1
|
Guo X, Bai J, Wang X, Guo S, Shang Z, Shao Z. Evoking the Cancer-immunity cycle by targeting the tumor-specific antigens in Cancer immunotherapy. Int Immunopharmacol 2025; 154:114576. [PMID: 40168803 DOI: 10.1016/j.intimp.2025.114576] [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: 02/08/2025] [Revised: 03/17/2025] [Accepted: 03/27/2025] [Indexed: 04/03/2025]
Abstract
Cancer-related deaths continue to rise, largely due to the suboptimal efficacy of current treatments. Fortunately, immunotherapy has emerged as a promising alternative, offering new hope for cancer patients. Among various immunotherapy approaches, targeting tumor-specific antigens (TSAs) has gained particular attention due to its demonstrated success in clinical settings. Despite these advancements, there are still gaps in our understanding of TSAs. Therefore, this review explores the life cycle of TSAs in cancer, the methods used to identify them, and recent advances in TSAs-targeted cancer therapies. Enhancing medical professionals' understanding of TSAs will help facilitate the development of more effective TSAs-based cancer treatments.
Collapse
Affiliation(s)
- Xiaomeng Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Junqiang Bai
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Xinmiao Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Shutian Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Department of Oral and Maxillofacial-Head and Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Zhe Shao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China; Day Surgery Center, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
2
|
Zhang Y, Yan L, Sun H, Zhang Z, Shen F, Sun L. Targeted Delivery of Personalized Cancer Vaccines Based on Antibody-Antigen Complexes. Vaccines (Basel) 2025; 13:324. [PMID: 40266219 PMCID: PMC11946472 DOI: 10.3390/vaccines13030324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/15/2025] [Accepted: 03/16/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Personalized cancer vaccines based on tumor neoantigens show great potential in cancer immunotherapy due to their high safety and specificity. However, it is inherently difficult to realize the efficiently targeted delivery of personalized cancer vaccines to antigen-presenting cells (APCs). METHODS This study aimed to address these challenges by developing and evaluating a personalized cancer vaccine based on antibody-antigen complexes, which was designed to enhance antitumor effects by increasing the utilization of tumor neoantigens by APCs. Mice were immunized with a carrier protein, keyhole limpet hemocyanin (KLH), to induce the production of antibodies against KLH. Subsequently, mice were immunized with KLH loaded with tumor neoantigens and the immunoadjuvant CpG ODN and underwent immunological analysis to evaluate the immune and antitumor effects. RESULTS The results showed that preimmunization with KLH could promote the uptake of the personalized KLH-based tumor vaccine, which was enhanced by dendritic cells (DCs) and macrophages (Mφs), by strengthening the T-cell immune responses to tumors. CONCLUSIONS Collectively, this work provides a new idea for the targeted delivery of personalized cancer vaccines.
Collapse
Affiliation(s)
- Yaling Zhang
- School of Life Sciences, Shanghai University, Shanghai 200444, China (L.Y.)
| | - Lingling Yan
- School of Life Sciences, Shanghai University, Shanghai 200444, China (L.Y.)
| | - He Sun
- School of Life Sciences, Shanghai University, Shanghai 200444, China (L.Y.)
| | - Ziyi Zhang
- School of Life Sciences, Shanghai University, Shanghai 200444, China (L.Y.)
| | - Fengyun Shen
- Institute of Materiobiology, Department of Chemistry, College of Science, Shanghai University, Shanghai 200444, China
| | - Lele Sun
- Institute of Materiobiology, Department of Chemistry, College of Science, Shanghai University, Shanghai 200444, China
| |
Collapse
|
3
|
Rosenberg-Mogilevsky A, Siegfried Z, Karni R. Generation of tumor neoantigens by RNA splicing perturbation. Trends Cancer 2025; 11:12-24. [PMID: 39578174 DOI: 10.1016/j.trecan.2024.10.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/16/2024] [Accepted: 10/23/2024] [Indexed: 11/24/2024]
Abstract
Immunotherapy has revolutionized cancer treatment, but the limited availability of tumor-specific neoantigens still remains a challenge. The potential of alternative mRNA splicing-derived neoantigens as a source of new immunotherapy targets has gained significant attention. Tumors exhibit unique splicing changes and splicing factor mutations which are prevalent in various cancers and play a crucial role in neoantigen production. We present advances in splicing modulation approaches, including small-molecule drugs, decoy and splice-switching antisense oligonucleotides (SSOs), CRISPR, small interfering RNAs (siRNAs), and nonsense-mediated RNA decay (NMD) inhibition, that can be adapted to enhance antitumor immune responses. Finally, we explore the clinical implications of these approaches, highlighting their potential to transform cancer immunotherapy and broaden its efficacy.
Collapse
Affiliation(s)
- Adi Rosenberg-Mogilevsky
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University and Hadassah Medical School, Jerusalem, Israel
| | - Zahava Siegfried
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University and Hadassah Medical School, Jerusalem, Israel
| | - Rotem Karni
- Department of Biochemistry and Molecular Biology, Institute for Medical Research Israel-Canada (IMRIC), Hebrew University and Hadassah Medical School, Jerusalem, Israel.
| |
Collapse
|
4
|
Huang Z, Han Z, Zheng K, Zhang Y, Liang Y, Zhu X, Zhou J. Development and application of a predictive model for survival and drug therapy based on COVID-19-related lncRNAs in non-small cell lung cancer. Medicine (Baltimore) 2024; 103:e40629. [PMID: 39654255 PMCID: PMC11631024 DOI: 10.1097/md.0000000000040629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 12/29/2023] [Accepted: 11/04/2024] [Indexed: 12/12/2024] Open
Abstract
Numerous studies have substantiated the pivotal role of long non-coding RNAs (lncRNAs) in the progression of non-small cell lung cancer (NSCLC) and the prognosis of afflicted patients. Notably, individuals with NSCLC may exhibit heightened vulnerability to the novel coronavirus disease (COVID-19), resulting in a more unfavorable prognosis subsequent to infection. Nevertheless, the impact of COVID-19-related lncRNAs on NSCLC remains unexplored. The aim of our study was to develop an innovative model that leverages COVID-19-related lncRNAs to optimize the prognosis of NSCLC patients. Pertinent genes and patient data were procured from reputable databases, including TCGA, Finngen, and RGD. Through co-expression analysis, we identified lncRNAs associated with COVID-19. Subsequently, we employed univariate, LASSO, and multivariate COX regression techniques to construct a risk model based on these COVID-19-related lncRNAs. The validity of the risk model was assessed using KM analysis, PCA, and ROC. Furthermore, functional enrichment analysis was conducted to elucidate the functional pathways linked to the identified lncRNAs. Lastly, we performed TME analysis and predicted the drug sensitivity of the model. Based on risk scores, patients were categorized into high- and low-risk subgroups, revealing distinct clinicopathological factors, immune pathways, and chemotherapy sensitivity between the subgroups. Four COVID-19-related lncRNAs (AL161431.1, AC079949.1, AC123595.1, and AC108136.1) were identified as potential candidates for constructing prognostic prediction models for NSCLC. We also observed a positive correlation between risk score and MDSC, exclusion, and CAF. Additionally, two immune pathways associated with high-risk and low-risk subgroups were identified. Our findings further support the association between COVID-19 infection and neuroactive ligand-receptor interaction, as well as steroid metabolism in NSCLC. Moreover, we identified several highly sensitive chemotherapy drugs for NSCLC treatment. The developed model holds significant value in predicting the prognosis of NSCLC patients and guiding treatment decisions.
Collapse
Affiliation(s)
- Ziyuan Huang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Zenglei Han
- Department of Pathology, Qingdao Municipal Hospital, Qingdao, China
| | - Kairong Zheng
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Yidan Zhang
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Yanjun Liang
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Xiao Zhu
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China
| | - Jiajun Zhou
- Department of Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
| |
Collapse
|
5
|
Ji S, Fang H, Guan J, He K, Yang Q. Immunoproteomics Reveal Different Characteristics for the Prognostic Markers of Intratumoral-Infiltrating CD3+ T Lymphocytes and Immunoscore in Colorectal Cancer. J Transl Med 2024; 104:102159. [PMID: 39419351 DOI: 10.1016/j.labinv.2024.102159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/26/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) and immunoscoring based on densities of CD3+ and CD8+ TILs are both favorable prognostic markers in colorectal cancer (CRC). However, determination of the molecular features of TILs, particularly their immunoproteomic signatures would require the development of large scale in situ spatiotemporal technologies. Recently, a multiplex in situ digital spatial proteomic profiling (DSP) tool GeoMx DSP has been applied to identify biomarkers predictive of therapeutic responses and to understand disease mechanisms and progression. Taking advantage of this tool, we simultaneously characterized the spatial distribution and interactions of 42 immune proteins in tumor cells (TCs), CD3+ T stromal TILs (sTILs), and CD20+ B sTILs using tissue microarrays, and further studied their associations with CD3+ T TILs and immunoscores in CRC. First, our data showed that well-known immune checkpoints, such as PD-L1, PD-L2, and LAG3, were expressed at low levels, whereas some other immune proteins, such as CD11c, CD68, STING, and CD44, were highly expressed. Second, 8 spatial interactions were identified, including 5 interactions between TC and CD20+ B sTILs, 2 interactions between CD3+ T sTILs and CD20+ B sTILs, and 1 interaction among TC, CD3+ T sTILs, and CD20+ B sTILs. Third, the differential immune microlandscape in the spatial compartments was identified in tissues with positive CD3+ T intratumoral TILs and high immunoscores. Collectively, to our knowledge, our study is the first to provide in situ spatial immune characteristics at the proteomic level. Moreover, our findings provide direct evidence supporting the infiltration of CD3+ T sTILs from stoma to TC and shed important insights into better understanding and treating CRC patients related to different immune prognostic markers.
Collapse
Affiliation(s)
- Saiyan Ji
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huanying Fang
- Department of Clinical Laboratory, Shanghai Yangpu District Mental Health Center, Shanghai, China
| | - Jingjie Guan
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun He
- Medical Research and Laboratory Diagnostic Center, Central Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Qingyuan Yang
- Department of Pathology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
6
|
Goswami A, Goyal S, Khurana P, Singh K, Deb B, Kulkarni A. Small molecule innate immune modulators in cancer therapy. Front Immunol 2024; 15:1395655. [PMID: 39318624 PMCID: PMC11419979 DOI: 10.3389/fimmu.2024.1395655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/21/2024] [Indexed: 09/26/2024] Open
Abstract
Immunotherapy has proved to be a breakthrough in cancer treatment. So far, a bulk of the approved/late-stage cancer immunotherapy are antibody-based. Although these antibody-based drugs have demonstrated great promise, a majority of them are limited due to their access to extracellular targets, lack of oral bioavailability, tumor microenvironment penetration, induction of antibody dependent cytotoxicity etc. In recent times, there has been an increased research focus on the development of small molecule immunomodulators since they have the potential to overcome the aforementioned limitations posed by antibodies. Furthermore, while most biologics based therapeutics that are in clinical use are limited to modulating the adaptive immune system, very few clinically approved therapeutic modalities exist that modulate the innate immune system. The innate immune system, which is the body's first line of defense, has the ability to turn cold tumors hot and synergize strongly with existing adaptive immune modulators. In preclinical studies, small molecule innate immune modulators have demonstrated synergistic efficacy as combination modalities with current standard-of-care immune checkpoint antibodies. In this review, we highlight the recent advances made by small molecule innate immunomodulators in cancer immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | - Barnali Deb
- Aten Porus Lifesciences Pvt. Ltd., Bengaluru, India
| | - Aditya Kulkarni
- Aten Porus Lifesciences Pvt. Ltd., Bengaluru, India
- Avammune Therapeutics, Philadelphia, PA, United States
| |
Collapse
|
7
|
Lin S, Hong J, Wu S, Zhu C, Liu F, Lin W, Cai X, Ye Y, Chen Y. BCL2A1 neoepitope-elicited cytotoxic T lymphocytes are a promising individualized immunotherapy of pancreatic cancer. J Leukoc Biol 2024; 116:601-610. [PMID: 38626292 DOI: 10.1093/jleuko/qiae092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 03/16/2024] [Accepted: 04/01/2024] [Indexed: 04/18/2024] Open
Abstract
Conventional treatments have shown a limited efficacy for pancreatic cancer, and immunotherapy is an emerging option for treatment of this highly fatal malignancy. Neoantigen is critical to improving the efficacy of tumor-specific immunotherapy. The cancer and peripheral blood specimens from an HLA-A0201-positive pancreatic cancer patient were subjected to next-generation sequencing, and bioinformatics analyses were performed to screen high-affinity and highly stable neoepitopes. The activation of cytotoxic T lymphocytes (CTLs) by dendritic cells (DCs) loaded with mutBCL2A111-20 neoepitope targeting a BCL2A1 mutant epitope was investigated, and the cytotoxicity of mutBCL2A111-20 neoepitope-specific CTLs to pancreatic cancer cells was evaluated. The mutBCL2A111-20 neoepitope was found to present a high immunogenicity and induce CTLs activation and proliferation, and these CTLs were cytotoxic to mutBCL2A111-20 neoepitope-loaded T2 cells and pancreatic cancer PANC-1-Neo and A2-BxPC-3-Neo cells that overexpressed mutBCL2A111-20 neoepitopes, appearing to be a targeting neoepitope specificity. In addition, high BCL2A1 expression correlated with a low 5-yr progression-free interval among pancreatic cancer patients. Our findings provide experimental supports to individualized T cell therapy targeting mutBCL2A111-20 neoepitopes, and provide an option of immunotherapy for pancreatic cancer.
Collapse
Affiliation(s)
- Shengzhe Lin
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, No. 29, Xinquan Road, Gulou District, Fuzhou, Fujian, 350001, China
| | - Jingwen Hong
- School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, University Town, Fuzhou, Fujian, 350122, China
| | - Suxin Wu
- School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, University Town, Fuzhou, Fujian, 350122, China
| | - Chenlu Zhu
- School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, University Town, Fuzhou, Fujian, 350122, China
| | - Fang Liu
- Laboratory of Immuno-Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No. 420, Fuma Road, Jinan District, Fuzhou, Fujian, 350014, China
- Fujian Key Laboratory of Translational Cancer Medicine, No. 420, Fuma Road, Jinan District, Fuzhou, Fujian, 350014, China
| | - Wansong Lin
- School of Basic Medical Sciences, Fujian Medical University, No. 1, Xuefu North Road, University Town, Fuzhou, Fujian, 350122, China
- Laboratory of Immuno-Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No. 420, Fuma Road, Jinan District, Fuzhou, Fujian, 350014, China
- Fujian Key Laboratory of Translational Cancer Medicine, No. 420, Fuma Road, Jinan District, Fuzhou, Fujian, 350014, China
| | - Xinran Cai
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, No. 29, Xinquan Road, Gulou District, Fuzhou, Fujian, 350001, China
| | - Yunbin Ye
- Laboratory of Immuno-Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No. 420, Fuma Road, Jinan District, Fuzhou, Fujian, 350014, China
- Fujian Key Laboratory of Translational Cancer Medicine, No. 420, Fuma Road, Jinan District, Fuzhou, Fujian, 350014, China
| | - Yanling Chen
- Department of Hepatobiliary Surgery and Fujian Institute of Hepatobiliary Surgery, Fujian Medical University Union Hospital, No. 29, Xinquan Road, Gulou District, Fuzhou, Fujian, 350001, China
| |
Collapse
|
8
|
Shan H, Wang X, Yin F, Zhou Y, Mao L, Zhu X, Liu C. Combination of transcriptome and Mendelian inheritance reveals novel prognostic biomarker of CTLA-4-related lncRNAs and protective role of nitrogen metabolism pathway in lung adenocarcinoma development. BMC Cancer 2024; 24:1009. [PMID: 39143529 PMCID: PMC11323378 DOI: 10.1186/s12885-024-12777-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024] Open
Abstract
OBJECTIVE Since in the cancer setting, tumor cells may use cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) to evade the immune system. This study aimed to identify CTLA-4-related long non-coding RNAs (lncRNAs) and assess their roles in lung adenocarcinoma (LUAD) development. METHODS Clinical and genomic data were obtained from The Cancer Genome Atlas (TCGA), MSigDB and Gene Weaver. CTLA-4-related lncRNA-based gene signatures (CTLA4LncSigs) were identified using Cox regression, establishing a risk score model and an independent prognostic model. Enrichment analysis (GO/KEGG) was performed. Mendelian randomization (MR) analysis investigated the nitrogen metabolism and lung cancer relationship, with Bayesian weighted MR (BWMR) addressing uncertainties. Correlations with tumor microenvironment and drug sensitivity were explored. RESULTS Nineteen CTLA4LncSigs significantly influenced LUAD prognosis. The risk score demonstrated independence as a prognostic factor. Functional analysis revealed lncRNAs' impact on nitrogen metabolism. MR and BWMR confirmed the protective role of the nitrogen metabolism pathway in lung cancer. CONCLUSION Our study identifies CTLA-4-related lncRNAs associated with LUAD prognosis and uncovers a previously undiscovered protective role of the nitrogen metabolism pathway in combating LUAD development, providing new insights into potential therapeutic targets and prognostic biomarkers for this aggressive cancer subtype.
Collapse
Affiliation(s)
- Huisi Shan
- Department of Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Department of Radiation Oncology, Guangdong Second People's Hospital, Jinan University, Guangzhou, China
| | - Xiaocong Wang
- Department of Pathology, Qingdao Municipal Hospital Group, Qingdao, China
| | - Fei Yin
- Department of Clinical Laboratory, Qingdao Sixth People's Hospital, Qingdao, China
| | - Yiting Zhou
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
- Department of Internal Medicine, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Liuhan Mao
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China
| | - Xiao Zhu
- The Second Affiliated Hospital, Guangdong Medical University, Zhanjiang, China.
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, School of Ocean and Tropical Medicine, Guangdong Medical University, Zhanjiang, China.
| | - Caixin Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China.
| |
Collapse
|
9
|
Yang M, Cui M, Sun Y, Liu S, Jiang W. Mechanisms, combination therapy, and biomarkers in cancer immunotherapy resistance. Cell Commun Signal 2024; 22:338. [PMID: 38898505 PMCID: PMC11186190 DOI: 10.1186/s12964-024-01711-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024] Open
Abstract
Anti-programmed death 1/programmed death ligand 1 (anti-PD-1/PD-L1) antibodies exert significant antitumor effects by overcoming tumor cell immune evasion and reversing T-cell exhaustion. However, the emergence of drug resistance causes most patients to respond poorly to these immune checkpoint inhibitors (ICIs). Studies have shown that insufficient T-cell infiltration, lack of PD-1 expression, deficient interferon signaling, loss of tumor antigen presentation, and abnormal lipid metabolism are all considered to be closely associated with immunotherapy resistance. To address drug resistance in tumor immunotherapy, a lot of research has concentrated on developing combination therapy strategies. Currently, ICIs such as anti-PD-1 /PD-L1 antibody combined with chemotherapy and targeted therapy have been approved for clinical treatment. In this review, we analyze the mechanisms of resistance to anti-PD-1/PD-L1 therapy in terms of the tumor microenvironment, gut microbiota, epigenetic regulation, and co-inhibitory immune checkpoint receptors. We also discuss various promising combination therapeutic strategies to address resistance to anti-PD-1/PD-L1 drugs, including combining these therapies with traditional Chinese medicine, non-coding RNAs, targeted therapy, other ICIs, and personalized cancer vaccines. Moreover, we focus on biomarkers that predict resistance to anti-PD-1/PD-L1 therapy as well as combination therapy efficacy. Finally, we suggest ways to further expand the application of immunotherapy through personalized combination strategies using biomarker systems.
Collapse
Affiliation(s)
- Manshi Yang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Mengying Cui
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Yang Sun
- Department of Orthopaedic, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Shui Liu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, 130041, China
| | - Weibo Jiang
- Department of Orthopaedic, The Second Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
10
|
Subbarayan R, Srinivasan D, Balakrishnan R, Kumar A, Usmani SS, Srivastava N. DNA damage response and neoantigens: A favorable target for triple-negative breast cancer immunotherapy and vaccine development. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 389:104-152. [PMID: 39396845 DOI: 10.1016/bs.ircmb.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Triple-negative breast cancer (TNBC) poses a significant clinical challenge due to its aggressive nature and limited therapeutic options. The interplay between DNA damage response (DDR) mechanisms and the emergence of neoantigens represents a promising avenue for developing targeted immunotherapeutic strategies and vaccines for TNBC. The DDR is a complex network of cellular mechanisms designed to maintain genomic integrity. In TNBC, where genetic instability is a hallmark, dysregulation of DDR components plays a pivotal role in tumorigenesis and progression. This review explores the intricate relationship between DDR and neoantigens, shedding light on the potential vulnerabilities of TNBC cells. Neoantigens, arising from somatic mutations in cancer cells, represent unique antigens that can be recognized by the immune system. TNBC's propensity for genomic instability leads to an increased mutational burden, consequently yielding a rich repertoire of neoantigens. The convergence of DDR and neoantigens in TNBC offers a distinctive opportunity for immunotherapeutic targeting. Immunotherapy has revolutionized cancer treatment by harnessing the immune system to selectively target cancer cells. The unique immunogenicity conferred by DDR-related neoantigens in TNBC positions them as ideal targets for immunotherapeutic interventions. This review also explores various immunotherapeutic modalities, including immune checkpoint inhibitors (ICIs), adoptive cell therapies, and cancer vaccines, that leverage the DDR and neoantigen interplay to enhance anti-tumor immune responses. Moreover, the potential for developing vaccines targeting DDR-related neoantigens opens new frontiers in preventive and therapeutic strategies for TNBC. The rational design of vaccines tailored to the individual mutational landscape of TNBC holds promise for precision medicine approaches. In conclusion, the convergence of DDR and neoantigens in TNBC presents a compelling rationale for the development of innovative immunotherapies and vaccines. Understanding and targeting these interconnected processes may pave the way for personalized and effective interventions, offering new hope for patients grappling with the challenges posed by TNBCs.
Collapse
Affiliation(s)
- Rajasekaran Subbarayan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, FAHS, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Dhasarathdev Srinivasan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, FAHS, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Ranjith Balakrishnan
- Centre for Advanced Biotherapeutics and Regenerative Medicine, FAHS, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, India
| | - Ajeet Kumar
- Department of Psychiatry, Washington university School of Medicine, St louis, MO, United States
| | - Salman Sadullah Usmani
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States.
| | - Nityanand Srivastava
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, United States.
| |
Collapse
|
11
|
Jiani W, Qin T, Jie M. Tumor neoantigens and tumor immunotherapies. Aging Med (Milton) 2024; 7:224-230. [PMID: 38725698 PMCID: PMC11077340 DOI: 10.1002/agm2.12295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/01/2024] [Accepted: 03/28/2024] [Indexed: 05/12/2024] Open
Abstract
As a high-risk group of patients with cancer, the elderly exhibit limited efficacy with traditional treatments. Immunotherapy emerges as a promising adjunctive therapeutic approach that holds potential in addressing the needs of geriatric patients with cancer. Neoantigens, a unique class of tumor-specific antigens generated by non-synonymous mutations, are garnering increasing attention as targets for immunotherapy in clinical applications. Newly developed technologies, such as second-generation gene sequencing and mass spectrometry, have provided powerful technical support for the identification and prediction of neoantigens. At present, neoantigen-based immunotherapy has been extensively applied in clinical trials and has demonstrated both safety and efficacy, marking the beginning of a new era for cancer immunotherapy. This article reviews the conception, classification, inducers, and screening process of tumor neoantigens, as well as the application prospects and combination therapy strategies of neoantigen-based cancer immunotherapy.
Collapse
Affiliation(s)
- Wang Jiani
- Department of Biotherapy Center, Beijing Hospital, National Center of GerontologyInstitute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| | - Tan Qin
- Department of Biotherapy Center, Beijing Hospital, National Center of GerontologyInstitute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| | - Ma Jie
- Department of Biotherapy Center, Beijing Hospital, National Center of GerontologyInstitute of Geriatric Medicine, Chinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
12
|
Xiang N, Zhang K, Zhao Y, Xu C, Zhang X, Meng S. Characterization of antigen presentation capability for neoantigen-based products using targeted LC-MS/MS method. J Pharm Biomed Anal 2024; 240:115886. [PMID: 38184916 DOI: 10.1016/j.jpba.2023.115886] [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: 09/27/2023] [Revised: 11/15/2023] [Accepted: 11/26/2023] [Indexed: 01/09/2024]
Abstract
The generation of an immune response in neoantigen-based products relies on antigen presentation, which is closely analyzed by bioassays for T-cell functions such as tetramer or cytokine release. Mass spectrometry (MS) has the potential to directly assess the antigen-presenting capability of antigen-presenting cells (APCs), offering advantages such as speed, multi-target analysis, robustness, and ease of transferability. However, it has not been used for quality control of these products due to challenges in sensitivity, including the number of cells and peptide diversity. In this study, we describe the development and validation of an improved targeted LC-MS/MS method with high sensitivity for characterizing antigen presentation, which could be applied in the quality control of neoantigen-based products. The parameters for the extraction were carefully optimized by different short peptides. Highly sensitive targeted triple quadrupole mass spectrometry combined with ultra-high performance liquid chromatography (UHPLC) was employed using a selective ion monitoring mode (Multiple Reaction Monitoring, MRM). Besides, we successfully implemented robust quality control peptides to ensure the reliability and consistency of this method, which proved invaluable for different APCs. With reference to the guidelines from ICH Q2 (R2), M10, as well as considering the specific attributes of the product itself, we validated the method for selectivity, specificity, sensitivity, limit of detection (LOD), recovery rate, matrix effect, repeatability, and application in dendritic cells (DCs) associated with neoantigen-based products. The validation process yields satisfactory results. Combining this approach with T cell assays will comprehensively assess cell product quality attributes from physicochemical and biological perspectives.
Collapse
Affiliation(s)
- Nan Xiang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; The Cell Collection and Research Center, National Institutes for Food and Drug Control, No. 31 Huatuo St., Daxing District, Beijing, China
| | - Kehua Zhang
- The Cell Collection and Research Center, National Institutes for Food and Drug Control, No. 31 Huatuo St., Daxing District, Beijing, China
| | - Yinghua Zhao
- SCIEX China, 5F, Building 1, No. 24 Jiuxianqiao Middle Road, Chaoyang District, Beijing, China
| | - Chongfeng Xu
- The Cell Collection and Research Center, National Institutes for Food and Drug Control, No. 31 Huatuo St., Daxing District, Beijing, China
| | - Xiuqing Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Shufang Meng
- The Cell Collection and Research Center, National Institutes for Food and Drug Control, No. 31 Huatuo St., Daxing District, Beijing, China.
| |
Collapse
|
13
|
He X, Fan K, Gong H, Huang M, Zeng Q, Huang J, Peng X, Lai P, Lu Y, Wang H. Mechanism study of cross presentation of exogenous antigen induced by cholera toxin-like chimeric protein. Vaccine 2024; 42:1549-1560. [PMID: 38320931 DOI: 10.1016/j.vaccine.2024.01.075] [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: 05/14/2023] [Revised: 12/09/2023] [Accepted: 01/23/2024] [Indexed: 02/08/2024]
Abstract
Tumor subunit vaccines have great potential in personalized cancer immunotherapy. They are usually administered with adjuvant owing to their low immunogenicity. Cholera toxin (CT) is a biological adjuvant with diverse biological functions and a long history of use. Our earlier study revealed that a CT-like chimeric protein co-delivered with murine granulocyte-macrophage colony stimulating factor (mGM-CSF) and prostate cancer antigen epitope could co-stimulate dendritic cells (DCs) and enhance cross presentation of tumor epitope. To further study the molecular mechanism of CT-like chimeric protein in cross presentation, major histocompatibility complex class I (MHC I)-restricted epitope 257-264 of ovalbumin (OVAT) was used as a model antigen peptide in this study. Recombinant A subunit and pentameric B subunit of CT protein were respectively genetically constructed and purified. Then both assembled into AB5 chimeric protein in vitro. Three different chimeric biomacromolecules containing mGM-CSF and OVAT were constructed according to the different fusion sites and whether the endoplasmic reticulum (ER) retention sequence was included. It was found that A2 domain and B subunit of CT were both available for loading epitopes and retaining GM1 affinity. The binding activity of GM1 was positively correlated with antigen endocytosis. Once internalized, DCs became mature and cross-presented antigen. KDEL helped the whole molecule to be retained in the ER, and this improved the cross presentation of antigen on MHC I molecules. In conclusion, hexameric CT-like chimeric protein with dual effects of GM1 affinity and ER retention sequence were potential in improvement of cross presentation. The results laid a foundation for designing personalized tumor vaccine based on CT-like chimeric protein molecular structure.
Collapse
Affiliation(s)
- Xianying He
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, CN, China
| | - Kaixiang Fan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, CN, China
| | - Haiyan Gong
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, CN, China
| | - Mingqin Huang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, CN, China
| | - Qingsong Zeng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, CN, China
| | - Junjie Huang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, CN, China
| | - Ximing Peng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, CN, China
| | - Peifang Lai
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, CN, China
| | - Yujing Lu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, CN, China
| | - Huaqian Wang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, CN, China.
| |
Collapse
|
14
|
Pan W, Tao T, Qiu Y, Zhu X, Zhou X. Natural killer cells at the forefront of cancer immunotherapy with immune potency, genetic engineering, and nanotechnology. Crit Rev Oncol Hematol 2024; 193:104231. [PMID: 38070841 DOI: 10.1016/j.critrevonc.2023.104231] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 11/22/2023] [Accepted: 12/05/2023] [Indexed: 01/07/2024] Open
Abstract
Natural killer (NK) cells are vital components of the human immune system, acting as innate lymphocytes and playing a crucial role in immune surveillance. Their unique ability to independently eliminate target cells without antigen contact or antibodies has sparked interest in immunological research. This review examines recent NK cell developments and applications, encompassing immune functions, interactions with target cells, genetic engineering techniques, pharmaceutical interventions, and implications in cancers. Insights into NK cell regulation emerge, with a focus on promising genetic engineering like CAR-engineered NK cells, enhancing specificity against tumors. Immune checkpoint inhibitors also enhance NK cells' potential in cancer therapy. Nanotechnology's emergence as a tool for targeted drug delivery to improve NK cell therapies is explored. In conclusion, NK cells are pivotal in immunity, holding exciting potential in cancer immunotherapy. Ongoing research promises novel therapeutic strategies, advancing immunotherapy and medical interventions.
Collapse
Affiliation(s)
- Weiyi Pan
- Department of Immunology, School of Medicine, Nantong University, Nantong, China; School of Public Health, Southern Medical University, Guangzhou, China
| | - Tao Tao
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Yishu Qiu
- Department of Biology, College of Arts and Science, New York University, New York, USA
| | - Xiao Zhu
- Computational Systems Biology Lab (CSBL), The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.
| | - Xiaorong Zhou
- Department of Immunology, School of Medicine, Nantong University, Nantong, China.
| |
Collapse
|
15
|
Liu K, He S, Sun S, Zhang X, He Y, Quan F, Pang B, Xiao Y. Computational Quantification of Cancer Immunoediting. Cancer Immunol Res 2023; 11:1159-1167. [PMID: 37540180 DOI: 10.1158/2326-6066.cir-22-0926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/31/2023] [Accepted: 07/10/2023] [Indexed: 08/05/2023]
Abstract
The remarkable success of cancer immunotherapy has revolutionized cancer treatment, emphasizing the importance of tumor-immune interactions in cancer evolution and treatment. Cancer immunoediting describes the dual effect of tumor-immune interactions: inhibiting tumor growth by destroying tumor cells and facilitating tumor escape by shaping tumor immunogenicity. To better understand tumor-immune interactions, it is critical to develop computational methods to measure the extent of cancer immunoediting. In this review, we provide a comprehensive overview of the computational methods for quantifying cancer immunoediting. We focus on describing the basic ideas, computational processes, advantages, limitations, and influential factors. We also summarize recent advances in quantifying cancer immunoediting studies and highlight future research directions. As the methods for quantifying cancer immunoediting are continuously improved, future research will further help define the role of immunity in tumorigenesis and hopefully provide a basis for the design of new personalized cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Kun Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shengyuan He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Shangqin Sun
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Xinxin Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yanzhen He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Fei Quan
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Bo Pang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yun Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
16
|
Zhang C, Zhang C, Wang H. Immune-checkpoint inhibitor resistance in cancer treatment: Current progress and future directions. Cancer Lett 2023; 562:216182. [PMID: 37076040 DOI: 10.1016/j.canlet.2023.216182] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 04/07/2023] [Accepted: 04/11/2023] [Indexed: 04/21/2023]
Abstract
Cancer treatment has been advanced with the advent of immune checkpoint inhibitors (ICIs) exemplified by anti-cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), anti-programmed cell death protein 1 (PD-1) and programmed cell death ligand 1 (PD-L1) drugs. Patients have reaped substantial benefit from ICIs in many cancer types. However, few patients benefit from ICIs whereas the vast majority undergoing these treatments do not obtain survival benefit. Even for patients with initial responses, they may encounter drug resistance in their subsequent treatments, which limits the efficacy of ICIs. Therefore, a deepening understanding of drug resistance is critically important for the explorations of approaches to reverse drug resistance and to boost ICI efficacy. In the present review, different mechanisms of ICI resistance have been summarized according to the tumor intrinsic, tumor microenvironment (TME) and host classifications. We further elaborated corresponding strategies to battle against such resistance accordingly, which include targeting defects in antigen presentation, dysregulated interferon-γ (IFN-γ) signaling, neoantigen depletion, upregulation of other T cell checkpoints as well as immunosuppression and exclusion mediated by TME. Moreover, regarding the host, several additional approaches that interfere with diet and gut microbiome have also been described in reversing ICI resistance. Additionally, we provide an overall glimpse into the ongoing clinical trials that utilize these mechanisms to overcome ICI resistance. Finally, we summarize the challenges and opportunities that needs to be addressed in the investigation of ICI resistance mechanisms, with the aim to benefit more patients with cancer.
Collapse
Affiliation(s)
- Chenyue Zhang
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai Medical College, Shanghai, China
| | - Chenxing Zhang
- Department of Nephrology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haiyong Wang
- Department of Internal Medicine-Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.
| |
Collapse
|
17
|
Meng L, Teng Z, Yang S, Wang N, Guan Y, Chen X, Liu Y. Biomimetic nanoparticles for DC vaccination: a versatile approach to boost cancer immunotherapy. NANOSCALE 2023; 15:6432-6455. [PMID: 36916703 DOI: 10.1039/d2nr07071e] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Cancer immunotherapy, which harnesses the immune system to fight cancer, has begun to make a breakthrough in clinical applications. Dendritic cells (DCs) are the bridge linking innate and adaptive immunity and the trigger of tumor immune response. Considering the cumbersome process and poor efficacy of classic DC vaccines, there has been interest in transferring the field of in vitro-generated DC vaccines to nanovaccines. Conventional nanoparticles have insufficient targeting ability and are easily cleared by the reticuloendothelial system. Biological components have evolved very specific functions, which are difficult to fully reproduce with synthetic materials, making people interested in using the further understanding of biological systems to prepare nanoparticles with new and enhanced functions. Biomimetic nanoparticles are semi-biological or nature-derived delivery systems comprising one or more natural materials, which have a long circulation time in vivo and excellent performance of targeting DCs, and can mimic the antigen-presenting behavior of DCs. In this review, we introduce the classification, design, preparation, and challenges of different biomimetic nanoparticles, and discuss their application in activating DCs in vivo and stimulating T cell antitumor immunity. Incorporating biomimetic nanoparticles into cancer immunotherapy has shown outstanding advantages in precisely coaxing the immune system against cancer.
Collapse
Affiliation(s)
- Lingyang Meng
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - Zhuang Teng
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - Shuang Yang
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - Na Wang
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - YingHua Guan
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - Xiguang Chen
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
- Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266000, P.R. China
| | - Ya Liu
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| |
Collapse
|
18
|
Yin L, Li H, Shi L, Chen K, Pan H, Han W. Research advances in nanomedicine applied to the systemic treatment of colorectal cancer. Int J Cancer 2023; 152:807-821. [PMID: 35984398 DOI: 10.1002/ijc.34256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 01/06/2023]
Abstract
The systematic treatment of colorectal cancer (CRC) still has room for improvement. The efficacy of chemotherapy, that of anti-vascular therapy, and that of immunotherapy have been unsatisfactory. In recent years, nanomaterials have been used as carriers to improve the bioavailability of anticancer drugs. For the treatment of colorectal cancer, nanodrugs increase the possibility of more precise targeted delivery. However, the actual benefits may cover more aspects. Nanocarriers can produce synergistic effects with anticancer drugs, including the scavenging of reactive oxygen species and co-delivery of a variety of drugs. Currently, immunotherapy has very limited clinical applications in CRC. Modified nanocarriers can activate the immune microenvironment, which can be used for staging antigen recognition or the immune response. Cancer vaccines based on nanomaterials and modified immune checkpoint inhibitors have shown therapeutic potential in animal models. Considering the direct or indirect relationship between the intestinal microflora and CRC, a variety of nanodrugs that regulate microbial function have been explored as an anticancer strategy, and the special structure of microorganisms can also be used as a basis for improving the delivery of traditional nanoparticles (NPs). This review summarizes recent research performed on nanocarriers in in vivo and in vitro models and the synergistic anticancer effects of nanocarriers, focusing on the interaction between NPs and the body, resulting in enhanced efficacy and immune activation. Furthermore, this review describes the current trend of NPs used in the treatment of CRC.
Collapse
Affiliation(s)
- Luxi Yin
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haozhe Li
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Linlin Shi
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
19
|
Li J, Wu Y, Xiang J, Wang H, Zhuang Q, Wei T, Cao Z, Gu Q, Liu Z, Peng R. Fluoroalkane modified cationic polymers for personalized mRNA cancer vaccines. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2023; 456:140930. [PMID: 36531858 PMCID: PMC9743697 DOI: 10.1016/j.cej.2022.140930] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/12/2022] [Accepted: 12/10/2022] [Indexed: 06/17/2023]
Abstract
Messenger RNA (mRNA) vaccines, while demonstrating great successes in the fight against COVID-19, have been extensively studied in other areas such as personalized cancer immunotherapy based on tumor neoantigens. In addition to the design of mRNA sequences and modifications, the delivery carriers are also critical in the development of mRNA vaccines. In this work, we synthesized fluoroalkane-grafted polyethylenimine (F-PEI) for mRNA delivery. Such F-PEI could promote intracellular delivery of mRNA and activate the Toll-like receptor 4 (TLR4)-mediated signaling pathway. The nanovaccine formed by self-assembly of F-PEI and the tumor antigen-encoding mRNA, without additional adjuvants, could induce the maturation of dendritic cells (DCs) and trigger efficient antigen presentation, thereby eliciting anti-tumor immune responses. Using the mRNA encoding the model antigen ovalbumin (mRNAOVA), our F-PEI-based mRNAOVA cancer vaccine could delay the growth of established B16-OVA melanoma. When combined with immune checkpoint blockade therapy, the F-PEI-based MC38 neoantigen mRNA cancer vaccine was able to suppress established MC38 colon cancer and prevent tumor reoccurrence. Our work presents a new tool for mRNA delivery, promising not only for personalized cancer vaccines but also for other mRNA-based immunotherapies.
Collapse
Affiliation(s)
- Junyan Li
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, 199 Ren'ai Rd, Suzhou, Jiangsu 215123, China
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, 199 Ren'ai Rd, Suzhou, Jiangsu 215123, China
| | - Yuanyuan Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, 199 Ren'ai Rd, Suzhou, Jiangsu 215123, China
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, 199 Ren'ai Rd, Suzhou, Jiangsu 215123, China
| | - Jian Xiang
- WuXi AppTec (Suzhou) Co., Ltd., 1336 Wuzhong Avenue, Wuzhong District, Suzhou 215104, China
| | - Hairong Wang
- Children's Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Qi Zhuang
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, 199 Ren'ai Rd, Suzhou, Jiangsu 215123, China
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, 199 Ren'ai Rd, Suzhou, Jiangsu 215123, China
| | - Ting Wei
- InnoBM Pharmaceuticals Co., Ltd., Suzhou, Jiangsu 215000, China
| | - Zhiqin Cao
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, 199 Ren'ai Rd, Suzhou, Jiangsu 215123, China
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, 199 Ren'ai Rd, Suzhou, Jiangsu 215123, China
| | - Qingyang Gu
- WuXi AppTec (Suzhou) Co., Ltd., 1336 Wuzhong Avenue, Wuzhong District, Suzhou 215104, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, 199 Ren'ai Rd, Suzhou, Jiangsu 215123, China
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, 199 Ren'ai Rd, Suzhou, Jiangsu 215123, China
| | - Rui Peng
- Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, 199 Ren'ai Rd, Suzhou, Jiangsu 215123, China
- Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, 199 Ren'ai Rd, Suzhou, Jiangsu 215123, China
| |
Collapse
|
20
|
Identification and Validation of RELN Mutation as a Response Indicator for Immune Checkpoint Inhibitor Therapy in Melanoma and Non-Small Cell Lung Cancer. Cells 2022; 11:cells11233841. [PMID: 36497098 PMCID: PMC9741468 DOI: 10.3390/cells11233841] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 11/15/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022] Open
Abstract
Remarkable clinical benefits in several advanced cancers are observed under the treatment of immune checkpoint inhibitor (ICI) agents. However, only a smaller proportion of patients respond to the treatments. Reelin (RELN) is frequently mutated in the cancer genome. In this study, the RELN mutation association with ICI treatment efficacy in melanoma and non-small cell lung cancer (NSCLC) was elucidated. Data from 631 melanoma and 109 NSCLC patients with both ICI treatment data and pre-treatment mutational profiles were collected. In addition, from the Cancer Genome Atlas (TCGA) project, we also obtained both tumors to explore the immunologic features behind RELN mutations. Melanoma patients with RELN mutations exhibited a favorable ICI survival benefit when compared with wild-type patients (HR: 0.66, 95% CI: 0.51-0.87, p = 0.003). A higher response rate was also noticed in RELN-mutated patients (38.9% vs. 28.3%, p = 0.017). The association of RELN mutations with a preferable immunotherapy outcome and response was further confirmed in NSCLC. Further exploration demonstrated that favorable immunocyte infiltration and immune response signaling pathways were found in patients with RELN mutations. In this study, RELN mutations were identified to connect with a better immune microenvironment and an improved ICI efficacy in melanoma and NSCLC, which provides a potential biomarker for immunological feature evaluation and immunotherapeutic outcome prediction at the molecular level.
Collapse
|
21
|
Zhong X, Li C, Zhao G, Li M, Chen S, Cao Y, Wang Q, Sun J, Zhu S, Chang S. Photoacoustic mediated multifunctional tumor antigen trapping nanoparticles inhibit the recurrence and metastasis of ovarian cancer by enhancing tumor immunogenicity. J Nanobiotechnology 2022; 20:468. [PMID: 36329515 PMCID: PMC9632083 DOI: 10.1186/s12951-022-01682-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022] Open
Abstract
The hypoimmunogenicity of tumors is one of the main bottlenecks of cancer immunotherapy. Enhancing tumor immunogenicity can improve the efficacy of tumor immunotherapy by increasing antigen exposure and presentation, and establishing an inflammatory microenvironment. Here, a multifunctional antigen trapping nanoparticle with indocyanine green (ICG), aluminum hydroxide (Al(OH)3) and oxaliplatin (OXA) (PPIAO) has been developed for tumor photoacoustic/ultrasound dual-modality imaging and therapy. The combination of photothermal/photodynamic therapy and chemotherapy induced tumor antigen exposure and release through immunogenic death of tumor cells. A timely capture and storage of antigens by aluminum hydroxide enabled dendritic cells to recognize and present those antigens spatiotemporally. In an ovarian tumor model, the photoacoustic-mediated PPIAO NPs combination therapy achieved a transition from "cold tumor" to "hot tumor" that promoted more CD8+ T lymphocytes activation in vivo and intratumoral infiltration, and successfully inhibited the growth of primary and metastatic tumors. An in situ tumor vaccine effect was produced from the treated tumor tissue, assisting mice against the recurrence of tumor cells. This study provided a simple and effective personalized tumor vaccine strategy for better treatment of metastatic and recurrent tumors. The developed multifunctional tumor antigen trapping nanoparticles may be a promising nanoplatform for integrating multimodal imaging monitoring, tumor treatment, and tumor vaccine immunotherapy.
Collapse
Affiliation(s)
- Xiaowen Zhong
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Chenyang Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Guangzong Zhao
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Mengmeng Li
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Shuning Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Yang Cao
- Chongqing Key Laboratory of Ultrasound Molecular Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Qi Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Jiangchuan Sun
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China
| | - Shenyin Zhu
- Department of Pharmacy, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400042, People's Republic of China.
| | - Shufang Chang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, People's Republic of China.
| |
Collapse
|
22
|
Neoantigen Vaccines; Clinical Trials, Classes, Indications, Adjuvants and Combinatorial Treatments. Cancers (Basel) 2022; 14:cancers14205163. [PMID: 36291947 PMCID: PMC9600771 DOI: 10.3390/cancers14205163] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/02/2022] [Accepted: 10/18/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Personalized neoantigen vaccines are a diverse group of personally tailored cancer vaccines that strengthen patients´ own immune reaction against cancer antigens. We analyzed 147 neoantigen vaccine clinical trials from ClinicalTrials.gov database and showed that peptide vaccines were the dominating vaccine type, while there were multiple new neoantigen vaccine types in the field. We also showed that neoantigen vaccines were mostly used in the treatment of small cell lung cancer, non-small cell lung cancer, melanoma, and glioma. According to our results neoantigen vaccines work at their best when combined with other treatments such as immune checkpoint inhibitors, chemotherapy, and radiation therapy. The effect of some neoantigen vaccine types were also often promoted with adjuvant therapy where poly-ICLC were the most recurrent adjuvant choice. Abstract Personalized neoantigen vaccines are a highly specific cancer treatment designed to induce a robust cytotoxic T-cell attack against a patient’s cancer antigens. In this study, we searched ClinicalTrials.gov for neoantigen vaccine clinical trials and systematically analyzed them, a total of 147 trials. Peptide vaccines are the largest neoantigen vaccine type, comprising up to 41% of the clinical trials. However, mRNA vaccines are a growing neoantigen vaccine group, especially in the most recent clinical trials. The most common cancer types in the clinical trials are glioma, lung cancer, and malignant melanoma, being seen in more than half of the clinical trials. Small-cell lung cancer and non-small-cell lung cancer are the largest individual cancer types. According to the results from the clinical trials, neoantigen vaccines work best when combined with other cancer treatments, and popular combination treatments include immune checkpoint inhibitors, chemotherapy, and radiation therapy. Additionally, half of the clinical trials combined neoantigen vaccines with an adjuvant to boost the immune effects, with poly-ICLC being the most recurrent adjuvant choice. This study clarifies the rapid clinical trial development of personalized neoantigen vaccines as an emerging class of cancer treatment with increasingly diversified opportunities in classes, indications, and combinatorial treatments.
Collapse
|
23
|
Xu Y, Tao T, Li S, Tan S, Liu H, Zhu X. Prognostic model and immunotherapy prediction based on molecular chaperone-related lncRNAs in lung adenocarcinoma. Front Genet 2022; 13:975905. [PMID: 36313456 PMCID: PMC9606628 DOI: 10.3389/fgene.2022.975905] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 09/21/2022] [Indexed: 11/17/2022] Open
Abstract
Introduction: Molecular chaperones and long non-coding RNAs (lncRNAs) have been confirmed to be closely related to the occurrence and development of tumors, especially lung cancer. Our study aimed to construct a kind of molecular chaperone-related long non-coding RNAs (MCRLncs) marker to accurately predict the prognosis of lung adenocarcinoma (LUAD) patients and find new immunotherapy targets. Methods: In this study, we acquired molecular chaperone genes from two databases, Genecards and molecular signatures database (MsigDB). And then, we downloaded transcriptome data, clinical data, and mutation information of LUAD patients through the Cancer Genome Atlas (TCGA). MCRLncs were determined by Spearman correlation analysis. We used univariate, least absolute shrinkage and selection operator (LASSO) and multivariate Cox regression analysis to construct risk models. Kaplan-meier (KM) analysis was used to understand the difference in survival between high and low-risk groups. Nomogram, calibration curve, concordance index (C-index) curve, and receiver operating characteristic (ROC) curve were used to evaluate the accuracy of the risk model prediction. In addition, we used gene ontology (GO) enrichment analysis and kyoto encyclopedia of genes and genomes (KEGG) enrichment analyses to explore the potential biological functions of MCRLncs. Immune microenvironmental landscapes were constructed by using single-sample gene set enrichment analysis (ssGSEA), tumor immune dysfunction and exclusion (TIDE) algorithm, “pRRophetic” R package, and “IMvigor210” dataset. The stem cell index based on mRNAsi expression was used to further evaluate the patient’s prognosis. Results: Sixteen MCRLncs were identified as independent prognostic indicators in patients with LUAD. Patients in the high-risk group had significantly worse overall survival (OS). ROC curve suggested that the prognostic features of MCRLncs had a good predictive ability for OS. Immune system activation was more pronounced in the high-risk group. Prognostic features of the high-risk group were strongly associated with exclusion and cancer-associated fibroblasts (CAF). According to this prognostic model, a total of 15 potential chemotherapeutic agents were screened for the treatment of LUAD. Immunotherapy analysis showed that the selected chemotherapeutic drugs had potential application value. Stem cell index mRNAsi correlates with prognosis in patients with LUAD. Conclusion: Our study established a kind of novel MCRLncs marker that can effectively predict OS in LUAD patients and provided a new model for the application of immunotherapy in clinical practice.
Collapse
Affiliation(s)
- Yue Xu
- Marine Medical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Tao Tao
- Department of Gastroscope, Zibo Central Hospital, Zibo, China
| | - Shi Li
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China
| | - Shuzhen Tan
- Department of Dermatology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Haiyan Liu
- Department of Cardiovascular Medicine, Nanchong Central Hospital, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong, China
- *Correspondence: Haiyan Liu, ; Xiao Zhu,
| | - Xiao Zhu
- Marine Medical Research Institute, Guangdong Medical University, Zhanjiang, China
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China
- Laboratory of Molecular Diagnosis, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Haiyan Liu, ; Xiao Zhu,
| |
Collapse
|
24
|
Zhu M, Chen E, Yu S, Xu C, Yu Y, Cao X, Li W, Zhang P, Wang Y, Lian B, Zhang S, Qu Y, Huang L, Shi W, Cui Y, Qian L, Liu T. Genomic profiling and the impact of MUC19 mutation in hepatoid adenocarcinoma of the stomach. Cancer Commun (Lond) 2022; 42:1032-1035. [PMID: 35851588 PMCID: PMC9558685 DOI: 10.1002/cac2.12336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 05/22/2022] [Accepted: 07/04/2022] [Indexed: 12/04/2022] Open
Affiliation(s)
- Mengxuan Zhu
- Department of Medical Oncology, Zhongshan HospitalFudan UniversityShanghai20032P. R. China
- Center of Evidence‐based medicineFudan UniversityShanghai20032P. R. China
| | - Erbao Chen
- Hepato‐Pancreato‐Biliary SurgeryPeking University Shenzhen HospitalShenzhenGuangdong518036P. R. China
| | - Shan Yu
- Department of Medical Oncology, Zhongshan HospitalFudan UniversityShanghai20032P. R. China
- Center of Evidence‐based medicineFudan UniversityShanghai20032P. R. China
| | - Chen Xu
- Department of Pathology, Zhongshan HospitalFudan UniversityShanghai20032P. R. China
| | - Yiyi Yu
- Department of Medical Oncology, Zhongshan HospitalFudan UniversityShanghai20032P. R. China
| | - Xin Cao
- Institute of Clinical Science, Zhongshan HospitalFudan UniversityShanghai20032P. R. China
| | - Wei Li
- Department of Medical Oncology, Zhongshan HospitalFudan UniversityShanghai20032P. R. China
| | - Pengfei Zhang
- Department of Medical Oncology, Zhongshan HospitalFudan UniversityShanghai20032P. R. China
| | - Yan Wang
- Department of Medical Oncology, Zhongshan HospitalFudan UniversityShanghai20032P. R. China
| | | | | | - Yueting Qu
- Origimed Co. LtdShanghai20032P. R. China
| | | | - Weiwei Shi
- Origimed Co. LtdShanghai20032P. R. China
| | - Yuehong Cui
- Department of Medical Oncology, Zhongshan HospitalFudan UniversityShanghai20032P. R. China
- Center of Evidence‐based medicineFudan UniversityShanghai20032P. R. China
| | - Li Qian
- Department of Medical Oncology, Zhongshan HospitalFudan UniversityShanghai20032P. R. China
- Center of Evidence‐based medicineFudan UniversityShanghai20032P. R. China
- Cancer centerZhongshan hospitalFudan UniversityShanghai20032P. R. China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan HospitalFudan UniversityShanghai20032P. R. China
- Center of Evidence‐based medicineFudan UniversityShanghai20032P. R. China
- Cancer centerZhongshan hospitalFudan UniversityShanghai20032P. R. China
| |
Collapse
|
25
|
Yu G, He X, Li X, Wu Y. Driving neoantigen-based cancer vaccines for personalized immunotherapy into clinic: A burdensome journey to promising land. Biomed Pharmacother 2022; 153:113464. [DOI: 10.1016/j.biopha.2022.113464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/15/2022] [Accepted: 07/21/2022] [Indexed: 11/02/2022] Open
|
26
|
Kye Y, Nagineni L, Gadad S, Ramirez F, Riva H, Fernandez L, Samaniego M, Holland N, Yeh R, Takigawa K, Dhandayuthapani S, Chacon J. The Identification and Clinical Applications of Mutated Antigens in the Era of Immunotherapy. Cancers (Basel) 2022; 14:4255. [PMID: 36077792 PMCID: PMC9454936 DOI: 10.3390/cancers14174255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 11/17/2022] Open
Abstract
The era of personalized cancer therapy is here. Advances in the field of immunotherapy have paved the way for the development of individualized neoantigen-based therapies that can translate into favorable treatment outcomes and fewer side effects for patients. Addressing challenges related to the identification, access, and clinical application of neoantigens is critical to accelerating the development of individualized immunotherapy for cancer patients.
Collapse
Affiliation(s)
- Yae Kye
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Lokesh Nagineni
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Shrikanth Gadad
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
- L. Frederick Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
- Center of Emphasis in Cancer, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
- Mays Cancer Center, UT Health San Antonio MD Anderson Cancer Center, San Antonio, TX 78229, USA
| | - Fabiola Ramirez
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Hannah Riva
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Lorena Fernandez
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Michelle Samaniego
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Nathan Holland
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Rose Yeh
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Kei Takigawa
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Subramanian Dhandayuthapani
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
- L. Frederick Francis Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
- Center of Emphasis in Infectious Diseases, Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| | - Jessica Chacon
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| |
Collapse
|
27
|
Immunotherapeutic Strategies for Head and Neck Squamous Cell Carcinoma (HNSCC): Current Perspectives and Future Prospects. Vaccines (Basel) 2022; 10:vaccines10081272. [PMID: 36016159 PMCID: PMC9416402 DOI: 10.3390/vaccines10081272] [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: 06/29/2022] [Revised: 08/03/2022] [Accepted: 08/03/2022] [Indexed: 11/21/2022] Open
Abstract
Neoantigens are abnormal proteins produced by genetic mutations in somatic cells. Because tumour neoantigens are expressed only in tumour cells and have immunogenicity, they may represent specific targets for precision immunotherapy. With the reduction in sequencing cost, continuous advances in artificial intelligence technology and an increased understanding of tumour immunity, neoantigen vaccines and adoptive cell therapy (ACT) targeting neoantigens have become research hotspots. Approximately 900,000 patients worldwide are diagnosed with head and neck squamous cell carcinoma (HNSCC) each year. Due to its high mutagenicity and abundant lymphocyte infiltration, HNSCC naturally generates a variety of potential new antigen targets that may be used for HNSCC immunotherapies. Currently, the main immunotherapy for HNSCC is use of immune checkpoint inhibitors(ICIs). Neoantigen vaccines and adoptive cell therapy targeting neoantigens are extensions of immunotherapy for HNSCC, and a large number of early clinical trials are underway in combination with immune checkpoint inhibitors for the treatment of recurrent or metastatic head and neck squamous cell carcinoma (R/M HNSCC). In this paper, we review recent neoantigen vaccine trials related to the treatment of HNSCC, introduce adoptive cell therapy targeting neoantigens, and propose a potential treatment for HNSCC. The clinical application of immune checkpoint inhibitor therapy and its combination with neoantigen vaccines in the treatment of HNSCC are summarized, and the prospect of using neoantigen to treat HNSCC is discussed and proposed.
Collapse
|
28
|
Tan S, Kong Y, Xian Y, Gao P, Xu Y, Wei C, Lin P, Ye W, Li Z, Zhu X. The Mechanisms of Ferroptosis and the Applications in Tumor Treatment: Enemies or Friends? Front Mol Biosci 2022; 9:938677. [PMID: 35911967 PMCID: PMC9334798 DOI: 10.3389/fmolb.2022.938677] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Ferroptosis, as a newly discovered non-apoptotic cell death mode, is beginning to be explored in different cancer. The particularity of ferroptosis lies in the accumulation of iron dependence and lipid peroxides, and it is different from the classical cell death modes such as apoptosis and necrosis in terms of action mode, biochemical characteristics, and genetics. The mechanism of ferroptosis can be divided into many different pathways, so it is particularly important to identify the key sites of ferroptosis in the disease. Herein, based on ferroptosis, we analyze the main pathways in detail. More importantly, ferroptosis is linked to the development of different systems of the tumor, providing personalized plans for the examination, treatment, and prognosis of cancer patients. Although some mechanisms and side effects of ferroptosis still need to be studied, it is still a promising method for cancer treatment.
Collapse
Affiliation(s)
- Shuzheng Tan
- School of Laboratory Medicine and Biological Engineering, Hangzhou Medical College, Hangzhou, China
- Department of Dermatology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Ying Kong
- Department of Clinical Laboratory, Hubei No.3 People’s Hospital of Jianghan University, Wuhan, China
| | - Yongtong Xian
- Computational Oncology Laboratory, Guangdong Medical University, Zhanjiang, China
| | - Pengbo Gao
- Computational Oncology Laboratory, Guangdong Medical University, Zhanjiang, China
| | - Yue Xu
- Computational Oncology Laboratory, Guangdong Medical University, Zhanjiang, China
| | - Chuzhong Wei
- Computational Oncology Laboratory, Guangdong Medical University, Zhanjiang, China
| | - Peixu Lin
- Computational Oncology Laboratory, Guangdong Medical University, Zhanjiang, China
| | - Weilong Ye
- Computational Oncology Laboratory, Guangdong Medical University, Zhanjiang, China
| | - Zesong Li
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China
- *Correspondence: Zesong Li, ; Xiao Zhu,
| | - Xiao Zhu
- School of Laboratory Medicine and Biological Engineering, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Zesong Li, ; Xiao Zhu,
| |
Collapse
|
29
|
Su X, Zheng G, Gui Z, Yang X, Zhang L, Pan F. A Systematic Analysis Reveals the Prognostic and Immunological Role of Reptin/RUVBL2 in Human Tumors. Front Genet 2022; 13:911223. [PMID: 35754815 PMCID: PMC9213802 DOI: 10.3389/fgene.2022.911223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 05/16/2022] [Indexed: 12/03/2022] Open
Abstract
Reptin/RUVBL2 is involved in the remodeling of chromatin, DNA damage repair, and regulation of the cell cycle, all of which help to play essential roles in cancer. However, relevant pan-cancer analysis of Reptin is lacking. This study first investigated the potential oncogenic roles of Reptin and revealed a relationship between Reptin with clinicopathological characteristics and immune infiltration based on big data. Here, we showed that Reptin is overexpressed in many cancers. A significant association exists between the expression of Reptin and the prognosis of cancer cases. Reptin had a meaningful interaction with the immune infiltration of CD4+ Th1 cells and immune modulator genes in multiple cancer types. And negative correlation exists between Reptin and cancer-associated fibroblasts in BRCA, PRAD, TGCT, and THYM. A significant negative association exists between Reptin and regulatory T cells in TGCT and THCA. Moreover, Reptin is significantly associated with genomic heterogeneity, DNA mismatch repair genes, methyltransferase, and RNA modification genes in specific cancer types. Spliceosome, Hippo signaling pathway, DNA replication pathway, and acetyltransferase activity-associated functions were observed in the effect of Reptin on the tumor. This systematic analysis highlights Reptin as a vital cancer regulator among numerous genes and proved its potential prognosticator value and therapeutic target role for specific tumor types.
Collapse
Affiliation(s)
- Xiaoru Su
- Department of Clinical Laboratory, Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Gaoming Zheng
- Department of Clinical Laboratory, Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Zhifang Gui
- Department of Clinical Laboratory, Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Xiao Yang
- Department of Clinical Laboratory, Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Lahong Zhang
- Department of Clinical Laboratory, Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Feng Pan
- Department of Clinical Laboratory, Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
30
|
Li X, Li M, Huang M, Lin Q, Fang Q, Liu J, Chen X, Liu L, Zhan X, Shan H, Lu D, Li Q, Li Z, Zhu X. The multi-molecular mechanisms of tumor-targeted drug resistance in precision medicine. Biomed Pharmacother 2022; 150:113064. [PMID: 35658234 DOI: 10.1016/j.biopha.2022.113064] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/28/2022] [Accepted: 04/28/2022] [Indexed: 11/02/2022] Open
Abstract
Clinically, cancer drug therapy is still dominated by chemotherapy drugs. Although the emergence of targeted drugs has greatly improved the survival rate of patients with advanced cancer, drug resistance has always been a difficult problem in clinical cancer treatment. At the current level of medicine, most drugs cannot escape the fate of drug resistance. With the emergence and development of gene detection, liquid biopsy ctDNA technology, and single-cell sequencing technology, the molecular mechanism of tumor drug resistance has gradually emerged. Drugs can also be updated in response to drug resistance mechanisms and bring higher survival benefits. The use of new drugs often leads to new mechanisms of resistance. In this review, the multi-molecular mechanisms of drug resistance are introduced, and the overcoming of drug resistance is discussed from the perspective of the tumor microenvironment.
Collapse
Affiliation(s)
- Xinming Li
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China; Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Mingdong Li
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Meiying Huang
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Qianyi Lin
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Qiuping Fang
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Jianjiang Liu
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Xiaohui Chen
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Lin Liu
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Xuliang Zhan
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Huisi Shan
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Deshuai Lu
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Qinlan Li
- Cancer Research Center, Guangdong Medical University, Zhanjiang, China
| | - Zesong Li
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors,Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China.
| | - Xiao Zhu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China; Cancer Research Center, Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
31
|
Lu M, Xu L, Jian X, Tan X, Zhao J, Liu Z, Zhang Y, Liu C, Chen L, Lin Y, Xie L. dbPepNeo2.0: A Database for Human Tumor Neoantigen Peptides From Mass Spectrometry and TCR Recognition. Front Immunol 2022; 13:855976. [PMID: 35493528 PMCID: PMC9043652 DOI: 10.3389/fimmu.2022.855976] [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: 01/16/2022] [Accepted: 03/17/2022] [Indexed: 12/04/2022] Open
Abstract
Neoantigens are widely reported to induce T-cell response and lead to tumor regression, indicating a promising potential to immunotherapy. Previously, we constructed an open-access database, i.e., dbPepNeo, providing a systematic resource for human tumor neoantigens to storage and query. In order to expand data volume and application scope, we updated dbPepNeo to version 2.0 (http://www.biostatistics.online/dbPepNeo2). Here, we provide about 801 high-confidence (HC) neoantigens (increased by 170%) and 842,289 low-confidence (LC) HLA immunopeptidomes (increased by 107%). Notably, 55 class II HC neoantigens and 630 neoantigen-reactive T-cell receptor-β (TCRβ) sequences were firstly included. Besides, two new analytical tools are developed, DeepCNN-Ineo and BLASTdb. DeepCNN-Ineo predicts the immunogenicity of class I neoantigens, and BLASTdb performs local alignments to look for sequence similarities in dbPepNeo2.0. Meanwhile, the web features and interface have been greatly improved and enhanced.
Collapse
Affiliation(s)
- Manman Lu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China.,Shanghai-Ministry of Science and Technology (MOST) Key Laboratory of Health and Disease Genomics, Institute for Genome and Bioinformatics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Linfeng Xu
- Shanghai-Ministry of Science and Technology (MOST) Key Laboratory of Health and Disease Genomics, Institute for Genome and Bioinformatics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China.,School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Xingxing Jian
- Shanghai-Ministry of Science and Technology (MOST) Key Laboratory of Health and Disease Genomics, Institute for Genome and Bioinformatics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China.,Bioinformatics Center, National Clinical Research Centre for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxiu Tan
- Shanghai-Ministry of Science and Technology (MOST) Key Laboratory of Health and Disease Genomics, Institute for Genome and Bioinformatics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China.,Department of Bioinformatics and Biostatistics, Shanghai Jiao Tong University, Shanghai, China
| | - Jingjing Zhao
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China.,Shanghai-Ministry of Science and Technology (MOST) Key Laboratory of Health and Disease Genomics, Institute for Genome and Bioinformatics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Zhenhao Liu
- Shanghai-Ministry of Science and Technology (MOST) Key Laboratory of Health and Disease Genomics, Institute for Genome and Bioinformatics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Yu Zhang
- Shanghai-Ministry of Science and Technology (MOST) Key Laboratory of Health and Disease Genomics, Institute for Genome and Bioinformatics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China.,School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Chunyu Liu
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China.,Shanghai-Ministry of Science and Technology (MOST) Key Laboratory of Health and Disease Genomics, Institute for Genome and Bioinformatics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Lanming Chen
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Yong Lin
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Lu Xie
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China.,Shanghai-Ministry of Science and Technology (MOST) Key Laboratory of Health and Disease Genomics, Institute for Genome and Bioinformatics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China.,Bioinformatics Center, National Clinical Research Centre for Geriatric Disorders, Department of Geriatrics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
32
|
Yang S, Zou X, Yang H, Li J, Zhang A, Zhang L, Li C, Zhu L, Ma Z. Identification of Enhancer RNA CDK6-AS1 as a Potential Novel Prognostic Biomarker in Gastric Cancer. Front Genet 2022; 13:854211. [PMID: 35571043 PMCID: PMC9100412 DOI: 10.3389/fgene.2022.854211] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/01/2022] [Indexed: 12/24/2022] Open
Abstract
Background: This study aimed to confirm the role of enhancer RNAs (eRNAs) in gastric cancer and their clinical utility. Methods: We used Cox survival and relevance analysis to identify the candidate eRNAs in gastric cancer and performed Gene Ontology and Reactome pathway enrichment to determine the potential functions of eRNAs. Correlation between eRNA, tumor-infiltrating immune cells, and drug sensitivity was then analyzed. Results:CDK6-AS1, a long non-coding RNA cyclin-dependent kinase 6, may serve as a poor potential prognostic biomarker candidate in gastric cancer with a positive correlation with its target gene CDK6. The low CDK6-AS1 expression group showed more frequent mutated driver genes than the high expression group. Moreover, CDK6-AS1 is involved in a key oncogenic pathway of the cell cycle and RNA transcription. CDK6-AS1 also shows dysregulations and associations with prognosis at the pan-cancer level. This eRNA may also be associated with immune cell infiltration and drug sensitivity. Conclusion:CDK6-AS1 may be a potential prognostic biomarker and chemotherapeutic drug sensitivity predictor in gastric cancer.
Collapse
|
33
|
Nehra M, Uthappa UT, Kumar V, Kumar R, Dixit C, Dilbaghi N, Mishra YK, Kumar S, Kaushik A. Nanobiotechnology-assisted therapies to manage brain cancer in personalized manner. J Control Release 2021; 338:224-243. [PMID: 34418523 DOI: 10.1016/j.jconrel.2021.08.027] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/14/2021] [Accepted: 08/16/2021] [Indexed: 12/24/2022]
Abstract
There are numerous investigated factors that limit brain cancer treatment efficacy such as ability of prescribed therapy to cross the blood-brain barrier (BBB), tumor specific delivery of a therapeutics, transport within brain interstitium, and resistance of tumor cells against therapies. Recent breakthroughs in the field of nano-biotechnology associated with developing multifunctional nano-theranostic emerged as an effective way to manage brain cancer in terms of higher efficacy and least possible adverse effects. Keeping challenges and state-of-art accomplishments into consideration, this review proposes a comprehensive, careful, and critical discussion focused on efficient nano-enabled platforms including nanocarriers for drug delivery across the BBB and nano-assisted therapies (e.g., nano-immunotherapy, nano-stem cell therapy, and nano-gene therapy) investigated for brain cancer treatment. Besides therapeutic efficacy point-of-view, efforts are being made to explore ways projected to tune such developed nano-therapeutic for treating patients in personalized manner via controlling size, drug loading, delivery, and retention. Personalized brain tumor management based on advanced nano-therapies can potentially lead to excellent therapeutic benefits based on unique genetic signatures in patients and their individual disease profile. Moreover, applicability of nano-systems as stimulants to manage the brain cancer growth factors has also been discussed in photodynamic therapy and radiotherapy. Overall, this review offers a comprehensive information on emerging opportunities in nanotechnology for advancing the brain cancer treatment.
Collapse
Affiliation(s)
- Monika Nehra
- Department of Bio and Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana 125001, India; Department of Mechanical Engineering, University Institute of Engineering and Technology, Panjab University, Chandigarh 160014, India
| | - U T Uthappa
- Department of Environment and Energy Engineering, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea; Centre for Nano and Material Sciences, Jain University, Jain Global Campus, Bengaluru 562112, Karnataka, India
| | - Virendra Kumar
- Department of Bio and Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana 125001, India
| | - Rajesh Kumar
- Department of Mechanical Engineering, University Institute of Engineering and Technology, Panjab University, Chandigarh 160014, India
| | - Chandra Dixit
- Department of Chemistry, University of Connecticut, Storrs, CT, USA
| | - Neeraj Dilbaghi
- Department of Bio and Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana 125001, India
| | - Yogendra Kumar Mishra
- Smart Materials, NanoSYD, Mads Clausen Institute, University of Southern Denmark, Alsion 2, 6400, Sønderborg, Denmark
| | - Sandeep Kumar
- Department of Bio and Nano Technology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana 125001, India.
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Health Systems Engineering, Department of Environmental Engineering, Florida Polytechnic University, Lakeland, FL 33805-8531, United States.
| |
Collapse
|
34
|
Gao G, Li C, Fan W, Zhang M, Li X, Chen W, Li W, Liang R, Li Z, Zhu X. Brilliant glycans and glycosylation: Seq and ye shall find. Int J Biol Macromol 2021; 189:279-291. [PMID: 34389387 DOI: 10.1016/j.ijbiomac.2021.08.054] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/02/2021] [Accepted: 08/06/2021] [Indexed: 01/30/2023]
Abstract
Proteoglycosylation is the addition of monosaccharides or glycans to the protein peptide chain. This is a common post-translational modification of proteins with a variety of biological functions. At present, more than half of all biopharmaceuticals in clinic are modified by glycosylation. Most glycoproteins are potential drug targets and biomarkers for disease diagnosis. Therefore, in-depth study of glycan structure of glycoproteins will ultimately improve the sensitivity and specificity of glycoproteins for clinical disease detection. With the deepening of research, the function and application value of glycans and glycosylation has gradually emerged. This review systematically introduces the latest research progress of glycans and glycosylation. It encompasses six cancers, four viruses, and their latest discoveries in Alzheimer's disease, allergic diseases, congenital diseases, gastrointestinal diseases, inflammation, and aging.
Collapse
Affiliation(s)
- Guanwen Gao
- School of Laboratory Medicine, Bengbu Medical College, Bengbu, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Chen Li
- Department of Biology, Chemistry, Pharmacy, Free University of Berlin, Berlin 14195, Germany
| | - Wenguo Fan
- Department of Anesthesiology, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Mingtao Zhang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Xinming Li
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Wenqing Chen
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Weiquan Li
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Runzhang Liang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China
| | - Zesong Li
- Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), The First Affiliated Hospital of Shenzhen University, Shenzhen, China; Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China.
| | - Xiao Zhu
- School of Laboratory Medicine, Bengbu Medical College, Bengbu, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), The First Dongguan Affiliated Hospital, Guangdong Medical University, Dongguan, China; Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), The First Affiliated Hospital of Shenzhen University, Shenzhen, China; Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China.
| |
Collapse
|
35
|
Combining Cancer Vaccines with Immunotherapy: Establishing a New Immunological Approach. Int J Mol Sci 2021; 22:ijms22158035. [PMID: 34360800 PMCID: PMC8348347 DOI: 10.3390/ijms22158035] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Therapeutic cancer vaccines have become increasingly qualified for use in personalized cancer immunotherapy. A deeper understanding of tumor immunology and novel antigen delivery technologies has assisted in optimizing vaccine design. Therapeutic cancer vaccines aim to establish long-lasting immunological memory against tumor cells, thereby leading to effective tumor regression and minimizing non-specific or adverse events. However, due to several resistance mechanisms, significant challenges remain to be solved in order to achieve these goals. In this review, we describe our current understanding with respect to the use of the antigen repertoire in vaccine platform development. We also summarize various intrinsic and extrinsic resistance mechanisms behind the failure of cancer vaccine development in the past. Finally, we suggest a strategy that combines immune checkpoint inhibitors to enhance the efficacy of cancer vaccines.
Collapse
|
36
|
Abstract
Mesenchymal stem cells (MSCs), a kind of multipotent stem cells with self-renewal ability and multi-differentiation ability, have become the “practical stem cells” for the treatment of diseases. MSCs have immunomodulatory properties and can be used to treat autoimmune diseases, such as systemic lupus erythematosus (SLE) and Crohn’s disease. MSCs also can be used in cancer and aging. At present, many clinical experiments are using MSCs. MSCs can reduce the occurrence of inflammation and apoptosis of tissue cells, and promote the proliferation of endogenous tissue and organ cells, so as to achieve the effect of repairing tissue and organs. MSCs presumably also play an important role in Corona Virus Disease 2019 (COVID-19) infection.
Collapse
|
37
|
Wu Z, Li S, Zhu X. The Mechanism of Stimulating and Mobilizing the Immune System Enhancing the Anti-Tumor Immunity. Front Immunol 2021; 12:682435. [PMID: 34194437 PMCID: PMC8237941 DOI: 10.3389/fimmu.2021.682435] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/25/2021] [Indexed: 12/17/2022] Open
Abstract
Cancer immunotherapy is a kind of therapy that can control and eliminate tumors by restarting and maintaining the tumor-immune cycle and restoring the body's normal anti-tumor immune response. Although immunotherapy has great potential, it is currently only applicable to patients with certain types of tumors, such as melanoma, lung cancer, and cancer with high mutation load and microsatellite instability, and even in these types of tumors, immunotherapy is not effective for all patients. In order to enhance the effectiveness of tumor immunotherapy, this article reviews the research progress of tumor microenvironment immunotherapy, and studies the mechanism of stimulating and mobilizing immune system to enhance anti-tumor immunity. In this review, we focused on immunotherapy against tumor microenvironment (TME) and discussed the important research progress. TME is the environment for the survival and development of tumor cells, which is composed of cell components and non-cell components; immunotherapy for TME by stimulating or mobilizing the immune system of the body, enhancing the anti-tumor immunity. The checkpoint inhibitors can effectively block the inhibitory immunoregulation, indirectly strengthen the anti-tumor immune response and improve the effect of immunotherapy. We also found the checkpoint inhibitors have brought great changes to the treatment model of advanced tumors, but the clinical treatment results show great individual differences. Based on the close attention to the future development trend of immunotherapy, this study summarized the latest progress of immunotherapy and pointed out a new direction. To study the mechanism of stimulating and mobilizing the immune system to enhance anti-tumor immunity can provide new opportunities for cancer treatment, expand the clinical application scope and effective population of cancer immunotherapy, and improve the survival rate of cancer patients.
Collapse
Affiliation(s)
- Zhengguo Wu
- Department of Thoracic Surgery, Yantian District People’s Hospital, Shenzhen, China
| | - Shang Li
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang, China
| | - Xiao Zhu
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People’s Hospital (Shenzhen Institute of Translational Medicine), Shenzhen, China
- First Affiliated Hospital, Bengbu Medical College, Bengbu, China
| |
Collapse
|
38
|
Cancer neoantigens as potential targets for immunotherapy. Clin Exp Metastasis 2021; 39:51-60. [PMID: 33950415 PMCID: PMC8097110 DOI: 10.1007/s10585-021-10091-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 03/22/2021] [Indexed: 12/13/2022]
Abstract
Immune checkpoint inhibitors (ICIs) targeting the cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) and programed cell death protein 1 (PD-1) or its ligand PD-L1 have increased the survival and cure rates for patients with many cancer types in various disease settings. However, only 10–40% of cancer patients benefited from these ICIs, of whom ~ 20% have treatment interruption or discontinuation due to immune-related adverse events that can be severe and even fatal. Current efforts in precision immunotherapy are focused on improving biomarker-based patient selection for currently available ICIs and exploring rationale combination and novel strategies to expand the benefit of immunotherapy to more cancer patients. Neoantigens arise from ~ 10% of the non-synonymous somatic mutations in cancer cells, are important targets of T cell-mediated anti-tumor immunity for individual patients. Advances in next generation sequencing technology and computational bioinformatics have enable the identification of genomic alterations, putative neoantigens, and gene expression profiling in individual tumors for personal oncology in a rapid and cost-effective way. Among the genomic biomarkers, defective mismatch DNA repair (dMMR), microsatellite instability high (MSI-H) and high tumor mutational burden (H-TMB) have received FDA approvals for selecting patients for ICI treatment. All these biomarkers measure high neoantigen load and tumor antigenicity, supporting the current development of neoantigen-based personalized cancer vaccines for patients with high TMB tumor. Several studies have shown neoantigen vaccines are feasible, safe and have promising clinical activity in patients with high TMB tumors in both metastatic and adjuvant settings. This review summarizes the emerging data and technologies for neoantigen-based personalized immunotherapy.
Collapse
|
39
|
Zhang X, Zhong L, Zou Z, Liang G, Tang Z, Li K, Tan S, Huang Y, Zhu X. Clinical and Prognostic Pan-Cancer Analysis of N6-Methyladenosine Regulators in Two Types of Hematological Malignancies: A Retrospective Study Based on TCGA and GTEx Databases. Front Oncol 2021; 11:623170. [PMID: 33816257 PMCID: PMC8015800 DOI: 10.3389/fonc.2021.623170] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 01/29/2021] [Indexed: 12/11/2022] Open
Abstract
N6-methyladenosine (m6A) is one of the most active modification factors of mRNA, which is closely related to cell proliferation, differentiation, and tumor development. Here, we explored the relationship between the pathogenesis of hematological malignancies and the clinicopathologic parameters. The datasets of hematological malignancies and controls were obtained from the TCGA [AML (n = 200), DLBCL (n = 48)] and GTEx [whole blood (n = 337), blood vascular artery (n = 606)]. We analyzed the m6A factor expression differences in normal tissue and tumor tissue and their correlations, clustered the express obvious clinical tumor subtypes, determined the tumor risk score, established Cox regression model, performed univariate and multivariate analysis on all datasets. We found that the AML patients with high expression of IGF2BP3, ALKBH5, and IGF2BP2 had poor survival, while the DLBCL patients with high expression of METTL14 had poor survival. In addition, "Total" datasets analysis revealed that IGF2BP1, ALKBH5, IGF2BP2, RBM15, METTL3, and ZNF217 were potential oncogenes for hematologic system tumors. Collectively, the expressions of some m6A regulators are closely related to the occurrence and development of hematologic system tumors, and the intervention of specific regulatory factors may lead to a breakthrough in the treatment in the future.
Collapse
Affiliation(s)
- Xiangsheng Zhang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Guangdong Medical University, Zhanjiang, China
| | - Liye Zhong
- Department of Hematology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhilin Zou
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Guangdong Medical University, Zhanjiang, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Guosheng Liang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Guangdong Medical University, Zhanjiang, China
| | - Zhenye Tang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Guangdong Medical University, Zhanjiang, China
| | - Kai Li
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Guangdong Medical University, Zhanjiang, China
| | - Shuzhen Tan
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Guangdong Medical University, Zhanjiang, China
| | - Yongmei Huang
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Guangdong Medical University, Zhanjiang, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
- The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China
| | - Xiao Zhu
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Guangdong Medical University, Zhanjiang, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
- The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
40
|
Xu JY, Liu MT, Tao T, Zhu X, Fei FQ. The role of gut microbiota in tumorigenesis and treatment. Biomed Pharmacother 2021; 138:111444. [PMID: 33662679 DOI: 10.1016/j.biopha.2021.111444] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/17/2022] Open
Abstract
A large number of microbial communities exist in normal human intestinal tracts, which maintain a relatively stable dynamic balance under certain conditions. Gut microbiota are closely connected with human health and the occurrence of tumors. The colonization of certain intestinal bacteria on specific sites, gut microbiota disturbance and intestinal immune disorders can induce the occurrence of tumors. Meanwhile, gut microbiota can also play a role in tumor therapy by participating in immune regulation, influencing the efficacy of anti-tumor drugs, targeted therapy of engineered probiotics and fecal microbiota transplantation. This article reviews the role of gut microbiota in the occurrence, development, diagnosis and treatment of tumors. A better understanding of how gut microbiota affect tumors will help us find more therapies to treat the disease.
Collapse
Affiliation(s)
- Jia-Yi Xu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Min-Ting Liu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Tao Tao
- Department of Gastroenterology, Zibo Central Hospital, Zibo, China
| | - Xiao Zhu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.
| | - Fang-Qin Fei
- Department of Endocrinology, the First Affiliated Hospital of Huzhou University, Huzhou, China.
| |
Collapse
|
41
|
Polymer-based hydrogels with local drug release for cancer immunotherapy. Biomed Pharmacother 2021; 137:111333. [PMID: 33571834 DOI: 10.1016/j.biopha.2021.111333] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 12/23/2022] Open
Abstract
Immunotherapy that boosts the body's immune system to treat local and distant metastatic tumors has offered a new treatment option for cancer. However, cancer immunotherapy via systemic administration of immunotherapeutic agents often has two major issues of limited immune responses and potential immune-related adverse events in the clinic. Hydrogels, a class of three-dimensional network biomaterials with unique porous structures can achieve local delivery of drugs into tumors to trigger the antitumor immunity, resulting in amplified immunotherapy at lower dosages. In this review, we summarize the recent development of polymer-based hydrogels as drug release systems for local delivery of various immunotherapeutic agents for cancer immunotherapy. The constructions of polymer-based hydrogels and their local delivery of various drugs in tumors to achieve sole immunotherapy, and chemotherapy-, and phototherapy-combinational immunotherapy are introduced. Furthermore, a brief conclusion is given and existing challenges and further perspectives of polymer-based hydrogels for cancer immunotherapy are discussed.
Collapse
|
42
|
Cancer Vaccines: Antigen Selection Strategy. Vaccines (Basel) 2021; 9:vaccines9020085. [PMID: 33503926 PMCID: PMC7911511 DOI: 10.3390/vaccines9020085] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Unlike traditional cancer therapies, cancer vaccines (CVs) harness a high specificity of the host’s immunity to kill tumor cells. CVs can train and bolster the patient’s immune system to recognize and eliminate malignant cells by enhancing immune cells’ identification of antigens expressed on cancer cells. Various features of antigens like immunogenicity and avidity influence the efficacy of CVs. Therefore, the choice and application of antigens play a critical role in establishing and developing CVs. Tumor-associated antigens (TAAs), a group of proteins expressed at elevated levels in tumor cells but lower levels in healthy normal cells, have been well-studied and developed in CVs. However, immunological tolerance, HLA restriction, and adverse events are major obstacles that threaten TAA-based CVs’ efficacy due to the “self-protein” characteristic of TAAs. As “abnormal proteins” that are completely absent from normal cells, tumor-specific antigens (TSAs) can trigger a robust immune response against tumor cells with high specificity and without going through central tolerance, contributing to cancer vaccine development feasibility. In this review, we focus on the unique features of TAAs and TSAs and their application in vaccines, summarizing their performance in preclinical and clinical trials.
Collapse
|
43
|
How wide is the application of genetic big data in biomedicine. Biomed Pharmacother 2020; 133:111074. [PMID: 33378973 DOI: 10.1016/j.biopha.2020.111074] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/16/2020] [Accepted: 11/27/2020] [Indexed: 12/17/2022] Open
Abstract
In the era of big data, massive genetic data, as a new industry, has quickly swept almost all industries, especially the pharmaceutical industry. As countries around the world start to build their own gene banks, scientists study the data to explore the origins and migration of humans. Moreover, big data encourage the development of cancer therapy and bring good news to cancer patients. Big datum has been involved in the study of many diseases, and it has been found that analyzing diseases at the gene level can lead to more beneficial treatment options than ordinary treatments. This review will introduce the development of extensive data in medical research from the perspective of big data and tumor, neurological and psychiatric diseases, cardiovascular diseases, other applications and the development direction of big data in medicine.
Collapse
|
44
|
Zhu X, Li S, Xu B, Luo H. Cancer evolution: A means by which tumors evade treatment. Biomed Pharmacother 2020; 133:111016. [PMID: 33246226 DOI: 10.1016/j.biopha.2020.111016] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 11/07/2020] [Accepted: 11/11/2020] [Indexed: 12/17/2022] Open
Abstract
Although various methods have been tried to study and treat cancer, the cancer remains a major challenge for human medicine today. One important reason for this is the presence of cancer evolution. Cancer evolution is a process in which tumor cells adapt to the external environment, which can suppress the human immune system's ability to recognize and attack tumors, and also reduce the reproducibility of cancer research. Among them, heterogeneity of the tumor provides intrinsic motivation for this process. Recently, with the development of related technologies such as liquid biopsy, more and more knowledge about cancer evolution has been gained and interest in this topic has also increased. Therefore, starting from the causes of tumorigenesis, this paper introduces several tumorigenesis processes and pathways, as well as treatment options for different targets.
Collapse
Affiliation(s)
- Xiao Zhu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China.
| | - Shi Li
- Guangdong Key Laboratory of Urogenital Tumor Systems and Synthetic Biology, The First Affiliated Hospital of Shenzhen University, The Second People's Hospital of Shenzhen, Shenzhen, China; Shenzhen Key Laboratory of Genitourinary Tumor, Translational Medicine Institute of Shenzhen, The Second People's Hospital of Shenzhen, Shenzhen, China; College of Bioengineering, Chongqing University, Chongqing, China
| | - Bairui Xu
- The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjian, China
| | - Hui Luo
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjian, China.
| |
Collapse
|
45
|
Lin B, Du L, Li H, Zhu X, Cui L, Li X. Tumor-infiltrating lymphocytes: Warriors fight against tumors powerfully. Biomed Pharmacother 2020; 132:110873. [PMID: 33068926 DOI: 10.1016/j.biopha.2020.110873] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/30/2020] [Accepted: 10/07/2020] [Indexed: 12/18/2022] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) are infiltrating lymphocytes in tumor tissues. After isolation, screening and amplification in vitro, they will be implanted into patients and play a specific killing effect on tumors. Since TILs have not been genetically modified and come from the body of patients, there will be relatively few adverse reactions. This is also the advantage of TIL treatment. In recent years, its curative effect on solid tumors began to show its sharpness. However, due to the limitations of the immune microenvironment and the mutation of antigens, TIL's development was slowed down. This article reviews the research progress, biological characteristics, preparation and methods of enhancing the therapeutic effect of tumor-infiltrating lymphocytes, their roles in different tumors and prognosis, and emphasizes the important value of tumor-infiltrating lymphocytes in anti-tumor.
Collapse
Affiliation(s)
- Baisheng Lin
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Likun Du
- First Affiliated Hospital, Heilongjiang University of Traditional Chinese Medicine, Harbin, 150040, China
| | - Hongmei Li
- Department of Pathology, Guangdong Medical University, Dongguan, China
| | - Xiao Zhu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China.
| | - Liao Cui
- Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Xiaosong Li
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|