1
|
Zhang J, Song Z, Zhang Y, Zhang C, Xue Q, Zhang G, Tan F. Recent advances in biomarkers for predicting the efficacy of immunotherapy in non-small cell lung cancer. Front Immunol 2025; 16:1554871. [PMID: 40406096 PMCID: PMC12095235 DOI: 10.3389/fimmu.2025.1554871] [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: 01/03/2025] [Accepted: 04/18/2025] [Indexed: 05/26/2025] Open
Abstract
Lung cancer continues to be the primary cause of cancer-related deaths globally, with non-small cell lung cancer (NSCLC) accounting for approximately 85% of all instances. Recently, immune checkpoint inhibitors (ICIs) have transformed the treatment approach for NSCLC, however, only a subset of patients experiences significant benefits. Therefore, identifying reliable biomarkers to forecast the efficacy of ICIs is crucial for ensuring the safety and effectiveness of treatments, becoming a major focus of current research efforts. This review highlights the recent advances in predictive biomarkers for the efficacy of ICIs in the treatment of NSCLC, including PD-L1 expression, tertiary lymphoid structures (TLS), tumor-infiltrating lymphocytes (TILs), tumor genomic alterations, transcriptional signatures, circulating biomarkers, and the microbiome. Furthermore, it underscores the pivotal roles of liquid biopsy, sequencing technologies, and digital pathology in biomarker discovery. Special attention is given to the predictive value of TLS, circulating biomarkers, and transcriptional signatures. The review concludes that the integration of multiple biomarkers holds promise for achieving more accurate efficacy predictions and optimizing personalized immunotherapy strategies. By providing a comprehensive overview of the current progress, this review offers valuable insights into biomarker-based precision medicine for NSCLC and outlines future research directions.
Collapse
Affiliation(s)
- Jiacheng Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zehao Song
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuanjie Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Chentong Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guochao Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
2
|
Han X, Guan J, Guo L, Jiao Q, Wang K, Hou F, Liu S, Yang S, Huang C, Cong W, Wang H. A CT-based interpretable deep learning signature for predicting PD-L1 expression in bladder cancer: a two-center study. Cancer Imaging 2025; 25:27. [PMID: 40065444 PMCID: PMC11892212 DOI: 10.1186/s40644-025-00849-1] [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: 01/12/2025] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND To construct and assess a deep learning (DL) signature that employs computed tomography imaging to predict the expression status of programmed cell death ligand 1 in patients with bladder cancer (BCa). METHODS This retrospective study included 190 patients from two hospitals who underwent surgical removal of BCa (training set/external validation set, 127/63). We used convolutional neural network and radiomics machine learning technology to generate prediction models. We then compared the performance of the DL signature with the radiomics machine learning signature and selected the optimal signature to build a nomogram with the clinical model. Finally, the internal forecasting process of the DL signature was explained using Shapley additive explanation technology. RESULTS On the external validation set, the DL signature had an area under the curve of 0.857 (95% confidence interval: 0.745-0.932), and demonstrated superior prediction performance in comparison with the other models. SHAP expression images revealed that the prediction of PD-L1 expression status is mainly influenced by the tumor edge region, particularly the area close to the bladder wall. CONCLUSIONS The DL signature performed well in comparison with other models and proved to be a valuable, dependable, and interpretable tool for predicting programmed cell death ligand 1 expression status in patients with BCa.
Collapse
Affiliation(s)
- Xiaomeng Han
- Department of Radiology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong, 266003, China
| | - Jing Guan
- Department of Radiology, The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei, 050000, China
| | - Li Guo
- College of Computer Science and Technology, Qingdao University, Qingdao, Shandong, 266071, China
| | - Qiyan Jiao
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Kexin Wang
- Qingdao Medical College, Qingdao University, Qingdao, Shandong, 266071, China
| | - Feng Hou
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, 266003, China
| | - Shunli Liu
- Department of Radiology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong, 266003, China
| | - Shifeng Yang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250022, China
| | - Chencui Huang
- Department of Research Collaboration, R&d Center, Beijing Deepwise & League of PHD Technology Co., Ltd, Beijing, 100080, China
| | - Wenbin Cong
- Department of Radiology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong, 266003, China.
| | - Hexiang Wang
- Department of Radiology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong, 266003, China.
| |
Collapse
|
3
|
Mussa A, Ismail NH, Hamid M, Al-Hatamleh MAI, Bragoli A, Hajissa K, Mokhtar NF, Mohamud R, Uskoković V, Hassan R. Understanding the role of TNFR2 signaling in the tumor microenvironment of breast cancer. J Exp Clin Cancer Res 2024; 43:312. [PMID: 39609700 PMCID: PMC11603874 DOI: 10.1186/s13046-024-03218-1] [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: 08/21/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024] Open
Abstract
Breast cancer (BC) is the most frequently diagnosed malignancy among women. It is characterized by a high level of heterogeneity that emerges from the interaction of several cellular and soluble components in the tumor microenvironment (TME), such as cytokines, tumor cells and tumor-associated immune cells. Tumor necrosis factor (TNF) receptor 2 (TNFR2) appears to play a significant role in microenvironmental regulation, tumor progression, immune evasion, drug resistance, and metastasis of many types of cancer, including BC. However, the significance of TNFR2 in BC biology is not fully understood. This review provides an overview of TNFR2 biology, detailing its activation and its interactions with important signaling pathways in the TME (e.g., NF-κB, MAPK, and PI3K/Akt pathways). We discuss potential therapeutic strategies targeting TNFR2, with the aim of enhancing the antitumor immune response to BC. This review provides insights into role of TNFR2 as a major immune checkpoint for the future treatment of patients with BC.
Collapse
Affiliation(s)
- Ali Mussa
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
- Department of Biology, Faculty of Education, Omdurman Islamic University, P.O. Box 382, Omdurman, Sudan
| | - Nor Hayati Ismail
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
| | - Mahasin Hamid
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Hunan Province, Changsha, 410013, China
- Department of Zoology, Faculty of Sciences and Information Technology, University of Nyala, Nyala, 63311, Sudan
| | - Mohammad A I Al-Hatamleh
- Division of Hematology and Oncology, Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Anthony Bragoli
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Khalid Hajissa
- Department of Zoology, Faculty of Science and Technology, Omdurman Islamic University, P.O. Box 382, Omdurman, Sudan
| | - Noor Fatmawati Mokhtar
- Institute for Research in Molecular Medicine (iNFORMM), Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia.
| | - Vuk Uskoković
- TardigradeNano LLC, Irvine, CA, 92604, USA
- Division of Natural Sciences, Fullerton College, Fullerton, CA, 92832, USA
| | - Rosline Hassan
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu , Kelantan, 16150, Malaysia.
| |
Collapse
|
4
|
Cao Y, Zhu H, Li Z, Liu C, Ye J. CT Image-Based Radiomic Analysis for Detecting PD-L1 Expression Status in Bladder Cancer Patients. Acad Radiol 2024; 31:3678-3687. [PMID: 38556431 DOI: 10.1016/j.acra.2024.02.047] [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: 11/01/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 04/02/2024]
Abstract
RATIONALE AND OBJECTIVES The role of Programmed death-ligand 1 (PD-L1) expression is crucial in guiding immunotherapy selection. This study aims to develop and evaluate a radiomic model, leveraging Computed Tomography (CT) imaging, with the objective of predicting PD-L1 expression status in patients afflicted with bladder cancer. MATERIALS AND METHODS The study encompassed 183 subjects diagnosed with histologically confirmed bladder cancer, among which the PD-L1(+) cohort constituted 60.1% of the total population. Stratified random sampling was utilized at a 7:3 ratio. We employed five diverse machine learning algorithms-Decision Tree, Random Forest, Linear Support Vector Classification, Support Vector Machine, and Logistic Regression-to establish radiomic models on the training dataset. These models endeavored to predict PD-L1 expression status premised on radiomic features derived from region-of-interest segmentation. Subsequent to this, the predictive performance of these models was examined on a validation set employing the receiver operating characteristic (ROC) curve. The DeLong test was utilized to contrast ROC curves, thereby pinpointing the model with superior predictive accuracy. RESULTS 16 features were chosen for the model construction. All five models revealed strong performance in the training set (AUC, 0.920-1) and commendable predictive ability in the validation set (AUC, 0.753-0.766). As per the DeLong test, no statistically significant disparities were observed among any of the models (P > 0.05) in the validation set. Additional verification through the calibration curve and decision curve analysis indicated that the Logistic Regression model exhibited extraordinary precision and practicality. CONCLUSION Our machine learning model, grounded on radiomic features, demonstrated its proficiency in accurately distinguishing bladder cancer patients with high PD-L1 expression. Future research, incorporating more exhaustive datasets, could potentially augment the predictive efficiency of radiomic algorithms, thereby advancing their clinical utility.
Collapse
Affiliation(s)
- Ying Cao
- Department of Radiotherapy, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou 215028, China
| | - Hongyu Zhu
- Department of Radiotherapy, The Affiliated Suzhou Hospital of Nanjing University Medical School, Suzhou 215153, China
| | - Zhenkai Li
- Department of Radiology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou 215028, China
| | - Canyu Liu
- Department of Radiotherapy, Dushu Lake Hospital Affiliated to Soochow University, Suzhou 215127, China
| | - Juan Ye
- Department of Radiology, Suzhou Kowloon Hospital, Shanghai Jiaotong University School of Medicine, Suzhou 215028, China.
| |
Collapse
|
5
|
Chen J, Amoozgar Z, Liu X, Aoki S, Liu Z, Shin SM, Matsui A, Hernandez A, Pu Z, Halvorsen S, Lei PJ, Datta M, Zhu L, Ruan Z, Shi L, Staiculescu D, Inoue K, Munn LL, Fukumura D, Huang P, Sassi S, Bardeesy N, Ho WJ, Jain RK, Duda DG. Reprogramming the Intrahepatic Cholangiocarcinoma Immune Microenvironment by Chemotherapy and CTLA-4 Blockade Enhances Anti-PD-1 Therapy. Cancer Immunol Res 2024; 12:400-412. [PMID: 38260999 PMCID: PMC10985468 DOI: 10.1158/2326-6066.cir-23-0486] [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: 06/11/2023] [Revised: 11/05/2023] [Accepted: 01/19/2024] [Indexed: 01/24/2024]
Abstract
Intrahepatic cholangiocarcinoma (ICC) has limited therapeutic options and a dismal prognosis. Adding blockade of the anti-programmed cell death protein (PD)-1 pathway to gemcitabine/cisplatin chemotherapy has recently shown efficacy in biliary tract cancers but with low response rates. Here, we studied the effects of anti-cytotoxic T lymphocyte antigen (CTLA)-4 when combined with anti-PD-1 and gemcitabine/cisplatin in orthotopic murine models of ICC. This combination therapy led to substantial survival benefits and reduction of morbidity in two aggressive ICC models that were resistant to immunotherapy alone. Gemcitabine/cisplatin treatment increased tumor-infiltrating lymphocytes and normalized the ICC vessels and, when combined with dual CTLA-4/PD-1 blockade, increased the number of activated CD8+Cxcr3+IFNγ+ T cells. CD8+ T cells were necessary for the therapeutic benefit because the efficacy was compromised when CD8+ T cells were depleted. Expression of Cxcr3 on CD8+ T cells is necessary and sufficient because CD8+ T cells from Cxcr3+/+ but not Cxcr3-/- mice rescued efficacy in T cell‒deficient mice. Finally, rational scheduling of anti-CTLA-4 "priming" with chemotherapy followed by anti-PD-1 therapy achieved equivalent efficacy with reduced overall drug exposure. These data suggest that this combination approach should be clinically tested to overcome resistance to current therapies in ICC patients.
Collapse
Affiliation(s)
- Jiang Chen
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of General Surgery, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Zohreh Amoozgar
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Immuno-oncology Research and Development, Sanofi, Cambridge, Massachusetts
| | - Xin Liu
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shuichi Aoki
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Surgery, Tohoku Graduate School of Medicine, Sendai, Japan
| | - Zelong Liu
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Sarah M. Shin
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Aya Matsui
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Kanazawa University Institute of Medical, Pharmaceutical and Health Sciences Faculty of Medicine, Kanazawa, Japan
| | - Alexei Hernandez
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Zhangya Pu
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Xiangya Hospital, Central South University, Changsha, China
| | - Stefan Halvorsen
- Center of Computational and Integrative Biology (CCIB), Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Pin-Ji Lei
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Meenal Datta
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Aerospace and Mechanical Engineering, College of Engineering, University of Notre Dame, Notre Dame, Indiana
| | - Lingling Zhu
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- West China Hospital of Sichuan University, Chengdu, China
| | - Zhiping Ruan
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Jiaotong University, Xi'an, China
| | - Lei Shi
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Daniel Staiculescu
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Koetsu Inoue
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Surgery, Tohoku Graduate School of Medicine, Sendai, Japan
| | - Lance L. Munn
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dai Fukumura
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Peigen Huang
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Slim Sassi
- Center of Computational and Integrative Biology (CCIB), Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Department of Orthopedics, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Nabeel Bardeesy
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Won Jin Ho
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Rakesh K. Jain
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Dan G. Duda
- Edwin L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
6
|
Tostes K, Siqueira AP, Reis RM, Leal LF, Arantes LMRB. Biomarkers for Immune Checkpoint Inhibitor Response in NSCLC: Current Developments and Applicability. Int J Mol Sci 2023; 24:11887. [PMID: 37569262 PMCID: PMC10418476 DOI: 10.3390/ijms241511887] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 08/13/2023] Open
Abstract
Lung cancer has the highest mortality rate among all cancer types, resulting in over 1.8 million deaths annually. Immunotherapy utilizing immune checkpoint inhibitors (ICIs) has revolutionized the treatment of non-small cell lung cancer (NSCLC). ICIs, predominantly monoclonal antibodies, modulate co-stimulatory and co-inhibitory signals crucial for maintaining immune tolerance. Despite significant therapeutic advancements in NSCLC, patients still face challenges such as disease progression, recurrence, and high mortality rates. Therefore, there is a need for predictive biomarkers that can guide lung cancer treatment strategies. Currently, programmed death-ligand 1 (PD-L1) expression is the only established biomarker for predicting ICI response. However, its accuracy and robustness are not consistently reliable. This review provides an overview of potential biomarkers currently under development or in the validation stage that hold promise in improving the classification of responders and non-responders to ICI therapy in the near future.
Collapse
Affiliation(s)
- Katiane Tostes
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil; (K.T.)
| | - Aléxia Polo Siqueira
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil; (K.T.)
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil; (K.T.)
- Life and Health Sciences Research Institute (ICVS), Medical School, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s-PT Government Associate Laboratory, 4806-909 Guimarães, Portugal
| | - Leticia Ferro Leal
- Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil; (K.T.)
- Barretos School of Health Sciences, Dr. Paulo Prata-FACISB, Barretos 14785-002, São Paulo, Brazil
| | | |
Collapse
|
7
|
Manapov F, Nieto A, Käsmann L, Taugner J, Kenndoff S, Flörsch B, Guggenberger J, Hofstetter K, Kröninger S, Lehmann J, Kravutske H, Pelikan C, Belka C, Eze C. Five years after PACIFIC: update on multimodal treatment efficacy based on real-world reports. Expert Opin Investig Drugs 2023; 32:187-200. [PMID: 36780358 DOI: 10.1080/13543784.2023.2179479] [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: 04/03/2022] [Revised: 01/19/2023] [Accepted: 02/08/2023] [Indexed: 02/14/2023]
Abstract
INTRODUCTION The growing body of real-life data on maintenance treatment with durvalumab suggests that immunological markers of the cancer host interplay may have significant effects on the efficacy of multimodal therapy in patients with unresectable stage III NSCLC. AREAS COVERED We summarize real-world clinical data regarding this new tri-modal approach and report on potential biomarker landscape. EXPERT OPINION The obvious question posed in this context of a very heterogeneous inoperable stage III NSCLC disease is: How can we augment an ability to predict checkpoint inhibition success or failure? Which tools and biomarkers, which clinical metadata and genetic background are relevant and feasible? No single biomarker will ever fully dominate the unresectable stage III NSCLC space, so we advocate multilevel and multivariate analysis of biomarkers. In this particular opinion piece, we explore the impact of PD-L1 expression on tumor cells, neutrophil-to-lymphocyte ratio, EGFR and STK11 mutational status, interferon-gamma signature, and tumor-infiltrating lymphocytes among others.
Collapse
Affiliation(s)
- Farkhad Manapov
- Department of Radiotherapy and Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Alexander Nieto
- Department of Radiotherapy and Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Lukas Käsmann
- Department of Radiotherapy and Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Julian Taugner
- Department of Radiotherapy and Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Saskia Kenndoff
- Department of Radiotherapy and Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Benedikt Flörsch
- Department of Radiotherapy and Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Julian Guggenberger
- Department of Radiotherapy and Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Kerstin Hofstetter
- Department of Radiotherapy and Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Sophie Kröninger
- Department of Radiotherapy and Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Janina Lehmann
- Department of Radiotherapy and Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Helene Kravutske
- Department of Radiotherapy and Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| | - Carolyn Pelikan
- Helmholtz Zentrum München, Immunoanalytics - Tissue Control of Immunocytes, Munich, Germany
| | - Claus Belka
- Department of Radiotherapy and Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
- Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for Lung Research (DZL), Munich, Germany
- German Cancer Consortium (DKTK), partner site Munich, Munich, Germany
| | - Chukwuka Eze
- Department of Radiotherapy and Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
8
|
Chen J, Amoozgar Z, Liu X, Aoki S, Liu Z, Shin S, Matsui A, Pu Z, Lei PJ, Datta M, Zhu L, Ruan Z, Shi L, Staiculescu D, Inoue K, Munn LL, Fukumura D, Huang P, Bardeesy N, Ho WJ, Jain RK, Duda DG. Reprogramming Intrahepatic Cholangiocarcinoma Immune Microenvironment by Chemotherapy and CTLA-4 Blockade Enhances Anti-PD1 Therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.26.525680. [PMID: 36747853 PMCID: PMC9901023 DOI: 10.1101/2023.01.26.525680] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Intrahepatic cholangiocarcinoma (ICC) has limited therapeutic options and a dismal prognosis. Anti-PD-L1 immunotherapy combined with gemcitabine/cisplatin chemotherapy has recently shown efficacy in biliary tract cancers, but responses are seen only in a minority of patients. Here, we studied the roles of anti-PD1 and anti-CTLA-4 immune checkpoint blockade (ICB) therapies when combined with gemcitabine/cisplatin and the mechanisms of treatment benefit in orthotopic murine ICC models. We evaluated the effects of the combined treatments on ICC vasculature and immune microenvironment using flow cytometry analysis, immunofluorescence, imaging mass cytometry, RNA-sequencing, qPCR, and in vivo T-cell depletion and CD8+ T-cell transfer using orthotopic ICC models and transgenic mice. Combining gemcitabine/cisplatin with anti-PD1 and anti-CTLA-4 antibodies led to substantial survival benefits and reduction of morbidity in two aggressive ICC models, which were ICB-resistant. Gemcitabine/cisplatin treatment increased the frequency of tumor-infiltrating lymphocytes and normalized the ICC vessels, and when combined with dual CTLA-4/PD1 blockade, increased the number of activated CD8+Cxcr3+IFN-γ+ T-cells. Depletion of CD8+ but not CD4+ T-cells compromised efficacy. Conversely, CD8+ T-cell transfer from Cxcr3-/- versus Cxcr3+/+ mice into Rag1-/- immunodeficient mice restored the anti-tumor effect of gemcitabine/cisplatin/ICB combination therapy. Finally, rational scheduling of the ICBs (anti-CTLA-4 "priming") with chemotherapy and anti-PD1 therapy achieved equivalent efficacy with continuous dosing while reducing overall drug exposure. In summary, gemcitabine/cisplatin chemotherapy normalizes vessel structure, increases activated T-cell infiltration, and enhances anti-PD1/CTLA-4 immunotherapy efficacy in aggressive murine ICC. This combination approach should be clinically tested to overcome resistance to current therapies in ICC patients.
Collapse
Affiliation(s)
- Jiang Chen
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| | - Zohreh Amoozgar
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| | - Xin Liu
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School; 185 Cambridge Street, Simches Building, CPZN-4216, Boston, MA 02114, USA
| | - Shuichi Aoki
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| | - Zelong Liu
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| | - Sarah Shin
- Department of Medicine, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, 401 N. Broadway, Baltimore, MD 21231, USA
| | - Aya Matsui
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| | - Zhangya Pu
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| | - Pin-Ji Lei
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| | - Meenal Datta
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| | - Lingling Zhu
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| | - Zhiping Ruan
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| | - Lei Shi
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School; 185 Cambridge Street, Simches Building, CPZN-4216, Boston, MA 02114, USA
| | - Daniel Staiculescu
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| | - Koetsu Inoue
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| | - Lance L. Munn
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| | - Dai Fukumura
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| | - Peigen Huang
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| | - Nabeel Bardeesy
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School; 185 Cambridge Street, Simches Building, CPZN-4216, Boston, MA 02114, USA
| | - Won Jin Ho
- Department of Medicine, Johns Hopkins Sidney Kimmel Comprehensive Cancer Center, 401 N. Broadway, Baltimore, MD 21231, USA
| | - Rakesh. K. Jain
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| | - Dan G. Duda
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School; 100 Blossom Street, Cox-734, MA 02114, USA
| |
Collapse
|
9
|
Li J, Zhang Y, Liu Z, Han X. Editorial: Application of artificial intelligence in improving immunotherapeutic efficacy. Front Pharmacol 2022; 13:1100837. [PMID: 36582534 PMCID: PMC9793071 DOI: 10.3389/fphar.2022.1100837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Affiliation(s)
- Jie Li
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China,Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China,*Correspondence: Zaoqu Liu, ; Xinwei Han,
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China,Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China,Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China,*Correspondence: Zaoqu Liu, ; Xinwei Han,
| |
Collapse
|
10
|
Bruns IB, Beltman JB. Quantifying the contribution of transcription factor activity, mutations and microRNAs to CD274 expression in cancer patients. Sci Rep 2022; 12:4374. [PMID: 35289334 PMCID: PMC8921511 DOI: 10.1038/s41598-022-08356-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 03/03/2022] [Indexed: 12/15/2022] Open
Abstract
Immune checkpoint inhibitors targeting the programmed cell death protein 1 (PD-1)/programmed cell death protein ligand 1 (PD-L1) axis have been remarkably successful in inducing tumor remissions in several human cancers, yet a substantial number of patients do not respond to treatment. Because this may be partially due to the mechanisms giving rise to high PD-L1 expression within a patient, it is highly relevant to fully understand these mechanisms. In this study, we conduct a bioinformatic analysis to quantify the relative importance of transcription factor (TF) activity, microRNAs (miRNAs) and mutations in determining PD-L1 (CD274) expression at mRNA level based on data from the Cancer Genome Atlas. To predict individual CD274 levels based on TF activity, we developed multiple linear regression models by taking the expression of target genes of the TFs known to directly target PD-L1 as independent variables. This analysis showed that IRF1, STAT1, NFKB and BRD4 are the most important regulators of CD274 expression, explaining its mRNA levels in 90–98% of the patients. Because the remaining patients had high CD274 levels independent of these TFs, we next investigated whether mutations associated with increased CD274 mRNA levels, and low levels of miRNAs associated with negative regulation of CD274 expression could cause high CD274 levels in these patients. We found that mutations or miRNAs offered an explanation for high CD274 levels in 81–100% of the underpredicted patients. Thus, CD274 expression is largely explained by TF activity, and the remaining unexplained cases can largely be explained by mutations or low miRNA abundance.
Collapse
Affiliation(s)
- Imke B Bruns
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Joost B Beltman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands.
| |
Collapse
|
11
|
Chiu LC, Lin SM, Lo YL, Kuo SCH, Yang CT, Hsu PC. Immunotherapy and Vaccination in Surgically Resectable Non-Small Cell Lung Cancer (NSCLC). Vaccines (Basel) 2021; 9:689. [PMID: 34201650 PMCID: PMC8310081 DOI: 10.3390/vaccines9070689] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 06/10/2021] [Accepted: 06/22/2021] [Indexed: 12/15/2022] Open
Abstract
Early-stage NSCLC (stages I and II, and some IIIA diseases) accounts for approximately 30% of non-small cell lung cancer (NSCLC) cases, with surgery being its main treatment modality. The risk of disease recurrence and cancer-related death, however, remains high among NSCLC patients after complete surgical resection. In previous studies on the long-term follow-up of post-operative NSCLC, the results showed that the five-year survival rate was about 65% for stage IB and about 35% for stage IIIA diseases. Platinum-based chemotherapy with or without radiation therapy has been used as a neoadjuvant therapy or post-operative adjuvant therapy in NSCLC, but the improvement of survival is limited. Immune checkpoint inhibitors (ICIs) have effectively improved the 5-year survival of advanced NSCLC patients. Cancer vaccination has also been explored and used in the prevention of cancer or reducing disease recurrence in resected NSCLC. Here, we review studies that have focused on the use of immunotherapies (i.e., ICIs and vaccination) in surgically resectable NSCLC. We present the results of completed clinical trials that have used ICIs as neoadjuvant therapies in pre-operative NSCLC. Ongoing clinical trials investigating ICIs as neoadjuvant and adjuvant therapies are also summarized.
Collapse
Affiliation(s)
- Li-Chung Chiu
- Division of Thoracic Medicine, Department of Internal Medicine, College of Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (L.-C.C.); (S.-M.L.); (Y.-L.L.); (S.C.-H.K.); (C.-T.Y.)
- Department of Thoracic Medicine, New Taipei Municipal Tu Cheng Hospital, New Taipei City 23652, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Shu-Min Lin
- Division of Thoracic Medicine, Department of Internal Medicine, College of Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (L.-C.C.); (S.-M.L.); (Y.-L.L.); (S.C.-H.K.); (C.-T.Y.)
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Yu-Lun Lo
- Division of Thoracic Medicine, Department of Internal Medicine, College of Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (L.-C.C.); (S.-M.L.); (Y.-L.L.); (S.C.-H.K.); (C.-T.Y.)
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Scott Chih-Hsi Kuo
- Division of Thoracic Medicine, Department of Internal Medicine, College of Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (L.-C.C.); (S.-M.L.); (Y.-L.L.); (S.C.-H.K.); (C.-T.Y.)
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Cheng-Ta Yang
- Division of Thoracic Medicine, Department of Internal Medicine, College of Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (L.-C.C.); (S.-M.L.); (Y.-L.L.); (S.C.-H.K.); (C.-T.Y.)
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
- Department of Internal Medicine, Taoyuan Chang Gung Memorial Hospital, Taoyuan City 33378, Taiwan
- Department of Respiratory Therapy, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| | - Ping-Chih Hsu
- Division of Thoracic Medicine, Department of Internal Medicine, College of Medicine, Chang Gung Memorial Hospital at Linkou, Taoyuan City 33305, Taiwan; (L.-C.C.); (S.-M.L.); (Y.-L.L.); (S.C.-H.K.); (C.-T.Y.)
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan City 33302, Taiwan
| |
Collapse
|
12
|
Siewe N, Friedman A. TGF-β inhibition can overcome cancer primary resistance to PD-1 blockade: A mathematical model. PLoS One 2021; 16:e0252620. [PMID: 34061898 PMCID: PMC8168900 DOI: 10.1371/journal.pone.0252620] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
Immune checkpoint inhibitors have demonstrated, over the recent years, impressive clinical response in cancer patients, but some patients do not respond at all to checkpoint blockade, exhibiting primary resistance. Primary resistance to PD-1 blockade is reported to occur under conditions of immunosuppressive tumor environment, a condition caused by myeloid derived suppressor cells (MDSCs), and by T cells exclusion, due to increased level of T regulatory cells (Tregs). Since TGF-β activates Tregs, TGF-β inhibitor may overcome primary resistance to anti-PD-1. Indeed, recent mice experiments show that combining anti-PD-1 with anti-TGF-β yields significant therapeutic improvements compared to anti-TGF-β alone. The present paper introduces two cancer-specific parameters and, correspondingly, develops a mathematical model which explains how primary resistance to PD-1 blockade occurs, in terms of the two cancer-specific parameters, and how, in combination with anti-TGF-β, anti-PD-1 provides significant benefits. The model is represented by a system of partial differential equations and the simulations are in agreement with the recent mice experiments. In some cancer patients, treatment with anti-PD-1 results in rapid progression of the disease, known as hyperprogression disease (HPD). The mathematical model can also explain how this situation arises, and it predicts that HPD may be reversed by combining anti-TGF-β to anti-PD-1. The model is used to demonstrate how the two cancer-specific parameters may serve as biomarkers in predicting the efficacy of combination therapy with PD-1 and TGF-β inhibitors.
Collapse
Affiliation(s)
- Nourridine Siewe
- School of Mathematical Sciences, College of Science, Rochester Institute of Technology, Rochester, New York, United States of America
| | - Avner Friedman
- Department of Mathematics, Mathematical Biosciences Institute, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
13
|
Cameselle-García S, Abdulkader-Sande S, Sánchez-Ares M, Rodríguez-Carnero G, Garcia-Gómez J, Gude-Sampedro F, Abdulkader-Nallib I, Cameselle-Teijeiro JM. PD-L1 expression and immune cells in anaplastic carcinoma and poorly differentiated carcinoma of the human thyroid gland: A retrospective study. Oncol Lett 2021; 22:553. [PMID: 34093774 PMCID: PMC8170268 DOI: 10.3892/ol.2021.12814] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 05/04/2021] [Indexed: 02/07/2023] Open
Abstract
Anaplastic thyroid carcinoma (ATC) and poorly differentiated thyroid carcinoma (PDTC) have limited treatment options, and immune profiling may help select patients for immunotherapy. The prevalence and relevance of programmed death-1 ligand (PD-L1) expression and the presence of immune cells in ATC and PDTC has not yet been well established. The present study investigated PD-L1 expression (clone 22C3) and cells in the tumor microenvironment (TME), including tumor-infiltrating lymphocytes (TILs), tumor-associated macrophages (TAMs) and dendritic cells, in whole tissue sections of 15 cases of ATC and 13 cases of PDTC. Immunohistochemical PD-L1 expression using a tumor proportion score (TPS) with a 1% cut-off was detected in 9/15 (60%) of ATC cases and 1/13 (7.7%) of PDTC cases (P=0.006). PD-L1 expression in TILs was limited to the ATC group (73.3 vs. 0% in ATC and PDTC, respectively). In the ATC group, the TPS for tumor positive PD-L1 expression revealed a non-significant trend towards worse survival, but no difference was observed when investigating PD-L1 expression in TILs and TAMs. In addition to increased PD-L1 expression, all ATC cases exhibited significantly increased CD3+ and CD8+ T cells, CD68+ and CD163+ macrophages, and S100+ dendritic cells compared with the PDTC cases. Loss of mutL homolog 1 and PMS1 homolog 2 expression was observed in one ATC case with the highest PD-L1 expression, as well as in the only PDTC case positive for PD-L1. Notably, the latter was the only PDTC case exhibiting positivity for p53 and a cellular microenvironment similar to ATC. The current results indicated that PD-L1 expression was frequent in ATC, but rare in PDTC. In addition to PD-L1, the present study suggested that microsatellite instability may serve a role in both the TME and the identification of immunotherapy candidates among patients with PDTC.
Collapse
Affiliation(s)
- Soledad Cameselle-García
- Department of Medical Oncology, University Hospital Complex of Ourense, Galician Healthcare Service, 32005 Ourense, Spain
| | - Sámer Abdulkader-Sande
- Department of Pathology, Clinical University Hospital of Santiago de Compostela, Health Research Institute of Santiago de Compostela, Galician Healthcare Service, 15706 Santiago de Compostela, Spain
| | - María Sánchez-Ares
- Department of Pathology, Clinical University Hospital of Santiago de Compostela, Health Research Institute of Santiago de Compostela, Galician Healthcare Service, 15706 Santiago de Compostela, Spain
| | - Gemma Rodríguez-Carnero
- Department of Endocrinology and Nutrition, Clinical University Hospital of Santiago de Compostela, Health Research Institute of Santiago de Compostela, Galician Healthcare Service, 15706 Santiago de Compostela, Spain
| | - Jesús Garcia-Gómez
- Department of Medical Oncology, University Hospital Complex of Ourense, Galician Healthcare Service, 32005 Ourense, Spain
| | - Francisco Gude-Sampedro
- Department of Epidemiology, Clinical University Hospital of Santiago de Compostela, Health Research Institute of Santiago de Compostela, Galician Healthcare Service, 15706 Santiago de Compostela, Spain.,School of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Ihab Abdulkader-Nallib
- Department of Pathology, Clinical University Hospital of Santiago de Compostela, Health Research Institute of Santiago de Compostela, Galician Healthcare Service, 15706 Santiago de Compostela, Spain.,School of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - José Manuel Cameselle-Teijeiro
- Department of Pathology, Clinical University Hospital of Santiago de Compostela, Health Research Institute of Santiago de Compostela, Galician Healthcare Service, 15706 Santiago de Compostela, Spain.,School of Medicine, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| |
Collapse
|
14
|
Greisen SR, Deleuran B. Checkpoint Molecules in Rheumatology-or the Benefits of Being Exhausted. Curr Rheumatol Rep 2021; 23:22. [PMID: 33651184 DOI: 10.1007/s11926-021-00991-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW This review will focus on the most common co-inhibitory molecules, emphasizing the importance of these in relation to rheumatic disease. RECENT FINDINGS Checkpoint molecules are pivotal in determining the outcome of antigen activation. Checkpoint molecules consist of co-stimulatory and co-inhibitory molecules, where the first activates and the latter inhibits the antigen presentation process. Studies show that increased activity of co-inhibitory molecules is associated with a good prognosis in rheumatic diseases. Opposite, when cancer patients are treated with antibodies blocking the inhibitory pathways, autoimmune diseases, including arthritis, develop as immune-related adverse events (IrAE). This emphasizes the importance of these pathways in autoimmune disease. Co-inhibitory molecules are becoming increasingly interesting as future treatment targets in rheumatic conditions. Treatments with antibodies blocking these pathways result in IrAE, often manifesting as autoimmune rheumatic diseases. Therefore, a need to get acquainted with these molecules is growing so we can cope with future challenges in rheumatic diseases.
Collapse
Affiliation(s)
- Stinne Ravn Greisen
- Department of Biomedicine, Aarhus University, Skou-building, C.F. Møllers Alle 6, DK-8000, Aarhus C, Denmark. .,Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark.
| | - Bent Deleuran
- Department of Biomedicine, Aarhus University, Skou-building, C.F. Møllers Alle 6, DK-8000, Aarhus C, Denmark.,Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
15
|
Haugh AM, Salama AKS, Johnson DB. Advanced Melanoma: Resistance Mechanisms to Current Therapies. Hematol Oncol Clin North Am 2021; 35:111-128. [PMID: 33759769 PMCID: PMC7991196 DOI: 10.1016/j.hoc.2020.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Novel therapeutic agents introduced over the past decade, including immune checkpoint inhibitors and targeted therapies, have revolutionized the management of metastatic melanoma and significantly improved patient outcomes. Although robust and durable responses have been noted in some cases, treatment is often limited by innate or acquired resistance to these agents. This article provides an overview of known and suspected mechanisms involved with acquired resistance to BRAF/MEK inhibitors as well as developing insights into innate and acquired resistance to checkpoint inhibitors in patients with melanoma.
Collapse
Affiliation(s)
- Alexandra M Haugh
- Department of Medicine, Vanderbilt University Medical Center, 719 Thompson Lane, Suite 20400, Nashville, TN 37204, USA
| | - April K S Salama
- Department of Medicine, Duke University Medical Center, 20 Duke Medicine Cir, Durham, NC 27710, USA
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Vanderbilt Ingram Cancer Center, 777 PRB, 2220 Pierce Avenue, Nashville, TN 37232, USA.
| |
Collapse
|
16
|
Pavan A, Bragadin AB, Calvetti L, Ferro A, Zulato E, Attili I, Nardo G, Dal Maso A, Frega S, Menin AG, Fassan M, Calabrese F, Pasello G, Guarneri V, Aprile G, Conte P, Rosell R, Indraccolo S, Bonanno L. Role of next generation sequencing-based liquid biopsy in advanced non-small cell lung cancer patients treated with immune checkpoint inhibitors: impact of STK11, KRAS and TP53 mutations and co-mutations on outcome. Transl Lung Cancer Res 2021; 10:202-220. [PMID: 33569305 PMCID: PMC7867770 DOI: 10.21037/tlcr-20-674] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 11/12/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND Characterization of tumor-related genetic alterations is promising for the screening of new predictive markers in non-small cell lung cancer (NSCLC). Aim of the study was to evaluate prognostic and predictive role of most frequent tumor-associated genetic alterations detected in plasma before starting immune checkpoint inhibitors (ICIs). METHODS Between January 2017 and October 2019, advanced NSCLC patients were prospectively screened with plasma next-generation sequencing (NGS) while included in two trials: VISION (NCT02864992), using Guardant360® test, and MAGIC (Monitoring Advanced NSCLC through plasma Genotyping during Immunotherapy: Clinical feasibility and application), using Myriapod NGS-IL 56G Assay. A control group of patients not receiving ICIs was analyzed. RESULTS A total of 103 patients receiving ICIs were analyzed: median overall survival (OS) was 20.8 (95% CI: 16.7-24.9) months and median immune-related progression free disease (irPFS) 4.2 (95% CI: 2.3-6.1) months. TP53 mutations in plasma negatively affected OS both in patients treated with ICIs and in control group (P=0.001 and P=0.009), indicating a prognostic role. STK11 mutated patients (n=9) showed a trend for worse OS only if treated with ICIs. The presence of KRAS/STK11 co-mutation and KRAS/STK11/TP53 co-mutation affected OS only in patients treated with ICIs (HR =10.936, 95% CI: 2.337-51.164, P=0.002; HR =17.609, 95% CI: 3.777-82.089, P<0.001, respectively), indicating a predictive role. CONCLUSIONS Plasma genotyping demonstrated prognostic value of TP53 mutations and predictive value of KRAS/STK11 and KRAS/STK11/TP53 co-mutations.
Collapse
Affiliation(s)
- Alberto Pavan
- Medical Oncology 2, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
| | - Andrea Boscolo Bragadin
- Department of Surgery, Oncology and Gastroenterology, Università degli Studi di Padova, Padova, Italy
| | - Lorenzo Calvetti
- Department of Oncology, San Bortolo General Hospital, ULSS8 Berica - East District, Vicenza, Italy
| | - Alessandra Ferro
- Medical Oncology 2, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, Università degli Studi di Padova, Padova, Italy
| | - Elisabetta Zulato
- Immunology and Molecular Oncology Unit, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
| | - Ilaria Attili
- Medical Oncology 2, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, Università degli Studi di Padova, Padova, Italy
| | - Giorgia Nardo
- Immunology and Molecular Oncology Unit, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
| | - Alessandro Dal Maso
- Medical Oncology 2, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, Università degli Studi di Padova, Padova, Italy
| | - Stefano Frega
- Medical Oncology 2, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
| | - Andrea Giovanni Menin
- Department of Pathology, San Bortolo General Hospital, ULSS8 Berica - East District, Vicenza, Italy
| | - Matteo Fassan
- Department of Medicine, Surgical Pathology and Cytopathology Unit, Università degli Studi di Padova, Padova, Italy
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, Università degli Studi di Padova, Padova, Italy
| | - Giulia Pasello
- Medical Oncology 2, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
| | - Valentina Guarneri
- Medical Oncology 2, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, Università degli Studi di Padova, Padova, Italy
| | - Giuseppe Aprile
- Department of Oncology, San Bortolo General Hospital, ULSS8 Berica - East District, Vicenza, Italy
| | - PierFranco Conte
- Medical Oncology 2, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, Università degli Studi di Padova, Padova, Italy
| | - Rafael Rosell
- Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Germans Trias I Pujol Health Sciences Institute and Hospital Badalona, Barcelona, Spain
| | - Stefano Indraccolo
- Immunology and Molecular Oncology Unit, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
| | - Laura Bonanno
- Medical Oncology 2, Istituto Oncologico Veneto IOV - IRCCS, Padova, Italy
| |
Collapse
|
17
|
Lassalle S, Nahon-Esteve S, Frouin E, Boulagnon-Rombi C, Josselin N, Cassoux N, Barnhill R, Scheller B, Baillif S, Hofman P. PD-L1 Expression in 65 Conjunctival Melanomas and Its Association with Clinical Outcome. Int J Mol Sci 2020; 21:ijms21239147. [PMID: 33266349 PMCID: PMC7731195 DOI: 10.3390/ijms21239147] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/17/2020] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
Conjunctival melanoma (CM) iss a rare and aggressive tumour that is increasing in frequency. The prognostic value of PD-L1 expression, alone or in combination with CD8 and PD-1 expression and the BRAF and NRAS status, has not been determined in CM to date. We evaluated the expression of PD-L1, CD8, PD-1 in CM and investigated whether there was an association between the expression of these markers and the BRAF and NRAS molecular profile as well as some clinico-pathological criteria. A total of sixty-five CM were assessed for PD-L1, PD-1, and CD8 expression by immunohistochemistry (IHC) and for BRAF and NRAS genomic alterations using molecular biology techniques and anti-BRAF and anti-NRAS antibodies. PD-L1 expression in tumour cells (TC) was very low or absent but detected in tumour-infiltrating immune cells (IC). A correlation was observed between the expression of PD-L1, CD8, and PD-1 in IC. No correlation between PD-L1 expression (in tumour and/or immune cells) and BRAF or NRAS mutations was observed. PD-L1 expression in IC correlated with a higher pTNM stage and PD-L1 expression in TC with worse disease-specific survival. PD-L1 expression is a potential prognostic biomarker that correlates with poor prognosis in CM patients.
Collapse
Affiliation(s)
- Sandra Lassalle
- Laboratory of Clinical and Experimental Pathology, Centre Hospitalier Universitaire de Nice, University Côte d’Azur, Pasteur 1 Hospital, 30 avenue de la voie Romaine CS 51069, 06001 Nice CEDEX 1, France;
- Institute of Research on Cancer and Aging of Nice (IRCAN), INSERM U1081/CNRS UMR7284, Medical School 28, Avenue de Valombrose, 06107 Nice CEDEX 2, France
- FHU OncoAge, Centre Hospitalier Universitaire de Nice, University Côte d’Azur, Pasteur Hospital, 30 avenue de la voie Romaine CS 51069, 06001 Nice CEDEX 1, France
- Hospital-Integrated Biobank (BB 0033-00025), Laboratory of Clinical and Experimental Pathology, Pasteur 1 Hospital, 30 avenue de la voie Romaine CS 51069, 06001 Nice CEDEX 1, France
| | - Sacha Nahon-Esteve
- Department of Ophthalmology, Pasteur 2 Hospital, 30 avenue de la voie Romaine CS 51069, 06001 Nice CEDEX 1, France; (S.N.-E.); (S.B.)
| | - Eric Frouin
- Laboratory of Pathology, Centre Hospitalier Universitaire de Poitiers, 2 rue de la Milétrie, CS 90577, 86021 Poitiers CEDEX, France;
| | - Camille Boulagnon-Rombi
- Laboratory of Pathology, Centre Hospitalier Universitaire de Reims, avenue du Général Koenig, 51092 Reims CEDEX, France;
| | - Nicolas Josselin
- Institut d’Histo-Pathologie, 55 rue Amiral du Chaffault, CS 50424, 44104 Nantes CEDEX 4, France;
| | - Nathalie Cassoux
- Department of Ophthalmology, Institut Curie, 26 rue d’Ulm, 75248 Paris CEDEX 5, France;
| | - Raymond Barnhill
- Department of Pathology, Institut Curie, 26 rue d’Ulm, 75248 Paris CEDEX 5, France;
- Faculty of Medicine University of Paris Descartes, 15 rue de l’École de Médecine, 75006 Paris, France
| | - Boris Scheller
- Department of Epidemiology and Biostatistics, CLCC CAL, 33 avenue de Valombrose, 06189 Nice CEDEX 2, France;
| | - Stéphanie Baillif
- Department of Ophthalmology, Pasteur 2 Hospital, 30 avenue de la voie Romaine CS 51069, 06001 Nice CEDEX 1, France; (S.N.-E.); (S.B.)
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Centre Hospitalier Universitaire de Nice, University Côte d’Azur, Pasteur 1 Hospital, 30 avenue de la voie Romaine CS 51069, 06001 Nice CEDEX 1, France;
- Institute of Research on Cancer and Aging of Nice (IRCAN), INSERM U1081/CNRS UMR7284, Medical School 28, Avenue de Valombrose, 06107 Nice CEDEX 2, France
- FHU OncoAge, Centre Hospitalier Universitaire de Nice, University Côte d’Azur, Pasteur Hospital, 30 avenue de la voie Romaine CS 51069, 06001 Nice CEDEX 1, France
- Hospital-Integrated Biobank (BB 0033-00025), Laboratory of Clinical and Experimental Pathology, Pasteur 1 Hospital, 30 avenue de la voie Romaine CS 51069, 06001 Nice CEDEX 1, France
- Correspondence: ; Tel.: +33-4-92-03-88-55; Fax: +33-4-92-03-87-50
| |
Collapse
|
18
|
Hasan S. An Overview of Promising Biomarkers in Cancer Screening and Detection. Curr Cancer Drug Targets 2020; 20:831-852. [PMID: 32838718 DOI: 10.2174/1568009620666200824102418] [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: 05/13/2020] [Revised: 07/03/2020] [Accepted: 07/13/2020] [Indexed: 11/22/2022]
Abstract
Applications of biomarkers have been proved in oncology screening, diagnosis, predicting response to treatment as well as monitoring the progress of the disease. Considering the crucial role played by them during different disease stages, it is extremely important to evaluate, validate, and assess them to incorporate them into routine clinical care. In this review, the role of few most promising and successfully used biomarkers in cancer detection, i.e. PD-L1, E-Cadherin, TP53, Exosomes, cfDNA, EGFR, mTOR with regard to their structure, mode of action, and reports signifying their pathological significance, are addressed. Also, an overview of some successfully used biomarkers for cancer medicine has been presented. The study also summarizes biomarker-driven personalized cancer therapy i.e., approved targets and indications, as per the US FDA. The review also highlights the increasingly prominent role of biomarkers in drug development at all stages, with particular reference to clinical trials. The increasing utility of biomarkers in clinical trials is clearly evident from the trend shown, wherein ~55 percent of all oncology clinical trials in 2019 were seen to involve biomarkers, as opposed to ~ 15 percent in 2001, which clearly proves the essence and applicability of biomarkers for synergizing clinical information with tumor progression. Still, there are significant challenges in the implementation of these possibilities with strong evidence in cost-- effective manner.
Collapse
Affiliation(s)
- Saba Hasan
- Amity Institute of Biotechnology, Amity University, Uttar Pradesh, Lucknow, India
| |
Collapse
|