1
|
Zaccariotto GDC, Bistaffa MJ, Zapata AMM, Rodero C, Coelho F, Quitiba JV, Lima L, Sterman R, Cardoso VDO, Zucolotto V. Cancer Nanovaccines: Mechanisms, Design Principles, and Clinical Translation. ACS NANO 2025; 19:16204-16223. [PMID: 40202241 PMCID: PMC12060653 DOI: 10.1021/acsnano.4c15765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 04/01/2025] [Accepted: 04/01/2025] [Indexed: 04/10/2025]
Abstract
Cancer immunotherapy has transformed the landscape of oncological treatment by employing various strategies to teach the immune system to eliminate tumors. Among these, cancer nanovaccines are an emerging strategy that utilizes nanotechnology to enhance immune activation in response to tumor antigens. This review addresses the principles behind the different technologies in this field aimed at generating a robust and effective immune response. The diversity of strategies adopted for the design of nanovaccines is discussed, including the types of active agents, nanocarriers, their functionalizations, and the incorporation of adjuvants. Furthermore, strategies to optimize nanoparticle formulations to enhance the antigen presentation, target immune cells, and organs and promote strong and durable antitumor responses are explored. Finally, we analyze the current state of clinical application, highlighting ongoing clinical trials and the future potential of cancer nanovaccines. The insights presented in this review aim to guide future research and development efforts in the field, contributing to the advancement of more effective and targeted nanovaccines in the fight against cancer.
Collapse
Affiliation(s)
- Gabriel de Camargo Zaccariotto
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Maria Julia Bistaffa
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Angelica Maria Mazuera Zapata
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Camila Rodero
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Fernanda Coelho
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - João Victor
Brandão Quitiba
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Lorena Lima
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | - Raquel Sterman
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| | | | - Valtencir Zucolotto
- Nanomedicine
and Nanotoxicology Group, São Carlos Institute of Physics, University of São Paulo, São Paulo 13566-590, Brazil
| |
Collapse
|
2
|
Alghazali T, Ahmed AT, Hussein UAR, Sanghvi G, Uthirapathy S, Edan RT, Lal M, Shit D, Naidu KS, Al-Hamairy AK. Noncoding RNA (ncRNA)-mediated regulation of TLRs: critical regulator of inflammation in tumor microenvironment. Med Oncol 2025; 42:144. [PMID: 40163200 DOI: 10.1007/s12032-025-02690-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/13/2025] [Indexed: 04/02/2025]
Abstract
Toll-like receptors (TLRs) are central components of the innate immune system as they recognize molecular patterns associated with pathogens and cellular damage and initiate immune responses using MyD88- and TRIF-dependent pathways. In contrast to being very useful for immune defense, dysregulated TLR signaling may be involved in diseases, such as cancer and autoimmune conditions. In cancer, TLRs create an environment that supports tumorigenesis and growth. In addition to this, a class of multifunctional noncoding RNAs (ncRNAs), including miRNAs, lncRNAs, and circRNAs, regulate gene expression without encoding proteins. MiRNAs regulate gene expression in a fine-tuned manner, while lncRNAs and circRNAs do so via diverse mechanisms. Notably, these ncRNAs interact, where lncRNAs and circRNAs function as competing endogenous RNAs and ceRNA, affecting miRNA activity. This interaction has a vital role in cancer pathology, in influencing that of various oncogenes and tumor suppressors in the tumor microenvironment; hence, modulation of ncRNAs could also be a great promising therapeutic approach. In this context, interplay between TLRs and ncRNAs is of paramount importance as they influence various parameters of the tumor microenvironment. TLR signaling works upon the expression of ncRNAs, while ncRNAs work back to regulate TLR signaling in return. An example of this includes miRNA targeting of components of the TLR; lncRNAs induced by TLR signaling possibly would favor tumor progression. Pharmacological interventions directed toward inhibiting these TLR pathways could be the model to halt malignancy by hampering pro-tumor inflammation and boosting immune responses against neoplasms. Hence, the review will highlight the complicated contrast of ncRNAs and TLRs within human cancer. By connecting the mechanisms, the researchers may study more about tumorigenesis and gather up new, innovative notions regarding therapeutic targeting.
Collapse
Affiliation(s)
| | | | | | - Gaurav Sanghvi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, 360003, Gujarat, India
| | - Subasini Uthirapathy
- Pharmacy Department, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Reem Turki Edan
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | - Madan Lal
- Department of Medicine, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Debasish Shit
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Ahmed Khudhair Al-Hamairy
- Anesthesia Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| |
Collapse
|
3
|
Ji Y, Xiao C, Fan T, Deng Z, Wang D, Cai W, Li J, Liao T, Li C, He J. The epigenetic hallmarks of immune cells in cancer. Mol Cancer 2025; 24:66. [PMID: 40038722 PMCID: PMC11881328 DOI: 10.1186/s12943-025-02255-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 01/30/2025] [Indexed: 03/06/2025] Open
Abstract
Targeting the dysregulation of epigenetic mechanisms in cancer has emerged as a promising therapeutic strategy. Although the significant rationale progress of epigenetic therapies in blocking cancer cells, how epigenetic regulation shapes tumor microenvironment (TME) and establishes antitumor immunity remains less understood. Recent study focus has been put on the epigenetic-mediated changes in the fate of immune cells, including the differentiation, expansion, recruitment, functionalization, and exhaustion of T cells, natural killer (NK) cells, tumor-associated macrophages (TAMs), dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs), and B cells within the TME. Here, we review the latest molecular and clinical insights into how DNA modifications, histone modification, and epitranscriptome-related regulations shape immune cells of various cancers. We also discuss opportunities for leveraging epigenetic therapies to improve cancer immunotherapies. This review provides the epigenetic foundations of cancer immunity and proposes the future direction of combination therapies.
Collapse
Affiliation(s)
- Yu Ji
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- 4+4 Medical Doctor Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Di Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wenpeng Cai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jia Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tianle Liao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
4
|
Afra F, Eftekhar SP, Farid AS, Ala M. Non-coding RNAs in cancer immunotherapy: A solution to overcome immune resistance? PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 209:215-240. [PMID: 39461753 DOI: 10.1016/bs.pmbts.2024.02.003] [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: 10/29/2024]
Abstract
With the rapid advancement in immunotherapy, cancer immune resistance has become more evident, which demands new treatment approaches to achieve greater efficacy. Non-coding RNAs (ncRNAs) are a heterogeneous group of RNAs that are not translated to proteins but instead regulate different stages of gene expression. Recent studies have increasingly supported the critical role of ncRNAs in immune cell-cancer cell cross-talk, and numerous ncRNAs have been implicated in the immune evasion of cancer cells. Cancer cells take advantage of ncRNAs to modulate several signaling pathways and upregulate the expression of immune checkpoints and anti-inflammatory mediators, thereby dampening the anti-tumor response of M1 macrophages, dendritic cells, cytotoxic T cells, and natural killer cells or potentiating the immunosuppressive properties of M2 macrophages, regulatory T cells, and myeloid-derived suppressive cells. Upregulation of immunosuppressive ncRNAs or downregulation of immunogenic ncNRAs is a major driver of resistance to immune checkpoint inhibitors, cancer vaccines, and other means of cancer immunotherapy, making ncRNAs ideal targets for treatment. In addition, ncRNAs released by cancer cells have been demonstrated to possess prognostic values for patients who undergo cancer immunotherapy. Future clinical trials are urged to consider the potential of ncRNAs in cancer immunotherapy.
Collapse
Affiliation(s)
- Fatemeh Afra
- Clinical Pharmacy Department, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Parsa Eftekhar
- Student Research Committee, Health Research Center, Babol University of Medical Sciences, Babol, Iran
| | - Amir Salehi Farid
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Moein Ala
- Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
5
|
Li S, Zhang J, Ma M, Zhang M, Li L, Chen W, Li S. NaAsO 2 regulates TLR4/MyD88/NF-κB signaling pathway through DNMT1/SOCS1 to cause apoptosis and inflammation in hepatic BRL-3A cells. Biol Trace Elem Res 2024; 202:258-267. [PMID: 36988786 DOI: 10.1007/s12011-023-03648-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 03/22/2023] [Indexed: 03/30/2023]
Abstract
The exact molecular mechanism of arsenic-induced liver injury has not been fully elucidated. The aim of the study was to investigate the potential mechanism of NaAsO2-induced cytotoxicity in BRL-3A cells and to provide a basis for the mechanism of arsenic poisoning. BRL-3A cells were treated with different doses of NaAsO2, DNMT1 inhibitor (DC_517), TLR4 inhibitor (TAK-242), and transfection of SOCS1 plasmid. Cell activity, apoptosis, inflammation and protein expression of DNMT1, SOCS1, TLR4, MyD88, and NF-κB were detected by CCK8 assay, Annexin V-FITC and Western blot, respectively. With increasing NaAsO2 doses, BAX and caspase-3 expression increased, Bcl-2 expression decreased, pro-inflammatory factors TNF-α, IL-1β, and IL-6 increased, and cell activity decreased causing increased apoptosis. When BRL-3A was intervened with 10, and 20 μmol/L NaAsO2, DNMT1 expression was elevated, SOCS1 expression was decreased, and TLR4, MyD88, p-IκBα/IκBα, and p-p65/p65 expression were elevated. After the combination of NaAsO2 and DC_517, compared to the NaAsO2 group, apoptosis and inflammation were attenuated, SOCS1 expression was elevated and TLR4, MyD88, p-IκBα/IκBα and p-p65/p65 expression was decreased. Apoptosis and inflammation were attenuated after co-treatment of SOCS1 high expression with NaAsO2 compared to the NaAsO2 group. In addition, TLR4, MyD88, p-IκBα/IκBα and p-p65/p65 expression was reduced. When NaAsO2 and TAK-242 were combined, apoptosis and inflammation were attenuated. Besides MyD88, p-IκBα/IκBα and p-p65/p65 expression was reduced compared to the NaAsO2 group. We found that NaAsO2 induce apoptosis and inflammation in BLR-3A cells, which may be related to inhibit SOCS1 through regulation of DNMT1 and thus activating the TLR4/MyD88/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Sheng Li
- Department of Public Health, School of Medicine, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Jingyi Zhang
- Department of Public Health, School of Medicine, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Mingxiao Ma
- Department of Public Health, School of Medicine, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Mengyao Zhang
- Department of Public Health, School of Medicine, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Linzhi Li
- Department of Public Health, School of Medicine, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Weixin Chen
- Department of Public Health, School of Medicine, Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Shugang Li
- School of Public Health, Capital Medical University, No. 10 Xitoutiao, Youanmenwai, Beijing, China.
| |
Collapse
|
6
|
Liu R, Zhao E, Yu H, Yuan C, Abbas MN, Cui H. Methylation across the central dogma in health and diseases: new therapeutic strategies. Signal Transduct Target Ther 2023; 8:310. [PMID: 37620312 PMCID: PMC10449936 DOI: 10.1038/s41392-023-01528-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 05/23/2023] [Accepted: 05/25/2023] [Indexed: 08/26/2023] Open
Abstract
The proper transfer of genetic information from DNA to RNA to protein is essential for cell-fate control, development, and health. Methylation of DNA, RNAs, histones, and non-histone proteins is a reversible post-synthesis modification that finetunes gene expression and function in diverse physiological processes. Aberrant methylation caused by genetic mutations or environmental stimuli promotes various diseases and accelerates aging, necessitating the development of therapies to correct the disease-driver methylation imbalance. In this Review, we summarize the operating system of methylation across the central dogma, which includes writers, erasers, readers, and reader-independent outputs. We then discuss how dysregulation of the system contributes to neurological disorders, cancer, and aging. Current small-molecule compounds that target the modifiers show modest success in certain cancers. The methylome-wide action and lack of specificity lead to undesirable biological effects and cytotoxicity, limiting their therapeutic application, especially for diseases with a monogenic cause or different directions of methylation changes. Emerging tools capable of site-specific methylation manipulation hold great promise to solve this dilemma. With the refinement of delivery vehicles, these new tools are well positioned to advance the basic research and clinical translation of the methylation field.
Collapse
Affiliation(s)
- Ruochen Liu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China
- Jinfeng Laboratory, Chongqing, 401329, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, 400715, China
| | - Erhu Zhao
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China
- Jinfeng Laboratory, Chongqing, 401329, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, 400715, China
| | - Huijuan Yu
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China
| | - Chaoyu Yuan
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China
| | - Muhammad Nadeem Abbas
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China
- Jinfeng Laboratory, Chongqing, 401329, China
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, 400715, China
| | - Hongjuan Cui
- State Key Laboratory of Resource Insects, Medical Research Institute, Southwest University, Chongqing, 400715, China.
- Jinfeng Laboratory, Chongqing, 401329, China.
- Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Chongqing, 400716, China.
- Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
7
|
Esparcia-Pinedo L, Romero-Laorden N, Alfranca A. Tertiary lymphoid structures and B lymphocytes: a promising therapeutic strategy to fight cancer. Front Immunol 2023; 14:1231315. [PMID: 37622111 PMCID: PMC10445545 DOI: 10.3389/fimmu.2023.1231315] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/24/2023] [Indexed: 08/26/2023] Open
Abstract
Tertiary lymphoid structures (TLSs) are clusters of lymphoid cells with an organization that resembles that of secondary lymphoid organs. Both structures share common developmental characteristics, although TLSs usually appear in chronically inflamed non-lymphoid tissues, such as tumors. TLSs contain diverse types of immune cells, with varying degrees of spatial organization that represent different stages of maturation. These structures support both humoral and cellular immune responses, thus the correlation between the existence of TLS and clinical outcomes in cancer patients has been extensively studied. The finding that TLSs are associated with better prognosis in some types of cancer has led to the design of therapeutic strategies based on promoting the formation of these structures. Agents such as chemokines, cytokines, antibodies and cancer vaccines have been used in combination with traditional antitumor treatments to enhance TLS generation, with good results. The induction of TLS formation therefore represents a novel and promising avenue for the treatment of a number of tumor types.
Collapse
Affiliation(s)
- Laura Esparcia-Pinedo
- Immunology Department, Hospital Universitario de La Princesa and Instituto de Investigación Sanitaria Princesa, Madrid, Spain
- Department of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Nuria Romero-Laorden
- Medical Oncology Department, Hospital Universitario de La Princesa and Instituto de Investigación Sanitaria Princesa, Madrid, Spain
- Cátedra Universidad Autónoma de Madrid (UAM)-Fundación Instituto Roche de Medicina Personalizada de Precisión, Madrid, Spain
| | - Arantzazu Alfranca
- Immunology Department, Hospital Universitario de La Princesa and Instituto de Investigación Sanitaria Princesa, Madrid, Spain
- Department of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Cátedra Universidad Autónoma de Madrid (UAM)-Fundación Instituto Roche de Medicina Personalizada de Precisión, Madrid, Spain
- Centro de Investigación Biomédica en Red Cardiovascular, CIBERCV, Madrid, Spain
| |
Collapse
|
8
|
Xie Z, Zhou Z, Yang S, Zhang S, Shao B. Epigenetic regulation and therapeutic targets in the tumor microenvironment. MOLECULAR BIOMEDICINE 2023; 4:17. [PMID: 37273004 DOI: 10.1186/s43556-023-00126-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 04/02/2023] [Indexed: 06/06/2023] Open
Abstract
The tumor microenvironment (TME) is crucial to neoplastic processes, fostering proliferation, angiogenesis and metastasis. Epigenetic regulations, primarily including DNA and RNA methylation, histone modification and non-coding RNA, have been generally recognized as an essential feature of tumor malignancy, exceedingly contributing to the dysregulation of the core gene expression in neoplastic cells, bringing about the evasion of immunosurveillance by influencing the immune cells in TME. Recently, compelling evidence have highlighted that clinical therapeutic approaches based on epigenetic machinery modulate carcinogenesis through targeting TME components, including normalizing cells' phenotype, suppressing cells' neovascularization and repressing the immunosuppressive components in TME. Therefore, TME components have been nominated as a promising target for epigenetic drugs in clinical cancer management. This review focuses on the mechanisms of epigenetic modifications occurring to the pivotal TME components including the stroma, immune and myeloid cells in various tumors reported in the last five years, concludes the tight correlation between TME reprogramming and tumor progression and immunosuppression, summarizes the current advances in cancer clinical treatments and potential therapeutic targets with reference to epigenetic drugs. Finally, we summarize some of the restrictions in the field of cancer research at the moment, further discuss several interesting epigenetic gene targets with potential strategies to boost antitumor immunity.
Collapse
Affiliation(s)
- Zhuojun Xie
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, South Renmin Road, Sichuan, 610041, Chengdu, China
| | - Zirui Zhou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, South Renmin Road, Sichuan, 610041, Chengdu, China
| | - Shuxian Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, South Renmin Road, Sichuan, 610041, Chengdu, China
| | - Shiwen Zhang
- Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, South Renmin Road, Sichuan, 610041, Chengdu, China.
| | - Bin Shao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No. 14, Section 3, South Renmin Road, Sichuan, 610041, Chengdu, China.
| |
Collapse
|
9
|
Pathania AS. Crosstalk between Noncoding RNAs and the Epigenetics Machinery in Pediatric Tumors and Their Microenvironment. Cancers (Basel) 2023; 15:2833. [PMID: 37345170 DOI: 10.3390/cancers15102833] [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: 02/22/2023] [Revised: 05/08/2023] [Accepted: 05/10/2023] [Indexed: 06/23/2023] Open
Abstract
According to the World Health Organization, every year, an estimated 400,000+ new cancer cases affect children under the age of 20 worldwide. Unlike adult cancers, pediatric cancers develop very early in life due to alterations in signaling pathways that regulate embryonic development, and environmental factors do not contribute much to cancer development. The highly organized complex microenvironment controlled by synchronized gene expression patterns plays an essential role in the embryonic stages of development. Dysregulated development can lead to tumor initiation and growth. The low mutational burden in pediatric tumors suggests the predominant role of epigenetic changes in driving the cancer phenotype. However, one more upstream layer of regulation driven by ncRNAs regulates gene expression and signaling pathways involved in the development. Deregulation of ncRNAs can alter the epigenetic machinery of a cell, affecting the transcription and translation profiles of gene regulatory networks required for cellular proliferation and differentiation during embryonic development. Therefore, it is essential to understand the role of ncRNAs in pediatric tumor development to accelerate translational research to discover new treatments for childhood cancers. This review focuses on the role of ncRNA in regulating the epigenetics of pediatric tumors and their tumor microenvironment, the impact of their deregulation on driving pediatric tumor progress, and their potential as effective therapeutic targets.
Collapse
Affiliation(s)
- Anup S Pathania
- Department of Biochemistry and Molecular Biology & The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
10
|
Du Y, Shan C, You Y, Chen M, Zhu L, Shu G, Han G, Wu L, Ji J, Yu H, Du Y. NIR-II fluorescence imaging-guided hepatocellular carcinoma treatment via IR-1061-acridine and lenvatinib co-loaded thermal-sensitive micelles and anti-PD-1 combinational therapy. CHEMICAL ENGINEERING JOURNAL 2023; 454:140437. [DOI: 10.1016/j.cej.2022.140437] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
11
|
Tang WW, Bauer KM, Barba C, Ekiz HA, O’Connell RM. miR-aculous new avenues for cancer immunotherapy. Front Immunol 2022; 13:929677. [PMID: 36248881 PMCID: PMC9554277 DOI: 10.3389/fimmu.2022.929677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/18/2022] [Indexed: 01/25/2023] Open
Abstract
The rising toll of cancer globally necessitates ingenuity in early detection and therapy. In the last decade, the utilization of immune signatures and immune-based therapies has made significant progress in the clinic; however, clinical standards leave many current and future patients without options. Non-coding RNAs, specifically microRNAs, have been explored in pre-clinical contexts with tremendous success. MicroRNAs play indispensable roles in programming the interactions between immune and cancer cells, many of which are current or potential immunotherapy targets. MicroRNAs mechanistically control a network of target genes that can alter immune and cancer cell biology. These insights provide us with opportunities and tools that may complement and improve immunotherapies. In this review, we discuss immune and cancer cell-derived miRNAs that regulate cancer immunity and examine miRNAs as an integral part of cancer diagnosis, classification, and therapy.
Collapse
Affiliation(s)
- William W. Tang
- Divison of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, United States
- Hunstman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Kaylyn M. Bauer
- Divison of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, United States
- Hunstman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Cindy Barba
- Divison of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, United States
- Hunstman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| | - Huseyin Atakan Ekiz
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, İzmir, Turkey
| | - Ryan M. O’Connell
- Divison of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT, United States
- Hunstman Cancer Institute, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
12
|
Godoy-Tena G, Ballestar E. Epigenetics of Dendritic Cells in Tumor Immunology. Cancers (Basel) 2022; 14:cancers14051179. [PMID: 35267487 PMCID: PMC8909611 DOI: 10.3390/cancers14051179] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 12/14/2022] Open
Abstract
Dendritic cells (DCs) are professional antigen-presenting cells with the distinctive property of inducing the priming and differentiation of naïve CD4+ and CD8+ T cells into helper and cytotoxic effector T cells to develop efficient tumor-immune responses. DCs display pathogenic and tumorigenic antigens on their surface through major histocompatibility complexes to directly influence the differentiation of T cells. Cells in the tumor microenvironment (TME), including cancer cells and other immune-infiltrated cells, can lead DCs to acquire an immune-tolerogenic phenotype that facilitates tumor progression. Epigenetic alterations contribute to cancer development, not only by directly affecting cancer cells, but also by their fundamental role in the differentiation of DCs that acquire a tolerogenic phenotype that, in turn, suppresses T cell-mediated responses. In this review, we focus on the epigenetic regulation of DCs that have infiltrated the TME and discuss how knowledge of the epigenetic control of DCs can be used to improve DC-based vaccines for cancer immunotherapy.
Collapse
Affiliation(s)
- Gerard Godoy-Tena
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Barcelona, Spain;
| | - Esteban Ballestar
- Epigenetics and Immune Disease Group, Josep Carreras Research Institute (IJC), 08916 Barcelona, Spain;
- Epigenetics in Inflammatory and Metabolic Diseases Laboratory, Health Science Center (HSC), East China Normal University (ECNU), Shanghai 200241, China
- Correspondence:
| |
Collapse
|
13
|
Lamrayah M, Phelip C, Coiffier C, Lacroix C, Willemin T, Trimaille T, Verrier B. A Polylactide-Based Micellar Adjuvant Improves the Intensity and Quality of Immune Response. Pharmaceutics 2022; 14:pharmaceutics14010107. [PMID: 35057003 PMCID: PMC8778782 DOI: 10.3390/pharmaceutics14010107] [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: 11/16/2021] [Revised: 12/24/2021] [Accepted: 12/27/2021] [Indexed: 11/21/2022] Open
Abstract
Micelles from amphiphilic polylactide-block-poly(N-acryloxysuccinimide-co-N-vinylpyrrolidone) (PLA-b-P(NAS-co-NVP)) block copolymers of 105 nm in size were characterized and evaluated in a vaccine context. The micelles were non-toxic in vitro (both in dendritic cells and HeLa cells). In vitro fluorescence experiments combined with in vivo fluorescence tomography imaging, through micelle loading with the DiR near infrared probe, suggested an efficient uptake of the micelles by the immune cells. The antigenic protein p24 of the HIV-1 was successfully coupled on the micelles using the reactive N-succinimidyl ester groups on the micelle corona, as shown by SDS-PAGE analyses. The antigenicity of the coupled antigen was preserved and even improved, as assessed by the immuno-enzymatic (ELISA) test. Then, the performances of the micelles in immunization were investigated and compared to different p24-coated PLA nanoparticles, as well as Alum and MF59 gold standards, following a standardized HIV-1 immunization protocol in mice. The humoral response intensity (IgG titers) was substantially similar between the PLA micelles and all other adjuvants over an extended time range (one year). More interestingly, this immune response induced by PLA micelles was qualitatively higher than the gold standards and PLA nanoparticles analogs, expressed through an increasing avidity index over time (>60% at day 365). Taken together, these results demonstrate the potential of such small-sized micellar systems for vaccine delivery.
Collapse
Affiliation(s)
- Myriam Lamrayah
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
- Correspondence: (M.L.); (T.T.)
| | - Capucine Phelip
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| | - Céline Coiffier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| | - Céline Lacroix
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| | - Thibaut Willemin
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| | - Thomas Trimaille
- Laboratoire Ingénierie des Matériaux Polymères (IMP), CNRS UMR 5223, Université Lyon 1, Université de Lyon, 69622 Villeurbanne, France
- Correspondence: (M.L.); (T.T.)
| | - Bernard Verrier
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR 5305, Université Lyon 1, Université de Lyon, 69367 Lyon, France; (C.P.); (C.C.); (C.L.); (T.W.); (B.V.)
| |
Collapse
|
14
|
He T, Luo Y, Zhang Q, Men Z, Su T, Fan L, Chen H, Shen T. Hyalase-Mediated Cascade Degradation of a Matrix Barrier and Immune Cell Penetration by a Photothermal Microneedle for Efficient Anticancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:26790-26799. [PMID: 34061496 DOI: 10.1021/acsami.1c06725] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
For melanoma with high lethality and metastasis rate, traditional therapy has limited effects; local photothermal therapy (PTT) synergetic with immune therapy for cancer treatment can perhaps improve the situation. However, because of the natural existence of the tumor matrix barrier, the penetration depth of drugs and immune cells often dampens the efficacy of cancer treatment. Herein, we report an innovative synergetic PTT and immune therapy through dissolving microneedles for the codelivery of the hyaluronidase-modified semiconductor polymer nanoparticles containing poly(cyclopentadithiophene-alt-benzothiadiazole) and immune adjuvant polyinosinic-polycytidylic acid (PIC). Benefiting from the dissolution of an extracellular matrix of hyaluronidase, the semiconductor polymer nanoparticles and PIC penetrate the tumor deeply, under synergetic therapy with PTT, activating the immune cells and enhancing the T-cell immune response for inhibition of tumor growth and metastasis. This study provides a promising platform for effective melanoma treatment and a novel strategy to overcome the stromal barrier.
Collapse
Affiliation(s)
- Ting He
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, P.R. China
| | - Yu Luo
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, P.R. China
- Frontier Institute of Medical & Pharmaceutical Science and Technology, College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, No. 333 Longteng Road, Shanghai 201620, P.R. China
| | - Qin Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, P.R. China
| | - Zening Men
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, P.R. China
| | - Tong Su
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, P.R. China
| | - Linpeng Fan
- Institute for Frontier Materials, Deakin University, Geelong, Victoria 3216, Australia
| | - Hangrong Chen
- State Key Laboratory of High-Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai 200050, P.R. China
| | - Teng Shen
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University, No. 826 Zhangheng Road, Shanghai 201203, P.R. China
| |
Collapse
|
15
|
Xing Y, Ruan G, Ni H, Qin H, Chen S, Gu X, Shang J, Zhou Y, Tao X, Zheng L. Tumor Immune Microenvironment and Its Related miRNAs in Tumor Progression. Front Immunol 2021; 12:624725. [PMID: 34084160 PMCID: PMC8167795 DOI: 10.3389/fimmu.2021.624725] [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: 11/02/2020] [Accepted: 04/29/2021] [Indexed: 12/11/2022] Open
Abstract
MiRNA is a type of small non-coding RNA, by regulating downstream gene expression that affects the progression of multiple diseases, especially cancer. MiRNA can participate in the biological processes of tumor, including proliferation, invasion and escape, and exhibit tumor enhancement or inhibition. The tumor immune microenvironment contains numerous immune cells. These cells include lymphocytes with tumor suppressor effects such as CD8+ T cells and natural killer cells, as well as some tumor-promoting cells with immunosuppressive functions, such as regulatory T cells and myeloid-derived suppressor cells. MiRNA can affect the tumor immune microenvironment by regulating the function of immune cells, which in turn modulates the progression of tumor cells. Investigating the role of miRNA in regulating the tumor immune microenvironment will help elucidate the specific mechanisms of interaction between immune cells and tumor cells, and may facilitate the use of miRNA as a predictor of immune disorders in tumor progression. This review summarizes the multifarious roles of miRNA in tumor progression through regulation of the tumor immune microenvironment, and provides guidance for the development of miRNA drugs to treat tumors and for the use of miRNA as an auxiliary means in tumor immunotherapy.
Collapse
Affiliation(s)
- Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Aoyama S, Nakagawa R, Mulé JJ, Mailloux AW. Inducible Tertiary Lymphoid Structures: Promise and Challenges for Translating a New Class of Immunotherapy. Front Immunol 2021; 12:675538. [PMID: 34054863 PMCID: PMC8160316 DOI: 10.3389/fimmu.2021.675538] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/27/2021] [Indexed: 12/12/2022] Open
Abstract
Tertiary lymphoid structures (TLS) are ectopically formed aggregates of organized lymphocytes and antigen-presenting cells that occur in solid tissues as part of a chronic inflammation response. Sharing structural and functional characteristics with conventional secondary lymphoid organs (SLO) including discrete T cell zones, B cell zones, marginal zones with antigen presenting cells, reticular stromal networks, and high endothelial venues (HEV), TLS are prominent centers of antigen presentation and adaptive immune activation within the periphery. TLS share many signaling axes and leukocyte recruitment schemes with SLO regarding their formation and function. In cancer, their presence confers positive prognostic value across a wide spectrum of indications, spurring interest in their artificial induction as either a new form of immunotherapy, or as a means to augment other cell or immunotherapies. Here, we review approaches for inducible (iTLS) that utilize chemokines, inflammatory factors, or cellular analogues vital to TLS formation and that often mirror conventional SLO organogenesis. This review also addresses biomaterials that have been or might be suitable for iTLS, and discusses remaining challenges facing iTLS manufacturing approaches for clinical translation.
Collapse
Affiliation(s)
- Shota Aoyama
- Department of Surgery, Institute of Gastroenterology, Tokyo Women’s Medical University, Tokyo, Japan
| | - Ryosuke Nakagawa
- Department of Surgery, Institute of Gastroenterology, Tokyo Women’s Medical University, Tokyo, Japan
| | - James J. Mulé
- Immunology Program, Moffitt Cancer Center, Tampa, FL, United States
- Cutaneous Oncology Program, Moffitt Cancer Center, Tampa, FL, United States
| | - Adam W. Mailloux
- Department of Microbiology and Immunology, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
17
|
Xing Y, Wang Z, Lu Z, Xia J, Xie Z, Jiao M, Liu R, Chu Y. MicroRNAs: immune modulators in cancer immunotherapy. IMMUNOTHERAPY ADVANCES 2021; 1:ltab006. [PMID: 35919742 PMCID: PMC9327120 DOI: 10.1093/immadv/ltab006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/18/2021] [Accepted: 05/07/2021] [Indexed: 12/27/2022] Open
Abstract
Summary
MicroRNA (miRNA) is a class of endogenous small non-coding RNA of 18–25 nucleotides and plays regulatory roles in both physiological and pathological processes. Emerging evidence support that miRNAs function as immune modulators in tumors. MiRNAs as tumor suppressors or oncogenes are also found to be able to modulate anti-tumor immunity or link the crosstalk between tumor cells and immune cells surrounding. Based on the specific regulating function, miRNAs can be used as predictive, prognostic biomarkers, and therapeutic targets in immunotherapy. Here, we review new findings about the role of miRNAs in modulating immune responses, as well as discuss mechanisms underlying their dysregulation, and their clinical potentials as indicators of tumor prognosis or to sensitize cancer immunotherapy.
Collapse
Affiliation(s)
- Yun Xing
- Shanghai Fifth People’s Hospital and Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Zhiqiang Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Zhou Lu
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
- Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai 200032, P.R. China
| | - Jie Xia
- Shanghai Fifth People’s Hospital and Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Zhangjuan Xie
- Shanghai Fifth People’s Hospital and Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Mengxia Jiao
- Shanghai Fifth People’s Hospital and Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Ronghua Liu
- Shanghai Fifth People’s Hospital and Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
| | - Yiwei Chu
- Shanghai Fifth People’s Hospital and Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, P.R. China
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, P.R. China
| |
Collapse
|
18
|
Wang Y, Lu C, Huang H, Yao S, Xu C, Ye Y, Gui S, Li G. A lipid-soluble extract of Pinellia pedatisecta Schott orchestrates intratumoral dendritic cell-driven immune activation through SOCS1 signaling in cervical cancer. JOURNAL OF ETHNOPHARMACOLOGY 2021; 267:112837. [PMID: 32276009 DOI: 10.1016/j.jep.2020.112837] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/14/2020] [Accepted: 04/01/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pinellia pedatisecta Schott extract (PE) is generated from Pinellia pedatisecta Schott, a traditional Chinese medicinal plant. PE suppresses cervical tumor growth and exhibits effects on dendritic cells (DCs) that lead to modulation of antitumor CD4+ and CD8+ responses. AIMS To explore the underlying mechanisms by which PE modulates tumor-associated dendritic cell (TADC) activation and function. METHODS DCs and TADCs were generated from murine bone marrow and exposed to PE solutions at different doses, as well as to repeated doses separated at different time intervals. Quantitative PCR, Western blot analysis, flow cytometry, and gene silencing were used to analyze the modulatory effects of PE on the SOCS1/JAK2/STAT pathways. Furthermore, we separated human cervical tumor-infiltrated DCs (TIDCs) and conducted an ex-vivo stimulation model to observe the effect of PE. For phenotypic analysis of cultured DCs and ex vivo human specimens, we used flow cytometry to detect the molecular markers associated with cell function. RESULTS In cultured TADCs and human cervical TIDCs, maturation- and functional markers (MHCII, CD80, CD83, CD86, and IL-12) were downregulated, whereas SOCS1 was upregulated. PE enhanced the expression of CD80, CD86, and IL-12 in cervical TIDCs, which induced increased expression of CD107a, GZMB, and perforin in CTLs, and furthermore induced apoptosis in a larger number of tumor cells. In cultured TADCs, PE downregulated SOCS1 expression and activated the phosphorylation of JAK2, STAT1, STAT4, and STAT5 in both dose- and time-dependent manners. The effects of PE upregulating MHCII, CD80, CD86, IL-12 on TADCs were blocked after SOCS1 silencing. CONCLUSIONS In this study, PE restored the impaired function of cervical TIDCs, thereby eliciting further antitumor CTL responses. The effects of PE on TADCs were mediated through inhibition of SOCS1 and activation of downstream JAK2-STAT1/STAT4/STAT5 pathways. PE may be a potent and effective immunomodulatory drug for antitumor treatment via the blockade of SOCS1 signaling in DCs.
Collapse
Affiliation(s)
- Yumeng Wang
- Department of Integration of Western and Traditional Medicine, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200090, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, 200011, China
| | - Chong Lu
- Department of Integration of Western and Traditional Medicine, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200090, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, 200011, China
| | - Haixia Huang
- Department of Integration of Western and Traditional Medicine, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200090, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, 200011, China
| | - Sheng Yao
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Congjian Xu
- Department of Integration of Western and Traditional Medicine, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200090, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, 200011, China
| | - Yang Ye
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Suiqi Gui
- Department of Integration of Western and Traditional Medicine, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200090, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, 200011, China
| | - Guiling Li
- Department of Integration of Western and Traditional Medicine, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, 200090, China; Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Fudan University, Shanghai, 200011, China.
| |
Collapse
|
19
|
Tittarelli A, Navarrete M, Lizana M, Hofmann-Vega F, Salazar-Onfray F. Hypoxic Melanoma Cells Deliver microRNAs to Dendritic Cells and Cytotoxic T Lymphocytes through Connexin-43 Channels. Int J Mol Sci 2020; 21:ijms21207567. [PMID: 33066331 PMCID: PMC7589225 DOI: 10.3390/ijms21207567] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/05/2020] [Accepted: 10/09/2020] [Indexed: 12/13/2022] Open
Abstract
Alterations in microRNA (miRNA) profiles, induced by tumor microenvironment stressors, like hypoxia, allow cancer cells to acquire immune-resistance phenotypes. Indeed, hypoxia-induced miRNAs have been implicated in cancer progression through numerous cancer cell non-autonomous mechanisms, including the direct transfer of hypoxia-responsive miRNA from cancer to immune cells via extracellular vesicles. Connexin-43 (Cx43)-constituted gap junctions (GJs) have also been involved in miRNA intercellular mobilization, in other biological processes. In this report, we aimed to evaluate the involvement of Cx43-GJs in the shift of miRNAs induced by hypoxia, from hypoxic melanoma cells to dendritic cells and melanoma-specific cytotoxic T lymphocytes (CTLs). Using qRT-PCR arrays, we identified that miR-192-5p was strongly induced in hypoxic melanoma cells. Immune cells acquired this miRNA after co-culture with hypoxic melanoma cells. The transfer of miR-192-5p was inhibited when hypoxic melanoma cells expressed a dominant negative Cx43 mutant or when Cx43 expression was silenced using specific short-hairpin RNAs. Interestingly, miR-192-5p levels on CTLs after co-culture with hypoxic melanoma cells were inversely correlated with the cytotoxic activity of T cells and with ZEB2 mRNA expression, a validated immune-related target of miR-192-5p, which is also observed in vivo. Altogether, our data suggest that hypoxic melanoma cells may suppress CTLs cytotoxic activity by transferring hypoxia-induced miR-192-5p through a Cx43-GJs driven mechanism, constituting a resistance strategy for immunological tumor escape.
Collapse
Affiliation(s)
- Andrés Tittarelli
- Programa Institucional de Fomento a la Investigación, Desarrollo e Innovación (PIDi), Universidad Tecnológica Metropolitana (UTEM), Santiago 8940577, Chile
- Correspondence: ; Tel.: +56-2-2787-7903
| | - Mariela Navarrete
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (M.N.); (M.L.); (F.H.-V.); (F.S.-O.)
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| | - Marcelo Lizana
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (M.N.); (M.L.); (F.H.-V.); (F.S.-O.)
| | - Francisca Hofmann-Vega
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (M.N.); (M.L.); (F.H.-V.); (F.S.-O.)
| | - Flavio Salazar-Onfray
- Disciplinary Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile; (M.N.); (M.L.); (F.H.-V.); (F.S.-O.)
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile
| |
Collapse
|
20
|
Mei X, Shi W, Zhao W, Luo H, Zhang Y, Wang Y, Sheng Z, Wang D, Zhu XQ, Huang W. Fasciola gigantica excretory-secretory products (FgESPs) modulate the differentiation and immune functions of buffalo dendritic cells through a mechanism involving DNMT1 and TET1. Parasit Vectors 2020; 13:355. [PMID: 32680546 PMCID: PMC7368760 DOI: 10.1186/s13071-020-04220-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/07/2020] [Indexed: 12/15/2022] Open
Abstract
Background Fasciola gigantica infection threatens the health of both humans and animals in the world. The excretory/secretory products (ESPs) of this fluke has been reported to impair the activation and maturation of immune cells. We have previously shown the influence of F. gigantica ESPs (FgESPs) on the maturation of buffalo dendritic cells (DCs). However, the underlying mechanisms remain unclear. The objective of this study was to investigate the potency of FgESPs in shifting the differentiation and immune functions of buffalo DCs. Methods Buffalo DCs were incubated with FgESPs directly or further co-cultured with lymphocytes in vitro. qRT-PCR was employed to determine the gene expression profile of DCs or the mixed cells, and an ELISA was used to measure cytokine levels in the supernatants. Hoechst and Giemsa staining assays, transmission electron microscopy, caspase-3/7 activity test and histone methylation test were performed to determine DC phenotyping, apoptosis and methylation. To investigate the mechanism involved with DNA methylation, a Co-IP assay and immunofluorescent staining assay were performed to observe if there was any direct interaction between FgESPs and DNMT1/TET1 in buffalo DCs, while RNAi technology was employed to knockdown DNMT1 and TET1 in order to evaluate any different influence of FgESPs on DCs when these genes were absent. Results qRT-PCR and ELISA data together demonstrated the upregulation of DC2 and Th2/Treg markers in DCs alone and DCs with a mixed lymphocyte reaction (MLR), suggesting a bias of DC2 that potentially directed Th2 differentiation in vitro. DC apoptosis was also found and evidenced morphologically and biochemically, which might be a source of tolerogenic DCs that led to Treg differentiation. In addition, FgESPs induced methylation level changes of histones H3K4 and H3K9, which correlate with DNA methylation. Co-IP and immunofluorescent subcellular localization assays showed no direct interaction between the FgESPs and DNMT1/TET1 in buffalo DCs. The productions of IL-6 and IL-12 were found separately altered by the knockdown of DNMT1 and TET1 in DCs after FgESPs treatment. Conclusions FgESPs may induce the DC2 phenotype or the apoptosis of buffalo DCs to induce the downstream Th2/Treg response of T cells, possibly through a DNMT1- or TET1-dependent manner(s).![]()
Collapse
Affiliation(s)
- Xuefang Mei
- School of Animal Science and Technology, Guangxi University, Nanning, 530005, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Wei Shi
- School of Preclinical Medicine, Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Wenping Zhao
- School of Animal Science and Technology, Guangxi University, Nanning, 530005, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Honglin Luo
- Guangxi Key Laboratory for Aquatic Genetic Breeding and Healthy Aquaculture, Guangxi Institute of Fishery Sciences, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Yaoyao Zhang
- School of Animal Science and Technology, Guangxi University, Nanning, 530005, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Yurui Wang
- School of Animal Science and Technology, Guangxi University, Nanning, 530005, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Zhaoan Sheng
- School of Animal Science and Technology, Guangxi University, Nanning, 530005, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Dongying Wang
- School of Animal Science and Technology, Guangxi University, Nanning, 530005, Guangxi Zhuang Autonomous Region, People's Republic of China.
| | - Xing-Quan Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Veterinary Parasitology of Gansu Province, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, Gansu, People's Republic of China. .,Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University College of Veterinary Medicine, Yangzhou, 225009, Jiangsu, People's Republic of China.
| | - Weiyi Huang
- School of Animal Science and Technology, Guangxi University, Nanning, 530005, Guangxi Zhuang Autonomous Region, People's Republic of China.
| |
Collapse
|
21
|
Yang F, Shi K, Jia YP, Hao Y, Peng JR, Qian ZY. Advanced biomaterials for cancer immunotherapy. Acta Pharmacol Sin 2020; 41:911-927. [PMID: 32123302 PMCID: PMC7468530 DOI: 10.1038/s41401-020-0372-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/27/2020] [Indexed: 02/05/2023]
Abstract
Immunotherapy, as a powerful strategy for cancer treatment, has achieved tremendous efficacy in clinical trials. Despite these advancements, there is much to do in terms of enhancing therapeutic benefits and decreasing the side effects of cancer immunotherapy. Advanced nanobiomaterials, including liposomes, polymers, and silica, play a vital role in the codelivery of drugs and immunomodulators. These nanobiomaterial-based delivery systems could effectively promote antitumor immune responses and simultaneously reduce toxic adverse effects. Furthermore, nanobiomaterials may also combine with each other or with traditional drugs via different mechanisms, thus giving rise to more accurate and efficient tumor treatment. Here, an overview of the latest advancement in these nanobiomaterials used for cancer immunotherapy is given, describing outstanding systems, including lipid-based nanoparticles, polymer-based scaffolds or micelles, inorganic nanosystems, and others.
Collapse
Affiliation(s)
- Fan Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Kun Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Yan-Peng Jia
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Ying Hao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Jin-Rong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Zhi-Yong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
22
|
The DNMT1/miR-34a/FOXM1 Axis Contributes to Stemness of Liver Cancer Cells. JOURNAL OF ONCOLOGY 2020; 2020:8978930. [PMID: 32308683 PMCID: PMC7142390 DOI: 10.1155/2020/8978930] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/08/2020] [Accepted: 02/18/2020] [Indexed: 12/11/2022]
Abstract
Background Whether DNA methyltransferase 1 (DNMT1)/miR-34a/FoxM1 signaling promotes the stemness of liver cancer stem cells (LCSCs) remains unclear. This study aimed to assess whether methylation-based silencing of miR-34a by DNMT1 contributes to stemness features via FoxM1 upregulation in LCSCs. Methods The CD133+ subgroup of MHCC97H cells sorted by MACS was used as LCSCs. DNMT1, BMI1, SOX2, and OCT4 mRNA levels, and miR-34a amounts were determined by qRT-PCR. DNMT1, CD44, and FoxM1 proteins were analyzed by immunoblot. Sphere and colony formation abilities were detected by respective assays. CD133+ cell percentages were assessed by flow cytometry. In vivo oncogenicity was evaluated using a tumor xenograft model in mice. The effects of DNMT1/miR-34a signaling on the stemness of LCSCs were examined by knockdown or overexpression of DNMT1 and/or transfection of miR-34a mimic or inhibitor using lentivirus-delivery systems. FoxM1 association with miR-34a was detected by a reporter assay. Results We here showed that LCSCs exhibited elevated DNMT1 activity and expression, lower miR-34a expression with higher promoter methylation, and stronger stemness, compared with the parental liver cancer cells. DNMT1 knockdown repressed DNMT1, increased miR-34a amounts by promoter demethylation, and reduced stemness in LCSCs, whereas DNMT1 overexpression had the opposite effects in liver cancer cells. Transfection with miR-34a mimic repressed the stemness of LCSCs, while miR-34a inhibitor significantly downregulated miR-34a and enhanced stemness, without affecting DNMT1 in liver cancer cells. MiR-34a mimic rescued the effects of DNMT1 overexpression on the stemness of LCSCs, without affecting DNMT1 expression. Finally, FOXM1 was identified as a direct target by miR-34a in LCSCs. Conclusions We revealed that aberrant activation of DNMT1 causes miR-34a promoter methylation and suppression, leading to FoxM1 upregulation by disinhibition and promotion of LCSC stemness. These findings suggest that blockage of DNMT1/miR-34a-mediated FOXM1 upregulation might suppress liver cancer by targeting LCSCs.
Collapse
|
23
|
Eppler HB, Jewell CM. Biomaterials as Tools to Decode Immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1903367. [PMID: 31782844 PMCID: PMC7124992 DOI: 10.1002/adma.201903367] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 09/23/2019] [Indexed: 05/02/2023]
Abstract
The immune system has remarkable capabilities to combat disease with exquisite selectivity. This feature has enabled vaccines that provide protection for decades and, more recently, advances in immunotherapies that can cure some cancers. Greater control over how immune signals are presented, delivered, and processed will help drive even more powerful options that are also safe. Such advances will be underpinned by new tools that probe how immune signals are integrated by immune cells and tissues. Biomaterials are valuable resources to support this goal, offering robust, tunable properties. The growing role of biomaterials as tools to dissect immune function in fundamental and translational contexts is highlighted. These technologies can serve as tools to understand the immune system across molecular, cellular, and tissue length scales. A common theme is exploiting biomaterial features to rationally direct how specific immune cells or organs encounter a signal. This precision strategy, enabled by distinct material properties, allows isolation of immunological parameters or processes in a way that is challenging with conventional approaches. The utility of these capabilities is demonstrated through examples in vaccines for infectious disease and cancer immunotherapy, as well as settings of immune regulation that include autoimmunity and transplantation.
Collapse
Affiliation(s)
- Haleigh B Eppler
- Fischell Department of Bioengineering, 8278 Paint Brach Drive, College Park, MD, 20742, USA
- Biological Sciences Training Program, 1247 Biology Psychology Building, College Park, MD, 20742, USA
| | - Christopher M Jewell
- Fischell Department of Bioengineering, 8278 Paint Brach Drive, College Park, MD, 20742, USA
- Biological Sciences Training Program, 1247 Biology Psychology Building, College Park, MD, 20742, USA
- Robert E. Fischell Institute for Biomedical Devices, 8278 Paint Branch Drive, College Park, MD, 20742, USA
- United States Department of Veterans Affairs, VA Maryland Health Care System, 10. N Green Street, Baltimore, MD, 21201, USA
- Department of Microbiology and Immunology, University of Maryland Medical School, 685 West Baltimore Street, HSF-I Suite 380, Baltimore, MD, 21201, USA
- Marlene and Stewart Greenebaum Cancer Center, 22 South Greene Street, Baltimore, MD, 21201, USA
| |
Collapse
|
24
|
Phung CD, Tran TH, Kim JO. Engineered nanoparticles to enhance natural killer cell activity towards onco-immunotherapy: a review. Arch Pharm Res 2020; 43:32-45. [DOI: 10.1007/s12272-020-01218-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/20/2020] [Indexed: 12/12/2022]
|
25
|
Lin YX, Wang Y, Blake S, Yu M, Mei L, Wang H, Shi J. RNA Nanotechnology-Mediated Cancer Immunotherapy. Theranostics 2020; 10:281-299. [PMID: 31903120 PMCID: PMC6929632 DOI: 10.7150/thno.35568] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/06/2019] [Indexed: 12/19/2022] Open
Abstract
RNA molecules (e.g., siRNA, microRNA, and mRNA) have shown tremendous potential for immunomodulation and cancer immunotherapy. They can activate both innate and adaptive immune system responses by silencing or upregulating immune-relevant genes. In addition, mRNA-based vaccines have recently been actively pursued and tested in cancer patients, as a form of treatment. Meanwhile, various nanomaterials have been developed to enhance RNA delivery to the tumor and immune cells. In this review article, we summarize recent advances in the development of RNA-based therapeutics and their applications in cancer immunotherapy. We also highlight the variety of nanoparticle platforms that have been used for RNA delivery to elicit anti-tumor immune responses. Finally, we provide our perspectives of potential challenges and opportunities of RNA-based nanotherapeutics in clinical translation towards cancer immunotherapy.
Collapse
Affiliation(s)
- Yao-Xin Lin
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yi Wang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Sara Blake
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Tufts University, Medford, MA 02155, USA
| | - Mian Yu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Lin Mei
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong 510006, China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing 100190, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
26
|
Amon L, Lehmann CHK, Baranska A, Schoen J, Heger L, Dudziak D. Transcriptional control of dendritic cell development and functions. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 349:55-151. [PMID: 31759434 DOI: 10.1016/bs.ircmb.2019.10.001] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dendritic cells (DCs) are major regulators of adaptive immunity, as they are not only capable to induce efficient immune responses, but are also crucial to maintain peripheral tolerance and thereby inhibit autoimmune reactions. DCs bridge the innate and the adaptive immune system by presenting peptides of self and foreign antigens as peptide MHC complexes to T cells. These properties render DCs as interesting target cells for immunomodulatory therapies in cancer, but also autoimmune diseases. Several subsets of DCs with special properties and functions have been described. Recent achievements in understanding transcriptional programs on single cell level, together with the generation of new murine models targeting specific DC subsets, advanced our current understanding of DC development and function. Thus, DCs arise from precursor cells in the bone marrow with distinct progenitor cell populations splitting the monocyte populations and macrophage populations from the DC lineage, which upon lineage commitment can be separated into conventional cDC1, cDC2, and plasmacytoid DCs (pDCs). The DC populations harbor intrinsic programs enabling them to react for specific pathogens in dependency on the DC subset, and thereby orchestrate T cell immune responses. Similarities, but also varieties, between human and murine DC subpopulations are challenging, and will require further investigation of human specimens under consideration of the influence of the tissue micromilieu and DC subset localization in the future.
Collapse
Affiliation(s)
- Lukas Amon
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Christian H K Lehmann
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Baranska
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Janina Schoen
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Lukas Heger
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Diana Dudziak
- Laboratory of Dendritic Cell Biology, Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
27
|
Sang MM, Liu FL, Wang Y, Luo RJ, Huan XX, Han LF, Zhang ZT, Feng F, Qu W, Liu W, Zheng F. A novel redox/pH dual-responsive and hyaluronic acid-decorated multifunctional magnetic complex micelle for targeted gambogic acid delivery for the treatment of triple negative breast cancer. Drug Deliv 2019; 25:1846-1857. [PMID: 30334478 PMCID: PMC6225507 DOI: 10.1080/10717544.2018.1486472] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Gambogic acid (GA) is a naturally derived potent anticancer agent with extremely poor biocompatibility. In the present study, a novel of redox/pH dual-responsive multifunctional magnetic complex micelle (sPEG/HA/CSO-SS-Hex/Fe3O4/GA), which consisted of a reducible hexadecanol-modified chitosan oligosaccharide polymer micelle (CSO-SS-Hex) coated with hyaluronic acid (HA) and DCA grafted sheddable PEG-PLL (sPEG) copolymers and loaded with gambogic acid (GA) and Fe3O4 nanoparticles were developed for parenteral delivery for the treatment of triple negative breast cancer (TNBC). The ex vivo study showed that the sPEG shielded cationic HA/CSO-SS-Hex/Fe3O4/GA core at physiological pH but quickly shed off to re-expose the core due to its charge reversible property. The sPEG/HA/CSO-SS-Hex/Fe3O4/GA micelles effectively facilitated tumor-targeted GA delivery by HA, which is a targeting ligand for CD44 receptor of TNBC cells, meanwhile increase GA uptake at the acidic condition but diminished the drug uptake at neutral pH. The in vitro cellular uptake study and in vivo biodistribution and antitumor activity of the formulations were determined, all results showed that the complex micelle enhanced TNBC tumor cellular uptake and fast drug release due to the combined effect of magnet targeting, CD44 receptor-mediated internalization and redox/pH dual-responsive drug release. Hence, tumor-targeted delivery of GA with redox/pH dual-responsive multifunctional magnetic complex micelle sPEG/HA/CSO-SS-Hex/Fe3O4/GA might have potential implications for the chemotherapy of TNBC.
Collapse
Affiliation(s)
- Mang Mang Sang
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Fu Lei Liu
- c Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Yang Wang
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Ren Jie Luo
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Xiao Xian Huan
- c Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Ling Fei Han
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Zhong Tao Zhang
- c Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Feng Feng
- c Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Wei Qu
- c Department of Natural Medicinal Chemistry , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Wenyuan Liu
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| | - Feng Zheng
- a Key Laboratory of Drug Quality Control and Pharmacovigilance, Ministry of Education , China Pharmaceutical University , Nanjing , People's Republic of China.,b Department of Pharmaceutical Analysis , China Pharmaceutical University , Nanjing , People's Republic of China
| |
Collapse
|
28
|
Yong SB, Chung JY, Song Y, Kim J, Ra S, Kim YH. Non-viral nano-immunotherapeutics targeting tumor microenvironmental immune cells. Biomaterials 2019; 219:119401. [PMID: 31398571 DOI: 10.1016/j.biomaterials.2019.119401] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 12/12/2022]
Abstract
The tumor microenvironmental immune cells (TMICs) consists of myeloid cells (tumor-associated macrophages, dendritic cells, myeloid-derived suppressor cells, etc.) and lymphocytes (T cells and B cells), all of which could be immunologically suppressed through their interactions with cancer cells. Immunological understanding of the tumor microenvironment (TME) has led to great success in the development of clinical cancer immunotherapeutic. The most advanced cancer immunotherapies are chimeric antigen receptor-modified T cells (CAR-T cells) and checkpoint inhibiting antibodies blocking CTLA4, PD-1 and PD-L1. However, many hurdles remain that should be addressed for improved therapeutic efficacy and reduced side effects such as cytokine release syndrome and patient-death. In recent decades, nanoparticles have been demonstrated as an efficient drug delivery tool due to their ease of modification, biocompatibility and intrinsic tumor targeting effect, and also been applied for cancer immunotherapy. In this review, we briefly introduce the immunosuppressive functions of TMICs and review recent advances in the development of TMIC-targeted nanotherapeutics for cancer immunotherapy. Tumor-associated macrophage (TAM)-targeted systems have shown to deplete or repolarize macrophages to M1 state for anti-tumoral immune responses. Tumor-infiltrating T cell (TIT)-targeted strategies have provided the activation of effector T cells and suppression of regulatory T cells in tumor, overcoming the current hurdles of single regimen checkpoint inhibitors. Lastly, recent studies on dendritic cell-targeted mRNA vaccination are discussed and the future perspectives of nano-immunotherapeutic for next-generation of cancer immunotherapy is emphasized.
Collapse
Affiliation(s)
- Seok-Beom Yong
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, BK 21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 133-791, Seoul, Republic of Korea
| | - Jee Young Chung
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, BK 21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 133-791, Seoul, Republic of Korea
| | - Yoonsung Song
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, BK 21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 133-791, Seoul, Republic of Korea
| | - Jaehyun Kim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, BK 21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 133-791, Seoul, Republic of Korea
| | - Sehee Ra
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, BK 21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 133-791, Seoul, Republic of Korea
| | - Yong-Hee Kim
- Department of Bioengineering, Institute for Bioengineering and Biopharmaceutical Research, BK 21 Plus Future Biopharmaceutical Human Resources Training and Research Team, Hanyang University, 133-791, Seoul, Republic of Korea.
| |
Collapse
|
29
|
Trimaille T, Lacroix C, Verrier B. Self-assembled amphiphilic copolymers as dual delivery system for immunotherapy. Eur J Pharm Biopharm 2019; 142:232-239. [PMID: 31229673 DOI: 10.1016/j.ejpb.2019.06.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 05/03/2019] [Accepted: 06/19/2019] [Indexed: 01/07/2023]
Abstract
Subunit vaccines using recombinant antigens appear as the privileged vaccination technology for safety reasons but still require the development of carriers/adjuvants ensuring optimal immunogenicity and efficacy. Micelles from self-assembled amphiphilic copolymers have recently emerged as highly relevant and promising candidates owing to their ease of preparation, low size (entering in lymphatic capillaries for reaching lymph nodes), size/surface tunability and chemical versatility enabling introduction of stimuli (e.g. pH) responsive features and biofunctionalization with dedicated molecules. In particular, research efforts have increasingly focused on dendritic cells (DCs) targeting and activation by co-delivering (with antigen) ligands of pattern recognition receptors (PRRs, e.g. toll-like receptors). Such strategy has appeared as one of the most effective for eliciting CD 8+ T-cell response, which is crucial in the eradication of tumors and numerous infectious diseases. In this short review, we highlight the recent advances in such micelle-based carriers in subunit vaccination and how their precise engineering can be a strong asset for guiding and controlling immune responses.
Collapse
Affiliation(s)
- Thomas Trimaille
- Aix Marseille Univ, CNRS, Institut de Chimie Radicalaire, Marseille, France.
| | - Céline Lacroix
- Université Lyon 1, CNRS, UMR 5305, Biologie Tissulaire et Ingénierie Thérapeutique, IBCP, 69367 Lyon, France
| | - Bernard Verrier
- Université Lyon 1, CNRS, UMR 5305, Biologie Tissulaire et Ingénierie Thérapeutique, IBCP, 69367 Lyon, France
| |
Collapse
|
30
|
Wang M, Song J, Zhou F, Hoover AR, Murray C, Zhou B, Wang L, Qu J, Chen WR. NIR-Triggered Phototherapy and Immunotherapy via an Antigen-Capturing Nanoplatform for Metastatic Cancer Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1802157. [PMID: 31131193 PMCID: PMC6523374 DOI: 10.1002/advs.201802157] [Citation(s) in RCA: 196] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 02/21/2019] [Indexed: 05/13/2023]
Abstract
Combined phototherapy and immunotherapy demonstrates strong potential in the treatment of metastatic cancers. An upconversion nanoparticle (UCNP) based antigen-capturing nanoplatform is designed to synergize phototherapies and immunotherapy. In particular, this nanoplatform is constructed via self-assembly of DSPE-PEG-maleimide and indocyanine green (ICG) onto UCNPs, followed by loading of the photosensitizer rose bengal (RB). ICG significantly enhances the RB-based photodynamic therapy efficiency of UCNP/ICG/RB-mal upon activation by a near-infrared (NIR) laser, simultaneously achieving selective photothermal therapy. Most importantly, tumor-derived protein antigens, arising from phototherapy-treated tumor cells, can be captured and retained in situ, due to the functionality of maleimide, which further enhance the tumor antigen uptake and presentation by antigen-presenting cells. The synergized photothermal, photodynamic, and immunological effects using light-activated UCNP/ICG/RB-mal induces a tumor-specific immune response. In the experiments, intratumoral administration of UCNP/ICG/RB-mal, followed by noninvasive irradiation with an NIR laser, destroys primary tumors and inhibits untreated distant tumors, using a poorly immunogenic, highly metastatic 4T1 mammary tumor model. With the simultaneous use of anti-CTLA-4, about 84% of the treated tumor-bearing mice achieve long-term survival and 34% of mice develop tumor-specific immunity. Overall, this antigen-capturing nanoplatform provides a promising approach for the treatment of metastatic cancers.
Collapse
Affiliation(s)
- Meng Wang
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong ProvinceCollege of Optoelectronic EngineeringShenzhen UniversityShenzhen518060P. R. China
| | - Jun Song
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong ProvinceCollege of Optoelectronic EngineeringShenzhen UniversityShenzhen518060P. R. China
| | - Feifan Zhou
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong ProvinceCollege of Optoelectronic EngineeringShenzhen UniversityShenzhen518060P. R. China
| | - Ashley R. Hoover
- Center of Interdisciplinary Biomedical Education and ResearchCollege of Mathematics and ScienceUniversity of Central OklahomaOK73034USA
| | - Cynthia Murray
- Center of Interdisciplinary Biomedical Education and ResearchCollege of Mathematics and ScienceUniversity of Central OklahomaOK73034USA
| | - Benqing Zhou
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong ProvinceCollege of Optoelectronic EngineeringShenzhen UniversityShenzhen518060P. R. China
| | - Lu Wang
- Center of Interdisciplinary Biomedical Education and ResearchCollege of Mathematics and ScienceUniversity of Central OklahomaOK73034USA
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong ProvinceCollege of Optoelectronic EngineeringShenzhen UniversityShenzhen518060P. R. China
| | - Wei R. Chen
- Center of Interdisciplinary Biomedical Education and ResearchCollege of Mathematics and ScienceUniversity of Central OklahomaOK73034USA
| |
Collapse
|
31
|
Abstract
Cancer immunotherapy has shown impressive clinical results in the last decade, improving both solid and hematologic cancer patients' overall survival. Nevertheless, most of the molecular aspects underlying the response to this approach are still under investigation. miRNAs in particular have been described as regulators of a plethora of different immunologic processes and thus have the potential to be key in the future developments of immunotherapy. In this review, we summarize and discuss the emerging role of miRNAs in the diagnosis and therapeutics of the four principal cancer immunotherapy approaches: immune checkpoint blockade, adoptive cell therapy, cancer vaccines, and cytokine therapy. In particular, this review is focused on potential roles for miRNAs to be adjuvants in soluble factor- and cell-based therapies, with the aim of helping to increase specificity and decrease toxicity, and on the potential for rationally identified miRNA-based diagnostic approaches to aid in precision clinical immunooncology.
Collapse
|
32
|
Chen Z, Liu L, Liang R, Luo Z, He H, Wu Z, Tian H, Zheng M, Ma Y, Cai L. Bioinspired Hybrid Protein Oxygen Nanocarrier Amplified Photodynamic Therapy for Eliciting Anti-tumor Immunity and Abscopal Effect. ACS NANO 2018; 12:8633-8645. [PMID: 30005164 DOI: 10.1021/acsnano.8b04371] [Citation(s) in RCA: 256] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
An ideal cancer therapeutic strategy is expected to possess potent ability to not only ablate primary tumors but also prevent distance metastasis and relapse. In this study, human serum albumin was hybridized with hemoglobin by intermolecular disulfide bonds to develop a hybrid protein oxygen nanocarrier with chlorine e6 encapsulated (C@HPOC) for oxygen self-sufficient photodynamic therapy (PDT). C@HPOC realized the tumor-targeted co-delivery of photosensitizer and oxygen, which remarkably relieved tumor hypoxia. C@HPOC was favorable for more efficient PDT and enhanced infiltration of CD8+ T cells in tumors. Moreover, oxygen-boosted PDT of C@HPOC induced immunogenic cell death, with the release of danger-associated molecular patterns to activate dendritic cells, T lymphocytes, and natural killer cells in vivo. Notably, C@HPOC-mediated immunogenic PDT could destroy primary tumors and effectively suppress distant tumors and lung metastasis in a metastatic triple-negative breast cancer model by evoking systemic anti-tumor immunity. This study provides a paradigm of oxygen-augmented immunogenic PDT for metastatic cancer treatment.
Collapse
Affiliation(s)
- Zhikuan Chen
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| | - Lanlan Liu
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| | - Ruijing Liang
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , P. R. China
| | - Zhenyu Luo
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , P. R. China
- University of Chinese Academy of Sciences , Beijing 100049 , P. R. China
| | - Huamei He
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , P. R. China
| | - Zhihao Wu
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , P. R. China
| | - Hao Tian
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Key Laboratory for Nanomedicine , Guangdong Medical University , Dongguan 523808 , P. R. China
| | - Mingbin Zheng
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , P. R. China
- Dongguan Key Laboratory of Drug Design and Formulation Technology, Key Laboratory for Nanomedicine , Guangdong Medical University , Dongguan 523808 , P. R. China
| | - Yifan Ma
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , P. R. China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Shenzhen Engineering Laboratory of Nanomedicine and Nanoformulations , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , P. R. China
| |
Collapse
|
33
|
Abstract
Cancer immunotherapy is a powerful, growing treatment approach to cancer that can be combined with chemotherapy, radiotherapy, and oncosurgery. Modulating the immune system to enhance anticancer response by several strategies has yielded improved cancer survival. Despite this progress, the success rate for immunotherapy has been below expectations due to unpredictable efficacy and off-target side effects from systemic dosing. Nanotechnology offers numerous different materials and targeting properties to overcome many of these challenges in immunotherapy. In this chapter, we review current immunotherapy and its challenges as well as the latest nanotechnology applications in cancer immunotherapy.
Collapse
Affiliation(s)
- C Tilden Hagan
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; UNC/NCSU Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Yusra B Medik
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Andrew Z Wang
- Laboratory of Nano- and Translational Medicine, Lineberger Comprehensive Cancer Center, Carolina Center for Cancer Nanotechnology Excellence, Carolina Institute of Nanomedicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States; Department of Radiation Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| |
Collapse
|
34
|
Liu L, He H, Liang R, Yi H, Meng X, Chen Z, Pan H, Ma Y, Cai L. ROS-Inducing Micelles Sensitize Tumor-Associated Macrophages to TLR3 Stimulation for Potent Immunotherapy. Biomacromolecules 2018; 19:2146-2155. [PMID: 29669207 DOI: 10.1021/acs.biomac.8b00239] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
One approach to cancer immunotherapy is the repolarization of immunosuppressive tumor-associated macrophages (TAMs) to antitumor M1 macrophages. The present study developed galactose-functionalized zinc protoporphyrin IX (ZnPP) grafted poly(l-lysine)- b-poly(ethylene glycol) polypeptide micelles (ZnPP PM) for TAM-targeted immunopotentiator delivery, which aimed at in vivo repolarization of TAMs to antitumor M1 macrophages. The outcomes revealed that ROS-inducing ZnPP PM demonstrated specificity for the in vitro and in vivo targeting of macrophages, elevated the level of ROS, and lowered STAT3 expression in BM-TAMs. Poly I:C (PIC, a TLR3 agonist)-loaded ZnPP PM (ZnPP PM/PIC) efficiently repolarized TAMs to M1 macrophages, which were reliant on ROS generation. Further, ZnPP PM/PIC substantially elevated the activated NK cells and T lymphocytes in B16-F10 melanoma tumors, which caused vigorous tumor regression. Therefore, the TAM-targeted transport of an immunologic adjuvant with ZnPP-grafted nanovectors may be a potential strategy to repolarize TAMs to M1 macrophages in situ for effective cancer immunotherapy.
Collapse
Affiliation(s)
- Lanlan Liu
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , People's Republic of China.,University of Chinese Academy of Sciences , Beijing 100049 , People's Republic of China
| | - Huamei He
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , People's Republic of China
| | - Ruijing Liang
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , People's Republic of China
| | - Huqiang Yi
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , People's Republic of China
| | - Xiaoqing Meng
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , People's Republic of China.,University of Chinese Academy of Sciences , Beijing 100049 , People's Republic of China
| | - Zhikuan Chen
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , People's Republic of China.,University of Chinese Academy of Sciences , Beijing 100049 , People's Republic of China
| | - Hong Pan
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , People's Republic of China.,University of Chinese Academy of Sciences , Beijing 100049 , People's Republic of China
| | - Yifan Ma
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , People's Republic of China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS Key Lab for Health Informatics, Institute of Biomedicine and Biotechnology , Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences , Shenzhen 518055 , People's Republic of China
| |
Collapse
|
35
|
Yan S, Liu G, Jin C, Wang Z, Duan Q, Xu J, Xu D. MicroRNA-6869-5p acts as a tumor suppressor via targeting TLR4/NF-κB signaling pathway in colorectal cancer. J Cell Physiol 2018; 233:6660-6668. [PMID: 29206292 DOI: 10.1002/jcp.26316] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2017] [Accepted: 12/04/2017] [Indexed: 12/18/2022]
Abstract
Many studies have implicated that microRNAs (miRNAs), as non-coding RNAs, play important roles in the development and progression of colorectal cancer (CRC). However, little is known about the role of a newly identified miRNA, miR-6869-5p, in CRC. We aim to investigate the modifying effects and underlying mechanisms of miR-6869-5 in colorectal carcinogenesis and progression. Significantly reduced levels of miR-6869-5p were observed in both serum exosomes tumor tissue samples from patients with CRC. The prediction of targets of miR-6869-5p in databases of targetscan, microRNA. ORG and miRDBA revealed that toll-like receptor 4 (TLR4) is a potential target for this miRNA. MiR-6869-5p could inhibit cell proliferation and the production of inflammatory cytokines (TNF-α and IL-6) in CRC cells via directly targeting TLR4. The protective effect of miR-6869-5p from colorectal carcinogenesis was dependent on TLR4/NF-κB signaling pathway. In addition, the 3-year survival was poor among CRC patients with decreased levels of miR-6869-5p in serum exosomes. Thus, miR-6869-5p may serve as a tumor suppressor in CRC, and serum exosomal miR-6869-5p is a promising circulating biomarker for the prediction of CRC prognosis.
Collapse
Affiliation(s)
- Shushan Yan
- Department of Gastrointestinal and Anal Diseases Surgery, The Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Guoyan Liu
- Department of Dermatology, The Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Chengwen Jin
- Functional Laboratory, Clinical Medicine College of Weifang Medical University, Weifang, China
| | - Zengfang Wang
- Department of Gynecology and Obstetrics, Weifang Hospital of Maternal and Child Health, Weifang, China
| | - Quanhong Duan
- Department of Gastrointestinal and Anal Diseases Surgery, The Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Jiang Xu
- Department of Rehabilitation, Huai'an Second People's Hospital, The Affiliated Hospital of Xuzhou Medical University, Huai'an, China
| | - Donghua Xu
- Department of Rheumatology and Immunology, The Affiliated Hospital of Weifang Medical University, Weifang, China.,Clinical Medicine College, Weifang Medical University, Weifang, China
| |
Collapse
|
36
|
Shen H, Sun T, Hoang HH, Burchfield JS, Hamilton GF, Mittendorf EA, Ferrari M. Enhancing cancer immunotherapy through nanotechnology-mediated tumor infiltration and activation of immune cells. Semin Immunol 2017; 34:114-122. [PMID: 28947107 PMCID: PMC5705528 DOI: 10.1016/j.smim.2017.09.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/10/2017] [Accepted: 09/11/2017] [Indexed: 12/11/2022]
Abstract
Cancer immunotherapy has become arguably the most promising advancement in cancer research and therapy in recent years. The efficacy of cancer immunotherapy is critically dependent on specific physiological and physical processes - collectively referred to as transport barriers - including the activation of T cells by antigen presenting cells, T cells migration to and penetration into the tumor microenvironment, and movement of nutrients and other immune cells through the tumor microenvironment. Nanotechnology-based approaches have great potential to help overcome these transport barriers. In this review, we discuss the ways that nanotechnology is being leveraged to improve the efficacy and potency of various cancer immunotherapies.
Collapse
Affiliation(s)
- Haifa Shen
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA; Department of Cell and Developmental Biology, Weill Cornell Medical College, New York, NY 10065, USA
| | - Tong Sun
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Hanh H Hoang
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Jana S Burchfield
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Gillian F Hamilton
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA
| | - Elizabeth A Mittendorf
- Department of Breast Surgical Oncology, the University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mauro Ferrari
- Houston Methodist Research Institute, 6670 Bertner Avenue, Houston, TX 77030, USA; Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA.
| |
Collapse
|
37
|
Hargadon KM. Strategies to Improve the Efficacy of Dendritic Cell-Based Immunotherapy for Melanoma. Front Immunol 2017; 8:1594. [PMID: 29209327 PMCID: PMC5702020 DOI: 10.3389/fimmu.2017.01594] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 11/06/2017] [Indexed: 12/20/2022] Open
Abstract
Melanoma is a highly aggressive form of skin cancer that frequently metastasizes to vital organs, where it is often difficult to treat with traditional therapies such as surgery and radiation. In such cases of metastatic disease, immunotherapy has emerged in recent years as an exciting treatment option for melanoma patients. Despite unprecedented successes with immune therapy in the clinic, many patients still experience disease relapse, and others fail to respond at all, thus highlighting the need to better understand factors that influence the efficacy of antitumor immune responses. At the heart of antitumor immunity are dendritic cells (DCs), an innate population of cells that function as critical regulators of immune tolerance and activation. As such, DCs have the potential to serve as important targets and delivery agents of cancer immunotherapies. Even immunotherapies that do not directly target or employ DCs, such as checkpoint blockade therapy and adoptive cell transfer therapy, are likely to rely on DCs that shape the quality of therapy-associated antitumor immunity. Therefore, understanding factors that regulate the function of tumor-associated DCs is critical for optimizing both current and future immunotherapeutic strategies for treating melanoma. To this end, this review focuses on advances in our understanding of DC function in the context of melanoma, with particular emphasis on (1) the role of immunogenic cell death in eliciting tumor-associated DC activation, (2) immunosuppression of DC function by melanoma-associated factors in the tumor microenvironment, (3) metabolic constraints on the activation of tumor-associated DCs, and (4) the role of the microbiome in shaping the immunogenicity of DCs and the overall quality of anti-melanoma immune responses they mediate. Additionally, this review highlights novel DC-based immunotherapies for melanoma that are emerging from recent progress in each of these areas of investigation, and it discusses current issues and questions that will need to be addressed in future studies aimed at optimizing the function of melanoma-associated DCs and the antitumor immune responses they direct against this cancer.
Collapse
Affiliation(s)
- Kristian M. Hargadon
- Hargadon Laboratory, Department of Biology, Hampden-Sydney College, Hampden-Sydney, VA, United States
| |
Collapse
|
38
|
Liu L, Yi H, He H, Pan H, Cai L, Ma Y. Tumor associated macrophage-targeted microRNA delivery with dual-responsive polypeptide nanovectors for anti-cancer therapy. Biomaterials 2017; 134:166-179. [PMID: 28463694 DOI: 10.1016/j.biomaterials.2017.04.043] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 04/21/2017] [Accepted: 04/23/2017] [Indexed: 11/26/2022]
Abstract
Repolarizing Tumor-associated macrophages (TAMs) to anti-tumor M1 macrophages with microRNA (miR) is a plausible approach for cancer treatment. However, how to achieve TAM-targeted miR delivery remains a challenge. The present study generated redox/pH dual-responsive hybrid polypeptide nanovectors, which consisted of self-crosslinked redox-responsive nanoparticles based on galactose-functionalized n-butylamine-poly(l-lysine)-b-poly(l-cysteine) polypeptides (GLC) coated with DCA-grafted sheddable PEG-PLL (sPEG) copolymers. The ex vivo study showed that sPEG shielded cationic GLC core at physiological pH but quickly shed off to re-expose GLC due to it charge reversible property. Encapsulation with sPEG/GLC nanovectors effectively facilitated macrophage-targeted miR delivery at the acidic condition but diminished miR uptake at neutral pH. Administration of miR155-loaded sPEG/GLC (sPEG/GLC/155) nanocomplexes increased miR155 expression in TAMs about 100-400 folds both in vitro and in vivo. sPEG/GLC/155 also effectively repolarized immunosuppressive TAMs to anti-tumor M1 macrophages through elevating M1 macrophage markers (IL-12, iNOS, MHC II) and suppressing M2 macrophage markers (Msr2 and Arg1) in TAMs. Moreover, the treatment of sPEG/GLC/155 significantly increased activated T lymphocytes and NK cells in tumors, which consequently led to robust tumor regression. Hence, TAM-targeted delivery of miR with redox/pH dual-responsive sPEG/GLC nanovectors could be a promising approach to re-polarize TAMs to M1 macrophages in situ and induce tumor regression.
Collapse
Affiliation(s)
- Lanlan Liu
- Key Lab of Health Informatics of Chinese Academy of Sciences, Guangdong Key Laboratory of Nanomedicine, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, PR China
| | - Huqiang Yi
- Key Lab of Health Informatics of Chinese Academy of Sciences, Guangdong Key Laboratory of Nanomedicine, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, PR China; Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, PR China
| | - Huamei He
- Key Lab of Health Informatics of Chinese Academy of Sciences, Guangdong Key Laboratory of Nanomedicine, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, PR China
| | - Hong Pan
- Key Lab of Health Informatics of Chinese Academy of Sciences, Guangdong Key Laboratory of Nanomedicine, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, PR China
| | - Lintao Cai
- Key Lab of Health Informatics of Chinese Academy of Sciences, Guangdong Key Laboratory of Nanomedicine, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, PR China
| | - Yifan Ma
- Key Lab of Health Informatics of Chinese Academy of Sciences, Guangdong Key Laboratory of Nanomedicine, Shenzhen Institutes of Advanced Technology, Chinese Academy of Science, Shenzhen 518055, PR China.
| |
Collapse
|