51
|
Skeltved N, Nordmaj MA, Berendtsen NT, Dagil R, Stormer EMR, Al-Nakouzi N, Jiang K, Aicher A, Heeschen C, Gustavsson T, Choudhary S, Gögenur I, Christensen JP, Theander TG, Daugaard M, Salanti A, Nielsen MA. Bispecific T cell-engager targeting oncofetal chondroitin sulfate induces complete tumor regression and protective immune memory in mice. J Exp Clin Cancer Res 2023; 42:106. [PMID: 37118819 PMCID: PMC10142489 DOI: 10.1186/s13046-023-02655-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/28/2023] [Indexed: 04/30/2023] Open
Abstract
BACKGROUND The malaria protein VAR2CSA binds oncofetal chondroitin sulfate (ofCS), a unique chondroitin sulfate, expressed on almost all mammalian cancer cells. Previously, we produced a bispecific construct targeting ofCS and human T cells based on VAR2CSA and anti-CD3 (V-aCD3Hu). V-aCD3Hu showed efficacy against xenografted tumors in immunocompromised mice injected with human immune cells at the tumor site. However, the complex effects potentially exerted by the immune system as a result of the treatment cannot occur in mice without an immune system. Here we investigate the efficacy of V-aCD3Mu as a monotherapy and combined with immune checkpoint inhibitors in mice with a fully functional immune system. METHODS We produced a bispecific construct consisting of a recombinant version of VAR2CSA coupled to an anti-murine CD3 single-chain variable fragment. Flow cytometry and ELISA were used to check cell binding capabilities and the therapeutic effect was evaluated in vitro in a killing assay. The in vivo efficacy of V-aCD3Mu was then investigated in mice with a functional immune system and established or primary syngeneic tumors in the immunologically "cold" 4T1 mammary carcinoma, B16-F10 malignant melanoma, the pancreatic KPC mouse model, and in the immunologically "hot" CT26 colon carcinoma model. RESULTS V-aCD3Mu had efficacy as a monotherapy, and the combined treatment of V-aCD3Mu and an immune checkpoint inhibitor showed enhanced effects resulting in the complete elimination of solid tumors in the 4T1, B16-F10, and CT26 models. This anti-tumor effect was abscopal and accompanied by a systemic increase in memory and activated cytotoxic and helper T cells. The combined treatment also led to a higher percentage of memory T cells in the tumor without an increase in regulatory T cells. In addition, we observed partial protection against re-challenge in a melanoma model and full protection in a breast cancer model. CONCLUSIONS Our findings suggest that V-aCD3Mu combined with an immune checkpoint inhibitor renders immunologically "cold" tumors "hot" and results in tumor elimination. Taken together, these data provide proof of concept for the further clinical development of V-aCD3 as a broad cancer therapy in combination with an immune checkpoint inhibitor.
Collapse
Affiliation(s)
- Nanna Skeltved
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mie A Nordmaj
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark
| | - Nicolai T Berendtsen
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark
| | - Robert Dagil
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark
| | - Emilie M R Stormer
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark
| | - Nader Al-Nakouzi
- Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
| | - Ke Jiang
- Center for Single-Cell Omics and Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Alexandra Aicher
- Precision Immunotherapy, Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Christopher Heeschen
- Center for Single-Cell Omics and Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Pancreatic Cancer Heterogeneity, Candiolo Cancer Institute - FPO - IRCCS, Candiolo (Torino), Italy
| | - Tobias Gustavsson
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark
- Var2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Swati Choudhary
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark
- Var2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Ismail Gögenur
- Department of Clinical Medicine, University of Copenhagen and Center for Surgical Science, Zealand University Hospital, Copenhagen, Denmark
| | - Jan P Christensen
- Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thor G Theander
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mads Daugaard
- Vancouver Prostate Centre, University of British Columbia, Vancouver, Canada
- Var2 Pharmaceuticals ApS, Copenhagen, Denmark
| | - Ali Salanti
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark.
| | - Morten A Nielsen
- Centre for Medical Parasitology, Department of Infectious Diseases, University of Copenhagen and, Copenhagen University Hospital, Copenhagen, Denmark.
| |
Collapse
|
52
|
Zhang J, Cui X, Huang Y, Xu X, Feng C, Li J. Anticancer Effect of STING Agonist-Encapsulated Liposomes on Breast Cancer. Molecules 2023; 28:molecules28093740. [PMID: 37175150 PMCID: PMC10179927 DOI: 10.3390/molecules28093740] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Breast cancer is one of the most common cancers worldwide, posing a serious threat to human health. Recently, innate immunity has become a widely discussed topic in antitumor research. The STING pathway is an important component of innate immunity, and several STING agonists have been developed and applied in antitumor research. Dimeric amidobenzimidazole (diABZI) is one STING agonist and is a nucleotide analog with low serological stability and cell membrane permeability. In this study, we prepared diABZI-encapsulated liposomes (dLNPs) using the ammonium sulfate gradient method. The average particle size of the dLNPs was 99.76 ± 0.230 nm, and the encapsulation efficiency was 58.29 ± 0.53%. Additionally, in vivo and in vitro assays showed that the dLNPs had a sustained-release effect and that the circulation time in vivo was longer than 48 h. The expression of IFN-β and IFN-γ was elevated in mice treated with dLNPs. Moreover, we found that dLNPs can recruit CD8+ T cells to tumor tissue and exert antitumor effects. The dLNPs-treated group showed the most significant efficacy: the average tumor volume was 231.46 mm3, which decreased by 78.16% and 54.47% compared to the PBS group and diABZI group. Meanwhile, the hemolysis rate of the dLNPs was 2%, showing high biocompatibility. In conclusion, dLNPs can effectively suppress tumor growth and possess great potential in breast cancer therapy.
Collapse
Affiliation(s)
- Jibing Zhang
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Xiao Cui
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Yujiao Huang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Xiangdong Xu
- Liaocheng Inspection and Testing Center, Liaocheng 252000, China
| | - Changshun Feng
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| | - Jun Li
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng 252059, China
| |
Collapse
|
53
|
Chaib M, Tanveer UA, Makowski L. Myeloid cells in the era of cancer immunotherapy: Top 3 unanswered questions. Pharmacol Ther 2023; 244:108370. [PMID: 36871784 PMCID: PMC10798582 DOI: 10.1016/j.pharmthera.2023.108370] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 02/09/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Myeloid cells are increasingly being recognized as central players orchestrating or suppressing antitumor immune responses. With the advent of high-resolution analytical methods such as single-cell technologies, we now appreciate the heterogeneity and complexity of the myeloid compartment in the context of cancer. Because of their highly plastic nature, targeting myeloid cells has shown promising results either as a monotherapy or in combination with immunotherapy in preclinical models and cancer patients. However, the complexity of myeloid cell cellular crosstalk and molecular networks contributes to our poor understanding of the different myeloid cell subsets in tumorigenesis, which makes targeting myeloid cells challenging. Here, we summarize varied myeloid cell subsets and their contribution to tumor progression with a main focus on mononuclear phagocytes. The top three unanswered questions challenging the field of myeloid cells and cancer in the era of cancer immunotherapy are addressed. Through these questions, we discuss how myeloid cell origin and identity influence their function and disease outcomes. Different therapeutic strategies used to target myeloid cells in cancer are also addressed. Finally, the durability of myeloid cell targeting is interrogated by examining the complexity of resultant compensatory cellular and molecular mechanisms.
Collapse
Affiliation(s)
- Mehdi Chaib
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Ubaid A Tanveer
- Division of Hematology Oncology, Department of Medicine, College of Medicine, USA; Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Liza Makowski
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Division of Hematology Oncology, Department of Medicine, College of Medicine, USA; Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
54
|
Yang Y, Wang X, Lu J, Dong Z, Hu R, Chen W, Hu S, Lu G, Huang B, Dong S, Wang L, Wang C. Construction of a Prognostic Model for Predicting Colorectal Cancer Prognosis and Response to Immunotherapy Based on Cuproptosis-Associated lncRNAs. JOURNAL OF ONCOLOGY 2023; 2023:2733232. [PMID: 36968641 PMCID: PMC10033210 DOI: 10.1155/2023/2733232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 03/17/2023]
Abstract
Colorectal cancer (CRC) is a common and highly lethal gastrointestinal malignancy. Immunotherapy has shown positive efficacy in the treatment of CRC; however, only a minority of patients benefit from immunotherapy. The aim of this study is to construct a cuproptosis-related lncRNA (CRLs) risk score model to predict the prognosis and immune infiltration of CRC patients. Firstly, we synthetically analyzed 19 cuproptosis-related genes (CRGs) from CRC samples derived from the TCGA and obtained 33 CRLs that were significantly associated with prognosis. Next, we defined three cuproptosis modification patterns via consensus clustering analysis (C1, C2, and C3). Further analysis showed that there were significant differences in the abundance of B cells, NK cells, fibroblasts, monocytes, CD8+ cells, bone marrow dendritic cells, and cytotoxic lymphocytes in different clusters. In addition, the LASSO regression screened out 6 individual CRLs (AC009315.1, PLS3-AS1, ZEB1-AS1, AC007608.3, AC010789.2, and AC010207.1) closely related to the prognosis of CRC. We found that the low-risk group had better survival prognoses in patients. Furthermore, the high-risk group had lower immune scores and exhibited lower CD8+ T cell infiltration. Moreover, the low-risk group had lower immune exclusion, immune dysfunction and TIDE scores than the high-risk group. Interestingly, the lncRNAs in our risk model were positively associated with most immune checkpoints. CD274 (PD-L1), CTLA4, and HAVCR2 (TIM3) were positively correlated with risk scores. Moreover, MSI-H patients had lower risk scores than MSI-L patients, and IPS scores were significantly higher in the low CRLs score group. In conclusion, we constructed a novel risk score model with6 lncRNAs related to cuproptosis, which may be a potential biomarker for evaluating the prognosis and immune treatment for CRC.
Collapse
Affiliation(s)
- Yi Yang
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Jinan University Institute of Obesity and Metabolic Disorders, Guangzhou, Guangdong, China
| | - Xiaoli Wang
- West General Department, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jin Lu
- Key Laboratory of Computational Medicine and Intelligent Health of Anhui Higher Education Institutes, Bengbu Medical College, Bengbu, Anhui, China
| | - Zhiyong Dong
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Ruixiang Hu
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Wenhui Chen
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Songhao Hu
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Guanhua Lu
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Biao Huang
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Shiliang Dong
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Lu Wang
- Institute of Precision Cancer Medicine and Pathology, Jinan University Medical College, Guangzhou, Guangdong, China
| | - Cunchuan Wang
- Department of Bariatric Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
- Jinan University Institute of Obesity and Metabolic Disorders, Guangzhou, Guangdong, China
| |
Collapse
|
55
|
Chang J, Wu H, Wu J, Liu M, Zhang W, Hu Y, Zhang X, Xu J, Li L, Yu P, Zhu J. Constructing a novel mitochondrial-related gene signature for evaluating the tumor immune microenvironment and predicting survival in stomach adenocarcinoma. J Transl Med 2023; 21:191. [PMID: 36915111 PMCID: PMC10012538 DOI: 10.1186/s12967-023-04033-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/03/2023] [Indexed: 03/16/2023] Open
Abstract
BACKGROUND The incidence and mortality of gastric cancer ranks fifth and fourth worldwide among all malignancies, respectively. Accumulating evidences have revealed the close relationship between mitochondrial dysfunction and the initiation and progression of stomach cancer. However, rare prognostic models for mitochondrial-related gene risk have been built up in stomach cancer. METHODS In current study, the expression and prognostic value of mitochondrial-related genes in stomach adenocarcinoma (STAD) patients were systematically analyzed to establish a mitochondrial-related risk model based on available TCGA and GEO databases. The tumor microenvironment (TME), immune cell infiltration, tumor mutation burden, and drug sensitivity of gastric adenocarcinoma patients were also investigated using R language, GraphPad Prism 8 and online databases. RESULTS We established a mitochondrial-related risk prognostic model including NOX4, ALDH3A2, FKBP10 and MAOA and validated its predictive power. This risk model indicated that the immune cell infiltration in high-risk group was significantly different from that in the low-risk group. Besides, the risk score was closely related to TME signature genes and immune checkpoint molecules, suggesting that the immunosuppressive tumor microenvironment might lead to poor prognosis in high-risk groups. Moreover, TIDE analysis demonstrated that combined analysis of risk score and immune score, or stromal score, or microsatellite status could more effectively predict the benefit of immunotherapy in STAD patients with different stratifications. Finally, rapamycin, PD-0325901 and dasatinib were found to be more effective for patients in the high-risk group, whereas AZD7762, CEP-701 and methotrexate were predicted to be more effective for patients in the low-risk group. CONCLUSIONS Our results suggest that the mitochondrial-related risk model could be a reliable prognostic biomarker for personalized treatment of STAD patients.
Collapse
Affiliation(s)
- Jingjia Chang
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Hao Wu
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Jin Wu
- Department of Pathology, Laboratory of Translational Medicine Research, Deyang People's Hospital, Deyang, China.,Key Laboratory of Tumor Molecular Research of Deyang, Deyang, China.,Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Ming Liu
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Wentao Zhang
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Yanfen Hu
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Xintong Zhang
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Jing Xu
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Li Li
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China
| | - Pengfei Yu
- Department of Gastrointestinal Surgery, Xijing Hospital, Air Force Military Medical University, Xi'an, 710032, Shaanxi, China.
| | - Jianjun Zhu
- Department of Cell Biology and Medical Genetics, School of Basic Medical Science, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
56
|
Aria H, Rezaei M. Immunogenic cell death inducer peptides: A new approach for cancer therapy, current status and future perspectives. Biomed Pharmacother 2023; 161:114503. [PMID: 36921539 DOI: 10.1016/j.biopha.2023.114503] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/23/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Immunogenic Cell Death (ICD) is a type of cell death that kills tumor cells by stimulating the adaptive immune response against other tumor cells. ICD depends on the endoplasmic reticulum (ER) stress and the secretion of Damage-Associated Molecular Patterns (DAMP) by the dying tumor cell. DAMPs recruit innate immune cells such as Dendritic Cells (DC), triggering a cancer-specific immune response such as cytotoxic T lymphocytes (CTLs) to eliminate remaining cancer cells. ICD is accompanied by several hallmarks in dying cells, such as surface translocation of ER chaperones, calreticulin (CALR), and extracellular secretion of DAMPs such as high mobility group protein B1 (HMGB1) and adenosine triphosphate (ATP). Therapeutic peptides can kill bacteria and tumor cells thus affecting the immune system. They have high specificity and affinity for their targets, small size, appropriate cell membrane penetration, short half-life, and simple production processes. Peptides are interesting agents for immunomodulation since they may overcome the limitations of other therapeutics. Thus, the development of peptides affecting the TME and active antitumoral immunity has been actively pursued. On the other hand, several peptides have been recently identified to trigger ICD and anti-cancer responses. In the present review, we review previous studies on peptide-induced ICD, their mechanism, their targets, and markers. They include anti-microbial peptides (AMPs), cationic or mitochondrial targeting, checkpoint inhibitors, antiapoptotic inhibitors, and "don't eat me" inhibitor peptides. Also, peptides will be investigated potentially inducing ICD that is divided into ER stressors, ATPase inhibitors, and anti-microbial peptides.
Collapse
Affiliation(s)
- Hamid Aria
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marzieh Rezaei
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
57
|
Davola ME, Cormier O, Vito A, El-Sayes N, Collins S, Salem O, Revill S, Ask K, Wan Y, Mossman K. Oncolytic BHV-1 Is Sufficient to Induce Immunogenic Cell Death and Synergizes with Low-Dose Chemotherapy to Dampen Immunosuppressive T Regulatory Cells. Cancers (Basel) 2023; 15:cancers15041295. [PMID: 36831636 PMCID: PMC9953776 DOI: 10.3390/cancers15041295] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 02/22/2023] Open
Abstract
Immunogenic cell death (ICD) can switch immunologically "cold" tumors "hot", making them sensitive to immune checkpoint inhibitor (ICI) therapy. Many therapeutic platforms combine multiple modalities such as oncolytic viruses (OVs) and low-dose chemotherapy to induce ICD and improve prognostic outcomes. We previously detailed many unique properties of oncolytic bovine herpesvirus type 1 (oBHV) that suggest widespread clinical utility. Here, we show for the first time, the ability of oBHV monotherapy to induce bona fide ICD and tumor-specific activation of circulating CD8+ T cells in a syngeneic murine model of melanoma. The addition of low-dose mitomycin C (MMC) was necessary to fully synergize with ICI through early recruitment of CD8+ T cells and reduced infiltration of highly suppressive PD-1+ Tregs. Cytokine and gene expression analyses within treated tumors suggest that the addition of MMC to oBHV therapy shifts the immune response from predominantly anti-viral, as evidenced by a high level of interferon-stimulated genes, to one that stimulates myeloid cells, antigen presentation and adaptive processes. Collectively, these data provide mechanistic insights into how oBHV-mediated therapy modalities overcome immune suppressive tumor microenvironments to enable the efficacy of ICI therapy.
Collapse
Affiliation(s)
- Maria Eugenia Davola
- Department of Medicine, Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Olga Cormier
- Department of Medicine, Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Alyssa Vito
- Department of Medicine, Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Nader El-Sayes
- Department of Medicine, Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Susan Collins
- Department of Medicine, Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Omar Salem
- Department of Medicine, Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Spencer Revill
- Department of Medicine, Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4K1, Canada
- Firestone Institute for Respiratory Health, St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada
| | - Kjetil Ask
- Department of Medicine, Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4K1, Canada
- Firestone Institute for Respiratory Health, St. Joseph’s Healthcare Hamilton, Hamilton, ON L8N 4A6, Canada
| | - Yonghong Wan
- Department of Medicine, Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Karen Mossman
- Department of Medicine, Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON L8S 4K1, Canada
- Correspondence: ; Tel.: +1-905-525-9140 (ext. 23542)
| |
Collapse
|
58
|
Li Z, Yang H, Liu J, Li L, Wang X. TOMM34 serves as a candidate therapeutic target associated with immune cell infiltration in colon cancer. Front Oncol 2023; 13:947364. [PMID: 36845719 PMCID: PMC9948080 DOI: 10.3389/fonc.2023.947364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 01/27/2023] [Indexed: 02/11/2023] Open
Abstract
Background Colon cancer represents one of the most pervasive digestive malignancies worldwide. Translocase of the outer mitochondrial membrane 34 (TOMM34) is considered an oncogene and is implicated in tumor proliferation. However, the correlation between TOMM34 and immune cell infiltration in colon cancer has not been investigated. Materials and methods Based on multiple open online databases, we performed integrated bioinformatics analysis of TOMM34 to evaluate the prognostic value of TOMM34 and its correlation with immune cell infiltration. Results TOMM34 gene and protein expression levels were elevated in tumor tissues compared with normal tissues. Survival analysis revealed that upregulation of TOMM34 was significantly associated with poorer survival time in colon cancer. High TOMM34 expression was dramatically related to low levels of B cells, CD8+ T cells, neutrophils, dendritic cells, PD-1, PD-L1 and CTLA-4. Conclusions Our results confirmed that high expression of TOMM34 in tumor tissue correlates with immune cell infiltration and worse prognosis in colon cancer patients. TOMM34 may serve as a potential prognostic biomarker for colon cancer diagnosis and prognosis prediction.
Collapse
Affiliation(s)
- Zhigui Li
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Hongzhao Yang
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Jianbo Liu
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Li Li
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaodong Wang
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, China,*Correspondence: Xiaodong Wang,
| |
Collapse
|
59
|
Lan SY, Ding Y, Wang C, Fang J, Ren C, Liu JL, Kang H, Chang Y. High Level of Ubiquitin Conjugate Enzyme E2O Indicates Poor Prognosis of Patients with Hepatocellular Carcinoma. Curr Med Sci 2023; 43:93-103. [PMID: 36269535 DOI: 10.1007/s11596-022-2609-1] [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: 01/11/2022] [Accepted: 03/14/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Ubiquitin conjugate enzyme E2O (UBE2O) is a ubiquitin-conjugating enzyme that has been reported to be involved in tumorigenesis. This study investigated the role of UBE2O in hepatocellular carcinoma (HCC). METHODS The expression of UBE2O was detected using qRT-PCR, Western blotting, and immunohistochemical staining. Cell proliferation and Transwell assays were used to detect proliferation, migration, and invasion of HCC cells, respectively. Bioinformatic analysis was performed to analyze the relationship between UBE2O and the clinical features, prognosis, and immune cell infiltration of HCC. RESULTS UBE2O was significantly over-expressed in HCC tissues. High expression of UBE2O was associated with poor tumor grade and poor prognosis. Functional experiments showed that down-regulation of UBE2O inhibited HCC cell proliferation, migration, and invasion. Co-expression gene analysis and gene set enrichment analysis showed that UBE2O was associated with protein hydrolysis, cell cycle, and cancer-related pathways in HCC. The results of immune analysis revealed that the expression of UBE2O was positively correlated with the immune infiltration and expression of immune-related chemokines of HCC. CONCLUSIONS UBE2O is significantly correlated with the prognosis of HCC and may be a valuable prognostic biomarker for HCC.
Collapse
Affiliation(s)
- Si-Yu Lan
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Yang Ding
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Chun Wang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Jun Fang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Chao Ren
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Jia-Liang Liu
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Hui Kang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China
| | - Ying Chang
- Department of Gastroenterology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China. .,Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Diseases, Wuhan, 430071, China.
| |
Collapse
|
60
|
Yu X, Luo B, Lin J, Zhu Y. Alternative splicing event associated with immunological features in bladder cancer. Front Oncol 2023; 12:966088. [PMID: 36686818 PMCID: PMC9851621 DOI: 10.3389/fonc.2022.966088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 11/30/2022] [Indexed: 01/07/2023] Open
Abstract
Bladder cancer (BLCA) is the most prevalent urinary tumor with few treatments. Alternative splicing (AS) is closely related to tumor development and tumor immune microenvironment. However, the comprehensive analysis of AS and prognosis and immunological features in BLCA is still lacking. In this study, we downloaded RNA-Seq data and clinical information from The Cancer Genome Atlas (TCGA) database, and AS events were acquired from the TCGA Splice-seq. A total of eight prognostic AS events (C19orf57|47943|ES, ANK3|11845|AP, AK9|77203|AT, GRIK2|77096|AT, DYM|45472|ES, PTGER3|3415|AT, ACTG1|44120|RI, and TRMU|62711|AA) were identified by univariate analysis and least absolute shrinkage and selection operator (LASSO) regression analysis to construct a risk score model. The Kaplan-Meier analysis revealed that the high-risk group had a worse prognosis compared with the low-risk group. The area under the receiver operating characteristic (ROC) curves (AUCs) for this risk score model in 1, 3, and 5 years were 0.698, 0.742, and 0.772, respectively. One of the prognostic AS event-related genes, TRMU, was differentially expressed between tumor and normal tissues in BLCA. The single-sample gene set enrichment analysis (ssGSEA) and CIBERSORT algorithm showed that both the risk score model and TRMU were significantly associated with tumor immune microenvironment and immune status (immune cells, immune-related pathway, and immune checkpoint) in BLCA patients. The TIMER database confirmed the relationship between the expression of TRMU and immune cells and checkpoint genes. Furthermore, Cytoscape software 3.8.0 was used to construct the regulatory network between AS and splicing factors (SFs). Our study demonstrated that AS events were powerful biomarkers to predict the prognosis and immune status in BLCA, which may be potential therapeutic targets in BLCA.
Collapse
Affiliation(s)
- Xinbo Yu
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bixian Luo
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianwei Lin
- Department of General Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Zhu
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China,*Correspondence: Yu Zhu,
| |
Collapse
|
61
|
Wang N, Zhao L, Zhang D, Kong F. Research progress on the immunomodulatory mechanism of acupuncture in tumor immune microenvironment. Front Immunol 2023; 14:1092402. [PMID: 36865562 PMCID: PMC9971227 DOI: 10.3389/fimmu.2023.1092402] [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: 11/08/2022] [Accepted: 02/03/2023] [Indexed: 02/16/2023] Open
Abstract
With the constantly deeper understanding of individualized precision therapy, immunotherapy is increasingly developed and personalized. The tumor immune microenvironment (TIME) mainly consists of infiltrating immune cells, neuroendocrine cells, extracellular matrix, lymphatic vessel network, etc. It is the internal environment basis for the survival and development of tumor cells. As a characteristic treatment of traditional Chinese medicine, acupuncture has shown potentially beneficial impacts on TIME. The currently available information demonstrated that acupuncture could regulate the state of immunosuppression through a range of pathways. An effective way to understand the mechanisms of action of acupuncture was to analyze the response following treatment of the immune system. This research reviewed the mechanisms of acupuncture regulating tumor immunological status based on innate and adaptive immunity.
Collapse
Affiliation(s)
- Na Wang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Lu Zhao
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Dou Zhang
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Fanming Kong
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
62
|
Zhou S, Lu J, Liu S, Shao J, Liu Z, Li J, Xiao W. Role of the tumor microenvironment in malignant melanoma organoids during the development and metastasis of tumors. Front Cell Dev Biol 2023; 11:1166916. [PMID: 37152280 PMCID: PMC10154581 DOI: 10.3389/fcell.2023.1166916] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/11/2023] [Indexed: 05/09/2023] Open
Abstract
Malignant melanoma (MM) is the most metastatic and aggressive form of skin cancer, and carries a high risk of death. Immune-checkpoint inhibitor therapy and molecular-targeted therapy can prolong the survival of patients with advanced MM significantly. However, the low response rate and inevitable drug resistance prevent further improvements in efficacy, which is closely related to the tumor microenvironment (TME). The TME refers to the tumor stroma, including fibroblasts, keratinocytes, immune cells, soluble molecules, and extracellular matrix (ECM). The dynamic interaction between the TME and tumor cells is very important for the growth, local invasion, and metastatic spread of tumor cells. A patient-derived organoid (PDO) model involves isolation of tumor tissue from patients with MM and culturing it in vitro in a three-dimensional pattern. Compared with traditional cultivation methods, the PDO model preserves the heterogeneity of the tissue structure of MM and demonstrates the interaction between MM cells and the TME. It can reproduce the characteristics of proliferation, migration, and invasion of MM cells, and better simulate the structural function of MM in vivo. This review explores the role of each TME component in development of the PDO model. This review will provide a reference for research on the drug screening and targeted treatment using PDOs, particularly for the immunotherapy of MM.
Collapse
|
63
|
Pal K, Sheth RA. Engineering the Tumor Immune Microenvironment through Minimally Invasive Interventions. Cancers (Basel) 2022; 15:196. [PMID: 36612192 PMCID: PMC9818918 DOI: 10.3390/cancers15010196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
The tumor microenvironment (TME) is a unique landscape that poses several physical, biochemical, and immune barriers to anti-cancer therapies. The rapidly evolving field of immuno-engineering provides new opportunities to dismantle the tumor immune microenvironment by efficient tumor destruction. Systemic delivery of such treatments can often have limited local effects, leading to unwanted offsite effects such as systemic toxicity and tumor resistance. Interventional radiologists use contemporary image-guided techniques to locally deliver these therapies to modulate the immunosuppressive TME, further accelerating tumor death and invoking a better anti-tumor response. These involve local therapies such as intratumoral drug delivery, nanorobots, nanoparticles, and implantable microdevices. Physical therapies such as photodynamic therapy, electroporation, hyperthermia, hypothermia, ultrasound therapy, histotripsy, and radiotherapy are also available for local tumor destruction. While the interventional radiologist can only locally manipulate the TME, there are systemic offsite recruitments of the immune response. This is known as the abscopal effect, which leads to more significant anti-tumoral downstream effects. Local delivery of modern immunoengineering methods such as locoregional CAR-T therapy combined with immune checkpoint inhibitors efficaciously modulates the immunosuppressive TME. This review highlights the various advances and technologies available now to change the TME and revolutionize oncology from a minimally invasive viewpoint.
Collapse
Affiliation(s)
| | - Rahul A. Sheth
- Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
64
|
Hashemi M, Ghadyani F, Hasani S, Olyaee Y, Raei B, Khodadadi M, Ziyarani MF, Basti FA, Tavakolpournegari A, Matinahmadi A, Salimimoghadam S, Aref AR, Taheriazam A, Entezari M, Ertas YN. Nanoliposomes for doxorubicin delivery: Reversing drug resistance, stimuli-responsive carriers and clinical translation. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
65
|
Zhu S, Wang Y, Tang J, Cao M. Radiotherapy induced immunogenic cell death by remodeling tumor immune microenvironment. Front Immunol 2022; 13:1074477. [PMID: 36532071 PMCID: PMC9753984 DOI: 10.3389/fimmu.2022.1074477] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022] Open
Abstract
Emerging evidence indicates that the induction of radiotherapy(RT) on the immunogenic cell death (ICD) is not only dependent on its direct cytotoxic effect, changes in the tumor immune microenvironment also play an important role in it. Tumor immune microenvironment (TIME) refers to the immune microenvironment that tumor cells exist, including tumor cells, inflammatory cells, immune cells, various signaling molecules and extracellular matrix. TIME has a barrier effect on the anti-tumor function of immune cells, which can inhibit all stages of anti-tumor immune response. The remodeling of TIME caused by RT may affect the degree of immunogenicity, and make it change from immunosuppressive phenotype to immunostimulatory phenotype. It is of great significance to reveal the causes of immune escape of tumor cells, especially for the treatment of drug-resistant tumor. In this review, we focus on the effect of RT on the TIME, the mechanism of RT in reversing the TIME to suppress intrinsic immunity, and the sensitization effect of the remodeling of TIME caused by RT on the effectiveness of immunotherapy.
Collapse
|
66
|
Identification of immune subtypes and their prognosis and molecular implications in colorectal cancer. PLoS One 2022; 17:e0278114. [PMID: 36417424 PMCID: PMC9683557 DOI: 10.1371/journal.pone.0278114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 10/12/2022] [Indexed: 11/25/2022] Open
Abstract
Immune composition is commonly heterogeneous and varies among colorectal cancer (CRC) patients. A comprehensive immune classification may act as important characteristics to predict CRC prognosis. Thus, we aimed to identify novel immune specific subtypes to guide future therapies. Unsupervised clustering was used to classify CRC samples into different immune subtypes based on abundances of immune cell populations, during which TCGA and GSE17536 datasets were used as training and validation sets, respectively. The associations between the immune subtypes and patient prognosis were investigated. Further, we identified differentially expressed genes (DEGs) between immune high and low subtypes, followed by functional enrichment analyses of DEGs. The expression levels of 74 immunomodulators (IMs) across immune subtypes were analyzed. As a result, we clustered CRC samples into three distinct immune subtypes (immune high, moderate, and low). Patients with immune-high subtype showed the best prognosis, and patients with immune-low subtype had the worst survival in both TCGA and GSE17536 cohorts. A group of 2735 up-regulated DEGs were identified across immune high and low subtypes. The main DEGs were the members of complement components, chemokines, immunoglobulins, and immunosuppressive genes that are involved in immune modulation-related pathways (e.g., cytokine-cytokine receptor interaction) or GO terms (e.g., adaptive immune response and T cell activation). The expression levels of 63 IMs were significantly varied across immune subtypes. In conclusion, this study provides a conceptual framework and molecular characteristics of CRC immune subtypes, which may accurately predict prognosis and offer novel targets for personalized immunotherapy through modifying subtype-specific tumor immune microenvironment.
Collapse
|
67
|
Oladejo M, Nguyen HM, Silwal A, Reese B, Paulishak W, Markiewski MM, Wood LM. Listeria-based immunotherapy directed against CD105 exerts anti-angiogenic and anti-tumor efficacy in renal cell carcinoma. Front Immunol 2022; 13:1038807. [PMID: 36439126 PMCID: PMC9692019 DOI: 10.3389/fimmu.2022.1038807] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/19/2022] [Indexed: 07/29/2023] Open
Abstract
Targeting tumor-associated angiogenesis is currently at the forefront of renal cell carcinoma (RCC) therapy, with sunitinib and bevacizumab leading to increased survival in patients with metastatic RCC (mRCC). However, resistance often occurs shortly after initiation of therapy, suggesting that targeting the tumor-associated vascular endothelium may not be sufficient to eradicate RCC. This study reports the therapeutic efficacy of a Listeria (Lm)-based vaccine encoding an antigenic fragment of CD105 (Lm-LLO-CD105A) that targets both RCC tumor cells and the tumor-associated vasculature. Lm-LLO-CD105A treatment reduced primary tumor growth in both subcutaneous and orthotopic models of murine RCC. The vaccine conferred anti-tumor immunity and remodeled the tumor microenvironment (TME), resulting in increased infiltration of polyfunctional CD8+ and CD4+ T cells and reduced infiltration of immunosuppressive cell types within the TME. We further provide evidence that the therapeutic efficacy of Lm-LLO-CD105A is mediated by CD8+ T cells and is dependent on the robust antigenic expression of CD105 by RCC tumor cells. The result from this study demonstrates the safety and promising therapeutic efficacy of targeting RCC-associated CD105 expression with Lm-based immunotherapy.
Collapse
|
68
|
Zhang W, Li S, Li C, Li T, Huang Y. Remodeling tumor microenvironment with natural products to overcome drug resistance. Front Immunol 2022; 13:1051998. [PMID: 36439106 PMCID: PMC9685561 DOI: 10.3389/fimmu.2022.1051998] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 10/13/2022] [Indexed: 09/01/2023] Open
Abstract
With cancer incidence rates continuing to increase and occurrence of resistance in drug treatment, there is a pressing demand to find safer and more effective anticancer strategy for cancer patients. Natural products, have the advantage of low toxicity and multiple action targets, are always used in the treatment of cancer prevention in early stage and cancer supplement in late stage. Tumor microenvironment is necessary for cancer cells to survive and progression, and immune activation is a vital means for the tumor microenvironment to eliminate cancer cells. A number of studies have found that various natural products could target and regulate immune cells such as T cells, macrophages, mast cells as well as inflammatory cytokines in the tumor microenvironment. Natural products tuning the tumor microenvironment via various mechanisms to activate the immune response have immeasurable potential for cancer immunotherapy. In this review, it highlights the research findings related to natural products regulating immune responses against cancer, especially reveals the possibility of utilizing natural products to remodel the tumor microenvironment to overcome drug resistance.
Collapse
Affiliation(s)
- Wanlu Zhang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Shubo Li
- Liaoning Center for Animal Disease Control and Prevention, Liaoning Agricultural Development Service Center, Shenyang, China
| | - Chunting Li
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Tianye Li
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Yongye Huang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| |
Collapse
|
69
|
Murugan D, Murugesan V, Panchapakesan B, Rangasamy L. Nanoparticle Enhancement of Natural Killer (NK) Cell-Based Immunotherapy. Cancers (Basel) 2022; 14:cancers14215438. [PMID: 36358857 PMCID: PMC9653801 DOI: 10.3390/cancers14215438] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
Simple Summary Natural killer cells are a part of the native immune response to cancer. NK cell-based immunotherapies are an emerging strategy to kill tumor cells. This paper reviews the role of NK cells, their mechanism of action for killing tumor cells, and the receptors which could serve as potential targets for signaling. In this review, the role of nanoparticles in NK cell activation and increased cytotoxicity of NK cells against cancer are highlighted. Abstract Natural killer (NK) cells are one of the first lines of defense against infections and malignancies. NK cell-based immunotherapies are emerging as an alternative to T cell-based immunotherapies. Preclinical and clinical studies of NK cell-based immunotherapies have given promising results in the past few decades for hematologic malignancies. Despite these achievements, NK cell-based immunotherapies have limitations, such as limited performance/low therapeutic efficiency in solid tumors, the short lifespan of NK cells, limited specificity of adoptive transfer and genetic modification, NK cell rejection by the patient’s immune system, insignificant infiltration of NK cells into the tumor microenvironment (TME), and the expensive nature of the treatment. Nanotechnology could potentially assist with the activation, proliferation, near-real time imaging, and enhancement of NK cell cytotoxic activity by guiding their function, analyzing their performance in near-real time, and improving immunotherapeutic efficiency. This paper reviews the role of NK cells, their mechanism of action in killing tumor cells, and the receptors which could serve as potential targets for signaling. Specifically, we have reviewed five different areas of nanotechnology that could enhance immunotherapy efficiency: nanoparticle-assisted immunomodulation to enhance NK cell activity, nanoparticles enhancing homing of NK cells, nanoparticle delivery of RNAi to enhance NK cell activity, genetic modulation of NK cells based on nanoparticles, and nanoparticle activation of NKG2D, which is the master regulator of all NK cell responses.
Collapse
Affiliation(s)
- Dhanashree Murugan
- School of Biosciences & Technology (SBST), Vellore Institute of Technology (VIT), Vellore 632014, India
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Vasanth Murugesan
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India
- School of Advanced Sciences (SAS), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Balaji Panchapakesan
- Small Systems Laboratory, Department of Mechanical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA
- Correspondence: (B.P.); (L.R.)
| | - Loganathan Rangasamy
- Drug Discovery Unit (DDU), Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), Vellore Institute of Technology (VIT), Vellore 632014, India
- Correspondence: (B.P.); (L.R.)
| |
Collapse
|
70
|
Yang LK, Lin CX, Li SH, Liang JJ, Xiao LL, Xie GH, Liu HW, Liao X. Novel IKZF3 transcriptomic signature correlates with positive outcomes of skin cutaneous melanoma: A pan-cancer analysis. Front Genet 2022; 13:1036402. [PMID: 36353107 PMCID: PMC9638148 DOI: 10.3389/fgene.2022.1036402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/07/2022] [Indexed: 11/27/2022] Open
Abstract
To investigate the potential relationship between Ikaros family genes and skin cutaneous melanoma (SKCM), we undertook a pan-cancer analysis of the transcriptional signature and clinical data of melanoma through multiple databases. First, 10,327 transcriptomic samples from different cancers were included to determine the overall characteristics and clinical prognoses associated with Ikaros gene expression across cancer types. Second, differentially expressed genes analysis, prognostic evaluation, and gene set enrichment analysis were employed to investigate the role of Ikaros (IKZF) genes in SKCM. Third, we evaluated the relationship between Ikaros family genes and SKCM immune infiltrates and verified the findings using the GEO single-cell sequencing dataset. The results show that Ikaros genes were widely expressed among different cancer types with independently similar patterns as follows: 1. IKZF1 and IKZF3, and 2. IKZF2 and IKZF4–5. IKZF2 and IKZF5 were downregulated in the primary tumor, and IKZF1–3 expression decreased significantly as the T-stage or metastasis increased in SKCM. Moreover, high IKZF1–3 expression was associated with better overall survival, disease-specific survival, and progression-free interval. IKZF3 is an independent prognostic factor of SKCM. Among Ikaros genes, the expression of IKZF1 and IKZF3 positively correlated with the infiltration level of CD4+ T cells and CD8+ T cells, B cells, and Tregs in SKCM and negatively correlated with the infiltration level of M0 and M1 macrophages. Moreover, single-cell sequencing data analysis revealed that IKZF1 and IKZF3 were mainly expressed by immune cells. Correlation analysis shows the immune factors and drug responses associated with IKZF3 expression. In conclusion, the present study is the first, to our knowledge, to identify a pan-cancer genomic signature of the Ikaros gene family among different cancers. Expression of these family members, particularly high levels of IKZF3, indicate positive immunological status and beneficial clinical outcomes of SKCM. IKZF3 may therefore serve as potential targets for immunotherapy of melanoma.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xuan Liao
- *Correspondence: Hong-Wei Liu, ; Xuan Liao,
| |
Collapse
|
71
|
Pęczek P, Gajda M, Rutkowski K, Fudalej M, Deptała A, Badowska-Kozakiewicz AM. Cancer-associated inflammation: pathophysiology and clinical significance. J Cancer Res Clin Oncol 2022; 149:2657-2672. [PMID: 36260158 PMCID: PMC9579684 DOI: 10.1007/s00432-022-04399-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 10/05/2022] [Indexed: 11/28/2022]
Abstract
Purpose Cancer cells, despite stemming from the own cells of their host, usually elicit an immune response. This response usually enables elimination of cancer at its earliest stages. However, some tumors develop mechanisms of escaping immune destruction and even profiting from tumor-derived inflammation. Methods We summarized the roles of different immune cell populations in various processes associated with cancer progression and possible methods of reshaping tumor-associated inflammation to increase the efficacy of cancer therapy. Results Changes in various signaling pathways result in attraction of immunosuppressive, pro-tumorigenic cells, such as myeloid-derived suppressor cells, tumor-associated macrophages, and neutrophils, while at the same time suppressing the activity of lymphocytes, which have the potential of destroying cancer cells. These changes promote tumor progression by increasing angiogenesis and growth, accelerating metastasis, and impairing drug delivery to the tumor site. Conclusion Due to its multi-faceted role in cancer, tumor-associated inflammation can serve as a valuable therapy target. By increasing it, whether through decreasing overall immunosuppression with immune checkpoint inhibitor therapy or through more specific methods, such as cancer vaccines, oncolytic viruses, or chimeric antigen receptor T cells, cancer-derived immunosuppression can be overcome, resulting in immune system destroying cancer cells. Even changes occurring in the microbiota can influence the shape of antitumor response, which could provide new attractive diagnostic or therapeutic methods. Interestingly, also decreasing the distorted tumor-associated inflammation with non-steroidal anti-inflammatory drugs can lead to positive outcomes.
Collapse
Affiliation(s)
- Piotr Pęczek
- Department of Cancer Prevention, Students' Scientific Organization of Cancer Cell Biology, Medical University of Warsaw, Warsaw, Poland
| | - Monika Gajda
- Department of Cancer Prevention, Students' Scientific Organization of Cancer Cell Biology, Medical University of Warsaw, Warsaw, Poland
| | - Kacper Rutkowski
- Department of Cancer Prevention, Students' Scientific Organization of Cancer Cell Biology, Medical University of Warsaw, Warsaw, Poland
| | - Marta Fudalej
- Department of Cancer Prevention, Medical University of Warsaw, Erazma Ciołka 27, Warsaw, Poland.,Department of Oncology and Haematology, Central Clinical Hospital of the Ministry of Interior and Administration, Warsaw, Poland
| | - Andrzej Deptała
- Department of Cancer Prevention, Medical University of Warsaw, Erazma Ciołka 27, Warsaw, Poland.,Department of Oncology and Haematology, Central Clinical Hospital of the Ministry of Interior and Administration, Warsaw, Poland
| | | |
Collapse
|
72
|
High Expression of COL10A1 Is an Independent Predictive Poor Prognostic Biomarker and Associated with Immune Infiltration in Advanced Gastric Cancer Microenvironment. JOURNAL OF ONCOLOGY 2022; 2022:1463316. [PMID: 36276283 PMCID: PMC9584694 DOI: 10.1155/2022/1463316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022]
Abstract
Medical technology has become more and more sophisticated recently, which, however, fails to contribute to a better prognosis for patients suffering advanced gastric cancer (GC). Hence, new biomarkers specific to GC diagnosis and prognosis shall be identified urgently. This study screened differentially expressed genes (DEGs) between 375 GC samples and 32 paracancer tissue samples from TCGA datasets. The expression of Collagen type X alpha 1 (COL10A1) in GC was analyzed. The chi-square test assisted in analyzing the relevance of COL10A1 to the clinicopathologic characteristics. The Kaplan-Meier method helped to assess the survival curves and log-rank tests assisted in the examination of the differences. The Cox proportional hazard regression model served for analyzing the risk factors for GC. Then, we developed a nomogram that contained the COL10A1 expression and clinical information. Finally, how COL10A1 expression was associated with the immune infiltration was also evaluated. In this study, 7179 upregulated and 3771 downregulated genes were identified. Among them, COL10A1 expression was distinctly increased in GC specimens compared with nontumor specimens. High COL10A1 expression exhibited an obvious relation to tumor T and pathologic stage. ROC assays confirmed the diagnostic value of COL10A1 expression in screening GC samples from normal samples. Survival data displayed that patients with high COL10A1 expression exhibited a shorter OS and DSS than those with low COL10A1 expression. We obtained a predictive nomogram, which could better predict the COL10A1 expression by virtue of discrimination and calibration. The prognostic value of COL10A1 expression was further confirmed in GSE84426 datasets. Immune assays revealed that COL10A1 expression was associated with tumor-filtrating immune cells, like CD8 T cells, cytotoxic cells, DC, eosinophils, iDC, macrophages, mast cells, NK CD56dim cells, NK cells, pDC, T helper cells, Tem, Th1 cells, Th17 cells, and Treg. Overall, we firstly proved that COL10A1 may be a novel and valuable prognostic and diagnostic factor for GC patients. In addition, COL10A1 has potential to be an immune indicator in GC.
Collapse
|
73
|
Xiao J, Li Y, Liu Y, Chen Y, He Z, Peng S, Yin Y. The involvement of homeobox-C 4 in predicting prognosis and unraveling immune landscape across multiple cancers via integrated analysis. Front Genet 2022; 13:1021473. [DOI: 10.3389/fgene.2022.1021473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 09/22/2022] [Indexed: 11/13/2022] Open
Abstract
Background: There has been growing evidence that the aberrantly expressed Homeobox-C 4 (HOXC4) plays crucial roles in the development of some cancer types. However, it remains unclear as far as its expression patterns and prognostic significance are concerned, as is tumor immunity.Methods: To investigate the expression levels and prognostic implications of HOXC4, multiple data sources were used in conjunction with quantitative real-time polymerase chain reaction (qRT-PCR) verification. Afterward, diverse immunological-related analyses, along with anti-cancer drug sensitivity, were performed in a number of cancer types. A further exploration of the underlying mechanisms of HOXC4 in tumorigenesis and immunity was carried out using the Gene Set Enrichment Analysis (GSEA) and the Gene Set Variation Analysis (GSVA).Results: Based on extensive database mining, HOXC4 was ubiquitously expressed across 21 tumor cell lines and significantly higher than that of normal tissues in 21 tumor types. The outcome of survival analysis including overall survival (OS), disease-free interval (DFI), disease-specific survival (DSS) and progression-free interval (PFI) revealed that upregulation of HOXC4 expression in several cancers was associated with worse prognosis. Additionally, HOXC4 was observed to correlate closely with colon adenocarcinoma (COAD), head and neck squamous cell carcinoma (HNSC), lower grade glioma (LGG), liver hepatocellular carcinoma (LIHC), rectum adenocarcinoma (READ), and thyroid carcinoma (THCA) in terms of tumor immune cells infiltration. As a result of our comprehensive pan-cancer study, we have identified a significant link between the expression of HOXC4 and the efficacy of immunotherapy-related treatments, together with anti-cancer drug sensitivity. As a final note, HOXC4 was found to modulate multiple signaling pathways involved in tumorigenesis and immunity.Conclusion: HOXC4 has been implicated in our study for the first time as an oncogene in cancers with a poor prognosis, potentially laying the groundwork for promising clinical biomarkers and immunotherapy approaches.
Collapse
|
74
|
Liu Q, Zhang W, Jiao R, Lv Z, Lin X, Xiao Y, Zhang K. Rational Nanomedicine Design Enhances Clinically Physical Treatment-Inspired or Combined Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2203921. [PMID: 36002305 PMCID: PMC9561875 DOI: 10.1002/advs.202203921] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/03/2022] [Indexed: 05/19/2023]
Abstract
Independent of tumor type and non-invasive or minimally-invasive feature, current physical treatments including ultrasound therapy, microwave ablation (MWA), and radiofrequency ablation (RFA) are widely used as the local treatment methods in clinics for directly killing tumors and activating systematic immune responses. However, the activated immune responses are inadequate and incompetent for tumor recession, and the incomplete thermal ablation even aggravates the immunosuppressive tumor microenvironment (ITM), resulting in the intractable tumor recurrence and metastasis. Intriguingly, nanomedicine provides a powerful platform as they can elevate energy utilization efficiency and augment oncolytic effects for mitigating ITM and potentiating the systematic immune responses. Especially after combining with clinical immunotherapy, the anti-tumor killing effect by activating or enhancing the human anti-tumor immune system is reached, enabling the effective prevention against tumor recurrence and metastasis. This review systematically introduces the cutting-edge progress and direction of nanobiotechnologies and their corresponding nanomaterials. Moreover, the enhanced physical treatment efficiency against tumor progression, relapse, and metastasis via activating or potentiating the autologous immunity or combining with exogenous immunotherapeutic agents is exemplified, and their rationales are analyzed. This review offers general guidance or directions to enhance clinical physical treatment from the perspectives of immunity activation or magnification.
Collapse
Affiliation(s)
- Qiaoqiao Liu
- Department of RadiologyLiuzhou People's Hospital Affiliated to Guangxi Medical UniversityNo. 8 Wenchang RoadLiuzhou545006P. R. China
- Central LaboratoryShanghai Tenth People's HospitalTongji University School of MedicineShanghai200072P. R. China
- National Center for International Research of Bio‐targeting TheranosticsGuangxi Key Laboratory of Bio‐targeting TheranosticsGuangxi Medical UniversityNo. 22 Shuangyong Road 22Nanning530021P. R. China
| | - Wei Zhang
- Department of RadiologyLiuzhou People's Hospital Affiliated to Guangxi Medical UniversityNo. 8 Wenchang RoadLiuzhou545006P. R. China
| | - Rong Jiao
- National Center for International Research of Bio‐targeting TheranosticsGuangxi Key Laboratory of Bio‐targeting TheranosticsGuangxi Medical UniversityNo. 22 Shuangyong Road 22Nanning530021P. R. China
| | - Zheng Lv
- Department of RadiologyLiuzhou People's Hospital Affiliated to Guangxi Medical UniversityNo. 8 Wenchang RoadLiuzhou545006P. R. China
- Central LaboratoryShanghai Tenth People's HospitalTongji University School of MedicineShanghai200072P. R. China
| | - Xia Lin
- National Center for International Research of Bio‐targeting TheranosticsGuangxi Key Laboratory of Bio‐targeting TheranosticsGuangxi Medical UniversityNo. 22 Shuangyong Road 22Nanning530021P. R. China
| | - Yunping Xiao
- Department of RadiologyLiuzhou People's Hospital Affiliated to Guangxi Medical UniversityNo. 8 Wenchang RoadLiuzhou545006P. R. China
| | - Kun Zhang
- Department of RadiologyLiuzhou People's Hospital Affiliated to Guangxi Medical UniversityNo. 8 Wenchang RoadLiuzhou545006P. R. China
- Central LaboratoryShanghai Tenth People's HospitalTongji University School of MedicineShanghai200072P. R. China
- National Center for International Research of Bio‐targeting TheranosticsGuangxi Key Laboratory of Bio‐targeting TheranosticsGuangxi Medical UniversityNo. 22 Shuangyong Road 22Nanning530021P. R. China
| |
Collapse
|
75
|
Liu JY, Yao J, Liu JJ, He T, Wang FJ, Xie TY, Cui JX, Yang XD. LASTR is a novel prognostic biomarker and predicts response to cancer immunotherapy in gastric cancer. Front Oncol 2022; 12:1020255. [PMID: 36249015 PMCID: PMC9557225 DOI: 10.3389/fonc.2022.1020255] [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: 08/16/2022] [Accepted: 09/07/2022] [Indexed: 11/24/2022] Open
Abstract
Gastric cancer (GC), a malignant tumor of digestive tract, is characterized by a high death rate. Thus, it is of particular importance to clarify the mechanisms of GC and gain new molecular targets for the sake of preventing and treating GC. It was reported that long non-coding RNAs (IncRNAs) are prognostic factors to cancer. Ferroptosis refers to a process of programmed cell death dependent on iron. This study sets out to investigate the expression and function of ferroptosis-related lncRNA (FRlncRNA) in GC. TCGA datasets offered RNA-seq data for 375 GC patients and clinical data for 443 GC patients. Based on Pearson’s correlation analysis, we studied their expression and identified the FRlncRNAs. Differentially expressed prognosis related to FRlncRNA were determined with the help of the Wilcoxon test and univariate Cox regression analysis. To evaluate the accuracy of the prognostic capacity, researchers used the Kaplan-Meier technique, as well as univariate and multivariate Cox regression and receiver operating characteristic (ROC) curve studies. We also carried out the real-time PCR and CCK8 assays to examine the expression and function of FRlncRNA. In this study, we identified 50 ferroptosis-related DEGs which were involved in tumor progression. In addition, we identified 33 survival-related FRlncRNAs. Among them, lncRNA associated with SART3 regulation of splicing(LASTR) was confirmed to be highly expressed in GC specimens compared to non-tumor specimens in this cohort. Survival assays illuminated that the high LASTR expression predicted a shorter overall survival and progression-free survival of GC patients. Based on multivariate Cox regression analyses, it was confirmed that the GC had a worse chance of surviving the disease overall if their tumors expressed LASTR, which was an independent prognostic indication. Then, Loss-of-function tests showed that knocking down LASTR had a significant effect on reducing the proliferation of GC cells. Finally, we found that the expression of LASTR was negatively associated with CD8 T cells, T cells, Th17 cells, and T helper cells. Overall, our findings identified a novel survival-related FRlncRNA, LASTR which possibly can serve as a novel prognostic biomarker predicting response to cancer immunotherapy and therapeutic target for GC patients.
Collapse
Affiliation(s)
- Jun-Yan Liu
- Department of General Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jing Yao
- Department of General Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jia-Jia Liu
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tao He
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fang-Jie Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Army Medical Center (Daping Hospital), Army Medical University, Chongqing, China
| | - Tian-Yu Xie
- Department of General Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
| | - Jian-Xin Cui
- Department of General Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Jian-Xin Cui, ; Xiao-Dong Yang,
| | - Xiao-Dong Yang
- Department of General Surgery, The First Medical Center, Chinese People's Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Jian-Xin Cui, ; Xiao-Dong Yang,
| |
Collapse
|
76
|
Liu J, Tao H, Yuan T, Li J, Li J, Liang H, Huang Z, Zhang E. Immunomodulatory effects of regorafenib: Enhancing the efficacy of anti-PD-1/PD-L1 therapy. Front Immunol 2022; 13:992611. [PMID: 36119072 PMCID: PMC9479218 DOI: 10.3389/fimmu.2022.992611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/18/2022] [Indexed: 11/14/2022] Open
Abstract
Anti-PD-1/PD-L1 therapy has shown significant benefits in the treatment of a variety of malignancies. However, not all cancer patients can benefit from this strategy due to drug resistance. Therefore, there is an urgent need for methods that can effectively improve the efficacy of anti-PD-1/PD-L1 therapy. Combining anti-PD-1/PD-L1 therapy with regorafenib has been demonstrated as an effective method to enhance its therapeutic effect in several clinical studies. In this review, we describe common mechanisms of resistance to anti-PD-1/PD-L1 therapy, including lack of tumor immunogenicity, T cell dysfunction, and abnormal expression of PD-L1. Then, we illustrate the role of regorafenib in modifying the tumor microenvironment (TME) from multiple aspects, which is different from other tyrosine kinase inhibitors. Regorafenib not only has immunomodulatory effects on various immune cells, but can also regulate PD-L1 and MHC-I on tumor cells and promote normalization of abnormal blood vessels. Therefore, studies on the synergetic mechanism of the combination therapy may usher in a new era for cancer treatment and help us identify the most appropriate individuals for more precise treatment.
Collapse
Affiliation(s)
- Junjie Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haisu Tao
- Department of Hepatobiliary Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Tong Yuan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiang Li
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian Li
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Erlei Zhang, ; Zhiyong Huang, ; Huifang Liang,
| | - Zhiyong Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Erlei Zhang, ; Zhiyong Huang, ; Huifang Liang,
| | - Erlei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Erlei Zhang, ; Zhiyong Huang, ; Huifang Liang,
| |
Collapse
|
77
|
Aria H, Rezaei M, Nazem S, Daraei A, Nikfar G, Mansoori B, Bahmanyar M, Tavassoli A, Vakil MK, Mansoori Y. Purinergic receptors are a key bottleneck in tumor metabolic reprogramming: The prime suspect in cancer therapeutic resistance. Front Immunol 2022; 13:947885. [PMID: 36072596 PMCID: PMC9444135 DOI: 10.3389/fimmu.2022.947885] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
ATP and other nucleoside phosphates have specific receptors named purinergic receptors. Purinergic receptors and ectonucleotidases regulate various signaling pathways that play a role in physiological and pathological processes. Extracellular ATP in the tumor microenvironment (TME) has a higher level than in normal tissues and plays a role in cancer cell growth, survival, angiogenesis, metastasis, and drug resistance. In this review, we investigated the role of purinergic receptors in the development of resistance to therapy through changes in tumor cell metabolism. When a cell transforms to neoplasia, its metabolic processes change. The metabolic reprogramming modified metabolic feature of the TME, that can cause impeding immune surveillance and promote cancer growth. The purinergic receptors contribute to therapy resistance by modifying cancer cells' glucose, lipid, and amino acid metabolism. Limiting the energy supply of cancer cells is one approach to overcoming resistance. Glycolysis inhibitors which reduce intracellular ATP levels may make cancer cells more susceptible to anti-cancer therapies. The loss of the P2X7R through glucose intolerance and decreased fatty acid metabolism reduces therapeutic resistance. Potential metabolic blockers that can be employed in combination with other therapies will aid in the discovery of new anti-cancer immunotherapy to overcome therapy resistance. Therefore, therapeutic interventions that are considered to inhibit cancer cell metabolism and purinergic receptors simultaneously can potentially reduce resistance to treatment.
Collapse
Affiliation(s)
- Hamid Aria
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Marzieh Rezaei
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shima Nazem
- Department of Laboratory Medicine, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abdolreza Daraei
- Department of Medical Genetics, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Ghasem Nikfar
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Behnam Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Maryam Bahmanyar
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Alireza Tavassoli
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Mohammad Kazem Vakil
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
- Department of Medical Genetics, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
78
|
Liu J, Li W, Wu L. Pan-cancer analysis suggests histocompatibility minor 13 is an unfavorable prognostic biomarker promoting cell proliferation, migration, and invasion in hepatocellular carcinoma. Front Pharmacol 2022; 13:950156. [PMID: 36046831 PMCID: PMC9421072 DOI: 10.3389/fphar.2022.950156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/21/2022] [Indexed: 02/05/2023] Open
Abstract
Histocompatibility Minor 13 (HM13) encoding the signal peptide peptidase plays an important role in maintaining protein homeostasis but its role in tumors remains unclear. In this study, 33 tumor RNA-seq datasets were extracted from The Cancer Genome Atlas (TCGA) database, and the pan-cancer expression profile of HM13 was evaluated in combination with The Genotype-Tissue Expression (GTEx) datasets. The prognostic significance of abnormal HM13 pan-cancer expression was evaluated by univariate Cox regression and Kaplan-Meier analyses. Co-expression analysis was performed to examine the correlation between abnormal pan-cancer expression of HM13 and immune cell infiltration, immune checkpoint, molecules related to RNA modification, tumor mutational burden (TMB), microsatellite instability (MSI), and other related molecules. CellMiner database was used to evaluate the relationship between the expression of HM13 and drug sensitivity. The results showed overexpression of HM13 in almost all tumors except kidney chromophobe (KICH). Abnormally high expression of HM13 in adrenocortical carcinoma (ACC), kidney renal papillary cell carcinoma (KIRP), uveal melanoma (UVM), liver hepatocellular carcinoma (LIHC), brain lower grade glioma (LGG), head and neck squamous cell carcinoma (HNSC), and kidney renal clear cell carcinoma (KIRC) was associated with poor prognosis. Expression of HM13 correlated strongly with pan-cancer immune checkpoint gene expression and immune cell infiltration. Drug sensitivity analysis indicated that the expression of HM13 was an excellent predictor of drug sensitivity. We verified that both mRNA and protein levels of HM13 were abnormally upregulated in HCC tissues, and were independent risk factors for poor prognosis. Furthermore, interference with HM13 expression in Huh-7 and HCCLM3 cells significantly inhibited proliferation, migration, and invasion. Therefore, our findings demonstrate that HM13 is a potential pan-cancer prognostic marker, thus providing a new dimension for understanding tumor development.
Collapse
Affiliation(s)
- Jun Liu
- Department of Clinical Laboratory, Yue Bei People’s Hospital, Shantou University Medical College, Shaoguan, Guangdong, China
- Medical Research Center, Yue Bei People’s Hospital, Shantou University Medical College, Shaoguan, Guangdong, China
| | - Wenli Li
- Reproductive Medicine Center, Yue Bei People’s Hospital, Shantou University Medical College, Shaoguan, Guangdong, China
| | - Liangyin Wu
- Department of Clinical Laboratory, Yue Bei People’s Hospital, Shantou University Medical College, Shaoguan, Guangdong, China
| |
Collapse
|
79
|
Ouyang Y, Huang J, Wang Y, Tang F, Hu Z, Zeng Z, Zhang S. Bioinformatic analysis of RNA-seq data from TCGA database reveals prognostic significance of immune-related genes in colon cancer. Medicine (Baltimore) 2022; 101:e29962. [PMID: 35945793 PMCID: PMC9351934 DOI: 10.1097/md.0000000000029962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The tumor immune microenvironment is of crucial importance in cancer progression and anticancer immune responses. Thus, systematic exploration of the expression landscape and prognostic significance of immune-related genes (IRGs) to assist in the prognosis of colon cancer is valuable and significant. The transcriptomic data of 470 colon cancer patients were obtained from The Cancer Genome Atlas database and the differentially expressed genes were analyzed. After an intersection analysis, the hub IRGs were identified and a prognostic index was further developed using multivariable Cox analysis. In addition, the discriminatory ability and prognostic significance of the constructed model were validated and the characteristics of IRGs associated overall survival were analyzed to elucidate the underlying molecular mechanisms. A total of 465 differentially expressed IRGs and 130 survival-associated IRGs were screened. Then, 46 hub IRGs were identified by an intersection analysis. A regulatory network displayed that most of these genes were unfavorable for the prognosis of colon cancer and were regulated by transcription factors. After a least absolute shrinkage and selection operator regression analysis, 14 hub IRGs were ultimately chose to construct a prognostic index. The validation results illustrated that this model could act as an independent indicator to moderately separate colon cancer patients into low- and high-risk groups. This study ascertained the prognostic significance of IRGs in colon cancer and successfully constructed an IRG-based prognostic signature for clinical prediction. Our results provide promising insight for the exploration of diagnostic markers and immunotherapeutic targets in colon cancer.
Collapse
Affiliation(s)
- Yan Ouyang
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province/Immune Cells and Antibody Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Jiangtao Huang
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province/Immune Cells and Antibody Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Yun Wang
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province/Immune Cells and Antibody Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Fuzhou Tang
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province/Immune Cells and Antibody Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| | - Zuquan Hu
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province/Immune Cells and Antibody Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education of China, Guizhou Medical University, Guiyang, China
- *Correspondence: Zuquan Hu, Department of Medical Biotechnology, School of Biology and Engineering, Guizhou Medical University, Guiyang, Guizhou, China (e-mail: )
| | - Zhu Zeng
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province/Immune Cells and Antibody Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Engineering Center of Cellular Immunotherapy of Guizhou Province, Guizhou Medical University, Guiyang, China
| | - Shichao Zhang
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province/Immune Cells and Antibody Engineering Research Center of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
| |
Collapse
|
80
|
Tang YL, Li GS, Li DM, Tang D, Huang JZ, Feng H, He RQ, Huang ZG, Dang YW, Kong JL, Gan TQ, Zhou HF, Zeng JJ, Chen G. The clinical significance of integrin subunit alpha V in cancers: from small cell lung carcinoma to pan-cancer. BMC Pulm Med 2022; 22:300. [PMID: 35927660 PMCID: PMC9354352 DOI: 10.1186/s12890-022-02095-8] [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: 04/14/2022] [Accepted: 07/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Little is known about the relationship between integrin subunit alpha V (ITGAV) and cancers, including small cell lung cancer (SCLC). METHODS Using large sample size from multiple sources, the clinical roles of ITGAV expression in SCLC were explored using differential expression analysis, receiver operating characteristic curves, Kaplan-Meier curves, etc. RESULTS: Decreased mRNA (SMD = - 1.05) and increased protein levels of ITGAV were detected in SCLC (n = 865). Transcription factors-ZEB2, IK2F1, and EGR2-may regulate ITGAV expression in SCLC, as they had ChIP-Seq (chromatin immunoprecipitation followed by sequencing) peaks upstream of the transcription start site of ITGAV. ITGAV expression made it feasible to distinguish SCLC from non-SCLC (AUC = 0.88, sensitivity = 0.78, specificity = 0.84), and represented a risk role in the prognosis of SCLC (p < 0.05). ITGAV may play a role in cancers by influencing several immunity-related signaling pathways and immune cells. Further, the extensive pan-cancer analysis verified the differential expression of ITGAV and its clinical significance in multiple cancers. CONCLUSION ITGAV served as a potential marker for prognosis and identification of cancers including SCLC.
Collapse
Affiliation(s)
- Yu-Lu Tang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Guo-Sheng Li
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Dong-Ming Li
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Deng Tang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Jie-Zhuang Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Hao Feng
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Rong-Quan He
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Zhi-Guang Huang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Yi-Wu Dang
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Jin-Liang Kong
- Ward of Pulmonary and Critical Care Medicine, Department of Respiratory Medicine, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Ting-Qing Gan
- Department of Medical Oncology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Hua-Fu Zhou
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Jing-Jing Zeng
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Gang Chen
- Department of Pathology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, People's Republic of China.
| |
Collapse
|
81
|
Li D, Pang X, Zhu X, Shanzhou Q, Wen G, Ma D. Low-dose radiotherapy combined with immunotherapy for suboral adenoid cystic carcinoma with bilateral lung metastasis: A case report and literature review. Oncol Lett 2022; 24:279. [PMID: 35814824 PMCID: PMC9260714 DOI: 10.3892/ol.2022.13399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/30/2022] [Indexed: 11/23/2022] Open
Abstract
Adenoid cystic carcinoma (ACC) is a type of malignant tumor arising from the salivary glands. The tumor is characterized by a predilection for recurrence and metastasis. At present, there is no effective treatment for ACC complicated with lung metastasis. The present study reported on a case of suboral ACC with bilateral lung metastasis and the clinical features and treatment options were discussed based on a literature review. A 55-year-old female presented with suboral ACC accompanied with bilateral lung metastases. The main symptoms were masses in the right mandible and shortness of breath. Low-dose radiotherapy (LDRT) combined with immunotherapy were administered to control lung metastases. The patient is currently in a stable condition and is receiving immunotherapy. LDRT combined with immunotherapy is a feasible treatment option for such patients. Our experience with this case and the information from the literature review provide insight into the therapeutic approach for this relatively rare entity.
Collapse
Affiliation(s)
- Danyang Li
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Xuezhou Pang
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Xinxin Zhu
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Qiyue Shanzhou
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Guo Wen
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Daiyuan Ma
- Department of Oncology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
82
|
Cornelison C, Fadel S. Clickable Biomaterials for Modulating Neuroinflammation. Int J Mol Sci 2022; 23:8496. [PMID: 35955631 PMCID: PMC9369181 DOI: 10.3390/ijms23158496] [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] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 02/04/2023] Open
Abstract
Crosstalk between the nervous and immune systems in the context of trauma or disease can lead to a state of neuroinflammation or excessive recruitment and activation of peripheral and central immune cells. Neuroinflammation is an underlying and contributing factor to myriad neuropathologies including neurodegenerative diseases like Alzheimer's disease and Parkinson's disease; autoimmune diseases like multiple sclerosis; peripheral and central nervous system infections; and ischemic and traumatic neural injuries. Therapeutic modulation of immune cell function is an emerging strategy to quell neuroinflammation and promote tissue homeostasis and/or repair. One such branch of 'immunomodulation' leverages the versatility of biomaterials to regulate immune cell phenotypes through direct cell-material interactions or targeted release of therapeutic payloads. In this regard, a growing trend in biomaterial science is the functionalization of materials using chemistries that do not interfere with biological processes, so-called 'click' or bioorthogonal reactions. Bioorthogonal chemistries such as Michael-type additions, thiol-ene reactions, and Diels-Alder reactions are highly specific and can be used in the presence of live cells for material crosslinking, decoration, protein or cell targeting, and spatiotemporal modification. Hence, click-based biomaterials can be highly bioactive and instruct a variety of cellular functions, even within the context of neuroinflammation. This manuscript will review recent advances in the application of click-based biomaterials for treating neuroinflammation and promoting neural tissue repair.
Collapse
Affiliation(s)
- Chase Cornelison
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA;
| | | |
Collapse
|
83
|
Lv B, Wang Y, Ma D, Cheng W, Liu J, Yong T, Chen H, Wang C. Immunotherapy: Reshape the Tumor Immune Microenvironment. Front Immunol 2022; 13:844142. [PMID: 35874717 PMCID: PMC9299092 DOI: 10.3389/fimmu.2022.844142] [Citation(s) in RCA: 186] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 06/13/2022] [Indexed: 12/12/2022] Open
Abstract
Tumor immune microenvironment (TIME) include tumor cells, immune cells, cytokines, etc. The interactions between these components, which are divided into anti-tumor and pro-tumor, determine the trend of anti-tumor immunity. Although the immune system can eliminate tumor through the cancer-immune cycle, tumors appear to eventually evade from immune surveillance by shaping an immunosuppressive microenvironment. Immunotherapy reshapes the TIME and restores the tumor killing ability of anti-tumor immune cells. Herein, we review the function of immune cells within the TIME and discuss the contribution of current mainstream immunotherapeutic approaches to remolding the TIME. Changes in the immune microenvironment in different forms under the intervention of immunotherapy can shed light on better combination treatment strategies.
Collapse
Affiliation(s)
- Bingzhe Lv
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yunpeng Wang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Dongjiang Ma
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Wei Cheng
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Jie Liu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Tao Yong
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Hao Chen
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China.,Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Chen Wang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, China.,Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
84
|
Zhang Y, Zou J, Chen R. An M0 macrophage-related prognostic model for hepatocellular carcinoma. BMC Cancer 2022; 22:791. [PMID: 35854246 PMCID: PMC9294844 DOI: 10.1186/s12885-022-09872-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 07/04/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The role of M0 macrophages and their related genes in the prognosis of hepatocellular carcinoma (HCC) remains poorly characterized. METHODS Multidimensional bioinformatic methods were used to construct a risk score model using M0 macrophage-related genes (M0RGs). RESULTS Infiltration of M0 macrophages was significantly higher in HCC tissues than in normal liver tissues (P = 2.299e-07). Further analysis revealed 35 M0RGs that were associated with HCC prognosis; two M0RGs (OLA1 and ATIC) were constructed and validated as a prognostic signature for overall survival of patients with HCC. Survival analysis revealed the positive relationship between the M0RG signature and unfavorable prognosis. Correlation analysis showed that this risk model had positive associations with clinicopathological characteristics, somatic gene mutations, immune cell infiltration, immune checkpoint inhibitor targets, and efficacy of common drugs. CONCLUSIONS The constructed M0RG-based risk model may be promising for the clinical prediction of prognoses and therapeutic responses in patients with HCC.
Collapse
Affiliation(s)
- Yiya Zhang
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Ju Zou
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Department of Infectious Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Ruochan Chen
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,Department of Infectious Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
85
|
Shan Q, Zhang Y, Liang Z. Clustering analysis and prognostic signature of lung adenocarcinoma based on the tumor microenvironment. Sci Rep 2022; 12:12059. [PMID: 35835908 PMCID: PMC9283441 DOI: 10.1038/s41598-022-15971-4] [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: 02/18/2022] [Accepted: 07/01/2022] [Indexed: 02/05/2023] Open
Abstract
Because of immunotherapy failure in lung adenocarcinoma, we have tried to find new potential biomarkers for differentiating different tumor subtypes and predicting prognosis. We identified two subtypes based on tumor microenvironment-related genes in this study. We used seven methods to analyze the immune cell infiltration between subgroups. Further analysis of tumor mutation load and immune checkpoint expression among different subgroups was performed. The least absolute shrinkage and selection operator Cox regression was applied for further selection. The selected genes were used to construct a prognostic 14-gene signature for LUAD. Next, a survival analysis and time-dependent receiver operating characteristics were performed to verify and evaluate the model. Gene set enrichment analyses and immune analysis in risk groups was also performed. According to the expression of genes related to the tumor microenvironment, lung adenocarcinoma can be divided into cold tumors and hot tumors. The signature we built was able to predict prognosis more accurately than previously known models. The signature-based tumor microenvironment provides further insight into the prediction of lung adenocarcinoma prognosis and may guide individualized treatment.
Collapse
Affiliation(s)
- Qingqing Shan
- Department of Respiration, Chengdu First People's Hospital, Chengdu, 610041, China
| | - Yifan Zhang
- Department of Respiration, Chengdu First People's Hospital, Chengdu, 610041, China
| | - Zongan Liang
- Department of Respiration, West China Hospital of Sichuan University, 37# Guo Xue Xiang, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
86
|
Construction of a Novel Clinical Stage-Related Gene Signature for Predicting Outcome and Immune Response in Hepatocellular Carcinoma. J Immunol Res 2022; 2022:6535009. [PMID: 35865652 PMCID: PMC9296277 DOI: 10.1155/2022/6535009] [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: 05/22/2022] [Revised: 06/23/2022] [Accepted: 06/28/2022] [Indexed: 11/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) with high heterogeneity is one of the most frequent malignant tumors. However, there were no studies to create a clinical stage-related gene signature for HCC patients. Differentially expressed genes (DEGs) associated with clinical stage of HCC were analyzed based on TCGA datasets. Functional enrichment analysis was carried out by the use of stage-related DEGs. Then, the least absolute shrinkage and selection operator (LASSO) regression and univariate Cox regression were performed to reduce the overfit and the number of genes for further analysis. Next, survival and ROC assays were carried out to demonstrate the model using TCGA. Functional analysis and immune microenvironment analysis related to stage-related DEGs were performed. Reverse transcriptase polymerase chain reaction (RT-PCR) and Cell Counting Kit-8 (CCK-8) assays were applied to examine the expression and function of PNCK in HCC. In this research, there were 21 DEGs between HCC specimens with stage (I-II) and HCC specimens with stage (III-IV), including 20 increased genes and 1 decreased genes. A novel seven-gene signature (including PITX2, PNCK, GLIS1, SCNN1G, MMP1, ZNF488, and SHISA9) was created for the prediction of outcomes of HCC patients. The ROC curves confirmed the prognostic value of the new model. Cox assays demonstrated that the seven-gene signature can independently forecast overall survival. The immune analysis revealed that patients with low risk score exhibited more immune activities. Moreover, we confirmed that PNCK expressions were distinctly increased in HCC, and its silence suppressed the proliferation of HCC cells. Overall, our research offered a robust and reliable gene signature which displayed an important value in the prediction of overall survival of HCC patients and might deliver more effective personalized therapies.
Collapse
|
87
|
Zhang B, Tang B, Lv J, Gao J, Qin L. Systematic analyses to explore immune gene sets-based signature in hepatocellular carcinoma, in which IGF2BP3 contributes to tumor progression. Clin Immunol 2022; 241:109073. [PMID: 35817291 DOI: 10.1016/j.clim.2022.109073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 06/17/2022] [Accepted: 07/02/2022] [Indexed: 11/16/2022]
Abstract
Tumor immune microenvironment (TIME) is of critical importance for the development and therapeutic response of hepatocellular carcinoma (HCC). However, limited studies have investigated immune-related indicators for clinical supervision and decision. The current study aimed to develop an improved prognostic signature based on TIME. HCC patients from TCGA and ICGC database were classified into three subtypes (Immunity High, Immunity Medium and Immunity Low) according to ssGSEA scores of 29 immune gene sets. Differentially expressed immune-related genes (DE IRGs) between Immune High and Low groups were screened with an adjusted P < 0.05. Weighted gene co-expression network analysis (WGCNA) was used to establish gene co-expression modules of differentially expressed genes (DEGs) between tumor and normal tissues. 45 survival-related immune genes (SRIGs) were identified at points of intersection between hub genes and DE IRGs. By performing Cox regression and LASSO analysis, 3 of the 45 SRIGs were screened to establish a prognostic model. Patients with high risk scores exhibited worse survival outcome and poorer response to chemotherapy. Potential mechanisms of chemotherapy resistance also have been discussed. More significantly, high -risk patients showed increased immune cell infiltration and checkpoints, which suggested a benefit of immunotherapy. In addition, knockdown of IGF2BP3 was determined to significantly inhibit cell proliferation and migration in HCC. Our immune-related model may be an effective tool for precise diagnosis and treatment of HCC. It may help to select patients suitable for chemotherapy, and immunotherapy.
Collapse
Affiliation(s)
- Baohui Zhang
- Department of Physiology, School of Life Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, PR China
| | - Bufu Tang
- Departmcent of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| | - Jiarui Lv
- Department of Physiology, School of Life Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, PR China
| | - Jianyao Gao
- Department of Radiation Oncology, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ling Qin
- Department of Physiology, School of Life Science, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, Liaoning Province 110122, PR China.
| |
Collapse
|
88
|
Rossi A, Belmonte B, Carnevale S, Liotti A, De Rosa V, Jaillon S, Piconese S, Tripodo C. Stromal and Immune Cell Dynamics in Tumor Associated Tertiary Lymphoid Structures and Anti-Tumor Immune Responses. Front Cell Dev Biol 2022; 10:933113. [PMID: 35874810 PMCID: PMC9304551 DOI: 10.3389/fcell.2022.933113] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
Tertiary lymphoid structures (TLS) are ectopic lymphoid organs that have been observed in chronic inflammatory conditions including cancer, where they are thought to exert a positive effect on prognosis. Both immune and non-immune cells participate in the genesis of TLS by establishing complex cross-talks requiring both soluble factors and cell-to-cell contact. Several immune cell types, including T follicular helper cells (Tfh), regulatory T cells (Tregs), and myeloid cells, may accumulate in TLS, possibly promoting or inhibiting their development. In this manuscript, we propose to review the available evidence regarding specific aspects of the TLS formation in solid cancers, including 1) the role of stromal cell composition and architecture in the recruitment of specific immune subpopulations and the formation of immune cell aggregates; 2) the contribution of the myeloid compartment (macrophages and neutrophils) to the development of antibody responses and the TLS formation; 3) the immunological and metabolic mechanisms dictating recruitment, expansion and plasticity of Tregs into T follicular regulatory cells, which are potentially sensitive to immunotherapeutic strategies directed to costimulatory receptors or checkpoint molecules.
Collapse
Affiliation(s)
- Alessandra Rossi
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care “G. D’Alessandro”, University of Palermo, Palermo, Italy
| | | | - Antonietta Liotti
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale, Consiglio Nazionale Delle Ricerche, Naples, Italy
| | - Veronica De Rosa
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale, Consiglio Nazionale Delle Ricerche, Naples, Italy
| | - Sebastien Jaillon
- RCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Silvia Piconese
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
- IRCCS Fondazione Santa Lucia, Unità di Neuroimmunologia, Rome, Italy
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Rome, Italy
- *Correspondence: Silvia Piconese,
| | - Claudio Tripodo
- Tumor Immunology Unit, Department of Sciences for Health Promotion and Mother-Child Care “G. D’Alessandro”, University of Palermo, Palermo, Italy
- Histopathology Unit, FIRC Institute of Molecular Oncology (IFOM), Milan, Italy
| |
Collapse
|
89
|
Shi L, Zhu W, Huang Y, Zhuo L, Wang S, Chen S, Zhang B, Ke B. Cancer-associated fibroblast-derived exosomal microRNA-20a suppresses the PTEN/PI3K-AKT pathway to promote the progression and chemoresistance of non-small cell lung cancer. Clin Transl Med 2022; 12:e989. [PMID: 35857905 PMCID: PMC9299573 DOI: 10.1002/ctm2.989] [Citation(s) in RCA: 93] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 07/02/2022] [Accepted: 07/07/2022] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs) contributes to overall tumor progression. In the current survey, we explored the ability of microRNA-20a (miR-20a) within these CAF-derived exosomes to influence non-small-cell lung cancer (NSCLC) progression. MATERIALS AND METHODS Normal tissue-associated fibroblasts (NAFs) and CAFs were collected from samples of NSCLC patient tumors and paracancerous lung tissues. Exosomes derived from these cells were then characterized via Western blotting, nanoparticle tracking analyses, and transmission electron microscopy. The expression of miR-20a was assessed via qPCR and fluorescence in situ hybridization (FISH). CCK-8, EdU uptake, and colony formation assessments were used for evaluating tumor proliferation, while Hoechst staining was performed to monitor the in vitro apoptotic death of tumor cells. A model of xenograft tumor established in nude mice was also used to evaluate in vivo tumor responses. RESULTS CAF-derived exosomes exhibited miR-20a upregulation and promoted NSCLC cell proliferation and resistance to cisplatin (DDP). Mechanistically, CAF-derived exosomes were discovered to transmit miR-20a to tumor cells wherein it was able to target PTEN to enhance DDP resistance and proliferation. Associated PTEN downregulation following exosome-derived miR-20a treatment enhanced PI3K/AKT pathway activation. CONCLUSION The achieved outcomes explain that CAFs can release miR-20a-containing exosomes capable of promoting NSCLC progression and chemoresistance, highlighting this pathway as a possible therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Lin Shi
- Department of Traditional Chinese MedicineZhujiang Hospital of Southern Medical UniversityGuangzhouChina
| | - Weiliang Zhu
- Department of Cancer CenterZhujiang Hospital of Southern Medical UniversityGuangzhouChina
| | - Yuanyuan Huang
- Department of VIP RegionState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center of Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Lin Zhuo
- Department of Traditional Chinese MedicineZhujiang Hospital of Southern Medical UniversityGuangzhouChina
| | - Siyun Wang
- Department of Traditional Chinese MedicineZhujiang Hospital of Southern Medical UniversityGuangzhouChina
| | - Shaobing Chen
- Department of Traditional Chinese MedicineZhujiang Hospital of Southern Medical UniversityGuangzhouChina
| | - Bei Zhang
- Department of VIP RegionState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center of Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| | - Bin Ke
- Department of VIP RegionState Key Laboratory of Oncology in South ChinaCollaborative Innovation Center of Cancer MedicineSun Yat‐sen University Cancer CenterGuangzhouChina
| |
Collapse
|
90
|
Zhao Q, Gao S, Chen X, Zhu X. POC1A, prognostic biomarker of immunosuppressive microenvironment in cancer. Aging (Albany NY) 2022; 14:5195-5210. [PMID: 35748773 PMCID: PMC9271305 DOI: 10.18632/aging.204141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 06/14/2022] [Indexed: 11/25/2022]
Abstract
POC1 centriolar protein A (POC1A) effect in pan-cancer remains uncertain. The POC1A expression in normal and tumor tissues underwent analysis in this study utilizing data from the Genotype-Tissue Expression (GTEx) project and the Cancer Genome Atlas (TCGA) database. POC1A prognostic value and clinicopathological features were assessed utilizing the TCGA cohort. The relationship between immunological cell infiltration and POC1A of TCGA samples downloaded from TIMER2 and ImmuCellAI databases were observed. The relation between POC1A and immunological checkpoints genes, microsatellite instability (MSI) as well as tumor mutation burden (TMB) was also evaluated. Additionally, gene set enrichment analysis (GSEA) was utilized for exploring POC1A potential molecular mechanism in pan-cancer. In almost all 33 tumors, POCA1 showed a high expression. In most cases, high POC1A expression was linked significantly with a poor prognosis. Additionally, Tumor immune infiltration and tumors microenvironment were correlated with the expression of POC1A. In addition, TMB and MSI, as well as immune checkpoint genes in pan-cancer, were related to POC1A expression. In GSEA analysis, POC1A is implicated in cell cycle and immune-related pathways. These results might elucidate the crucial roles of POC1A in pan-cancer as a prognostic biomarker and immunotherapy target.
Collapse
Affiliation(s)
- Qi Zhao
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Shuping Gao
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Xin Chen
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Xiyan Zhu
- Department of Thoracic Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| |
Collapse
|
91
|
Zhu T, Han J, Yang L, Cai Z, Sun W, Hua Y, Xu J. Immune Microenvironment in Osteosarcoma: Components, Therapeutic Strategies and Clinical Applications. Front Immunol 2022; 13:907550. [PMID: 35720360 PMCID: PMC9198725 DOI: 10.3389/fimmu.2022.907550] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/26/2022] [Indexed: 12/21/2022] Open
Abstract
Osteosarcoma is a primary malignant tumor that tends to threaten children and adolescents, and the 5-year event-free survival rate has not improved significantly in the past three decades, bringing grief and economic burden to patients and society. To date, the genetic background and oncogenesis mechanisms of osteosarcoma remain unclear, impeding further research. The tumor immune microenvironment has become a recent research hot spot, providing novel but valuable insight into tumor heterogeneity and multifaceted mechanisms of tumor progression and metastasis. However, the immune microenvironment in osteosarcoma has been vigorously discussed, and the landscape of immune and non-immune component infiltration has been intensively investigated. Here, we summarize the current knowledge of the classification, features, and functions of the main infiltrating cells, complement system, and exosomes in the osteosarcoma immune microenvironment. In each section, we also highlight the complex crosstalk network among them and the corresponding potential therapeutic strategies and clinical applications to deepen our understanding of osteosarcoma and provide a reference for imminent effective therapies with reduced adverse effects.
Collapse
Affiliation(s)
- Tianyi Zhu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, Shanghai, China
| | - Jing Han
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, Shanghai, China
| | - Liu Yang
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, Shanghai, China
| | - Zhengdong Cai
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, Shanghai, China
| | - Wei Sun
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, Shanghai, China
| | - Yingqi Hua
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, Shanghai, China
| | - Jing Xu
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Bone Tumor Institution, Shanghai, China
| |
Collapse
|
92
|
Shen F, Sun L, Wang L, Peng R, Fan C, Liu Z. Framework Nucleic Acid Immune Adjuvant for Transdermal Delivery Based Chemo-immunotherapy for Malignant Melanoma Treatment. NANO LETTERS 2022; 22:4509-4518. [PMID: 35594186 DOI: 10.1021/acs.nanolett.2c01332] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Despite the tremendous progresses of cancer immunotherapy, its current clinical responses rate in melanoma remains to be improved. Here, we have reported a skin penetrating tetrahedral framework nucleic acid immune adjuvant (FNAIA) to transdermally deliver chemotherapy drugs into melanoma to induce the immunogenic death of tumor cells and expose tumor antigens, which with assistance of CpG oligodeoxynucleotide incorporated in FNAIA could trigger systemic tumor-specific immune responses. Compared with free CpG, FNAIA could penetrate deeper into subcutaneous tumor tissues and more effectively stimulate dendritic cell maturation. Notably, doxorubicin-loaded FNAIA locally applied on the intact skin above the melanoma could effectively inhibit the growth of mouse B16F10 melanoma and increase tumor CD8+ T cell infiltration. Moreover, combined with immune checkpoint inhibitor, the growth of distant tumors could also be effectively inhibited, suggesting that this strategy could induce systemic immune responses. Therefore, this work provides a new idea for non-invasive treatment of skin cancer.
Collapse
Affiliation(s)
- Fengyun Shen
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Lele Sun
- School of Life Sciences, Shanghai University, Shanghai 200444, China
| | - Lihua Wang
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Rui Peng
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai 201240, China
| | - Zhuang Liu
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Institute of Functional Nano and Soft Materials (FUNSOM), Collaborative Innovation Center of Suzhou Nano Science and Technology, Soochow University, Suzhou, Jiangsu 215123, China
| |
Collapse
|
93
|
ROS-triggered nanoinducer based on dermatan sulfate enhances immunogenic cell death in melanoma. J Control Release 2022; 348:22-33. [PMID: 35461966 DOI: 10.1016/j.jconrel.2022.04.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/23/2022] [Accepted: 04/15/2022] [Indexed: 12/18/2022]
Abstract
Due to its complexity, diversity and heterogeneity, melanoma is a kind of malignant tumor. It has been proved that the enhancement of anti-tumor immune response such as immunogenic cell death (ICD) is an important therapeutic strategy. In previous studies, we confirmed that dermatan sulfate (DS) from skin tissue could specifically homing to melanoma B16F10 cells. In this study, we propose a nanoinducer (DOX/ADS NP) based on a functional DS for melanoma. This nanosystem is composed of DS as framework, aromatic thioketal derivative (ATK) as functional grafting unit and doxorubicin (DOX) designed as an ICD inducer. Through the intermolecular interaction between DOX and ATK, DOX/ADS NP with specific-homing, high-loading and ROS-triggering release was obtained via self-assemble. Compared with free DOX and non-functionalized nanomedicine, DOX/ADS NP could release DOX into B16F10 cells better, and strongly induce the translocation of calreticulin (CRT) to the cell membrane. CRT is a marker of ICD, also as a "eat me" signal to stimulate the maturation and antigen presentation of dendritic cells. Therefore, a series of subsequent immune responses were activated: maturation of dendritic cells, T cells proliferation, increased tumor-infiltrating CTLs and the ratio of CTLs to Tregs, and up-regulated cytotoxic cytokine expression. In conclusion, DOX/ADS NP promoted ICD-associated immune response through more specific targeting effect and sensitive responsive DOX release, achieving better inhibitory effect on melanoma than free DOX and other nanoformulation. This biomimetic ICD nanoinducer based on DS is expected to provide new strategies and references for the treatment of melanoma.
Collapse
|
94
|
Wang H, Shao R, Liu W, Peng S, Bai S, Fu B, Zhao C, Lu Y. Integrative analysis identifies CXCL11 as an immune-related prognostic biomarker correlated with cell proliferation and immune infiltration in multiple myeloma microenvironment. Cancer Cell Int 2022; 22:187. [PMID: 35568859 PMCID: PMC9107742 DOI: 10.1186/s12935-022-02608-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/02/2022] [Indexed: 02/07/2023] Open
Abstract
Purpose The interaction between tumor cells and tumor microenvironment (TME) has an important impact on progression and prognosis of multiple myeloma (MM), and has been proven to be promising therapeutic targets. This study intended to explore the relationship between TME and prognosis and identify valuable biomarkers of MM. Methods The transcriptomic and clinical information of MM retrieved from the Gene Expression Omnibus (GEO) were used to establish the model. The curve of Kaplan–Meier survival and the time-dependent receiver operating characteristic (ROC) were used to appraise the predictive ability. A nomogram was established for clinical application. Furthermore, the CIBERSORT algorithm was used to investigate the relation between IRGPI with the infiltration of immune cells. We also used histology, as well as in vitro and in vivo experiments to validate these findings. Results The results demonstrated an immune-related gene-based prognostic index (IRGPI) combined with clinical information. Patients were separated into high- and low-risk groups based on risk score, which had significantly difference in survival status and immune infiltrations. Furthermore, we identified CXCL11 as a key factor, which positively promotes the progression of MM and correlate with macrophage M2-like polarization and tumor immune cells infiltration. Conclusion Our findings suggest the IRGPI significantly demonstrate the differential prognosis and prediction of immune cells infiltration. It provides some insights into the complex interaction between myeloma tumor cells and the TME, as well as in the development of a novel biomarker target for anti-MM therapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02608-9.
Collapse
Affiliation(s)
- Huizhong Wang
- Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China.,State Key Laboratory of Oncology in South China, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China.,Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
| | - Ruonan Shao
- Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China.,State Key Laboratory of Oncology in South China, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China.,Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
| | - Wenjian Liu
- Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China.,State Key Laboratory of Oncology in South China, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China.,Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
| | - Shumei Peng
- Department of Pediatrics, Guangdong Women and Children Hospital, Guangzhou, 510060, China
| | - Shenrui Bai
- Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China.,State Key Laboratory of Oncology in South China, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China.,Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
| | - Bibo Fu
- Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China.,State Key Laboratory of Oncology in South China, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China.,Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
| | - Congling Zhao
- Department of Pediatrics, Guangdong Women and Children Hospital, Guangzhou, 510060, China.
| | - Yue Lu
- Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China. .,State Key Laboratory of Oncology in South China, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China. .,Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China.
| |
Collapse
|
95
|
Bettano K, Zielstorff M, Sevilla R, Yang R, Zhou H, Rosahl T, Zhang-Hoover J, Moy LY, Zhang W. A bioluminescence reporter mouse model for visualizing and quantifying CD8+ T cells in vivo. Neoplasia 2022; 27:100781. [PMID: 35381456 PMCID: PMC8980487 DOI: 10.1016/j.neo.2022.100781] [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: 11/12/2021] [Accepted: 02/24/2022] [Indexed: 11/17/2022]
Abstract
Cytotoxic CD8+ T cells are the primary effector cells mediating anti-tumor responses. In vivo monitoring of CD8+ T cells has broad implications for the development of novel cancer therapies. Here we describe the development of a genetically engineered mouse model (GEMM) in which CD8+ T cells are labeled with an optical reporter, enabling in vivo, longitudinal monitoring using bioluminescence imaging (BLI). Firefly luciferase (Luc2), human diphtheria toxin receptor (DTR), and enhanced green fluorescence protein (eGFP) cDNAs are engineered under the CD8α promoter to generate a transgenic mouse line. Luciferase mRNA and CD8α mRNA were generally correlated in various tissues from these mice. Sorted splenic CD8+ T cells, CD4+ T cells and CD3- non-T cells verified that the luciferase signal is specific to CD8+ T cells. In vivo imaging showed that luciferase signal was detected in various immune organs, such as lymph nodes, thymus, and spleen, and the detection was confirmed by ex vivo examination. Administration of diphtheria toxin markedly reduced luciferase signal systemically, confirming the function of the DTR. In the MC38 mouse syngeneic model, we observed significant increases in CD8+ T cells with mDX400 treatment, an anti PD-1 mouse monoclonal antibody that correlated with tumor growth inhibition. This novel reporter GEMM is a valuable drug discovery tool for profiling compounds and understanding mechanisms of action in immunotherapy of cancer.
Collapse
|
96
|
Synthesis, characterization, and evaluation of chloroaluminium phthalocyanine incorporated in poly(ε-caprolactone) nanoparticles for photodynamic therapy. Photodiagnosis Photodyn Ther 2022; 38:102850. [PMID: 35395414 DOI: 10.1016/j.pdpdt.2022.102850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/21/2022] [Accepted: 04/03/2022] [Indexed: 01/08/2023]
Abstract
BACKGROUND The use of nanotechnology has been widely used in biomedical science, which consists of orthopedic implants, tissue engineering, cancer therapy and drug elution from nanoparticle systems, such as poly-caprolactone (PCL) nanoparticles, which stand out mainly for their biocompatibility, being considered as effective carriers for photosensitizing drugs (PS) in photodynamic therapy (PDT) protocols. METHODS This manuscript describes the synthesis and characterization of PCL nanoparticles for controlled release of the drug chloro-aluminum phthalocyanine (ClAlPc) as a photosensitizer for application in PDT. The PCL-ClAlPc nanoparticles were developed by the nanoprecipitation process. The structure and morphology of the nanoparticles were studied with scanning electron microscopy (SEM) and with Fourier transform infrared (FTIR). The size of nanomaterials was studied using the Dynamic Light Scattering (DLS) method. Photophysical and photochemical characterizations were performed. Subsequently, photobiological studies were also used to characterize the system. RESULTS The nanoparticles had an average diameter of 384.7 ± 138.6 nm and a polydispersity index of 0.153. SEM analysis revealed that the system formed a spherical shape typical of these delivery systems. Charging efficiency was 82.1% ± 1.2%. The phthalocyanine-loaded PCL nanoparticles maintained their photophysical behavior after encapsulation. Cell viability was determined after the dark toxicity test, and it was possible to observe that there was no evidence of toxicity in the dark, for all concentrations tested. The assay also revealed that adenocarcinoma cells treated with free ClAlPc and in the nanoformulation showed 100% cell death when subjected to PDT protocols. The intracellular location of the photosensitizer indicated a high potential for accumulation in the cytoplasm and nucleus. CONCLUSIONS From the photophysical, photochemical and photobiological analyzes obtained, it was possible to observe that the development of PCL nanoparticles encapsulated with ClAlPc, by the nanoprecipitation method was adequate and that the in vivo release study is efficient to reduce the release rate and attenuate the burst of PS loaded on PCL nanoparticles. The results reinforce that the use of this system as drug delivery systems is useful in PDT protocols.
Collapse
|
97
|
Xue Y, Ning B, Liu H, Jia B. Construction of immune-related lncRNA signature to predict aggressiveness, immune landscape, and drug resistance of colon cancer. BMC Gastroenterol 2022; 22:127. [PMID: 35300596 PMCID: PMC8928673 DOI: 10.1186/s12876-022-02200-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 02/24/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Colon cancer remains one of the most common malignancies across the world. Thus far, a biomarker, which can comprehensively predict the survival outcomes, clinical characteristics, and therapeutic sensitivity, is still lacking. METHODS We leveraged transcriptomic data of colon cancer from the existing datasets and constructed immune-related lncRNA (irlncRNA) pairs. After integrating with clinical survival data, we performed differential analysis and identified 11 irlncRNAs signature using Lasso regression analysis. We next plotted the 1-, 5-, and 10-year curve lines of receiver operating characteristics, calculated the areas under the curve, and recognized the optimal cutoff point. Then, we validated the pair-risk model in terms of the survival outcomes of the patients involved. Moreover, we tested the reliability of the model for predicting tumor aggressiveness and therapeutic susceptibility of colon cancer. Additionally, we reemployed the 11 of irlncRNAs involved in the pair-risk model to construct an expression-risk model to predict the prognostic outcomes of the patients involved. RESULTS We recognized a total of 377 differentially expressed irlncRNAs (DEirlcRNAs), including 28 low-expressed and 349 high-expressed irlncRNAs in colon cancer patients. After performing a univariant Cox analysis, we identified 115 risk irlncRNAs that were significantly correlated with survival outcomes of patients involved. By taking the overlap of the DEirlcRNAs and the risk irlncRNAs, we ultimately recognized 55 irlncRNAs as core irlncRNAs. Then, we established a Cox HR model (pair-risk model) as well as an expression HR model (exp-risk model) based on 11 of the 55 core irlncRNAs. We found that both of the two models significantly outperformed the commonly used clinical characteristics, including age, T, N, and M stages when predicting survival outcomes. Moreover, we validated the pair-risk model as a potential tool for studying the tumor microenvironment of colon cancer and drug susceptibility. Additionally, we noticed that combinational use of the pair-risk model and the exp-risk model yielded a more robust approach for predicting the survival outcomes of patients with colon cancer. CONCLUSIONS We recognized 11 irlncRNAs and created a pair-risk model and an exp-risk model, which have the potential to predict clinical characteristics of colon cancer, either solely or conjointly.
Collapse
Affiliation(s)
- Yonggan Xue
- Department of General Surgery, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Bobin Ning
- Department of General Surgery, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Hongyi Liu
- Department of General Surgery, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China
| | - Baoqing Jia
- Department of General Surgery, Chinese PLA General Hospital, No. 28, Fuxing Road, Haidian District, Beijing, 100853, People's Republic of China.
| |
Collapse
|
98
|
Pan-Cancer Analysis of Microfibrillar-Associated Protein 2 (MFAP2) Based on Bioinformatics and qPCR Verification. JOURNAL OF ONCOLOGY 2022; 2022:8423173. [PMID: 35211173 PMCID: PMC8863482 DOI: 10.1155/2022/8423173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 12/24/2021] [Accepted: 12/30/2021] [Indexed: 11/18/2022]
Abstract
MFAP2 has been reported to play an oncogenic role in several types of human cancers. However, the expression profile of MFAP2 in various cancers and its impact on prognosis and immune infiltration remain unclear. In this study, the mRNA expression and protein expression of MFAP2 in normal tissues, tumor cell lines, and 33 malignant tumor tissues were analyzed comprehensively using Genotype-Tissue Expression (GTEx), Cancer Cell Line Encyclopedia (CCLE), and The Cancer Genome Atlas (TCGA), Oncomine and UALCAN databases, and the expression of MFAP2 in different grades and stages of cancers was assessed using Gene Expression Profiling Interactive Analysis 2 (GEPIA2) and Tumor and Immune System Interaction Database (TISIDB). In general, MFAP2 showed distinct expression in most tumor and normal tissues, closely associated with higher tumor grade, higher tumor stage, and poor survival in multiple cancers. A search of the UALCAN database and the cBioPortal database revealed that this difference in mRNA level expression could be partly attributed to abnormal DNA methylation and mutations at the genomic level. In addition, MFAP2 expression was also associated with tumor mutation burden, microsatellite instability, and neoantigens in different cancer types. More importantly, the TIMER and TISIDB databases also showed that MFAP2 levels were significantly correlated with immune infiltration abundance and immune-related gene markers, as well as ESTIMATE scores. By qPCR, MFAP2 expression was validated in four kinds of tumor tissue samples. The present study combined several databases and performed a pan-cancer analysis of the expression profile, methylation, and mutation for MFAP2 and its implications for prognosis and immune infiltration, suggesting that MFAP2 could contribute to malignant properties of many tumors. MFAP2 may be an important biomarker with prognostic value and has the potential to be a target for tumor immunotherapy.
Collapse
|
99
|
Shi L, Cao J, Lei X, Shi Y, Wu L. Multi-omics data identified TP53 and LRP1B as key regulatory gene related to immune phenotypes via EPCAM in HCC. Cancer Med 2022; 11:2145-2158. [PMID: 35150083 PMCID: PMC9119357 DOI: 10.1002/cam4.4594] [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: 03/19/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 12/16/2022] Open
Abstract
Background Many studies showed that the prognosis of hepatocellular carcinoma (HCC) was significantly associated with the expressions of TP53 and LRP1B. However, the potential influence of the two genes on the malignant progression of HCC is still to be expounded. Methods According to the correlation analysis between immune cells and expression levels of TP53 and LRP1B, we filtered the immune cells to perform unsupervised clustering analysis. Integration of multi‐omic data analysis identified genetic alteration and epigenetic alteration. In addition, pathway analysis was used to explore the potential function of the differentially expressed mRNAs. According to the differentially expressed genes, we established an interaction network to seek the hub gene. Least absolute shrinkage and selection operator (LASSO) regression analysis was used to build a prognosis model. Results The unsupervised clustering analysis showed that the cluster A1 showed the highest immune cell levels and the cluster B2 showed the lowest immune cell levels. Multi‐omics data analysis identified that somatic mutations, copy number variations, and DNA methylation levels had significant differences between cluster A1 and cluster B2. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis found that the upregulated mRNAs in the cluster A1 were mainly concentrated in T cell activation, external side of plasma membrane, receptor ligand activity, and cytokine−cytokine receptor interaction. Importantly, the EPCAM was identified as a critical node in the lncRNAs–miRNAs–mRNAs regulatory network correlated with the immune phenotypes. In addition, based on differentially expressed genes between cluster A1 and cluster B2, the prognostic model established by LASSO could predict the overall survival (OS) of HCC accurately. Conclusions The results indicated that the TP53 and LRP1B acted as the key genes in regulating the immune phenotypes of HCC via EPCAM.
Collapse
Affiliation(s)
- Liang Shi
- Department of Clinical Laboratory Medicine, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jie Cao
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xin Lei
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yifen Shi
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lili Wu
- Department of Clinical Blood Transfusion, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Department of Clinical Laboratory, The Central Hospital of Wenzhou, Wenzhou, China
| |
Collapse
|
100
|
Dual-RNA controlled delivery system inhibited tumor growth by apoptosis induction and TME activation. J Control Release 2022; 344:97-112. [DOI: 10.1016/j.jconrel.2022.02.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 02/13/2022] [Accepted: 02/15/2022] [Indexed: 12/27/2022]
|