lianyungang, 222006, Jiangsu, China.
- Department of Laboratory Medicine, Lianyungang Clinical College, Bengbu Medical University & the Second People'S Hospital of Lianyungang, Lianyungang, 222006, Jiangsu, China.
| |
Collapse
|
8
|
Zhou H, Zheng Z, Fan C, Zhou Z. Mechanisms and strategies of immunosenescence effects on non-small cell lung cancer (NSCLC) treatment: A comprehensive analysis and future directions. Semin Cancer Biol 2025; 109:44-66. [PMID: 39793777 DOI: 10.1016/j.semcancer.2025.01.001] [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: 09/20/2024] [Revised: 12/29/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
Non-small cell lung cancer (NSCLC), the most prevalent form of lung cancer, remains a leading cause of cancer-related mortality worldwide, particularly among elderly individuals. The phenomenon of immunosenescence, characterized by the progressive decline in immune cell functionality with aging, plays a pivotal role in NSCLC progression and contributes to the diminished efficacy of therapeutic interventions in older patients. Immunosenescence manifests through impaired immune surveillance, reduced cytotoxic responses, and increased chronic inflammation, collectively fostering a pro-tumorigenic microenvironment. This review provides a comprehensive analysis of the molecular, cellular, and genetic mechanisms of immunosenescence and its impact on immune surveillance and the tumor microenvironment (TME) in NSCLC. We explore how aging affects various immune cells, including T cells, B cells, NK cells, and macrophages, and how these changes compromise the immune system's ability to detect and eliminate tumor cells. Furthermore, we address the challenges posed by immunosenescence to current therapeutic strategies, particularly immunotherapy, which faces significant hurdles in elderly patients due to immune dysfunction. The review highlights emerging technologies, such as single-cell sequencing and CRISPR-Cas9, which offer new insights into immunosenescence and its potential as a therapeutic target. Finally, we outline future research directions, including strategies for rejuvenating the aging immune system and optimizing immunotherapy for older NSCLC patients, with the goal of improving treatment efficacy and survival outcomes. These efforts hold promise for the development of more effective, personalized therapies for elderly patients with NSCLC.
Collapse
Affiliation(s)
- Huatao Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China
| | - Zilong Zheng
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China.
| | - Zijing Zhou
- Department of Pulmonary and Critical Care Medicine, the Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha 410011, China.
| |
Collapse
|
9
|
Narita D, Hishinuma E, Ebina-Shibuya R, Miyauchi E, Matsukawa N, Motoike IN, Kinoshita K, Koshiba S, Tsukita Y, Notsuda H, Kimura N, Saito R, Murakami K, Fujino N, Ichikawa T, Yamada M, Tamada T, Sugiura H. Histological and genetic features and therapeutic responses of lung cancers explored via the global analysis of their metabolome profile. Lung Cancer 2025; 200:108082. [PMID: 39884221 DOI: 10.1016/j.lungcan.2025.108082] [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: 08/25/2024] [Revised: 01/01/2025] [Accepted: 01/06/2025] [Indexed: 02/01/2025]
Abstract
BACKGROUND Lung cancer is the deadliest disease globally, with more than 120,000 diagnosed cases and more than 75,000 deaths annually in Japan. Several treatment options for advanced lung cancer are available, and the discovery of biomarkers will be useful for personalized medicine. Using metabolome analysis, we aimed to identify biomarkers for diagnosis and treatment response by examining the changes in metabolites associated with lung cancer progression. METHODS Plasma samples from patients with recurrent or metastatic non-small cell lung carcinomas diagnosed at Tohoku University Hospital between 2019 and 2024 were used in this study. Metabolomic analysis was performed using the Biocrates Life Sciences MxP Quant 500 kit. Multivariate, principal component, and orthogonal partial least squares discriminant analyses were performed. RESULTS The triglyceride and phosphatidylcholine concentrations were higher in the patients with early than in those with advanced lung adenocarcinomas. However, the cholesterol ester concentrations were higher for the patients with advanced lung cancer. The concentrations of hexosylceramide were higher in patients with early lung adenocarcinoma than in those with squamous cell carcinoma. Relative to epidermal growth factor receptor (EGFR)-mutation negative cases, the EGFR-mutation positive cases showed marked differences between the ceramide and triglyceride concentrations. For the best therapeutic effect of EGFR-TKI treatment, the hexosylceramide (HexCer) (d18:1/24:0), ceramide (Cer) (d18:2/22:0), and ceramide (Cer) (d18:2/24:0) concentrations were higher for the stable and progressive disease groups. The concentrations of phosphatidylcholine (PC) ae C42:2, sphingomyelin (SM) C24:1, and lysophosphatidylcholine (lysoPC) a C18:2 were higher in the partial response group treated with immune checkpoint inhibitors and chemotherapy. CONCLUSION Metabolomic analysis may be useful for the diagnosis and treatment of lung cancer and may provide clues for new therapeutic strategies. PC ae C42:2, SM C24:1, and lysoPC a C18:2 can serve as predictive biomarkers for monitoring the therapeutic effects of the combination of immune checkpoint inhibitors and chemotherapy.
Collapse
Affiliation(s)
- Daisuke Narita
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Eiji Hishinuma
- Advanced Research Center for Innovations in Next Generation Medicine, Tohoku University, Sendai, Japan; Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Risa Ebina-Shibuya
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Eisaku Miyauchi
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naomi Matsukawa
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Ikuko N Motoike
- Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Systems Bioinformatics, Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Kengo Kinoshita
- Advanced Research Center for Innovations in Next Generation Medicine, Tohoku University, Sendai, Japan; Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan; Systems Bioinformatics, Graduate School of Information Sciences, Tohoku University, Sendai, Japan
| | - Seizo Koshiba
- Advanced Research Center for Innovations in Next Generation Medicine, Tohoku University, Sendai, Japan; Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Yoko Tsukita
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hirotsugu Notsuda
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Nozomu Kimura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryota Saito
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Koji Murakami
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoya Fujino
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiro Ichikawa
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mitsuhiro Yamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tsutomu Tamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hisatoshi Sugiura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
10
|
Huang J, Zhang X, Zhang H, Li Y, Huang H, Li Z, Qiu Z, Wu H, Huang D, Xu X, Bian J. Addressing Clinical Limitations of Glutaminase Inhibitors: Novel Strategies for Osimertinib-Resistant Lung Cancer by Exploiting Glutamine Metabolic Dependency. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411479. [PMID: 39680480 PMCID: PMC11809341 DOI: 10.1002/advs.202411479] [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: 09/18/2024] [Revised: 11/12/2024] [Indexed: 12/18/2024]
Abstract
Overcoming acquired resistance to Osimertinib remains a critical challenge in treating NSCLC. This research indicates that Osimertinib-resistant cells exhibit a strong dependence on glutamine metabolism. However, targeting GLS1 shows limited anticancer effects, probably because it cannot fully block the glutamine metabolic pathway. The investigation reveals that a more effective strategy involves simultaneously inhibiting both ASCT2 and GLS1. After confirming the efficacy of this dual-targeting approach against Osimertinib-resistant cells in preclinical models, the potential of utilizing a broad-spectrum glutamine metabolism antagonist is further explored to achieve superior antitumor efficacy. DON, broad-spectrum glutamine antagonist, presents toxicity issues. Herein, the high NQO1 expression in Osimertinib-resistant NSCLC cells is leveraged to design an NQO1-responsive DON prodrug, 10e (LBJ-10e). This prodrug demonstrates superior safety compared to natural DON and greater antitumor activity against resistant tumors compared to the clinical phase II drug DRP104. These findings may address the clinical limitations of GLS1 allosteric inhibitors and underscore prodrug strategies in effectively treating Osimertinib-resistant lung cancer, providing a foundation for future clinical trials.
Collapse
Affiliation(s)
- Jiali Huang
- Jiangsu Key Laboratory of Drug Design and OptimizationDepartment of Medicinal ChemistryChina Pharmaceutical UniversityNanjingJiangsu210009China
- Department of Biomedical EngineeringSchool of EngineeringChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Xiankang Zhang
- Jiangsu Key Laboratory of Drug Design and OptimizationDepartment of Medicinal ChemistryChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Hui Zhang
- Jiangsu Key Laboratory of Drug Design and OptimizationDepartment of Medicinal ChemistryChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Yu Li
- Jiangsu Key Laboratory of Drug Design and OptimizationDepartment of Medicinal ChemistryChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Huidan Huang
- Center of Drug Screening & EvaluationWannan Medical CollegeWuhuAnhui241000China
| | - Zhiyu Li
- Jiangsu Key Laboratory of Drug Design and OptimizationDepartment of Medicinal ChemistryChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Zhixia Qiu
- Jiangsu Key Laboratory of Drug Design and OptimizationDepartment of Medicinal ChemistryChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Hongxi Wu
- Jiangsu Key Laboratory of Drug Design and OptimizationDepartment of Medicinal ChemistryChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Dechun Huang
- Department of Biomedical EngineeringSchool of EngineeringChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Xi Xu
- Jiangsu Key Laboratory of Drug Design and OptimizationDepartment of Medicinal ChemistryChina Pharmaceutical UniversityNanjingJiangsu210009China
| | - Jinlei Bian
- Jiangsu Key Laboratory of Drug Design and OptimizationDepartment of Medicinal ChemistryChina Pharmaceutical UniversityNanjingJiangsu210009China
| |
Collapse
|
11
|
Sardarabadi P, Lee KY, Sun WL, Kojabad AA, Liu CH. Investigating T Cell Immune Dynamics and IL-6's Duality in a Microfluidic Lung Tumor Model. ACS APPLIED MATERIALS & INTERFACES 2025; 17:4354-4367. [PMID: 39471283 PMCID: PMC11758792 DOI: 10.1021/acsami.4c09065] [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: 06/02/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/01/2024]
Abstract
Interleukin 6 (IL-6), produced by immune cells, is crucial in promoting T cell trafficking to infection and inflammation sites, influencing various physiological and pathological processes. Concentrations of IL-6 and other cytokines and chemokines can influence T cell differentiation and activation. Understanding the dual faces of IL-6 within the tumor microenvironment is crucial to understanding its role. A flow-based microsystem was designed to investigate CD4+ T cell activation in response to different IL-6 gradients in an under-control 3D culture. The study found that cancer cells' response to varying IL-6 concentrations was dynamic and dose-sensitive, with immune cell migration rates showing sensitivity to the IL-6 gradient. A549 cell expansion increases gradually and time-dependently with 50 ng of IL-6, while Jurkat cell migration follows a time-dependent pattern. However, when a total of 100 ng IL-6 concentration is applied, A549 cells expand rapidly, potentially influencing Jurkat cell migration. Jurkat cell mobility is lower, possibly due to increased A549 cell presence and heightened cell-cell interactions. Different IL-6 concentration gradients can modulate the expression of some CD markers like CD69 and programed cell death protein 1 in CD4+ T cells, suggesting that IL-6 concentration gradients affect immune cell phenotypes. This suggests that IL-6 plays a crucial role in activating T helper cells and may be involved in the later phases of inflammation. Also, the increased levels of IFN-γ and TNF-α highlight IL-6's impact on T cell inflammatory response. This study emphasizes the intricate effects of IL-6 on T cell activation, phenotype, cytokine production, and phenotypic heterogeneity, providing valuable insights into immune response modulation in an experimental setting.
Collapse
Affiliation(s)
- Parvaneh Sardarabadi
- Institute
of Nanoengineering and Microsystems, National
Tsing Hua University, Hsinchu 30044, Taiwan,
R.O.C
| | - Kang-Yun Lee
- Division
of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho
Hospital, Taipei Medical University, New Taipei City 235, Taiwan, R.O.C
- Division
of Pulmonary Medicine, Department of Internal Medicine, School of
Medicine, College of Medicine, Taipei Medical
University, Taipei 110, Taiwan, R.O.C
- TMU
Research Center for Thoracic Medicine, Taipei
Medical University, Taipei 110, Taiwan, R.O.C
| | - Wei-Lun Sun
- Pythia
Biotech LTD., New Taipei City 23561, Taiwan,
R.O.C
| | - Amir Asri Kojabad
- Department
of Hematology, School of Allied Medical Sciences, Iran University of Medical Sciences, Tehran 14535, Iran
| | - Cheng-Hsien Liu
- Institute
of Nanoengineering and Microsystems, National
Tsing Hua University, Hsinchu 30044, Taiwan,
R.O.C
- Department
of Power Mechanical Engineering, National
Tsing Hua University, Hsinchu 30044, Taiwan,
R.O.C
- College
of Semiconductor Research, National Tsing
Hua University, Hsinchu 30044, Taiwan, R.O.C
| |
Collapse
|
12
|
Alden SL, Charmsaz S, Li HL, Tsai HL, Danilova L, Munjal K, Brancati M, Warner A, Howe K, Griffin E, Nakazawa M, Thoburn C, Gizzi J, Hernandez A, Gross NE, Coyne EM, Hallab E, Shin SS, Durham J, Lipson EJ, Ged Y, Baretti M, Hoffman-Censits J, Seiwert TY, Guha A, Bansal S, Tang L, Chandler GS, Mohindra R, Garonce-Hediger R, Jaffee EM, Ho WJ, Kao C, Yarchoan M. Pan-tumor analysis to investigate the obesity paradox in immune checkpoint blockade. J Immunother Cancer 2025; 13:e009734. [PMID: 39832896 PMCID: PMC11748946 DOI: 10.1136/jitc-2024-009734] [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: 05/22/2024] [Accepted: 10/07/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Obesity is a risk factor for developing cancer but is also associated with improved outcomes after treatment with immune checkpoint inhibitors (ICIs), a phenomenon called the obesity paradox. To interrogate mechanisms of divergent immune responses in obese and non-obese patients, we examined the relationship among obesity status, clinical responses, and immune profiles from a diverse, pan-tumor cohort of patients treated with ICI-based therapy. METHODS From June 2021 to March 2023, we prospectively collected serial peripheral blood samples from patients with advanced or metastatic solid tumors who received ICI as standard of care at Johns Hopkins. Patients were stratified by obesity status at treatment initiation, with obesity defined as body mass index (BMI)≥30 at treatment initiation and BMI≥18.5 and <30 considered non-obese; underweight patients (BMI<18.5) were excluded. We evaluated the concentration of 37 cytokines and used cytometry by time of flight to characterize immune cell clusters and cell-surface expression markers at baseline and on-treatment. RESULTS We enrolled 94 patients, of whom 30 (32%) were obese and 64 (68%) were non-obese. Compared with non-obese patients, obese patients had superior progression-free survival (HR: 0.44 (95% CI: 0.24 to 0.81), p=0.01) and overall survival (OS) (HR: 0.24 (95% CI: 0.07 to 0.80), p=0.02). Obese patients had lower serum IL-15 levels at treatment baseline and lower on-treatment levels of IL-6, IL-8, and IL-15. Low on-treatment IL-6 was associated with improved OS (HR: 0.27 (95% CI: 0.08 to 0.88), p=0.03), as was low on-treatment IL-8 (HR: 0.19 (95% CI: 0.05 to 0.70), p=0.01). Obese patients demonstrated lower levels of T effector cells with reduced expression of cytotoxicity markers and higher expression of exhaustion markers at baseline and on-treatment. CONCLUSIONS Obese and non-obese patients with cancer have divergent immunological responses to ICIs. Obesity is associated with reduced levels of certain inhibitory cytokines and higher expression of T-cell exhaustion markers. ICI-based therapy may more effectively reverse T-cell dysfunction in obese patients, potentially contributing to the paradoxically improved responses in this population.
Collapse
Affiliation(s)
- Stephanie L Alden
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Soren Charmsaz
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Howard L Li
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Hua-Ling Tsai
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ludmila Danilova
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Cancer Convergence Institute, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kabeer Munjal
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Madelena Brancati
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Aanika Warner
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kathryn Howe
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ervin Griffin
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mari Nakazawa
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chris Thoburn
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jennifer Gizzi
- The Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alexei Hernandez
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Nicole E Gross
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Erin M Coyne
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Elsa Hallab
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Sarah S Shin
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jennifer Durham
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Evan J Lipson
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Yasser Ged
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Marina Baretti
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jean Hoffman-Censits
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Tanguy Y Seiwert
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Aditi Guha
- Genentech Inc, South San Francisco, California, USA
| | | | - Laura Tang
- Genentech Inc, South San Francisco, California, USA
| | | | | | | | - Elizabeth M Jaffee
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
- Cancer Convergence Institute, Johns Hopkins University, Baltimore, Maryland, USA
| | - Won Jin Ho
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
- Cancer Convergence Institute, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chester Kao
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Mark Yarchoan
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
- Cancer Convergence Institute, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
13
|
Shi Y, McKenery A, Dolan M, Mastri M, Hill JW, Dommer A, Benzekry S, Long M, Abrams SI, Puzanov I, Ebos JML. Acquired resistance to PD-L1 inhibition enhances a type I IFN-regulated secretory program in tumors. EMBO Rep 2025; 26:521-559. [PMID: 39663510 PMCID: PMC11772817 DOI: 10.1038/s44319-024-00333-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 11/10/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024] Open
Abstract
Therapeutic inhibition of programmed cell death ligand (PD-L1) is linked to alterations in interferon (IFN) signaling. Since IFN-regulated intracellular signaling can control extracellular secretory programs in tumors to modulate immunity, we examined IFN-related secretory changes in tumor cells following resistance to PD-L1 inhibition. Here we report an anti-PD-L1 treatment-induced secretome (PTIS) in tumor models of acquired resistance that is regulated by type I IFNs. These secretory changes can suppress activation of T cells ex vivo while diminishing tumor cell cytotoxicity, revealing that tumor-intrinsic treatment adaptations can exert broad tumor-extrinsic effects. When reimplanted in vivo, resistant tumor growth can slow or stop when PTIS components are disrupted individually, or when type I IFN signaling machinery is blocked. Interestingly, genetic and therapeutic disruption of PD-L1 in vitro can only partially recapitulate the PTIS phenotype highlighting the importance of developing in vivo-based resistance models to more faithfully mimic clinically-relevant treatment failure. Together, this study shows acquired resistance to immune-checkpoint inhibitors 'rewires' tumor secretory programs controlled by type I IFNs that, in turn, can protect from immune cell attack.
Collapse
Affiliation(s)
- Yuhao Shi
- Department of Experimental Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Amber McKenery
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Melissa Dolan
- Department of Experimental Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Michalis Mastri
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - James W Hill
- Jacobs School of Medicine and Biomedical Sciences, SUNY at Buffalo, Buffalo, USA
| | - Adam Dommer
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Sebastien Benzekry
- Computational Pharmacology and Clinical Oncology (COMPO), Inria Sophia Antipolis-Méditerranée, Centre de Recherches en Cancérologie de Marseille, Inserm U1068, CNRS UMR7258, Institut Paoli-Calmettes, Faculté de Pharmacie, Aix-Marseille University, Marseille, France
| | - Mark Long
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Scott I Abrams
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - John M L Ebos
- Department of Experimental Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA.
| |
Collapse
|
14
|
Yao J, Lin X, Zhang X, Xie M, Ma X, Bao X, Song J, Liang Y, Wang Q, Xue X. Predictive biomarkers for immune checkpoint inhibitors therapy in lung cancer. Hum Vaccin Immunother 2024; 20:2406063. [PMID: 39415535 PMCID: PMC11487980 DOI: 10.1080/21645515.2024.2406063] [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: 04/23/2024] [Revised: 09/05/2024] [Accepted: 09/15/2024] [Indexed: 10/18/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have changed the treatment mode of lung cancer, extending the survival time of patients unprecedentedly. Once patients respond to ICIs, the median duration of response is usually longer than that achieved with cytotoxic or targeted drugs. Unfortunately, there is still a large proportion of lung cancer patients do not respond to ICI. Effective biomarkers are crucial for identifying lung cancer patients who can benefit from them. The first predictive biomarker is programmed death-ligand 1 (PD-L1), but its predictive value is limited to specific populations. With the development of single-cell sequencing and spatial imaging technologies, as well as the use of deep learning and artificial intelligence, the identification of predictive biomarkers has been greatly expanded. In this review, we will dissect the biomarkers used to predict ICIs efficacy in lung cancer from the tumor-immune microenvironment and host perspectives, and describe cutting-edge technologies to further identify biomarkers.
Collapse
Affiliation(s)
- Jie Yao
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xuwen Lin
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xin Zhang
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| | - Mei Xie
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xidong Ma
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xinyu Bao
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| | - Jialin Song
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| | - Yiran Liang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Qiqi Wang
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xinying Xue
- Department of Respiratory and Critical Care, Emergency and Critical Care Medical Center, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
- Department of Respiratory and Critical Care, Shandong Second Medical University, Weifang, Shandong, China
| |
Collapse
|
15
|
Strouse J, Chan KK, Baccile R, He G, Louden DKN, Giurcanu M, Singh A, Rieth J, Abdel-Wahab N, Katsumoto TR, Singh N, Rouhani S, Reid P. Impact of steroid-sparing immunosuppressive agents on tumor outcome in the context of cancer immunotherapy with highlight on melanoma: a systematic literature review and meta-analysis. Front Immunol 2024; 15:1499478. [PMID: 39737191 PMCID: PMC11682972 DOI: 10.3389/fimmu.2024.1499478] [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: 09/20/2024] [Accepted: 11/25/2024] [Indexed: 01/01/2025] Open
Abstract
Background The impact of steroid-sparing immunosuppressive agents (SSIAs) for immune-related adverse events (irAEs) on tumor outcome is not well-known. This systematic review evaluates tumor outcomes for corticosteroid (CS) monotherapy versus CS with SSIA (CS-SSIA) for irAE treatment with a focus on melanoma. Methods Search was conducted through 1/5/23 using PubMed, Embase, Cochrane CENTRAL, and Web of Science. We included case series, retrospective/prospective observational studies and interventional clinical trials. Individual-level data was analyzed using KM curves and Cox regression for overall survival (OS) and progression free survival (PFS). Time to SSIA was treated as a time-varying exposure using landmark analysis (landmark timepoint=3 months after irAE) to account for immortal time bias. For group-level data, meta-analysis compared the use of SSIA to No SSIA for irAEs. Results Of twenty-two publications with individual-level data, 147 patients with any cancer (57 CS, 90 CS-SSIA) and 65 with melanoma (18 CS, 47 CS-SSIA) underwent landmark analysis. Twenty-two publications underwent group-level evaluation and four were included in the meta-analysis. CS-SSIA versus CS showed higher risk of all-cause mortality and progression (HR 2.75, 95%CI: 1.44-5.27, p<0.01 and HR 1.75, 95%CI: 1.07-2.85, p=0.03, respectively). Melanoma showed worse OS and PFS for CS-SSIA versus CS (HR 5.68, 95%CI: 1.31-24.67, p=0.02 and HR 2.68, 95%CI: 1.12-6.40, p=0.03, respectively). In the meta-analysis of group-level data (n=2558), we found worse OS and PFS for CS-SSIA versus No SSIA (HR 1.58, 95%CI: 1.25; 2.01, p<0.01 and 1.70, 95%CI: 1.25-2.33, p<0.01). Tumor necrosis factor-alpha inhibitors (TNFi) were the most common SSIA. In the melanoma cohort, TNFi had worse OS and PFS versus CS (HR 6.46, 95%CI: 1.43-29.19, p = 0.02 and HR 7.49, 95%CI: 2.29-24.48, p<0.01, respectively). TNFi versus Other SSIAs showed a trend toward worse OS and worse PFS (HR 6.96, 95%CI: 0.90-53.65, p=0.06 and HR 21.5, 95%CI: 2.63-175.8, p<0.01, respectively). Meta-analysis showed a concern for TNFi compared to Other SSIA (HR 1.56, 95%CI: 1.17-2.09, p<0.01 respectively). Conclusions While our results raise concern about the effects of CS-SSIA and TNFi for irAE therapy on tumor outcomes, prospective randomized controlled trials are needed to definitively assess the effect of SSIAs on tumor outcomes.
Collapse
Affiliation(s)
- Jennifer Strouse
- Division of Immunology, University of Iowa, Iowa City, IA, United States
| | - Karmela Kimi Chan
- Division of Rheumatology, Department of Medicine, Hospital for Special Surgery, New York, NY, United States
| | - Rachel Baccile
- Center for Health and The Social Sciences, University of Chicago, Chicago, IL, United States
| | - Gong He
- Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI, United States
| | - Diana K. N. Louden
- University Libraries, University of Washington, Seattle, WA, United States
| | - Mihai Giurcanu
- Department of Public Health Sciences, University of Chicago, Chicago, IL, United States
| | - Arohi Singh
- University of Chicago Medicine, Chicago, IL, United States
| | - John Rieth
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, University of Iowa Health Care, Iowa City, IA, United States
| | - Noha Abdel-Wahab
- Section of Rheumatology & Clinical Immunology, Department of General Internal Medicine, and Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Rheumatology & Rehabilitation, Assiut University Hospitals, Assiut University Faculty of Medicine, Asyut, Egypt
| | - Tamiko R. Katsumoto
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University, Stanford, CA, United States
| | - Namrata Singh
- Division of Rheumatology, University of Washington, Seattle, WA, United States
| | - Sherin Rouhani
- Mass General Cancer Center, Massachusetts General Hospital, Boston, MA, United States
| | - Pankti Reid
- Division of Rheumatology, Department of Medicine, University of Chicago, Chicago, IL, United States
| |
Collapse
|
16
|
Akdogan O, Turkmen S, Uyar GC, Yucel KB, Tufekci B, Gurler F, Yazici O, Ozdemir N, Ozet A, Karakaya C, Sutcuoglu O. Impact of Serum GDF-15 and IL-6 on Immunotherapy Response in Cancer: A Prospective Study. Cancers (Basel) 2024; 16:4146. [PMID: 39766045 PMCID: PMC11674841 DOI: 10.3390/cancers16244146] [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: 11/12/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Immunotherapy has transformed cancer treatment; however, predicting treatment response remains challenging. Serum biomarkers can help identify patients who are most likely to benefit from immunotherapy. OBJECTIVE This study evaluates the relationship between serum growth differentiation factor 15 (GDF-15) and interleukin-6 (IL-6) levels and treatment outcomes in cancer patients undergoing second-line immunotherapy. METHODS We conducted a prospective observational study involving 85 patients with non-small-cell lung cancer (NSCLC), renal cell carcinoma (RCC), or malignant melanoma treated with nivolumab. The baseline serum levels of GDF-15 and IL-6 were measured by using ELISA kits. The primary endpoints were progression-free survival (PFS) and overall survival (OS), with cachexia as a secondary outcome. RESULTS Elevated GDF-15 levels were significantly associated with shorter PFS (HR: 0.55, 95% CI: 0.32-0.96, p = 0.032) and OS (HR: 0.47, 95% CI: 0.25-0.90, p = 0.020). Higher IL-6 levels correlated with shorter PFS, though statistical significance was not achieved. Additionally, high GDF-15 levels were linked to increased cachexia incidence (p = 0.037). CONCLUSION Our findings indicate that GDF-15 could serve as a prognostic biomarker for immunotherapy response and may also be a target for cachexia management. Further studies should explore its potential to guide clinical decision making in oncology.
Collapse
Affiliation(s)
- Orhun Akdogan
- Department of Medical Oncology, Gazi University, 06560 Ankara, Türkiye; (F.G.); (O.Y.); (N.O.); (A.O.)
| | - Sena Turkmen
- Department of Medical Biochemistry, Gazi University, 06560 Ankara, Türkiye; (S.T.); (C.K.)
| | - Galip Can Uyar
- Department of Medical Oncology, Ankara Etlik City Hospital, 06170 Ankara, Türkiye; (G.C.U.); (K.B.Y.); (O.S.)
| | - Kadriye Bir Yucel
- Department of Medical Oncology, Ankara Etlik City Hospital, 06170 Ankara, Türkiye; (G.C.U.); (K.B.Y.); (O.S.)
| | - Busra Tufekci
- Department of Internal Medicine, Gazi University, 06560 Ankara, Türkiye;
| | - Fatih Gurler
- Department of Medical Oncology, Gazi University, 06560 Ankara, Türkiye; (F.G.); (O.Y.); (N.O.); (A.O.)
| | - Ozan Yazici
- Department of Medical Oncology, Gazi University, 06560 Ankara, Türkiye; (F.G.); (O.Y.); (N.O.); (A.O.)
| | - Nuriye Ozdemir
- Department of Medical Oncology, Gazi University, 06560 Ankara, Türkiye; (F.G.); (O.Y.); (N.O.); (A.O.)
| | - Ahmet Ozet
- Department of Medical Oncology, Gazi University, 06560 Ankara, Türkiye; (F.G.); (O.Y.); (N.O.); (A.O.)
| | - Cengiz Karakaya
- Department of Medical Biochemistry, Gazi University, 06560 Ankara, Türkiye; (S.T.); (C.K.)
| | - Osman Sutcuoglu
- Department of Medical Oncology, Ankara Etlik City Hospital, 06170 Ankara, Türkiye; (G.C.U.); (K.B.Y.); (O.S.)
| |
Collapse
|
17
|
Lyu W, Cheng X, Yu Z, Dong R, Sheng Z, Zhang T, Yin X, Shen F. Early-stage diagnosis of ovarian cancer via digital immunoassay on a SlipChip. Talanta 2024; 280:126782. [PMID: 39216422 DOI: 10.1016/j.talanta.2024.126782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/20/2024] [Accepted: 08/24/2024] [Indexed: 09/04/2024]
Abstract
Ovarian cancer (OC) is one of the three major gynecologic malignancies and has the highest mortality rate because of the late diagnosis. Liquid biopsy based on serum protein biomarkers has demonstrated great potential for early diagnosis but remains limited by the analysis performance of conventional immunoassay technologies, such as chemiluminescence, and biomarkers, such as CA125. To address this challenge and achieve accurate early-stage diagnosis of OC, we developed a digital immunoassay on a SlipChip (DiSC) for quantitative analysis of a potential serum protein biomarker, Spondin-1 (SPON1). The DiSC system achieved a limit of detection (LoD) of 23 fg/μL for digital quantification of SPON1. The DiSC system was utilized to quantify the serum level of SPON1 in 357 clinical serum samples, including 63 from patients with benign ovarian tumors and 294 from patients with malignant ovarian cancer, ranging from stages I to IV. SPON1 concentrations were significantly different in samples from patients with malignant ovarian cancer. Notably, significantly different SPON1 levels were observed in early stages (I and II), in lymph node-negative cases (N0), and before metastasis (M0), suggesting that SPON1 could serve as a sensitive diagnostic biomarker for early-stage OC. The differential diagnostic model based on SPON1 levels quantified using DiSC demonstrated an area under the curve (AUC) of 0.8187 for early-stage OC, a significant improvement over CA-125 (AUC = 0.6967). For OC of all stages, the AUC was 0.8225, which could be further increased to 0.8750 when combined with CA-125. This showcases the potential of SPON1 as a novel biomarker for sensitive early-stage diagnosis of ovarian cancer and the capability of the DiSC system in discovering low-abundance biomarkers for disease diagnosis.
Collapse
Affiliation(s)
- Weiyuan Lyu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xinrui Cheng
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Ziqing Yu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Ruirui Dong
- Affiliated Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, 214002, China
| | - Zheyi Sheng
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Ting Zhang
- Affiliated Women's Hospital of Jiangnan University, Jiangnan University, Wuxi, 214002, China
| | - Xia Yin
- State Key Laboratory for Oncogenes and Related Genes, Shanghai Key Laboratory of Gynecologic Oncology, Department of Obstetrics and Gynecology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Feng Shen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China.
| |
Collapse
|
18
|
Zhang SJ, Wu C, Walt DR. A Multiplexed Digital Platform Enables Detection of Attomolar Protein Levels with Minimal Cross-Reactivity. ACS NANO 2024; 18:29891-29901. [PMID: 39422558 DOI: 10.1021/acsnano.4c10340] [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] [Indexed: 10/19/2024]
Abstract
Protein-based biomarkers are essential for disease diagnostics, yet their low abundance in biofluids often presents significant detection challenges for traditional enzyme-linked immunosorbent assay (ELISA) techniques. While various ultrasensitive methods such as digital ELISA have improved sensitivity, multiplex assays still suffer from considerable cross-reactivities that can compromise result accuracies. To address this challenge, we have developed barcoded Molecular On-bead Signal Amplification for Individual Counting (barcoded MOSAIC), a multiplexed digital ELISA technology that markedly reduces cross-reactivity by pairing barcoded detection antibodies with specific bead types. This approach enables the simultaneous detection of eight analytes from less than 9 μL of blood, with sensitivities ranging from midpicomolar to low-attomolar levels and a collective dynamic range exceeding seven logs across multiple analytes within a single multiplex assay. Additionally, barcoded MOSAIC is compatible with standard immunoassay reagents and workflows, utilizing a rapid, automatable flow cytometric readout for quantification, which makes it a highly accessible benchtop platform that is readily adoptable by both research and clinical laboratories, setting the stage for future translation into point-of-care applications.
Collapse
Affiliation(s)
- Stephanie J Zhang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| | - Connie Wu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| | - David R Walt
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| |
Collapse
|
19
|
Shen S, Hong Y, Huang J, Qu X, Sooranna SR, Lu S, Li T, Niu B. Targeting PD-1/PD-L1 in tumor immunotherapy: Mechanisms and interactions with host growth regulatory pathways. Cytokine Growth Factor Rev 2024; 79:16-28. [PMID: 39179486 DOI: 10.1016/j.cytogfr.2024.08.001] [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: 04/16/2024] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/26/2024]
Abstract
Tumor immunotherapy has garnered considerable attention, emerging as a new standard of care in cancer treatment. The conventional targets, such as VEGF and EGFR, have been extended to others including BRAF and PD-1/PD-L1, which have shown significant potential in recent cancer treatments. This review aims to succinctly overview the impact and mechanisms of therapies that modulate PD-1/PD-L1 expression by targeting VEGF, EGFR, LAG-3, CTLA-4 and BRAF. We investigated how modulation of PD-1/PD-L1 expression impacts growth factor signaling, shedding light on the interplay between immunomodulatory pathways and growth factor networks within the tumor microenvironment. By elucidating these interactions, we aim to provide insights into novel potential synergistic therapeutic strategies for cancer immunotherapy.
Collapse
Affiliation(s)
- Songyu Shen
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China
| | - Yihan Hong
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China
| | - Jiajun Huang
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China
| | - Xiaosheng Qu
- Guangxi Botanical Garden of Medicinal Plants, Nanning, Guangxi 530023, China
| | - Suren Rao Sooranna
- Department of Metabolism, Digestion and Reproduction, Imperial College London, 369 Fulham Road, London SW10 9NH, United Kingdom
| | - Sheng Lu
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, 169 Changle West Rd, Xi'an 710032, China.
| | - Bing Niu
- School of life Science, Shanghai University, 99 Shangda Road, 200444, China.
| |
Collapse
|
20
|
Chen T, Wang M, Chen Y, Cao Y, Liu Y. Advances in predictive biomarkers associated with immunotherapy in extensive-stage small cell lung cancer. Cell Biosci 2024; 14:117. [PMID: 39267195 PMCID: PMC11391723 DOI: 10.1186/s13578-024-01283-9] [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: 05/06/2024] [Accepted: 08/05/2024] [Indexed: 09/14/2024] Open
Abstract
Small cell lung cancer (SCLC) is a highly malignant and poor-prognosis cancer, with most cases diagnosed at the extensive stage (ES). Amidst a landscape marked by limited progress in treatment modalities for ES-SCLC over the past few decades, the integration of immune checkpoint inhibitors (ICIs) with platinum-based chemotherapy has provided a milestone approach for improving prognosis, emerging as the new standard for initial therapy in ES-SCLC. However, only a minority of SCLC patients can benefit from ICIs, which frequently come with varying degrees of immune-related adverse events (irAEs). Therefore, it is crucial to investigate predictive biomarkers to screen potential beneficiaries of ICIs, mitigate the risk of side effects, and improve treatment precision. This review summarized potential biomarkers for predicting ICI response in ES-SCLC, with a primary focus on markers sourced from tumor tissue or peripheral blood samples. The former mainly included PD-L1 expression, tumor mutational burden (TMB), along with cellular or molecular components related to the tumor microenvironment (TME) and antigen presentation machinery (APM), molecular subtypes of SCLC, and inflammatory gene expression profiles. Circulating biomarkers predominantly comprised circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), cytokines, plasma autoantibodies, inflammation-related parameters, and blood TMB. We synthesized and analyzed the research progress of these potential markers. Notably, investigations into PD-L1 expression and TMB have been the most extensive, exhibiting preliminary predictive efficacy in salvage immunotherapy; however, consistent conclusions have yet to be reached across studies. Additionally, novel predictive markers developed based on TME composition, APM, transcriptomic and genomic features provide promising tools for precision immunotherapy. Circulating biomarkers offer the advantages of convenience, non-invasiveness, and a comprehensive reflection of tumor molecular characteristics. They may serve as alternative options for predicting immunotherapy efficacy in SCLC. However, there is a scarcity of studies, and the significant heterogeneity in research findings warrants attention.
Collapse
Affiliation(s)
- Tong Chen
- 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, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Mingzhao Wang
- 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, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yanchao Chen
- 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, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yang Cao
- 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, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yutao Liu
- 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, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
21
|
Zheng J, Xu S, Wang G, Shi Y. Applications of CT-based radiomics for the prediction of immune checkpoint markers and immunotherapeutic outcomes in non-small cell lung cancer. Front Immunol 2024; 15:1434171. [PMID: 39238640 PMCID: PMC11374640 DOI: 10.3389/fimmu.2024.1434171] [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: 05/17/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
In recent years, there has been significant research interest in the field of immunotherapy for non-small cell lung cancer (NSCLC) within the academic community. Given the observed variations in individual responses, despite similarities in histopathologic type, immunohistochemical index, TNM stage, or mutation status, the identification of a reliable biomarker for early prediction of therapeutic responses is of utmost importance. Conventional medical imaging techniques primarily focus on macroscopic tumor monitoring, which may no longer adequately fulfill the requirements of clinical diagnosis and treatment. CT (computerized tomography) or PEF/CT-based radiomics has the potential to investigate the molecular-level biological attributes of tumors, such as PD-1/PD-L1 expression and tumor mutation burden, which offers a novel approach to assess the effectiveness of immunotherapy and forecast patient prognosis. The utilization of cutting-edge radiological imaging techniques, including radiomics, PET/CT, machine learning, and artificial intelligence, demonstrates significant potential in predicting diagnosis, treatment response, immunosuppressive characteristics, and immune-related adverse events. The current review highlights that CT scan-based radiomics is a reliable and feasible way to predict the benefits of immunotherapy in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Jie Zheng
- Department of Radiology, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, Zhejiang, China
| | - Shuang Xu
- Department of Radiology, Redcliffe Hospital, The University of Queensland, Redcliffe, QLD, Australia
| | - Guoyu Wang
- Department of Radiology, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, Zhejiang, China
| | - Yiming Shi
- Department of Radiology, Taizhou Central Hospital, Taizhou University Hospital, Taizhou, Zhejiang, China
| |
Collapse
|
22
|
Markussen A, Johansen JS, Larsen FO, Theile S, Hasselby JP, Willemoe GL, Lorentzen T, Madsen K, Høgdall E, Poulsen TS, Wilken EE, Geertsen P, Behrens CP, Svane IM, Nielsen D, Chen IM. Nivolumab with or without Ipilimumab Combined with Stereotactic Body Radiotherapy in Patients with Metastatic Biliary Tract Cancer: A Randomized Phase 2 Study. Clin Cancer Res 2024; 30:3428-3437. [PMID: 38874506 DOI: 10.1158/1078-0432.ccr-24-0286] [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: 01/24/2024] [Revised: 04/02/2024] [Accepted: 06/11/2024] [Indexed: 06/15/2024]
Abstract
PURPOSE The purpose of this study was to evaluate the clinical benefits of nivolumab with/without ipilimumab combined with stereotactic body radiotherapy (SBRT) in patients with pretreated metastatic biliary tract cancer (mBTC). PATIENTS AND METHODS The study was a phase 2 randomized trial with Simon's optimal two-stage design requiring 36 evaluable patients per group after second stage. Sixty-one patients were included from September 2018 to January 2022 and randomized (1:1) to receive SBRT (15 Gy × 1 on day 1 to a primary or metastatic lesion) and nivolumab (3 mg/kg intravenously on day 1 and every 2 weeks) with/without ipilimumab (1 mg/kg intravenously on day 1 and every 6 weeks). Primary endpoint was clinical benefit rate (CBR), defined as the percentage of patients with complete response, partial response, or stable disease. Decision to continue accrual into the second stage depended on the CBR from the first stage. RESULTS Forty-two patients received SBRT/nivolumab/ipilimumab with a CBR of 31.0% [95% confidence interval (CI), 17.6-47.1]. Five patients (11.9%) achieved partial response with median duration of 4.4 months (range, 1.1-21.5). Nineteen patients received SBRT/nivolumab. This group was closed after the initial stage based on a CBR of 10.5% (95% CI, 1.3-33.1). Adverse events were graded with National Cancer Institute Common Terminology Criteria for Adverse Events version 4.0. Grade ≥3 treatment-related adverse events occurred in 13 (31%) and 3 (16%) patients in the SBRT/nivolumab/ipilimumab and SBRT/nivolumab groups, respectively. One patient died from immune-related hepatitis in the SBRT/nivolumab/ipilimumab group. CONCLUSIONS Combining SBRT, nivolumab, and ipilimumab is well tolerated, feasible, and shows response in a subgroup of patients with mBTC.
Collapse
Affiliation(s)
- Alice Markussen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Julia S Johansen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Medicine, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
| | - Finn O Larsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Susann Theile
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Jane P Hasselby
- Department of Pathology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Gro L Willemoe
- Department of Pathology, Copenhagen University Hospital-Rigshospitalet, Copenhagen, Denmark
| | - Torben Lorentzen
- Department of Gastroenterology, Unit of Surgical Ultrasound, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Kasper Madsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Estrid Høgdall
- Department of Pathology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Tim S Poulsen
- Department of Pathology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Eva E Wilken
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Poul Geertsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Claus P Behrens
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Health Technology, Technical University of Denmark, Roskilde, Denmark
| | - Inge M Svane
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
- National Center for Cancer Immune Therapy, Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| | - Dorte Nielsen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen, Denmark
| | - Inna M Chen
- Department of Oncology, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
| |
Collapse
|
23
|
Horaguchi S, Nakahara Y, Igarashi Y, Kouro T, Wei F, Murotani K, Udagawa S, Higashijima N, Matsuo N, Murakami S, Kato T, Kondo T, Xiang H, Kasajima R, Himuro H, Tsuji K, Mano Y, Komahashi M, Miyagi Y, Saito H, Azuma K, Uehara S, Sasada T. Prognostic Significance of Plasma Neutrophil Extracellular Trap Levels in Patients with Non-Small Cell Lung Cancer Treated with Immune Checkpoint Inhibitors. Biomedicines 2024; 12:1831. [PMID: 39200295 PMCID: PMC11351864 DOI: 10.3390/biomedicines12081831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 09/02/2024] Open
Abstract
Neutrophil extracellular traps (NETs) released from neutrophils are related to cancer progression. However, the relationship between the therapeutic effects of immune checkpoint inhibitors (ICIs) such as anti-PD-1 and anti-PD-L1 antibodies and plasma NET concentration in patients with non-small cell lung cancer (NSCLC) is poorly understood. In this study, concentrations of citrullinated histone H3 (CitH3), a surrogate marker of NETs, in plasma before/after treatment were examined in patients with advanced or recurrent NSCLC undergoing ICI treatment (n = 185). The clinical significances of NET levels before/after treatment and posttreatment changes were statistically evaluated. As a result, multivariate Cox analysis showed that high NET levels before treatment were statistically significant predictors of unfavorable overall survival (OS; p < 0.001, HR 1.702, 95% CI 1.356-2.137) and progression-free survival (PFS; p < 0.001, HR 1.566, 95% CI 1.323-1.855). The Kaplan-Meier curves showed significant separation between the high- and low-NET groups in OS (p = 0.002) and PFS (p < 0.001). Additionally, high NET levels after treatment were also significantly associated with worse OS (p < 0.001) and PFS (p < 0.001) by multivariate Cox analysis. Notably, the pretreatment NET levels were significantly correlated with the plasma levels of NET-related inflammatory cytokines, such as IL-6 and IL-8, and with NET-related gene expression and immune-suppressive profile in peripheral blood mononuclear cells. Our findings suggest that NETs released from activated neutrophils might reduce the clinical efficacy of ICIs in patients with NSCLC.
Collapse
Affiliation(s)
- Shun Horaguchi
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (S.H.); (T.K.); (F.W.); (H.H.); (K.T.); (Y.M.); (M.K.)
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yoshiro Nakahara
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama 241-8515, Japan; (Y.N.); (S.M.); (T.K.); (T.K.); (H.S.)
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara 252-0375, Japan
| | - Yuka Igarashi
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
| | - Taku Kouro
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (S.H.); (T.K.); (F.W.); (H.H.); (K.T.); (Y.M.); (M.K.)
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
| | - Feifei Wei
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (S.H.); (T.K.); (F.W.); (H.H.); (K.T.); (Y.M.); (M.K.)
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
| | - Kenta Murotani
- Biostatistics Center, Kurume University School of Medicine, Kurume 830-0011, Japan;
| | - Seiichi Udagawa
- Mathematics Section, Division of Natural Sciences, Nihon University School of Medicine, Tokyo 173-0032, Japan;
| | - Naoko Higashijima
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
| | - Norikazu Matsuo
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume 830-0011, Japan; (N.M.); (K.A.)
| | - Shuji Murakami
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama 241-8515, Japan; (Y.N.); (S.M.); (T.K.); (T.K.); (H.S.)
| | - Terufumi Kato
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama 241-8515, Japan; (Y.N.); (S.M.); (T.K.); (T.K.); (H.S.)
| | - Tetsuro Kondo
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama 241-8515, Japan; (Y.N.); (S.M.); (T.K.); (T.K.); (H.S.)
| | - Huihui Xiang
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (H.X.); (R.K.); (Y.M.)
| | - Rika Kasajima
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (H.X.); (R.K.); (Y.M.)
| | - Hidetomo Himuro
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (S.H.); (T.K.); (F.W.); (H.H.); (K.T.); (Y.M.); (M.K.)
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
| | - Kayoko Tsuji
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (S.H.); (T.K.); (F.W.); (H.H.); (K.T.); (Y.M.); (M.K.)
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
| | - Yasunobu Mano
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (S.H.); (T.K.); (F.W.); (H.H.); (K.T.); (Y.M.); (M.K.)
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
| | - Mitsuru Komahashi
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (S.H.); (T.K.); (F.W.); (H.H.); (K.T.); (Y.M.); (M.K.)
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yohei Miyagi
- Molecular Pathology and Genetics Division, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (H.X.); (R.K.); (Y.M.)
| | - Haruhiro Saito
- Department of Thoracic Oncology, Kanagawa Cancer Center, Yokohama 241-8515, Japan; (Y.N.); (S.M.); (T.K.); (T.K.); (H.S.)
| | - Koichi Azuma
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume 830-0011, Japan; (N.M.); (K.A.)
| | - Shuichiro Uehara
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Tetsuro Sasada
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (S.H.); (T.K.); (F.W.); (H.H.); (K.T.); (Y.M.); (M.K.)
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama 241-8515, Japan; (Y.I.); (N.H.)
| |
Collapse
|
24
|
Wang X, Chen D, Ma Y, Mo D, Yan F. Variation of peripheral blood-based biomarkers for response of anti-PD-1 immunotherapy in non-small-cell lung cancer. Clin Transl Oncol 2024; 26:1934-1943. [PMID: 38451413 PMCID: PMC11249409 DOI: 10.1007/s12094-024-03416-5] [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: 02/01/2024] [Accepted: 02/20/2024] [Indexed: 03/08/2024]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) for non-small-cell lung cancer (NSCLC) are on the rise, but unfortunately, only a small percentage of patients benefit from them in the long term. Thus, it is crucial to identify biomarkers that can forecast the efficacy of immunotherapy. METHODS We retrospectively studied 224 patients with NSCLC who underwent anti-PD-1 therapy. The role of biomarkers and clinical characteristics were assessed in a prognostic model. RESULTS Only 14.3% of patients had both programmed death ligand 1 (PD-L1) and tumor mutational burden (TMB) outcomes, highlighting the need to investigate more available biomarkers. Our analysis found a correlation between histological PD-L1 TPS and hematological PD-1 expression. Analysis of hematological biomarkers revealed that elevated expression of CD4/CD8 and LYM% are positively associated with effective immunotherapy, while PD-1+ on T cells, NLR, and MLR have a negative impact. Moreover, high level of ΔCEA%, CYFRA21-1 and LDH may suggest ineffective ICIs. We also observed that disparate immunotherapy drugs didn't significantly impact prognosis. Lastly, by comparing squamous carcinoma and adenocarcinoma cohorts, ΔCEA%, CD3+PD-1+, CD4+PD-1+, and CD4/CD8 are more important in predicting the prognosis of adenocarcinoma patients, while age is more significant for squamous carcinoma patients. CONCLUSION Our research has yielded encouraging results in identifying a correlation between immunotherapy's response and clinical characteristics, peripheral immune cell subsets, and biochemical and immunological biomarkers. The screened hematological detection panel could be used to forecast an NSCLC patient's response to anti-PD-1 immunotherapy with an accuracy rate of 76.3%, which could help customize suitable therapeutic decision-making.
Collapse
Affiliation(s)
- Xiaoming Wang
- Department of Clinical Laboratory, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Baizi Ting No.42, Nanjing, 210009, Jiangsu, China
| | - Dayu Chen
- Department of Clinical Laboratory, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Baizi Ting No.42, Nanjing, 210009, Jiangsu, China
| | - Yuyan Ma
- Department of Clinical Laboratory, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Baizi Ting No.42, Nanjing, 210009, Jiangsu, China
| | - Dongping Mo
- Department of Clinical Laboratory, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Baizi Ting No.42, Nanjing, 210009, Jiangsu, China
| | - Feng Yan
- Department of Clinical Laboratory, Nanjing Medical University Affiliated Cancer Hospital & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Baizi Ting No.42, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
25
|
Goswami M, Toney NJ, Pitts SC, Celades C, Schlom J, Donahue RN. Peripheral immune biomarkers for immune checkpoint inhibition of solid tumours. Clin Transl Med 2024; 14:e1814. [PMID: 39162097 PMCID: PMC11333946 DOI: 10.1002/ctm2.1814] [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: 05/30/2024] [Revised: 07/29/2024] [Accepted: 08/04/2024] [Indexed: 08/21/2024] Open
Abstract
BACKGROUND With the rapid adoption of immunotherapy for the treatment of cancer comes the pressing need for readily accessible biomarkers to guide immunotherapeutic strategies and offer insights into outcomes with specific treatments. Regular sampling of solid tumour tissues outside of melanoma for immune monitoring is not often feasible; conversely, routine, frequent interrogation of circulating immune biomarkers is entirely possible. As immunotherapies and immune checkpoint inhibitors, in particular, are more widely used in first-line, neoadjuvant, and metastatic settings, the discovery and validation of peripheral immune biomarkers are urgently needed across solid tumour types for improved prediction and prognostication of clinical outcomes in response to immunotherapy, as well as elucidation of mechanistic underpinnings of the intervention. Careful experimental design, encompassing both retrospective and prospective studies, is required in such biomarker identification studies, and concerted efforts are essential for their advancement into clinical settings. CONCLUSION In this review, we summarize shared immune features between the tumour microenvironment and systemic circulation, evaluate exploratory peripheral immune biomarker studies, and discuss associations between candidate biomarkers with clinical outcomes. We also consider integration of multiple peripheral immune parameters for better prediction and prognostication and discuss considerations in study design to further evaluate the clinical utility of candidate peripheral immune biomarkers for immunotherapy of solid tumours. HIGHLIGHTS Peripheral immune biomarkers are critical for improved prediction and prognostication of clinical outcomes for patients with solid tumours treated with immune checkpoint inhibition. Candidate peripheral biomarkers, such as cytokines, soluble factors, and immune cells, have potential as biomarkers to guide immunotherapy of solid tumours. Multiple peripheral immune parameters may be integrated to improve prediction and prognostication. The potential of peripheral immune biomarkers to guide immunotherapy of solid tumours requires critical work in biomarker discovery, validation, and standardization.
Collapse
Affiliation(s)
- Meghali Goswami
- Center for Immuno‐Oncology, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Nicole J. Toney
- Center for Immuno‐Oncology, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Stephanie C. Pitts
- Center for Immuno‐Oncology, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Carolina Celades
- Center for Immuno‐Oncology, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Jeffrey Schlom
- Center for Immuno‐Oncology, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Renee N. Donahue
- Center for Immuno‐Oncology, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
26
|
Yi M, Li T, Niu M, Zhang H, Wu Y, Wu K, Dai Z. Targeting cytokine and chemokine signaling pathways for cancer therapy. Signal Transduct Target Ther 2024; 9:176. [PMID: 39034318 PMCID: PMC11275440 DOI: 10.1038/s41392-024-01868-3] [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: 02/28/2024] [Revised: 04/30/2024] [Accepted: 05/11/2024] [Indexed: 07/23/2024] Open
Abstract
Cytokines are critical in regulating immune responses and cellular behavior, playing dual roles in both normal physiology and the pathology of diseases such as cancer. These molecules, including interleukins, interferons, tumor necrosis factors, chemokines, and growth factors like TGF-β, VEGF, and EGF, can promote or inhibit tumor growth, influence the tumor microenvironment, and impact the efficacy of cancer treatments. Recent advances in targeting these pathways have shown promising therapeutic potential, offering new strategies to modulate the immune system, inhibit tumor progression, and overcome resistance to conventional therapies. In this review, we summarized the current understanding and therapeutic implications of targeting cytokine and chemokine signaling pathways in cancer. By exploring the roles of these molecules in tumor biology and the immune response, we highlighted the development of novel therapeutic agents aimed at modulating these pathways to combat cancer. The review elaborated on the dual nature of cytokines as both promoters and suppressors of tumorigenesis, depending on the context, and discussed the challenges and opportunities this presents for therapeutic intervention. We also examined the latest advancements in targeted therapies, including monoclonal antibodies, bispecific antibodies, receptor inhibitors, fusion proteins, engineered cytokine variants, and their impact on tumor growth, metastasis, and the tumor microenvironment. Additionally, we evaluated the potential of combining these targeted therapies with other treatment modalities to overcome resistance and improve patient outcomes. Besides, we also focused on the ongoing research and clinical trials that are pivotal in advancing our understanding and application of cytokine- and chemokine-targeted therapies for cancer patients.
Collapse
Affiliation(s)
- Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China
| | - Tianye Li
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310000, People's Republic of China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Haoxiang Zhang
- Department of Hepatopancreatobiliary Surgery, Fujian Provincial Hospital, Fuzhou, 350001, People's Republic of China
| | - Yuze Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Zhijun Dai
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310000, People's Republic of China.
| |
Collapse
|
27
|
Liu Z, Lei T, Guo Y, Zheng C. The impact of sarcopenia on the efficacy of PD-1 inhibitors in non-small cell lung cancer and potential strategies to overcome resistance. Front Pharmacol 2024; 15:1377666. [PMID: 39101140 PMCID: PMC11294093 DOI: 10.3389/fphar.2024.1377666] [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: 01/28/2024] [Accepted: 06/18/2024] [Indexed: 08/06/2024] Open
Abstract
Recent studies have revealed that sarcopenia can adversely affect the efficacy of PD-1 inhibitors in the treatment of non-small cell lung cancer (NSCLC). PD-1 inhibitors are immune checkpoint inhibitors widely used in the treatment of various cancers. However, NSCLC patients may have poorer outcomes when receiving PD-1 inhibitor treatment, and sarcopenia may affect the efficacy of PD-1 inhibitors through immune and metabolic mechanisms. In this article, we summarize the reported negative impact of sarcopenia on the effectiveness of PD-1 inhibitors in the treatment of NSCLC in recent years. Based on existing research results, we analyze the possible mechanisms by which sarcopenia affects the efficacy of PD-1 inhibitors and discuss possible strategies to address this issue. This could help to understand the impact of sarcopenia on the treatment of PD-1 inhibitors and provide more accurate expectations of treatment outcomes for clinicians and patients. Additionally, we present tailored intervention strategies for sarcopenic patients undergoing PD-1 inhibitor therapy, aiming to optimize treatment efficacy and enhance patient quality of life. Nevertheless, further research is warranted to elucidate the mechanisms through which sarcopenia impacts PD-1 inhibitors and to identify more efficacious intervention approaches for improving the effectiveness of PD-1 inhibitor treatment in sarcopenic patients.
Collapse
Affiliation(s)
- Zhenchao Liu
- School of Pharmacy, Qingdao University, Qingdao, Shandong, China
| | - Tianxiang Lei
- Institute of Integrative Medicine, Qingdao University, Qingdao, Shandong, China
| | - Yunliang Guo
- Institute of Integrative Medicine, Qingdao University, Qingdao, Shandong, China
| | - Chongwen Zheng
- Department of Neurology, The 2nd Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
28
|
Alnemri AE, Tekumalla S, Moroco AE, Vathiotis I, Tuluc M, Gargano S, Zhan T, Cognetti DM, Curry JM, Argiris A, Linnenbach A, South AP, Harshyne LA, Johnson JM, Luginbuhl AJ. Predictive capacity of immune-related adverse events and cytokine profiling in neoadjuvant immune checkpoint inhibitor trials for head and neck squamous cell carcinoma. Cancer Med 2024; 13:e7370. [PMID: 38847087 PMCID: PMC11157197 DOI: 10.1002/cam4.7370] [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: 01/29/2024] [Revised: 05/10/2024] [Accepted: 05/27/2024] [Indexed: 06/10/2024] Open
Abstract
OBJECTIVES Certain low-level immune-related adverse events (irAEs) have been associated with survival benefits in patients with various solid tumors on immune checkpoint inhibitors (ICIs). We aimed to investigate the association between irAEs and response to neoadjuvant ICIs in patients with head and neck squamous cell carcinoma (HNSCC) and to identify differences in circulating cytokine levels based on irAE status. METHODS This was a retrospective cohort study including three neoadjuvant clinical trials from July 2017 to January 2022: NCT03238365 (nivolumab ± tadalafil), NCT03854032 (nivolumab ± BMS986205), NCT03618654 (durvalumab ± metformin). The presence and type of irAEs, pathologic treatment response, and survival were compared. Canonical linear discriminant analysis (LDA) was performed to identify combinations of circulating cytokines predictive of irAEs using plasma sample multiplex assay. RESULTS Of 113 participants meeting inclusion criteria, 32 (28.3%) developed irAEs during treatment or follow-up. Positive p16 status was associated with irAEs (odds ratio [OR] 2.489; 95% CI 1.069-6.119; p = 0.043). irAEs were associated with pathologic treatment response (OR 3.73; 95% CI 1.34-10.35; p = 0.011) and with higher OS in the combined cohort (HR 0.319; 95% CI 0.113-0.906; p = 0.032). Patients with irAEs within the nivolumab cohort had significant elevations of select cytokines pre-treatment. Canonical LDA identified key drivers of irAEs among all trials, which were highly predictive of future irAE status. CONCLUSIONS irAEs are associated with response to neoadjuvant ICI therapy in HNSCC and can serve as clinical indicators for improved clinical outcomes. irAEs can be predicted by concentrations of several circulating cytokines prior to treatment.
Collapse
Affiliation(s)
- Angela E. Alnemri
- Department of Otolaryngology – Head & Neck SurgeryThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Sruti Tekumalla
- Department of Otolaryngology – Head & Neck SurgeryThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Annie E. Moroco
- Department of Otolaryngology – Head & Neck SurgeryThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Ioannis Vathiotis
- Department of Medical OncologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Madalina Tuluc
- Department of PathologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Stacey Gargano
- Department of PathologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Tingting Zhan
- Department of BiostatisticsThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - David M. Cognetti
- Department of Otolaryngology – Head & Neck SurgeryThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Joseph M. Curry
- Department of Otolaryngology – Head & Neck SurgeryThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Athanassios Argiris
- Department of Medical OncologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Alban Linnenbach
- Department of Dermatology and Cutaneous BiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Andrew P. South
- Department of Otolaryngology – Head & Neck SurgeryThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- Department of Pharmacology, Physiology and Cancer BiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Larry A. Harshyne
- Department of Microbiology & ImmunologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Jennifer M. Johnson
- Department of Otolaryngology – Head & Neck SurgeryThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
- Department of Medical OncologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Adam J. Luginbuhl
- Department of Otolaryngology – Head & Neck SurgeryThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
29
|
Jin SX, Liu BN, Ji HJ, Wu JR, Li BL, Gao XL, Li N, Zheng ZD, Du C. Serum cytokines and creatinine/cystatin C ratio as prognostic biomarkers in advanced cancer patients treated with anti-PD-1/PD-L1 therapy. Support Care Cancer 2024; 32:370. [PMID: 38776028 PMCID: PMC11111483 DOI: 10.1007/s00520-024-08525-z] [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: 02/11/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE Immune checkpoint inhibitors (ICIs), specifically targeting the programmed cell death protein-1 or its ligand (PD-1/PD-L1), have been extensively used in the treatment of a spectrum of malignancies, although the predictive biomarkers remain to be elucidated. This study aims to investigate the association between baseline circulating levels of cytokines and the creatinine/cystatin C ratio (CCR) with the treatment outcomes of ICIs in patients with advanced cancer. METHODS The pre-treatment circulating levels of 10 cytokines (PD-L1, CTLA4, CXCL10, LAG3, HGF, CCL2, MIG, GRANB, IL-18, and IL-6) were measured via automated capillary-based immunoassay platform in the serum of 65 advanced cancer patients treated with anti-PD-1/PD-L1-based systemic therapy and 10 healthy volunteers. The levels of cytokines and CCR were quantified and categorized into high and low groups based on the median value. The associations of serum cytokines and CCR with response to treatment, survival, and immune-related adverse events were assessed. RESULTS Elevated circulating levels of 6 cytokines (PD-L1, CXCL10, HGF, CCL2, MIG, and IL-6) were observed in cancer patients compared with that in healthy volunteers. The correlation coefficients between cytokines, CCR and nutritional risk index were also calculated. In the cancer cohort (N = 65), low circulating HGF (P = 0.023, P = 0.029), low IL-6 (P = 0.002, P < 0.001), and high CCR (P = 0.031, P = 0.008) were associated with significantly improved progression-free survival (PFS) and overall survival (OS). Multi-variable COX analyses adjusted for clinicopathological factors revealed that low HGF, low IL-6, and high CCR were independent favorable prognostic factors for PFS (P = 0.028, P = 0.010, and P = 0.015, respectively) and OS (P = 0.043, P = 0.003, and P = 0.026, respectively). Grade 2 irAEs occurred more frequently in patients with low levels of circulating CCL2 and LAG3. CONCLUSIONS Pre-treatment circulating levels of serum IL-6, HGF, and CCR may serve as independent predictive and prognostic biomarkers in advanced cancer patients treated with ICIs-based systemic therapy. These findings might help to identify potential patients who would benefit from these therapies.
Collapse
Affiliation(s)
- Shan-Xiu Jin
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China
| | - Bo-Na Liu
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China
| | - Hong-Juan Ji
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jing-Ran Wu
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China
| | - Bao-Lei Li
- Department of Oncology, Anshan Tumor Hospital, Anshan, China
| | - Xiao-Li Gao
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China
| | - Na Li
- Department of Gynaecology and Obstetrics, The First Hospital of Jilin University, Jilin, China.
| | - Zhen-Dong Zheng
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China.
| | - Cheng Du
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China.
| |
Collapse
|
30
|
Lin M, Zhao A, Chen B. Potential mechanism of Chai Gui Zexie Decoction for NSCLC treatment assessed using network pharmacology, bioinformatics, and molecular docking: An observational study. Medicine (Baltimore) 2024; 103:e38204. [PMID: 38758858 PMCID: PMC11098237 DOI: 10.1097/md.0000000000038204] [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: 02/01/2024] [Accepted: 04/19/2024] [Indexed: 05/19/2024] Open
Abstract
To explore the potential mechanism of Chai Gui Zexie Decoction for non-small cell lung cancer (NSCLC) treatment using network pharmacology, bioinformatics, and molecular docking. The active ingredients of Chai Gui Zexie Decoction and the associated predicted targets were screened using the TCMSP database. NSCLC-related targets were obtained from GeneCards and OMIM. Potential action targets, which are intersecting drug-predicted targets and disease targets, were obtained from Venny 2.1. The protein-protein interaction network was constructed by importing potential action targets into the STRING database, and the core action targets and core ingredients were obtained via topological analysis. The core action targets were entered into the Metascape database, and Gene Ontology annotation analysis and Kyoto Encyclopedia of Genes and Genomes pathway analysis were performed. Differentially expressed genes were screened using the Gene Expression Omnibus, and the key targets were obtained by validating the core action targets. The key targets were input into The Tumor IMmune Estimation Resource for immune cell infiltration analysis. Finally, the molecular docking of key targets and core ingredients was performed. We obtained 60 active ingredients, 251 drug prediction targets, and 2133 NSCLC-related targets. Meanwhile, 147 potential action targets were obtained, and 47 core action targets and 40 core ingredients were obtained via topological analysis. We detected 175 pathways related to NSCLC pharmaceutical therapy. In total, 1249 Gene Ontology items were evaluated. Additionally, 3102 differential genes were screened, and tumor protein P53, Jun proto-oncogene, interleukin-6, and mitogen-activated protein kinase 3 were identified as the key targets. The expression of these key targets in NSCLC was correlated with macrophage, CD4+ T, CD8+ T, dendritic cell, and neutrophil infiltration. The molecular docking results revealed that the core ingredients have a potent affinity for the key targets. Chai Gui Zexie Decoction might exert its therapeutic effect on NSCLC through multiple ingredients, targets, and signaling pathways.
Collapse
Affiliation(s)
- Manbian Lin
- Department of Medical Oncology, Fuzhou Hospital of Traditional Chinese Medicine Affiliated to Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Aiping Zhao
- Department of Internal Medicine, The Affiliated People’s Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Bishan Chen
- Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
31
|
Maiorano BA, Schinzari G, Carbone C, Piro G, Rossi E, Di Maio M, Di Giacomo A, Maiello E. Prognostic role of circulating cytokines and inflammation indexes for avelumab maintenance in metastatic urothelial carcinoma. Front Immunol 2024; 15:1401214. [PMID: 38799450 PMCID: PMC11116647 DOI: 10.3389/fimmu.2024.1401214] [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: 03/14/2024] [Accepted: 04/18/2024] [Indexed: 05/29/2024] Open
Abstract
Background Avelumab maintenance after first-line platinum-based chemotherapy represents a cornerstone for the treatment of metastatic urothelial carcinoma (mUC). However, identifying prognostic biomarkers is paramount for optimizing patients' benefits while minimizing toxicity. Cytokines represent circulating mediators of the complex interaction between cancer, the immune system, and inflammation. Inflammation, a hallmark of cancer, can be expressed by circulating factors. In different tumor subtypes, peripheral blood biomarkers, such as circulating cytokines, and systemic inflammatory indexes, have been addressed as potential prognostic factors for immune checkpoint inhibitors. However, their role in mUC still needs to be determined. Methods Between February 2021 and April 2023, we prospectively collected plasma cytokines and inflammation indexes in 28 patients with mUC before starting avelumab as first-line maintenance. The primary endpoint was the relationship between baseline cytokines and inflammatory indexes with the clinical benefit (CB), defined as the number of Responders. Secondary endpoints included the correlation of baseline cytokines and inflammatory indexes with progression-free survival (PFS), overall survival (OS), and the number and grade of immune-related adverse events. Results High pre-treatment levels of interferon (IFN)-γ and interleukin (IL)-2, and low levels of IL-6, IL-8, neutrophil-to-lymphocyte ratio (NLR), lymphocyte-to-monocyte ratio (LMR), and systemic-inflammation index (SII) were associated with clinical benefit and longer survival. In the multivariate analysis, low IL-8, NLR, and SII levels maintained a positive prognostic value for OS. Conclusion Our data suggest that, in mUC patients receiving avelumab, pre-treatment levels of plasma cytokines and inflammatory indexes may serve as potential prognostic biomarkers for response and efficacy. In particular, patients with signs of pre-therapeutic inflammation showed a significantly lower response and survival to avelumab. On the contrary, low systemic inflammation and high levels of cytokines characterized responders and longer survivors.
Collapse
Affiliation(s)
- Brigida Anna Maiorano
- Oncology Unit, IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Giovanni Schinzari
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Carmine Carbone
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Geny Piro
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Ernesto Rossi
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli Istituti di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | | | | | - Evaristo Maiello
- Oncology Unit, IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, Italy
| |
Collapse
|
32
|
Spagnolo CC, Pepe F, Ciappina G, Nucera F, Ruggeri P, Squeri A, Speranza D, Silvestris N, Malapelle U, Santarpia M. Circulating biomarkers as predictors of response to immune checkpoint inhibitors in NSCLC: Are we on the right path? Crit Rev Oncol Hematol 2024; 197:104332. [PMID: 38580184 DOI: 10.1016/j.critrevonc.2024.104332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/25/2024] [Indexed: 04/07/2024] Open
Abstract
Immune checkpoints inhibitors (ICIs) have markedly improved the therapeutic management of advanced NSCLC and, more recently, they have demonstrated efficacy also in the early-stage disease. Despite better survival outcomes with ICIs compared to standard chemotherapy, a large proportion of patients can derive limited clinical benefit from these agents. So far, few predictive biomarkers, including the programmed death-ligand 1 (PD-L1), have been introduced in clinical practice. Therefore, there is an urgent need to identify novel biomarkers to select patients for immunotherapy, to improve efficacy and avoid unnecessary toxicity. A deeper understanding of the mechanisms involved in antitumor immunity and advances in the field of liquid biopsy have led to the identification of a wide range of circulating biomarkers that could potentially predict response to immunotherapy. Herein, we provide an updated overview of these circulating biomarkers, focusing on emerging data from clinical studies and describing modern technologies used for their detection.
Collapse
Affiliation(s)
- Calogera Claudia Spagnolo
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Via S. Pansini, Naples 80131, Italy
| | - Giuliana Ciappina
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy
| | - Francesco Nucera
- Respiratory Medicine Unit, Department of Biomedical Sciences, Dentistry and Morphological and Functional Imaging (BIOMORF), University of Messina, Messina 98122, Italy
| | - Paolo Ruggeri
- Respiratory Medicine Unit, Department of Biomedical Sciences, Dentistry and Morphological and Functional Imaging (BIOMORF), University of Messina, Messina 98122, Italy
| | - Andrea Squeri
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy
| | - Desirèe Speranza
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Via S. Pansini, Naples 80131, Italy
| | - Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina 98122, Italy.
| |
Collapse
|
33
|
Dai L, Tan Q, Li L, Lou N, Zheng C, Yang J, Huang L, Wang S, Luo R, Fan G, Xie T, Yao J, Zhang Z, Tang L, Shi Y, Han X. High-Throughput Antigen Microarray Identifies Longitudinal Prognostic Autoantibody for Chemoimmunotherapy in Advanced Non-Small Cell Lung Cancer. Mol Cell Proteomics 2024; 23:100749. [PMID: 38513890 PMCID: PMC11070596 DOI: 10.1016/j.mcpro.2024.100749] [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: 08/11/2023] [Revised: 02/03/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024] Open
Abstract
Chemoimmunotherapy has evolved as a standard treatment for advanced non-small cell lung cancer (aNSCLC). However, inevitable drug resistance has limited its efficacy, highlighting the urgent need for biomarkers of chemoimmunotherapy. A three-phase strategy to discover, verify, and validate longitudinal predictive autoantibodies (AAbs) for aNSCLC before and after chemoimmunotherapy was employed. A total of 528 plasma samples from 267 aNSCLC patients before and after anti-PD1 immunotherapy were collected, plus 30 independent formalin-fixed paraffin-embedded samples. Candidate AAbs were firstly selected using a HuProt high-density microarray containing 21,000 proteins in the discovery phase, followed by validation using an aNSCLC-focused microarray. Longitudinal predictive AAbs were chosen for ELISA based on responders versus non-responders comparison and progression-free survival (PFS) survival analysis. Prognostic markers were also validated using immunohistochemistry and publicly available immunotherapy datasets. We identified and validated a panel of two AAbs (MAX and DHX29) as pre-treatment biomarkers and another panel of two AAbs (MAX and TAPBP) as on-treatment predictive markers in aNSCLC patients undergoing chemoimmunotherapy. All three AAbs exhibited a positive correlation with early responses and PFS (p < 0.05). The kinetics of MAX AAb showed an increasing trend in responders (p < 0.05) and a tendency to initially increase and then decrease in non-responders (p < 0.05). Importantly, MAX protein and mRNA levels effectively discriminated PFS (p < 0.05) in aNSCLC patients treated with immunotherapy. Our results present a longitudinal analysis of changes in prognostic AAbs in aNSCLC patients undergoing chemoimmunotherapy.
Collapse
Affiliation(s)
- Liyuan Dai
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Qiaoyun Tan
- 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 Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Lin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Ning Lou
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Cuiling Zheng
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Jianliang Yang
- 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 Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Liling Huang
- 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 Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Shasha Wang
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Rongrong Luo
- Department of Clinical Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Guangyu Fan
- 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 Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Tongji Xie
- 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 Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Jiarui Yao
- 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 Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Zhishang Zhang
- 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 Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Le Tang
- 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 Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China
| | - Yuankai Shi
- 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 Key Laboratory of Clinical Study on Anticancer Molecular Targeted Drugs, Beijing, China.
| | - Xiaohong Han
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
34
|
Mirjačić Martinović K, Vuletić A, Tišma Miletić N, Matković S, Gavrilović D, Ninković A, Jurišić V, Babović N. Circulating IL-6 is associated with disease progression in BRAFwt metastatic melanoma patients receiving anti-PD-1 therapy. J Clin Pathol 2024; 77:343-351. [PMID: 36754615 DOI: 10.1136/jcp-2022-208615] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/27/2023] [Indexed: 02/10/2023]
Abstract
AIMS Despite efficacy of anti-PD-1 blockade in treatment of metastatic melanoma (MM), many patients achieve rapid disease progression (DP). Therefore, the aim of this study is to better define biomarkers for DP by analysing levels of circulating cytokines TGF-β, IFN-γ, IL-6, IL-8 and IL-10 in MM patients prior to anti-PD-1 therapy. METHODS Cytokine levels were evaluated before therapy with pembrolizumab in peripheral blood of BRAF wild-type (wt) MM patients by ELISA method. RESULTS In this study, we give pretherapy levels for circulating TGF-β, IFN-γ, IL-6, IL-8 and IL-10 in BRAFwt MM patients and analyse them according to metastasis stage (M1a+M1 b, M1c, M1d groups), lactate dehydrogenase (LDH) level and occurrence of DP. Increased IL-6 level was found in M1d group (central nervous system metastasis), while LDH+patients (LDH ≥460 IU/L) have increased IL-6 and IL-8 values that correlate with LDH level. Also, IL-6 correlates with C reactive protein values. Furthermore, patients with DP have significantly higher IL-6 level compared with non-DP patients. Conversely, the other analysed cytokines are similar in investigated groups of MM patients. By receiver operating characteristics curve analysis, pretherapy IL-6 level was found to be a biomarker for the occurrence of DP with cut-off value of 3.02 pg/mL. Patients in M1d stage are prevalent in the group with IL-6 ≥3.02 pg/mL that is characterised with reduced progression-free survival and higher pretherapy IL-8 and LDH. CONCLUSION The evidence in this study implies that baseline IL-6 could be a biomarker of DP and poor prognosis in BRAFwt MM patients treated with pembrolizumab.
Collapse
Affiliation(s)
- Katarina Mirjačić Martinović
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, Belgrade, Serbia
| | - Ana Vuletić
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, Belgrade, Serbia
| | - Nevena Tišma Miletić
- Department of Experimental Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, Belgrade, Serbia
| | - Suzana Matković
- Department of Medical Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, Belgrade, Serbia
| | - Dušica Gavrilović
- Data Centre, Institute of Oncology and Radiology of Serbia, Pasterova 14, Belgrade, Serbia
| | - Aleksandra Ninković
- Department of Biochemistry, Institute of Oncology and Radiology of Serbia, Pasterova 14, Belgrade, Serbia
| | - Vladimir Jurišić
- Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nada Babović
- Department of Medical Oncology, Institute of Oncology and Radiology of Serbia, Pasterova 14, Belgrade, Serbia
| |
Collapse
|
35
|
Gao F, You X, Yang L, Zou X, Sui B. Boosting immune responses in lung tumor immune microenvironment: A comprehensive review of strategies and adjuvants. Int Rev Immunol 2024; 43:280-308. [PMID: 38525925 DOI: 10.1080/08830185.2024.2333275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 02/12/2024] [Accepted: 03/15/2024] [Indexed: 03/26/2024]
Abstract
The immune system has a substantial impact on the growth and expansion of lung malignancies. Immune cells are encompassed by a stroma comprising an extracellular matrix (ECM) and different cells like stromal cells, which are known as the tumor immune microenvironment (TIME). TME is marked by the presence of immunosuppressive factors, which inhibit the function of immune cells and expand tumor growth. In recent years, numerous strategies and adjuvants have been developed to extend immune responses in the TIME, to improve the efficacy of immunotherapy. In this comprehensive review, we outline the present knowledge of immune evasion mechanisms in lung TIME, explain the biology of immune cells and diverse effectors on these components, and discuss various approaches for overcoming suppressive barriers. We highlight the potential of novel adjuvants, including toll-like receptor (TLR) agonists, cytokines, phytochemicals, nanocarriers, and oncolytic viruses, for enhancing immune responses in the TME. Ultimately, we provide a summary of ongoing clinical trials investigating these strategies and adjuvants in lung cancer patients. This review also provides a broad overview of the current state-of-the-art in boosting immune responses in the TIME and highlights the potential of these approaches for improving outcomes in lung cancer patients.
Collapse
Affiliation(s)
- Fei Gao
- Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Xiaoqing You
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Liu Yang
- Department of Oncology, Da Qing Long Nan Hospital, Daqing, Heilongjiang Province, China
| | - Xiangni Zou
- Department of Nursing, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| | - Bowen Sui
- Department of Oncology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, China
| |
Collapse
|
36
|
Kimura N, Tsukita Y, Ebina-Shibuya R, Miyauchi E, Yamada M, Narita D, Saito R, Inoue C, Fujino N, Ichikawa T, Tamada T, Sugiura H. Peripheral blood biomarkers associated with combination of immune checkpoint blockade plus chemotherapy in NSCLC. Cancer Biomark 2024:CBM230301. [PMID: 38669521 DOI: 10.3233/cbm-230301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2024]
Abstract
BACKGROUND Biomarkers predicting clinical outcomes of treating non-small cell lung cancer (NSCLC) with combination of immune checkpoint inhibitors (ICIs) and chemotherapy would be valuable. OBJECTIVE This study aims to seek predictors of combination of ICI/chemotherapy response in NSCLC patients using peripheral blood samples. METHODS Patients diagnosed with advanced NSCLC between July 2019 and May 2021 receiving combination of ICI/chemotherapy were included and assessed for partial responses (PR), stable disease (SD) or progressive disease (PD). We measured circulating immune cells, plasma cytokines and chemokines. RESULTS Nineteen patients were enrolled. The proportions of circulating natural killer (NK) cells within CD45 + cells, programmed death 1 (PD-1) + Tim-3 + T cells within CD4 + cells, and the amount of chemokine C-X-C ligand (CXCL10) in the plasma were significantly elevated in PR relative to SD/PD patients (median 8.1%-vs-2.1%, P = 0.0032; median 1.2%-vs-0.3%, P = 0.0050; and median 122.6 pg/ml-vs-76.0 pg/ml, P = 0.0125, respectively). Patients with 2 or 3 elevated factors had longer progression-free survival than patients with 0 or only one (not reached-vs-5.6 months, P = 0.0002). CONCLUSIONS We conclude that NK cells, CD4 + PD-1 + Tim-3 + T cells, and CXCL10 levels in pre-treatment peripheral blood may predict the efficacy of combination of ICI/chemotherapy in NSCLC.
Collapse
Affiliation(s)
- Nozomu Kimura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yoko Tsukita
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Risa Ebina-Shibuya
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Eisaku Miyauchi
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mitsuhiro Yamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Daisuke Narita
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryota Saito
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Chihiro Inoue
- Department of Anatomic Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoya Fujino
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiro Ichikawa
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tsutomu Tamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hisatoshi Sugiura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
37
|
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
|
38
|
Saylor PJ, Kozin SV, Matsui A, Goldberg SI, Aoki S, Shigeta K, Mamessier E, Smith MR, Michaelson MD, Lee RJ, Duda DG. The radiopharmaceutical radium-223 has immunomodulatory effects in patients and facilitates anti-programmed death receptor-1 therapy in murine models of bone metastatic prostate cancer. Radiother Oncol 2024; 192:110091. [PMID: 38224917 PMCID: PMC10905770 DOI: 10.1016/j.radonc.2024.110091] [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: 06/27/2023] [Revised: 01/03/2024] [Accepted: 01/07/2024] [Indexed: 01/17/2024]
Abstract
BACKGROUND & PURPOSE Radium-223 (Ra223) improves survival in metastatic prostate cancer (mPC), but its impact on systemic immunity is unclear, and biomarkers of response are lacking. We examined markers of immunomodulatory activity during standard clinical Ra223 and studied the impact of Ra223 on response to immune checkpoint inhibition (ICI) in preclinical models. MATERIALS & METHODS We conducted a single-arm biomarker study of Ra223 in 22 bone mPC patients. We measured circulating immune cell subsets and a panel of cytokines before and during Ra223 therapy and correlated them with overall survival (OS). Using two murine mPC models-orthotopic PtenSmad4-null and TRAMP-C1 grafts in syngeneic immunocompetent mice-we tested the efficacy of combining Ra223 with ICI. RESULTS Above-median level of IL-6 at baseline was associated with a median OS of 358 versus 947 days for below levels; p = 0.044, from the log-rank test. Baseline PlGF and PSA inversely correlated with OS (p = 0.018 and p = 0.037, respectively, from the Cox model). Ra223 treatment was associated with a mild decrease in some peripheral immune cell populations and a shift in the proportion of MDSCs from granulocytic to myeloid. In mice, Ra223 increased the proliferation of CD8+ and CD4+ helper T cells without leading to CD8+ T cell exhaustion in the mPC lesions. In one of the models, combining Ra223 and anti-PD-1 antibody significantly prolonged survival, which correlated with increased CD8+ T cell infiltration in tumor tissue. CONCLUSION The inflammatory cytokine IL-6 and the angiogenic biomarker PlGF at baseline were promising outcome biomarkers after standard Ra223 treatment. In mouse models, Ra223 increased intratumoral CD8+ T cell infiltration and proliferation and could improve OS when combined with anti-PD-1 ICI.
Collapse
Affiliation(s)
- Philip J Saylor
- MGH Cancer Center, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Sergey V Kozin
- Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Aya Matsui
- Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Saveli I Goldberg
- Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Shuichi Aoki
- Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Kohei Shigeta
- Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Emilie Mamessier
- Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Matthew R Smith
- MGH Cancer Center, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - M Dror Michaelson
- MGH Cancer Center, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Richard J Lee
- MGH Cancer Center, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Dan G Duda
- Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
39
|
Quagliariello V, Passariello M, Bisceglia I, Paccone A, Inno A, Maurea C, Rapuano Lembo R, Manna L, Iovine M, Canale ML, Scherillo M, Ascierto PA, Gabrielli D, De Lorenzo C, Maurea N. Combinatorial immune checkpoint blockade increases myocardial expression of NLRP-3 and secretion of H-FABP, NT-Pro-BNP, interleukin-1β and interleukin-6: biochemical implications in cardio-immuno-oncology. Front Cardiovasc Med 2024; 11:1232269. [PMID: 38322766 PMCID: PMC10844473 DOI: 10.3389/fcvm.2024.1232269] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 01/10/2024] [Indexed: 02/08/2024] Open
Abstract
Background Immune checkpoint blockade in monotherapy or combinatorial regimens with chemotherapy or radiotherapy have become an integral part of oncology in recent years. Monoclonal antibodies against CTLA-4 or PD-1 or PDL-1 are the most studied ICIs in randomized clinical trials, however, more recently, an anti-LAG3 (Lymphocyte activation gene-3) antibody, Relatlimab, has been approved by FDA in combination with Nivolumab for metastatic melanoma therapy. Moreover, Atezolizumab is actually under study in association with Ipilimumab for therapy of metastatic lung cancer. Myocarditis, vasculitis and endothelitis are rarely observed in these patients on monotherapy, however new combination therapies could expose patients to more adverse cardiovascular events. Methods Human cardiomyocytes co-cultured with human peripheral blood lymphocytes (hPBMCs) were exposed to monotherapy and combinatorial ICIs (PD-L1 and CTLA-4 or PD-1 and LAG-3 blocking agents, at 100 nM) for 48 h. After treatments, cardiac cell lysis and secretion of biomarkers of cardiotoxicity (H-FABP, troponin-T, BNP, NT-Pro-BNP), NLRP3-inflammasome and Interleukin 1 and 6 were determined through colorimetric and enzymatic assays. Mitochondrial functions were studied in cardiomyocyte cell lysates through quantification of intracellular Ca++, ATP content and NADH:ubiquinone oxidoreductase core subunit S1 (Ndufs1) levels. Histone deacetylases type 4 (HDAC-4) protein levels were also determined in cardiomyocyte cell lysates to study potential epigenetic changes induced by immunotherapy regimens. Results Both combinations of immune checkpoint inhibitors exert more potent cardiotoxic side effects compared to monotherapies against human cardiac cells co-cultured with human lymphocytes. LDH release from cardiac cells was 43% higher in PD-L1/CTLA-4 blocking agents, and 35.7% higher in PD-1/LAG-3 blocking agents compared to monotherapies. HDAC4 and intracellular Ca++ levels were increased, instead ATP content and Ndufs1 were reduced in myocardial cell lysates (p < 0.001 vs. untreated cells). Troponin-T, BNP, NT-Pro-BNP and H-FABP, were also strongly increased in combination therapy compared to monotherapy regimen. NLRP3 expression, IL-6 and IL-1β levels were also increased by PDL-1/CTLA-4 and PD-1/LAG-3 combined blocking agents compared to untreated cells and monotherapies. Conclusions Data of the present study, although in vitro, indicate that combinatorial immune checkpoint blockade, induce a pro- inflammatory phenotype, thus indicating that these therapies should be closely monitored by the multidisciplinary team consisting of oncologists, cardiologists and immunologists.
Collapse
Affiliation(s)
- V. Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - M. Passariello
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
| | - I. Bisceglia
- Servizi Cardiologici Integrati, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, Rome, Italy
| | - A. Paccone
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - A. Inno
- Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale Sacro Cuore Don Calabria, Negrar, Italy
| | - C. Maurea
- Medical Oncology, Ospedale del Mare, Naples, Italy
| | - R. Rapuano Lembo
- Department of Molecular Medicine, Ceinge-Biotecnologie Avanzate s.c.a.r.l., Naples, Italy
| | - L. Manna
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
| | - M. Iovine
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| | - M. L. Canale
- U.O.C. Cardiologia, Ospedale Versilia, Lido di Camaiore (LU), Camaiore, Italy
| | - M. Scherillo
- Cardiologia Interventistica e UTIC, A.O. San Pio, Presidio Ospedaliero Gaetano Rummo, Benevento, Italy
| | - P. A. Ascierto
- Melanoma Cancer Immunotherapy and Innovative Therapy Unit, Istituto Nazionale Tumori IRCCS Fondazione "G. Pascale", Naples, Italy
| | - D. Gabrielli
- U.O.C. Cardiologia, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, Roma – Fondazione per il Tuo Cuore – Heart Care Foundation, Firenze, Italy
| | - C. De Lorenzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
- Department of Molecular Medicine, Ceinge-Biotecnologie Avanzate s.c.a.r.l., Naples, Italy
| | - N. Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, Italy
| |
Collapse
|
40
|
Wu YX, Tian BY, Ou XY, Wu M, Huang Q, Han RK, He X, Chen SL. A novel model for predicting prognosis and response to immunotherapy in nasopharyngeal carcinoma patients. Cancer Immunol Immunother 2024; 73:14. [PMID: 38236288 PMCID: PMC10796600 DOI: 10.1007/s00262-023-03626-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/30/2023] [Indexed: 01/19/2024]
Abstract
Blood-based biomarkers of immune checkpoint inhibitors (ICIs) response in patients with nasopharyngeal carcinoma (NPC) are lacking, so it is necessary to identify biomarkers to select NPC patients who will benefit most or least from ICIs. The absolute values of lymphocyte subpopulations, biochemical indexes, and blood routine tests were determined before ICIs-based treatments in the training cohort (n = 130). Then, the least absolute shrinkage and selection operator (Lasso) Cox regression analysis was developed to construct a prediction model. The performances of the prediction model were compared to TNM stage, treatment, and Epstein-Barr virus (EBV) DNA using the concordance index (C-index). Progression-free survival (PFS) was estimated by Kaplan-Meier (K-M) survival curve. Other 63 patients were used for validation cohort. The novel model composed of histologic subtypes, CD19+ B cells, natural killer (NK) cells, regulatory T cells, red blood cells (RBC), AST/ALT ratio (SLR), apolipoprotein B (Apo B), and lactic dehydrogenase (LDH). The C-index of this model was 0.784 in the training cohort and 0.735 in the validation cohort. K-M survival curve showed patients with high-risk scores had shorter PFS compared to the low-risk groups. For predicting immune therapy responses, the receiver operating characteristic (ROC), decision curve analysis (DCA), net reclassifcation improvement index (NRI) and integrated discrimination improvement index (IDI) of this model showed better predictive ability compared to EBV DNA. In this study, we constructed a novel model for prognostic prediction and immunotherapeutic response prediction in NPC patients, which may provide clinical assistance in selecting those patients who are likely to gain long-lasting clinical benefits to anti-PD-1 therapy.
Collapse
Affiliation(s)
- Ya-Xian Wu
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Bo-Yu Tian
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Xin-Yuan Ou
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Meng Wu
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Qi Huang
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Run-Kun Han
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510006, Guangdong, People's Republic of China
| | - Xia He
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510006, Guangdong, People's Republic of China.
| | - Shu-Lin Chen
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510006, Guangdong, People's Republic of China.
- Research Center for Translational Medicine, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
41
|
Nakahara Y, Kouro T, Motoyama S, Miura M, Fujita K, Igarashi Y, Higashijima N, Matsuo N, Himuro H, Wei F, Horaguchi S, Tsuji K, Mano Y, Komahashi M, Saito H, Azuma K, Sasada T. Circulating IL-6 and not its circulating signaling components sIL-6R and sgp130 demonstrate clinical significance in NSCLC patients treated with immune checkpoint inhibitors. Front Cell Dev Biol 2024; 11:1324898. [PMID: 38469154 PMCID: PMC10926441 DOI: 10.3389/fcell.2023.1324898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/11/2023] [Indexed: 03/13/2024] Open
Abstract
Introduction: Clinical roles of plasma IL-6 levels have been reported in patients with various cancers, including non-small cell lung cancer (NSCLC), treated with immune checkpoint inhibitors (ICIs). However, the roles of other IL-6 signaling components, soluble IL-6 receptor (sIL-6R) and soluble gp130 (sgp130), in the plasma have not been elucidated. Methods: Blood was collected from 106 patients with NSCLC before initiation of ICI treatment (anti-PD-1 or anti-PD-L1 antibody). Plasma levels of IL-6, sIL-6R, sgp130, and their complexes were assessed by Cox regression hazard model to evaluate their clinical significance. The clinical role of IL-6 or IL-6R genetic polymorphisms was also analyzed. Results: Cox regression analysis showed that higher plasma IL-6 levels significantly predicted unfavorable overall survival (OS; hazard ratio [HR] 1.34, 95% confidence interval [CI] 1.05-1.68, p = 0.012) in NSCLC patients treated with ICIs. However, plasma sIL-6R and sgp130 levels showed no prognostic significance (p = 0.882 and p = 0.934, respectively). In addition, the estimated concentrations of binary IL-6:sIL-6R and ternary IL-6:sIL-6R:sgp130 complexes and their ratios (binary/ternary complex) were not significantly associated with OS (p = 0.647, p = 0.727, and p = 0.273, respectively). Furthermore, the genetic polymorphisms of IL-6 (-634G>C) and IL-6R (48892A>C) showed no clinical role by Kaplan-Meier survival analysis (p = 0.908 and p = 0.639, respectively). Discussion: These findings demonstrated the clinical significance of plasma levels of IL-6, but not of other IL-6 signaling components, sIL-6R and sgp130, suggesting that classical IL-6 signaling, but not trans-signaling, may be related to anti-tumor immune responses in cancer patients treated with ICIs.
Collapse
Affiliation(s)
- Yoshiro Nakahara
- Department of Respiratory Medicine, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara, Kanagawa, Japan
| | - Taku Kouro
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, Japan
| | - Satoru Motoyama
- Department of Comprehensive Cancer Control, Akita University Graduate School of Medicine, Akita, Japan
- Division of Esophageal Surgery, Akita University Hospital, Akita, Japan
- Department of Gastroenterological Surgery, Japanese Red Cross Akita Hospital, Akita, Japan
| | - Masatomo Miura
- Department of Pharmacy, Akita University Hospital, Akita, Japan
| | - Kazuma Fujita
- Department of Pharmacy, Akita University Hospital, Akita, Japan
| | - Yuka Igarashi
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Naoko Higashijima
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Norikazu Matsuo
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Hidetomo Himuro
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, Japan
| | - Feifei Wei
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, Japan
| | - Shun Horaguchi
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, Japan
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Kayoko Tsuji
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, Japan
| | - Yasunobu Mano
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, Japan
| | - Mitsuru Komahashi
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, Japan
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Haruhiro Saito
- Department of Respiratory Medicine, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Koichi Azuma
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Tetsuro Sasada
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, Japan
| |
Collapse
|
42
|
Li J, Shi W, Xiong J, Huang Y, He Y, Zhou Y, Yang Z, Peng Y. Toxicity spectrum and risk factors for chemo-immunotherapy in locally advanced or metastatic lung cancer. Clin Exp Immunol 2023; 214:162-169. [PMID: 37696500 PMCID: PMC10714187 DOI: 10.1093/cei/uxad105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 06/11/2023] [Accepted: 09/07/2023] [Indexed: 09/13/2023] Open
Abstract
Chemo-immunotherapy has become the best first-line treatment for advanced lung cancer patients without oncogenic drivers. However, it may also lead to an increased incidence and severity of treatment-related adverse events. In this retrospective study, lung cancer patients administrated with either anti-PD-1 or anti-PD-L1 treatment plus chemotherapy were included. Data on demographic characteristics, disease characteristics, treatment strategies, laboratory results, and clinical outcomes were collected from the Electronic Medical Records System and evaluation scales. Chi-square, univariate, and multivariate logistic regression analyses were used to identify the risk factors for immune-related adverse events (irAEs). A total of 116 patients were included in the study, and the majority experienced treatment-related adverse events. Adverse events of any grade were reported in 114 (98.3%) patients, with 73 (62.9%) experiencing Grade 3 or higher events. The most frequent adverse events were anemia (67.2%), decreased appetite (62.9%), and alopecia (53.4%). Fifty-four (46.6%) patients were diagnosed with irAEs, with hypothyroidism (28.4%) being the most commonly reported. Multivariable analysis demonstrated a significant correlation between the number of treatment cycles, elevated baseline levels of thyroid stimulating hormone (TSH) and interleukin-6 (IL-6) with irAEs (OR = 1.222, P = 0.009, OR = 1.945, P = 0.016, OR = 1.176, P = 0.004), and IL-6 was identified as a strong predictor of severe irAEs (OR = 1.084, P = 0.014). Our study demonstrated the safety of chemo-immunotherapy in lung cancer patients without additional toxicity. The number of treatment cycles, higher baseline levels of TSH and IL-6 were identified as potential clinical biomarkers for irAEs.
Collapse
Affiliation(s)
- Jinjin Li
- Department of Cancer Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Wenhao Shi
- Department of Cancer Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jin Xiong
- Department of Cancer Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yusheng Huang
- Department of Cancer Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yi He
- Department of Cancer Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yan Zhou
- Department of Cancer Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Zhenzhou Yang
- Department of Cancer Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Chongqing Clinical Research Center for Geriatrics and Gerontology, Chongqing, China
| | - Yuan Peng
- Department of Cancer Center, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
- Guchengtai Community Health Center of Chengxi District, Xining, China
| |
Collapse
|
43
|
Wang C, Pan Y, Liu Y, Guo B, Shi J, Rong G, Guo Z, Li Z, Yang Q, Nie J, Han W. Long-term complete remission and peripheral biomarkers in Hodgkin lymphoma patients after decitabine-plus-camrelizumab epi-immunotherapy and treatment cessation. MedComm (Beijing) 2023; 4:e428. [PMID: 38020717 PMCID: PMC10665599 DOI: 10.1002/mco2.428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 12/01/2023] Open
Abstract
Patients with relapsed/refractory classical Hodgkin lymphoma (cHL) achieve complete response (CR) after decitabine-plus-camrelizumab therapy, while long-term outcome especially after treatment discontinuation remains unclear. We present a retrospective analysis of 87 relapsed/refractory cHL patients who acquired CR after decitabine-plus-camrelizumab. Patients were divided into two groups and received consolidation treatment every 3-4 or 6-12 weeks, and 1-year of continuous CR was guaranteed for treatment cessation. At a median follow-up of 5.3 years, the median relapse-free survival (RFS) after achieving CR with decitabine-plus-camrelizumab therapy was 4.5 years, and patients underwent consolidation per 3-4 weeks might have longer RFS. The baseline percentage of peripheral central memory T cells was not associated with RFS, while patients with higher pretreatment serum levels of interleukin-6 (IL-6) and lactate dehydrogenase (LDH) had significantly shorter RFS and increased risk for disease recurrence. Fifty-seven patients completed and discontinued decitabine-plus-camrelizumab, and their median RFS had not been reached. The 2-year RFS rate after treatment cessation was 78% (95% CI, 67-90%). Patients in the high-risk subgroup with higher pretreatment IL-6 and LDH levels showed poor treatment-free remission. Moreover, decitabine-plus-camrelizumab therapy was safe and cost-effective. In conclusion, patients who obtained CR with decitabine-plus-camrelizumab and received consolidation per 3-4 weeks can achieve long-term remission after treatment discontinuation.
Collapse
Affiliation(s)
- Chunmeng Wang
- The Second School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
- Department of Bio‐therapeuticthe First Medical Centre, Chinese PLA General HospitalBeijingChina
- Department of Bio‐therapeuticthe Fifth Medical Centre, Chinese PLA General HospitalBeijingChina
| | - Yuting Pan
- Chinese People's Liberation Army Medical SchoolChinese PLA General HospitalBeijingChina
| | - Yang Liu
- Department of Bio‐therapeuticthe First Medical Centre, Chinese PLA General HospitalBeijingChina
- Department of Bio‐therapeuticthe Fifth Medical Centre, Chinese PLA General HospitalBeijingChina
| | - Bing Guo
- Department of Bio‐therapeuticthe Fifth Medical Centre, Chinese PLA General HospitalBeijingChina
| | - Jinhong Shi
- Department of Bio‐therapeuticthe Fifth Medical Centre, Chinese PLA General HospitalBeijingChina
| | - Guanghua Rong
- Department of Bio‐therapeuticthe Fifth Medical Centre, Chinese PLA General HospitalBeijingChina
| | - Zhipeng Guo
- Department of Bio‐therapeuticthe Fifth Medical Centre, Chinese PLA General HospitalBeijingChina
| | - Zhifang Li
- Department of Bio‐therapeuticthe Fifth Medical Centre, Chinese PLA General HospitalBeijingChina
| | - Qingming Yang
- Department of Bio‐therapeuticthe First Medical Centre, Chinese PLA General HospitalBeijingChina
- Department of Bio‐therapeuticthe Fifth Medical Centre, Chinese PLA General HospitalBeijingChina
| | - Jing Nie
- Department of Bio‐therapeuticthe First Medical Centre, Chinese PLA General HospitalBeijingChina
| | - Weidong Han
- The Second School of Clinical MedicineSouthern Medical UniversityGuangzhouChina
- Department of Bio‐therapeuticthe First Medical Centre, Chinese PLA General HospitalBeijingChina
- Department of Bio‐therapeuticthe Fifth Medical Centre, Chinese PLA General HospitalBeijingChina
- Changping LaboratoryBeijingChina
| |
Collapse
|
44
|
Shibata Y, Kishida T, Kouro T, Wei F, Igarashi Y, Himuro H, Noguchi T, Koizumi M, Suzuki T, Osaka K, Saigusa Y, Sasada T. Immune mediators as predictive biomarkers for anti-PD-1 antibody therapy in urothelial carcinoma. Front Pharmacol 2023; 14:1269935. [PMID: 38026978 PMCID: PMC10679331 DOI: 10.3389/fphar.2023.1269935] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction: This study aimed to identify immune mediators, including cytokines, chemokines, and growth factors, in the plasma for predicting treatment efficacy and immune-related adverse events (irAEs) in advanced urothelial carcinoma (aUC) treated with immune checkpoint inhibitors (ICIs). Methods: We enrolled 57 patients with aUC who were treated with the anti-programmed cell death protein 1 (PD-1) antibody pembrolizumab after the failure of platinum-based chemotherapy between February 2018 and December 2020. Plasma levels of 73 soluble immune mediators were measured before and 6 weeks after initiating pembrolizumab therapy. The association of estimated soluble immune mediators with clinical outcomes, including overall survival (OS), progression-free survival (PFS), anti-tumor responses, and irAEs, were statistically evaluated. Results: In the multivariate analysis, levels of 18 factors at baseline and 12 factors during treatment were significantly associated with OS. Regarding PFS, baseline levels of 17 factors were significantly associated with PFS. Higher levels of interleukin (IL)-6, IL-8, soluble tumor necrosis factor receptor 1 (sTNF-R1), and IL-12 (p40), both at baseline and post-treatment, were significantly associated with worse OS. Conversely, low IL-6 and high TWEAK levels at baseline were associated with irAEs. Among identified factors, interferon (IFN) γ and IL-12 (p40) were repeatedly identified; high baseline levels of these factors were risk factors for worse OS and PFS, as well as progressive disease. Notably, using correlation and principal component analysis, factors significantly associated with clinical outcomes were broadly classified into three groups exhibiting similar expression patterns. Discussion: Measuring plasma levels of soluble immune mediators, such as IL-6, IL-8, sTNF-R1, IFNγ, and IL-12 (p40), could be recommended for predicting prognosis and irAEs in ICI-treated patients with aUC.
Collapse
Affiliation(s)
- Yosuke Shibata
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Takeshi Kishida
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Taku Kouro
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Feifei Wei
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Yuka Igarashi
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Hidetomo Himuro
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Takeaki Noguchi
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Mitsuyuki Koizumi
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Takahisa Suzuki
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Kimito Osaka
- Department of Urology, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| | - Yusuke Saigusa
- Department of Biostatistics, School of Medicine, Yokohama City University, Yokohama, Kanagawa, Japan
| | - Tetsuro Sasada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Kanagawa, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Kanagawa, Japan
| |
Collapse
|
45
|
Samart P, Heenatigala Palliyage G, Issaragrisil S, Luanpitpong S, Rojanasakul Y. Musashi-2 in cancer-associated fibroblasts promotes non-small cell lung cancer metastasis through paracrine IL-6-driven epithelial-mesenchymal transition. Cell Biosci 2023; 13:205. [PMID: 37941042 PMCID: PMC10631049 DOI: 10.1186/s13578-023-01158-5] [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: 06/28/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023] Open
Abstract
BACKGROUND Lung cancer, the most common cause of cancer-related mortality worldwide, is predominantly associated with advanced/metastatic disease. The interaction between tumor cells and cancer-associated fibroblasts (CAFs) in tumor microenvironment is known to be essential for regulating tumor progression and metastasis, but the underlying mechanisms, particularly the role of RNA-binding protein Musashi-2 (MSI2) in CAFs in promoting non-small cell lung cancer (NSCLC) invasiveness and metastatic spread, remain obscure. METHODS Genomic and proteomic database analyses were performed to evaluate the potential clinical significance of MSI2 in NSCLC tumor and stromal clinical specimens. Molecular approaches were used to modify MSI2 in CAFs and determine its functional role in NSCLC cell motility in vitro using 2D and 3D models, and in metastasis in a xenograft mouse model using live-cell imaging. RESULTS MSI2, both gene and protein, is upregulated in NSCLC tissues and is associated with poor prognosis and high metastatic risk in patients. Interestingly, MSI2 is also upregulated in NSCLC stroma and activated fibroblasts, including CAFs. Depletion of MSI2 in CAFs by CRISPR-Cas9 strongly inhibits NSCLC cell migration and invasion in vitro, and attenuates local and distant metastatic spread of NSCLC cells in vivo. The crosstalk between CAFs and NSCLC cells occurs via paracrine signaling, which is regulated by MSI2 in CAFs via IL-6. The secreted IL-6 promotes epithelial-mesenchymal transition in NSCLC cells, which drives metastasis. CONCLUSION Our findings reveal for the first time that MSI2 in CAFs is important in CAF-mediated NSCLC cell invasiveness and metastasis via IL-6 paracrine signaling. Therefore, targeting the MSI2/IL-6 axis in CAFs could be effective in combating NSCLC metastasis.
Collapse
Affiliation(s)
- Parinya Samart
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, 26506, USA
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Siriraj Hospital, Bangkoknoi, Bangkok, 10700, Thailand
| | | | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Siriraj Hospital, Bangkoknoi, Bangkok, 10700, Thailand
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, 2 Siriraj Hospital, Bangkoknoi, Bangkok, 10700, Thailand.
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences, West Virginia University, Morgantown, WV, 26506, USA.
- WVU Cancer Institute, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
46
|
Soler MF, Abaurrea A, Azcoaga P, Araujo AM, Caffarel MM. New perspectives in cancer immunotherapy: targeting IL-6 cytokine family. J Immunother Cancer 2023; 11:e007530. [PMID: 37945321 PMCID: PMC10649711 DOI: 10.1136/jitc-2023-007530] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2023] [Indexed: 11/12/2023] Open
Abstract
Chronic inflammation has been recognized as a canonical cancer hallmark. It is orchestrated by cytokines, which are master regulators of the tumor microenvironment (TME) as they represent the main communication bridge between cancer cells, the tumor stroma, and the immune system. Interleukin (IL)-6 represents a keystone cytokine in the link between inflammation and cancer. Many cytokines from the IL-6 family, which includes IL-6, oncostatin M, leukemia inhibitory factor, IL-11, IL-27, IL-31, ciliary neurotrophic factor, cardiotrophin 1, and cardiotrophin-like cytokine factor 1, have been shown to elicit tumor-promoting roles by modulating the TME, making them attractive therapeutic targets for cancer treatment.The development of immune checkpoint blockade (ICB) immunotherapies has radically changed the outcome of some cancers including melanoma, lung, and renal, although not without hurdles. However, ICB shows limited efficacy in other solid tumors. Recent reports support that chronic inflammation and IL-6 cytokine signaling are involved in resistance to immunotherapy. This review summarizes the available preclinical and clinical data regarding the implication of IL-6-related cytokines in regulating the immune TME and the response to ICB. Moreover, the potential clinical benefit of combining ICB with therapies targeting IL-6 cytokine members for cancer treatment is discussed.
Collapse
Affiliation(s)
- Maria Florencia Soler
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, Donostia-San Sebastian, Spain
| | - Andrea Abaurrea
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, Donostia-San Sebastian, Spain
| | - Peio Azcoaga
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, Donostia-San Sebastian, Spain
| | - Angela M Araujo
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, Donostia-San Sebastian, Spain
| | - Maria M Caffarel
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, Donostia-San Sebastian, Spain
- Ikerbasque Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
47
|
Peng Y, Qi Q, Zhu M, Zhang Y, Bao Y, Liu Y. Plasma levels of 12 different cytokines correlate to PD-1 inhibitor combined chemotherapy responses in advanced non-small-cell lung cancer patient. Int Immunopharmacol 2023; 124:110888. [PMID: 37690239 DOI: 10.1016/j.intimp.2023.110888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/17/2023] [Accepted: 08/29/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND Targeted anti-programmed death receptor 1 (PD-1) monoclonal antibodies, when combined with chemotherapy, have shown improved outcomes in non-small cell lung cancer (NSCLC). However, it is important to note that not all patients benefit from this treatment, and there is a pressing need for more reliable efficacy measures and potential predictors of outcome. Cytokines, which are important molecules in the immune system, have been considered as potential biomarkers in clinical settings, but their precise clinical use remains unclear. In this study, our objective was to assess whether the levels of cytokines in the patient's blood sample are associated with tumor response to anti-PD-1 monoclonal antibodies combined with chemotherapy as well as the survival of patients with advanced non-small cell lung cancer. MATERIALS AND METHODS A total of 12 plasma cytokines were measured in advanced NSCLC patients (n = 35) and healthy individuals (n = 26) using multi-microsphere flow immunofluorescence. The relationship between cytokine levels and clinical response was analyzed using nonparametric Wilcoxon matched-pair ranked tests. Progression-free survival (PFS) time was recorded for all patients through radiographic outcome assessment and telephone follow-up. Survival curves were generated using the Kaplan-Meier and log-rank tests, and the thresholds for cytokines were determined using receiver operating characteristic analysis (ROC). RESULTS The expression levels of interleukin IL-6, IL-1 β, IFN-γ, IL-12p70, and TNF-α were significantly lower in the control group than those in the NSCLC group (p = 0.001, p = 0.0028, p = 0.019, p = 0.0001, p = 0.0021). High IL-10 levels at baseline and after 4 cycles of treatment conferred a worse prognosis; in addition, high TNF-α levels in patients after two cycles of immunochemotherapy suggested drug resistance. High levels of IL-6 and IFN-γ in patients undergoing four cycles of immunochemotherapy were associated with worse PFS. CONCLUSIONS Our study suggests that cytokines can serve as detection indicators for predicting efficacy in non-small cell lung cancer patients undergoing anti-PD-1 combined with chemotherapy treatment. Elevated levels of IL-10, TNF-α, IL-6, and IFN-γ in the plasma may indicate a higher likelihood of experiencing a worse clinical outcome.
Collapse
Affiliation(s)
- Yun Peng
- Clinical Oncology Laboratory, Changzhou Tumor Hospital, Changzhou, Jiangsu Province 213002, China
| | - Qiufeng Qi
- Clinical Oncology Laboratory, Changzhou Tumor Hospital, Changzhou, Jiangsu Province 213002, China
| | - Ming Zhu
- Clinical Oncology Laboratory, Changzhou Tumor Hospital, Changzhou, Jiangsu Province 213002, China
| | - Yaping Zhang
- Clinical Oncology Laboratory, Changzhou Tumor Hospital, Changzhou, Jiangsu Province 213002, China
| | - Yanqing Bao
- Clinical Oncology Laboratory, Changzhou Tumor Hospital, Changzhou, Jiangsu Province 213002, China
| | - Yongping Liu
- Clinical Oncology Laboratory, Changzhou Tumor Hospital, Changzhou, Jiangsu Province 213002, China; Department of Oncology, Changzhou Tumor Hospital, Changzhou, Jiangsu Province 213002, China.
| |
Collapse
|
48
|
Vita E, Stefani A, Piro G, Mastrantoni L, Cintoni M, Cicchetti G, Sparagna I, Monaca F, Horn G, Russo J, Barone D, Di Salvatore M, Trisolini R, Lococo F, Mazzarella C, Cancellieri A, Carbone C, Larici AR, Mele MC, Pilotto S, Milella M, Tortora G, Bria E. Leptin-mediated meta-inflammation may provide survival benefit in patients receiving maintenance immunotherapy for extensive-stage small cell lung cancer (ES-SCLC). Cancer Immunol Immunother 2023; 72:3803-3812. [PMID: 37668709 PMCID: PMC10576666 DOI: 10.1007/s00262-023-03533-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 08/20/2023] [Indexed: 09/06/2023]
Abstract
BACKGROUND Only few ES-SCLC patients experience long-term survival benefit by maintenance IT. Adipokines-induced metabolic meta-inflammation has been related to enhanced responsiveness to IT in obese patients; however, their prognostic role in SCLC is currently controversial. METHODS Pre-treatment CT scan was used for determining distribution of abdominal adiposity, and blood samples were collected at fasting for measuring glycemia, insulin, ghrelin, leptin and adipokines (TNF-α, IFN-γ, IL-6 and MCP-1). Patients with known history of DM type II or metabolic syndrome with HOMA index > 2.5 were considered insulin resistant (IR). RESULTS In ES-SCLC pts receiving maintenance IT, increased leptin concentration and higher leptin/visceral adipose tissue (VAT) ratio were significantly associated with prolonged PFS. By applying a hierarchical clustering algorithm, we identified a cluster of patients characterized by higher leptin values and lower pro-inflammatory cytokines (TNF-α, IFN-γ and IL-6) who experienced longer PFS (13.2 vs 8.05 months; HR: 0.42 [0.18-0.93] p = 0.02) and OS (18.04 vs 12.09 mo; HR: 0.53 [0.25-1.29] p = 0.07). CONCLUSIONS Adipokines can play a crucial role to determining effectiveness of anti-cancer immunotherapy. The role of metabolic immune dysfunctions needs further pre-clinical validation and is currently investigated in the larger prospective cohort.
Collapse
Affiliation(s)
- Emanuele Vita
- UOSD Oncologia Toraco-Polmonare, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
- Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Alessio Stefani
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Geny Piro
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Luca Mastrantoni
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marco Cintoni
- UOC Nutrizione Clinica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giuseppe Cicchetti
- UOC Radiologia Toracica e Cardiovascolare, Advanced Radiodiagnostics Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ileana Sparagna
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Federico Monaca
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Guido Horn
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Jacopo Russo
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Diletta Barone
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Mariantonietta Di Salvatore
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Rocco Trisolini
- Università Cattolica del Sacro Cuore, Rome, Italy
- UOC Pneumologia Interventistica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Filippo Lococo
- Università Cattolica del Sacro Cuore, Rome, Italy
- UOC Chirurgia Toracica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Ciro Mazzarella
- UOC Radioterapia Oncologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Alessandra Cancellieri
- UOC Anatomia Patologica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Carmine Carbone
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Anna Rita Larici
- Università Cattolica del Sacro Cuore, Rome, Italy
- UOC Radiologia Toracica e Cardiovascolare, Advanced Radiodiagnostics Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Maria Cristina Mele
- Università Cattolica del Sacro Cuore, Rome, Italy
- UOC Nutrizione Clinica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Sara Pilotto
- UOC Oncologia Medica, Verona University Hospital Trust, Verona, Italy
| | - Michele Milella
- UOC Oncologia Medica, Verona University Hospital Trust, Verona, Italy
| | - Giampaolo Tortora
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Emilio Bria
- UOSD Oncologia Toraco-Polmonare, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
49
|
Zhu X, Yu B, Shen Y, Zhao Y, Fu X, Zhu Y, Gu G, Liu C. Screening biomarkers for predicting the efficacy of immunotherapy in patients with PD-L1 overexpression. J Cancer Res Clin Oncol 2023; 149:12965-12976. [PMID: 37468609 PMCID: PMC10587271 DOI: 10.1007/s00432-023-05160-9] [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: 05/31/2023] [Accepted: 07/09/2023] [Indexed: 07/21/2023]
Abstract
PURPOSE Immunotherapy plays an important role in non-small cell lung cancer (NSCLC); in particular, immune checkpoint inhibitors (ICIs) therapy has good therapeutic effects in PD-L1-positive patients. This study aims to screen NSCLC patients with PD-L1-positive expression and select effective biomarkers for ICI immunotherapy. METHODS Collected tumor samples from the Affiliated Cancer Hospital of Xinjiang Medical University and 117 patients with stage III-IV NSCLC were included in the study. All patients were on first- or second-line therapy and not on targeted therapy. Based on the molecular profiles and clinical features, we screened biomarkers for predicting the efficacy of immunotherapy in patients with PD-L1 overexpression. RESULTS 117 NSCLC patients receiving ICIs immunotherapy were enrolled. First, we found that immunotherapy was more effective in patients with positive PD-L1 expression. Second, we found that ROS1 gene mutations, KRAS gene mutations, tumor stage, and the endocrine system diseases history are independent prognostic factors for PD-L1 positive patients. Then we combined independent risk factors and constructed a new Nomogram to predict the therapeutic efficacy of ICIs immunotherapy in PD-L1 positive patients. The Nomogram integrates these factors into a prediction model, and the predicted C-statistic of 3 months, 6 months and 12 months are 0.85, 0.84 and 0.85, which represents the high predictive accuracy of the model. CONCLUSIONS We have established a model that can predict the efficacy of ICIs immunotherapy in PD-L1 positive patients. The model consists of ROS1 gene mutations, KRAS gene mutations, tumor staging, and endocrine system disease history, and has good predictive ability.
Collapse
Affiliation(s)
- Xiaodan Zhu
- Department of Pulmonary Medicine, Affiliated Cancer Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Xinshi District, Urumqi, 830000, Xinjiang, China
| | - Bo Yu
- Department of Medicine, Beijing USCI Medical Laboratory, No. 65, Xingshikou Road, Haidian District, Beijing, 100195, China
| | - Yanli Shen
- Department of Pulmonary Medicine, Affiliated Cancer Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Xinshi District, Urumqi, 830000, Xinjiang, China
| | - Yan Zhao
- Department of Pulmonary Medicine, Affiliated Cancer Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Xinshi District, Urumqi, 830000, Xinjiang, China
| | - Xiyujing Fu
- Department of Pulmonary Medicine, Affiliated Cancer Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Xinshi District, Urumqi, 830000, Xinjiang, China
| | - Yunji Zhu
- Department of Pulmonary Medicine, Affiliated Cancer Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Xinshi District, Urumqi, 830000, Xinjiang, China
| | - Guomin Gu
- Department of Pulmonary Medicine, Affiliated Cancer Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Xinshi District, Urumqi, 830000, Xinjiang, China.
| | - Chunling Liu
- Department of Pulmonary Medicine, Affiliated Cancer Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Xinshi District, Urumqi, 830000, Xinjiang, China.
| |
Collapse
|
50
|
Röhl L, Wellhausen J, Berszin M, Krücken I, Zebralla V, Pirlich M, Wiegand S, Dietz A, Wald T, Wichmann G. Immune checkpoint blockade induced shifts in cytokine expression patterns in peripheral blood of head and neck cancer patients are linked to outcome. Front Immunol 2023; 14:1237623. [PMID: 37849764 PMCID: PMC10577218 DOI: 10.3389/fimmu.2023.1237623] [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: 06/09/2023] [Accepted: 09/12/2023] [Indexed: 10/19/2023] Open
Abstract
Background Immune-checkpoint blockade (ICB) of programmed-death-1 (PD-1) with pembrolizumab or nivolumab is approved for treating recurrent/metastatic (R/M) head and neck squamous cell carcinoma (HNSCC). NadiHN and ADRISK are phase IIB trials investigating in locally advanced (LA) HNSCC having low or high risk of recurrence the potential benefits from adding nivolumab to post-operative radiotherapy or pembrolizumab to cisplatin-based radio-chemotherapy. Methods Along five randomized controlled ICB trials including NadiHN and ADRISK, blood samples were taken before and after starting ICB in n=25 patients. Concentrations of vascular endothelial growth factor A (VEGF), CCL2 (MCP-1), interleukin-6 (IL-6), IL-8, interferon-gamma (IFN-γ), and CXCL10 (IP-10) pre- and post-ICB in EDTA-anticoagulated plasma and serum were compared. We used receiver operating characteristic (ROC) curves to identify optimal cutoff for defining subgroups before analyzing overall survival (OS) applying Kaplan-Meier plots and multivariate Cox regression. Results We detected huge heterogeneity between cytokine patterns in pre-and post-ICB plasma and serum. We observed high correlation between concentrations of some cytokines. Despite absent systematic OS differences after ICB with pembrolizumab or nivolumab or between LA-HNSCC versus R/M HNSCC patients, we noticed improved outcome of patients having lower IFN-γ concentrations pre- and post-ICB and following ICB reduced concentrations of VEGF, IL-6, and IL-8 but not MCP-1. Contrarily, increases in IL-6, IL-8, and VEGF levels correlated with impaired outcome. Multivariate Cox regression revealed five independent OS predictors among cytokines; using natural logarithms of their hazard ratios to estimate an individual's risk of dying, three cytokine-expression pattern (CEP)-risk groups with no death within mean (95% confidence interval) follow-up of 29.2 (22.1-36.2) months and median OS of 11.3 (8.8-13.8) and 2.9 (0.4-5.4) months were found. Conclusion Whereas individual pre- or post-ICB cytokine concentrations in serum or plasma alone failed to predict the survivor group, CEP-risk groups may support the identification of individual patients with long-lasting benefit from ICB.
Collapse
Affiliation(s)
- Louisa Röhl
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
| | - Jana Wellhausen
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
| | - Michael Berszin
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
| | - Irene Krücken
- Institute of Pathology, University Hospital Leipzig, Leipzig, Germany
| | - Veit Zebralla
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
| | - Markus Pirlich
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
| | - Susanne Wiegand
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
| | - Andreas Dietz
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
| | - Theresa Wald
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
| | - Gunnar Wichmann
- Department of Otorhinolaryngology, Head and Neck surgery, University Hospital Leipzig, Leipzig, Germany
| |
Collapse
|