1
|
Qiao Y, Xie D, Li Z, Cao S, Zhao D. Global research trends on biomarkers for cancer immunotherapy: Visualization and bibliometric analysis. Hum Vaccin Immunother 2025; 21:2435598. [PMID: 39773010 PMCID: PMC11730411 DOI: 10.1080/21645515.2024.2435598] [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: 09/05/2024] [Revised: 11/08/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
The global burden of cancer continues to grow, posing a significant public health challenge. Although cancer immunotherapy has shown significant efficacy, the response rate is not high. Therefore, the objective of our research was to identify the latest research trends and hotspots on biomarkers from 1993 to 2023. Data were collected from the database Web of Science core collection. Bibliometric analysis and visualization were conducted with CiteSpace(6.3.1), VOSviewer (v1.6.20), R-bibliometrix(v4.3.3), and Microsoft Excel(2019). A total of 2686 literatures were retrieved. The sheer annual volume of publications has shown a rapid upward trend since 2015. The United States has generated the most publications and Harvard University ranked as a leading institution. The global biomarker research on immune checkpoint inhibitors (ICIs) revealed regional differences and in-depth explorations should be promoted in developing countries. Although China has become the second largest country in terms of publication, the average citation per paper and the total link strength were both lower than the other countries. The research on biomarkers mainly concentrated upon the following aspects: PD-1/PD-L1, CTLA-4, gene expression, adverse events, total mutational burden (TMB), body mass index (BMI), gut microbiota, cd8(+)/cd4(+) t-cells, and blood-related biomarkers such as lactate dehydrogenase (LDH), neutrophil-lymphocyte ratio (NLR), cytokines. Furthermore, "artificial intelligence" and "machine learning" have become the most important research hotspot over the last 2 y, which will help us to identify useful biomarkers from complex big data and provide a basis for precise medicine for malignant tumors.
Collapse
Affiliation(s)
- Yuan Qiao
- Department of Clinical Pharmacy, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| | - Dong Xie
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhengxiang Li
- Department of Pharmacy, Tianjin Medical University General Hospital, Tianjin, China
| | - Shaohua Cao
- Department of Clinical Pharmacy, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| | - Dong Zhao
- Department of Clinical Laboratory, Yan’an University Affiliated Hospital, Yan’an, Shaanxi, China
| |
Collapse
|
2
|
von Itzstein MS, Liu J, Mu-Mosley H, Fattah F, Park JY, SoRelle JA, Farrar JD, Gwin ME, Hsiehchen D, Gloria-McCutchen Y, Wakeland EK, Cole S, Bhalla S, Kainthla R, Puzanov I, Switzer B, Daniels GA, Zakharia Y, Shaheen M, Zhang J, Xie Y, Gerber DE. Racial Differences in Systemic Immune Parameters in Individuals With Lung Cancer. JTO Clin Res Rep 2025; 6:100751. [PMID: 39619775 PMCID: PMC11605181 DOI: 10.1016/j.jtocrr.2024.100751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/10/2024] [Accepted: 10/13/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction Racial and ethnic disparities in the presentation and outcomes of lung cancer are widely known. To evaluate potential factors contributing to these observations, we measured systemic immune parameters in Black and White patients with lung cancer. Methods Patients scheduled to receive cancer immunotherapy were enrolled in a multi-institutional prospective biospecimen collection registry. Clinical and demographic information were obtained from electronic medical records. Pretreatment peripheral blood samples were collected and analyzed for cytokines using a multiplex panel and for immune cell populations using mass cytometry. Differences between Black and White patients were determined and corrected for multiple comparisons. Results A total of 187 patients with NSCLC (Black, 19; White, 168) were included in the analysis. Compared with White patients, Black patients had greater comorbidity (median Charlson Comorbidity Index 5 versus 3; p = 0.04) and were more likely to have received previous chemotherapy (79% versus 47%; p = 0.03). Black patients had significantly lower levels of CCL23 and CCL27 and significantly higher levels of CCL8, CXCL1, CCL26, CCL25, CCL1, IL-1b, CXCL16, and IFN-γ (all p < 0.05, false discovery rate < 0.1). Black patients also exhibited greater populations of nonclassical CD16+ monocytes, NKT-like cells, CD4+ cells, CD38+ monocytes, and CD57+ gamma delta T cells (all p < 0.05). Conclusions Black and White patients with lung cancer exhibit several differences in immune parameters, with Black patients exhibiting greater levels of numerous proinflammatory cytokines and cell populations. The etiology and clinical significance of these differences warrant further evaluation.
Collapse
Affiliation(s)
- Mitchell S. von Itzstein
- Department of Internal Medicine (Division of Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, Texas
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jialiang Liu
- Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hong Mu-Mosley
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Farjana Fattah
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jason Y. Park
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jeffrey A. SoRelle
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - J. David Farrar
- Department of Immunology, University of Texas Southwestern Medical Center. Dallas, Texas
| | - Mary E. Gwin
- Department of Internal Medicine (Division of Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, Texas
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - David Hsiehchen
- Department of Internal Medicine (Division of Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, Texas
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yvonne Gloria-McCutchen
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Edward K. Wakeland
- Department of Immunology, University of Texas Southwestern Medical Center. Dallas, Texas
| | - Suzanne Cole
- Department of Internal Medicine (Division of Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, Texas
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sheena Bhalla
- Department of Internal Medicine (Division of Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, Texas
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Radhika Kainthla
- Department of Internal Medicine (Division of Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, Texas
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Igor Puzanov
- Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | | | - Gregory A. Daniels
- Moores Cancer Center, University of California San Diego Health, San Diego, California
| | | | | | | | - Yang Xie
- Quantitative Biomedical Research Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Peter O’Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, Texas
| | - David E. Gerber
- Department of Internal Medicine (Division of Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, Texas
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
- Peter O’Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
3
|
Ogutu S, Mohammed M, Mwambi H. Cytokine profiles as predictors of HIV incidence using machine learning survival models and statistical interpretable techniques. Sci Rep 2024; 14:29895. [PMID: 39622992 PMCID: PMC11612445 DOI: 10.1038/s41598-024-81510-y] [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: 07/05/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024] Open
Abstract
HIV remains a critical global health issue, with an estimated 39.9 million people living with the virus worldwide by the end of 2023 (according to WHO). Although the epidemic's impact varies significantly across regions, Africa remains the most affected. In the past decade, considerable efforts have focused on developing preventive measures, such as vaccines and pre-exposure prophylaxis, to combat sexually transmitted HIV. Recently, cytokine profiles have gained attention as potential predictors of HIV incidence due to their involvement in immune regulation and inflammation, presenting new opportunities to enhance preventative strategies. However, the high-dimensional, time-varying nature of cytokine data collected in clinical research, presents challenges for traditional statistical methods like the Cox proportional hazards (PH) model to effectively analyze survival data related to HIV. Machine learning (ML) survival models offer a robust alternative, especially for addressing the limitations of the PH model's assumptions. In this study, we applied survival support vector machine (SSVM) and random survival forest (RSF) models using changes or means in cytokine levels as predictors to assess their association with HIV incidence, evaluate variable importance, measure predictive accuracy using the concordance index (C-index) and integrated Brier score (IBS) and interpret the model's predictions using Shapley additive explanations (SHAP) values. Our results indicated that RSFs models outperformed SSVMs models, with the difference covariate model performing better than the mean covariate model. The highest C-index for SSVM was 0.7180 under the difference covariate model, while for RSF, it reached 0.8801 under the difference covariate model using the log-rank split rule. Key cytokines identified as positive predictors of HIV incidence included TNF-A, BASIC-FGF, IL-5, MCP-3, and EOTAXIN, while 29 cytokines were negative predictors. Baseline factors such as condom use frequency, treatment status, number of partners, and sexual activity also emerged as significant predictors. This study underscored the potential of cytokine profiles for predicting HIV incidence and highlighted the advantages of RSFs models in analyzing high-dimensional, time-varying data over SSVMs. It further through ablation studies emphasized the importance of selecting key features within mean and difference based covariate models to achieve an optimal balance between model complexity and predictive accuracy.
Collapse
Affiliation(s)
- Sarah Ogutu
- School of Mathematics, Statistics and Computer Science, University of KwaZulu-Natal, Pietermaritzburg, 3201, South Africa.
| | - Mohanad Mohammed
- School of Mathematics, Statistics and Computer Science, University of KwaZulu-Natal, Pietermaritzburg, 3201, South Africa
- School of Nursing and Public Health, University of KwaZulu-Natal, Pietermaritzburg, 3201, South Africa
| | - Henry Mwambi
- School of Mathematics, Statistics and Computer Science, University of KwaZulu-Natal, Pietermaritzburg, 3201, South Africa
| |
Collapse
|
4
|
Shuang Z, Xingyu X, Yue C, Mingjing Y. Explainable Machine Learning Predictions for the Benefit From Chemotherapy in Advanced Non-Small Cell Lung Cancer Without Available Targeted Mutations. THE CLINICAL RESPIRATORY JOURNAL 2024; 18:e70044. [PMID: 39696772 DOI: 10.1111/crj.70044] [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: 06/18/2024] [Revised: 10/16/2024] [Accepted: 12/08/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is a global health challenge. Chemotherapy remains the standard therapy for advanced NSCLC without mutations, but drug resistance often reduces effectiveness. Developing more effective methods to predict and monitor chemotherapy benefits early is crucial. METHODS We carried out a retrospective cohort study of NSCLC patients without targeted mutations who received chemotherapy at West China Hospital from 2009 to 2013. We identified variables associated with chemotherapy outcomes and built four predictive models by machine learning. Shapley additive explanations (SHAP) interpreted the best model's predictions. The Kaplan-Meier method assessed key variables' impact on 5-year overall survival. RESULTS The study enrolled 461 NSCLC patients. Eight variables were selected for the model: differentiation, surgery history, neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), total bilirubin (TBIL), total protein (TP), alanine aminotransferase (ALT), and lactate dehydrogenase (LDH). The extreme gradient boosting (Xgboost) model exhibited superior discriminatory ability in predicting complete response (CR) probabilities to chemotherapy, with an AUC of 0.78. SHAP plots showed surgery history and high differentiation were related to CR benefits from chemotherapy. Absence of surgery, higher NLR, higher PLR, and higher LDH were all independent prognostic factors for poor survivals in NSCLC patients without mutations receiving chemotherapy. CONCLUSIONS By machine learning, we developed a predictive model to assess chemotherapy benefits in NSCLC patients without targeted mutations, utilizing eight readily available and non-invasive clinical indicators. Demonstrating satisfactory predictive performance and clinical practicability, this model may help clinicians identify patients' tendency to benefit from chemotherapy, potentially improving their prognosis.
Collapse
Affiliation(s)
- Zhao Shuang
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiong Xingyu
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Cheng Yue
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yu Mingjing
- Department of Respiratory and Critical Care Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
5
|
Fan C, Li Y, Jiang A, Zhao R. Machine Learning-enhanced Signature of Metastasis-related T Cell Marker Genes for Predicting Overall Survival in Malignant Melanoma. J Immunother 2024:00002371-990000000-00125. [PMID: 39506915 DOI: 10.1097/cji.0000000000000544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/10/2024] [Indexed: 11/08/2024]
Abstract
In this study, we aimed to investigate disparities in the tumor immune microenvironment (TME) between primary and metastatic malignant melanoma (MM) using single-cell RNA sequencing (scRNA-seq) and to identify metastasis-related T cell marker genes (MRTMGs) for predicting patient survival using machine learning techniques. We identified 6 distinct T cell clusters in 10×scRNA-seq data utilizing the Uniform Manifold Approximation and Projection (UMAP) algorithm. Four machine learning algorithms highlighted SRGN, PMEL, GPR143, EIF4A2, and DSP as pivotal MRTMGs, forming the foundation of the MRTMGs signature. A high MRTMGs signature was found to be correlated with poorer overall survival (OS) and suppression of antitumor immunity in MM patients. We developed a nomogram that combines the MRTMGs signature with the T stage and N stage, which accurately predicts 1-year, 3-year, and 5-year OS probabilities. Furthermore, in an immunotherapy cohort, a high MRTMG signature was associated with an unfavorable response to anti-programmed death 1 (PD-1) therapy. In conclusion, primary and metastatic MM display distinct TME landscapes with different T cell subsets playing crucial roles in metastasis. The MRTMGs signature, established through machine learning, holds potential as a valuable biomarker for predicting the survival of MM patients and their response to anti-PD-1 therapy.
Collapse
Affiliation(s)
- Chaoxin Fan
- Department of Oncology, Xi'an People's Hospital (Xi'an Fourth Hospital)
| | - Yimeng Li
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi
| | - Aimin Jiang
- Shandong Provincial Key Laboratory of Precision Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences
| | - Rui Zhao
- Department of Clinical Nutrition, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
6
|
Dai Z, Chen C, Zhou Z, Zhou M, Xie Z, Liu Z, Liu S, Chen Y, Li J, Liu B, Shen J. Circulating Biomarkers Predict Immunotherapeutic Response in Hepatocellular Carcinoma Using a Machine Learning Method. J Hepatocell Carcinoma 2024; 11:2133-2144. [PMID: 39493265 PMCID: PMC11531708 DOI: 10.2147/jhc.s474593] [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: 06/04/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
Background Immune checkpoint inhibitor (ICI) therapy is a promising treatment for cancer. However, the response rate to ICI therapy in hepatocellular carcinoma (HCC) patients is low (approximately 30%). Thus, an approach to predict whether a patient will benefit from ICI therapy is required. This study aimed to design a classifier based on circulating indicators to identify patients suitable for ICI therapy. Methods This retrospective study included HCC patients who received immune checkpoint inhibitor therapy between March 2017 and September 2023 at Nanjing Drum Tower Hospital and Jinling Hospital. The levels of the 17 serum biomarkers and baseline patients' characters were assessed to discern meaningful circulating indicators related with survival benefits using random forest. A prognostic model was then constructed to predict survival of patients after treatment. Results A total of 369 patients (mean age 56, median follow-up duration 373 days,) were enrolled in this study. Among the 17 circulating biomarkers, 11 were carefully selected to construct a classifier. Receiver operating characteristic (ROC) analysis yielded an area under the curve (AUC) of 0.724. Notably, patients classified into the low-risk group exhibited a more positive prognosis (P = 0.0079; HR, 0.43; 95% CI 0.21-0.87). To enhance efficacy, we incorporated 11 clinical features. The extended model incorporated 12 circulating indicators and 5 clinical features. The AUC of the refined classifier improved to 0.752. Patients in the low-risk group demonstrated superior overall survival compared with those in the high-risk group (P = 0.026; HR 0.39; 95% CI 0.11-1.37). Conclusion Circulating biomarkers are useful in predicting therapeutic outcomes and can help in making clinical decisions regarding the use of ICI therapy.
Collapse
Affiliation(s)
- Zhiyan Dai
- Department of Precision Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Chao Chen
- Department of Oncology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Department of Oncology, Jinling Hospital, Clinical College of Nanjing Medical University, Nanjing, 21002, People’s Republic of China
| | - Ziyan Zhou
- Department of Precision Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Department of Oncology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Mingzhen Zhou
- Department of Precision Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Department of Oncology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Zhengyao Xie
- Department of Precision Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Ziyao Liu
- Department of Precision Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Siyuan Liu
- Department of Precision Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Yiqiang Chen
- Department of Precision Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Jingjing Li
- Department of Precision Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Baorui Liu
- Department of Oncology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| | - Jie Shen
- Department of Precision Medicine, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
- Department of Oncology, Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing, People’s Republic of China
| |
Collapse
|
7
|
Hirasawa Y, Kubota Y, Mura E, Suzuki R, Tsurui T, Iriguchi N, Ishiguro T, Ohkuma R, Shimokawa M, Ariizumi H, Horiike A, Wada S, Yamashita T, Ariyoshi T, Goto S, Otsuka K, Murakami M, Kiuchi Y, Yoshimura K, Tsunoda T. Chemotherapy combined with immune checkpoint inhibitors may overcome the detrimental effect of high neutrophil-to-lymphocyte ratio prior to treatment in esophageal cancer patients. Front Oncol 2024; 14:1449941. [PMID: 39464714 PMCID: PMC11502307 DOI: 10.3389/fonc.2024.1449941] [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: 06/16/2024] [Accepted: 09/16/2024] [Indexed: 10/29/2024] Open
Abstract
Introduction Immune checkpoint inhibitors (ICIs) have emerged as a promising treatment option for esophageal cancer (EC). Although ICIs enable long-term survival in some patients, the efficacy of ICIs varies widely among patients. Therefore, predictive biomarkers are necessary for identifying patients who are most likely to benefit from ICIs to improve the efficacy of the treatment. We retrospectively analyzed the outcomes of combination therapy, including nivolumab plus ipilimumab or chemotherapy plus anti-programmed cell death 1 (PD-1) antibodies in our institute to identify biomarkers. Methods Twenty-seven patients received nivolumab plus ipilimumab, and thirty-six patients received chemotherapy plus anti-PD-1 antibodies were included in this study. We analyzed patient characteristics, efficacy, and safety. Multivariable analysis of biomarkers evaluated the correlation among overall survival (OS), progression-free survival (PFS), and the following variables: body mass index, performance status, neutrophil-to-lymphocyte ratio (NLR), C-reactive protein level, and albumin level before treatment. Results In multivariable analysis, albumin level was significantly correlated with PFS in the cisplatin plus 5-fluorouracil (CF) plus pembrolizumab group. NLR and albumin level were significantly correlated with OS in the nivolumab plus ipilimumab group. Other variables, including PS, BMI, and CRP did not correlate with any of the outcomes. Conclusions High NLR in EC patients prior to treatment was significantly less effective for ICIs. In chemotherapy combined with ICIs, NLR before the treatment was not associated with treatment efficacy, suggesting combination chemotherapy may be beneficial for EC patients with high NLR. NLR may be an indicator of immunocompetence in anti-tumor immunity and a convenient predictive biomarker for selecting appropriate treatments including ICIs.
Collapse
Affiliation(s)
- Yuya Hirasawa
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- Division of Medical Pharmacology, Department of Pharmacology, Showa University School of Medicine, Tokyo, Japan
| | - Yutaro Kubota
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Emiko Mura
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Risako Suzuki
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- Division of Medical Pharmacology, Department of Pharmacology, Showa University School of Medicine, Tokyo, Japan
| | - Toshiaki Tsurui
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- Division of Medical Pharmacology, Department of Pharmacology, Showa University School of Medicine, Tokyo, Japan
- Department of Clinical Immuno-Oncology, Clinical Research Institute of Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
| | - Nana Iriguchi
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Tomoyuki Ishiguro
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Ryotaro Ohkuma
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Masahiro Shimokawa
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Hirotsugu Ariizumi
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Atsushi Horiike
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| | - Satoshi Wada
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- Department of Clinical Diagnostic Oncology, Clinical Research Institute of Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
| | - Takeshi Yamashita
- Showa University Hospital Esophageal Cancer Center, Esophageal Surgery, Tokyo, Japan
| | - Tomotake Ariyoshi
- Showa University Hospital Esophageal Cancer Center, Esophageal Surgery, Tokyo, Japan
| | - Satoru Goto
- Showa University Hospital Esophageal Cancer Center, Esophageal Surgery, Tokyo, Japan
| | - Koji Otsuka
- Showa University Hospital Esophageal Cancer Center, Esophageal Surgery, Tokyo, Japan
| | - Masahiko Murakami
- Showa University Hospital Esophageal Cancer Center, Esophageal Surgery, Tokyo, Japan
| | - Yuji Kiuchi
- Division of Medical Pharmacology, Department of Pharmacology, Showa University School of Medicine, Tokyo, Japan
- Pharmacological Research Center, Showa University, Tokyo, Japan
| | - Kiyoshi Yoshimura
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
- Department of Clinical Immuno-Oncology, Clinical Research Institute of Clinical Pharmacology and Therapeutics, Showa University, Tokyo, Japan
| | - Takuya Tsunoda
- Division of Medical Oncology, Department of Medicine, Showa University School of Medicine, Tokyo, Japan
| |
Collapse
|
8
|
Murata D, Azuma K, Murotani K, Kawahara A, Nishii Y, Tokito T, Sasada T, Hoshino T. Characterization of pre- and on-treatment soluble immune mediators and the tumor microenvironment in NSCLC patients receiving PD-1/L1 inhibitor monotherapy. Cancer Immunol Immunother 2024; 73:214. [PMID: 39235457 PMCID: PMC11377373 DOI: 10.1007/s00262-024-03781-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/14/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Despite the favorable therapeutic efficacy observed with ICI monotherapy, the majority of non-small cell lung cancer (NSCLC) patients do not respond. Therefore, identifying patients who could optimally benefit from ICI treatment remains a challenge. METHODS Among 183 patients with advanced or recurrent NSCLC who received ICI monotherapy, we analyzed 110 patients whose pre- and post-treatment plasma samples were available. Seventy-three soluble immune mediators were measured at ICI initiation and 6 weeks later. To identify useful biomarkers, we analyzed the association of pre-treatment levels and on-treatment changes of soluble immune mediators with survival of patients. The associations of pre-treatment or on-treatment biomarkers with irAE development, PD-L1 expression, CD8+ TIL density, and neutrophil to lymphocyte ratio (NLR) were also analyzed. RESULTS Univariate analysis showed that pre-treatment biomarkers included 6 immune mediators, whereas on-treatment biomarkers included 8 immune mediators. Multivariate analysis showed that pre-treatment biomarkers included 4 immune mediators (CCL19, CCL21, CXCL5, CXCL10), whereas on-treatment biomarkers included 5 immune mediators (CCL7, CCL19, CCL23, CCL25, IL-32). IrAE development was associated with on-treatment change in CCL23. PD-L1 expression was associated with the pre-treatment levels of TNFSF13B and the on-treatment change in CCL25. CD8+ TIL density was associated with the pre-treatment CXCL10 level, whereas NLR was correlated with pre-treatment levels of CCL13 and CCL17. CONCLUSION We identified several soluble immune mediators as pre-treatment and on-treatment biomarkers of survival in patients with NSCLC treated with ICI monotherapy. Some of these biomarkers were associated with other possible predictors, including irAE development, PD-L1 expression, CD8+ TIL density and NLR. Further large-scale studies are needed to establish biomarkers for patients with NSCLC who received ICI monotherapy.
Collapse
Affiliation(s)
- Daiki Murata
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, Fukuoka, 830-0011, Japan
| | - Koichi Azuma
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, Fukuoka, 830-0011, Japan.
| | - Kenta Murotani
- Biostatistics Center, Kurume University School of Medicine, Fukuoka, Japan
| | - Akihiko Kawahara
- Department of Diagnostic Pathology, Kurume University School of Medicine, Fukuoka, Japan
| | - Yuuya Nishii
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, Fukuoka, 830-0011, Japan
| | - Takaaki Tokito
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, Fukuoka, 830-0011, Japan
| | - Tetsuro Sasada
- Cancer Vaccine and Immunotherapy Center and Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Kanagawa, Japan
| | - Tomoaki Hoshino
- Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, 67 Asahi-Machi, Kurume, Fukuoka, 830-0011, Japan
| |
Collapse
|
9
|
Sun T, Liu J, Yuan H, Li X, Yan H. Construction of a risk prediction model for lung infection after chemotherapy in lung cancer patients based on the machine learning algorithm. Front Oncol 2024; 14:1403392. [PMID: 39184040 PMCID: PMC11341396 DOI: 10.3389/fonc.2024.1403392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/23/2024] [Indexed: 08/27/2024] Open
Abstract
Purpose The objective of this study was to create and validate a machine learning (ML)-based model for predicting the likelihood of lung infections following chemotherapy in patients with lung cancer. Methods A retrospective study was conducted on a cohort of 502 lung cancer patients undergoing chemotherapy. Data on age, Body Mass Index (BMI), underlying disease, chemotherapy cycle, number of hospitalizations, and various blood test results were collected from medical records. We used the Synthetic Minority Oversampling Technique (SMOTE) to handle unbalanced data. Feature screening was performed using the Boruta algorithm and The Least Absolute Shrinkage and Selection Operator (LASSO). Subsequently, six ML algorithms, namely Logistic Regression (LR), Random Forest (RF), Gaussian Naive Bayes (GNB), Multi-layer Perceptron (MLP), Support Vector Machine (SVM), and K-Nearest Neighbors (KNN) were employed to train and develop an ML model using a 10-fold cross-validation methodology. The model's performance was evaluated through various metrics, including the area under the receiver operating characteristic curve (ROC), accuracy, sensitivity, specificity, F1 score, calibration curve, decision curves, clinical impact curve, and confusion matrix. In addition, model interpretation was performed by the Shapley Additive Explanations (SHAP) analysis to clarify the importance of each feature of the model and its decision basis. Finally, we constructed nomograms to make the predictive model results more readable. Results The integration of Boruta and LASSO methodologies identified Gender, Smoke, Drink, Chemotherapy cycles, pleural effusion (PE), Neutrophil-lymphocyte count ratio (NLR), Neutrophil-monocyte count ratio (NMR), Lymphocytes (LYM) and Neutrophil (NEUT) as significant predictors. The LR model demonstrated superior performance compared to alternative ML algorithms, achieving an accuracy of 81.80%, a sensitivity of 81.1%, a specificity of 82.5%, an F1 score of 81.6%, and an AUC of 0.888(95%CI(0.863-0.911)). Furthermore, the SHAP method identified Chemotherapy cycles and Smoke as the primary decision factors influencing the ML model's predictions. Finally, this study successfully constructed interactive nomograms and dynamic nomograms. Conclusion The ML algorithm, combining demographic and clinical factors, accurately predicted post-chemotherapy lung infections in cancer patients. The LR model performed well, potentially improving early detection and treatment in clinical practice.
Collapse
Affiliation(s)
- Tao Sun
- Department of Hematology and Oncology Laboratory, The Central Hospital of Shaoyang, Shaoyang, Hunan, China
| | - Jun Liu
- Department of Scientific Research, The First Affiliated Hospital of Shaoyang University, Shaoyang, Hunan, China
| | - Houqin Yuan
- Department of Hematology and Oncology Laboratory, The Central Hospital of Shaoyang, Shaoyang, Hunan, China
| | - Xin Li
- Department of Hematology and Oncology Laboratory, The Central Hospital of Shaoyang, Shaoyang, Hunan, China
| | - Hui Yan
- Department of Hematology and Oncology Laboratory, The Central Hospital of Shaoyang, Shaoyang, Hunan, China
| |
Collapse
|
10
|
Yang W, Chen C, Ouyang Q, Han R, Sun P, Chen H. Machine learning models for predicting of PD-1 treatment efficacy in Pan-cancer patients based on routine hematologic and biochemical parameters. Cancer Cell Int 2024; 24:258. [PMID: 39034386 PMCID: PMC11265142 DOI: 10.1186/s12935-024-03439-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 07/08/2024] [Indexed: 07/23/2024] Open
Abstract
Immune checkpoint blockade therapy targeting the programmed death-1(PD-1) pathway has shown remarkable efficacy and durable response in patients with various cancer types. Early prediction of therapeutic efficacy is important for optimizing treatment plans and avoiding potential side effects. In this work, we developed an efficient machine learning prediction method using routine hematologic and biochemical parameters to predict the efficacy of PD-1 combination treatment in Pan-Cancer patients. A total of 431 patients with nasopharyngeal carcinoma, esophageal cancer and lung cancer who underwent PD-1 checkpoint inhibitor combination therapy were included in this study. Patients were divided into two groups: progressive disease (PD) and disease control (DC) groups. Hematologic and biochemical parameters were collected before and at the third week of PD-1 therapy. Six machine learning models were developed and trained to predict the efficacy of PD-1 combination therapy at 8-12 weeks. Analysis of 57 blood biomarkers before and after three weeks of PD-1 combination therapy through statistical analysis, heatmaps, and principal component analysis did not accurately predict treatment outcome. However, with machine learning models, both the AdaBoost classifier and GBDT demonstrated high levels of prediction efficiency, with clinically acceptable AUC values exceeding 0.7. The AdaBoost classifier exhibited the highest performance among the 6 machine learning models, with a sensitivity of 0.85 and a specificity of 0.79. Our study demonstrated the potential of machine learning to predict the efficacy of PD-1 combination therapy based on changes in hematologic and biochemical parameters.
Collapse
Affiliation(s)
- Wenjian Yang
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
- Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, 310024, China
| | - Cui Chen
- Department of Oncology, The First Affiliated Hospital, Sun Yat-Sen University, 58 Zhongshan Road II, Guangzhou, 510080, China
| | - Qiangqiang Ouyang
- College of Electronic Engineering, South China Agricultural University, Guangzhou, 510642, Guangdong, China
| | - Runkun Han
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Peng Sun
- Department of Medical Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| | - Hao Chen
- Department of Clinical Laboratory, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|
11
|
von Itzstein MS, Liu J, Mu-Mosley H, Fattah F, Park JY, SoRelle JA, Farrar JD, Gwin ME, Hsiehchen D, Gloria-McCutchen Y, Wakeland EK, Cole S, Bhalla S, Kainthla R, Puzanov I, Switzer B, Daniels GA, Zakharia Y, Shaheen M, Zhang J, Xie Y, Gerber DE. Differences in systemic immune parameters in individuals with lung cancer according to race. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.597754. [PMID: 38915535 PMCID: PMC11195092 DOI: 10.1101/2024.06.07.597754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Introduction Racial and ethnic disparities in the presentation and outcomes of lung cancer are widely known. To evaluate potential factors contributing to these observations, we measured systemic immune parameters in Black and White patients with lung cancer. Methods Patients scheduled to receive cancer immunotherapy were enrolled in a multi-institutional prospective biospecimen collection registry. Clinical and demographic information were obtained from electronic medical records. Pre-treatment peripheral blood samples were collected and analyzed for cytokines using a multiplex panel and for immune cell populations using mass cytometry. Differences between Black and White patients were determined and corrected for multiple comparisons. Results A total of 187 patients with non-small cell lung cancer (Black, 19; White, 168) were included in the analysis. There were no significant differences in baseline characteristics between Black and White patients. Compared to White patients, Black patients had significantly lower levels of CCL23 and CCL27, and significantly higher levels of CCL8, CXCL1, CCL26, CCL25, CCL1, IL-1 b, CXCL16, and IFN-γ (all P <0.05, FDR<0.1). Black patients also exhibited greater populations of non-classical CD16+ monocytes, NKT-like cells, CD4+ cells, CD38+ monocytes, and CD57+ gamma delta T cells (all P <0.05). Conclusions Black and White patients with lung cancer exhibit several differences in immune parameters, with Black patients exhibiting greater levels of numerous pro-inflammatory cytokines and cell populations. The etiology and clinical significance of these differences warrant further evaluation.
Collapse
|
12
|
Daye D, Parker R, Tripathi S, Cox M, Brito Orama S, Valentin L, Bridge CP, Uppot RN. CASCADE: Context-Aware Data-Driven AI for Streamlined Multidisciplinary Tumor Board Recommendations in Oncology. Cancers (Basel) 2024; 16:1975. [PMID: 38893096 PMCID: PMC11171258 DOI: 10.3390/cancers16111975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 05/18/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
This study addresses the potential of machine learning in predicting treatment recommendations for patients with hepatocellular carcinoma (HCC). Using an IRB-approved retrospective study of patients discussed at a multidisciplinary tumor board, clinical and imaging variables were extracted and used in a gradient-boosting machine learning algorithm, XGBoost. The algorithm's performance was assessed using confusion matrix metrics and the area under the Receiver Operating Characteristics (ROC) curve. The study included 140 patients (mean age 67.7 ± 8.9 years), and the algorithm was found to be predictive of all eight treatment recommendations made by the board. The model's predictions were more accurate than those based on published therapeutic guidelines by ESMO and NCCN. The study concludes that a machine learning model incorporating clinical and imaging variables can predict treatment recommendations made by an expert multidisciplinary tumor board, potentially aiding clinical decision-making in settings lacking subspecialty expertise.
Collapse
Affiliation(s)
- Dania Daye
- Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (M.C.); (L.V.); (C.P.B.); (R.N.U.)
- Harvard Medical School, Boston, MA 02115, USA;
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA
| | | | - Satvik Tripathi
- Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (M.C.); (L.V.); (C.P.B.); (R.N.U.)
- Harvard Medical School, Boston, MA 02115, USA;
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA
| | - Meredith Cox
- Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (M.C.); (L.V.); (C.P.B.); (R.N.U.)
- Harvard Medical School, Boston, MA 02115, USA;
| | | | - Leonardo Valentin
- Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (M.C.); (L.V.); (C.P.B.); (R.N.U.)
- Professional Hospital Guaynabo, Guaynabo 00971, Puerto Rico
| | - Christopher P. Bridge
- Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (M.C.); (L.V.); (C.P.B.); (R.N.U.)
- Harvard Medical School, Boston, MA 02115, USA;
- Athinoula A. Martinos Center for Biomedical Imaging, Charlestown, MA 02129, USA
| | - Raul N. Uppot
- Massachusetts General Hospital, Boston, MA 02114, USA; (S.T.); (M.C.); (L.V.); (C.P.B.); (R.N.U.)
- Harvard Medical School, Boston, MA 02115, USA;
| |
Collapse
|
13
|
Sinha T, Khan A, Awan M, Bokhari SFH, Ali K, Amir M, Jadhav AN, Bakht D, Puli ST, Burhanuddin M. Artificial Intelligence and Machine Learning in Predicting the Response to Immunotherapy in Non-small Cell Lung Carcinoma: A Systematic Review. Cureus 2024; 16:e61220. [PMID: 38939246 PMCID: PMC11210434 DOI: 10.7759/cureus.61220] [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] [Accepted: 05/27/2024] [Indexed: 06/29/2024] Open
Abstract
Non-small cell lung carcinoma (NSCLC) is a prevalent and aggressive form of lung cancer, with a poor prognosis for metastatic disease. Immunotherapy, particularly immune checkpoint inhibitors (ICIs), has revolutionized the management of NSCLC, but response rates are highly variable. Identifying reliable predictive biomarkers is crucial to optimize patient selection and treatment outcomes. This systematic review aimed to evaluate the current state of artificial intelligence (AI) and machine learning (ML) applications in predicting the response to immunotherapy in NSCLC. A comprehensive literature search identified 19 studies that met the inclusion criteria. The studies employed diverse AI/ML techniques, including deep learning, artificial neural networks, support vector machines, and gradient boosting methods, applied to various data modalities such as medical imaging, genomic data, clinical variables, and immunohistochemical markers. Several studies demonstrated the ability of AI/ML models to accurately predict immunotherapy response, progression-free survival, and overall survival in NSCLC patients. However, challenges remain in data availability, quality, and interpretability of these models. Efforts have been made to develop interpretable AI/ML techniques, but further research is needed to improve transparency and explainability. Additionally, translating AI/ML models from research settings to clinical practice poses challenges related to regulatory approval, data privacy, and integration into existing healthcare systems. Nonetheless, the successful implementation of AI/ML models could enable personalized treatment strategies, improve treatment outcomes, and reduce unnecessary toxicities and healthcare costs associated with ineffective treatments.
Collapse
Affiliation(s)
- Tanya Sinha
- Internal Medicine, Tribhuvan University, Kathmandu, NPL
| | - Aiman Khan
- Medicine, Liaquat College of Medicine and Dentistry, Karachi, PAK
| | - Manahil Awan
- General Practice, Liaquat National Hospital and Medical College, Karachi, PAK
| | | | - Khawar Ali
- Medicine and Surgery, King Edward Medical University, Lahore, PAK
| | - Maaz Amir
- Medicine and Surgery, King Edward Medical University, Lahore, PAK
| | - Aneesh N Jadhav
- Pediatrics, Bharat Ratna Dr. Babasaheb Ambedkar Memorial Hospital, Mumbai, IND
| | - Danyal Bakht
- Medicine and Surgery, Mayo Hospital, Lahore, PAK
| | - Sai Teja Puli
- Internal Medicine, Bhaskar Medical College, Hyderabad, IND
| | | |
Collapse
|
14
|
Augustin RC, Luke JJ. Rapidly Evolving Pre- and Post-surgical Systemic Treatment of Melanoma. Am J Clin Dermatol 2024; 25:421-434. [PMID: 38409643 PMCID: PMC11552441 DOI: 10.1007/s40257-024-00852-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/07/2024] [Indexed: 02/28/2024]
Abstract
With the development of effective BRAF-targeted and immune-checkpoint immunotherapies for metastatic melanoma, clinical trials are moving these treatments into earlier adjuvant and perioperative settings. BRAF-targeted therapy is a standard of care in resected stage III-IV melanoma, while anti-programmed death-1 (PD1) immunotherapy is now a standard of care option in resected stage IIB through IV disease. With both modalities, recurrence-free survival and distant-metastasis-free survival are improved by a relative 35-50%, yet no improvement in overall survival has been demonstrated. Neoadjuvant anti-PD1 therapy improves event-free survival by approximately an absolute 23%, although improvements in overall survival have yet to be demonstrated. Understanding which patients are most likely to recur and which are most likely to benefit from treatment is now the highest priority question in the field. Biomarker analyses, such as gene expression profiling of the primary lesion and circulating DNA, are preliminarily exciting as potential biomarkers, though each has drawbacks. As in the setting of metastatic disease, markers that inform positive outcomes include interferon-γ gene expression, PD-L1, and high tumor mutational burden, while negative predictors of outcome include circulating factors such as lactate dehydrogenase, interleukin-8, and C-reactive protein. Integrating and validating these markers into clinically relevant models is thus a high priority. Melanoma therapeutics continues to advance with combination adjuvant approaches now investigating anti-PD1 with lymphocyte activation gene 3 (LAG3), T-cell immunoreceptor with Ig and ITIM domains (TIGIT), and individualized neoantigen therapies. How this progress will be integrated into the management of a unique patient to reduce recurrence, limit toxicity, and avoid over-treatment will dominate clinical research and patient care over the next decade.
Collapse
Affiliation(s)
- Ryan C Augustin
- UPMC Hillman Cancer Center, 5150 Centre Ave. Room 1.27C, Pittsburgh, PA, 15232, USA
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Jason J Luke
- UPMC Hillman Cancer Center, 5150 Centre Ave. Room 1.27C, Pittsburgh, PA, 15232, USA.
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
15
|
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
|
16
|
Hong CS, Diergaarde B, Whiteside TL. Small extracellular vesicles in plasma carry luminal cytokines that remain undetectable by antibody-based assays in cancer patients and healthy donors. BJC REPORTS 2024; 2:16. [PMID: 38938748 PMCID: PMC11210721 DOI: 10.1038/s44276-024-00037-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 06/29/2024]
Abstract
Background Small (30-150nm) extracellular vesicles (sEV), also known as exosomes, play a key role in cell-to-cell signaling. They are produced by all cells, circulate freely and are present in all body fluids. Evidence indicates that cytokines are present on the surface and/or in the lumen of sEV. The contribution of intravesicular cytokines to cytokine levels in plasma are unknown. Methods sEV were isolated by ultrafiltration/size exclusion chromatography from pre-cleared plasma obtained from patients with head and neck squamous cell carcinoma (HNSCC) and healthy donors (HDs). Multiplex immunoassays were used to measure cytokine levels in paired untreated and detergent-treated (0.5% Triton X-100) plasma and plasma-derived detergent-treated sEV. Non-parametric tests were used to assess differences in cytokine levels. Results The presence of cytokines in sEV isolated from patients' and HDs' plasma was confirmed by immunoblots and on-bead flow cytometry. sEV-associated cytokines were functional in various in vitro assays. Levels of cytokines in sEV varied among the HNSCC patients and were generally significantly higher than the levels observed in sEV from HDs. Compared to untreated plasma, levels for the majority (40/51) of the evaluated proteins were significantly higher in detergent-treated plasma (P<0.0001-0.03). In addition, levels of 24/51 proteins in sEV, including IL6, TNFRII, IL-17a, IFNa and IFNg, were significantly positively correlated with the difference between levels detected in detergent-treated plasma and untreated plasma. Discussion The data indicate that sEV-associated cytokines account for the differences in cytokine levels measured in detergent-treated versus untreated plasma. Ab-based assays using untreated plasma detect only soluble cytokines and miss cytokines carried in the lumen of sEV. Permeabilization of sEV with a mild detergent allows for Ab-based detection of sEV-associated and soluble cytokines in plasma. The failure to detect cytokines carried in the sEV lumen leads to inaccurate estimates of cytokine levels in body fluids.
Collapse
Affiliation(s)
- Chang Sook Hong
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA 15213 USA
| | - Brenda Diergaarde
- Department of Human Genetics, School of Public Health, University of Pittsburgh and UPMC Hillman Cancer Center, Pittsburgh, PA 15213 USA
| | - Theresa L. Whiteside
- Departments of Pathology, Immunology and Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213 USA
| |
Collapse
|
17
|
Wei F, Sasada T. Circulating cytokine signatures as a soluble biomarker of immune checkpoint inhibitor therapy in non-small-cell lung cancer. Genes Immun 2024; 25:89-91. [PMID: 38097745 DOI: 10.1038/s41435-023-00236-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 02/18/2024]
Affiliation(s)
- Feifei Wei
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan.
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan.
| | - Tetsuro Sasada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan.
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center, Yokohama, Japan.
| |
Collapse
|
18
|
Lippenszky L, Mittendorf KF, Kiss Z, LeNoue-Newton ML, Napan-Molina P, Rahman P, Ye C, Laczi B, Csernai E, Jain NM, Holt ME, Maxwell CN, Ball M, Ma Y, Mitchell MB, Johnson DB, Smith DS, Park BH, Micheel CM, Fabbri D, Wolber J, Osterman TJ. Prediction of Effectiveness and Toxicities of Immune Checkpoint Inhibitors Using Real-World Patient Data. JCO Clin Cancer Inform 2024; 8:e2300207. [PMID: 38427922 PMCID: PMC10919473 DOI: 10.1200/cci.23.00207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/15/2023] [Accepted: 01/17/2024] [Indexed: 03/03/2024] Open
Abstract
PURPOSE Although immune checkpoint inhibitors (ICIs) have improved outcomes in certain patients with cancer, they can also cause life-threatening immunotoxicities. Predicting immunotoxicity risks alongside response could provide a personalized risk-benefit profile, inform therapeutic decision making, and improve clinical trial cohort selection. We aimed to build a machine learning (ML) framework using routine electronic health record (EHR) data to predict hepatitis, colitis, pneumonitis, and 1-year overall survival. METHODS Real-world EHR data of more than 2,200 patients treated with ICI through December 31, 2018, were used to develop predictive models. Using a prediction time point of ICI initiation, a 1-year prediction time window was applied to create binary labels for the four outcomes for each patient. Feature engineering involved aggregating laboratory measurements over appropriate time windows (60-365 days). Patients were randomly partitioned into training (80%) and test (20%) sets. Random forest classifiers were developed using a rigorous model development framework. RESULTS The patient cohort had a median age of 63 years and was 61.8% male. Patients predominantly had melanoma (37.8%), lung cancer (27.3%), or genitourinary cancer (16.4%). They were treated with PD-1 (60.4%), PD-L1 (9.0%), and CTLA-4 (19.7%) ICIs. Our models demonstrate reasonably strong performance, with AUCs of 0.739, 0.729, 0.755, and 0.752 for the pneumonitis, hepatitis, colitis, and 1-year overall survival models, respectively. Each model relies on an outcome-specific feature set, though some features are shared among models. CONCLUSION To our knowledge, this is the first ML solution that assesses individual ICI risk-benefit profiles based predominantly on routine structured EHR data. As such, use of our ML solution will not require additional data collection or documentation in the clinic.
Collapse
Affiliation(s)
- Levente Lippenszky
- Science and Technology Organization—Artificial Intelligence & Machine Learning, GE HealthCare, Budapest, Hungary/San Ramon, CA
| | | | - Zoltán Kiss
- Science and Technology Organization—Artificial Intelligence & Machine Learning, GE HealthCare, Budapest, Hungary/San Ramon, CA
| | - Michele L. LeNoue-Newton
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
| | - Pablo Napan-Molina
- Science and Technology Organization—Artificial Intelligence & Machine Learning, GE HealthCare, Budapest, Hungary/San Ramon, CA
| | - Protiva Rahman
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
- Health Outcomes and Biomedical Informatics, University of Florida, Tallahassee, FL
| | - Cheng Ye
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
| | - Balázs Laczi
- Science and Technology Organization—Artificial Intelligence & Machine Learning, GE HealthCare, Budapest, Hungary/San Ramon, CA
| | - Eszter Csernai
- Science and Technology Organization—Artificial Intelligence & Machine Learning, GE HealthCare, Budapest, Hungary/San Ramon, CA
| | - Neha M. Jain
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- OneOncology, Nashville, TN
| | - Marilyn E. Holt
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Sarah Cannon Research Institute, Nashville, TN
| | - Christina N. Maxwell
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | - Madeleine Ball
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt University School of Medicine, Nashville, TN
| | - Yufang Ma
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Pharmaceutical Services, Vanderbilt University Medical Center, Nashville, TN
| | - Margaret B. Mitchell
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Boston, MA
| | - Douglas B. Johnson
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - David S. Smith
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, TN
| | - Ben H. Park
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Christine M. Micheel
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Daniel Fabbri
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
| | - Jan Wolber
- Pharmaceutical Diagnostics, GE HealthCare, Chalfont St Giles, United Kingdom
| | - Travis J. Osterman
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
19
|
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
|