1
|
Tulsian K, Thakker D, Vyas VK. Overcoming chimeric antigen receptor-T (CAR-T) resistance with checkpoint inhibitors: Existing methods, challenges, clinical success, and future prospects : A comprehensive review. Int J Biol Macromol 2025; 306:141364. [PMID: 39988153 DOI: 10.1016/j.ijbiomac.2025.141364] [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: 08/09/2024] [Revised: 11/20/2024] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Immune checkpoint blockade is, as of today, the most successful form of cancer immunotherapy, with more than 43 % of cancer patients in the US eligible to receive it; however, only up to 12.5 % of patients respond to it. Similarly, adoptive cell therapy using bioengineered chimeric antigen receptorT (CAR-T) cells and T-cell receptor (TCR) cells has provided excellent responses against liquid tumours, but both forms of immunotherapy have encountered challenges within a tumour microenvironment that is both lacking in tumour-specific T-cells and is strongly immunosuppressive toward externally administered CAR-T and TCR cells. This review focuses on understanding approved checkpoint blockade and adoptive cell therapy at both biological and clinical levels before delving into how and why their combination holds significant promise in overcoming their individual shortcomings. The advent of next-generation checkpoint inhibitors has further strengthened the immune checkpoint field, and a special section explores how these inhibitors can address existing hurdles in combining checkpoint blockade with adoptive cell therapy and homing in on our cancer target for long-term immunity.
Collapse
Affiliation(s)
- Kartik Tulsian
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Dhinal Thakker
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India
| | - Vivek K Vyas
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad, 382481, Gujarat, India.
| |
Collapse
|
2
|
He S, Luo S, Cai B, Chen J, Zhang Y, Zhao F, Liu Q, Liu T, Wang W, Peng T, Lu X, Zheng S. Divergent roles of PKM2 in regulating PD-L1 and PD-L2 expression and their implications in human and mouse cancer models. Acta Biochim Biophys Sin (Shanghai) 2025. [PMID: 39980348 DOI: 10.3724/abbs.2025019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2025] Open
Abstract
Cancer cells evade immune detection through checkpoint molecules like PD-L1 and PD-L2 which suppress T-cell activation. While PD-L1 is well-studied, the role of PD-L2 remains unclear. Pyruvate kinase M2 (PKM2), a metabolic enzyme, influences immune checkpoint regulation, but its role in PD-L1 and PD-L2 modulation is not well defined. Here, we investigate the role of pyruvate kinase M2 (PKM2) in modulating the immune checkpoint molecules PD-L1 and PD-L2 via GATA3 in cancer cells, with insights from both human and mouse models. We find that PKM2 enhances PD-L1 expression while inhibiting PD-L2, a dual regulatory mechanism that facilitates immune evasion. Knockdown and overexpression experiments revealed GATA3 as a key mediator. PKM2 knockout reduced GATA3 level, leading to decreased PD-L1 and increased PD-L2 expression. Chromatin immunoprecipitation (ChIP)-qPCR demonstrates that GATA3 functions as a direct transcription factor capable of binding to the promoters of PD-L1 and PD-L2. In silico analyses of 81 esophageal squamous cell carcinoma (ESCC) cases from the TCGA database demonstrate that PKM2 mRNA is unrelated to PD-L1 and PD-L2 expression but is negatively correlated with CD8 + T-cell infiltration in ESCC. To further validate these findings, we establish a xenograft model using immune-competent C57/BL6N mice, where knockdown of PKM2 results in significant downregulation of both PD-L1 and PD-L2 expression. Collectively, these findings underscore the divergent roles of PKM2 in regulating immune checkpoint expression in human and mouse cancer models and suggest that targeting the PKM2-GATA3 axis could enhance cancer immunotherapy by fine-tuning PD-L1 and PD-L2 levels.
Collapse
Affiliation(s)
- Shuo He
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
- Department of Pathology, Basic Medicine College, Xinjiang Medical University, Urumqi 830017, China
| | - Shujuan Luo
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
- Department of Pathology, Basic Medicine College, Xinjiang Medical University, Urumqi 830017, China
| | - Bangwu Cai
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
- Department of Pathology, Basic Medicine College, Xinjiang Medical University, Urumqi 830017, China
| | - Jiao Chen
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
| | - Yao Zhang
- Beijing Beanstalk International Bilingual School, Beijing, 100016
| | - Feng Zhao
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Disease, Operation Management Department, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011
| | - Qing Liu
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
| | - Tao Liu
- Department of Clinical Laboratory, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
| | - Wei Wang
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
| | - Tianyuan Peng
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
| | - Xiaomei Lu
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
| | - Shutao Zheng
- State Key Laboratory of Pathogenesis, Prevention, Treatment of Central Asian High Incidence Diseases, Clinical Medical Research Institute, First Affiliated Hospital of Xinjiang Medical University, Xinjiang Uygur Autonomous Region, Urumqi 830011, China
- Department of Pathology, Basic Medicine College, Xinjiang Medical University, Urumqi 830017, China
| |
Collapse
|
3
|
Wei X, Xiong X, Chen Z, Chen B, Zhang C, Zhang W. MicroRNA155 in non-small cell lung cancer: a potential therapeutic target. Front Oncol 2025; 15:1517995. [PMID: 39963112 PMCID: PMC11830606 DOI: 10.3389/fonc.2025.1517995] [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: 10/27/2024] [Accepted: 01/09/2025] [Indexed: 02/20/2025] Open
Abstract
Lung cancer (LC) is the second most commonly diagnosed cancer among both men and women, and it stands as the leading cause of cancer-related mortality, characterized by high rates of morbidity and mortality. Among its subtypes, non-small cell lung cancer (NSCLC) is the most prevalent and one of the most challenging malignant tumors to treat. To date, various therapeutic approaches, including surgery, radiotherapy, and chemotherapy, have been employed in the management of lung cancer; however, due to its aggressive nature, the survival rates remain low. Consequently, exploring novel treatment strategies is of paramount importance. MicroRNAs (miRNAs), a large family of non-coding RNAs, play crucial roles in regulating several key biological processes, including cell proliferation, differentiation, inflammation, and apoptosis. Among these, microRNA155(miR-155) is one of the most conserved and versatile miRNAs, predominantly overexpressed in various diseases, including malignant tumors. This review elucidates the biological functions and roles of miR-155 in NSCLC and discusses its potential significance as a therapeutic target for future research directions and clinical applications.
Collapse
Affiliation(s)
- Xiangju Wei
- The First Clinical College, Xuzhou Medical University, Xuzhou, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xianmin Xiong
- The First Clinical College, Xuzhou Medical University, Xuzhou, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Ze Chen
- The First Clinical College, Xuzhou Medical University, Xuzhou, China
| | - Bi Chen
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Cantang Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wenhui Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
4
|
Prochera A, Muppirala AN, Kuziel GA, Soualhi S, Shepherd A, Sun L, Issac B, Rosenberg HJ, Karim F, Perez K, Smith KH, Archibald TH, Rakoff-Nahoum S, Hagen SJ, Rao M. Enteric glia regulate Paneth cell secretion and intestinal microbial ecology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589545. [PMID: 38659931 PMCID: PMC11042301 DOI: 10.1101/2024.04.15.589545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Glial cells of the enteric nervous system (ENS) interact closely with the intestinal epithelium and secrete signals that influence epithelial cell proliferation and barrier formation in vitro. Whether these interactions are important in vivo, however, is unclear because previous studies reached conflicting conclusions [1]. To better define the roles of enteric glia in steady state regulation of the intestinal epithelium, we characterized the glia in closest proximity to epithelial cells and found that the majority express PLP1 in both mice and humans. To test their functions using an unbiased approach, we genetically depleted PLP1+ cells in mice and transcriptionally profiled the small and large intestines. Surprisingly, glial loss had minimal effects on transcriptional programs and the few identified changes varied along the gastrointestinal tract. In the ileum, where enteric glia had been considered most essential for epithelial integrity, glial depletion did not drastically alter epithelial gene expression but caused a modest enrichment in signatures of Paneth cells, a secretory cell type important for innate immunity. In the absence of PLP1+ glia, Paneth cell number was intact, but a subset appeared abnormal with irregular and heterogenous cytoplasmic granules, suggesting a secretory deficit. Consistent with this possibility, ileal explants from glial-depleted mice secreted less functional lysozyme than controls with corresponding effects on fecal microbial composition. Collectively, these data suggest that enteric glia do not exert broad effects on the intestinal epithelium but have an essential role in regulating Paneth cell function and gut microbial ecology.
Collapse
Affiliation(s)
- Aleksandra Prochera
- Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA
| | - Anoohya N Muppirala
- Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA
| | - Gavin A Kuziel
- Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA
- Division of Infectious Diseases, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA
- Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Salima Soualhi
- Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA
| | - Amy Shepherd
- Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA
| | - Liang Sun
- Research Computing, Department of Information Technology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Biju Issac
- Research Computing, Department of Information Technology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Harry J Rosenberg
- Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Farah Karim
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, USA
| | - Kristina Perez
- Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA
| | - Kyle H Smith
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Tonora H Archibald
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Seth Rakoff-Nahoum
- Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA
- Division of Infectious Diseases, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA
- Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
| | - Susan J Hagen
- Department of Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Meenakshi Rao
- Division of Gastroenterology, Department of Pediatrics, Boston Children's Hospital and Harvard Medical School, 300 Longwood Ave, Boston, MA 02115, USA
| |
Collapse
|
5
|
Zhu L, Qu Y, Yang J, Shao T, Kuang J, Liu C, Qi Y, Li M, Li Y, Zhang S, Wang J, Liu Y, Liu J, Hu Y, Zhu L, Hou T. PD-L2 act as an independent immune checkpoint in colorectal cancer beyond PD-L1. Front Immunol 2024; 15:1486888. [PMID: 39687608 PMCID: PMC11646888 DOI: 10.3389/fimmu.2024.1486888] [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: 08/27/2024] [Accepted: 11/11/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction Immunotherapy, especially immune checkpoint blockade (ICB), holds promise as a therapeutic strategy in colorectal cancer (CRC) by harnessing the patient's immune system to target malignant cells. Particularly, the PD-1/PD-L1 axis is widely recognized for its critical role in tumor microenvironment immunosuppression. Antibodies targeting PD-1 or PD-L1 have shown sustained efficacy against various cancers, including CRC. Nonetheless, many CRC patients exhibit limited responses to such immunotherapy, and the resistance mechanisms remain incompletely understood. Methods We conducted experiments with C57BL/6 mice, and used the MC38 cell line for ICB treatment studies in syngeneic mouse models. Gene and protein analyses were performed using qPCR, Western Blot, and flow cytometry, with bioinformatics for clinical data survival analysis. Results In this study, we reveal that targeting PD-L2 emerges as a complementary therapeutic strategy to PD-1/PD-L1 blockade in CRC. Although PD-L2 is also inducible by IFNγ, like PD-L1, it displays a unique spatial distribution within the tumor microenvironment, implying discrete roles in immune evasion. Additionally, we uncovered a significant correlation between PD-L1 and PD-L2 expression levels and the infiltration of various immune cells, encompassing multiple dendritic cell (DC) subtypes. This correlation implies an enhanced antigen presentation process that may be unleashed by blocking these two immune checkpoints. Discussion Our results highlight the significance of PD-L2 as an essential immune checkpoint alongside PD-L1 and emphasize its potential as a target for bolstering antitumor immunity in colorectal cancer.
Collapse
Affiliation(s)
- Lvyun Zhu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Ying Qu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Junru Yang
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Tong Shao
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Jingyu Kuang
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Chuanyang Liu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Yanhua Qi
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Ming Li
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
- Laboratory of Liquid Propellant Application Technology, Jiuquan Satellite Launch Centre, Jiuquan, China
| | - Yingying Li
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Sujuan Zhang
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingyang Wang
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Yu Liu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Jiali Liu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Yanming Hu
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lingyun Zhu
- Department of Biology and Chemistry, College of Sciences, National University of Defense Technology, Changsha, Hunan, China
| | - Tao Hou
- Department of Oncology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
6
|
Chen L, Huang L, Gu Y, Li C, Sun P, Xiang Y. Novel post-translational modifications of protein by metabolites with immune responses and immune-related molecules in cancer immunotherapy. Int J Biol Macromol 2024; 277:133883. [PMID: 39033895 DOI: 10.1016/j.ijbiomac.2024.133883] [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: 07/01/2023] [Revised: 06/30/2024] [Accepted: 07/13/2024] [Indexed: 07/23/2024]
Abstract
Tumour immunotherapy is an effective and essential treatment for cancer. However, the heterogeneity of tumours and the complex and changeable tumour immune microenvironment (TME) creates many uncertainties in the clinical application of immunotherapy, such as different responses to tumour immunotherapy and significant differences in individual efficacy. It makes anti-tumour immunotherapy face many challenges. Immunometabolism is a critical determinant of immune cell response to specific immune effector molecules, significantly affecting the effects of tumour immunotherapy. It is attributed mainly to the fact that metabolites can regulate the function of immune cells and immune-related molecules through the protein post-translational modifications (PTMs) pathway. This study systematically summarizes a variety of novel protein PTMs including acetylation, propionylation, butyrylation, succinylation, crotonylation, malonylation, glutarylation, 2-hydroxyisobutyrylation, β-hydroxybutyrylation, benzoylation, lactylation and isonicotinylation in the field of tumour immune regulation and immunotherapy. In particular, we elaborate on how different PTMs in the TME can affect the function of immune cells and lead to immune evasion in cancer. Lastly, we highlight the potential treatment with the combined application of target-inhibited protein modification and immune checkpoint inhibitors (ICIs) for improved immunotherapeutic outcomes.
Collapse
Affiliation(s)
- Lihua Chen
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China; National Clinical Research Center for Obstetric & Gynecologic Diseases, PR China
| | - Lixiang Huang
- Laboratory of Gynecologic Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, PR China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fuzhou 350001, Fujian, PR China
| | - Yu Gu
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China; National Clinical Research Center for Obstetric & Gynecologic Diseases, PR China
| | - Chen Li
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China; National Clinical Research Center for Obstetric & Gynecologic Diseases, PR China
| | - Pengming Sun
- Laboratory of Gynecologic Oncology, Department of Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou 350001, Fujian, PR China; Fujian Key Laboratory of Women and Children's Critical Diseases Research, Fuzhou 350001, Fujian, PR China.
| | - Yang Xiang
- Department of Obstetrics and Gynecology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, PR China; National Clinical Research Center for Obstetric & Gynecologic Diseases, PR China.
| |
Collapse
|
7
|
Scibilia KR, Schlicke P, Schneller F, Kuttler C. Predicting resistance and pseudoprogression: are minimalistic immunoediting mathematical models capable of forecasting checkpoint inhibitor treatment outcomes in lung cancer? Math Biosci 2024; 376:109287. [PMID: 39218211 DOI: 10.1016/j.mbs.2024.109287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 08/15/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The increased application of immune checkpoint inhibitors (ICIs) targeting PD-1/PD-L1 in lung cancer treatment generates clinical need to reliably predict individual patients' treatment outcomes. METHODS To bridge the prediction gap, we examine four different mathematical models in the form of ordinary differential equations, including a novel delayed response model. We rigorously evaluate their individual and combined predictive capabilities with regard to the patients' progressive disease (PD) status through equal weighting of model-derived outcome probabilities. RESULTS Fitting the complete treatment course, the novel delayed response model (R2=0.938) outperformed the simplest model (R2=0.865). The model combination was able to reliably predict patient PD outcome with an overall accuracy of 77% (sensitivity = 70%, specificity = 81%), solely through calibration with primary tumor longest diameter measurements. It autonomously identified a subset of 51% of patients where predictions with an overall accuracy of 81% (sensitivity = 81%, specificity = 81%) can be achieved. All models significantly outperformed a fully data-driven machine learning-based approach. IMPLICATIONS These modeling approaches provide a dynamic baseline framework to support clinicians in treatment decisions by identifying different treatment outcome trajectories with already clinically available measurement data. LIMITATIONS AND FUTURE DIRECTIONS Conjoint application of the presented approach with other predictive tools and biomarkers, as well as further disease information (e.g. metastatic stage), could further enhance treatment outcome prediction. We believe the simple model formulations allow widespread adoption of the developed models to other cancer types. Similar models can easily be formulated for other treatment modalities.
Collapse
Affiliation(s)
- Kevin Robert Scibilia
- Department of Computer Science, TUM School of Computation, Information and Technology, Technical University of Munich, Boltzmannstr. 3, Garching, 85747, Germany
| | - Pirmin Schlicke
- Department of Mathematics, TUM School of Computation, Information and Technology, Technical University of Munich, Boltzmannstr. 3, Garching, 85747, Germany.
| | - Folker Schneller
- Department of Internal Medicine III, Klinikum Rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Ismaninger Str. 22, Munich, 81675, Germany
| | - Christina Kuttler
- Department of Mathematics, TUM School of Computation, Information and Technology, Technical University of Munich, Boltzmannstr. 3, Garching, 85747, Germany
| |
Collapse
|
8
|
Cysneiros MADPC, Cirqueira MB, Barbosa LDF, Chaves de Oliveira Ê, Morais LK, Wastowski IJ, Floriano VG. Immune cells and checkpoints in pancreatic adenocarcinoma: Association with clinical and pathological characteristics. PLoS One 2024; 19:e0305648. [PMID: 38954689 PMCID: PMC11218951 DOI: 10.1371/journal.pone.0305648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 06/03/2024] [Indexed: 07/04/2024] Open
Abstract
INTRODUCTION Pancreatic adenocarcinoma is an extremely aggressive neoplasm, with many challenges to be overcome in order to achieve a truly effective treatment. It is characterized by a mostly immunosuppressed environment, with dysfunctional immune cells and active immunoinhibitory pathways that favor tumor evasion and progression. Thus, the study and understanding of the tumor microenvironment and the various cells subtypes and their functional capacities are essential to achieve more effective treatments, especially with the use of new immunotherapeutics. METHODS Seventy cases of pancreatic adenocarcinoma divided into two groups 43 with resectable disease and 27 with unresectable disease were analyzed using immunohistochemical methods regarding the expression of programmed cell death ligand 1 (PD-L1), programmed cell death ligand 2 (PD-L2), and human leukocyte antigen G (HLA-G) molecules as well as the populations of CD4+ and CD8+ T lymphocytes, regulatory T cells (Tregs), and M2 macrophages (MM2). Several statistical tests, including multivariate analyses, were performed to examine how those immune cells and immunoinhibitory molecules impact the evolution and prognosis of pancreatic adenocarcinoma. RESULTS CD8+ T lymphocytes and M2 macrophages predominated in the group operated on, and PD-L2 expression predominated in the unresectable group. PD-L2 was associated with T stage, lymph node metastasis, and clinical staging, while in survival analysis, PD-L2 and HLA-G were associated with a shorter survival. In the inoperable cases, Tregs cells, MM2, PD-L1, PD-L2, and HLA-G were positively correlated. CONCLUSIONS PD-L2 and HLA-G expression correlated with worse survival in the cases studied. Tumor microenvironment was characterized by a tolerant and immunosuppressed pattern, mainly in unresectable lesions, where a broad positive influence was observed between immunoinhibitory cells and immune checkpoint proteins expressed by tumor cells.
Collapse
Affiliation(s)
| | - Magno Belém Cirqueira
- Diagnostic and Therapeutic Support Division of Clinical Hospital, Federal University of Goias, Goiania, Brazil
| | | | | | - Lucio Kenny Morais
- Surgery Department of Medicine College, Federal University of Goias, Goiania, Brazil
| | | | - Vitor Gonçalves Floriano
- Clinics Department of Medicine College, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
9
|
Sun Y, Yang J, Chen Y, Guo Y, Xiong J, Guo X, Zhang Y, Gu L, Tong M, Wang W, Sun J. PD-L2 Expression in Breast Cancer Promotes Tumor Development and Progression. J Immunol Res 2024; 2024:3145695. [PMID: 38983273 PMCID: PMC11233179 DOI: 10.1155/2024/3145695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 06/01/2024] [Accepted: 06/10/2024] [Indexed: 07/11/2024] Open
Abstract
Background This work focused on investigating the role of programmed death ligand 2 (PD-L2) in the progression of breast cancer by utilizing breast cancer specimens and cells. Materials and Methods The serum levels of soluble PD-L2 (sPD-L2) in breast cancer patients and healthy individuals were analyzed by means of the enzyme-linked immunosorbent assay, and the PD-L2 levels within 416 resected breast cancer specimens were assessed through immunohistochemistry. Concurrently, in vitro cell experiments and in vivo animal experiments were carried out to analyze the relationship between PD-L2 and the invasion and migration of breast cancer. Results The concentration of sPD-L2 in breast cancer patients significantly increased compared to that in the control groups. Additionally, breast cancer patients with high concentrations of sPD-L2 had higher Ki67 values (≥30%) and tumor grades. PD-L2 was expressed in 79.09% of the cancer samples, which exhibited a positive correlation with the progesterone receptor (PR) and the human epidermal growth factor receptor 2 (HER2). Furthermore, we discovered that knockdown of PD-L2 inhibited the migratory and invasive abilities of both MCF-7 and MDA-MB231 cells. Conclusion Our findings demonstrated that knockdown of PD-L2 suppressed tumor growth, providing novel insights into important biological functions.
Collapse
Affiliation(s)
- Yuling Sun
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in OncologySuzhou Vocational Health College, Suzhou 215009, China
| | - Jie Yang
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in OncologySuzhou Vocational Health College, Suzhou 215009, China
| | - Yachun Chen
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in OncologySuzhou Vocational Health College, Suzhou 215009, China
| | - Yundi Guo
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in OncologySuzhou Vocational Health College, Suzhou 215009, China
| | - Jian Xiong
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in OncologySuzhou Vocational Health College, Suzhou 215009, China
| | - Xuqin Guo
- Center for Drug Metabolism and PharmacokineticsCollege of Pharmaceutical SciencesSoochow University, Suzhou 215123, China
| | - Yawen Zhang
- Center for Drug Metabolism and PharmacokineticsCollege of Pharmaceutical SciencesSoochow University, Suzhou 215123, China
| | - Li Gu
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in OncologySuzhou Vocational Health College, Suzhou 215009, China
| | - Min Tong
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in OncologySuzhou Vocational Health College, Suzhou 215009, China
| | - Weipeng Wang
- Center for Drug Metabolism and PharmacokineticsCollege of Pharmaceutical SciencesSoochow University, Suzhou 215123, China
| | - Jing Sun
- Jiangsu Province Engineering Research Center of Molecular Target Therapy and Companion Diagnostics in OncologySuzhou Vocational Health College, Suzhou 215009, China
| |
Collapse
|
10
|
Kuol N, Godlewski J, Kmiec Z, Vogrin S, Fraser S, Apostolopoulos V, Nurgali K. Cholinergic signaling influences the expression of immune checkpoint inhibitors, PD-L1 and PD-L2, and tumor hallmarks in human colorectal cancer tissues and cell lines. BMC Cancer 2023; 23:971. [PMID: 37828429 PMCID: PMC10568879 DOI: 10.1186/s12885-023-11410-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/16/2023] [Indexed: 10/14/2023] Open
Abstract
BACKGROUND Cancer cells express immunosuppressive molecules, such as programmed death ligands (PD-L)1 and PD-L2, enabling evasion from the host's immune system. Cancer cells synthesize and secrete acetylcholine (ACh), acting as an autocrine or paracrine hormone to promote their proliferation, differentiation, and migration. METHODS We correlated the expression of PD-L1, PD-L2, cholinergic muscarinic receptor 3 (M3R), alpha 7 nicotinic receptor (α7nAChR), and choline acetyltransferase (ChAT) in colorectal cancer (CRC) tissues with the stage of disease, gender, age, risk, and patient survival. The effects of a muscarinic receptor blocker, atropine, and a selective M3R blocker, 4-DAMP, on the expression of immunosuppressive and cholinergic markers were evaluated in human CRC (LIM-2405, HT-29) cells. RESULTS Increased expression of PD-L1, M3R, and ChAT at stages III-IV was associated with a high risk of CRC and poor survival outcomes independent of patients' gender and age. α7nAChR and PD-L2 were not changed at any CRC stages. Atropine and 4-DAMP suppressed the proliferation and migration of human CRC cells, induced apoptosis, and decreased PD-L1, PD-L2, and M3R expression in CRC cells via inhibition of EGFR and phosphorylation of ERK. CONCLUSIONS The expression of immunosuppressive and cholinergic markers may increase the risk of recurrence of CRC. These markers might be used in determining prognosis and treatment regimens for CRC patients. Blocking cholinergic signaling may be a potential therapeutic for CRC through anti-proliferation and anti-migration via inhibition of EGFR and phosphorylation of ERK. These effects allow the immune system to recognize and eliminate cancer cells.
Collapse
Affiliation(s)
- Nyanbol Kuol
- Institute for Health and Sport, Victoria University, Melbourne, Australia.
- Department of Physiology and Cell Biology, University of Nevada, Reno, USA.
| | | | - Zbigniew Kmiec
- Department of Histology, Medical University of Gdansk, Gdansk, Poland
| | - Sara Vogrin
- Department of Medicine Western Health, University of Melbourne, Melbourne, Australia
| | - Sarah Fraser
- Institute for Health and Sport, Victoria University, Melbourne, Australia
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne, Australia
- Immunology Program, Australian Institute of Musculoskeletal Sciences, Melbourne, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, Melbourne, Australia
- Department of Medicine Western Health, University of Melbourne, Melbourne, Australia
- Regenerative Medicine Program, Australian Institute of Musculoskeletal Sciences, Melbourne, Australia
| |
Collapse
|
11
|
Wagner A, Schlicke P, Fritz M, Kuttler C, Oden JT, Schumann C, Wohlmuth B. A phase-field model for non-small cell lung cancer under the effects of immunotherapy. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:18670-18694. [PMID: 38052574 DOI: 10.3934/mbe.2023828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Formulating mathematical models that estimate tumor growth under therapy is vital for improving patient-specific treatment plans. In this context, we present our recent work on simulating non-small-scale cell lung cancer (NSCLC) in a simple, deterministic setting for two different patients receiving an immunotherapeutic treatment. At its core, our model consists of a Cahn-Hilliard-based phase-field model describing the evolution of proliferative and necrotic tumor cells. These are coupled to a simplified nutrient model that drives the growth of the proliferative cells and their decay into necrotic cells. The applied immunotherapy decreases the proliferative cell concentration. Here, we model the immunotherapeutic agent concentration in the entire lung over time by an ordinary differential equation (ODE). Finally, reaction terms provide a coupling between all these equations. By assuming spherical, symmetric tumor growth and constant nutrient inflow, we simplify this full 3D cancer simulation model to a reduced 1D model. We can then resort to patient data gathered from computed tomography (CT) scans over several years to calibrate our model. Our model covers the case in which the immunotherapy is successful and limits the tumor size, as well as the case predicting a sudden relapse, leading to exponential tumor growth. Finally, we move from the reduced model back to the full 3D cancer simulation in the lung tissue. Thereby, we demonstrate the predictive benefits that a more detailed patient-specific simulation including spatial information as a possible generalization within our framework could yield in the future.
Collapse
Affiliation(s)
- Andreas Wagner
- School of Computation, Information and Technology, Technical University of Munich, Munich, Bavaria, Germany
| | - Pirmin Schlicke
- School of Computation, Information and Technology, Technical University of Munich, Munich, Bavaria, Germany
| | - Marvin Fritz
- Computational Methods for PDEs, Johann Radon Institute for Computational and Applied Mathematics, Linz, Upper Austria, Austria
| | - Christina Kuttler
- School of Computation, Information and Technology, Technical University of Munich, Munich, Bavaria, Germany
| | - J Tinsley Oden
- Oden Institute for Computational Engineering and Sciences, The University of Texas at Austin, Austin, Texas, United States of America
| | - Christian Schumann
- Clinic of Pneumology, Thoracic Oncology, Sleep and Respiratory Critical Care, Klinikverbund Allgäu, Kempten, Bavaria, Germany
| | - Barbara Wohlmuth
- School of Computation, Information and Technology, Technical University of Munich, Munich, Bavaria, Germany
| |
Collapse
|
12
|
Wang F, Xia T, Li Z, Gao X, Fang X. Current status of clinical trial research and application of immune checkpoint inhibitors for non-small cell lung cancer. Front Oncol 2023; 13:1213297. [PMID: 37727216 PMCID: PMC10505960 DOI: 10.3389/fonc.2023.1213297] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 08/17/2023] [Indexed: 09/21/2023] Open
Abstract
Immunotherapy has emerged as a hot topic in the treatment of non-small cell lung cancer (NSCLC) with remarkable success. Compared to chemotherapy patients, the 5-year survival rate for immunotherapy patients is 3-fold higher, approximately 4%-5% versus 15%-16%, respectively. Immunotherapies include chimeric antigen receptor T-cell (CAR-T) therapy, tumor vaccines, immune checkpoint inhibitors, and so forth. Among them, immune checkpoint inhibitors are in the spotlight. Common immune checkpoint inhibitors (ICIs) currently in clinical use include programmed death receptor-1(PD-1)/programmed death ligand-1(PD-L1) and cytotoxic T lymphocyte-associated antigen 4(CTLA-4). This article focuses on monotherapy and combination therapy of CTLA-4 and PD-1/PD-L1 immune checkpoint inhibitors. In particular, the combination therapy of ICIs includes the combination of ICIs and chemotherapy, the combination therapy of dual ICIs, the combination of ICIs and anti-angiogenic drugs, the combination of ICIs and radiotherapy, and the combination of ICIs inhibitors and tumor vaccines and so forth. This article focuses on the combination therapy of ICIs with chemotherapy, the combination therapy of dual ICIs, and the combination therapy of ICIs with anti-angiogenic drugs. The efficacy and safety of ICIs as single agents in NSCLC have been demonstrated in many trials. However, ICIs plus chemotherapy regimens offer significant advantages in the treatment of NSCLC with little to no dramatic increase in toxicity, while combined dual ICIs significantly reduce the adverse effects (AEs) of chemotherapy. ICIs plus anti-angiogenic agents regimen improves anti-tumor activity and safety and is expected to be the new paradigm for the treatment of advanced NSCLC. Despite some limitations, these agents have achieved better overall survival rates. In this article, we review the current status and progress of research on ICIs in NSCLC in recent years, aiming to better guide the individualized treatment of NSCLC patients.
Collapse
Affiliation(s)
- Fuli Wang
- Department of Oncology, Lianyungang Clinical College Affiliated to Bengbu Medical College, Lianyungang, China
- Department of Oncology, Gaochun Hospital Afliated to Jiangsu University, Nanjing, China
| | - Teng Xia
- Department of Oncology, Gaochun Hospital Afliated to Jiangsu University, Nanjing, China
| | - Zhiqiang Li
- Department of Oncology, Lianyungang Clinical College Affiliated to Bengbu Medical College, Lianyungang, China
| | - Xuzhu Gao
- Department of Oncology, Lianyungang Clinical College Affiliated to Bengbu Medical College, Lianyungang, China
- Department of Oncology, Gaochun Hospital Afliated to Jiangsu University, Nanjing, China
| | - Xinjian Fang
- Department of Oncology, Lianyungang Clinical College Affiliated to Bengbu Medical College, Lianyungang, China
- Department of Oncology, Gaochun Hospital Afliated to Jiangsu University, Nanjing, China
| |
Collapse
|
13
|
Tang L, Zhang Z, Fan J, Xu J, Xiong J, Tang L, Jiang Y, Zhang S, Zhang G, Luo W, Xu Y. Comprehensively analysis of immunophenotyping signature in triple-negative breast cancer patients based on machine learning. Front Pharmacol 2023; 14:1195864. [PMID: 37426809 PMCID: PMC10328722 DOI: 10.3389/fphar.2023.1195864] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 06/05/2023] [Indexed: 07/11/2023] Open
Abstract
Immunotherapy is a promising strategy for triple-negative breast cancer (TNBC) patients, however, the overall survival (OS) of 5-years is still not satisfactory. Hence, developing more valuable prognostic signature is urgently needed for clinical practice. This study established and verified an effective risk model based on machine learning methods through a series of publicly available datasets. Furthermore, the correlation between risk signature and chemotherapy drug sensitivity were also performed. The findings showed that comprehensive immune typing is highly effective and accurate in assessing prognosis of TNBC patients. Analysis showed that IL18R1, BTN3A1, CD160, CD226, IL12B, GNLY and PDCD1LG2 are key genes that may affect immune typing of TNBC patients. The risk signature plays a robust ability in prognosis prediction compared with other clinicopathological features in TNBC patients. In addition, the effect of our constructed risk model on immunotherapy response was superior to TIDE results. Finally, high-risk groups were more sensitive to MR-1220, GSK2110183 and temsirolimus, indicating that risk characteristics could predict drug sensitivity in TNBC patients to a certain extent. This study proposes an immunophenotype-based risk assessment model that provides a more accurate prognostic assessment tool for patients with TNBC and also predicts new potential compounds by performing machine learning algorithms.
Collapse
|
14
|
Manso T, Kushwaha A, Abdollahi N, Duroux P, Giudicelli V, Kossida S. Mechanisms of action of monoclonal antibodies in oncology integrated in IMGT/mAb-DB. Front Immunol 2023; 14:1129323. [PMID: 37215135 PMCID: PMC10196129 DOI: 10.3389/fimmu.2023.1129323] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 04/07/2023] [Indexed: 05/24/2023] Open
Abstract
Background Cancer cells activate different immune checkpoint (IC) pathways in order to evade immunosurveillance. Immunotherapies involving ICs either block or stimulate these pathways and enhance the efficiency of the immune system to recognize and attack cancer cells. In this way, the development of monoclonal antibodies (mAbs) targeting ICs has significant success in cancer treatment. Recently, a systematic description of the mechanisms of action (MOA) of the mAbs has been introduced in IMGT/mAb-DB, the IMGT® database dedicated to mAbs for therapeutic applications. The characterization of these antibodies provides a comprehensive understanding of how mAbs work in cancer. Methods In depth biocuration taking advantage of the abundant literature data as well as amino acid sequence analyses from mAbs managed in IMGT/2Dstructure-DB, the IMGT® protein database, allowed to define a standardized and consistent description of the MOA of mAbs targeting immune checkpoints in cancer therapy. Results A fine description and a standardized graphical representation of the MOA of selected mAbs are integrated within IMGT/mAb-DB highlighting two main mechanisms in cancer immunotherapy, either Blocking or Agonist. In both cases, the mAbs enhance cytotoxic T lymphocyte (CTL)-mediated anti-tumor immune response (Immunostimulant effect) against tumor cells. On the one hand, mAbs targeting co-inhibitory receptors may have a functional Fc region to increase anti-tumor activity by effector properties that deplete Treg cells (Fc-effector function effect) or may have limited FcγR binding to prevent Teff cells depletion and reduce adverse events. On the other hand, agonist mAbs targeting co-stimulatory receptors may bind to FcγRs, resulting in antibody crosslinking (FcγR crosslinking effect) and substantial agonism. Conclusion In IMGT/mAb-DB, mAbs for cancer therapy are characterized by their chains, domains and sequence and by several therapeutic metadata, including their MOA. MOAs were recently included as a search criterion to query the database. IMGT® is continuing standardized work to describe the MOA of mAbs targeting additional immune checkpoints and novel molecules in cancer therapy, as well as expanding this study to other clinical domains.
Collapse
|
15
|
Lv J, Jiang Z, Yuan J, Zhuang M, Guan X, Liu H, Yin Y, Ma Y, Liu Z, Wang H, Wang X. Pan-cancer analysis identifies PD-L2 as a tumor promotor in the tumor microenvironment. Front Immunol 2023; 14:1093716. [PMID: 37006239 PMCID: PMC10060638 DOI: 10.3389/fimmu.2023.1093716] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/19/2023] [Indexed: 03/18/2023] Open
Abstract
BACKGROUND Programmed cell death protein 1 (PD-1) receptor has two ligands,programmed death-ligand 1 (PD-L1) and PD-L2. When compared with PD-L1, PD-L2 has not received much attention, and its role remains unclear. METHODS The expression profiles of pdcd1lg2 (PD-L2-encoding gene) mRNA and PD-L2 protein were analyzed using TCGA, ICGC, and HPA databases. Kaplan-Meier and Cox regression analyses were used to assess the prognostic significance of PD-L2. We used GSEA, Spearman's correlation analysis and PPI network to explore the biological functions of PD-L2. PD-L2-associated immune cell infiltration was evaluated using the ESTIMATE algorithm and TIMER 2.0. The expressions of PD-L2 in tumor-associated macrophages (TAMs) in human colon cancer samples, and in mice in an immunocompetent syngeneic setting were verified using scRNA-seq datasets, multiplex immunofluorescence staining, and flow cytometry. After fluorescence-activated cell sorting, flow cytometry and qRT-PCR and transwell and colony formation assays were used to evaluate the phenotype and functions of PD-L2+TAMs. Immune checkpoint inhibitors (ICIs) therapy prediction analysis was performed using TIDE and TISMO. Last, a series of targeted small-molecule drugs with promising therapeutic effects were predicted using the GSCA platform. RESULTS PD-L2 was expressed in all the common human cancer types and deteriorated outcomes in multiple cancers. PPI network and Spearman's correlation analysis revealed that PD-L2 was closely associated with many immune molecules. Moreover, both GSEA results of KEGG pathways and GSEA results for Reactome analysis indicated that PD-L2 expression played an important role in cancer immune response. Further analysis showed that PD-L2 expression was strongly associated with the infiltration of immune cells in tumor tissue in almost all cancer types, among which macrophages were the most positively associated with PD-L2 in colon cancer. According to the results mentioned above, we verified the expression of PD-L2 in TAMs in colon cancer and found that PD-L2+TAMs population was not static. Additionally, PD-L2+TAMs exhibited protumor M2 phenotype and increased the migration, invasion, and proliferative capacity of colon cancer cells. Furthermore, PD-L2 had a substantial predictive value for ICIs therapy cohorts. CONCLUSION PD-L2 in the TME, especially expressed on TAMs, could be applied as a potential therapeutic target.
Collapse
Affiliation(s)
- Jingfang Lv
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Jiang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Junhu Yuan
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meng Zhuang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Guan
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hengchang Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yefeng Yin
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiming Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zheng Liu
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongying Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xishan Wang
- Department of Colorectal Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
TRUONG NC, HUYNH NT, PHAM KD, PHAM PV. Roles of cancer stem cells in cancer immune surveillance. MINERVA BIOTECHNOLOGY AND BIOMOLECULAR RESEARCH 2023. [DOI: 10.23736/s2724-542x.23.02944-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
|
17
|
Yang X, Yu Y, Wang Z, Wu P, Su X, Wu Z, Gan J, Zhang D. NOX4 has the potential to be a biomarker associated with colon cancer ferroptosis and immune infiltration based on bioinformatics analysis. Front Oncol 2022; 12:968043. [PMID: 36249057 PMCID: PMC9554470 DOI: 10.3389/fonc.2022.968043] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 09/07/2022] [Indexed: 11/20/2022] Open
Abstract
Background Colon cancer (CC) is a common tumor, but its pathogenesis is still not well understood. Competitive endogenous RNA (ceRNA) theory, ferroptosis and tumor immune infiltration may be the mechanisms of the development of cancer. The purpose of the study is to seek genes connected with both immunity and ferroptosis, and provide important molecular basis for early noninvasive diagnosis and immunotherapy of CC. Methods We extracted messenger RNA (mRNA), microRNA (miRNA), and long noncoding RNA (lncRNA) data of CC from The Cancer Genome Atlas database (TCGA), identified the differentially expressed mRNA (DEmRNA), miRNA (DEmiRNA) and lncRNA (DElncRNA), then constructed a ceRNA network. Venn overlap analysis was used to identify genes associated with immunity and ferroptosis in ceRNA network. The expression and prognosis of target genes were analyzed via Gene Expression Profiling Interactive Analysis (GEPIA) and PrognoScan database, and we analysed the related functions and signaling pathways of target genes by enrichment analysis. The correlation between target genes and tumor immune infiltrating was explored by CIBERSORT and spearman correlation analysis. Finally, the expression of target genes was detected via quantitative reverse transcription-PCR (qRT-PCR) in CC and normal colon tissues. Results Results showed that there were 4 DElncRNA, 4 DEmiRNA and 126 DEmRNA in ceRNA network. NADPH oxidase 4 protein (NOX4) was a DEmRNA associated with immunity and ferroptosis in ceRNA network. NOX4 was highly expressed in CC and connected with unfavourable prognosis. NOX4 was obviously enriched in pathways connected with carcinogenesis and significantly correlated with six kinds of immune cells. Immune checkpoints and NOX4 spearman correlation analysis showed that the expression of NOX4 was positively related to programmed cell death protein 1 (PD-1)-PDCD1, programmed cell death-Ligand 1 (PD-L1)-CD274 and cytotoxic T-lymphocyte-associated protein 4 (CTLA4). Conclusions To conclude, our study suggests that NOX4 is associated with both ferroptosis and tumor immunity, and might be a biomarker associated with the carcinogenesis, prognosis of CC and a potential target of CC immunotherapy.
Collapse
Affiliation(s)
- Xiaoping Yang
- Key Laboratory of Digestive Diseases of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yi Yu
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Zirui Wang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Pingfan Wu
- Department of Pathology, The 940th Hospital of the Joint Logistic Support of the People’s Liberation Army, Lanzhou, China
| | - Xiaolu Su
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, China
| | - Zhiping Wu
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
| | - Jianxin Gan
- Department of general surgery, Lanzhou University Second Hospital, Lanzhou, China
| | - Dekui Zhang
- Key Laboratory of Digestive Diseases of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
- Department of Gastroenterology, Lanzhou University Second Hospital, Lanzhou, China
- *Correspondence: Dekui Zhang,
| |
Collapse
|
18
|
Yan Y, Feng X, Li C, Lerut T, Li H. Treatments for resectable esophageal cancer: from traditional systemic therapy to immunotherapy. Chin Med J (Engl) 2022; 135:2143-2156. [PMID: 36525602 PMCID: PMC9771193 DOI: 10.1097/cm9.0000000000002371] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Indexed: 12/23/2022] Open
Abstract
ABSTRACT Esophageal cancer (EC) has a high incidence and poor prognosis. The two major histological types, squamous cell carcinoma and adenocarcinoma, differ in their epidemiology and treatment options. Patients with locally advanced EC benefit from multimodal therapy concepts including neoadjuvant chemotherapy, neoadjuvant chemoradiotherapy, and perioperative chemotherapy. Currently, immunotherapy for the solid tumor is a hot spot. Treatment with adjuvant immune checkpoint inhibitors (ICIs) is the first immunotherapy for resectable EC listed in the latest National Comprehensive Cancer Network Guidelines for the Esophageal and Esophagogastric Junction Cancers. Recent clinical trials have established ICIs for three treatment models of resectable EC. Their short-term results demonstrated ideal efficacy and tolerable toxicity, though some concerns remain. This review summarizes the novel data on the ICIs for resectable EC and lists the registered related clinical trials. Hopefully, this review can provide a reference for ongoing research on the treatment options for resectable EC.
Collapse
Affiliation(s)
- Yan Yan
- Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xijia Feng
- Department of Thoracic Surgery, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chengqiang Li
- Department of Thoracic Surgery, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Toni Lerut
- Department of Thoracic Surgery, University Hospital Gasthuisberg, University of Leuven, Leuven 3000, Belgium
| | - Hecheng Li
- Department of Thoracic Surgery, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| |
Collapse
|
19
|
Yang Z, Simovic MO, Liu B, Burgess MB, Cap AP, DalleLucca JJ, Li Y. Indices of complement activation and coagulation changes in trauma patients. Trauma Surg Acute Care Open 2022; 7:e000927. [PMID: 36117727 PMCID: PMC9476135 DOI: 10.1136/tsaco-2022-000927] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 08/10/2022] [Indexed: 11/26/2022] Open
Abstract
Objectives Early complementopathy and coagulopathy are shown often after trauma. However, the prevalence of any interplay between complement cascade (ComC) and coagulation cascade (CoaC) after trauma remains unclear. This study intended to explore whether complement-coagulation crosstalk exists, which may provide a reliable guide to clinical implications in trauma patients. Methods This single-center cohort study of trauma patients enrolled 100 patients along with 20 healthy volunteers. Blood samples from patients were collected at admission, 45, 90, 135 minutes, and 18 hours after admission. Demographic characteristics were recorded, blood levels of ComC and CoaC factors, and inflammatory cytokines were measured by ELISA, clot-based assays, or luminex multiplex assay, and partial thromboplastin (PT) and partial thromboplastin time (PTT) were assessed using a Behring blood coagulation system. Results Compared with the healthy controls, plasma levels of complement factors (C5b-9 and Bb) and 11 tested inflammatory cytokines increased in moderately and severely injured patients as early as 45 minutes after admission and sustained higher levels up to 18 hours after admission. C5b-9 correlated positively to patients’ hospital stay. In parallel, the consumption of coagulation factors I, II, X, and XIII was shown throughout the first 18 hours after admission in moderately and severely injured patients, whereas PT, PTT, D-dimer, factor VII, and factor VIII values significantly increased from the admission to 135 minutes in moderately and severely injured patients. Along with an inverse correlation between plasma Bb, factors I and II, a positive correlation between C5b-9, Bb, D-dimer, PT, and PTT was evident. Conclusions This study demonstrates trauma-induced early activation of plasma cascades including ComC, CoaC, and fibrinolytic cascade, and their correlation between plasma cascades in severe trauma patients. Our study suggests that the simultaneous modulation of plasma cascades might benefit clinical outcomes for trauma patients. Level of evidence Prospective study, level III.
Collapse
Affiliation(s)
- Zhangsheng Yang
- Department of Organ Function Support, US Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Milomir O Simovic
- Department of Organ Function Support, US Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Bin Liu
- Department of Organ Function Support, US Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Matthew B Burgess
- Department of Organ Function Support, US Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | - Andrew P Cap
- Department of Organ Function Support, US Army Institute of Surgical Research, Fort Sam Houston, Texas, USA
| | | | - Yansong Li
- Department of Organ Function Support, US Army Institute of Surgical Research, Fort Sam Houston, Texas, USA.,Trauma Research, UTHSCSA, San Antonio, Texas, USA.,Geneva Foundation, Tacoma, Washington, USA
| |
Collapse
|
20
|
Development of a human phage display-derived anti-PD-1 scFv antibody: an attractive tool for immune checkpoint therapy. BMC Biotechnol 2022; 22:22. [PMID: 35996120 PMCID: PMC9396865 DOI: 10.1186/s12896-022-00752-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/18/2022] [Indexed: 12/13/2022] Open
Abstract
Background The PD-1 checkpoint pathway plays a major role in tumor immune evasion and the development of the tumor microenvironment. Clinical studies show that therapeutic antibodies blocking the PD-1 pathway can restore anti-tumor or anti-virus immune responses by the reinvigoration of exhausted T cells. Because of the promising results of anti-PD-1 monoclonal antibodies in cancer treatment, autoimmune disorders, and infectious diseases, the PD-1 has emerged as an encouraging target for different diseases. Results In the present study, we employed a human semi-synthetic phage library for isolation of some scFvs against the extracellular domain of PD-1 protein by panning process. After the panning, a novel anti-PD-1 scFv (SS107) was found that exhibited specific binding to PD-1 antigen and stimulated Jurkat T cells. The selected anti-PD-1 scFv could restore the production of IL-2 and IFN-γ by Jurkat T cells that were co-cultured with PD-L1 positive tumor cells. Conclusion This anti-PD-1 scFv with high specificity and the ability to reactivate exhausted T cells has the potential to be developed as an anti-cancer agent or to be used in combination with other therapeutic approaches.
Supplementary Information The online version contains supplementary material available at 10.1186/s12896-022-00752-8.
Collapse
|
21
|
Talaat IM, Elemam NM, Zaher S, Saber-Ayad M. Checkpoint molecules on infiltrating immune cells in colorectal tumor microenvironment. Front Med (Lausanne) 2022; 9:955599. [PMID: 36072957 PMCID: PMC9441912 DOI: 10.3389/fmed.2022.955599] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 07/29/2022] [Indexed: 11/19/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent cancer types worldwide, with a high mortality rate due to metastasis. The tumor microenvironment (TME) contains multiple interactions between the tumor and the host, thus determining CRC initiation and progression. Various immune cells exist within the TME, such as tumor-infiltrating lymphocytes (TILs), tumor-associated macrophages (TAMs), and tumor-associated neutrophils (TANs). The immunotherapy approach provides novel opportunities to treat solid tumors, especially toward immune checkpoints. Despite the advances in the immunotherapy of CRC, there are still obstacles to successful treatment. In this review, we highlighted the role of these immune cells in CRC, with a particular emphasis on immune checkpoint molecules involved in CRC pathogenesis.
Collapse
Affiliation(s)
- Iman M. Talaat
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Pathology Department, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Noha M. Elemam
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
| | - Shroque Zaher
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Maha Saber-Ayad
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Department of Pharmacology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
22
|
Alfaro Alfaro ÁE, Murillo Castillo B, Cordero García E, Tascón J, Morales AI. Colon Cancer Pharmacogenetics: A Narrative Review. PHARMACY 2022; 10:95. [PMID: 36005935 PMCID: PMC9413567 DOI: 10.3390/pharmacy10040095] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 07/22/2022] [Accepted: 07/27/2022] [Indexed: 12/16/2022] Open
Abstract
Currently, metastatic colon cancer is treated with monotherapeutic regimens such as folinic acid, fluorouracil, and oxaliplatin (FOLFOX), capecitabine and oxaliplatin (CapeOX), and leucovorin, fluorouracil, and irinotecan hydrochloride (FOLFIRI). Other treatments include biological therapies and immunotherapy with drugs such as bevacizumab, panitumumab, cetuximab, and pembrolizumab. After the research, it was found that some mutations make those treatments not as effective in all patients. In this bibliographic review, we investigated the pharmacogenetic explanations for how mutations in the genes coding for rat sarcoma virus (RAS) and rapidly accelerated fibrosarcoma (RAF) reduce the effectiveness of these treatments and allow the continued proliferation of tumors. Furthermore, we note that patients with mutations in the dihydropyrimidine dehydrogenase (DPDY) gene usually require lower doses of therapies such as 5-fluorouracyl (5-FU) and capecitabine to avoid severe adverse effects. Some other mutations in the thymidylate synthase gene (TSYM), methylenetetrahydrofolate reductase gene (MTHFR), and ATP binding cassette transporter B (ABCB1 and ABCB2) affect efficacy and security of the treatments. It is important to address the clinical implication of the oncologist in the study of gene mutations than can influence in the antitumoral response and safety of colon cancer treatments.
Collapse
Affiliation(s)
| | | | | | - Javier Tascón
- Toxicology Unit, Universidad de Salamanca, 37007 Salamanca, Spain
| | - Ana I. Morales
- Toxicology Unit, Universidad de Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
23
|
Williams-Hall R, Berry P, Williamson N, Barclay M, Roberts A, Gater A, Tolley C, Bradley H, Ward A, Hsia E, Zuraw Q, DeLong P, Touma Z, Strand V. Generation of evidence supporting the content validity of SF-36, FACIT-F, and LupusQoL, and novel patient-reported symptom items for use in patients with systemic lupus erythematosus (SLE) and SLE with lupus nephritis (LN). Lupus Sci Med 2022; 9:e000712. [PMID: 36007978 PMCID: PMC9422858 DOI: 10.1136/lupus-2022-000712] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/01/2022] [Indexed: 11/03/2022]
Abstract
OBJECTIVE SLE and lupus nephritis (LN) have significant impacts on the health-related quality of life of patients living with the condition, which are important to capture from the patient's perspective using patient-reported outcomes (PROs). The objectives of this study were to evaluate the content validity of PROs commonly used in SLE and LN (36-Item Short Form Health Survey (SF-36), Functional Assessment of Chronic Illness Therapy-Fatigue (FACIT-F) and Lupus Quality of Life (LupusQoL), as well as novel PRO symptom severity items measuring skin rash, joint pain, joint stiffness and swelling of the legs and/or feet, in both populations. METHODS Qualitative, semi-structured, cognitive interviews were conducted with 48 participants (SLE=28, LN=20). Understanding and relevance of symptom and impact PRO concepts from existing PROs were assessed, alongside novel PRO symptom severity items with different recall periods (24 hours vs 7 days) and response scales (Numerical Rating Scale (NRS) vs Verbal Rating Scale). Interviews were conducted in multiple rounds to allow for modifications to the novel PRO items. Analysis of verbatim interview transcripts was performed. RESULTS Symptom and impact concepts assessed by the SF-36, FACIT-F, and LupusQoL were well understood by both participants with SLE and LN (≥90.0%), with most considered relevant by over half of the participants asked (≥51.9%). All participants asked (100%) understood the novel PRO symptom severity items, and the majority (≥90.0%) considered the symptoms relevant. Minor modifications to the novel PRO items were made between rounds to improve clarity based on participant feedback. The selected 7-day recall period and NRS in the final iteration of the PRO items were understood and relevant. No differences in interview findings between the SLE and LN samples were identified. CONCLUSIONS Findings provide evidence of content validity for concepts assessed by the SF-36, FACIT-F, LupusQoL and the novel PRO symptom severity items, supporting use of these PROs to comprehensively assess disease impact in future SLE and LN clinical trials.
Collapse
Affiliation(s)
| | - Pamela Berry
- Patient Reported Outcomes, Janssen Global Services LLC, Titusville, Florida, USA
| | | | | | - Anna Roberts
- Patient-Centered Outcomes, Adelphi Values, Bollington, UK
| | - Adam Gater
- Patient-Centered Outcomes, Adelphi Values, Bollington, UK
| | - Chloe Tolley
- Patient-Centered Outcomes, Adelphi Values, Bollington, UK
| | - Helena Bradley
- Patient-Centered Outcomes, Adelphi Values, Bollington, UK
| | - Amy Ward
- Patient-Centered Outcomes, Adelphi Values, Bollington, UK
| | - Elizabeth Hsia
- Immunology Clinical Development, Janssen Research & Development LLC, Spring House, Pennsylvania, USA
- Division of Rheumatology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Qing Zuraw
- Immunology Clinical Development, Janssen Research & Development LLC, Spring House, Pennsylvania, USA
| | - Patricia DeLong
- Patient Reported Outcomes, Janssen Global Services LLC, Titusville, Florida, USA
| | - Zahi Touma
- Division of Rheumatology, University of Toronto, Toronto, Ontario, Canada
| | - Vibeke Strand
- Division of Immunology/Rheumatology, Stanford University, Palo Alto, California, USA
| |
Collapse
|
24
|
Wang Q, He Y, Li W, Xu X, Hu Q, Bian Z, Xu A, Tu H, Wu M, Wu X. Soluble Immune Checkpoint-Related Proteins in Blood Are Associated With Invasion and Progression in Non-Small Cell Lung Cancer. Front Immunol 2022; 13:887916. [PMID: 35874720 PMCID: PMC9296827 DOI: 10.3389/fimmu.2022.887916] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundImmune checkpoint inhibition therapy has been achieved significant success in the treatment of non-small cell lung cancer (NSCLC). However, the role of soluble immune checkpoint- related proteins in NSCLC remains obscure.MethodsWe evaluated the circulating levels of 14 immune checkpoint-related proteins panel (BTLA, LAG-3, GITR, IDO, PD-L2, PD-L1, PD-1, HVEM, Tim-3, CD28, CD27, CD80, CD137 and CTLA-4) and their associations with the risk of invasive disease and the risk of NSCLC in 43 pre-invasive (AIS), 81 invasive NSCLC (IAC) patients and matched 35 healthy donors using a multiplex Luminex assay. Gene expression in tumors from TCGA were analyzed to elucidate potential mechanisms. The multivariate logistic regression model was applied in the study. ROC(receiver operator characteristic) curve and calibration curve were used in the performance evaluation.ResultsWe found that sCD27, sCD80, CD137 and sPDL2 levels were significantly increased in IAC cases compared to AIS cases (P= 1.05E-06, 4.44E-05, 2.30E-05 and 1.16E-06, respectively), whereas sPDL1 and sPDL2 levels were significantly increased in NSCLC cases compared to healthy controls (P=3.25E-05 and 1.49E-05, respectively). Unconditional univariate logistic regression analysis indicated that increased sCD27, sCD80, sCD137, and sPDL2 were significantly correlated with the risk of invasive diseases. The model with clinical variables, sCD27 and sPDL2 demonstrated the best performance (AUC=0.845) in predicting the risk of IAC. CD27 and PDCD1LG2 (PDL2) showed significant association with cancer invasion signature in TCGA dataset.ConclusionOur study provides evidence that soluble immune checkpoint-related proteins may associate with the risk of IAC, and we further established an optimized multivariate predictive model, which highlights their potential application in the treatment of NSCLC patients. Future studies may apply these biomarkers to test their predictive value of survival and treatment outcome during immunotherapy in NSCLC patients.
Collapse
Affiliation(s)
- Qinchuan Wang
- Center for Biostatistics, Bioinformatics and Big Data, The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- Department of Surgical Oncology, Affiliated Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Yue He
- Center for Biostatistics, Bioinformatics and Big Data, The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Wanlu Li
- Center for Biostatistics, Bioinformatics and Big Data, The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Xiaohang Xu
- Center for Biostatistics, Bioinformatics and Big Data, The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Qingfeng Hu
- Center for Biostatistics, Bioinformatics and Big Data, The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Zilong Bian
- Center for Biostatistics, Bioinformatics and Big Data, The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Andi Xu
- Center for Biostatistics, Bioinformatics and Big Data, The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Huakang Tu
- Center for Biostatistics, Bioinformatics and Big Data, The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
| | - Ming Wu
- Department of Thoracic Surgery, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- *Correspondence: Xifeng Wu, ; Ming Wu,
| | - Xifeng Wu
- Center for Biostatistics, Bioinformatics and Big Data, The Second Affiliated Hospital and School of Public Health, Zhejiang University School of Medicine, Hangzhou, China
- The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Hangzhou, China
- *Correspondence: Xifeng Wu, ; Ming Wu,
| |
Collapse
|
25
|
Nguyen TT, Shin DH, Sohoni S, Singh SK, Rivera-Molina Y, Jiang H, Fan X, Gumin J, Lang FF, Alvarez-Breckenridge C, Godoy-Vitorino F, Zhu L, Zheng WJ, Zhai L, Ladomersky E, Lauing KL, Alonso MM, Wainwright DA, Gomez-Manzano C, Fueyo J. Reshaping the tumor microenvironment with oncolytic viruses, positive regulation of the immune synapse, and blockade of the immunosuppressive oncometabolic circuitry. J Immunother Cancer 2022; 10:e004935. [PMID: 35902132 PMCID: PMC9341188 DOI: 10.1136/jitc-2022-004935] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Oncolytic viruses are considered part of immunotherapy and have shown promise in preclinical experiments and clinical trials. Results from these studies have suggested that tumor microenvironment remodeling is required to achieve an effective response in solid tumors. Here, we assess the extent to which targeting specific mechanisms underlying the immunosuppressive tumor microenvironment optimizes viroimmunotherapy. METHODS We used RNA-seq analyses to analyze the transcriptome, and validated the results using Q-PCR, flow cytometry, and immunofluorescence. Viral activity was analyzed by replication assays and viral titration. Kyn and Trp metabolite levels were quantified using liquid chromatography-mass spectrometry. Aryl hydrocarbon receptor (AhR) activation was analyzed by examination of promoter activity. Therapeutic efficacy was assessed by tumor histopathology and survival in syngeneic murine models of gliomas, including Indoleamine 2,3-dioxygenase (IDO)-/- mice. Flow cytometry was used for immunophenotyping and quantification of cell populations. Immune activation was examined in co-cultures of immune and cancer cells. T-cell depletion was used to identify the role played by specific cell populations. Rechallenge experiments were performed to identify the development of anti-tumor memory. RESULTS Bulk RNA-seq analyses showed the activation of the immunosuppressive IDO-kynurenine-AhR circuitry in response to Delta-24-RGDOX infection of tumors. To overcome the effect of this pivotal pathway, we combined Delta-24-RGDOX with clinically relevant IDO inhibitors. The combination therapy increased the frequency of CD8+ T cells and decreased the rate of myeloid-derived suppressor cell and immunosupressive Treg tumor populations in animal models of solid tumors. Functional studies demonstrated that IDO-blockade-dependent activation of immune cells against tumor antigens could be reversed by the oncometabolite kynurenine. The concurrent targeting of the effectors and suppressors of the tumor immune landscape significantly prolonged the survival in animal models of orthotopic gliomas. CONCLUSIONS Our data identified for the first time the in vivo role of IDO-dependent immunosuppressive pathways in the resistance of solid tumors to oncolytic adenoviruses. Specifically, the IDO-Kyn-AhR activity was responsible for the resurface of local immunosuppression and resistance to therapy, which was ablated through IDO inhibition. Our data indicate that combined molecular and immune therapy may improve outcomes in human gliomas and other cancers treated with virotherapy.
Collapse
Affiliation(s)
- Teresa T Nguyen
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Dong Ho Shin
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Sagar Sohoni
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sanjay K Singh
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yisel Rivera-Molina
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hong Jiang
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xuejun Fan
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Joy Gumin
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Filipa Godoy-Vitorino
- Department of Microbiology and Medical Zoology, University of Puerto Rico School of Medicine, San Juan, Puerto Rico
| | - Lisha Zhu
- The University of Texas Health Science Center at Houston School of Biomedical Informatics, Houston, Texas, USA
| | - W Jim Zheng
- The University of Texas Health Science Center at Houston School of Biomedical Informatics, Houston, Texas, USA
| | - Lijie Zhai
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Erik Ladomersky
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Kristen L Lauing
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Marta M Alonso
- Pediatrics, Clínica Universidad de Navarra, Pamplona, Spain
- Program of Solid Tumors, CIMA, Pamplona, Spain
| | - Derek A Wainwright
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Medicine-Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Candelaria Gomez-Manzano
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| | - Juan Fueyo
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, Texas, USA
| |
Collapse
|
26
|
Tissue and circulating PD-L2: moving from health and immune-mediated diseases to head and neck oncology. Crit Rev Oncol Hematol 2022; 175:103707. [PMID: 35569724 DOI: 10.1016/j.critrevonc.2022.103707] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 05/04/2022] [Accepted: 05/09/2022] [Indexed: 12/21/2022] Open
Abstract
Amongst the chief targets of immune-checkpoint inhibitors (ICIs), namely the Programmed cell death protein 1 (PD-1)/PD-Ligands (Ls) axis, most research has focused on PD-L1, while to date PD-L2 is still under-investigated. However, emerging data support PD-L2 relevant expression in malignancies of the head and neck area, mostly in head and neck squamous cell carcinoma (HNSCC) and salivary gland cancers (SGCs). In this context, ICIs have achieved highly heterogeneous outcomes, emphasizing an urgent need for the identification of predictive biomarkers. With the present review, we aimed at describing PD-L2 biological significance by focusing on its tissue expression, its binding to PD-1 and RGMb receptors, and its impact on physiological and anti-cancer immune response. Specifically, we reported PD-L2 expression rates and significant clinical correlates among different head and neck cancer histotypes. Finally, we described the biology of soluble PD-L2 form and its potential application as a prognostic and/or predictive circulating biomarker.
Collapse
|
27
|
Fan JY, Huang Y, Li Y, Muluh TA, Fu SZ, Wu JB. Bacteria in cancer therapy: A new generation of weapons. Cancer Med 2022; 11:4457-4468. [PMID: 35522104 PMCID: PMC9741989 DOI: 10.1002/cam4.4799] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 12/15/2022] Open
Abstract
Tumors are presently a major threat to human life and health. Malignant tumors are conventionally treated through radiotherapy and chemotherapy. However, traditional therapies yield unsatisfactory results due to high toxicity to the normal cells, inability to treat deep tumor tissues, and the possibility of inducing drug resistance in the tumor cells. This has caused immunotherapy to emerge as an effective and alternate treatment strategy. To overcome the limitations of the conventional treatments as well as to avert the risk of various drug resistance and cytotoxicity, bacterial anti-tumor immunotherapy has raised the interest of researchers. This therapeutic strategy employs bacteria to specifically target and colonize the tumor tissues with preferential accumulation and proliferation. Such bacterial accumulation initiates a series of anti-tumor immune responses, effectively eliminating the tumor cells. This immunotherapy can use the bacteria alone or concomitantly with the other methods. For example, the bacteria can deliver the anti-cancer effect mediators by regulating the expression of the bacterial genes or by synthesizing the bioengineered bacterial complexes. This review will discuss the mechanism of utilizing bacteria in treating tumors, especially in terms of immune mechanisms. This could help in better integrating the bacterial method with other treatment options, thereby, providing a more effective, reliable, and unique treatment therapy for tumors.
Collapse
Affiliation(s)
- Jun Ying Fan
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China
| | - Yuan Huang
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China
| | - Yi Li
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China
| | - Tobias Achu Muluh
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China
| | - Shao Zhi Fu
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China,Department of Nuclear MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China
| | - Jing Bo Wu
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China,Department of Nuclear MedicineThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuanP.R. China,Academician (Expert) Workstation of Sichuan ProvinceLuzhouSichuanP.R. China
| |
Collapse
|
28
|
Sasikumar PG, Ramachandra M. Small Molecule Agents Targeting PD-1 Checkpoint Pathway for Cancer Immunotherapy: Mechanisms of Action and Other Considerations for Their Advanced Development. Front Immunol 2022; 13:752065. [PMID: 35585982 PMCID: PMC9108255 DOI: 10.3389/fimmu.2022.752065] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 03/29/2022] [Indexed: 12/20/2022] Open
Abstract
Pioneering success of antibodies targeting immune checkpoints such as programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) has changed the outlook of cancer therapy. Although these antibodies show impressive durable clinical activity, low response rates and immune-related adverse events are becoming increasingly evident in antibody-based approaches. For further strides in cancer immunotherapy, novel treatment strategies including combination therapies and alternate therapeutic modalities are highly warranted. Towards this discovery and development of small molecule, checkpoint inhibitors are actively being pursued, and the efforts have culminated in the ongoing clinical testing of orally bioavailable checkpoint inhibitors. This review focuses on the small molecule agents targeting PD-1 checkpoint pathway for cancer immunotherapy and highlights various chemotypes/scaffolds and their characterization including binding and functionality along with reported mechanism of action. The learnings from the ongoing small molecule clinical trials and crucial points to be considered for their clinical development are also discussed.
Collapse
|
29
|
Batchu S, Hakim A, Henry OS, Madzo J, Atabek U, Spitz FR, Hong YK. Transcriptome-guided resolution of tumor microenvironment interactions in pheochromocytoma and paraganglioma subtypes. J Endocrinol Invest 2022; 45:989-998. [PMID: 35088383 DOI: 10.1007/s40618-021-01729-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 12/19/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Pheochromocytomas and paragangliomas (PCPG) are rare catecholamine-secreting endocrine tumors deriving from chromaffin cells of the embryonic neural crest. Although distinct molecular PCPG subtypes have been elucidated, certain characteristics of these tumors have yet to be fully examined, namely the tumor microenvironment (TME). To further understand tumor-stromal interactions in PCPG subtypes, the present study deconvoluted bulk tumor gene expression to examine ligand-receptor interactions. METHODS RNA-sequencing data primary solid PCPG tumors were derived from The Cancer Genome Atlas (TCGA). Tumor purity was estimated using two robust algorithms. The tumor purity estimates and bulk tumor expression values allowed for non-negative linear regression to predict the average expression of each gene in the stromal and tumor compartments for each PCPG molecular subtype. The predicted expression values were then used in conjunction with a previously curated ligand-receptor database and scoring system to evaluate top ligand-receptor interactions. RESULTS Across all PCPG subtypes compared to normal samples, tumor-to-tumor signaling between bone morphogenic proteins 7 (BMP7) and 15 (BMP15) and cognate receptors ACVR2B and BMPR1B was increased. In addition, tumor-to-stroma signaling was enriched for interactions between predicted tumor-originating delta-like ligand 3 (DLL3) and predicted stromal NOTCH receptors. Stroma-to-tumor signaling was enriched for interactions between ephrins A1 and A4 with ephrin receptors EphA5, EphA7, and EphA8. Pseudohypoxia subtype tumors displayed increased predicted stromal expression of genes related to immune-exhausted T-cell response, including those for inhibitory receptors HAVCR2 and CTLA4. CONCLUSION The current exploratory study predicted stromal and tumor through compartmental deconvolution and yielded previously unrecognized interactions and putative biomarkers in PCPG.
Collapse
Affiliation(s)
- S Batchu
- Cooper Medical School at Rowan University, 401 Broadway, Camden, NJ, 08103, USA.
| | - A Hakim
- Department of Surgery, Cooper University Hospital, Camden, NJ, USA
| | - O S Henry
- Cooper Medical School at Rowan University, 401 Broadway, Camden, NJ, 08103, USA
| | - J Madzo
- Coriell Institute, Camden, NJ, USA
| | - U Atabek
- Department of Surgery, Cooper University Hospital, Camden, NJ, USA
| | - F R Spitz
- Department of Surgery, Cooper University Hospital, Camden, NJ, USA
| | - Y K Hong
- Department of Surgery, Cooper University Hospital, Camden, NJ, USA
| |
Collapse
|
30
|
Zhong Z, Vong CT, Chen F, Tan H, Zhang C, Wang N, Cui L, Wang Y, Feng Y. Immunomodulatory potential of natural products from herbal medicines as immune checkpoints inhibitors: Helping to fight against cancer via multiple targets. Med Res Rev 2022; 42:1246-1279. [PMID: 35028953 PMCID: PMC9306614 DOI: 10.1002/med.21876] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 12/03/2021] [Accepted: 12/15/2021] [Indexed: 12/19/2022]
Abstract
Immunotherapy sheds new light to cancer treatment and is satisfied by cancer patients. However, immunotoxicity, single-source antibodies, and single-targeting stratege are potential challenges to the success of cancer immunotherapy. A huge number of promising lead compounds for cancer treatment are of natural origin from herbal medicines. The application of natural products from herbal medicines that have immunomodulatory properties could alter the landscape of immunotherapy drastically. The present study summarizes current medication for cancer immunotherapy and discusses the potential chemicals from herbal medicines as immune checkpoint inhibitors that have a broad range of immunomodulatory effects. Therefore, this review provides valuable insights into the efficacy and mechanism of actions of cancer immunotherapies, including natural products and combined treatment with immune checkpoint inhibitors, which could confer an improved clinical outcome for cancer treatment.
Collapse
Affiliation(s)
- Zhangfeng Zhong
- School of Chinese MedicineThe University of Hong KongPokfulamHong KongChina
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauTaipaMacao SARChina
| | - Chi Teng Vong
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauTaipaMacao SARChina
| | - Feiyu Chen
- School of Chinese MedicineThe University of Hong KongPokfulamHong KongChina
| | - Horyue Tan
- School of Chinese MedicineThe University of Hong KongPokfulamHong KongChina
| | - Cheng Zhang
- School of Chinese MedicineThe University of Hong KongPokfulamHong KongChina
| | - Ning Wang
- School of Chinese MedicineThe University of Hong KongPokfulamHong KongChina
| | - Liao Cui
- Guangdong Key Laboratory for Research and Development of Natural DrugsGuangdong Medical UniversityZhanjiangGuangdongChina
| | - Yitao Wang
- Macau Centre for Research and Development in Chinese Medicine, Institute of Chinese Medical SciencesUniversity of MacauTaipaMacao SARChina
| | - Yibin Feng
- School of Chinese MedicineThe University of Hong KongPokfulamHong KongChina
| |
Collapse
|
31
|
Ok Atılgan A, Yılmaz Akçay E, Özen Ö, Haberal Reyhan AN, Ayhan A. The Overexpression of Programmed Death-Ligand 2 in Uterine Adenosarcoma: Correlation with High-Grade Morphology, Mutant Type TP53 Expression and Clinical Outcomes. Int J Surg Pathol 2022; 31:352-364. [PMID: 35466759 DOI: 10.1177/10668969221095189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Immunotherapy involving the programmed death-1 (PD-1)/the programmed death-ligand (PD-1/PD-L) blockade is an understudied tumor therapy approach in cases of adenosarcoma. PD-L1 and PD-L2, and tumor protein p53 (p53) were examined in 20 uterine adenosarcoma cases, and tumor-infiltrating lymphocytes and tumor-associated macrophages were counted in tumor tissue using immunohistochemistry. While CPS PD-L1 positivity with 1% and 10% cut-off values was observed in 40% and 10% of tumors, respectively, CPS PD-L2 positivity with 1%, 10% and 50% cut-off values was observed in 100%, 85% and 50% of the tumors, respectively. The CPS PD-L2 positivity with a 50% cut-off value was positively correlated with tumor grade and the presence of sarcomatous overgrowth and lymphovascular invasion (LVI) (p = 0.025, p = 0.025, and p = 0.025, respectively). Nine of 11 high-grade adenosarcomas and none of the low-grade adenosarcomas showed mutant type p53 expression (p = 0.000). However, PD-L1 expression and tumor-infiltrating immune cells did not correlate with clinicopathological parameters. The CPS PD-L2 positivity with a 50% cut-off value was also positively correlated with mutant type p53 expression (p = 0.024) and tumor-associated macrophages density (p = 0.024). The CPS PD-L2 positivity with a 50% cut-off value and mutant type p53 expression were associated with shorter disease-free survival and shorter overall survival. The high density of tumor-associated macrophages and low density of tumor-infiltrating lymphocytes were also associated with shorter disease-free survival and overall survival (p < 0.05).These results suggested that the CPS PD-L2 positivity with a 50% cut-off value, p53 mutation and tumor microenvironment played an essential role in the progression of uterine adenosarcomas.
Collapse
Affiliation(s)
- Alev Ok Atılgan
- Department of Pathology, Faculty of Medicine, Baskent University, Bahcelievler, Ankara, Turkey
| | - Eda Yılmaz Akçay
- Department of Pathology, Faculty of Medicine, Baskent University, Bahcelievler, Ankara, Turkey
| | - Özlem Özen
- Department of Pathology, Faculty of Medicine, Baskent University, Bahcelievler, Ankara, Turkey
| | - A. Nihan Haberal Reyhan
- Department of Pathology, Faculty of Medicine, Baskent University, Bahcelievler, Ankara, Turkey
| | - Ali Ayhan
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Baskent University, Faculty of Medicine, Bahcelievler, Ankara, Turkey
| |
Collapse
|
32
|
Ehtesham M, Fortune K, Shabbir MA, Peredo-Wende R. Sjogren syndrome presenting as atrioventricular block in an adult. BMJ Case Rep 2022; 15:e247337. [PMID: 35396234 PMCID: PMC8996011 DOI: 10.1136/bcr-2021-247337] [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] [Accepted: 04/01/2022] [Indexed: 11/03/2022] Open
Abstract
A woman in her late teens with a history of Juvenile idiopathic arthritis (JIA) and ongoing sicca symptoms presented with syncope. Upon admission, she was found to be bradycardic with a second-degree atrioventricular (AV) block. After infectious, structural and metabolic aetiologies had been ruled out, she was worked up for rheumatologic causes.Our patient had elevated titres of anti-Sjogren syndrome (SS) antibodies anti-Ro antibodies and was diagnosed with AV block secondary to SS. She was treated with a permanent pacemaker. Patient was followed up in clinic where she denied further syncopal episodes and was started on secretagogues for sicca symptoms.
Collapse
Affiliation(s)
- Moiz Ehtesham
- Internal Medicine, Albany Medical Center Hospital, Albany, New York, USA
| | - Kathryn Fortune
- Internal Medicine, Albany Medical Center Hospital, Albany, New York, USA
| | | | | |
Collapse
|
33
|
Takamochi K, Hara K, Hayashi T, Kohsaka S, Takahashi F, Suehara Y, Shimokawa M, Suzuki K. Clinical relevance of PD-L2 expression in surgically resected lung adenocarcinoma. Lung Cancer 2022; 168:50-58. [DOI: 10.1016/j.lungcan.2022.04.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/13/2022] [Accepted: 04/20/2022] [Indexed: 01/22/2023]
|
34
|
Abdel-Salam LO, El Hanbuli H, Abdelhafez DN. Tumoral and Stromal Pdl1 and Pdl2 Checkpoints Immunohistochemical Expression in Pancreatic Ductal Adenocarcinoma, a Promising Field Of Study. Open Access Maced J Med Sci 2022. [DOI: 10.3889/oamjms.2022.9070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is world-widely considered as one of the most malignant tumors. Programmed cell death protein 1 (PD-1), via its ligands PDL1 and PDL2 plays a critical role in cancer immunoediting. The ligands are expressed in many solid tumors and there is an emerging hope of using anti-PDL in cancer immunotherapy.
Material and methods:
This study included 40 patients with PDAC who underwent pancreaticoduodenectomy. PDL1 and PDL2 pancreatic expression were evaluated in these patients using immunohistochemical staining and correlated their expression levels with each patient’s reported clinicopathological features.
Results:
There were significant relations between high tumoral PDL1 expression and the PDAC tumor histologic grade (p= 0.021) and the tumor status (T) (p= 0.022), while the stromal expression of PDL1 showed non-significant relation with any of the studied features. There were significant relations between high tumoral PDL2 expression and tumor stage (p=0.012), while the stromal expression of PDL2 showed significant relation with tumor status, lymph node status, tumor stage and the presence lympho-vascular invasion with P value equal 0.001, 0.009, 0.009, 0.045 respectively.
Conclusion:
This study showed that in PDAC patients high tumoral PDL1 and PDL2 expression was associated with some important prognostic factors, while only stromal PDL2 expression was significantly associated with most of the studied prognostic features emphasizing a role of both markers in the prognosis of this neoplasm.
Collapse
|
35
|
Zhu D, Wu S, Li Y, Zhang Y, Chen J, Ma J, Cao L, Lyu Z, Hou T. Ferroptosis-related gene SLC1A5 is a novel prognostic biomarker and correlates with immune infiltrates in stomach adenocarcinoma. Cancer Cell Int 2022; 22:124. [PMID: 35305616 PMCID: PMC8933927 DOI: 10.1186/s12935-022-02544-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 03/07/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Stomach adenocarcinoma (STAD) is associated with high morbidity and mortality rates. Ferroptosis is an iron-dependent form of cell death, which plays an important role in the development of many cancers. Tumor-associated competing endogenous RNAs (ceRNAs) regulate tumorigenesis and development. Our study aimed to construct ceRNA networks and explore the relationship between ferroptosis-related genes in the ceRNA network and immune infiltration in STAD. METHODS Based on the interactions among long noncoding RNAs (lncRNAs), microRNAs (miRNAs), and messenger RNAs (mRNAs), a ceRNA network was constructed to illustrate the relationships among lncRNAs, miRNAs, and mRNAs. Subsequently, gene ontology (GO) and Kyoto encyclopedia of genes and genomes (KEGG) functional enrichment analyses were carried out to explore the functions and interactions of the differentially expressed (DE) mRNAs related to the ceRNA network. Differential expression and prognostic analysis of ferroptosis-related genes in the ceRNA network were performed using the R package "limma" and "survminer." The correlation between ferroptosis-related genes and tumor-infiltrating immune cells was analyzed using Spearman correlation analysis and CIBERSORT. Quantitative real-time PCR (qRT-PCR) was used to validate the expression of ferroptosis-related genes in STAD cells lines. RESULTS A ceRNA network consisting of 29 DElncRNAs, 31 DEmiRNAs, and 182 DEmRNAs was constructed. These DEmRNAs were significantly enriched in pathways related to the occurrence and development of STAD. The ferroptosis-related gene SLC1A5 was upregulated in STAD (P < 0.001) and was associated with better prognosis (P = 0.049). The CIBERSORT database and Spearman correlation analysis indicated that SLC1A5 was correlated with eight types of tumor-infiltrating immune cells and immune checkpoints, including PD-L1(CD-274) and PD-1(PDCD1). The SLC1A5 mRNA was found to be highly expressed in STAD cells lines. CONCLUSIONS Our study provides insights into the function of ceRNAs in STAD and identifies biomarkers for the development of therapies for STAD. The ferroptosis-related gene SLC1A5 in the ceRNA network was associated with both tumor-infiltrating immune cells and immune checkpoints in the tumor microenvironment, suggesting that SLC1A5 may be a novel prognostic marker and a potential target for STAD immunotherapy in the future.
Collapse
Affiliation(s)
- Dandan Zhu
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
- Guangdong Clinical Laboratory Center, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Sifan Wu
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
- Department of Laboratory Medicine, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Yafang Li
- Guangdong Clinical Laboratory Center, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
- Department of Laboratory Medicine, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Yu Zhang
- Medical Department, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Jierong Chen
- Department of Laboratory Medicine, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Jianhong Ma
- Department of Laboratory Medicine, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Lixue Cao
- Research Center of Medical Sciences, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China.
| | - Zejian Lyu
- Department of Gastrointestinal Surgery, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China.
| | - Tieying Hou
- School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China.
- Guangdong Clinical Laboratory Center, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- Medical Department, Guangdong Provincial People's Hospital; Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
36
|
Raftopoulou S, Valadez-Cosmes P, Mihalic ZN, Schicho R, Kargl J. Tumor-Mediated Neutrophil Polarization and Therapeutic Implications. Int J Mol Sci 2022; 23:3218. [PMID: 35328639 PMCID: PMC8951452 DOI: 10.3390/ijms23063218] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/25/2022] [Accepted: 03/09/2022] [Indexed: 01/04/2023] Open
Abstract
Neutrophils are immune cells with reported phenotypic and functional plasticity. Tumor-associated neutrophils display many roles during cancer progression. Several tumor microenvironment (TME)-derived factors orchestrate neutrophil release from the bone marrow, recruitment and functional polarization, while simultaneously neutrophils are active stimulators of the TME by secreting factors that affect immune interactions and subsequently tumor progression. Successful immunotherapies for many cancer types and stages depend on the targeting of tumor-infiltrating lymphocytes. Neutrophils impact the success of immunotherapies, such as immune checkpoint blockade therapies, by displaying lymphocyte suppressive properties. The identification and characterization of distinct neutrophil subpopulations or polarization states with pro- and antitumor phenotypes and the identification of the major TME-derived factors of neutrophil polarization would allow us to harness the full potential of neutrophils as complementary targets in anticancer precision therapies.
Collapse
Affiliation(s)
| | | | | | | | - Julia Kargl
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria; (S.R.); (P.V.-C.); (Z.N.M.); (R.S.)
| |
Collapse
|
37
|
Walia HK, Sharma P, Singh N, Sharma S. Immunotherapy in Small Cell Lung Cancer Treatment: a Promising Headway for Future Perspective. Curr Treat Options Oncol 2022; 23:268-294. [PMID: 35226309 DOI: 10.1007/s11864-022-00949-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2022] [Indexed: 12/24/2022]
Abstract
Despite advancements in clinical research, both prognosis and treatment for SCLC patients are still in the nascent stage. SCLC is a fatal disease with high tumor mutational burden and is strongly associated with exposure to tobacco. This leads to the development of potential neo-antigens, inhibition of immune responses, and development of paraneoplastic disorders. Surgery, radiation, and chemotherapy are widely accepted treatments for cancer globally, and most recently, immunotherapy has now become the "fourth pillar" of SCLC treatment. Various immune checkpoint pathways regulate the activation of T cells at multiple stages during an immune response. T cell checkpoint inhibitors such as anti-PD1 (pembrolizumab, nivolumab), anti-PDL1, and anti-CTLA-4 (tremelimumab, ipilimumab) have potential to show anti-cancer activity along with the promise to prolong survival in patients with SCLC. Treatment with the CTLA-4-specific antibodies can restore the immune response by increasing the accumulation and survival of T-cells whereas monoclonal antibodies block either PD-1 or its ligands that prevent downregulation of effector T-cell, which enables the T-cells to mediate the death of tumor cells. Furthermore, monoclonal antibody in combination with chemotherapy has attained quite a focus to enhance the survival of SCLC patients. Apart from this, various immunotherapeutic approaches have been evaluated in the clinical trials for SCLC patients such as TLR9 agonist, anti-CD47, anti-ganglioside therapy, and anti-Notch signaling. The current review focuses on the rationale as well as on the clinical studies of immunotherapy in SCLC along with the clinical end results of certain immunotherapeutic agents and novel therapeutic combinations in SCLC patients.
Collapse
Affiliation(s)
- Harleen Kaur Walia
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, Punjab, 147004, India
| | - Parul Sharma
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, Punjab, 147004, India
| | - Navneet Singh
- Department of Pulmonary Medicine, Post-Graduate Institute of Medical Education & Research, Chandigarh, 160012, India
| | - Siddharth Sharma
- Department of Biotechnology, Thapar Institute of Engineering & Technology, Patiala, Punjab, 147004, India.
| |
Collapse
|
38
|
Expression of Immunomodulatory Checkpoint Molecules in Drug-Resistant Neuroblastoma: An Exploratory Study. Cancers (Basel) 2022; 14:cancers14030751. [PMID: 35159017 PMCID: PMC8833944 DOI: 10.3390/cancers14030751] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 01/27/2023] Open
Abstract
Simple Summary Neuroblastoma is a common childhood cancer with poor prognosis. Prior studies suggest that inhibition of molecules called checkpoint proteins, which normally prevent one’s own immune system from attacking itself, has been successfully used for treatment of multiple advanced adult cancers but has yet to be fully explored in neuroblastoma. Cancer can hijack these pathways to prevent the immune system from recognizing and destroying cancer cells. We investigated checkpoint protein expression in pediatric neuroblastoma and its role in drug resistance. We created drug-resistant neuroblastoma cell lines and compared expression of checkpoint proteins between drug-resistant and parental cell lines. In total, 13 checkpoint proteins were expressed by all cell lines regardless of drug resistance. Although PD-L1 and checkpoint proteins do not necessarily impart drug resistance, they may be potential targets for drug therapy. Benchmarking checkpoint proteins provides the basis for future studies identifying targets for directed therapy and biomarkers for cancer detection or prognosis. Abstract Neuroblastoma is a common childhood cancer with poor prognosis when at its advanced stage. Checkpoint molecule inhibition is successful in treating multiple advanced adult cancers. We investigated PD-L1 and other checkpoint molecule expression to determine their roles in drug resistance and usefulness as targets for drug therapy. We developed three doxorubicin-resistant (DoxR) cell lines from parental cell lines. Matrigel in vitro invasion assays were used to compare invasiveness. Western blot assays were used to compare PD-L1 expression. Immuno-oncology checkpoint protein panels were used to compare concentrations of 17 checkpoint molecules both cellular and soluble. PD-L1 and 12 other checkpoint molecules were present in all cell lysates of each cell line without significantly different levels. Three were solubilized in the media of each cell line. PD-L1 is expressed in all DoxR and parental neuroblastoma cells and may be a potential target for drug therapy although its role in drug resistance remains unclear. Benchmarking checkpoint molecules provides the basis for future studies identifying targets for directed therapy and biomarkers for cancer detection or prognosis.
Collapse
|
39
|
Núñez Abad M, Calabuig-Fariñas S, Lobo de Mena M, Torres-Martínez S, García González C, García García JÁ, Iranzo González-Cruz V, Camps Herrero C. Programmed Death-Ligand 1 (PD-L1) as Immunotherapy Biomarker in Breast Cancer. Cancers (Basel) 2022; 14:307. [PMID: 35053471 PMCID: PMC8773553 DOI: 10.3390/cancers14020307] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/22/2021] [Accepted: 01/05/2022] [Indexed: 12/21/2022] Open
Abstract
Breast cancer constitutes the most common malignant neoplasm in women around the world. Approximately 12% of patients are diagnosed with metastatic stage, and between 5 and 30% of early or locally advanced BC patients will relapse, making it an incurable disease. PD-L1 ligation is an immune inhibitory molecule of the activation of T cells, playing a relevant role in numerous types of malignant tumors, including BC. The objective of the present review is to analyze the role of PD-L1 as a biomarker in the different BC subtypes, adding clinical trials with immune checkpoint inhibitors and their applicable results. Diverse trials using immunotherapy with anti-PD-1/PD-L1 in BC, as well as prospective or retrospective cohort studies about PD-L1 in BC, were included. Despite divergent results in the reviewed studies, PD-L1 seems to be correlated with worse prognosis in the hormone receptor positive subtype. Immune checkpoints inhibitors targeting the PD-1/PD-L1 axis have achieved great response rates in TNBC patients, especially in combination with chemotherapy, making immunotherapy a new treatment option in this scenario. However, the utility of PD-L1 as a predictive biomarker in the rest of BC subtypes remains unclear. In addition, predictive differences have been found in response to immunotherapy depending on the stage of the tumor disease. Therefore, a better understanding of tumor microenvironment, as well as identifying new potential biomarkers or combined index scores, is necessary in order to make a better selection of the subgroups of BC patients who will derive benefit from immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Martín Núñez Abad
- Department of Medical Oncology, Hospital General Universitario de Valencia, 46014 Valencia, Spain; (M.L.d.M.); (C.G.G.); (C.C.H.)
| | - Silvia Calabuig-Fariñas
- Molecular Oncology Laboratory, Fundación Investigación, Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.C.-F.); (S.T.-M.)
- Unidad Mixta TRIAL, Centro Investigación Príncipe Felipe-Fundación Investigación, Hospital General Universitario de Valencia, 46014 Valencia, Spain
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
- Department of Pathology, Universitat de València, 46010 Valencia, Spain
| | - Miriam Lobo de Mena
- Department of Medical Oncology, Hospital General Universitario de Valencia, 46014 Valencia, Spain; (M.L.d.M.); (C.G.G.); (C.C.H.)
| | - Susana Torres-Martínez
- Molecular Oncology Laboratory, Fundación Investigación, Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.C.-F.); (S.T.-M.)
- Unidad Mixta TRIAL, Centro Investigación Príncipe Felipe-Fundación Investigación, Hospital General Universitario de Valencia, 46014 Valencia, Spain
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
| | - Clara García González
- Department of Medical Oncology, Hospital General Universitario de Valencia, 46014 Valencia, Spain; (M.L.d.M.); (C.G.G.); (C.C.H.)
| | | | - Vega Iranzo González-Cruz
- Department of Medical Oncology, Hospital General Universitario de Valencia, 46014 Valencia, Spain; (M.L.d.M.); (C.G.G.); (C.C.H.)
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
- Department of Medicine, Universitat de València, 46010 Valencia, Spain
| | - Carlos Camps Herrero
- Department of Medical Oncology, Hospital General Universitario de Valencia, 46014 Valencia, Spain; (M.L.d.M.); (C.G.G.); (C.C.H.)
- Molecular Oncology Laboratory, Fundación Investigación, Hospital General Universitario de Valencia, 46014 Valencia, Spain; (S.C.-F.); (S.T.-M.)
- Unidad Mixta TRIAL, Centro Investigación Príncipe Felipe-Fundación Investigación, Hospital General Universitario de Valencia, 46014 Valencia, Spain
- Centro de Investigación Biomédica en Red Cáncer, CIBERONC, 28029 Madrid, Spain
- Department of Medicine, Universitat de València, 46010 Valencia, Spain
| |
Collapse
|
40
|
Kumar AR, Devan AR, Nair B, Vinod BS, Nath LR. Harnessing the immune system against cancer: current immunotherapy approaches and therapeutic targets. Mol Biol Rep 2021; 48:8075-8095. [PMID: 34671902 PMCID: PMC8605995 DOI: 10.1007/s11033-021-06752-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 09/15/2021] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy is a rapidly evolving concept that has been given the tag "fifth pillar" of cancer therapy while radiation therapy, chemotherapy, surgery and targeted therapy remain the other four pillars. This involves the stimulation of the immune system to control tumor growth and it specifically targets the neoplastic cells rather than the normal cells. Conventional chemotherapy has many limitations which include drug resistance, recurrence of cancer and severe adverse effects. Immunology has made major treatment breakthroughs for several cancers such as colorectal cancer, prostate cancer, breast cancer, lung cancer, liver cancer, kidney cancer, stomach cancer, acute lymphoblastic leukaemia etc. Currently, therapeutic strategies harnessing the immune system involve Checkpoint inhibitors, Chimeric antigen receptor T cells (CAR T cells), Monoclonal antibodies, Cancer vaccines, Cytokines, Radio-immunotherapy and Oncolytic virus therapy. The molecular characterization of several tumor antigens (TA) indicates that these TA can be utilized as promising candidates in cancer immunotherapy strategies. Here in this review, we highlight and summarize the different categories of emerging cancer immunotherapies along with the immunologically recognized tumor antigens involved in the tumor microenvironment.
Collapse
Affiliation(s)
- Ayana R Kumar
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - Aswathy R Devan
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - Bhagyalakshmi Nair
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India
| | - Balachandran S Vinod
- Department of Biochemistry, Sree Narayana College, Kollam, Kerala, 691001, India.
| | - Lekshmi R Nath
- Department of Pharmacognosy, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Science Campus, Ponekkara P. O., Kochi, Kerala, 682041, India.
| |
Collapse
|
41
|
Chen DY, Huang YH, Chen YM, Chen JJW, Yang TY, Chang GC, Tang KT. ANA positivity and complement level in pleural fluid are potential diagnostic markers in discriminating lupus pleuritis from pleural effusion of other aetiologies. Lupus Sci Med 2021; 8:8/1/e000562. [PMID: 34785570 PMCID: PMC8596033 DOI: 10.1136/lupus-2021-000562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/27/2021] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Lupus pleuritis is the most common pulmonary manifestation of systemic lupus erythematosus (SLE). We aimed to compare various biomarkers in discriminating between pleural effusions due to lupus pleuritis and other aetiologies. METHODS We determined in 59 patients (16 patients with SLE and 43 patients without SLE) pleural fluid levels of high-mobility group box 1, soluble receptor for advanced glycation end products (sRAGE), adenosine deaminase (ADA), interleukin (IL) 17A, tumour necrosis factor-α, antinuclear antibodies (ANA), and complements C3 and C4. RESULTS We found significant differences in the pleural fluid level of sRAGE, ADA, IL-17A, C3 and C4, and in the proportion of ANA positivity, among lupus pleuritis and other groups with pleural effusion. Specifically, ANA positivity (titre ≥1: 80) achieved a high sensitivity of 91%, specificity of 83% and negative predictive value (NPV) of 97% in discriminating lupus pleuritis from non-lupus pleural effusion. A parallel combination of the level of C3 (<24 mg/dL) and C4 (<3 mg/dL) achieved a sensitivity of 82%, specificity of 89% and NPV of 93% in discriminating lupus pleuritis from non-lupus exudative pleural effusion. CONCLUSIONS In conclusion, ANA, C3 and C4 in pleural fluid are useful in discriminating lupus pleuritis from pleural effusion due to other aetiologies with high NPV.
Collapse
Affiliation(s)
- Der-Yuan Chen
- Translational Medicine Laboratory, China Medical University Hospital, Taichung, Taiwan.,Rheumatology and Immunology Center, China Medical University Hospital, Taichung, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan
| | - Yen-Hsiang Huang
- Division of Chest Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Ming Chen
- Department of Medical Research, Taichung Veterans General Hospital, Taichung, Taiwan.,Faculty of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.,Ph.D. Program in Translational Medicine and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Jeremy J W Chen
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan
| | - Tsung-Ying Yang
- Division of Chest Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Faculty of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Gee-Chen Chang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung, Taiwan.,Faculty of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.,Division of Pulmonary Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Kuo-Tung Tang
- Faculty of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan .,Ph.D. Program in Translational Medicine and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan.,Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung, Taiwan
| |
Collapse
|
42
|
Programmed Cell Death Protein Ligand 2 Is a Potential Biomarker That Predicts the Efficacy of Immunotherapy. DISEASE MARKERS 2021; 2021:9453692. [PMID: 34754345 PMCID: PMC8572643 DOI: 10.1155/2021/9453692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/16/2021] [Indexed: 11/18/2022]
Abstract
Immune checkpoint inhibitor (ICI) responses vary, and biomarkers for predicting responders are urgently needed. Growing evidence points to the association between programmed cell death protein ligand 2 (PDL2) and ICI benefits, while clinical evidences were lacking. Thus, we consolidated five public ICI-treated cohorts to investigate the association between PDL2 expression and ICI treatment prognosis. Immune cell signatures and IFN-γ signatures are investigated in The Cancer Genome Atlas (TCGA) dataset and later in ICI-treated cohorts to explore the association between PDL2 and antitumor immunity in the tumor microenvironment (TME). We found that immune cell signatures and IFN-γ signatures were enriched in the PDL2-high group in TCGA pooled cohorts and most cancers. Consistently, in ICI-treated cohorts, patients with high PDL2 expression experienced longer overall survival time (OS) and were more likely responsive to ICIs than patients with low PDL2 expression. Immune cell scores of the high PDL2 expression patients were significantly higher (P < 0.05) than those of the low PDL2 expression patients in ICI-treated cohorts. In conclusion, our findings suggest that PDL2 is a potential predictive biomarker for ICIs.
Collapse
|
43
|
Zhou M, Zhang X, Li T, Chen Y. Dysregulated ferroptosis-related genes indicate potential clinical benefits for anti-PD-1/PD-L1 immunotherapy in lung adenocarcinoma. J Clin Lab Anal 2021; 35:e24086. [PMID: 34752672 PMCID: PMC8649384 DOI: 10.1002/jcla.24086] [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: 08/20/2021] [Revised: 09/30/2021] [Accepted: 10/20/2021] [Indexed: 11/08/2022] Open
Abstract
Background Ferroptosis is an iron‐dependent programmed cell death mechanism that influences the development of malignancy. Lung adenocarcinoma (LUAD) is the most common type of lung cancer with no known cure. Anti–PD‐1/PD‐L immunotherapy is effective for patients with partial LUAD. Therefore, there is an immediate requirement of novel markers to predict the individualised benefits of immunotherapy. Methods We manually collected the ferroptosis‐related gene (FERG) set and employed the Wilcoxon rank‐sum test to identify the differentially expressed FERGs. Subsequently, we constructed a recursive partitioning and regression tree (RPART) model to predict the benefits of anti–PD‐1/PD‐L1 immunotherapy. Subsequently, the ROC curve and AUC were used to evaluate the model efficiency in an independent dataset. Results In this study, we found that the dysregulated FERGs were closely associated with multiple metabolic processes in LUAD. Furthermore, we identified three ferroptosis‐related tumour subtypes (F1, F3 and F3). The F3 subtype exhibited higher immunoactivity and lower tumour purity, mutation count and aneuploidy and had better survival outcomes compared with the other two subtypes, implying that FERGs played an important role in intertumoral immune heterogeneity. We further explored the role of FERGs in the anti–PD‐1/PD‐L1 immunotherapy. We identified a set of three‐FERGs signature (CD44, G6PD and ZEB1) that acted as a promising indicator (AUC = 0.697) for the prediction of the benefits of anti–PD‐1/PD‐L1 immunotherapy. Conclusion Ferroptosis, as emerging programmed cell death mechanism, was associated with cancer development. We used ferroptosis‐related genes to predict the immunotherapy benefits that may facilitate the development of individualised anti‐cancer treatment strategies.
Collapse
Affiliation(s)
- Min Zhou
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Xiuxiu Zhang
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Tianyang Li
- The Second Clinical Medical College, Wenzhou Medical University, Wenzhou, China
| | - Yan Chen
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
44
|
Wu J, Sun Y, Li J, Ai M, You L, Shi J, Yu F. Analysis of Prognostic Alternative Splicing Reveals the Landscape of Immune Microenvironment in Thyroid Cancer. Front Oncol 2021; 11:763886. [PMID: 34733794 PMCID: PMC8558422 DOI: 10.3389/fonc.2021.763886] [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: 08/24/2021] [Accepted: 09/28/2021] [Indexed: 11/28/2022] Open
Abstract
Background The incidence of thyroid cancer (THCA) continues to increase in recent decades. Accumulating evidence showed that the unbalanced alternative splicing (AS) promotes the occurrence of cancers and leads to poor prognosis of patients. However, the research on alternative splicing events in THCA is lacking, and its underlying mechanism is not fully understood. This study identifies a novel prognostic signature based on AS events to reveal the relationship of AS with tumor immune microenvironment. Methods Based on the AS data, transcriptional data, and clinical information, the differentially expressed alternative splicings (DEASs) were screened out. Least absolute shrinkage and selection operator (LASSO) regression and multi-Cox regression analyses were employed to identify prognostic results related to AS events and establish a prognostic signature. The predictive ability of the signature was assessed by Kaplan-Meier (K-M) survival curve, risk plots, and receiver operating characteristic (ROC) curves. Furthermore, correlations between tumor-infiltrating immune cells, immune checkpoints, immune score and prognostic signature were analyzed. Results According to the LASSO regression analysis, a total of five AS events were selected to construct the signature. K-M survival curve showed that the higher the risk score, the worse the OS of the patients. Risk plots further confirmed this result. ROC curves indicated the high predictive efficiency of the prognostic signature. As for tumor immune microenvironment, patients in the high-risk group had a higher proportion of immune cells, including plasma cell, CD8+ T cell, macrophages (M0 and M2), and activated dendritic cell. Immune checkpoint proteins, such as PDCD1LG2, HAVCR2, CD274, etc., were significantly higher in the high-risk group. We also found that the ESTIMATE score, stromal score, and immune score were lower in the high-risk group, while the result of tumor purity was the opposite. Conclusions Collectively, a prognostic signature consisting of five AS events in THCA was established. Furthermore, there was an inextricable correlation between immune cell infiltration, immune checkpoint proteins, and AS events. This study will provide a basis for THCA immunotherapy in the future.
Collapse
Affiliation(s)
- Jian Wu
- Department of Otorhinolaryngology-Head and Neck Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Yifang Sun
- Department of Ophthalmology, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Junzheng Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Maomao Ai
- Department of Otorhinolaryngology-Head and Neck Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Lihua You
- Department of Otorhinolaryngology-Head and Neck Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| | - Jianbo Shi
- Department of Otorhinolaryngology-Head and Neck Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China.,Department of Otorhinolaryngology, The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, China
| | - Feng Yu
- Department of Otorhinolaryngology-Head and Neck Surgery, Guangzhou Red Cross Hospital, Jinan University, Guangzhou, China
| |
Collapse
|
45
|
Jimbu L, Mesaros O, Neaga A, Nanut AM, Tomuleasa C, Dima D, Bocsan C, Zdrenghea M. The Potential Advantage of Targeting Both PD-L1/PD-L2/PD-1 and IL-10-IL-10R Pathways in Acute Myeloid Leukemia. Pharmaceuticals (Basel) 2021; 14:1105. [PMID: 34832887 PMCID: PMC8620891 DOI: 10.3390/ph14111105] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/17/2021] [Accepted: 10/25/2021] [Indexed: 12/30/2022] Open
Abstract
Tumor cells promote the suppression of host anti-tumor type 1 T cell responses by various mechanisms, including the upregulation of surface inhibitory molecules such as programmed death ligand (PD-L)-1, and the production of immunosuppressive cytokines such as interleukin-10 (IL-10). There are over 2000 trials investigating PD-L1 and/or its receptor programmed-death 1 (PD-1) blockade in cancer, leading to the approval of PD-1 or PD-L1 inhibitors in several types of solid cancers and in hematological malignancies. The available data suggest that the molecule PD-L1 on antigen-presenting cells suppresses type 1 T cell immune responses such as cytotoxicity, and that the cytokine IL-10, in addition to downregulating immune responses, increases the expression of inhibitory molecule PD-L1. We hypothesize that the manipulation of both the co-inhibitory network (with anti-PD-L1 blocking antibodies) and suppressor network (with anti-IL-10 blocking antibodies) is an attractive immunotherapeutic intervention for acute myeloid leukemia (AML) patients ineligible for standard treatment with chemotherapy and hematopoietic stem cell transplantation, and with less severe adverse reactions. The proposed combination of these two immunotherapies represents a new approach that can be readily translated into the clinic to improve the therapeutic efficacy of AML disease treatment.
Collapse
Affiliation(s)
- Laura Jimbu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania; (O.M.); (A.N.); (A.M.N.); (C.T.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania;
| | - Oana Mesaros
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania; (O.M.); (A.N.); (A.M.N.); (C.T.); (M.Z.)
- “Octavian Fodor” Regional Institute of Gastroenterology and Hepatology, 19-21 Croitorilor Str., 400162 Cluj-Napoca, Romania
| | - Alexandra Neaga
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania; (O.M.); (A.N.); (A.M.N.); (C.T.); (M.Z.)
| | - Ana Maria Nanut
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania; (O.M.); (A.N.); (A.M.N.); (C.T.); (M.Z.)
| | - Ciprian Tomuleasa
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania; (O.M.); (A.N.); (A.M.N.); (C.T.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania;
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania;
| | - Corina Bocsan
- Department of Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania;
| | - Mihnea Zdrenghea
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Babes Str., 400012 Cluj-Napoca, Romania; (O.M.); (A.N.); (A.M.N.); (C.T.); (M.Z.)
- Department of Hematology, Ion Chiricuta Oncology Institute, 34-36 Republicii Str., 400015 Cluj-Napoca, Romania;
| |
Collapse
|
46
|
Ravindran Menon D, Li Y, Yamauchi T, Osborne DG, Vaddi PK, Wempe MF, Zhai Z, Fujita M. EGCG Inhibits Tumor Growth in Melanoma by Targeting JAK-STAT Signaling and Its Downstream PD-L1/PD-L2-PD1 Axis in Tumors and Enhancing Cytotoxic T-Cell Responses. Pharmaceuticals (Basel) 2021; 14:1081. [PMID: 34832863 PMCID: PMC8618268 DOI: 10.3390/ph14111081] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 12/13/2022] Open
Abstract
Over the last decade, therapies targeting immune checkpoints, such as programmed death-1 (PD-1), have revolutionized the field of cancer immunotherapy. However, low response rates and immune-related adverse events remain a major concern. Here, we report that epigallocatechin gallate (EGCG), the most abundant catechin in green tea, inhibits melanoma growth by modulating an immune response against tumors. In vitro experiments revealed that EGCG treatment inhibited interferon-gamma (IFN-γ)-induced PD-L1 and PD-L2 expression and JAK-STAT signaling. We confirmed that this effect was driven by inhibiting STAT1 gene expression and STAT1 phosphorylation, thereby downregulating the PD-L1/PD-L2 transcriptional regulator IRF1 in both human and mouse melanoma cells. Animal studies revealed that the in vivo tumor-inhibitory effect of EGCG was through CD8+ T cells and that the inhibitory effect of EGCG was comparable to anti-PD-1 therapy. However, their mechanisms of action were different. Dissimilar to anti-PD-1 treatment that blocks PD-1/PD-L1 interaction, EGCG inhibited JAK/STAT signaling and PD-L1 expression in tumor cells, leading to the re-activation of T cells. In summary, we demonstrate that EGCG enhances anti-tumor immune responses by inhibiting JAK-STAT signaling in melanoma. EGCG could be used as an alternative treatment strategy to target the PD-L1/PD-L2-PD-1 axis in cancers.
Collapse
Affiliation(s)
- Dinoop Ravindran Menon
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (D.R.M.); (Y.L.); (T.Y.); (D.G.O.); (P.K.V.); (Z.Z.)
| | - Yang Li
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (D.R.M.); (Y.L.); (T.Y.); (D.G.O.); (P.K.V.); (Z.Z.)
| | - Takeshi Yamauchi
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (D.R.M.); (Y.L.); (T.Y.); (D.G.O.); (P.K.V.); (Z.Z.)
| | - Douglas Grant Osborne
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (D.R.M.); (Y.L.); (T.Y.); (D.G.O.); (P.K.V.); (Z.Z.)
| | - Prasanna Kumar Vaddi
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (D.R.M.); (Y.L.); (T.Y.); (D.G.O.); (P.K.V.); (Z.Z.)
| | - Michael F Wempe
- Department of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Zili Zhai
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (D.R.M.); (Y.L.); (T.Y.); (D.G.O.); (P.K.V.); (Z.Z.)
| | - Mayumi Fujita
- Department of Dermatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; (D.R.M.); (Y.L.); (T.Y.); (D.G.O.); (P.K.V.); (Z.Z.)
- Department of Veterans Affairs Medical Center, VA Eastern Colorado Health Care System, Aurora, CO 80045, USA
- Department of Immunology & Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| |
Collapse
|
47
|
Jeong S, Lee N, Park MJ, Jeon K, Song W. Currently Used Laboratory Methodologies for Assays Detecting PD-1, PD-L1, PD-L2 and Soluble PD-L1 in Patients with Metastatic Breast Cancer. Cancers (Basel) 2021; 13:cancers13205225. [PMID: 34680373 PMCID: PMC8534186 DOI: 10.3390/cancers13205225] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Several methods targeting the programmed death protein-1 (PD-1) axis have been developed and evaluated for the detection of immune checkpoint levels that are strongly involved in immunotherapy for patients with metastatic breast cancer. Variations in different assays used in diverse studies have affected their result interpretation and clinical utility. When applying these assays to the laboratory, a comprehensive understanding of the characteristics of them should be recognized. We reviewed applied laboratory techniques for detecting PD-1, PD-ligand (L)1, PD-L2, and soluble PD-L1, which are important for selecting metastatic cancer patients for immunotherapy. Advances in methodologies according to the epoch are also investigated to gain insight into immunologic techniques and to facilitate appropriate laboratory settings for evaluating the PD-1 axis status, which are useful for estimating outcomes and planning patient-tailored immunotherapy strategies. Abstract Approximately 20% of breast cancer (BC) patients suffer from distant metastasis. The incidence and prevalence rates of metastatic BC have increased annually. Immune checkpoint inhibitors are an emerging area of treatment, especially for metastatic patients with poor outcomes. Several antibody drugs have been developed and approved for companion testing of the programmed death protine-1 (PD-1) axis. We reviewed currently used laboratory methodologies for assays determining PD-1 axis to provide a comprehensive understanding of principles, advantages, and drawbacks involved in their implementation. The most commonly used method is immunohistochemistry (92.9%) for PD-L1 expression using tissue samples (96.4%). The commonly used anti-PD-L1 antibody clone were commercially available 22C3 (30.8%), SP142 (19.2%), SP263 (15.4%), and E1L3N (11.5%). Enzyme-linked immunosorbent assay and electrochemiluminescent immunoassay that target soluble PD-ligand (L)1 were developed and popularized in 2019–2021, in contrast to 2016–2018. Easy accessibility and non-invasiveness due to the use of blood samples, quantitative outputs, and relatively rapid turnaround times make them more preferable. Regarding scoring methods, a combination of tumor and immune cells (45.5% in 2016–2018 to 57.1% in 2019–2021) rather than each cell alone became more popular. Information about antibody clones, platforms, scoring methods, and related companion drugs is recommended for reporting PD-L1 expression.
Collapse
Affiliation(s)
- Seri Jeong
- Department of Laboratory Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07440, Korea; (S.J.); (N.L.); (M.-J.P.)
| | - Nuri Lee
- Department of Laboratory Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07440, Korea; (S.J.); (N.L.); (M.-J.P.)
| | - Min-Jeong Park
- Department of Laboratory Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07440, Korea; (S.J.); (N.L.); (M.-J.P.)
| | - Kibum Jeon
- Department of Laboratory Medicine, Hangang Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07440, Korea;
| | - Wonkeun Song
- Department of Laboratory Medicine, Kangnam Sacred Heart Hospital, Hallym University College of Medicine, Seoul 07440, Korea; (S.J.); (N.L.); (M.-J.P.)
- Correspondence: ; Tel.: +82-2-829-5259
| |
Collapse
|
48
|
Liang SK, Chien LH, Chang GC, Tsai YH, Su WC, Chen YM, Huang MS, Lin HC, Fang WT, Hung HH, Jiang SS, Chen CY, Chen KY, Chang IS, Hsiung CA, Chen CJ, Yang PC. Programmed Death Ligand 2 Gene Polymorphisms Are Associated With Lung Adenocarcinoma Risk in Female Never-Smokers. Front Oncol 2021; 11:753788. [PMID: 34631591 PMCID: PMC8497977 DOI: 10.3389/fonc.2021.753788] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/08/2021] [Indexed: 12/24/2022] Open
Abstract
Objectives Lung cancer in never-smokers is a distinct disease associated with a different genomic landscape, pathogenesis, risk factors, and immune checkpoint inhibitor responses compared to those observed in smokers. This study aimed to identify novel single nucleotide polymorphisms (SNPs) of programmed death-1 (encoded by PDCD1) and its ligands, programmed death ligand 1 (CD274) and 2 (PDCD1LG2), associated with lung cancer risk in never-smoking women. Materials and Methods During September 2002 and July 2012, we enrolled never-smoking female patients with lung adenocarcinoma (LUAD) (n=1153) and healthy women (n=1022) from six tertiary hospitals in Taiwan. SNP data were obtained and analyzed from the genome-wide association study dataset and through an imputation method. The expression quantitative trait loci (eQTL) analysis was performed in both tumor and non-tumor tissues for the correlation between genetic expression and identified SNPs. Results A total of 12 PDCD1LG2 SNPs related to LUAD risk were identified in never-smoking women, including rs2381282, rs4742103, rs4237162, rs4742104, rs12237624, rs78096119, rs6476988, rs7857315, rs10975178, rs7854413, rs56001683, and rs7858319. Among them, six tagged PDCD1LG2 SNPs rs2381282, rs4742103, rs4237162, rs4742104, rs78096119, and rs56001683 were significantly associated with LUAD risk. Specifically, two PDCD1LG2 SNPs, rs12237624 and rs78096119, were associated with previous pulmonary tuberculosis infection in relation to LUAD susceptibility. Through an eQTL assay, we found that rs2381282 (p < 0.001), rs12237624 (p = 0.019), and rs78096119 (p = 0.019) were associated with the expression levels of programed death ligand 2. Conclusions Novel SNPs of programed death ligand 2 associated with lung adenocarcinoma risk were identified. Among them, two SNPs were associated with pulmonary tuberculosis infection in relation to lung adenocarcinoma susceptibility. These SNPs may help to stratify high-risk populations of never-smokers during lung cancer screening.
Collapse
Affiliation(s)
- Sheng-Kai Liang
- Department of Internal Medicine, National Taiwan University Hospital, Hsinchu Branch, Hsinchu, Taiwan.,Department of Medicine, National Taiwan University Cancer Center, Taipei, Taiwan
| | - Li-Hsin Chien
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Gee-Chen Chang
- Division of Chest Medicine, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan.,School of Medicine, Chung Shan Medical University, Taichung, Taiwan.,Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ying-Huang Tsai
- Division of Pulmonary and Critical Care Medicine, Linkou Chang Gung Memorial Hospital, Chang Gung Medical Foundation, Taoyuan, Taiwan
| | - Wu-Chou Su
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yuh-Min Chen
- Department of Chest Medicine, Taipei Veterans General Hospital, School of Medicine, National Yang-Ming University, and Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan
| | - Ming-Shyan Huang
- Department of Internal Medicine, E-Da Cancer Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Hsien-Chih Lin
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Wen-Tsen Fang
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan
| | - Hsiao-Han Hung
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan
| | - Shih-Sheng Jiang
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan
| | - Chih-Yi Chen
- Institute of Medicine, Chung Shan Medical University Hospital, and Division of Thoracic Surgery, Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Kuan-Yu Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - I-Shou Chang
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Taiwan.,Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Chao A Hsiung
- Division of Biostatistics and Bioinformatics, Institute of Population Health Sciences, National Health Research Institutes, Zhunan, Taiwan
| | - Chien-Jen Chen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,Graduate Institute of Epidemiology, College of Public Health, National Taiwan University, Taipei, Taiwan
| | - Pan-Chyr Yang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan.,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | | |
Collapse
|
49
|
Di Federico A, De Giglio A, Parisi C, Gelsomino F, Boni L, Ardizzoni A. Programmed Cell Death Protein-1 Inhibitors Versus Programmed Death-Ligand 1 Inhibitors in Addition to Chemotherapy for the First-Line Treatment of Advanced NSCLC: A Systematic Review and Meta-Analysis. JTO Clin Res Rep 2021; 2:100214. [PMID: 34590054 PMCID: PMC8474263 DOI: 10.1016/j.jtocrr.2021.100214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/01/2021] [Accepted: 07/18/2021] [Indexed: 01/20/2023] Open
Abstract
Introduction The addition of programmed cell death protein-1 (PD-1) or programmed death-ligand 1 (PD-L1) inhibitors to first-line chemotherapy (CT) improved the outcomes of advanced NSCLC. Nonetheless, no direct comparison exists between these combination treatments. Methods We performed a meta-analysis of randomized clinical trials to evaluate and compare the efficacy and safety of PD-(L)1 inhibitors in combination with first-line CT for advanced NSCLC. Results A total of eight randomized clinical trials were included. The addition of a PD-(L)1 inhibitor to CT improved progression-free survival, overall survival, and objective response rate compared with CT alone. The risk of grade greater than or equal to 3 treatment-related adverse events was slightly higher with the addition of a PD-(L)1 inhibitor to CT as compared with CT alone. A subgroup analysis according to the targeted receptor (PD-1 versus PD-L1) revealed that the addition of a PD-1 inhibitor to CT led to better objective response rate (p = 0.0001), progression-free survival (p = 0.006), and overall survival (p = 0.002) compared with that of a PD-L1 inhibitor. The risk of grade greater than or equal to 3 treatment-related adverse events was significantly increased with the addition of a PD-L1 inhibitor to CT, but not with the addition of a PD-1 inhibitor. A direct comparison using the meta-regression analysis confirmed the statistical significance of all previous findings. Conclusions On the basis of this meta-analysis, the addition of a PD-1 inhibitor to first-line CT revealed statistically significant better outcomes and less additional toxicity compared with that of a PD-L1 inhibitor, as compared with CT alone, in advanced NSCLC, regardless of PD-L1 status.
Collapse
Affiliation(s)
- Alessandro Di Federico
- Department of Specialized, Experimental and Diagnostic Medicine, University of Bologna, Bologna, Italy
- Division of Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero—Universitaria di Bologna, Bologna, Italy
| | - Andrea De Giglio
- Department of Specialized, Experimental and Diagnostic Medicine, University of Bologna, Bologna, Italy
- Division of Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero—Universitaria di Bologna, Bologna, Italy
- Corresponding author. Address for correspondence: Andrea De Giglio, MD, Department of Specialized, Experimental and Diagnostic Medicine, University of Bologna, Via Giuseppe Massarenti, 9, 40138 Bologna, Italy.
| | - Claudia Parisi
- Department of Specialized, Experimental and Diagnostic Medicine, University of Bologna, Bologna, Italy
- Division of Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero—Universitaria di Bologna, Bologna, Italy
| | - Francesco Gelsomino
- Department of Specialized, Experimental and Diagnostic Medicine, University of Bologna, Bologna, Italy
- Division of Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero—Universitaria di Bologna, Bologna, Italy
| | - Luca Boni
- Epidemiologia Clinica, Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS) Ospedale Policlinico San Martino—IST Nord CBA, Genova, Italy
| | - Andrea Ardizzoni
- Department of Specialized, Experimental and Diagnostic Medicine, University of Bologna, Bologna, Italy
- Division of Medical Oncology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Azienda Ospedaliero—Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
50
|
Paul S, Sa G. Curcumin as an Adjuvant to Cancer Immunotherapy. Front Oncol 2021; 11:675923. [PMID: 34485117 PMCID: PMC8415504 DOI: 10.3389/fonc.2021.675923] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 07/19/2021] [Indexed: 01/21/2023] Open
Abstract
The components of the immune system play a very sincere and crucial role in combating tumors. However, despite their firm efforts of elimination, tumor cells cleverly escape the surveillance process by adopting several immune evasion mechanisms. The conversion of immunogenicity of tumor microenvironment into tolerogenic is considered as a prime reason for tumor immune escape. Therapeutically, different immunotherapies have been adopted to block such immune escaping routes along with better clinical outcomes. Still, the therapies are haunted by several drawbacks. Over time, curcumin has been considered as a potential anti-cancer molecule. Its potentialities have been recorded against the standard hallmarks of cancer such as continuous proliferation, escaping apoptosis, continuous angiogenesis, insensitivity to growth inhibitors, tissue invasion, and metastasis. Hence, the diversity of curcumin functioning has already been established and exploration of its application with immunotherapies might open up a new avenue for scientists and clinicians. In this review, we briefly discuss the tumor's way of immune escaping, followed by various modern immunotherapies that have been used to encounter the escaping paths and their minute flaws. Finally, the conclusion has been drawn with the application of curcumin as a potential immune-adjuvant, which fearlessly could be used with immunotherapies for best outcomes.
Collapse
Affiliation(s)
| | - Gaurisankar Sa
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| |
Collapse
|