1
|
Potthoff TE, Walter C, Jeising D, Münter D, Verma A, Suero Molina E, Stummer W, Dugas M, Hartmann W, Dottermusch M, Altendorf L, Schüller U, Scheuermann S, Seitz C, Albert TK, Kerl K. Single-cell transcriptomics link gene expression signatures to clinicopathological features of gonadotroph and lactotroph PitNET. J Transl Med 2024; 22:1027. [PMID: 39548496 PMCID: PMC11566263 DOI: 10.1186/s12967-024-05821-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 10/31/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND Pituitary neuroendocrine tumors (PitNET) are among the most common intracranial tumors. Despite a frequent benign course, aggressive behavior can occur. Tumor behavior is known to be under the influence of the tumor microenvironment (TME). However, the relationship between TME cells and aggressive tumor behavior has not been adequately explored in PitNET. METHODS We performed differential expression analysis as well as gene expression program identification based on single-cell RNA sequencing to comparatively characterize the transcriptome of seven gonadotroph and three lactotroph PitNET and correlate it with clinical features using bulk RNA-seq data from an independent cohort of 134 PitNET. Tumor immune infiltration was quantified via immunostaining on tissue sections of gonadotroph and lactotroph PitNET. RESULTS In lactotroph PitNET, we detect a highly proliferative gene profile with significantly increased expression levels in aggressively growing tumors within bulk RNA-seq data of an independent cohort of 134 PitNET samples. We also report high intratumoral heterogeneity in gonadotroph PitNET (GoPN) and lactotroph PitNET (LaPN) and identify signatures of epithelial, endocrine, and immunological gene networks in both subtypes. A comparison of their TME composition shows enrichment of SPP1+ macrophages and CD4+ T cells in GoPN, as well as enrichment of CD4/CD8 double-negative T cells (DN) and natural killer cells (NK) in LaPN. Also notable is the presence of proliferative lymphocytes, the occurrence of which positively correlates with more aggressive tumor behavior in the bulk RNA-seq cohort. However, increased CD8+ T and NK cell abundances correlate significantly with reduced aggressiveness indicating potential anti-tumoral effects. CONCLUSIONS Our study expands the knowledge of the differences in cellular composition of gonadotroph and lactotroph PitNET subtypes. It lays the foundation for further studies on the influence of lymphoid cells on the variable aggressive behavior of PitNET. Regarding the treatment of drug-resistant lactotroph PitNET, proliferative lymphocytes, CD8+ T, and NK cells could represent potentially valuable targets for developing new cancer immunotherapies.
Collapse
Affiliation(s)
- T Elise Potthoff
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Carolin Walter
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
| | - Daniela Jeising
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Daniel Münter
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Archana Verma
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Eric Suero Molina
- Department of Neurosurgery, University Hospital of Münster, 48149, Münster, Germany
| | - Walter Stummer
- Department of Neurosurgery, University Hospital of Münster, 48149, Münster, Germany
| | - Martin Dugas
- Institute of Medical Informatics, University of Münster, 48149, Münster, Germany
- Institute of Medical Informatics, Heidelberg University Hospital, Heidelberg, Germany
| | - Wolfgang Hartmann
- Division of Translational Pathology, Gerhard Domagk Institute of Pathology, University Hospital Münster, 48149, Münster, Germany
| | - Matthias Dottermusch
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
| | - Lea Altendorf
- Department of Paediatric Haematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
- Research Institute Children's Cancer Center, 20251, Hamburg, Germany
| | - Ulrich Schüller
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
- Department of Paediatric Haematology and Oncology, University Medical Center Hamburg-Eppendorf, 20251, Hamburg, Germany
- Research Institute Children's Cancer Center, 20251, Hamburg, Germany
| | - Sophia Scheuermann
- DFG Cluster of Excellence 2180 'Image-Guided and Functional Instructed Tumor Therapy' (iFIT), University of Tübingen, 72076, Tübingen, Germany
- Department of Pediatric Hematology and Oncology, University Hospital Tübingen, 72076, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site, Tuebingen, Germany
| | - Christian Seitz
- DFG Cluster of Excellence 2180 'Image-Guided and Functional Instructed Tumor Therapy' (iFIT), University of Tübingen, 72076, Tübingen, Germany
- Department of Pediatric Hematology and Oncology, University Hospital Tübingen, 72076, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site, Tuebingen, Germany
| | - Thomas K Albert
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany
| | - Kornelius Kerl
- Department of Paediatric Haematology and Oncology, University Children's Hospital Münster, Albert-Schweitzer-Campus 1, 48149, Münster, Germany.
| |
Collapse
|
2
|
Li GS, He RQ, Huang ZG, Huang H, Yang Z, Liu J, Fu ZW, Huang WY, Zhou HF, Kong JL, Chen G. A novel prognostic signature of coagulation-related genes leveraged by machine learning algorithms for lung squamous cell carcinoma. Heliyon 2024; 10:e27595. [PMID: 38496840 PMCID: PMC10944263 DOI: 10.1016/j.heliyon.2024.e27595] [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: 04/05/2023] [Revised: 12/26/2023] [Accepted: 03/04/2024] [Indexed: 03/19/2024] Open
Abstract
Coagulation-related genes (CRGs) have been demonstrated to be essential for the development of certain tumors; however, little is known about CRGs in lung squamous cell carcinoma (LUSC). In this study, we adopted CRGs to construct a coagulation-related gene prognostic signature (CRGPS) using machine learning algorithms. Using a set of 92 machine learning integrated algorithms, the CRGPS was determined to be the optimal prognostic signature (median C-index = 0.600) for predicting the prognosis of an LUSC patient. The CRGPS was not only superior to traditional clinical parameters (e.g., T stage, age, and gender) and its commutative genes but also outperformed 19 preexisting prognostic signatures for LUSC on predictive accuracy. The CRGPS score was positively correlated with poor prognoses in patients with LUSC (hazard ratio > 1, p < 0.05), indicating its suitability as a prognostic marker for this disease. The CRGPS was observed to be inversely correlated with the degree of infiltration of natural killer cells. For some tumors, patients with lower CRGPS scores are more likely to experience enhanced immunotherapy effects (area under the curve = 0.70), which implies that the CRGPS can potentially predict immunotherapy efficacy. A high CRGPS score is predictive of an LUSC patient being sensitive to several drugs. Collectively, these findings indicate that the CRGPS may be a reliable indicator of the prognoses of patients with LUSC and may be useful for the clinical management of such patients.
Collapse
Affiliation(s)
- Guo-Sheng Li
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Zhi-Guang Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Hong Huang
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Zhen Yang
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Jun Liu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Zong-Wang Fu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Wan-Ying Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Hua-Fu Zhou
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Jin-Liang Kong
- Division of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, PR China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, PR China
| |
Collapse
|
3
|
Li T, Niu M, Zhang W, Qin S, Zhou J, Yi M. CAR-NK cells for cancer immunotherapy: recent advances and future directions. Front Immunol 2024; 15:1361194. [PMID: 38404574 PMCID: PMC10884099 DOI: 10.3389/fimmu.2024.1361194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 01/29/2024] [Indexed: 02/27/2024] Open
Abstract
Natural Killer (NK) cells, intrinsic to the innate immune system, are pivotal in combating cancer due to their independent cytotoxic capabilities in antitumor immune response. Unlike predominant treatments that target T cell immunity, the limited success of T cell immunotherapy emphasizes the urgency for innovative approaches, with a spotlight on harnessing the potential of NK cells. Despite tumors adapting mechanisms to evade NK cell-induced cytotoxicity, there is optimism surrounding Chimeric Antigen Receptor (CAR) NK cells. This comprehensive review delves into the foundational features and recent breakthroughs in comprehending the dynamics of NK cells within the tumor microenvironment. It critically evaluates the potential applications and challenges associated with emerging CAR-NK cell therapeutic strategies, positioning them as promising tools in the evolving landscape of precision medicine. As research progresses, the unique attributes of CAR-NK cells offer a new avenue for therapeutic interventions, paving the way for a more effective and precise approach to cancer treatment.
Collapse
Affiliation(s)
- Tianye Li
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Mengke Niu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weijiang Zhang
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Shuang Qin
- Department of Radiation Oncology, Hubei Cancer Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianwei Zhou
- Department of Gynecology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Obstetrics and Gynecology, Hangzhou, China
| | - Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
4
|
Meng X, Xu H, Liang Y, Liang M, Song W, Zhou B, Shi J, Du M, Gao Y. Enhanced CT-based radiomics model to predict natural killer cell infiltration and clinical prognosis in non-small cell lung cancer. Front Immunol 2024; 14:1334886. [PMID: 38283362 PMCID: PMC10811188 DOI: 10.3389/fimmu.2023.1334886] [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: 11/08/2023] [Accepted: 12/29/2023] [Indexed: 01/30/2024] Open
Abstract
Background Natural killer (NK) cells are crucial for tumor prognosis; however, their role in non-small-cell lung cancer (NSCLC) remains unclear. The current detection methods for NSCLC are inefficient and costly. Therefore, radiomics represent a promising alternative. Methods We analyzed the radiogenomics datasets to extract clinical, radiological, and transcriptome data. The effect of NK cells on the prognosis of NSCLC was assessed. Tumors were delineated using a 3D Slicer, and features were extracted using pyradiomics. A radiomics model was developed and validated using five-fold cross-validation. A nomogram model was constructed using the selected clinical variables and a radiomic score (RS). The CIBERSORTx database and gene set enrichment analysis were used to explore the correlations of NK cell infiltration and molecular mechanisms. Results Higher infiltration of NK cells was correlated with better overall survival (OS) (P = 0.002). The radiomic model showed an area under the curve of 0.731, with 0.726 post-validation. The RS differed significantly between high and low infiltration of NK cells (P < 0.01). The nomogram, using RS and clinical variables, effectively predicted 3-year OS. NK cell infiltration was correlated with the ICOS and BTLA genes (P < 0.001) and macrophage M0/M2 levels. The key pathways included TNF-α signaling via NF-κB and Wnt/β-catenin signaling. Conclusions Our radiomic model accurately predicted NK cell infiltration in NSCLC. Combined with clinical characteristics, it can predict the prognosis of patients with NSCLC. Bioinformatic analysis revealed the gene expression and pathways underlying NK cell infiltration in NSCLC.
Collapse
Affiliation(s)
- Xiangzhi Meng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haijun Xu
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yicheng Liang
- Department of Thoracic Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Mei Liang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Weijian Song
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Boxuan Zhou
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianwei Shi
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minjun Du
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yushun Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
5
|
Balzasch BM, Cerwenka A. Microenvironmental signals shaping NK-cell reactivity in cancer. Eur J Immunol 2023; 53:e2250103. [PMID: 37194594 DOI: 10.1002/eji.202250103] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/05/2023] [Accepted: 05/09/2023] [Indexed: 05/18/2023]
Abstract
Since the postulation of the "missing-self" concept, much progress has been made in defining requirements for NK-cell activation. Unlike T lymphocytes that process signals from receptors in a hierarchic manner dominated by the T-cell receptors, NK cells integrate receptor signals more "democratically." Signals originate not only the downstream of cell-surface receptors triggered by membrane-bound ligands or cytokines, but are also mediated by specialized microenvironmental sensors that perceive the cellular surrounding by detecting metabolites or the availability of oxygen. Thus, NK-cell effector functions are driven in an organ and disease-dependent manner. Here, we review the latest findings on how NK-cell reactivity in cancer is determined by the reception and integration of complex signals. Finally, we discuss how this knowledge can be exploited to guide novel combinatorial approaches for NK-cell-based anticancer therapies.
Collapse
Affiliation(s)
- Bianca M Balzasch
- Department of Immunobiochemistry, Mannheim Institute for Innate Immunosciences (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Adelheid Cerwenka
- Department of Immunobiochemistry, Mannheim Institute for Innate Immunosciences (MI3), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
6
|
Akshaya RL, Saranya I, Selvamurugan N. MicroRNAs mediated interaction of tumor microenvironment cells with breast cancer cells during bone metastasis. Breast Cancer 2023; 30:910-925. [PMID: 37578597 DOI: 10.1007/s12282-023-01491-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 08/07/2023] [Indexed: 08/15/2023]
Abstract
Breast cancer (BC) bone metastasis is primarily osteolytic and has limited therapeutic options. Metastasized BC cells prime the secondary environment in bone by forming a tumor niche, which favors their homing and colonization. The tumor microenvironment (TME) is primarily generated by the cancer cells. Bone TME is an intricate network of multiple cells, including altered bone, tumor, stromal, and immune cells. Recent findings highlight the significance of small non-coding microRNAs (miRNAs) in influencing TME during tumor metastasis. MiRNAs from TME-resident cells facilitate the interaction between the tumor and its microenvironment, thereby regulating the biological processes of tumors. These miRNAs can serve as oncogenes or tumor suppressors. Hence, both miRNA inhibitors and mimics are extensively utilized in pre-clinical trials for modulating the phenotypes of tumor cells and associated stromal cells. This review briefly summarizes the recent developments on the functional role of miRNAs secreted directly or indirectly from the TME-resident cells in facilitating tumor growth, progression, and metastasis. This information would be beneficial in developing novel targeted therapies for BC.
Collapse
Affiliation(s)
- R L Akshaya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603 103, Tamil Nadu, India
| | - I Saranya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603 103, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603 103, Tamil Nadu, India.
| |
Collapse
|
7
|
Kitamura T. Tumour-associated macrophages as a potential target to improve natural killer cell-based immunotherapies. Essays Biochem 2023; 67:1003-1014. [PMID: 37313600 PMCID: PMC10539946 DOI: 10.1042/ebc20230002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/15/2023]
Abstract
Adoptive transfer of natural killer (NK) cells has been proposed as a novel immunotherapy for malignant tumours resistant to current therapeutic modalities. Several clinical studies have demonstrated that the NK cell-infusion is well tolerated without severe side effects and shows promising results in haematological malignancies. However, patients with malignant solid tumours do not show significant responses to this therapy. Such disappointing results largely arise from the inefficient delivery of infused NK cells and the impairment of their functions in the tumour microenvironment (TME). Tumour-associated macrophages (TAMs) are the most abundant stromal cells in the TME of most solid tumours, and a high TAM density correlates with poor prognosis of cancer patients. Although our knowledge of the interactions between TAMs and NK cells is limited, many studies have indicated that TAMs suppress NK cell cytotoxicity against cancer cells. Therefore, blockade of TAM functions can be an attractive strategy to improve NK cell-based immunotherapies. On the other hand, macrophages are reported to activate NK cells under certain circumstances. This essay presents our current knowledge about mechanisms by which macrophages regulate NK cell functions and discusses possible therapeutic approaches to block macrophage-mediated NK cell suppression.
Collapse
Affiliation(s)
- Takanori Kitamura
- MRC Centre for Reproductive Health, The University of Edinburgh, Edinburgh, EH16 4TJ, United Kingdom
| |
Collapse
|
8
|
Zeng T, Zang W, Xiao H, Jiang Y, Lin S, Wang M, Li S, Li L, Li C, Lu C, Yang H. Carrier-Free Nanovaccine: An Innovative Strategy for Ultrahigh Melanoma Neoantigen Loading. ACS NANO 2023; 17:18114-18127. [PMID: 37695697 DOI: 10.1021/acsnano.3c04887] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
In personalized cancer immunotherapy, developing an effective neoantigen nanovaccine with high immunogenicity is a significant challenge. Traditional nanovaccine delivery systems often require nanocarriers, which can hinder the delivery of the neoantigen and cause significant toxicity. In this study, we present an innovative strategy of carrier-free nanovaccine achieved through direct self-assembly of 2'-fluorinated CpG (2'F-CpG) with melanoma neoantigen peptide (Obsl1). Molecular dynamics simulations demonstrated that the introduction of a fluorine atom into CpG increases the noncovalent interaction between 2'F-CpG and Obsl1, which enhanced the loading of Obsl1 on 2'F-CpG, resulting in the spontaneous formation of a hybrid 2'F-CpG/Obsl1 nanovaccine. This nanovaccine without extra nanocarriers showed ultrahigh Obsl1 loading up to 83.19 wt %, increasing the neoantigen peptide uptake by antigen-presenting cells (APCs). In C57BL/6 mice models, we demonstrated the long-term preventive and therapeutic effects of the prepared 2'F-CpG/Obsl1 nanovaccine against B16F10 melanoma. Immunocellular analysis revealed that the nanovaccine activated innate and adaptive immune responses to cancer cells. Hence, this study established a simple, safe, and effective preparation strategy for a carrier-free neoantigen nanovaccine, which could be adapted for the future design of personalized cancer vaccines in clinical settings.
Collapse
Affiliation(s)
- Tao Zeng
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Weijie Zang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Han Xiao
- State Key Laboratory of Structure of Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences Fuzhou, Fujian 350002, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yifan Jiang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Sang Lin
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Min Wang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Shiqing Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Liannishang Li
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Chunsen Li
- State Key Laboratory of Structure of Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences Fuzhou, Fujian 350002, People's Republic of China
- University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Chunhua Lu
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| | - Huanghao Yang
- MOE Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, People's Republic of China
| |
Collapse
|
9
|
Sozio F, Schioppa T, Laffranchi M, Salvi V, Tamassia N, Bianchetto-Aguilera FM, Tiberio L, Bonecchi R, Bosisio D, Parmentier M, Bottazzi B, Leone R, Russo E, Bernardini G, Garofalo S, Limatola C, Gismondi A, Sciumè G, Mantovani A, Del Prete A, Sozzani S. CCRL2 Expression by Specialized Lung Capillary Endothelial Cells Controls NK-cell Homing in Lung Cancer. Cancer Immunol Res 2023; 11:1280-1295. [PMID: 37343073 DOI: 10.1158/2326-6066.cir-22-0951] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 04/07/2023] [Accepted: 06/20/2023] [Indexed: 06/23/2023]
Abstract
Patterns of receptors for chemotactic factors regulate the homing of leukocytes to tissues. Here we report that the CCRL2/chemerin/CMKLR1 axis represents a selective pathway for the homing of natural killer (NK) cells to the lung. C-C motif chemokine receptor-like 2 (CCRL2) is a nonsignaling seven-transmembrane domain receptor able to control lung tumor growth. CCRL2 constitutive or conditional endothelial cell targeted ablation, or deletion of its ligand chemerin, were found to promote tumor progression in a Kras/p53Flox lung cancer cell model. This phenotype was dependent on the reduced recruitment of CD27- CD11b+ mature NK cells. Other chemotactic receptors identified in lung-infiltrating NK cells by single-cell RNA sequencing (scRNA-seq), such as Cxcr3, Cx3cr1, and S1pr5, were found to be dispensable in the regulation of NK-cell infiltration of the lung and lung tumor growth. scRNA-seq identified CCRL2 as the hallmark of general alveolar lung capillary endothelial cells. CCRL2 expression was epigenetically regulated in lung endothelium and it was upregulated by the demethylating agent 5-aza-2'-deoxycytidine (5-Aza). In vivo administration of low doses of 5-Aza induced CCRL2 upregulation, increased recruitment of NK cells, and reduced lung tumor growth. These results identify CCRL2 as an NK-cell lung homing molecule that has the potential to be exploited to promote NK cell-mediated lung immune surveillance.
Collapse
Affiliation(s)
- Francesca Sozio
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur-Italia, Rome, Italy
| | - Tiziana Schioppa
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Mattia Laffranchi
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur-Italia, Rome, Italy
| | - Valentina Salvi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Nicola Tamassia
- Department of Medicine, Section of General Pathology, University of Verona, Italy
| | | | - Laura Tiberio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Raffaella Bonecchi
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Daniela Bosisio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Marc Parmentier
- WELBIO and I.R.I.B.H.M., Université Libre de Bruxelles, Brussels, Belgium
| | | | - Roberto Leone
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Eleonora Russo
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur-Italia, Rome, Italy
| | - Giovanni Bernardini
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur-Italia, Rome, Italy
| | - Stefano Garofalo
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
- IRCCS Neuromed, Pozzilli (IS), Italy
| | - Angela Gismondi
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur-Italia, Rome, Italy
| | - Giuseppe Sciumè
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur-Italia, Rome, Italy
| | - Alberto Mantovani
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
- The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Silvano Sozzani
- Department of Molecular Medicine, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur-Italia, Rome, Italy
- IRCCS Neuromed, Pozzilli (IS), Italy
| |
Collapse
|
10
|
Kudling TV, Clubb JH, Pakola S, Quixabeira DC, Lähdeniemi IA, Heiniö C, Arias V, Havunen R, Cervera-Carrascon V, Santos JM, Sutinen E, Räsänen J, Borenius K, Mäyränpää MI, Aaltonen E, Sorsa S, Hemminki O, Kanerva A, Verschuren EW, Ilonen I, Hemminki A. Effective intravenous delivery of adenovirus armed with TNFα and IL-2 improves anti-PD-1 checkpoint blockade in non-small cell lung cancer. Oncoimmunology 2023; 12:2241710. [PMID: 37546696 PMCID: PMC10399490 DOI: 10.1080/2162402x.2023.2241710] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/24/2023] [Accepted: 07/24/2023] [Indexed: 08/08/2023] Open
Abstract
Lung cancer remains among the most difficult-to-treat malignancies and is the leading cause of cancer-related deaths worldwide. The introduction of targeted therapies and checkpoint inhibitors has improved treatment outcomes; however, most patients with advanced-stage non-small cell lung cancer (NSCLC) eventually fail these therapies. Therefore, there is a major unmet clinical need for checkpoint refractory/resistant NSCLC. Here, we tested the combination of aPD-1 and adenovirus armed with TNFα and IL-2 (Ad5-CMV-mTNFα/mIL-2) in an immunocompetent murine NSCLC model. Moreover, although local delivery has been standard for virotherapy, treatment was administered intravenously to facilitate clinical translation and putative routine use. We showed that treatment of tumor-bearing animals with aPD-1 in combination with intravenously injected armed adenovirus significantly decreased cancer growth, even in the presence of neutralizing antibodies. We observed an increased frequency of cytotoxic tumor-infiltrating lymphocytes, including tumor-specific cells. Combination treatment led to a decreased percentage of immunosuppressive tumor-associated macrophages and an improvement in dendritic cell maturation. Moreover, we observed expansion of the tumor-specific memory T cell compartment in secondary lymphoid organs in the group that received aPD-1 with the virus. However, although the non-replicative Ad5-CMV-mTNFα/mIL-2 virus allows high transgene expression in the murine model, it does not fully reflect the clinical outcome in humans. Thus, we complemented our findings using NSCLC ex vivo models fully permissive for the TNFα and IL-2- armed oncolytic adenovirus TILT-123. Overall, our data demonstrate the ability of systemically administered adenovirus armed with TNFα and IL-2 to potentiate the anti-tumor efficacy of aPD-1 and warrant further investigation in clinical trials.
Collapse
Affiliation(s)
- Tatiana V. Kudling
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - James H.A. Clubb
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Santeri Pakola
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Dafne C.A. Quixabeira
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Iris A.K. Lähdeniemi
- Translational Lung Cancer Research Group, Institute for Molecular Medicine Finland (FIMM), HiLife, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Camilla Heiniö
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Victor Arias
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Riikka Havunen
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Victor Cervera-Carrascon
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Joao M. Santos
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Eva Sutinen
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- Individualized Drug Therapy Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Department of Pulmonary Medicine, Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Jari Räsänen
- General Thoracic and Esophageal Surgery, Heart and Lung Center, Helsinki University Hospital and Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Kristian Borenius
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- General Thoracic and Esophageal Surgery, Heart and Lung Center, Helsinki University Hospital and Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikko I. Mäyränpää
- Pathology, University of Helsinki and Helsinki University Hospital (HUSLAB), Helsinki, Finland
| | - Eero Aaltonen
- Faculty of Medicine, Medicum, University of Helsinki, Helsinki, Finland
| | - Suvi Sorsa
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
| | - Otto Hemminki
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
- Department of Urology, Helsinki University Hospital, Helsinki, Finland
| | - Anna Kanerva
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
- Department of Gynecology and Obstetrics, Helsinki University Hospital, Helsinki, Finland
| | - Emmy W. Verschuren
- Translational Lung Cancer Research Group, Institute for Molecular Medicine Finland (FIMM), HiLife, University of Helsinki, Helsinki, Finland
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
| | - Ilkka Ilonen
- iCAN Digital Precision Cancer Medicine Flagship, Helsinki, Finland
- General Thoracic and Esophageal Surgery, Heart and Lung Center, Helsinki University Hospital and Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- TILT Biotherapeutics Ltd, Helsinki, Finland
- Comprehensive Cancer Center, Helsinki University Hospital (HUS), Helsinki, Finland
| |
Collapse
|
11
|
Del Prete A, Salvi V, Soriani A, Laffranchi M, Sozio F, Bosisio D, Sozzani S. Dendritic cell subsets in cancer immunity and tumor antigen sensing. Cell Mol Immunol 2023; 20:432-447. [PMID: 36949244 DOI: 10.1038/s41423-023-00990-6] [Citation(s) in RCA: 156] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/14/2023] [Indexed: 03/24/2023] Open
Abstract
Dendritic cells (DCs) exhibit a specialized antigen-presenting function and play crucial roles in both innate and adaptive immune responses. Due to their ability to cross-present tumor cell-associated antigens to naïve T cells, DCs are instrumental in the generation of specific T-cell-mediated antitumor effector responses in the control of tumor growth and tumor cell dissemination. Within an immunosuppressive tumor microenvironment, DC antitumor functions can, however, be severely impaired. In this review, we focus on the mechanisms of DC capture and activation by tumor cell antigens and the role of the tumor microenvironment in shaping DC functions, taking advantage of recent studies showing the phenotype acquisition, transcriptional state and functional programs revealed by scRNA-seq analysis. The therapeutic potential of DC-mediated tumor antigen sensing in priming antitumor immunity is also discussed.
Collapse
Affiliation(s)
- Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- Humanitas Clinical and Research Center-IRCCS Rozzano, Milano, Italy
| | - Valentina Salvi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Alessandra Soriani
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Mattia Laffranchi
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Francesca Sozio
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Daniela Bosisio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Silvano Sozzani
- Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
- IRCCS Neuromed, Pozzilli, IS, Italy.
| |
Collapse
|
12
|
Xu X, Xiang Y, Yang Y, Liu K, Cui Z, Tong X, Chen J, Hou F, Luo Z. The application of tumor cell-derived vesicles in oncology therapy. Clin Transl Oncol 2023; 25:364-374. [PMID: 36207510 DOI: 10.1007/s12094-022-02966-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/26/2022] [Indexed: 06/16/2023]
Abstract
Tumor cell-derived vesicles are released by tumor cells, have a phospholipid bilayer, and are widely distributed in various biological fluids. In recent years, it has been found that tumor cell-derived vesicles contain proteins, metabolites and nucleic acids and can be delivered to recipient cells to perform their physiological functions, such as mediating specific intercellular communication, activating or inhibiting signaling pathways, participating in regulating the modulation of tumor microenvironment and influencing tumor development, which can be used for early detection and diagnosis of cancer. In addition, tumor cell-derived vesicles exhibit multiple properties in tumor therapeutic applications and may serve as a new class of delivery systems. In this review, we elaborate on the application of tumor cell-derived vesicles in oncology therapy.
Collapse
Affiliation(s)
- Ximei Xu
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China.
| | - Yin Xiang
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Yang Yang
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Kai Liu
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Zhiwei Cui
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Xiaodong Tong
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Junliang Chen
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Fang Hou
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| | - Zhiqiang Luo
- The People's Hospital of Leshan, Leshan, 614000, Sichuan, China
| |
Collapse
|
13
|
Oliveira FMS, Cruz RE, Pinheiro GRG, Caliari MV. Comorbidities involving parasitic diseases: A look at the benefits and complications. Exp Biol Med (Maywood) 2022; 247:1819-1826. [PMID: 35876147 PMCID: PMC9679356 DOI: 10.1177/15353702221108387] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Parasitic infections acquired by the population cause substantial morbidity worldwide, with individuals from developing countries being most affected. Some parasites remain in the host for long periods, settling in different organs, manipulating the flow of nutrients and metabolites, and influencing the immune response, favoring their adaptation. The host attempts to counteract the metabolic and immunological alterations and the possible damage caused by infection. These metabolic and immunological changes experienced by the host can influence the progression of other existing morbidities or those that will be acquired in the future. Cancer and metabolic diseases are also frequent causes of morbidity in the world population. The large numbers of individuals affected by cancer and metabolic diseases and the high prevalence of morbidity caused by parasitic diseases favor the development of comorbidity involving these pathologies. This review provides an overview of major advances in research on cancer and metabolic diseases associated with parasitic infections. Information about hosts and parasites such as alterations of the immune response, metabolism and adaptation mechanisms of the parasites, and parasitic molecules with therapeutic potential is provided, as well as the beneficial results or complications related to the comorbidities discussed herein. We emphasize the need to conduct additional studies addressing comorbidities associated with parasitic infections to improve the understanding of the impact of this association on the progression of morbidities, as well as the possibility of the therapeutic use of and therapeutic approaches involving parasites.
Collapse
Affiliation(s)
- Fabrício Marcus Silva Oliveira
- Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Ruth Elizabeth Cruz
- Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Guilherme Rafael Gomide Pinheiro
- Department of Preventive Veterinary Medicine, Veterinary School, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Marcelo Vidigal Caliari
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil,Marcelo Vidigal Caliari.
| |
Collapse
|
14
|
Zhao Y, Bai Y, Shen M, Li Y. Therapeutic strategies for gastric cancer targeting immune cells: Future directions. Front Immunol 2022; 13:992762. [PMID: 36225938 PMCID: PMC9549957 DOI: 10.3389/fimmu.2022.992762] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Gastric cancer (GC) is a malignancy with a high incidence and mortality, and the emergence of immunotherapy has brought survival benefits to GC patients. Compared with traditional therapy, immunotherapy has the advantages of durable response, long-term survival benefits, and lower toxicity. Therefore, targeted immune cells are the most promising therapeutic strategy in the field of oncology. In this review, we introduce the role and significance of each immune cell in the tumor microenvironment of GC and summarize the current landscape of immunotherapy in GC, which includes immune checkpoint inhibitors, adoptive cell therapy (ACT), dendritic cell (DC) vaccines, reduction of M2 tumor-associated macrophages (M2 TAMs), N2 tumor-associated neutrophils (N2 TANs), myeloid-derived suppressor cells (MDSCs), effector regulatory T cells (eTregs), and regulatory B cells (Bregs) in the tumor microenvironment and reprogram TAMs and TANs into tumor killer cells. The most widely used immunotherapy strategies are the immune checkpoint inhibitor programmed cell death 1/programmed death-ligand 1 (PD-1/PD-L1) antibody, cytotoxic T lymphocyte–associated protein 4 (CTLA-4) antibody, and chimeric antigen receptor T (CAR-T) in ACT, and these therapeutic strategies have significant anti-tumor efficacy in solid tumors and hematological tumors. Targeting other immune cells provides a new direction for the immunotherapy of GC despite the relatively weak clinical data, which have been confirmed to restore or enhance anti-tumor immune function in preclinical studies and some treatment strategies have entered the clinical trial stage, and it is expected that more and more effective immune cell–based therapeutic methods will be developed and applied.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yuansong Bai
- Department of Oncology and Hematology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Meili Shen
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun, China
- *Correspondence: Yapeng Li, ; Meili Shen,
| | - Yapeng Li
- The National and Local Joint Engineering Laboratory for Synthesis Technology of High Performance Polymer, College of Chemistry, Jilin University, Changchun, China
- *Correspondence: Yapeng Li, ; Meili Shen,
| |
Collapse
|
15
|
Seliger B, Koehl U. Underlying mechanisms of evasion from NK cells as rational for improvement of NK cell-based immunotherapies. Front Immunol 2022; 13:910595. [PMID: 36045670 PMCID: PMC9422402 DOI: 10.3389/fimmu.2022.910595] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Natural killer (NK) cells belong to the family of innate immune cells with the capacity to recognize and kill tumor cells. Different phenotypes and functional properties of NK cells have been described in tumor patients, which could be shaped by the tumor microenvironment. The discovery of HLA class I-specific inhibitory receptors controlling NK cell activity paved the way to the fundamental concept of modulating immune responses that are regulated by an array of inhibitory receptors, and emphasized the importance to explore the potential of NK cells in cancer therapy. Although a whole range of NK cell-based approaches are currently being developed, there are still major challenges that need to be overcome for improved efficacy of these therapies. These include escape of tumor cells from NK cell recognition due to their expression of inhibitory molecules, immune suppressive signals of NK cells, reduced NK cell infiltration of tumors, an immune suppressive micromilieu and limited in vivo persistence of NK cells. Therefore, this review provides an overview about the NK cell biology, alterations of NK cell activities, changes in tumor cells and the tumor microenvironment contributing to immune escape or immune surveillance by NK cells and their underlying molecular mechanisms as well as the current status and novel aspects of NK cell-based therapeutic strategies including their genetic engineering and their combination with conventional treatment options to overcome tumor-mediated evasion strategies and improve therapy efficacy.
Collapse
Affiliation(s)
- Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
- *Correspondence: Barbara Seliger,
| | - Ulrike Koehl
- Fraunhofer Institute for Cell Therapy and Immunology, Leipzig, Germany
- Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
- Institute of Cellular Therapeutics, Hannover Medical School, Hannover, Germany
| |
Collapse
|
16
|
Hassanian H, Asadzadeh Z, Baghbanzadeh A, Derakhshani A, Dufour A, Rostami Khosroshahi N, Najafi S, Brunetti O, Silvestris N, Baradaran B. The expression pattern of Immune checkpoints after chemo/radiotherapy in the tumor microenvironment. Front Immunol 2022; 13:938063. [PMID: 35967381 PMCID: PMC9367471 DOI: 10.3389/fimmu.2022.938063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
As a disease with the highest disease-associated burden worldwide, cancer has been the main subject of a considerable proportion of medical research in recent years, intending to find more effective therapeutic approaches with fewer side effects. Combining conventional methods with newer biologically based treatments such as immunotherapy can be a promising approach to treating different tumors. The concept of "cancer immunoediting" that occurs in the field of the tumor microenvironment (TME) is the aspect of cancer therapy that has not been at the center of attention. One group of the role players of the so-called immunoediting process are the immune checkpoint molecules that exert either co-stimulatory or co-inhibitory effects in the anti-tumor immunity of the host. It involves alterations in a wide variety of immunologic pathways. Recent studies have proven that conventional cancer therapies, such as chemotherapy, radiotherapy, or a combination of them, i.e., chemoradiotherapy, alter the "immune compartment" of the TME. The mentioned changes encompass a wide range of variations, including the changes in the density and immunologic type of the tumor-infiltrating lymphocytes (TILs) and the alterations in the expression patterns of the different immune checkpoints. These rearrangements can have either anti-tumor immunity empowering or immune attenuating sequels. Thus, recognizing the consequences of various chemo(radio)therapeutic regimens in the TME seems to be of great significance in the evolution of therapeutic approaches. Therefore, the present review intends to summarize how chemo(radio)therapy affects the TME and specifically some of the most important, well-known immune checkpoints' expressions according to the recent studies in this field.
Collapse
Affiliation(s)
- Hamidreza Hassanian
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afshin Derakhshani
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB, Canada
- McCaig Insitute, Hotchkiss Brain Institute, and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
| | - Antoine Dufour
- McCaig Insitute, Hotchkiss Brain Institute, and Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB, Canada
- Departments of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | | | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Oronzo Brunetti
- Medical Oncology Unit, IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology “G. Barresi” University of Messina, Messina, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|