1
|
Shen Z, Meng X, Rautela J, Chopin M, Huntington ND. Adjusting the scope of natural killer cells in cancer therapy. Cell Mol Immunol 2025:10.1038/s41423-025-01297-4. [PMID: 40410571 DOI: 10.1038/s41423-025-01297-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 05/06/2025] [Indexed: 05/25/2025] Open
Abstract
Natural killer (NK) cells have evolved to detect abnormalities in tissues arising from infection with pathogens, genomic damage, or transformation and respond rapidly to the production of potent proinflammatory and cytolytic mediators. While this acute proinflammatory response is highly efficient at orchestrating sterilizing immunity to pathogens in a matter of days, cellular transformation often avoids the innate detection mechanisms of NK cells. When cellular transformation results in malignancy, tumor cells and/or the tumor microenvironment can evolve additional mechanisms to circumvent NK cell responses, and cancer is now a dominant disease burden worldwide. Here, we review recent advances in our understanding of the combined relationship between malignancies and natural killer (NK) cells, learn from recent clinical efforts in therapeutically targeting natural killer (NK) cells in cancer and outline some emerging therapeutic concepts that aim to improve the innate immune response against cancer.
Collapse
Affiliation(s)
- Zihen Shen
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Xiangpeng Meng
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Jai Rautela
- oNKo-Innate Pty Ltd., Moonee Ponds, VIC, Australia
| | - Michael Chopin
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Nicholas D Huntington
- Biomedicine Discovery Institute and the Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia.
- oNKo-Innate Pty Ltd., Moonee Ponds, VIC, Australia.
| |
Collapse
|
2
|
Wu N, Zhang Q, Tang R, Deng L, Cao Y, Fu B, Dong H, Huang Z, Wan L, He H, Lin Y, Liu J, Xia C, Li P. Ultrasound Visualization of Spatiotemporal Autophagy-Regulated Nanodroplets for Amplifying ICB in Melanoma via Remodeling Tumor Inflammatory Microenvironment. ACS APPLIED MATERIALS & INTERFACES 2025; 17:29364-29378. [PMID: 40331917 PMCID: PMC12100593 DOI: 10.1021/acsami.5c03394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 04/27/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025]
Abstract
Immune checkpoint blockade (ICB) therapy, represented by anti-PD-1/PD-L1 antibodies, is confronted with difficulties of unsatisfied response rates owing to the prevalence of "cold" immune tumor microenvironment (TME) in most cancers. Blocking cytoprotective autophagy has emerged as a potential strategy to remodel the inflammatory TME. Nevertheless, the dual roles of autophagy in tumor progression, coupled with the poor pharmacokinetic properties of small-molecule autophagy inhibitors, significantly restrict clinical applications. To address these challenges, a low-intensity focused ultrasound (LIFU) responsive phase-change nanodroplet delivery platform (SP@Lip-PEG) is elaborately developed to deliver specific autophagy inhibitor SAR405 for activating typical tumor-resident immune cells. The PEG-modified nanodroplets effectively accumulate into the tumor site. Upon LIFU activation, SP@Lip-PEG transforms into microbubbles through acoustic droplet vaporization (ADV) effects, enabling the controlled release of SAR405 under ultrasound imaging guidance. The released SAR405 significantly triggered the upregulation of proinflammatory factors CCL5 and CXCL10 through autophagy manipulation, creating an inflammatory TME to facilitate the recruitment of natural killer (NK) cells and CD8+ T cells, along with promoting dendritic cell (DC) maturation and synergistically enhancing ICB efficacy. With the high specificity of SAR405 and the controllable therapeutic process under LIFU irradiation, this noninvasive, efficient, and cost-effective drug delivery vector opened new horizons for conquering the clinical dilemma of rescuing ICB response rates.
Collapse
Affiliation(s)
- Nianhong Wu
- Department
of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular
Imaging and Therapy, The Second Affiliated
Hospital of Chongqing Medical University, Chongqing400010, China
| | - Qin Zhang
- Department
of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular
Imaging and Therapy, The Second Affiliated
Hospital of Chongqing Medical University, Chongqing400010, China
- Department
of Radiology, Chongqing Traditional Chinese
Medicine Hospital, Chongqing400021, China
- Chongqing
College of Traditional Chinese Medicine, Chongqing402760, China
| | - Rui Tang
- Department
of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular
Imaging and Therapy, The Second Affiliated
Hospital of Chongqing Medical University, Chongqing400010, China
| | - Liming Deng
- Department
of Ultrasound, The First Affiliated Hospital
of Chongqing Medical University, Chongqing400016, China
| | - Yuting Cao
- Department
of Ultrasound, The Second Xiangya Hospital, Central South University, Changsha, Hunan410011, China
| | - Benxin Fu
- Department
of Ultrasound, The Third Hospital of Mianyang,
Sichuan Mental Health Center, Mianyang621000, China
| | - Hongmei Dong
- Department
of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular
Imaging and Therapy, The Second Affiliated
Hospital of Chongqing Medical University, Chongqing400010, China
| | - Zeyan Huang
- Department
of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular
Imaging and Therapy, The Second Affiliated
Hospital of Chongqing Medical University, Chongqing400010, China
| | - Li Wan
- Department
of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular
Imaging and Therapy, The Second Affiliated
Hospital of Chongqing Medical University, Chongqing400010, China
| | - Hongye He
- Department
of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular
Imaging and Therapy, The Second Affiliated
Hospital of Chongqing Medical University, Chongqing400010, China
| | - Yi Lin
- Department
of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular
Imaging and Therapy, The Second Affiliated
Hospital of Chongqing Medical University, Chongqing400010, China
| | - Junjie Liu
- Department
of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular
Imaging and Therapy, The Second Affiliated
Hospital of Chongqing Medical University, Chongqing400010, China
| | - Chunyu Xia
- Department
of Pharmacy, The Second Affiliated Hospital
of Chongqing Medical University, Chongqing400010, China
| | - Pan Li
- Department
of Ultrasound and Chongqing Key Laboratory of Ultrasound Molecular
Imaging and Therapy, The Second Affiliated
Hospital of Chongqing Medical University, Chongqing400010, China
| |
Collapse
|
3
|
Qin X, Xu W, Wu J, Li M. Integration of single-cell and bulk RNA-sequencing data to construct and validate a signature based on NK cell marker genes to predict immunotherapy response and prognosis in colorectal cancer. Discov Oncol 2025; 16:134. [PMID: 39920524 PMCID: PMC11805743 DOI: 10.1007/s12672-025-01842-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/20/2025] [Indexed: 02/09/2025] Open
Abstract
We aimed to create a NK cell marker genes-based signature to predict immunotherapy response and prognosis in colorectal cancer. We integrated scRNA-seq data from four Gene Expression Omnibus (GEO) samples and performed Weighted gene correlation network analysis (WGCNA) based on 587 the Cancer Genome Atlas (TCGA) colorectal cancer samples to uncover NK cell-related genes. We identified 1080 NK cell-related core genes and 276 NK cell-related feature genes based on WGCNA and clustering and annotation of scRNA-seq data, respectively. Six key NK cell-related prognostic signature genes were obtained by univariate and LASSO regression analyses, including ADAM8, CTSD, CCL4, IL2RB, TTC38, and PLEK. Two validation cohorts from the GEO dataset, comprising 124 and 201 samples respectively, were used. The signature was significantly associated with overall survival and correlated with immune cell infiltration, immune and stromal scores, and immune checkpoint genes. Furthermore, the signature was associated with the homologous recombination deficiency (HRD) and T-cell receptor (TCR) scores. In conclusion, our study proposes a new prognostic signature based on NK cell marker genes, which may serve as a potential tool to predict overall survival and immunotherapy response for CRC patients.
Collapse
Affiliation(s)
- Xiaoyu Qin
- Department of Gastroenterology, Shanghai Pudong New Area Gongli Hospital, Shanghai, 200135, China
| | - Wenjuan Xu
- Department of General Surgery, Shanghai Punan Hospital, Pudong New District, Shanghai, 200125, China
| | - Jinxiu Wu
- Department of General Surgery, Shanghai Punan Hospital, Pudong New District, Shanghai, 200125, China
| | - Ming Li
- Department of General Surgery, Shanghai Punan Hospital, Pudong New District, Shanghai, 200125, China.
| |
Collapse
|
4
|
Hounye AH, Xiong L, Hou M. Integrated explainable machine learning and multi-omics analysis for survival prediction in cancer with immunotherapy response. Apoptosis 2025; 30:364-388. [PMID: 39633110 DOI: 10.1007/s10495-024-02050-4] [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] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
To demonstrate the efficacy of machine learning models in predicting mortality in melanoma cancer, we developed an interpretability model for better understanding the survival prediction of cancer. To this end, the optimal features were identified, ten different machine learning models were utilized to predict mortality across various datasets. Then we have utilized the important features identified by those machines learning methods to construct a new model named NKECLR to forecast mortality of patient with cancer. To explicitly clarify the model's decision-making process and uncover novel findings, an interpretable technique incorporating machine learning and SHapley Additive exPlanations (SHAP), as well as LIME, has been employed, and four genes EPGN, PHF11, RBM34, and ZFP36 were identified from those machine learning(ML). The experimental analysis conducted on training and validation datasets demonstrated that the proposed model has a good performance com- pared to existing methods with AUC value 81.8%, and 79.3%, respectively. Moreover, when combined our NKECLR with PD-L1, PD-1, and CTLA-4 the AUC value was 83%0. Finally, these findings have been applied to comprehend the response of drugs and immunotherapy. Our research introduced an innovative predictive NKECLR model utilizing natural killer(NK) cell marker genes for cohorts with melanoma cancer. The NKECLR model can effectively predict the survival of melanoma cancer cohorts and treatment results, revealing distinct immune cell infiltration in the high-risk group.
Collapse
Affiliation(s)
- Alphonse Houssou Hounye
- General surgery department of Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China.
| | - Li Xiong
- General surgery department of Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, Hunan, China.
| | - Muzhou Hou
- School of Mathematics and Statistics, Central South University, Changsha, 410083, China
| |
Collapse
|
5
|
Ehlers FAI, Blise KE, Betts CB, Sivagnanam S, Kooreman LFS, Hwang ES, Bos GMJ, Wieten L, Coussens LM. Natural killer cells occupy unique spatial neighborhoods in HER2 - and HER2 + human breast cancers. Breast Cancer Res 2025; 27:14. [PMID: 39856748 PMCID: PMC11762118 DOI: 10.1186/s13058-025-01964-4] [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: 08/25/2024] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Tumor-infiltrating lymphocytes are considered clinically beneficial in breast cancer, but the significance of natural killer (NK) cells is less well characterized. As increasing evidence has demonstrated that the spatial organization of immune cells in tumor microenvironments is a significant parameter for impacting disease progression as well as therapeutic responses, an improved understanding of tumor-infiltrating NK cells and their location within tumor contextures is needed to improve the design of effective NK cell-based therapies. In this study, we developed a multiplex immunohistochemistry (mIHC) antibody panel designed to quantitatively interrogate leukocyte lineages, focusing on NK cells and their phenotypes, in two independent breast cancer patient cohorts (n = 26 and n = 30). Owing to the clinical evidence supporting a significant role for NK cells in HER2+ breast cancer in mediating responses to Trastuzumab, we further evaluated HER2- and HER2+ specimens separately. Consistent with literature, we found that CD3+ T cells were the dominant leukocyte subset across breast cancer specimens. In comparison, NK cells, identified by CD56 or NKp46 expression, were scarce in all specimens with low granzyme B expression indicating reduced cytotoxic functionality. Whereas NK cell density and phenotype did not appear to be influenced by HER2 status, spatial analysis revealed distinct NK cells phenotypes regarding their proximity to neoplastic tumor cells that associated with HER2 status. Spatial cellular neighborhood analysis revealed multiple unique neighborhood compositions surrounding NK cells, where NK cells from HER2- tumors were more frequently found proximal to neoplastic tumor cells, whereas NK cells from HER2+ tumors were instead more frequently found proximal to CD3+ T cells. This study establishes the utility of quantitative mIHC to evaluate NK cells at the single-cell spatial proteomics level and illustrates how spatial characteristics of NK cell neighborhoods vary within the context of HER2- and HER2+ breast cancers.
Collapse
Affiliation(s)
- Femke A I Ehlers
- Department of Transplantation Immunology, Tissue Typing Laboratory, Maastricht University Medical Center+, Maastricht, 6229, HX, The Netherlands
- Department of Internal Medicine, Division of Hematology, Maastricht University Medical Center+, Maastricht, 6229 HX, The Netherlands
- GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, 6229 GT, The Netherlands
| | - Katie E Blise
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Courtney B Betts
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Shamilene Sivagnanam
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Loes F S Kooreman
- GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, 6229 GT, The Netherlands
- Department of Pathology, Maastricht University Medical Center+, Maastricht, 6229 HX, The Netherlands
| | - E Shelley Hwang
- Department of Surgery, Duke University Medical Center, Durham, NC, 27710, USA
| | - Gerard M J Bos
- Department of Internal Medicine, Division of Hematology, Maastricht University Medical Center+, Maastricht, 6229 HX, The Netherlands
- GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, 6229 GT, The Netherlands
| | - Lotte Wieten
- Department of Transplantation Immunology, Tissue Typing Laboratory, Maastricht University Medical Center+, Maastricht, 6229, HX, The Netherlands.
- GROW - School for Oncology and Reproduction, Maastricht University, Maastricht, 6229 GT, The Netherlands.
| | - Lisa M Coussens
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, 97239, USA.
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA.
- , 2720 S Moody Ave, #KC-CDCB, Portland, OR, 97201 - 5012, USA.
| |
Collapse
|
6
|
Liu Y, Li C, Deng W. Uncovering the Heterogeneity of Signaling Pathways in Skin Cutaneous Melanoma: Insights into Prognostic Values and Immune Interactions. Clin Cosmet Investig Dermatol 2025; 18:47-59. [PMID: 39802668 PMCID: PMC11725243 DOI: 10.2147/ccid.s500654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/01/2025] [Indexed: 01/16/2025]
Abstract
Background Signaling pathways play crucial roles in tumor cells. However, functional heterogeneity of signaling pathways in skin cutaneous melanoma (SKCM) has not been established. Methods Based on a recent computational pipeline, pathway activities between SKCM and normal samples were identified. Results The results showed that high activities in 12 pathways were associated with poor prognoses, while high activities in 17 pathways were associated with favorable prognoses. Interestingly, elevated metabolic pathway activity was unfavorable, whereas elevated immune activity was favorable for SKCM. Unfavorably elevated metabolic pathways strongly correlated with Wnt/beta-catenin signaling. Conversely, favorable pathways, such as glycosaminoglycan biosynthesis and keratan sulfate, were strongly correlated with anti-tumor pathways. Moreover, the activities of favorable pathways were strongly positively correlated with infiltrating CD8+ T cells, macrophages M1, immune score, and stromal score, all of which were favorable for SKCM. Conclusion Taken together, our study provides insights into the characteristics of several pathways in SKCM.
Collapse
Affiliation(s)
- Yufang Liu
- Department of Dermatology and Venereology, Fuyang People’s Hospital, Fuyang, Anhui, 236000, People’s Republic of China
| | - Chunyan Li
- Department of Dermatology and Venereology, Dermatology Hospital of Southern Medical University, Department of Dermatology, Guangzhou, People’s Republic of China
| | - Weiwei Deng
- Department of Dermatology and Venereology, Dermatology Hospital of Southern Medical University, Department of Dermatology, Guangzhou, People’s Republic of China
| |
Collapse
|
7
|
Hurtado M, Khajavi L, Essabbar A, Kammer M, Xie T, Coullomb A, Pradines A, Casanova A, Kruczynski A, Gouin S, Clermont E, Boutillet L, Senosain MF, Zou Y, Zhao S, Burq P, Mahfoudi A, Besse J, Launay P, Passioukov A, Chetaille E, Favre G, Maldonado F, Cruzalegui F, Delfour O, Mazières J, Pancaldi V. Transcriptomics profiling of the non-small cell lung cancer microenvironment across disease stages reveals dual immune cell-type behaviors. Front Immunol 2024; 15:1394965. [PMID: 39606240 PMCID: PMC11600981 DOI: 10.3389/fimmu.2024.1394965] [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: 03/02/2024] [Accepted: 09/24/2024] [Indexed: 11/29/2024] Open
Abstract
Background Lung cancer is the leading cause of cancer death worldwide, with poor survival despite recent therapeutic advances. A better understanding of the complexity of the tumor microenvironment is needed to improve patients' outcome. Methods We applied a computational immunology approach (involving immune cell proportion estimation by deconvolution, transcription factor activity inference, pathways and immune scores estimations) in order to characterize bulk transcriptomics of 62 primary lung adenocarcinoma (LUAD) samples from patients across disease stages. Focusing specifically on early stage samples, we validated our findings using an independent LUAD cohort with 70 bulk RNAseq and 15 scRNAseq datasets and on TCGA datasets. Results Through our methodology and feature integration pipeline, we identified groups of immune cells related to disease stage as well as potential immune response or evasion and survival. More specifically, we reported a duality in the behavior of immune cells, notably natural killer (NK) cells, which was shown to be associated with survival and could be relevant for immune response or evasion. These distinct NK cell populations were further characterized using scRNAseq data, showing potential differences in their cytotoxic activity. Conclusion The dual profile of several immune cells, most notably T-cell populations, have been discussed in the context of diseases such as cancer. Here, we report the duality of NK cells which should be taken into account in conjunction with other immune cell populations and behaviors in predicting prognosis, immune response or evasion.
Collapse
Affiliation(s)
- Marcelo Hurtado
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
| | - Leila Khajavi
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
| | - Abdelmounim Essabbar
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
| | - Michael Kammer
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Ting Xie
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
| | - Alexis Coullomb
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
| | - Anne Pradines
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
- Laboratory Medicine, Oncopole Claudius Regaud, Toulouse, France
| | - Anne Casanova
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
- Laboratory Medicine, Oncopole Claudius Regaud, Toulouse, France
| | | | - Sandrine Gouin
- Pulmonology Department, Larrey Hospital, University Hospital of Toulouse, Toulouse, France
| | - Estelle Clermont
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
- Laboratory Medicine, Oncopole Claudius Regaud, Toulouse, France
| | - Léa Boutillet
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
| | - Maria Fernanda Senosain
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical. Center, Nashville, TN, United States
| | - Yong Zou
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Cancer Early Detection and Prevention Initiative, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical. Center, Nashville, TN, United States
| | - Shillin Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Prosper Burq
- Data Science, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | | | - Jerome Besse
- Institut de Recherche Pierre Fabre, Toulouse, France
| | - Pierre Launay
- Institut de Recherche Pierre Fabre, Toulouse, France
| | | | | | - Gilles Favre
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
| | - Fabien Maldonado
- Division of Allergy, Pulmonary, and Critical Care Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | | | | | - Julien Mazières
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
- Pulmonology Department, Larrey Hospital, University Hospital of Toulouse, Toulouse, France
| | - Vera Pancaldi
- CRCT, Université de Toulouse, Institut national de la santé et de la recherche médicale (Inserm), Centre national de la recherche scientifique (CNRS), Université Toulouse III-Paul Sabatier, Centre de Recherches en cancérologie de Toulouse, Toulouse, France
- Life Sciences Department, Barcelona Supercomputing Center, Barcelona, Spain
| |
Collapse
|
8
|
Wessel RE, Ageeb N, Obeid JM, Mauldin IS, Goundry KA, Hanson GF, Hossain M, Lehman C, Gentzler RD, Wages NA, Slingluff Jr CL, Bullock TNJ, Dolatshahi S, Brown MG. Spatial colocalization and combined survival benefit of natural killer and CD8 T cells despite profound MHC class I loss in non-small cell lung cancer. J Immunother Cancer 2024; 12:e009126. [PMID: 39299754 PMCID: PMC11418484 DOI: 10.1136/jitc-2024-009126] [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] [Accepted: 08/13/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Major histocompatibility complex class I (MHC-I) loss is frequent in non-small cell lung cancer (NSCLC) rendering tumor cells resistant to T cell lysis. NK cells kill MHC-I-deficient tumor cells, and although previous work indicated their presence at NSCLC margins, they were functionally impaired. Within, we evaluated whether NK cell and CD8 T cell infiltration and activation vary with MHC-I expression. METHODS We used single-stain immunohistochemistry (IHC) and Kaplan-Meier analysis to test the effect of NK cell and CD8 T cell infiltration on overall and disease-free survival. To delineate immune covariates of MHC-I-disparate lung cancers, we used multiplexed immunofluorescence (mIF) imaging followed by multivariate statistical modeling. To identify differences in infiltration and intercellular communication between IFNγ-activated and non-activated lymphocytes, we developed a computational pipeline to enumerate single-cell neighborhoods from mIF images followed by multivariate discriminant analysis. RESULTS Spatial quantitation of tumor cell MHC-I expression revealed intratumoral and intertumoral heterogeneity, which was associated with the local lymphocyte landscape. IHC analysis revealed that high CD56+ cell numbers in patient tumors were positively associated with disease-free survival (HR=0.58, p=0.064) and overall survival (OS) (HR=0.496, p=0.041). The OS association strengthened with high counts of both CD56+ and CD8+ cells (HR=0.199, p<1×10-3). mIF imaging and multivariate discriminant analysis revealed enrichment of both CD3+CD8+ T cells and CD3-CD56+ NK cells in MHC-I-bearing tumors (p<0.05). To infer associations of functional cell states and local cell-cell communication, we analyzed spatial single-cell neighborhood profiles to delineate the cellular environments of IFNγ+/- NK cells and T cells. We discovered that both IFNγ+ NK and CD8 T cells were more frequently associated with other IFNγ+ lymphocytes in comparison to IFNγ- NK cells and CD8 T cells (p<1×10-30). Moreover, IFNγ+ lymphocytes were most often found clustered near MHC-I+ tumor cells. CONCLUSIONS Tumor-infiltrating NK cells and CD8 T cells jointly affected control of NSCLC tumor progression. Coassociation of NK and CD8 T cells was most evident in MHC-I-bearing tumors, especially in the presence of IFNγ. Frequent colocalization of IFNγ+ NK cells with other IFNγ+ lymphocytes in near-neighbor analysis suggests NSCLC lymphocyte activation is coordinately regulated.
Collapse
Affiliation(s)
- Remziye E Wessel
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Nardin Ageeb
- Department of Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Joseph M Obeid
- Department of Thoracic Surgery, Temple University Hospital, Philadelphia, Pennsylvania, USA
| | - Ileana S Mauldin
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Kate A Goundry
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Gabriel F Hanson
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Mahdin Hossain
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, USA
| | - Chad Lehman
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, USA
| | - Ryan D Gentzler
- Department of Medicine, Hematology and Oncology, University of Virginia, Charlottesville, Virginia, USA
| | - Nolan A Wages
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia, USA
| | | | - Timothy N J Bullock
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, USA
- Department of Pathology, University of Virginia, Charlottesville, Virginia, USA
| | - Sepideh Dolatshahi
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, USA
| | - Michael G Brown
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, Virginia, USA
- Department of Medicine, Nephrology Division, University of Virginia, Charlottesville, Virginia, USA
- Center for Immunity, Inflammation and Regenerative Medicine, University of Virginia School of Medicine, Charlottesville, Virginia, USA
- Department of Microbiology, Immunology and Cancer Biology, Univesity of Virginia, School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
9
|
Wang K, Peng B, Xu R, Lu T, Chang X, Shen Z, Shi J, Li M, Wang C, Zhou X, Xu C, Chang H, Zhang L. Comprehensive analysis of PPP4C's impact on prognosis, immune microenvironment, and immunotherapy response in lung adenocarcinoma using single-cell sequencing and multi-omics. Front Immunol 2024; 15:1416632. [PMID: 39026674 PMCID: PMC11254641 DOI: 10.3389/fimmu.2024.1416632] [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: 04/12/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Background Elevated PPP4C expression has been associated with poor prognostic implications for patients suffering from lung adenocarcinoma (LUAD). The extent to which PPP4C affects immune cell infiltration in LUAD, as well as the importance of associated genes in clinical scenarios, still requires thorough investigation. Methods In our investigation, we leveraged both single-cell and comprehensive RNA sequencing data, sourced from LUAD patients, in our analysis. This study also integrated datasets of immune-related genes from InnateDB into the framework. Our expansive evaluation employed various analytical techniques; these included pinpointing differentially expressed genes, constructing WGCNA, implementing Cox proportional hazards models. We utilized these methods to investigate the gene expression profiles of PPP4C within the context of LUAD and to clarify its potential prognostic value for patients. Subsequent steps involved validating the observed enhancement of PPP4C expression in LUAD samples through a series of experimental approaches. The array comprised immunohistochemistry staining, Western blotting, quantitative PCR, and a collection of cell-based assays aimed at evaluating the influence of PPP4C on the proliferative and migratory activities of LUAD cells. Results In lung cancer, elevated expression levels of PPP4C were observed, correlating with poorer patient prognoses. Validation of increased PPP4C levels in LUAD specimens was achieved using immunohistochemical techniques. Experimental investigations have substantiated the role of PPP4C in facilitating cellular proliferation and migration in LUAD contexts. Furthermore, an association was identified between the expression of PPP4C and the infiltration of immune cells in these tumors. A prognostic framework, incorporating PPP4C and immune-related genes, was developed and recognized as an autonomous predictor of survival in individuals afflicted with LUAD. This prognostic tool has demonstrated considerable efficacy in forecasting patient survival and their response to immunotherapeutic interventions. Conclusion The involvement of PPP4C in LUAD is deeply intertwined with the tumor's immune microenvironment. PPP4C's over-expression is associated with negative clinical outcomes, promoting both tumor proliferation and spread. A prognostic framework based on PPP4C levels may effectively predict patient prognoses in LUAD, as well as the efficacy of immunotherapy strategy. This research sheds light on the mechanisms of immune interaction in LUAD and proposes a new strategy for treatment.
Collapse
Affiliation(s)
- Kaiyu Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Peng
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ran Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Tong Lu
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaoyan Chang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhiping Shen
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jiaxin Shi
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Meifeng Li
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chenghao Wang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiang Zhou
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chengyu Xu
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hao Chang
- Department of Thoracic Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Linyou Zhang
- Department of Thoracic Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
10
|
Wessel RE, Ageeb N, Obeid JM, Mauldin I, Goundry KA, Hanson GF, Hossain M, Lehman C, Gentzler RD, Wages NA, Slingluff CL, Bullock TNJ, Dolatshahi S, Brown MG. Spatial colocalization and combined survival benefit of natural killer and CD8 T cells despite profound MHC class I loss in non-small cell lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.20.581048. [PMID: 38979183 PMCID: PMC11230195 DOI: 10.1101/2024.02.20.581048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Background MHC class I (MHC-I) loss is frequent in non-small cell lung cancer (NSCLC) rendering tumor cells resistant to T cell lysis. NK cells kill MHC-I-deficient tumor cells, and although previous work indicated their presence at NSCLC margins, they were functionally impaired. Within, we evaluated whether NK cell and CD8 T cell infiltration and activation vary with MHC-I expression. Methods We used single-stain immunohistochemistry (IHC) and Kaplan-Meier analysis to test the effect of NK cell and CD8 T cell infiltration on overall and disease-free survival. To delineate immune covariates of MHC-I-disparate lung cancers, we used multiplexed immunofluorescence (mIF) imaging followed by multivariate statistical modeling. To identify differences in infiltration and intercellular communication between IFNγ-activated and non-activated lymphocytes, we developed a computational pipeline to enumerate single cell neighborhoods from mIF images followed by multivariate discriminant analysis. Results Spatial quantitation of tumor cell MHC-I expression revealed intra- and inter-tumoral heterogeneity, which was associated with the local lymphocyte landscape. IHC analysis revealed that high CD56+ cell numbers in patient tumors were positively associated with disease-free survival (DFS) (HR=0.58, p=0.064) and overall survival (OS) (HR=0.496, p=0.041). The OS association strengthened with high counts of both CD56+ and CD8+ cells (HR=0.199, p<1×10-3). mIF imaging and multivariate discriminant analysis revealed enrichment of both CD3+CD8+ T cells and CD3-CD56+ NK cells in MHC-I-bearing tumors (p<0.05). To infer associations of functional cell states and local cell-cell communication, we analyzed spatial single cell neighborhood profiles to delineate the cellular environments of IFNγ+/- NK cells and T cells. We discovered that both IFNγ+ NK and CD8 T cells were more frequently associated with other IFNγ+ lymphocytes in comparison to IFNγ- NK cells and CD8 T cells (p<1×10-30). Moreover, IFNγ+ lymphocytes were most often found clustered near MHC-I+ tumor cells. Conclusions Tumor-infiltrating NK cells and CD8 T cells jointly affected control of NSCLC tumor progression. Co-association of NK and CD8 T cells was most evident in MHC-I-bearing tumors, especially in the presence of IFNγ. Frequent co-localization of IFNγ+ NK cells with other IFNγ+ lymphocytes in near-neighbor analysis suggests NSCLC lymphocyte activation is coordinately regulated.
Collapse
Affiliation(s)
- Remziye E Wessel
- Department of Biomedical Engineering, University of Virginia (UVA) School of Medicine, Charlottesville, Virginia 22908
| | - Nardin Ageeb
- Department of Biology, UVA, Charlottesville, Virginia 22908
| | - Joseph M Obeid
- Department of Thoracic Surgery, Temple University Hospital, Philadelphia, Pennsylvania 19140
| | - Ileana Mauldin
- Department of Surgery, UVA School of Medicine, Charlottesville, Virginia 22908
| | - Kate A Goundry
- Department of Biomedical Engineering, University of Virginia (UVA) School of Medicine, Charlottesville, Virginia 22908
| | - Gabriel F Hanson
- Department of Biomedical Engineering, University of Virginia (UVA) School of Medicine, Charlottesville, Virginia 22908
| | - Mahdin Hossain
- Beirne B. Carter Center for Immunology Research, UVA School of Medicine, Charlottesville, Virginia 22908
| | - Chad Lehman
- Beirne B. Carter Center for Immunology Research, UVA School of Medicine, Charlottesville, Virginia 22908
| | - Ryan D Gentzler
- Department of Medicine, Hematology and Oncology, UVA School of Medicine, Charlottesville, Virginia 22908
| | - Nolan A Wages
- Department of Biostatistics, Virginia Commonwealth University, Richmond, Virginia 23298-0032
| | - Craig L Slingluff
- Department of Surgery, UVA School of Medicine, Charlottesville, Virginia 22908
| | - Timothy N J Bullock
- Beirne B. Carter Center for Immunology Research, UVA School of Medicine, Charlottesville, Virginia 22908
- Department of Pathology, UVA School of Medicine, Charlottesville, Virginia 22908
| | - Sepideh Dolatshahi
- Department of Biomedical Engineering, University of Virginia (UVA) School of Medicine, Charlottesville, Virginia 22908
- Beirne B. Carter Center for Immunology Research, UVA School of Medicine, Charlottesville, Virginia 22908
| | - Michael G Brown
- Beirne B. Carter Center for Immunology Research, UVA School of Medicine, Charlottesville, Virginia 22908
- Department of Medicine, Nephrology, UVA School of Medicine, Charlottesville, Virginia 22908
- Center for Immunity, Inflammation and Regenerative Medicine, UVA School of Medicine, Charlottesville, Virginia 22908
- Department of Microbiology, Immunology and Cancer Biology, UVA School of Medicine, Charlottesville, Virginia 22908
| |
Collapse
|
11
|
Szentkereszty M, Ladányi A, Gálffy G, Tóvári J, Losonczy G. Density of tumor-infiltrating NK and Treg cells is associated with 5 years progression-free and overall survival in resected lung adenocarcinoma. Lung Cancer 2024; 192:107824. [PMID: 38761665 DOI: 10.1016/j.lungcan.2024.107824] [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: 01/08/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/20/2024]
Abstract
Surgical resection of pulmonary adenocarcinoma is considered to be curative but progression-free survival (PFS) has remained highly variable. Antitumor immune response may be important, however, the prognostic significance of tumor-infiltrating natural killer (NK) and regulatory T (Treg) lymphocytes is uncertain. Resected pulmonary adenocarcinoma tissues (n = 115) were studied by immunohistochemical detection of NKp46 and FoxP3 positivity to identify NK and Treg cells, respectively. Association of cell densities with clinicopathological features and progression-free survival (PFS) as well as overall survival (OS) were analyzed with a follow-up time of 60 months. Both types of immune cells were accumulated predominantly in tumor stroma. NK cell density showed association with female gender, non-smoking and KRAS wild-type status. According to Kaplan-Meier analysis, PFS and OS proved to be longer in patients with high NK or Treg cell densities (p = 0.0293 and p = 0.0375 for PFS, p = 0.0310 and p = 0.0448 for OS, respectively). Evaluating the prognostic effect of the combination of NK and Treg cell density values revealed that PFS and OS were significantly longer in NKhigh/Treghigh cases compared to the other groups combined (p = 0.0223 and p = 0.0325, respectively). Multivariate Cox regression analysis indicated that high NK cell density was independent predictor of longer PFS while high NK and high Treg cell densities both proved significant predictors of longer OS. The NKhigh/Treghigh combination also proved to be an independent prognostic factor for both PFS and OS. In conclusion, NK and Treg cells can be components of the innate and adaptive immune response at action against progression of pulmonary adenocarcinoma.
Collapse
Affiliation(s)
- Márton Szentkereszty
- Department of Pulmonology, Semmelweis University Clinical Center, Budapest, Hungary; Tumor Pathology Center, National Institute of Oncology, Budapest, Hungary
| | - Andrea Ladányi
- Tumor Pathology Center, National Institute of Oncology, Budapest, Hungary; National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary
| | - Gabriella Gálffy
- Department of Pulmonology, Semmelweis University Clinical Center, Budapest, Hungary; Pulmonology Hospital of Törökbálint, Törökbálint, Hungary
| | - József Tóvári
- National Tumor Biology Laboratory, National Institute of Oncology, Budapest, Hungary; Department of Experimental Pharmacology, National Institute of Oncology, Budapest, Hungary
| | - György Losonczy
- Department of Pulmonology, Semmelweis University Clinical Center, Budapest, Hungary.
| |
Collapse
|
12
|
Tan Y, Ding L, Li G. MCM4 acts as a biomarker for LUAD prognosis. J Cell Mol Med 2023; 27:3354-3362. [PMID: 37817427 PMCID: PMC10623528 DOI: 10.1111/jcmm.17819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 04/16/2023] [Accepted: 06/14/2023] [Indexed: 10/12/2023] Open
Abstract
MCM4 forms the pre-replication complex (MCM2-7) with five other minichromosome maintenance (MCM) proteins. This complex binds to replication origins at G1 stage in cell cycle process, playing a critical role in DNA replication initiation. Recently, MCM4 is reported to have a complex interaction with multiple cancer progression, including gastric, ovarian and cervical cancer. Here, this study mainly focused on the expression of MCM4 and its values in lung adenocarcinoma (LUAD). MCM4 was highly expressed in LUAD tumours and cells, and had an important effect on the overall survival. Overexpression of MCM4 promoted the proliferation, and suppressed the apoptosis in LUAD cells. However, MCM4 silence led to the opposite results. In vivo, knockdown of MCM4 inhibited tumour volume and weight in xenograft mouse model. As a member of DNA helicase, knockdown of MCM4 caused cell cycle arrest at G1 stage through inducing the expression of P21, a CDK inhibitor. These findings indicate that MCM4 may be a possible new therapeutic target for LUAD in the future.
Collapse
Affiliation(s)
- Yue Tan
- Branch of Minhang, Department of Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
| | - Lei Ding
- Department of Ultrasonic DiagnosisSecond Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Guiyuan Li
- Department of Oncology, School of Medicine, Tongji HospitalTongji UniversityShanghaiChina
| |
Collapse
|
13
|
Ma S, Caligiuri MA, Yu J. Harnessing Natural Killer Cells for Lung Cancer Therapy. Cancer Res 2023; 83:3327-3339. [PMID: 37531223 DOI: 10.1158/0008-5472.can-23-1097] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/13/2023] [Accepted: 07/31/2023] [Indexed: 08/04/2023]
Abstract
Lung cancer is the leading cause of cancer-related death worldwide. Although natural killer (NK) cells are garnering interest as a potential anticancer therapy because they selectively recognize and eliminate cancer cells, their use in treating solid tumors, including lung cancer, has been limited due to impediments to their efficacy, such as their limited ability to reach tumor tissues, the reduced antitumor activity of tumor-infiltrating NK cells, and the suppressive tumor microenvironment (TME). This comprehensive review provides an in-depth analysis of the cross-talk between the lung cancer TME and NK cells. We highlight the various mechanisms used by the TME to modulate NK-cell phenotypes and limit infiltration, explore the role of the TME in limiting the antitumor activity of NK cells, and discuss the current challenges and obstacles that hinder the success of NK-cell-based immunotherapy for lung cancer. Potential opportunities and promising strategies to address these challenges have been implemented or are being developed to optimize NK-cell-based immunotherapy for lung cancer. Through critical evaluation of existing literature and emerging trends, this review provides a comprehensive outlook on the future of NK-cell-based immunotherapy for treating lung cancer.
Collapse
Affiliation(s)
- Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Los Angeles, California
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Los Angeles, California
- Comprehensive Cancer Center, City of Hope, Los Angeles, California
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
- Hematologic Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Los Angeles, California
- Comprehensive Cancer Center, City of Hope, Los Angeles, California
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope, Los Angeles, California
| |
Collapse
|
14
|
Moreo E, Jarit-Cabanillas A, Robles-Vera I, Uranga S, Guerrero C, Gómez AB, Mata-Martínez P, Minute L, Araujo-Voces M, Felgueres MJ, Esteso G, Uranga-Murillo I, Arias M, Pardo J, Martín C, Valés-Gómez M, Del Fresno C, Sancho D, Aguiló N. Intravenous administration of BCG in mice promotes natural killer and T cell-mediated antitumor immunity in the lung. Nat Commun 2023; 14:6090. [PMID: 37794033 PMCID: PMC10551006 DOI: 10.1038/s41467-023-41768-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 09/12/2023] [Indexed: 10/06/2023] Open
Abstract
Intravesical administration of Bacillus Calmette-Guérin (BCG) was one of the first FDA-approved immunotherapies and remains a standard treatment for bladder cancer. Previous studies have demonstrated that intravenous (IV) administration of BCG is well-tolerated and effective in preventing tuberculosis infection in animals. Here, we examine IV BCG in several preclinical lung tumor models. Our findings demonstrate that BCG inoculation reduced tumor growth and prolonged mouse survival in models of lung melanoma metastasis and orthotopic lung adenocarcinoma. Moreover, IV BCG treatment was well-tolerated with no apparent signs of acute toxicity. Mechanistically, IV BCG induced tumor-specific CD8+ T cell responses, which were dependent on type 1 conventional dendritic cells, as well as NK cell-mediated immunity. Lastly, we also show that IV BCG has an additive effect on anti-PD-L1 checkpoint inhibitor treatment in mouse lung tumors that are otherwise resistant to anti-PD-L1 as monotherapy. Overall, our study demonstrates the potential of systemic IV BCG administration in the treatment of lung tumors, highlighting its ability to enhance immune responses and augment immune checkpoint blockade efficacy.
Collapse
Affiliation(s)
- Eduardo Moreo
- Grupo de Genética de Micobacterias, Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Facultad de Medicina, Universidad de Zaragoza, IIS-Aragon, Zaragoza, Spain
- CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Iñaki Robles-Vera
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Santiago Uranga
- Grupo de Genética de Micobacterias, Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Facultad de Medicina, Universidad de Zaragoza, IIS-Aragon, Zaragoza, Spain
- CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Claudia Guerrero
- Grupo de Genética de Micobacterias, Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Facultad de Medicina, Universidad de Zaragoza, IIS-Aragon, Zaragoza, Spain
- CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Belén Gómez
- Grupo de Genética de Micobacterias, Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Facultad de Medicina, Universidad de Zaragoza, IIS-Aragon, Zaragoza, Spain
- CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | | | - Luna Minute
- Hospital la Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Miguel Araujo-Voces
- Grupo de Genética de Micobacterias, Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Facultad de Medicina, Universidad de Zaragoza, IIS-Aragon, Zaragoza, Spain
- CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología (IUOPA), Universidad deOviedo, Oviedo, Spain
| | - María José Felgueres
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Gloria Esteso
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Iratxe Uranga-Murillo
- Grupo de Inmunoterapia, Inmunidad y Cáncer, Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Facultad de Medicina, Universidad de Zaragoza, IIS-Aragon, Zaragoza, Spain
- CIBER Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Maykel Arias
- Grupo de Inmunoterapia, Inmunidad y Cáncer, Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Facultad de Medicina, Universidad de Zaragoza, IIS-Aragon, Zaragoza, Spain
- CIBER Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Julián Pardo
- Grupo de Inmunoterapia, Inmunidad y Cáncer, Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Facultad de Medicina, Universidad de Zaragoza, IIS-Aragon, Zaragoza, Spain
- CIBER Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Martín
- Grupo de Genética de Micobacterias, Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Facultad de Medicina, Universidad de Zaragoza, IIS-Aragon, Zaragoza, Spain
- CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Mar Valés-Gómez
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Carlos Del Fresno
- Hospital la Paz Institute for Health Research (IdiPAZ), Madrid, Spain
| | - David Sancho
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Nacho Aguiló
- Grupo de Genética de Micobacterias, Departamento de Microbiología, Pediatría, Radiología y Salud Pública, Facultad de Medicina, Universidad de Zaragoza, IIS-Aragon, Zaragoza, Spain.
- CIBER Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
15
|
Liu Z, Ding M, Qiu P, Pan K, Guo Q. Natural killer cell-related prognostic risk model predicts prognosis and treatment outcomes in triple-negative breast cancer. Front Immunol 2023; 14:1200282. [PMID: 37520534 PMCID: PMC10373504 DOI: 10.3389/fimmu.2023.1200282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
Background Natural killer (NK) cells are crucial to the emergence, identification, and prognosis of cancers. The roles of NK cell-related genes in the tumor immune microenvironment (TIME) and immunotherapy treatment are unclear. Triple-negative breast cancer (TNBC) is a highly aggressive malignant tumor. Hence, this study was conducted to develop a reliable risk model related to NK cells and provide a novel system for predicting the prognosis of TNBC. Methods NK cell-related genes were collected from previous studies. Based on TCGA and GEO database, univariate and LASSO cox regression analysis were used to establish the NK cell-related gene signature. The patients with TNBC were separated to high-risk and low-risk groups. After that, survival analysis was conducted and the responses to immunotherapies were evaluated on the basis of the signature. Moreover, the drug sensitivity of some traditional chemotherapeutic drugs was assessed by using the "oncoPredict" R package. In addition, the expression levels of the genes involved in the signature were validated by using qRT-PCR in TNBC cell lines. Results The patients with TNBC were divided into high- and low-risk groups according to the median risk score of the 5-NK cell-related gene signature. The low-risk group was associated with a better clinical outcome. Besides, the differentially expressed genes between the different risk groups were enriched in the biological activities associated with immunity. The tumor immune cells were found to be highly infiltrated in the low-risk groups. In accordance with the TIDE score and immune checkpoint-related gene expression analysis, TNBC patients in the low-risk groups were suggested to have better responses to immunotherapies. Eventually, some classical anti-tumor drugs were shown to be less effective in high-risk groups than in low-risk groups. Conclusion The 5-NK cell-related gene signature exhibit outstanding predictive performance and provide fresh viewpoints for evaluating the success of immunotherapy. It will provide new insights to achieve precision and integrated treatment for TNBC in the future.
Collapse
Affiliation(s)
- Zundong Liu
- Stem Cell Laboratory, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Mingji Ding
- Department of Breast and Thyroid Surgery, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Pengjun Qiu
- Department of Breast and Thyroid Surgery, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Kelun Pan
- Department of Breast and Thyroid Surgery, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qiaonan Guo
- Department of Breast and Thyroid Surgery, Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
16
|
Morimoto T, Nakazawa T, Maeoka R, Nakagawa I, Tsujimura T, Matsuda R. Natural Killer Cell-Based Immunotherapy against Glioblastoma. Int J Mol Sci 2023; 24:ijms24032111. [PMID: 36768432 PMCID: PMC9916747 DOI: 10.3390/ijms24032111] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive and malignant primary brain tumor in adults. Despite multimodality treatment involving surgical resection, radiation therapy, chemotherapy, and tumor-treating fields, the median overall survival (OS) after diagnosis is approximately 2 years and the 5-year OS is poor. Considering the poor prognosis, novel treatment strategies are needed, such as immunotherapies, which include chimeric antigen receptor T-cell therapy, immune checkpoint inhibitors, vaccine therapy, and oncolytic virus therapy. However, these therapies have not achieved satisfactory outcomes. One reason for this is that these therapies are mainly based on activating T cells and controlling GBM progression. Natural killer (NK) cell-based immunotherapy involves the new feature of recognizing GBM via differing mechanisms from that of T cell-based immunotherapy. In this review, we focused on NK cell-based immunotherapy as a novel GBM treatment strategy.
Collapse
Affiliation(s)
- Takayuki Morimoto
- Department of Neurosurgery, Nara Medical University, Kashihara 634-8521, Japan
- Department of Neurosurgery, Nara City Hospital, Nara 630-8305, Japan
- Correspondence: (T.M.); (T.N.); Tel.: +81-744-22-3051 (T.M.); +81-745-84-9335 (T.N.)
| | - Tsutomu Nakazawa
- Department of Neurosurgery, Nara Medical University, Kashihara 634-8521, Japan
- Grandsoul Research Institute for Immunology, Inc., Uda 633-2221, Japan
- Clinic Grandsoul Nara, Uda 633-2221, Japan
- Correspondence: (T.M.); (T.N.); Tel.: +81-744-22-3051 (T.M.); +81-745-84-9335 (T.N.)
| | - Ryosuke Maeoka
- Department of Neurosurgery, Nara Medical University, Kashihara 634-8521, Japan
| | - Ichiro Nakagawa
- Department of Neurosurgery, Nara Medical University, Kashihara 634-8521, Japan
| | - Takahiro Tsujimura
- Grandsoul Research Institute for Immunology, Inc., Uda 633-2221, Japan
- Clinic Grandsoul Nara, Uda 633-2221, Japan
| | - Ryosuke Matsuda
- Department of Neurosurgery, Nara Medical University, Kashihara 634-8521, Japan
| |
Collapse
|
17
|
Vyas M, Requesens M, Nguyen TH, Peigney D, Azin M, Demehri S. Natural killer cells suppress cancer metastasis by eliminating circulating cancer cells. Front Immunol 2023; 13:1098445. [PMID: 36733396 PMCID: PMC9887278 DOI: 10.3389/fimmu.2022.1098445] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
Despite significant advances in cancer treatment, the metastatic spread of malignant cells to distant organs remains a major cause of cancer-related deaths. Natural killer (NK) cells play a crucial role in controlling tumor metastasis; however, the dynamics of NK cell-mediated clearance of metastatic tumors are not entirely understood. Herein, we demonstrate the cooperative role of NK and T cells in the surveillance of melanoma metastasis. We found that NK cells effectively limited the pulmonary seeding of B16 melanoma cells, while T cells played a primary role in restricting metastatic foci growth in the lungs. Although the metastatic foci in the lungs at the endpoint were largely devoid of NK cells, they played a prominent role in promoting T cell recruitment into the metastatic foci. Our data suggested that the most productive interaction between NK cells and metastatic cancer cells occurred when cancer cells were in circulation. Modifying the route of administration so that intravenously injected melanoma cells bypass the first liver passage resulted in significantly more melanoma metastasis to the lung. This finding indicated the liver as a prominent site where NK cells cleared melanoma cells to regulate their seeding in the lungs. Consistent with this notion, the liver and the lungs of the tumor-bearing mice showed dominance of NK and T cell activation, respectively. Thus, NK cells and T cells control pulmonary metastasis of melanoma cells by distinct mechanisms where NK cells play a critical function in shaping T cell-mediated in situ control of lung-seeded cancer cells. A precise understanding of the cooperative role of NK and T cells in controlling tumor metastasis will enable the development of the next generation of cancer immunotherapies.
Collapse
|
18
|
Ghasemi M, Abbasi L, Ghanbari Naeini L, Kokabian P, Nameh Goshay Fard N, Givtaj N. Dendritic cells and natural killer cells: The road to a successful oncolytic virotherapy. Front Immunol 2023; 13:950079. [PMID: 36703982 PMCID: PMC9871831 DOI: 10.3389/fimmu.2022.950079] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 09/02/2022] [Indexed: 01/11/2023] Open
Abstract
Every type of cancer tissue is theoretically more vulnerable to viral infection. This natural proclivity has been harnessed as a new anti-cancer therapy by employing oncolytic viruses (OVs) to selectively infect and destroy cancer cells while providing little or no harm with no toxicity to the host. Whereas the primary oncolytic capabilities of OVs initially sparked the greatest concern, the predominant focus of research is on the association between OVs and the host immune system. Numerous OVs are potent causal agents of class I MHC pathway-related chemicals, enabling early tumor/viral immune recognition and cytokine-mediated response. The modified OVs have been studied for their ability to bind to dendritic cells (DCs) by expressing growth factors, chemokines, cytokines, and defensins inside the viral genome. OVs, like reovirus, can directly infect DCs, causing them to release chemokines and cytokines that attract and excite natural killer (NK) cells. In addition, OVs can directly alter cancer cells' sensitivity to NK by altering the expression levels of NK cell activators and inhibitors on cancerous cells. Therefore, NK cells and DCs in modulating the therapeutic response should be considered when developing and improving future OV-based therapeutics, whether modified to express transgenes or used in combination with other drugs/immunotherapies. Concerning the close relationship between NK cells and DCs in the potential of OVs to kill tumor cells, we explore how DCs and NK cells in tumor microenvironment affect oncolytic virotherapy and summarize additional information about the interaction mentioned above in detail in this work.
Collapse
Affiliation(s)
- Matin Ghasemi
- Faculty of Medicine, Tonekabon Branch, Islamic Azad University, Tonekabon, Iran
| | - Laleh Abbasi
- Guilan University of Medical Sciences, Rasht, Iran
| | | | - Pajman Kokabian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Najmeh Nameh Goshay Fard
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Nozar Givtaj
- Rajaei Cardiovascular, Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran,*Correspondence: Nozar Givtaj,
| |
Collapse
|
19
|
Tumor immunology. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00003-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
20
|
Lindsay RS, Melssen MM, Stasiak K, Annis JL, Woods AN, Rodriguez AB, Brown MG, Engelhard VH. NK cells reduce anergic T cell development in early-stage tumors by promoting myeloid cell maturation. Front Oncol 2022; 12:1058894. [PMID: 36531040 PMCID: PMC9755581 DOI: 10.3389/fonc.2022.1058894] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/15/2022] [Indexed: 12/03/2022] Open
Abstract
Introduction Studies of NK cells in tumors have primarily focused on their direct actions towards tumor cells. We evaluated the impact of NK cells on expression of homing receptor ligands on tumor vasculature, intratumoral T cell number and function, and T cell activation in tumor draining lymph node. Methods Using an implantable mouse model of melanoma, T cell responses and homing receptor ligand expression on the vasculature were evaluated with and without NK cells present during the early stages of the tumor response by flow cytometry. Results NK cells in early-stage tumors are one source of IFNγ that augments homing receptor ligand expression. More significantly, NK cell depletion resulted in increased numbers of intratumoral T cells with an anergic phenotype. Anergic T cell development in tumor draining lymph node was associated with increased T-cell receptor signaling but decreased proliferation and effector cell activity, and an incomplete maturation phenotype of antigen presenting cells. These effects of NK depletion were similar to those of blocking CD40L stimulation. Discussion We conclude that an important function of NK cells is to drive proper APC maturation via CD40L during responses to early-stage tumors, reducing development of anergic T cells. The reduced development of anergic T cells resulting in improved tumor control and T cell responses when NK cells were present.
Collapse
Affiliation(s)
- Robin S. Lindsay
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Marit M. Melssen
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Katarzyna Stasiak
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Jessica L. Annis
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Amber N. Woods
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Anthony B. Rodriguez
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Michael G. Brown
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
- Division of Nephrology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Victor H. Engelhard
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
21
|
Flores BCT, Chawla S, Ma N, Sanada C, Kujur PK, Yeung R, Bellon MB, Hukari K, Fowler B, Lynch M, Chinen LTD, Ramalingam N, Sengupta D, Jeffrey SS. Microfluidic live tracking and transcriptomics of cancer-immune cell doublets link intercellular proximity and gene regulation. Commun Biol 2022; 5:1231. [DOI: 10.1038/s42003-022-04205-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 11/01/2022] [Indexed: 11/15/2022] Open
Abstract
AbstractCell–cell communication and physical interactions play a vital role in cancer initiation, homeostasis, progression, and immune response. Here, we report a system that combines live capture of different cell types, co-incubation, time-lapse imaging, and gene expression profiling of doublets using a microfluidic integrated fluidic circuit that enables measurement of physical distances between cells and the associated transcriptional profiles due to cell–cell interactions. We track the temporal variations in natural killer—triple-negative breast cancer cell distances and compare them with terminal cellular transcriptome profiles. The results show the time-bound activities of regulatory modules and allude to the existence of transcriptional memory. Our experimental and bioinformatic approaches serve as a proof of concept for interrogating live-cell interactions at doublet resolution. Together, our findings highlight the use of our approach across different cancers and cell types.
Collapse
|
22
|
Grottoli M, Carrega P, Zullo L, Dellepiane C, Rossi G, Parisi F, Barletta G, Zinoli L, Coco S, Alama A, Marconi S, Parodi M, Orecchia P, Bassi S, Vitale M, Mingari MC, Pfeffer U, Genova C, Pietra G. Immune Checkpoint Blockade: A Strategy to Unleash the Potential of Natural Killer Cells in the Anti-Cancer Therapy. Cancers (Basel) 2022; 14:cancers14205046. [PMID: 36291830 PMCID: PMC9599824 DOI: 10.3390/cancers14205046] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/09/2022] [Accepted: 10/12/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary Immune checkpoint blockade (ICB) with antibodies targeting CTLA-4 (Cytotoxic Lymphocyte Antigen 4) and/or programmed death-1 protein (PD-1)/programmed death ligand-1 (PD-L1) has significantly modified the therapeutic landscape of a broad range of human tumor types, including advanced non-small-cell lung cancer (NSCLC). Despite great advances of checkpoint immunotherapies, a minority of NSCLC patients (<20%) respond and/or experience long-term clinical benefits from these treatments. Limited response rates of T cell–based checkpoint immunotherapies suggest the presence of other checkpoints able to inhibit effective anti-tumor immune responses. Natural Killer (NK) cells represent a promising target for tumor immunotherapies, particularly against tumors that escape T-cell-mediated control. Like T cell function, NK cell function is also regulated by inhibitory immune-checkpoint molecules. In this review, we will provide an overview of the rationale, mechanisms of action, and clinical efficacy of these NK cell-based checkpoint therapy approaches. Finally, the future directions and current enhancements planned will be discussed. Abstract Immune checkpoint inhibitors (ICIs) immunotherapy has represented a breakthrough in cancer treatment. Clinical use of ICIs has shown an acceptable safety profile and promising antitumor activity. Nevertheless, some patients do not obtain clinical benefits after ICIs therapy. In order to improve and cure an increasing number of patients, the field has moved toward the discovery of new ICIs expressed by cells of innate immunity with an elevated inherent antitumor activity, such as natural killer cells. This review will focus on the recent findings concerning the role of classical and non-classical immune checkpoint molecules and receptors that regulate natural killer cell function, as potential targets, and their future clinical application.
Collapse
Affiliation(s)
- Melania Grottoli
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Paolo Carrega
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, 98122 Messina, Italy
| | - Lodovica Zullo
- UO Oncologia Medica 2 IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Chiara Dellepiane
- UO Oncologia Medica 2 IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Giovanni Rossi
- UO Oncologia Medica 2 IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Francesca Parisi
- UO Oncologia Medica 2 IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Giulia Barletta
- UO Oncologia Medica 2 IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Linda Zinoli
- DiMI, Department of Internal Medicine and Medical Specialties, University of Genova, 16132 Genova, Italy
| | - Simona Coco
- UOS Tumori Polmonari IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Angela Alama
- UOS Tumori Polmonari IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Silvia Marconi
- UOS Tumori Polmonari IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Monica Parodi
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Paola Orecchia
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Sara Bassi
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Massimo Vitale
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Maria Cristina Mingari
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- DiMES, Department of Experimental Medicine, University of Genova, 16132 Genova, Italy
| | - Ulrich Pfeffer
- Laboratory of Tumor Epigenetics IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Carlo Genova
- DiMI, Department of Internal Medicine and Medical Specialties, University of Genova, 16132 Genova, Italy
- UO Clinica di Oncologia Medica IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- Correspondence: (C.G.); (G.P.)
| | - Gabriella Pietra
- UO Immunologia IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
- DiMES, Department of Experimental Medicine, University of Genova, 16132 Genova, Italy
- Correspondence: (C.G.); (G.P.)
| |
Collapse
|
23
|
lncRNA MANCR Inhibits NK Cell Killing Effect on Lung Adenocarcinoma by Targeting miRNA-30d-5p. Cell Microbiol 2022. [DOI: 10.1155/2022/4928635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Background. NK cells are imperative in spontaneous antitumor response of various cancers. Currently, lncRNAs are considered important modulators of the tumor microenvironment. This study investigated the molecular mechanism by which mitotically associated long noncoding RNA (MANCR) controls killing effect of NK cells on lung adenocarcinoma (LUAD) in the tumor microenvironment. Methods. The interplay between MANCR and miRNA-30d-5p was analyzed by bioinformatics. Expression of MANCR mRNA and miRNA-30d-5p was examined using qRT-PCR. Dual-luciferase reporter and RIP assays were utilized to verify the targeted relationship between MANCR and miRNA-30d-5p. To investigate regulation of MANCR/miRNA-30d-5p axis in NK cell killing effect on LUAD cells, western blot tested the protein level of perforin and granzyme B. ELISA determined the level of IFN-γ. CytoTox 96 Non-Radioactive Cytotoxicity Assay kit was applied for cytotoxicity detection of NK cells. Perforin and granzyme B fluorescence intensity was measured via immunofluorescence, and cell apoptosis levels were also revealed via flow cytometry. Results. MANCR was found to be upregulated, while miRNA-30d-5p expression was downregulated in LUAD tissues. Overexpression of MANCR in LUAD cells significantly reduced NK cell IFN-γ secretion, expression of granzyme B and perforin, and NK cell killing effect. In addition, MANCR could target and downregulate miRNA-30d-5p expression, and miRNA-30d-5p overexpression reversed the inhibition of NK cell killing effect caused by MANCR overexpression. Conclusion. MANCR inhibited the killing effect of NK cells on LUAD via targeting and downregulating miRNA-30d-5p and provided new ideas for antitumor therapy based on tumor microenvironment.
Collapse
|
24
|
Comprehensive Analysis of Gene Signatures of m6ARNA Methylation Regulators in Lung Adenocarcinoma and Development of a Risk Scoring System. J Immunol Res 2022; 2022:7519838. [PMID: 36061307 PMCID: PMC9428682 DOI: 10.1155/2022/7519838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 07/15/2022] [Indexed: 12/02/2022] Open
Abstract
The recent application of targeted immunotherapy has greatly improved the clinical outcomes of patients with lung adenocarcinoma (LUAD), but drug resistance continues to emerge, and to evaluate and to improve patient prognosis are arduous. The diagnostic and prognostic value of N6-methyladenosine (M6A) in LUAD has attracted increasing attention. We systematically studied correlations among important M6A methylation regulators, tumor mutational burden (TMB), and immune infiltration in clinical and sequencing data from the LUAD cohort of the cancer genome map (TCGA). The molecular subtype clusters 1 and 2 were identified by the consensus clustering of 16 M6A regulatory factors. Clinical prognosis, M6A regulatory factor expression, TMB, pathway enrichment, and immune cell infiltration significantly differed between clusters 1 and 2. Compared with other clinical traits, a prognostic risk score system constructed using the M6A regulatory factors HNRNPA2B1 and HNRNPC can serve as an independent prognostic method for LUAD, with higher predictive sensitivity and specificity. Risk scores were significantly higher for cluster 2 than 1, which was consistent with the trend towards a better prognosis in cluster 1. Overall, our findings revealed an important role of M6A methylation regulators in LUAD, and our risk scoring system involving these regulators might help to screen groups at high risk for LUAD and provide important theoretical bioinformatic support for evaluating the prognosis of such patients.
Collapse
|
25
|
Zhao W, Liu L, Li X, Xu S. EphA10 drives tumor progression and immune evasion by regulating the MAPK/ERK cascade in lung adenocarcinoma. Int Immunopharmacol 2022; 110:109031. [PMID: 35839564 DOI: 10.1016/j.intimp.2022.109031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 07/01/2022] [Accepted: 07/02/2022] [Indexed: 12/24/2022]
Abstract
Backgrounds Lung adenocarcinoma is the most frequent histological type among patients with lung cancer. Ephrin receptor A10 (EphA10), a member of the receptor tyrosine kinase family, has been reported to participate in tumor progression, but its role in lung adenocarcinoma (LUAD) remains unknown. Methods Immunohistochemistry staining and real-time PCR were employed to determine the expression of EphA10 in clinical LUAD samples. EphA10 silencing or overexpression in LUAD cells was achieved by transduction of lentivirus. The effects of EphA10 on LUAD cells were evaluated by CCK-8, EdU staining, flow cytometry, Transwell, and Western blot. The in vivo tumor growth was assessed in the xenograft mice model. Results EphA10 was overexpressed in LUAD tissues. Higher EphA10 expression was observed in the tissues at the advanced tumor stage and was positively correlated with the EGFR. Mechanistically, silencing of EphA10 suppressed proliferation, migration, invasion, and epithelial-mesenchymal transition of LUAD cells. Additionally, EphA10 knockdown significantly reduced the PD-L1 expression in LUAD cells and enhanced NK cell-mediated anti-tumor effects. Furthermore, EphA10 activated the MAPK/ERK pathway, and U0126, an inhibitor of MEK, markedly reversed the promoting impacts of EphA10 overexpression on LUAD cells. Consistently, results from subcutaneous tumor xenografts in nude mice confirmed that EphA10 knockdown significantly inhibited tumor growth in vivo. Conclusions This work demonstrates that EphA10 drives tumor progression and immune evasion by regulating the MAPK/ERK cascade in LUAD, implying that EphA10 has the potential to be a therapeutic target in treating LUAD.
Collapse
Affiliation(s)
- Wenyue Zhao
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang 110001, Liaoning, People's Republic of China
| | - Lu Liu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang 110001, Liaoning, People's Republic of China
| | - Xuehao Li
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang 110001, Liaoning, People's Republic of China
| | - Shun Xu
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang 110001, Liaoning, People's Republic of China.
| |
Collapse
|
26
|
Natural killer cell awakening: unleash cancer-immunity cycle against glioblastoma. Cell Death Dis 2022; 13:588. [PMID: 35803912 PMCID: PMC9270460 DOI: 10.1038/s41419-022-05041-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/25/2022] [Accepted: 06/27/2022] [Indexed: 01/21/2023]
Abstract
Due to the negligence of the complex tumor immune microenvironment, traditional treatment for glioblastoma has reached its limitation and cannot achieve a satisfying outcome in the past decade. The emergence of immunotherapy based on the theory of cancer-immunity cycle has brought a new dawn to glioblastoma patients. However, the results of most phase II and phase III clinical trials are not optimistic due to the simple focus on T cells activation rather than other immune cells involved in anti-tumor immunity. NK cells play a critical role in both innate and adaptive immunity, having the ability to coordinate immune response in inflammation, autoimmune disease and cancer. They are expected to cooperate with T cells to maximize the anti-tumor immune effect and have great potential in treating glioblastoma. Here, we describe the traditional treatment methods and current immunotherapy strategies for glioblastoma. Then, we list a microenvironment map and discuss the reasons for glioblastoma inhibitory immunity from multiple perspectives. More importantly, we focus on the advantages of NK cells as potential immune regulatory cells and the ways to maximize their anti-tumor immune effect. Finally, our outlook on the directions and potential applications of NK cell-based therapy combining with the advance technologies is presented. This review depicts NK cell awakening as the precondition to unleash the cancer-immunity cycle against glioblastoma and elaborate this idea from biology to clinical treatment.
Collapse
|
27
|
Song P, Li W, Guo L, Ying J, Gao S, He J. Identification and Validation of a Novel Signature Based on NK Cell Marker Genes to Predict Prognosis and Immunotherapy Response in Lung Adenocarcinoma by Integrated Analysis of Single-Cell and Bulk RNA-Sequencing. Front Immunol 2022; 13:850745. [PMID: 35757748 PMCID: PMC9231585 DOI: 10.3389/fimmu.2022.850745] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/12/2022] [Indexed: 12/11/2022] Open
Abstract
Natural killer (NK) cells, the effectors of the innate immune system, have a remarkable influence on cancer prognosis and immunotherapy. In this study, a total of 1,816 samples from nine independent cohorts in public datasets were enrolled. We first conducted a comprehensive analysis of single-cell RNA-sequencing data of lung adenocarcinoma (LUAD) from the Gene Expression Omnibus (GEO) database and determined 189 NK cell marker genes. Subsequently, we developed a seven-gene prognostic signature based on NK cell marker genes in the TCGA LUAD cohort, which stratified patients into high-risk and low-risk groups. The predictive power of the signature was well verified in different clinical subgroups and GEO cohorts. With a multivariate analysis, the signature was identified as an independent prognostic factor. Low-risk patients had higher immune cell infiltration states, especially CD8+ T cells and follicular helper T cells. There existed a negative association between inflammatory activities and risk score, and the richness and diversity of the T-cell receptor (TCR) repertoire was higher in the low-risk groups. Importantly, analysis of an independent immunotherapy cohort (IMvigor210) revealed that low-risk patients had better immunotherapy responses and prognosis than high-risk patients. Collectively, our study developed a novel signature based on NK cell marker genes, which had a potent capability to predict the prognosis and immunotherapy response of LUAD patients.
Collapse
Affiliation(s)
- Peng 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
| | - Wenbin Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shugeng 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
| | - Jie He
- 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
|
28
|
Sugimura R, Chao Y. Deciphering Innate Immune Cell-Tumor Microenvironment Crosstalk at a Single-Cell Level. Front Cell Dev Biol 2022; 10:803947. [PMID: 35646915 PMCID: PMC9140036 DOI: 10.3389/fcell.2022.803947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 04/12/2022] [Indexed: 11/23/2022] Open
Abstract
The tumor microenvironment encompasses various innate immune cells which regulate tumor progression. Exploiting innate immune cells is a new frontier of cancer immunotherapy. However, the classical surface markers for cell-type classification cannot always well-conclude the phenotype, which will further hinge our understanding. The innate immune cells include dendritic cells, monocytes/macrophages, natural killer cells, and innate lymphoid cells. They play important roles in tumor growth and survival, in some cases promoting cancer, in other cases negating cancer. The precise characterization of innate immune cells at the single-cell level will boost the potential of cancer immunotherapy. With the development of single-cell RNA sequencing technology, the transcriptome of each cell in the tumor microenvironment can be dissected at a single-cell level, which paves a way for a better understanding of the cell type and its functions. Here, we summarize the subtypes and functions of innate immune cells in the tumor microenvironment based on recent literature on single-cell technology. We provide updates on recent achievements and prospects for how to exploit novel functions of tumor-associated innate immune cells and target them for cancer immunotherapy.
Collapse
|
29
|
Li P, Lai Y, Tian L, Zhou Q. The prognostic value of prognostic nutritional index in advanced cancer receiving PD-1/L1 inhibitors: A meta-analysis. Cancer Med 2022; 11:3048-3056. [PMID: 35297197 PMCID: PMC9385596 DOI: 10.1002/cam4.4668] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/14/2022] [Accepted: 01/28/2022] [Indexed: 02/05/2023] Open
Abstract
Purpose To study the prognostic value of the prognostic nutritional index (PNI) in advanced cancers receiving programmed death‐1/programmed death‐ligand 1 (PD‐1/L1) inhibitors. Methods Online electronic databases were comprehensively searched and available literature was retrieved. We extracted available data from included researches and pooled the hazard ratios (HRs) and 95% confidence intervals (CIs) to learn the prognostic value of PNI on overall survival (OS) or progression‐free survival (PFS); and meanwhile calculated the relative risk (RR) with 95% CI to study the relationship between PNI and treatment efficacy (objective response rate [ORR] or disease control rate [DCR]) in late staged cancer receiving PD‐1/L1 inhibitors. Results Nine studies were finally selected for this meta‐analysis. We obtained data regarding PNI on OS from all nine studies, and the pooled HR was 2.31 (95% CI 1.81–2.94, p = 0.000), showing a correlation between low PNI and worse OS. Eight studies reported a relationship between PNI and PFS, and combined results revealed shorter PFS in patients with lower PNI, with an HR of 1.75 (95% CI 1.40–2.18, p = 0.000). Four studies explored the association between PNI and ORR and two studies explored the influence of PNI on DCR. An association between PNI and ORR (RR = 0.47, p = 0.003) was observed, while no association between PNI and DCR (RR = 0.49, p = 0.103) was observed by pooling these studies. Conclusion In summary, this meta‐analysis indicated that a lower PNI was significantly correlated with decreased OS and PFS and played adverse roles in ORR in advanced cancer patients receiving PD‐1/L1 inhibitors. Therefore, PNI could be promising for predicting prognosis and treatment response in advanced malignancies treated with PD‐1/L1 inhibitors.
Collapse
Affiliation(s)
- Pengfei Li
- Department of Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yutian Lai
- Department of Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Long Tian
- Department of Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qinghua Zhou
- Department of Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
30
|
Zhang XZ, Chen MJ, Fan PM, Jiang W, Liang SX. BTG2 Serves as a Potential Prognostic Marker and Correlates with Immune Infiltration in Lung Adenocarcinoma. Int J Gen Med 2022; 15:2727-2745. [PMID: 35300128 PMCID: PMC8922043 DOI: 10.2147/ijgm.s340565] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/26/2022] [Indexed: 11/23/2022] Open
Abstract
Background B-cell translocation gene 2 (BTG2) has been revealed to be involved in the occurrence and development of multiple cancers. However, the role of BTG2 in lung adenocarcinoma (LUAD) is still ambiguous. Thus, this study aims to investigate the prognostic value of BTG2 and its correlation with immune infiltration in LUAD. Methods The expression of BTG2 in LUAD was analyzed using the TIMER and UALCAN databases. The correlations between BTG2 expression and clinicopathological factors were investigated using the UALCAN databases. The Kaplan–Meier plotter, GEPIA, and TCGA databases were employed to assess the prognostic value of BTG2. The STRING database and Cytoscape software were used to construct an interaction network and mine co-expression genes. The TISIDB database was examined for a correlation between BTG2 and driver genes in LUAD. Enrichment analysis of co-expressed genes and BTG2 was performed using the LinkedOmics database. Finally, the correlations between BTG2 and immune infiltrates were investigated using the TIMER, GEO, and TISIDB database. Results BTG2 was significantly downregulated in LUAD. The decreased expression of BTG2 in LUAD was significantly correlated with higher cancer stages and shorter duration of overall survival. The expressions of BTG2-related co-expression genes were associated with the prognosis in LUAD. The expression of BTG2 was closely associated with the mutations of TP53 and ROS1. Enrichment analysis revealed that BTG2 was significantly correlated with immune‐associated signaling pathways and function. In addition, the expression of BTG2 was found to be closely related to immune infiltration, multiple gene markers of immune cells, chemokines, and chemokine receptors. Conclusion Our findings have effectively demonstrated that BTG2 expression was downregulated in LUAD, indicating poor prognosis. Closely relating to immune cell infiltration, BTG2 may be a promising immune-related biomarker and molecular target for patients with LUAD.
Collapse
Affiliation(s)
- Xiao Zhen Zhang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Mao Jian Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, Guangdong, People’s Republic of China
- Department of Respiratory Oncology, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Ping Ming Fan
- Department of Breast-Thoracic Tumor Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou, 570102, Hainan, People’s Republic of China
| | - Wei Jiang
- Department of Respiratory Oncology, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
| | - Shi Xiong Liang
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, 530021, Guangxi Zhuang Autonomous Region, People’s Republic of China
- Correspondence: Shi Xiong Liang; Wei Jiang, Email ;
| |
Collapse
|
31
|
Ma Y, Zhang H. Genomics and Prognosis Analysis of N 6-Methyladenosine Regulators in Lung Adenocarcinoma. Front Genet 2021; 12:746666. [PMID: 34956315 PMCID: PMC8697852 DOI: 10.3389/fgene.2021.746666] [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: 07/24/2021] [Accepted: 10/11/2021] [Indexed: 12/09/2022] Open
Abstract
Objective: N6-methyladenosine (m6A) modification is involved in modulating various biological processes in human cancers. But the implication of m6A modification in lung adenocarcinoma (LUAD) is still unclear. Hence, this study conducted a comprehensive analysis of the expression and clinical implication of m6A regulators in LUAD. Methods: Consensus clustering analysis of 502 LUAD samples in the TCGA dataset was presented based on the expression profiles of 20 m6A regulators using ConsensusClusterPlus package. Overall survival (OS), activation of signaling pathways and tumor immunity (immune/stromal score, tumor purity, expression of HLA and immune checkpoints, and immune cell infiltration) were compared between m6A modification patterns. The m6A-related genes between patterns were identified and prognostic m6A-related genes were imported into LASSO-cox regression analysis. The m6A risk score was developed and its prognostic implication was evaluated and externally verified in the GSE30219 and GSE72094 dataset. Furthermore, a nomogram that contained independent prognostic indicators was established, followed by external verification. Results: Two m6A modification patterns were clustered across LUAD based on the expression similarity of the m6A regulators via consensus clustering analysis, with distinct OS, activation of signaling pathways and tumor immunity. Totally, 213 m6A-related genes that were identified by comparing two patterns were significantly related to LUAD prognosis. By LASSO method, we constructed the m6A risk score that was a reliable and independent prognostic factor for LUAD. Patients with low m6A risk score displayed a prominent survival advantage. After incorporating independent clinical features, we developed the prognostic nomogram that exhibited high predictive accuracy and the best clinical net benefit for OS. Conclusion: Collectively, our study may provide a clinically useful tool for precise prognostic management and optimization of immunotherapeutic strategies for LUAD patients.
Collapse
Affiliation(s)
- Yanpin Ma
- Department of Oncology, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Huping Zhang
- Department of Infectious Diseases, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
32
|
Chen C, Guo Q, Tang Y, Qu W, Zuo J, Ke X, Song Y. Screening and evaluation of the role of immune genes of brain metastasis in lung adenocarcinoma progression based on the TCGA and GEO databases. J Thorac Dis 2021; 13:5016-5034. [PMID: 34527340 PMCID: PMC8411151 DOI: 10.21037/jtd-21-935] [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: 05/18/2021] [Accepted: 07/06/2021] [Indexed: 12/19/2022]
Abstract
Background Brain metastasis was one of the factors leading to the poor long-term prognosis of patients with lung adenocarcinoma (LUAD). Methods The expression levels of immune genes in LUAD and LUAD brain metastases tissues were analyzed in GSE161116 dataset using the GEO2R, and the levels of differential immune genes in normal lung and LUAD tissues were verified. The biological functions and signaling mechanisms of the differential immune genes were explored via Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analysis. Cox regression analysis was used to screen the prognostic factors of LUAD patients, and a risk model was constructed. The role of the model was checked in the development of LUAD via receiver operating characteristic analysis, gene set enrichment analysis, and Cox regression analysis. Results Differentially expressed genes (DEGs) in brain metastasis were involved in the adaptive immune response, B cell differentiation, leukocyte migration, NF-kB signaling pathway, among others. The expression levels of TNFRSF11A, MS4A2, IL11, CAMP, MS4A1, and F2RL1 were independent factors affecting the poor prognosis of LUAD patients via Cox regression analysis and Akaike information criterion. In the constructed risk model, the overall survival of LUAD patients in the high-risk group was poor. The risk model was significantly related to the gender, clinical stage, T stage, lymph node metastasis, and survival status of LUAD patients. In addition, the risk model score was an independent risk factor that affected the poor prognosis of LUAD patients. TNFRSF11A, CAMP, F2RL1, IL11, MS4A1, and MS4A2 of the risk factors had diagnostic significance in LUAD brain metastasis and LUAD. The risk model participated in cytokinetic process, cell cycle, citrate cycle TCA cycle, etc. The risk model score was correlated with the levels of B cells memory, mast cells resting, macrophages M0, mast cells activated, neutrophils, eosinophils, T cells gamma delta, and immune cell markers. Conclusions The risk model based on the LUAD brain metastasis immune factors TNFRSF11A, MS4A2, IL11, CAMP, MS4A1, and F2RL1 was related to the diagnosis, poor prognosis, and immune infiltrating cells of LUAD patients, and is expected to provide a reference for the development of treatment strategies for LUAD patients.
Collapse
Affiliation(s)
- Cheng Chen
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Qiang Guo
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China.,Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yang Tang
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Wendong Qu
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jiebin Zuo
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xixian Ke
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yongxiang Song
- Department of Thoracic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
33
|
Gao F, He S, Jin A. MiRNAs and lncRNAs in NK cell biology and NK/T-cell lymphoma. Genes Dis 2021; 8:590-602. [PMID: 34291131 PMCID: PMC8278539 DOI: 10.1016/j.gendis.2020.08.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/07/2020] [Accepted: 08/19/2020] [Indexed: 12/15/2022] Open
Abstract
The important role of lncRNAs and miRNAs in directing immune responses has become increasingly clear. Recent evidence conforms that miRNAs and lncRNAs are involved in NK cell biology and diseases through RNA-protein, RNA-RNA, or RNA-DNA interactions. In this view, we summarize the contribution of miRNAs and lncRNAs to NK cell lineage development, activation and function, highlight the biological significance of functional miRNAs or lncRNAs in NKTL and discuss the potential of these miRNAs and lncRNAs as innovative biomarkers/targets for NKTL early diagnosis, target treatment and prognostic evaluations.
Collapse
Affiliation(s)
- FengXia Gao
- Department of Immunology, Chongqing Medical University, Chongqing, 400010, PR China
- Chongqing Key Laboratory of Tumor Immunology and Tumor Immunotherapy, Chongqing Medical University, No.1, Medical School Road, Yuzhong District, Chongqing, 400010, PR China
| | - SiRong He
- Department of Immunology, Chongqing Medical University, Chongqing, 400010, PR China
- Chongqing Key Laboratory of Tumor Immunology and Tumor Immunotherapy, Chongqing Medical University, No.1, Medical School Road, Yuzhong District, Chongqing, 400010, PR China
| | - AiShun Jin
- Department of Immunology, Chongqing Medical University, Chongqing, 400010, PR China
- Chongqing Key Laboratory of Tumor Immunology and Tumor Immunotherapy, Chongqing Medical University, No.1, Medical School Road, Yuzhong District, Chongqing, 400010, PR China
| |
Collapse
|
34
|
Inhibitors Targeting CDK9 Show High Efficacy against Osimertinib and AMG510 Resistant Lung Adenocarcinoma Cells. Cancers (Basel) 2021; 13:cancers13153906. [PMID: 34359807 PMCID: PMC8345430 DOI: 10.3390/cancers13153906] [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: 05/31/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Non-small cell lung cancer accounts for 80% of all lung cancer cases. While a subset of non-small cell lung cancer patients respond to immunotherapy, those who are treated with chemotherapy or targeted therapy develop resistance to the drugs. Thus, novel therapeutic strategies are needed to combat this disease. Here we show that inhibitors of the cyclin-dependent kinase 9 are highly effective in preventing the growth of a variety of lung cancer cell lines and lung cancer organoids with high potency. These inhibitors suppressed the expression of several genes like Sox2, Sox9, and Mcl1 that promote tumor growth, facilitating growth arrest. Since inhibitors of cyclin-dependent kinase 9 are undergoing clinical trials for hematological malignancies, our studies suggest that these inhibitors would be attractive candidates to combat non-small cell lung cancer. Abstract Non-small cell lung cancer has a 5-year survival rate of less than 12–15%, calling for the development of additional therapeutic strategies to combat this disease. Here we tested the efficacy of inhibiting cyclin-dependent kinase 9 (CDK9) on lung cancer cell lines with K-Ras and EGFR mutations and on lung cancer organoids. Three different CDK9 inhibitors reduced the viability and anchorage-independent growth of lung cancer cell lines at very low nanomolar to micromolar concentrations. CDK9 inhibition suppressed the expression of the anti-apoptotic protein, Mcl1, as well as the embryonic stem cell transcription factors, Sox2 and Sox9, which are pro-tumorigenic. In contrast, treatment with CDK9 inhibitors increased the levels of WT p53 and its downstream target p21 in K-Ras mutant cell lines. Furthermore, the CDK9 inhibitors could markedly reduce the viability of Osimertinib-resistant PC9 and AMG510-resistant H23 and H358 cells with comparable efficacy as the parental cells. CDK9 inhibitors could also significantly reduce the growth and viability of lung cancer organoids with high potency. Taken together, the data presented here strongly suggest that CDK9 inhibitors would be efficacious against K-Ras mutant and EGFR mutant NSCLCs, including those that develop resistance to targeted therapies.
Collapse
|
35
|
Ramachandran S, Verma AK, Dev K, Goyal Y, Bhatt D, Alsahli MA, Rahmani AH, Almatroudi A, Almatroodi SA, Alrumaihi F, Khan NA. Role of Cytokines and Chemokines in NSCLC Immune Navigation and Proliferation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5563746. [PMID: 34336101 PMCID: PMC8313354 DOI: 10.1155/2021/5563746] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/23/2021] [Accepted: 06/28/2021] [Indexed: 12/14/2022]
Abstract
With over a million deaths every year around the world, lung cancer is found to be the most recurrent cancer among all types. Nonsmall cell lung carcinoma (NSCLC) amounts to about 85% of the entire cases. The other 15% owes it to small cell lung carcinoma (SCLC). Despite decades of research, the prognosis for NSCLC patients is poorly understood with treatment options limited. First, this article emphasises on the part that tumour microenvironment (TME) and its constituents play in lung cancer progression. This review also highlights the inflammatory (pro- or anti-) roles of different cytokines (ILs, TGF-β, and TNF-α) and chemokine (CC, CXC, C, and CX3C) families in the lung TME, provoking tumour growth and subsequent metastasis. The write-up also pinpoints recent developments in the field of chemokine biology. Additionally, it covers the role of extracellular vesicles (EVs), as alternate carriers of cytokines and chemokines. This allows the cytokines/chemokines to modulate the EVs for their secretion, trafficking, and aid in cancer proliferation. In the end, this review also stresses on the role of these factors as prognostic biomarkers for lung immunotherapy, apart from focusing on inflammatory actions of these chemoattractants.
Collapse
Affiliation(s)
- Sowmya Ramachandran
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Main Campus, Penang, Malaysia
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Amit K Verma
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Kapil Dev
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Yamini Goyal
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Deepti Bhatt
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Mohammed A Alsahli
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Saleh A Almatroodi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraidah, Saudi Arabia
| | - Naushad Ahmad Khan
- Department of Biochemistry, Faculty of Medical Sciences, Alatoo International University, Bishkek, Kyrgyzstan
- Department of Trauma and Surgery, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
36
|
Yuan X, Rasul F, Nashan B, Sun C. Innate lymphoid cells and cancer: Role in tumor progression and inhibition. Eur J Immunol 2021; 51:2188-2205. [PMID: 34189723 PMCID: PMC8457100 DOI: 10.1002/eji.202049033] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/12/2021] [Accepted: 06/21/2021] [Indexed: 02/06/2023]
Abstract
Innate lymphoid cells (ILCs), a critical component of the immune system, have recently been nominated as emerging players associated with tumor progression and inhibition. ILCs are classified into five groups: natural killer (NK) cells, ILC1s, ILC2s, ILC3s, and lymphoid tissue inducer (LTis) cells. NK cells and ILC1s are mainly involved in antitumor activities due to their cytotoxic and cytokine production capabilities, respectively. The current understanding of the heterogeneous behavior of ILC2s and ILC3s in tumors is limited and incomplete. Mostly, their dual roles are modulated by their resident tissues, released cytokines, cancer types, and plasticity. Based on overlap RORγt and cytokine expression, the LTi cells were previously considered part of the ILC3s ontogeny, which are essential for the formation of the secondary lymphoid organs during embryogenesis. Indeed, these facts highlight the urgency in understanding the respective mechanisms that shape the phenotypes and responses of ILCs, either on the repressive or proliferative side in the tumor microenvironment (TME). This review aims to provide an updated view of ILCs biology with respect to tumorigenesis, including a description of ILC plasticity, their interaction with other immune cells and communication with components of the TME. Taken together, targeting ILCs for cancer immunotherapy could be a promising approach against tumors that needs to be further study.
Collapse
Affiliation(s)
- Xiaodong Yuan
- Transplant & Immunology Laboratory, Division of Life Sciences and Medicine, Department of Organ Transplantation Center, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Faiz Rasul
- Transplant & Immunology Laboratory, Division of Life Sciences and Medicine, Department of Organ Transplantation Center, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, P. R. China.,Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Björn Nashan
- Transplant & Immunology Laboratory, Division of Life Sciences and Medicine, Department of Organ Transplantation Center, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, P. R. China
| | - Cheng Sun
- Transplant & Immunology Laboratory, Division of Life Sciences and Medicine, Department of Organ Transplantation Center, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, P. R. China.,Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, P. R. China.,Institute of Immunology, University of Science and Technology of China, Hefei, Anhui, P. R. China
| |
Collapse
|
37
|
Hamilton G, Plangger A. The Impact of NK Cell-Based Therapeutics for the Treatment of Lung Cancer for Biologics: Targets and Therapy. Biologics 2021; 15:265-277. [PMID: 34262255 PMCID: PMC8273903 DOI: 10.2147/btt.s290305] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 06/15/2021] [Indexed: 12/20/2022]
Abstract
Lung cancer has a dismal prognosis and novel targeted therapies leave still room for major improvements and better outcomes. Immunotherapy targeting immune checkpoint (IC) proteins, either as single agents or in combination with chemotherapy, is active but responders constitute only approximately 10-15% of non-small cell lung cancer (NSCLC) patients. Other effector immune cells such as CAR-T cells or NK cells may help to overcome the limitations of the IC inhibitor therapies for lung cancer. NK cells can kill tumor cells without previous priming and are present in the circulatory system and lymphoid organs. Tissue-residing NK cells differ from peripheral effector cells and, in case of the lung, comprise CD56bright CD16-negative populations showing high cytokine release but low cytotoxicity in contrast to the circulating CD56dim CD16-positive NK cells exhibiting high cytotoxic efficacy. This local attenuation of NK cell killing potency seems due to a specific stage of NK differentiation, immunosuppressive factors as well as presence of myeloid-derived suppressor cells (MDSCs) and regulatory T cells (TREGs). Improved NK cell-based immunotherapies involve IL-2-stimulated effector cells, NK cells expanded with the help of cytokines, permanent NK cell lines, induced pluripotent stem cell-derived NK cells and NK cells armed with chimeric antigen receptors. Compared to CAR T cell therapy, NK cells administration is devoid of graft-versus-host disease (GvHD) and cytokine-release syndrome. Although NK cells are clearly active against lung cancer cells, the low-cytotoxicity differentiation state in lung tumors, the presence of immunosuppressive leucocyte populations, limited infiltration and adverse conditions of the microenvironment need to be overcome. This goal may be achieved in the future using large numbers of activated and armed NK cells as provided by novel methods in NK cell isolation, expansion and stimulation of cytotoxic activity, including combinations with monoclonal antibodies in antibody-dependent cytotoxicity (ADCC). This review discusses the basic characteristics of NK cells and the potential of NK cell preparations in cancer therapy.
Collapse
Affiliation(s)
- Gerhard Hamilton
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| | - Adelina Plangger
- Department of Vascular Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
38
|
Yu L, Cao S, Li J, Han B, Zhong H, Zhong R. Prognostic value and immune infiltration of a novel stromal/immune score-related P2RY12 in lung adenocarcinoma microenvironment. Int Immunopharmacol 2021; 98:107734. [PMID: 34175738 DOI: 10.1016/j.intimp.2021.107734] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 04/24/2021] [Accepted: 04/26/2021] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Increasing evidence highlights the clinical implications of P2RY12 that belongs to the family of G-protein coupled receptors in carcinogenesis. Here, this study was designed to explore the associations between P2RY12 and tumor immune microenvironment of lung adenocarcinoma (LUAD). METHODS Based on 352 LUAD samples from The Cancer Genome Atlas (TCGA), stromal and immune scores of each sample were estimated by ESTIMATE algorithm. Differential expression analysis was presented between stromal/immune high- and low-score groups. Protein-protein interaction (PPI) was then constructed by STRING database. Univariate and multivariate Cox regression analysis was utilized to screen prognosis-related factors. Co-expressed genes of P2RY12 were analyzed, followed by functional enrichment analysis. Furthermore, the correlation between P2RY12 and immune cell infiltrations was estimated using the TIMER database. P2RY12 expression was validated between 37 pairs of LUAD and normal tissues using RT-qPCR and immunohistochemistry. After overexpressing P2RY12, the proliferation and migration of A549 cells was detected by CCK-8 and scratch test. RESULTS 145 up- and 102 down-regulated stromal- and immune-related mRNAs were identified for LUAD. Based on 145 up-regulated mRNAs, a PPI network was conducted, consisting of 95 nodes and 210 relationship pairs. Ten hub genes were then identified. Among them, CCR8, CNR2, CXCR5, GPR18, GPR31 and P2RY12 were in association with overall survival of patients with LUAD. After adjusting other variables, P2RY12 expression was an independent prognostic factor for LUAD. 288 co-expressed genes of P2RY12 were determined and these co-expressed genes were primarily involved in immune-related biological processes or pathways. Moreover, P2RY12 was significantly correlated with M2 macrophage and dendritic cell infiltration. After validation, P2RY12 expression was significantly decreased in LUAD than normal tissues. Its overexpression distinctly suppressed proliferation and migration of A549 cells. CONCLUSION Our findings suggest that P2RY12 is an immune infiltration-related prognostic marker for LUAD.
Collapse
Affiliation(s)
- Lian Yu
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China.
| | - Shuhui Cao
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China.
| | - Jingwen Li
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China.
| | - Baohui Han
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China.
| | - Hua Zhong
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China.
| | - Runbo Zhong
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China.
| |
Collapse
|
39
|
Hess JB, Sutherland KD, Best SA. Exploring natural killer cell immunology as a therapeutic strategy in lung cancer. Transl Lung Cancer Res 2021; 10:2788-2805. [PMID: 34295678 PMCID: PMC8264324 DOI: 10.21037/tlcr-20-765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 10/19/2020] [Indexed: 11/06/2022]
Abstract
Cytotoxic immune cells are key in the control of tumor development and progression. Natural killer (NK) cells are the cytotoxic arm of the innate immune system with the capability to kill tumor cells and surveil tumor cell dissemination. As such, the interest in harnessing NK cells in tumor control is increasing in many solid tumor types, including lung cancer. Here, we review the pre-clinical models used to unveil the role of NK cells in immunosurveillance of solid tumors and highlight measures to enhance NK cell activity. Importantly, the development of NK immunotherapy is rapidly evolving. Enhancing the NK cell response can be achieved using two broad modalities: enhancing endogenous NK cell activity, or performing adoptive transfer of pre-activated NK cells to patients. Numerous clinical trials are evaluating the efficacy of NK cell immunotherapy in isolation or in combination with standard treatments, with encouraging initial results. Pre-clinical studies and early phase clinical trials suggest that patients with solid tumors, including lung cancer, have the potential to benefit from recent developments in NK cell immunotherapy.
Collapse
Affiliation(s)
- Jonas B Hess
- ACRF Cancer Biology and Stem Cell Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Kate D Sutherland
- ACRF Cancer Biology and Stem Cell Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Sarah A Best
- ACRF Cancer Biology and Stem Cell Division, The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria, Australia.,Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
40
|
Long S, Gu Y, An Y, Lin X, Chen X, Wang X, Liao C, Ouyang W, Wang N, He Z, Zhao X. Reovirus enhances cytotoxicity of natural killer cells against colorectal cancer via TLR3 pathway. J Transl Med 2021; 19:185. [PMID: 33933132 PMCID: PMC8088708 DOI: 10.1186/s12967-021-02853-y] [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: 01/31/2021] [Accepted: 04/21/2021] [Indexed: 02/08/2023] Open
Abstract
Background Cetuximab has been approved for use for first-line treatment of patients with wild-type KRAS metastatic colorectal cancer (CRC). However, treatment with cetuximab has shown limited efficacy as a CRC monotherapy. In addition, natural killer (NK) cell function is known to be severely attenuated in cancer patients. The goal of this study was to develop a new strategy to enhance antibody-dependent cell-mediated cytotoxicity (ADCC) mediated by NK cells, in combination with cetuximab against CRC cells. Methods Ex vivo expanded NK cells were stimulated with reovirus, and reovirus-activated NK cells mediated ADCC assay were performed on CRC cells in combination with cetuximab. The synergistic antitumor effects of reovirus-activated NK cells and cetuximab were tested on DLD-1 tumor-bearing mice. Finally, Toll-like receptor 3 (TLR3) knockdown in NK cells, along with chemical blockade of TLR3/dsRNA complex, and inhibition of the TLR3 downstream signaling pathway, were performed to explore the mechanisms by which reovirus enhances NK cell cytotoxicity. Results We first confirmed that exposure of NK cells to reovirus enhanced their cytotoxicity in a dose-dependent manner.We then investigated whether reovirus-activated NK cells exposed to cetuximab-bound CRC cells exhibited greater anti-tumor efficacy than either monotherapy. Co-culture of CRC cell lines with reovirus-activated NK cells indicated that NK cytotoxicity was significantly higher in combination with cetuximab, regardless of KRAS mutation status or EGFR expression level. We also found that reovirus activation of NK cells, in conjunction with cetuximab, resulted in significantly stronger anti-tumor efficacy.Finally, TLR3 knockdown, inhibition of TLR3/dsRNA complex or TBK1/IKKε demonstrated that activation of NK cells by reovirus was dependent on TLR3 and its downstream signaling pathway. Conclusions This study demonstrated that combination treatment of reovirus-activated NK cells with cetuximab synergistically enhances their anti-tumor cytotoxicity, suggesting a strong candidate strategy for clinical treatment of CRC. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02853-y.
Collapse
Affiliation(s)
- Shiqi Long
- Center for Stem Cell and Tissue Engineering Research/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, Guizhou, China.,Key Laboratory of Adult Stem Cell Transformation Research, Guiyang, 550004, China
| | - Yangzhuo Gu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yuanyuan An
- Center for Stem Cell and Tissue Engineering Research/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, Guizhou, China.,Key Laboratory of Adult Stem Cell Transformation Research, Guiyang, 550004, China
| | - Xiaojin Lin
- Center for Stem Cell and Tissue Engineering Research/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, Guizhou, China.,Key Laboratory of Adult Stem Cell Transformation Research, Guiyang, 550004, China
| | - Xiaoqing Chen
- Center for Stem Cell and Tissue Engineering Research/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, Guizhou, China.,Key Laboratory of Adult Stem Cell Transformation Research, Guiyang, 550004, China
| | - Xianyao Wang
- Center for Stem Cell and Tissue Engineering Research/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, Guizhou, China.,Key Laboratory of Adult Stem Cell Transformation Research, Guiyang, 550004, China
| | - Chunxiang Liao
- Center for Stem Cell and Tissue Engineering Research/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, Guizhou, China.,Key Laboratory of Adult Stem Cell Transformation Research, Guiyang, 550004, China
| | - Weiwei Ouyang
- Department of Oncology, The Affiliated Hospital of Guizhou Medical University and Guizhou Cancer Hospital, Guiyang, 550004, China
| | - Nianxue Wang
- Key Laboratory of Adult Stem Cell Transformation Research, Guiyang, 550004, China
| | - Zhixu He
- Center for Stem Cell and Tissue Engineering Research/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, Guizhou, China.,Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, Zunyi, 563000, Guizhou, China
| | - Xing Zhao
- Center for Stem Cell and Tissue Engineering Research/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, 550004, Guizhou, China. .,Key Laboratory of Adult Stem Cell Transformation Research, Guiyang, 550004, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
41
|
Horowitz NB, Mohammad I, Moreno-Nieves UY, Koliesnik I, Tran Q, Sunwoo JB. Humanized Mouse Models for the Advancement of Innate Lymphoid Cell-Based Cancer Immunotherapies. Front Immunol 2021; 12:648580. [PMID: 33968039 PMCID: PMC8100438 DOI: 10.3389/fimmu.2021.648580] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Innate lymphoid cells (ILCs) are a branch of the immune system that consists of diverse circulating and tissue-resident cells, which carry out functions including homeostasis and antitumor immunity. The development and behavior of human natural killer (NK) cells and other ILCs in the context of cancer is still incompletely understood. Since NK cells and Group 1 and 2 ILCs are known to be important for mediating antitumor immune responses, a clearer understanding of these processes is critical for improving cancer treatments and understanding tumor immunology as a whole. Unfortunately, there are some major differences in ILC differentiation and effector function pathways between humans and mice. To this end, mice bearing patient-derived xenografts or human cell line-derived tumors alongside human genes or human immune cells represent an excellent tool for studying these pathways in vivo. Recent advancements in humanized mice enable unparalleled insights into complex tumor-ILC interactions. In this review, we discuss ILC behavior in the context of cancer, the humanized mouse models that are most commonly employed in cancer research and their optimization for studying ILCs, current approaches to manipulating human ILCs for antitumor activity, and the relative utility of various mouse models for the development and assessment of these ILC-related immunotherapies.
Collapse
Affiliation(s)
- Nina B Horowitz
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States.,Department of Bioengineering, Stanford University School of Medicine and School of Engineering, Stanford, CA, United States
| | - Imran Mohammad
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Uriel Y Moreno-Nieves
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Ievgen Koliesnik
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Quan Tran
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - John B Sunwoo
- Department of Otolaryngology-Head and Neck Surgery, Stanford Cancer Institute and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
42
|
He A, Zhang R, Wang J, Huang Z, Liao W, Li Y, Wang C, Yang J, Feng Q, Wu L. TYK2 is a prognostic biomarker and associated with immune infiltration in the lung adenocarcinoma microenvironment. Asia Pac J Clin Oncol 2021; 18:e129-e140. [PMID: 33852776 DOI: 10.1111/ajco.13569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 01/18/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) remains a major disease with high morbidity and mortality. The Janus kinases (JAKs) play a significant part in cellular biological process, inflammation and immunity. The role of JAK family in LUAD is still ambiguous. METHODS Various bioinformatics web portals were applied to explore the prognostic value of JAK family and their correlation with immune infiltration in LUAD. RESULTS JAK1/2 was downregulated, whereas JAK3/TYK2 was upregulated in patients with LUAD compared with the healthy controls in subgroup analyses based on gender, age, smoking habits, cancer stage, TP53 mutation status, and nodal metastasis status. Drug sensitivity indicated that low expression of JAK3 and TYK2 were resistant to most of the small molecules or drugs. High TYK2 expression was associated with favorable overall survival and relapse free survival in LUAD. Moreover, univariate and multivariate analysis revealed that clinical stage, lymphatic node metastasis and TYK2 expression were the independent factors affecting the prognosis of LUAD patients. TYK2 expression in LUAD patients was positively associated with the abundance of immune cells (B cells, CD8+ T cells, CD4+ T cells, neutrophils, and dendritic cells) and immune biomarker sets. Moreover, TYK2 was mainly involved in RNA binding, transcriptional mis-regulation in cancer and cell cycles. We also identified several TYK2-associated miRNA or transcription factor targets in LUAD. CONCLUSION Our results indicated that TYK2 was a biomarker and associated with prognosis and immune infiltration in LUAD, laying a foundation for further study about the role of TYK2 in the carcinogenesis and progression of LUAD.
Collapse
Affiliation(s)
- Aoxiao He
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Rongguiyi Zhang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jiakun Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Zhihao Huang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Wenjun Liao
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yong Li
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Cong Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Jun Yang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Qian Feng
- Department of Emergency, Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Linquan Wu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| |
Collapse
|
43
|
Lione L, Salvatori E, Petrazzuolo A, Massacci A, Maggio R, Confroti A, Compagnone M, Aurisicchio L, Ciliberto G, Palombo F. Antitumor efficacy of a neoantigen cancer vaccine delivered by electroporation is influenced by microbiota composition. Oncoimmunology 2021; 10:1898832. [PMID: 33796408 PMCID: PMC7993125 DOI: 10.1080/2162402x.2021.1898832] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/05/2021] [Accepted: 02/24/2021] [Indexed: 01/22/2023] Open
Abstract
Cancer is a heterogeneous disease and its treatment remains unsatisfactory with inconstant therapeutic responses. This variability could be related, at least in part, to different and highly personalized gut microbiota compositions. Different studies have shown an impact of microbiota on antitumor therapy. It has been demonstrated that some gut bacteria influences the development and differentiation of immune cells, suggesting that different microbiota compositions could affect the efficacy of the antitumor vaccine. Emerging data suggest that recognition of neoantigens for the generation of neoantigen cancer vaccines (NCVs) could have a key role in the activity of clinical immunotherapies. However, it is still unknown whether there is a crosstalk between microbiota and NCV. This study aimed to understand the possible mechanisms of interaction between gut microbiota and NCV delivered by DNA-electroporation (DNA-EP). We found that decreased microbiota diversity induced by prolonged antibiotic (ATB) treatment is associated with higher intratumor specific immune responses and consequently to a better antitumor effect induced by NCV delivered by DNA-EP.
Collapse
Affiliation(s)
- Lucia Lione
- Cancer Immunology, Takis, Rome, Italy
- Università Magna Grecia, Catanzaro, Italy
| | | | - Adriana Petrazzuolo
- Cancer Immunology, Takis, Rome, Italy
- Equipe Labellisée Par La Ligue Contre Le Cancer, Université De Paris, Sorbonne Université, INSERM U1138, Centre De Recherche Des Cordeliers, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
- Biogem, Ariano Irpino, Italy
| | | | | | | | - Mirco Compagnone
- Université Paris Saclay, Faculty of Medicine Kremlin Bicêtre, France
| | - Luigi Aurisicchio
- Cancer Immunology, Takis, Rome, Italy
- Evvivax, Rome, Italy
- Biogem, Ariano Irpino, Italy
- Université Paris Saclay, Faculty of Medicine Kremlin Bicêtre, France
| | - Gennaro Ciliberto
- Cancer immunology, Neomatrix, Rome, Italy
- Scientific Directorate, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Fabio Palombo
- Cancer Immunology, Takis, Rome, Italy
- Université Paris Saclay, Faculty of Medicine Kremlin Bicêtre, France
| |
Collapse
|
44
|
Hou J, Zhong Q. A novel immunogenomic prognostic signature in lung squamous carcinoma. Medicine (Baltimore) 2021; 100:e24073. [PMID: 33466167 PMCID: PMC7808533 DOI: 10.1097/md.0000000000024073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 12/03/2020] [Indexed: 01/05/2023] Open
Abstract
Lung squamous carcinoma (LUSC) is a common subtype of lung cancer with limited available therapy and is thus associated with poor survival. Immune infiltrating cells and immune-related genes (IRGs) play a key role in the clinical outcomes of LUSC. In the present study, we aimed to develop a potential immunogenomic prognostic signature for patients with LUSC. The transcriptional profiles of 501 LUSC samples from The Cancer Genome Atlas (TCGA) and 2498 IRGs from the ImmPort database were used to develop the signature by Cox regression analysis. Ten differentially expressed and survival-associated IRGs were used to develop the risk signature, which could serve as an independent prognostic and predictive factor for patients with LUSC. Furthermore, this risk signature correlated with overall survival and clinical features, including age, in patients with LUSC. In addition, we identified 25 transcription factors that may regulate 15 survival-associated IRGs, using a regulatory network. Collectively, this immunogenomic signature could be a robust prognostic tool for patients with LUSC and holds great promise as individualized immunotherapy for LUSC.
Collapse
Affiliation(s)
- Jili Hou
- Department of Oncology, Zhuji People's Hospital of Zhejiang Province, The Zhuji Affiliated Hospital of Shaoxing University, Zhuji 311800
| | - Qiuying Zhong
- Department of Geriatrics, The Sixth Hospital of Wuhan, Affiliated Hospital of Jianghan University, Wuhan 430000, P.R.China
| |
Collapse
|
45
|
Fan T, Zhu M, Wang L, Liu Y, Tian H, Zheng Y, Tan F, Sun N, Li C, He J. Immune profile of the tumor microenvironment and the identification of a four-gene signature for lung adenocarcinoma. Aging (Albany NY) 2020; 13:2397-2417. [PMID: 33318300 PMCID: PMC7880407 DOI: 10.18632/aging.202269] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 10/22/2020] [Indexed: 12/11/2022]
Abstract
The composition and relative abundances of immune cells in the tumor microenvironment are key factors affecting the progression of lung adenocarcinomas (LUADs) and the efficacy of immunotherapy. Using the cancer gene expression dataset from The Cancer Genome Atlas (TCGA) program, we scored stromal and immune cells for tumor purity prediction by CIBERSORT and ESTMATE. Differential expression analysis was employed to identify 374 genes between the high-score group and the low-score group, which were utilized to conduct Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Protein-protein interaction (PPI) and Cox regression analysis were performed on the differentially expressed genes (DEGs) to identify four key tumor microenvironment (TME) -related genes (CCR2, CCR4, P2RY12, and P2RY13). The expression levels of the four DEGs differed significantly among LUAD patients of different ages, genders, and TNM stages. We found that the infiltration of resting memory CD4+ T cells, memory B cells, and M0 macrophages into the TME was co-regulated by these four DEGs. These four genes were closely related to the prognosis of LUAD and affected the infiltration of immune cells into the TME, which had predictive prognostic value in LUAD.
Collapse
Affiliation(s)
- Tao Fan
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,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 100021, China
| | - Mingchuang Zhu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,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 100021, China
| | - Liyu Wang
- 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 100021, China
| | - Yu Liu
- 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 100021, China
| | - He Tian
- 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 100021, China
| | - Yujia Zheng
- 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 100021, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Nan Sun
- 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 100021, China
| | - Chunxiang Li
- 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 100021, China
| | - Jie He
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan 430060, China.,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 100021, China
| |
Collapse
|
46
|
Tian WJ, Liu SS, Li BR. The Combined Detection of Immune Genes for Predicting the Prognosis of Patients With Non-Small Cell Lung Cancer. Technol Cancer Res Treat 2020; 19:1533033820977504. [PMID: 33256552 PMCID: PMC7711225 DOI: 10.1177/1533033820977504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Lung cancer is one of the leading causes of cancer-related death. In recent years, there has been an increasing interest in the fields of tumor and immunity. This study focused on the possible prognostic value of immune genes in non-small cell lung cancer patients. We used The Cancer Genome Atlas (TCGA) to download gene expression data and clinical information of lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC). The immune gene list was downloaded from the Immport database. We then constructed immune gene prognostic models on the basis of Cox regression analysis. We further evaluated the clinical significance of the models via survival analysis, receiver operating characteristic (ROC) curves, and independent prognostic factor analysis. Moreover, we analyzed the associations of prognostic models with both mutation burdens and neoantigens. Using the Gene Expression Omnibus (GEO) and Kaplan-Meier plotter databases, we evaluated the validity of the prognostic models. The prognostic model of LUAD included 13 immune genes, and the prognostic model of LUSC contained 10 immune genes. High-risk patients based on prognostic models had a lower 5-year survival rate than did low-risk patients. The ROC curve analysis demonstrated the prediction accuracy of the prognostic models, as the area under the curve (AUC) was 0.742, 0.707, and 0.711 for LUAD, and 0.668, 0.703, and 0.668 for LUSC, when the predicted survival times were 1, 3, and 5 years, respectively. The mutation burden analysis showed that mutation level was associated with the risk score in patients with LUAD. The analysis based on GEO and Kaplan-Meier plotter demonstrated the prognostic validity of the models. Therefore, immune gene-related models of LUAD and LUSC can predict prognosis. Further study of these genes may enable us to better distinguish between LUAD and LUSC and lead to improvement in immunotherapy for lung cancer.
Collapse
Affiliation(s)
- Wen-Juan Tian
- Department of Clinical Laboratory, Second Affiliated Hospital, 117799Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China.,School of Medicine, 117799Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Shan-Shan Liu
- Department of Clinical Laboratory, Second Affiliated Hospital, 117799Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China.,School of Medicine, 117799Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Bu-Rong Li
- Department of Clinical Laboratory, Second Affiliated Hospital, 117799Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| |
Collapse
|
47
|
Hu-Lieskovan S, Bhaumik S, Dhodapkar K, Grivel JCJB, Gupta S, Hanks BA, Janetzki S, Kleen TO, Koguchi Y, Lund AW, Maccalli C, Mahnke YD, Novosiadly RD, Selvan SR, Sims T, Zhao Y, Maecker HT. SITC cancer immunotherapy resource document: a compass in the land of biomarker discovery. J Immunother Cancer 2020; 8:e000705. [PMID: 33268350 PMCID: PMC7713206 DOI: 10.1136/jitc-2020-000705] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2020] [Indexed: 02/07/2023] Open
Abstract
Since the publication of the Society for Immunotherapy of Cancer's (SITC) original cancer immunotherapy biomarkers resource document, there have been remarkable breakthroughs in cancer immunotherapy, in particular the development and approval of immune checkpoint inhibitors, engineered cellular therapies, and tumor vaccines to unleash antitumor immune activity. The most notable feature of these breakthroughs is the achievement of durable clinical responses in some patients, enabling long-term survival. These durable responses have been noted in tumor types that were not previously considered immunotherapy-sensitive, suggesting that all patients with cancer may have the potential to benefit from immunotherapy. However, a persistent challenge in the field is the fact that only a minority of patients respond to immunotherapy, especially those therapies that rely on endogenous immune activation such as checkpoint inhibitors and vaccination due to the complex and heterogeneous immune escape mechanisms which can develop in each patient. Therefore, the development of robust biomarkers for each immunotherapy strategy, enabling rational patient selection and the design of precise combination therapies, is key for the continued success and improvement of immunotherapy. In this document, we summarize and update established biomarkers, guidelines, and regulatory considerations for clinical immune biomarker development, discuss well-known and novel technologies for biomarker discovery and validation, and provide tools and resources that can be used by the biomarker research community to facilitate the continued development of immuno-oncology and aid in the goal of durable responses in all patients.
Collapse
Affiliation(s)
- Siwen Hu-Lieskovan
- Huntsman Cancer Institute, Salt Lake City, UT, USA
- University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | - Kavita Dhodapkar
- Department of Pediatrics, Emory University, Atlanta, Georgia, USA
- Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, Georgia, USA
| | | | - Sumati Gupta
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | - Brent A Hanks
- Duke University Medical Center, Durham, North Carolina, USA
| | | | | | - Yoshinobu Koguchi
- Earle A Chiles Research Institute, Providence Cancer Institute, Portland, Oregon, USA
| | - Amanda W Lund
- Oregon Health and Science University, Portland, Oregon, USA
| | | | | | | | | | - Tasha Sims
- Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | | | | |
Collapse
|
48
|
Russick J, Joubert PE, Gillard-Bocquet M, Torset C, Meylan M, Petitprez F, Dragon-Durey MA, Marmier S, Varthaman A, Josseaume N, Germain C, Goc J, Dieu-Nosjean MC, Validire P, Fournel L, Zitvogel L, Bindea G, Lupo A, Damotte D, Alifano M, Cremer I. Natural killer cells in the human lung tumor microenvironment display immune inhibitory functions. J Immunother Cancer 2020; 8:jitc-2020-001054. [PMID: 33067317 PMCID: PMC7570244 DOI: 10.1136/jitc-2020-001054] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Natural killer (NK) cells play a crucial role in tumor immunosurveillance through their cytotoxic effector functions and their capacity to interact with other immune cells to build a coordinated antitumor immune response. Emerging data reveal NK cell dysfunction within the tumor microenvironment (TME) through checkpoint inhibitory molecules associated with a regulatory phenotype. OBJECTIVE We aimed at analyzing the gene expression profile of intratumoral NK cells compared with non-tumorous NK cells, and to characterize their inhibitory function in the TME. METHODS NK cells were sorted from human lung tumor tissue and compared with non- tumoral distant lungs. RESULTS In the current study, we identify a unique gene signature of NK cell dysfunction in human non-small cell lung carcinoma (NSCLC). First, transcriptomic analysis reveals significant changes related to migratory pattern with a downregulation of sphingosine-1-phosphate receptor 1 (S1PR1) and CX3C chemokine receptor 1 (CX3CR1) and overexpression of C-X-C chemokine receptor type 5 (CXCR5) and C-X-C chemokine receptor type 6 (CXCR6). Second, cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and killer cell lectin like receptor (KLRC1) inhibitory molecules were increased in intratumoral NK cells, and CTLA-4 blockade could partially restore MHC class II level on dendritic cell (DC) that was impaired during the DCs/NK cell cross talk. Finally, NK cell density impacts the positive prognostic value of CD8+ T cells in NSCLC. CONCLUSIONS These findings demonstrate novel molecular cues associated with NK cell inhibitory functions in NSCLC.
Collapse
Affiliation(s)
- Jules Russick
- Centre de Recherche des Cordeliers, Sorbonne Universite, Inserm, Universite de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France
| | - Pierre-Emmanuel Joubert
- Centre de Recherche des Cordeliers, Sorbonne Universite, Inserm, Universite de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France
| | - Mélanie Gillard-Bocquet
- Centre de Recherche des Cordeliers, Sorbonne Universite, Inserm, Universite de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France
| | - Carine Torset
- Centre de Recherche des Cordeliers, Sorbonne Universite, Inserm, Universite de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France
| | - Maxime Meylan
- Centre de Recherche des Cordeliers, Sorbonne Universite, Inserm, Universite de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France
| | - Florent Petitprez
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Marie-Agnes Dragon-Durey
- Centre de Recherche des Cordeliers, Sorbonne Universite, Inserm, Universite de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France.,Universite de Paris. Laboratoire d'immunologie, Hopital Europeen Georges Pompidou, APHP, Paris, France
| | - Solenne Marmier
- Centre de Recherche des Cordeliers, Sorbonne Universite, Inserm, Universite de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France
| | - Aditi Varthaman
- Centre de Recherche des Cordeliers, Sorbonne Universite, Inserm, Universite de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France
| | - Nathalie Josseaume
- Centre de Recherche des Cordeliers, Sorbonne Universite, Inserm, Universite de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France
| | - Claire Germain
- Centre de Recherche des Cordeliers, Sorbonne Universite, Inserm, Universite de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France
| | - Jérémy Goc
- Centre de Recherche des Cordeliers, Sorbonne Universite, Inserm, Universite de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France
| | - Marie-Caroline Dieu-Nosjean
- Sorbonne Universite, INSERM U1135, Centre d'Immunologie et des Maladies Infectieuses, Team Immune Microenvironment and Immunotherapy, F-75013, Paris, France
| | - Pierre Validire
- Department of Pathology, Institut Mutualiste Montsouris, Paris, France
| | - Ludovic Fournel
- Departments of Pathology and Thoracic Surgery, Hospital Cochin Assistance Publique Hopitaux de Paris, F-75014, Paris, France
| | - Laurence Zitvogel
- INSERM U1015, Gustave Roussy, 114 rue Edouard Vaillant, 94805, Villejuif Cedex, France.,Universite Paris Saclay, Le Kremlin-Bicêtre, France
| | - Gabriela Bindea
- Centre de Recherche des Cordeliers, Sorbonne Universite, Inserm, Universite de Paris, Team Laboratory of Integrative cancer immunology, F-75006, Paris, France
| | - Audrey Lupo
- Centre de Recherche des Cordeliers, Sorbonne Universite, Inserm, Universite de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France.,Departments of Pathology and Thoracic Surgery, Hospital Cochin Assistance Publique Hopitaux de Paris, F-75014, Paris, France
| | - Diane Damotte
- Centre de Recherche des Cordeliers, Sorbonne Universite, Inserm, Universite de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France.,Departments of Pathology and Thoracic Surgery, Hospital Cochin Assistance Publique Hopitaux de Paris, F-75014, Paris, France
| | - Marco Alifano
- Centre de Recherche des Cordeliers, Sorbonne Universite, Inserm, Universite de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France.,Departments of Pathology and Thoracic Surgery, Hospital Cochin Assistance Publique Hopitaux de Paris, F-75014, Paris, France
| | - Isabelle Cremer
- Centre de Recherche des Cordeliers, Sorbonne Universite, Inserm, Universite de Paris, Team Inflammation, complement and cancer, F-75006, Paris, France
| |
Collapse
|
49
|
Li G, Wang G, Guo Y, Li S, Zhang Y, Li J, Peng B. Development of a novel prognostic score combining clinicopathologic variables, gene expression, and mutation profiles for lung adenocarcinoma. World J Surg Oncol 2020; 18:249. [PMID: 32950055 PMCID: PMC7502202 DOI: 10.1186/s12957-020-02025-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/10/2020] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Integrating phenotypic and genotypic information to improve prognostic prediction is under active investigation for lung adenocarcinoma (LUAD). In this study, we developed a new prognostic model for event-free survival (EFS) and recurrence-free survival (RFS) based on the combination of clinicopathologic variables, gene expression, and mutation data. METHODS We enrolled a total of 408 patients from the Cancer Genome Atlas Lung Adenocarcinoma (TCGA-LUAD) project for the study. We pre-selected gene expression or mutation features and constructed 14 different input feature sets for predictive model development. We assessed model performance with multiple evaluation metrics including the distribution of C-index on testing dataset, risk score significance, and time-dependent AUC under competing risks scenario. We stratified patients into higher- and lower-risk subgroups by the final risk score and further investigated underlying immune phenotyping variations associated with the differential risk. RESULTS The model integrating all three types of data achieved the best prediction performance. The resultant risk score provided a higher-resolution risk stratification than other models within pathologically defined subgroups. The score could account for extra EFS-related variations that were not captured by clinicopathologic scores. Being validated for RFS prediction under a competing risks modeling framework, the score achieved a significantly higher time-dependent AUC as compared to that of the conventional clinicopathologic variables-based model (0.772 vs. 0.646, p value < 0.001). The higher-risk patients were characterized with transcriptional aberrations of multiple immune-related genes, and a significant depletion of mast cells and natural killer cells. CONCLUSIONS We developed a novel prognostic risk score with improved prediction accuracy, using clinicopathologic variables, gene expression and mutation profiles as input, for LUAD. Such score was a significant predictor of both EFS and RFS. TRIAL REGISTRATION This study was based on public open data from TCGA and hence the study objects were retrospectively registered.
Collapse
Affiliation(s)
- Guofeng Li
- Department of Thoracic Surgery, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Luohu District, Shenzhen, 518020, China
| | - Guangsuo Wang
- Department of Thoracic Surgery, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Luohu District, Shenzhen, 518020, China
| | - Yanhua Guo
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Yangpu District, Shanghai, 200433, China
| | - Shixuan Li
- Department of Thoracic Surgery, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Luohu District, Shenzhen, 518020, China
| | - Youlong Zhang
- Department of Biostatistics, HuaJia Biomedical Intelligence, Shenzhen Overseas Chinese High-Tech Venture Park, Nanshan District, Shenzhen, 518057, China
| | - Jialu Li
- Department of Biostatistics, HuaJia Biomedical Intelligence, Shenzhen Overseas Chinese High-Tech Venture Park, Nanshan District, Shenzhen, 518057, China.
| | - Bin Peng
- Department of Thoracic Surgery, Shenzhen People's Hospital, Second Clinical Medical College of Jinan University, Luohu District, Shenzhen, 518020, China.
| |
Collapse
|
50
|
Mazzaschi G, Minari R, Zecca A, Cavazzoni A, Ferri V, Mori C, Squadrilli A, Bordi P, Buti S, Bersanelli M, Leonetti A, Cosenza A, Ferri L, Rapacchi E, Missale G, Petronini PG, Quaini F, Tiseo M. Soluble PD-L1 and Circulating CD8+PD-1+ and NK Cells Enclose a Prognostic and Predictive Immune Effector Score in Immunotherapy Treated NSCLC patients. Lung Cancer 2020; 148:1-11. [PMID: 32768804 DOI: 10.1016/j.lungcan.2020.07.028] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/15/2020] [Accepted: 07/24/2020] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Upfront criteria to foresee immune checkpoint inhibitors (ICIs) efficacy are far from being identified. Thus, we integrated blood descriptors of pro-inflammatory/immunosuppressive or effective anti-tumor response to non-invasively define predictive immune profiles in ICI-treated advanced non-small cell lung cancer (NSCLC). METHODS Peripheral blood (PB) was prospectively collected at baseline from 109 consecutive NSCLC patients undergoing ICIs as first or more line treatment. Soluble PD-L1 (sPD-L1) (immunoassay), CD8+PD-1+ and NK (FACS) cells were assessed and interlaced to generate an Immune effector Score (IeffS). Lung Immune Prognostic Index (LIPI) was computed by LDH levels and derived Neutrophil-to-Lymphocyte Ratio (dNLR). All these parameters were correlated with survival outcome and treatment response. RESULTS High sPD-L1 and low CD8+PD-1+ and NK number had negative impact on PFS (P < 0.001), OS (P < 0.01) and ICI-response (P < 0.05). Thus, sPD-L1high, CD8+PD-1+low and NKlow were considered as risk factors encompassing IeffS, whose prognostic power outperformed that of individual features and slightly exceeded that of LIPI. Accordingly, the absence of these risk factors portrayed a favorable IeffS characterizing patients with significantly (P < 0.001) prolonged PFS (median NR vs 2.3 months) and OS (median NR vs 4.1) and greater benefit from ICIs (P < 0.01). We then combined each risk parameter composing IeffS and LIPI (LDHhigh, dNLRhigh), thus defining three distinct prognostic classes. A remarkable impact of IeffS-LIPI integration was documented on survival outcome (PFS, HR = 4.61; 95%CI = 2.32-9.18; P < 0.001; OS, HR=4.03; 95%CI=1.91-8.67; P < 0.001) and ICI-response (AUC=0.90, 95%CI=0.81-0.97, P < 0.001). CONCLUSION Composite risk models based on blood parameters featuring the tumor-host interaction might provide accurate prognostic scores able to predict ICI benefit in NSCLC patients.
Collapse
Affiliation(s)
- G Mazzaschi
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy; Department of Medicine & Surgery, University of Parma, Italy
| | - R Minari
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy.
| | - A Zecca
- Infectious Diseases and Hepatology Unit, Laboratory of Viral Immunopathology, University Hospital of Parma, Italy
| | - A Cavazzoni
- Department of Medicine & Surgery, University of Parma, Italy
| | - V Ferri
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - C Mori
- Infectious Diseases and Hepatology Unit, Laboratory of Viral Immunopathology, University Hospital of Parma, Italy
| | - A Squadrilli
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - P Bordi
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - S Buti
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - M Bersanelli
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - A Leonetti
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - A Cosenza
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - L Ferri
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - E Rapacchi
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - G Missale
- Department of Medicine & Surgery, University of Parma, Italy; Infectious Diseases and Hepatology Unit, Laboratory of Viral Immunopathology, University Hospital of Parma, Italy
| | - P G Petronini
- Department of Medicine & Surgery, University of Parma, Italy
| | - F Quaini
- Department of Medicine & Surgery, University of Parma, Italy
| | - M Tiseo
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy; Department of Medicine & Surgery, University of Parma, Italy
| |
Collapse
|