201
|
Xing M, Wang X, Kiken RA, He L, Zhang JY. Immunodiagnostic Biomarkers for Hepatocellular Carcinoma (HCC): The First Step in Detection and Treatment. Int J Mol Sci 2021; 22:6139. [PMID: 34200243 PMCID: PMC8201127 DOI: 10.3390/ijms22116139] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/27/2021] [Accepted: 05/27/2021] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) exerts huge effects on the health burden of the world because of its high mortality and poor prognosis. HCC is often clinically detected late in patients. If HCC could be detected and treated earlier, the survival rate of patients will be greatly improved. Therefore, identifying specific biomarkers is urgent and important for HCC. The liver is also recognized as an immune organ. The occurrence of HCC is related to exacerbation of immune tolerance and/or immunosurveillance escape. The host immune system plays an important role in the recognition and targeting of tumor cells in cancer immunotherapy, as can be seen from the clinical success of immune checkpoint inhibitors and chimeric antigen receptor (CAR) T cells. Thus, there is a pressing medical need to discover immunodiagnostic biomarkers specific to HCC for understanding the pathological mechanisms of HCC, especially for immunotherapy targets. We have reviewed the existing literature to summarize the immunodiagnostic markers of HCC, including autoantibodies against tumor-associated antigens (TAAs) and exosomes, to provide new insights into HCC and early detection of this deadly cancer.
Collapse
Affiliation(s)
- Mengtao Xing
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China;
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (X.W.); (R.A.K.)
| | - Xinzhi Wang
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (X.W.); (R.A.K.)
- Jiangsu Key Laboratory of Drug Screening, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Robert A. Kiken
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (X.W.); (R.A.K.)
| | - Ling He
- Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, China;
| | - Jian-Ying Zhang
- Department of Biological Sciences & NIH-Sponsored Border Biomedical Research Center, The University of Texas at El Paso, El Paso, TX 79968, USA; (X.W.); (R.A.K.)
| |
Collapse
|
202
|
Vu NB, Nguyen HT, Palumbo R, Pellicano R, Fagoonee S, Pham PV. Stem cell-derived exosomes for wound healing: current status and promising directions. Minerva Med 2021; 112:384-400. [PMID: 33263376 DOI: 10.23736/s0026-4806.20.07205-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Wound healing, especially of chronic wounds, is still an unmet therapeutic area since assessment and management are extremely complicated. Although many efforts have been made to treat wounds, all strategies have achieved limited results for chronic wounds. Stem cell-based therapy is considered a promising approach for complex wounds such as those occurring in diabetics. Mesenchymal stem cell transplantation significantly improves wound closure, angiogenesis and wound healing. However, cell therapy is complex, expensive and time-consuming. Recent studies have shown that stem cell-derived exosomes can be an exciting approach to treat wounds. Exosomes derived from mesenchymal stem cells can induce benefit in almost all stages of wound healing, including control of immune responses, inhibition of inflammation, promoting cell proliferation and angiogenesis, while reducing scar formation during the wound healing process. This review aimed at offering an updated overview of the use of exosomes in biological applications, such as wound healing, and addresses not only current applications but also new directions for this next-generation approach in wound healing.
Collapse
Affiliation(s)
- Ngoc B Vu
- Stem Cell Institute, University of Science, Ho Chi Minh, Vietnam
- Vietnam National University - Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Hoa T Nguyen
- Stem Cell Institute, University of Science, Ho Chi Minh, Vietnam
- Vietnam National University - Ho Chi Minh City, Ho Chi Minh, Vietnam
| | - Rosanna Palumbo
- Institute of Biostructure and Bioimaging (CNR), Naples, Italy
| | | | - Sharmila Fagoonee
- Institute of Biostructure and Bioimaging (CNR), Molecular Biotechnology Center, Turin, Italy
| | - Phuc V Pham
- Stem Cell Institute, University of Science, Ho Chi Minh, Vietnam -
- Vietnam National University - Ho Chi Minh City, Ho Chi Minh, Vietnam
- Laboratory of Stem Cell Research and Application, Ho Chi Minh, Vietnam
| |
Collapse
|
203
|
Mehanny M, Lehr CM, Fuhrmann G. Extracellular vesicles as antigen carriers for novel vaccination avenues. Adv Drug Deliv Rev 2021; 173:164-180. [PMID: 33775707 DOI: 10.1016/j.addr.2021.03.016] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/01/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023]
Abstract
Antigen delivery has always been a challenge in scientific practice of vaccine formulation. Yet, mammalian extracellular vesicles (EVs) or bacterial membrane vesicles (MVs) provide an innovative avenue for safe and effective delivery of antigenic material. They include intrinsically loaded antigens from EV-secreting cells or extrinsically loaded antigens onto pre-formed vesicles. Interestingly, many studies shed light on potential novel anti-cancer vaccination immunotherapy for therapeutic applications from mammalian cell host-derived EVs, as well as conventional vaccination for prophylactic applications using bacterial cell-derived MVs against infectious diseases. Here, we discuss the rationale, status quo and potential for both vaccine applications using EVs.
Collapse
|
204
|
Hou PP, Chen HZ. Extracellular vesicles in the tumor immune microenvironment. Cancer Lett 2021; 516:48-56. [PMID: 34082025 DOI: 10.1016/j.canlet.2021.05.032] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/23/2021] [Accepted: 05/25/2021] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles (EVs) have gained significant attention in recent decades as major mediators of intercellular communication that are involved in various essential physiological and pathological processes. They are secreted by almost all cell types and carry bioactive materials, such as proteins, lipids and nucleic acids, that can be transmitted from host cells to recipient cells, thereby eliciting phenotypic and functional alterations in the recipient cells. Recent evidence shows that EVs play essential roles in remodeling the tumor immune microenvironment (TIME). EVs derived from tumor cells and immune cells mediate mutual communication at proximal and distal sites, which determines tumor fate and antitumor therapeutic effectiveness. In this review, the current understanding of EVs and their roles in remodeling the TIME and modulating tumor-specific immunity are summarized. We mainly discuss the mutual regulation between tumor cells and tumor-infiltrating immune cells through the delivery of EVs in the TIME. We also describe the limitations of current studies and discuss directions for further research.
Collapse
Affiliation(s)
- Pei-Pei Hou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Hang-Zi Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, 361102, China.
| |
Collapse
|
205
|
Wang X, Zhou Y, Ding K. Roles of exosomes in cancer chemotherapy resistance, progression, metastasis and immunity, and their clinical applications (Review). Int J Oncol 2021; 59:44. [PMID: 34013358 PMCID: PMC8143748 DOI: 10.3892/ijo.2021.5224] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 04/21/2021] [Indexed: 12/18/2022] Open
Abstract
Exosomes are a type of vesicle that are secreted by cells, with a diameter of 40-100 nm, and that appear as a cystic shape under an electron microscope. Exosome cargo includes a variety of biologically active substances such as non-coding RNA, lipids and small molecule proteins. Exosomes can be taken up by neighboring cells upon secretion or by distant cells within the circulatory system, affecting gene expression of the recipient cells. The present review discusses the formation and secretion of exosomes, and how they can remodel the tumor microenvironment, enhancing cancer cell chemotherapy resistance and tumor progression. Exosome-mediated induction of tumor metastasis is also highlighted. More importantly, the review discusses the manner in which exosomes can change the metabolism of cancer cells and the immune system, which may help to devise novel therapeutic approaches for cancer treatment. With the development of nanotechnology, exosomes can also be used as biomarkers and for the delivery of chemical drugs, serving as a tool to diagnose and treat cancer.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, P.R. China
| | - Yuan Zhou
- Gruduate School, Tianjin Medical University, Tianjin 300070, P.R. China
| | - Kaiyang Ding
- Department of Hematology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, P.R. China
| |
Collapse
|
206
|
Guan MC, Ouyang W, Wang MD, Liang L, Li N, Fu TT, Shen F, Lau WY, Xu QR, Huang DS, Zhu H, Yang T. Biomarkers for hepatocellular carcinoma based on body fluids and feces. World J Gastrointest Oncol 2021; 13:351-365. [PMID: 34040698 PMCID: PMC8131906 DOI: 10.4251/wjgo.v13.i5.351] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/18/2021] [Accepted: 04/13/2021] [Indexed: 02/06/2023] Open
Abstract
Novel non-/minimally-invasive and effective approaches are urgently needed to supplement and improve current strategies for diagnosis and management of hepatocellular carcinoma (HCC). Overwhelming evidence from published studies on HCC has documented that multiple molecular biomarkers detected in body fluids and feces can be utilized in early-diagnosis, predicting responses to specific therapies, evaluating prognosis before or after therapy, as well as serving as novel therapeutic targets. Detection and analysis of proteins, metabolites, circulating nucleic acids, circulating tumor cells, and extracellular vesicles in body fluids (e.g., blood and urine) and gut microbiota (e.g., in feces) have excellent capabilities to improve different aspects of management of HCC. Numerous studies have been devoted in identifying more promising candidate biomarkers and therapeutic targets for diagnosis, treatment, and monitoring responses of HCC to conventional therapies, most of which may improve diagnosis and management of HCC in the future. This review aimed to summarize recent advances in utilizing these biomarkers in HCC and discuss their clinical significance.
Collapse
Affiliation(s)
- Ming-Cheng Guan
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Wei Ouyang
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Ming-Da Wang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital (Navy Medical University), Second Military Medical University, Shanghai 200438, China
| | - Lei Liang
- Department of Hepatobiliary, Pancreatic and Minimal Invasive Surgery, Zhejiang Provincial People’s Hospital (People’s Hospital of Hangzhou Medical College), Hangzhou 310000, Zhejiang Province, China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou 310000, Zhejiang Province, China
| | - Na Li
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Ting-Ting Fu
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Feng Shen
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital (Navy Medical University), Second Military Medical University, Shanghai 200438, China
| | - Wan-Yee Lau
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital (Navy Medical University), Second Military Medical University, Shanghai 200438, China
- Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Qiu-Ran Xu
- Department of Hepatobiliary, Pancreatic and Minimal Invasive Surgery, Zhejiang Provincial People’s Hospital (People’s Hospital of Hangzhou Medical College), Hangzhou 310000, Zhejiang Province, China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou 310000, Zhejiang Province, China
| | - Dong-Sheng Huang
- Department of Hepatobiliary, Pancreatic and Minimal Invasive Surgery, Zhejiang Provincial People’s Hospital (People’s Hospital of Hangzhou Medical College), Hangzhou 310000, Zhejiang Province, China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou 310000, Zhejiang Province, China
| | - Hong Zhu
- Department of Medical Oncology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| | - Tian Yang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital (Navy Medical University), Second Military Medical University, Shanghai 200438, China
- Department of Hepatobiliary, Pancreatic and Minimal Invasive Surgery, Zhejiang Provincial People’s Hospital (People’s Hospital of Hangzhou Medical College), Hangzhou 310000, Zhejiang Province, China
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Hangzhou 310000, Zhejiang Province, China
| |
Collapse
|
207
|
MAGEA4 Coated Extracellular Vesicles Are Stable and Can Be Assembled In Vitro. Int J Mol Sci 2021; 22:ijms22105208. [PMID: 34069064 PMCID: PMC8155938 DOI: 10.3390/ijms22105208] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/02/2021] [Accepted: 05/11/2021] [Indexed: 12/18/2022] Open
Abstract
Extracellular vesicles (EVs) are valued candidates for the development of new tools for medical applications. Vesicles carrying melanoma-associated antigen A (MAGEA) proteins, a subfamily of cancer-testis antigens, are particularly promising tools in the fight against cancer. Here, we have studied the biophysical and chemical properties of MAGEA4-EVs and show that they are stable under common storage conditions such as keeping at +4 °C and -80 °C for at least 3 weeks after purification. The MAGEA4-EVs can be freeze-thawed two times without losing MAGEA4 in detectable quantities. The attachment of MAGEA4 to the surface of EVs cannot be disrupted by high salt concentrations or chelators, but the vesicles are sensitive to high pH. The MAGEA4 protein can bind to the surface of EVs in vitro, using robust passive incubation. In addition, EVs can be loaded with recombinant proteins fused to the MAGEA4 open reading frame within the cells and also in vitro. The high stability of MAGEA4-EVs ensures their potential for the development of EV-based anti-cancer applications.
Collapse
|
208
|
Extracellular vesicles in immunomodulation and tumor progression. Nat Immunol 2021; 22:560-570. [PMID: 33753940 PMCID: PMC9389600 DOI: 10.1038/s41590-021-00899-0] [Citation(s) in RCA: 346] [Impact Index Per Article: 86.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 02/03/2021] [Indexed: 01/31/2023]
Abstract
Extracellular vesicles have emerged as prominent regulators of the immune response during tumor progression. EVs contain a diverse repertoire of molecular cargo that plays a critical role in immunomodulation. Here, we identify the role of EVs as mediators of communication between cancer and immune cells. This expanded role of EVs may shed light on the mechanisms behind tumor progression and provide translational diagnostic and prognostic tools for immunologists.
Collapse
|
209
|
Wu G, Geng H, Xu R, Deng M, Yang C, Xun C, Wang Y, Cai Q, Chen P. Preparation of a CaTiO 3/Al 3+/Pr 3+/Sm 3+ nanocomposite for enrichment of exosomes in human serum. Talanta 2021; 226:122186. [PMID: 33676717 DOI: 10.1016/j.talanta.2021.122186] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 01/28/2021] [Accepted: 01/30/2021] [Indexed: 10/22/2022]
Abstract
Exosomes (30-200 nm) play important roles in intercellular communication. Because their contents differ between healthy individuals and subjects diagnosed with various diseases, exosomes have been regarded as potential sources of biomarkers for clinical diagnosis. However, the accuracy of diagnosis by exosomal biomarkers is highly dependent on the extraction efficiency, yield, and the quality of exosomes. Hence, inexpensive, convenient, and fast exosome separation methods are required. In the present study, the CaTiO3/Al3+/Pr3+/Sm3+ nanocomposite was synthesized and applied in highly selective and efficient separation of exosomes. Notably, the developed material exhibited higher specificity and efficiency than commercially available TiO2. Moreover, CaTiO3/Al3+/Pr3+/Sm3+ could be reused at least three times without any significant decrease in efficiency. The synthesized material was also used for the extraction of exosomes from the serums of patients with Alzheimer's disease (AD) and healthy controls. The exosomes were subjected to two-dimensional gel electrophoresis (2-DE) separation and matrix-assisted laser desorption/ionization-time of flight (MALDI TOF/TOF) mass spectrometry analysis. It was found that five proteins in the exosomes were evidently upregulated, while one protein was downregulated. Among the detected proteins, serum amyloid P-component (SAP) has been reported to be closely related to pathogenesis of AD. The obtained results indicated that the developed method involving separation and analysis of serum exosomes could be used for disease diagnosis or postoperative clinical monitoring.
Collapse
Affiliation(s)
- Guangyao Wu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Hongchao Geng
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China
| | - Rongfang Xu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Min Deng
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Changcheng Yang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Chengfeng Xun
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Ying Wang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China
| | - Qingyun Cai
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, PR China.
| | - Ping Chen
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China; State Key Laboratory of Developmental Biology of Freshwater Fish, College of Life Sciences, Hunan Normal University, Changsha, 410081, PR China.
| |
Collapse
|
210
|
Lee NK, Kothandan VK, Kothandan S, Byun Y, Hwang SR. Exosomes and Cancer Stem Cells in Cancer Immunity: Current Reports and Future Directions. Vaccines (Basel) 2021; 9:vaccines9050441. [PMID: 34062950 PMCID: PMC8147426 DOI: 10.3390/vaccines9050441] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/21/2021] [Accepted: 04/27/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer stem cells (CSCs), which have the capacity to self-renew and differentiate into various types of cells, are notorious for their roles in tumor initiation, metastasis, and therapy resistance. Thus, underlying mechanisms for their survival provide key insights into developing effective therapeutic strategies. A more recent focus has been on exosomes that play a role in transmitting information between CSCs and non-CSCs, resulting in activating CSCs for cancer progression and modulating their surrounding microenvironment. The field of CSC-derived exosomes (CSCEXs) for different types of cancer is still under exploration. A deeper understanding and further investigation into CSCEXs’ roles in tumorigenicity and the identification of novel exosomal components are necessary for engineering exosomes for the treatment of cancer. Here, we review the features of CSCEXs, including surface markers, cargo, and biological or physiological functions. Further, reports on the immunomodulatory effects of CSCEXs are summarized, and exosome engineering for CSC-targeting is also discussed.
Collapse
Affiliation(s)
- Na-Kyeong Lee
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (N.-K.L.); (Y.B.)
| | - Vinoth Kumar Kothandan
- Department of Biomedical Sciences, Graduate School, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju 61452, Korea;
| | - Sangeetha Kothandan
- Department of Biotechnology, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai 600073, India;
| | - Youngro Byun
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea; (N.-K.L.); (Y.B.)
| | - Seung-Rim Hwang
- Department of Biomedical Sciences, Graduate School, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju 61452, Korea;
- College of Pharmacy, Chosun University, 309 Pilmun-daero, Dong-gu, Gwangju 61452, Korea
- Correspondence:
| |
Collapse
|
211
|
Xue D, Han J, Liu Y, Tuo H, Peng Y. Current perspectives on exosomes in the diagnosis and treatment of hepatocellular carcinoma (review). Cancer Biol Ther 2021; 22:279-290. [PMID: 33847207 PMCID: PMC8183537 DOI: 10.1080/15384047.2021.1898728] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The prognosis of hepatocellular carcinoma (HCC), a malignant tumor, is poor. Tumor recurrence and metastasis are the major challenges for the treatment of HCC. Various studies have demonstrated that exosomes, which are loaded with various biomolecules including nucleic acids, lipids, and proteins are involved in the recurrence and metastasis of HCC. Additionally, exosomes mediate various biological processes, such as immune response, cell apoptosis, angiogenesis, thrombosis, autophagy, and intercellular signal transduction. In cancer, exosomes regulate cancer cell differentiation, development, and drug resistance. Circular RNAs, microRNAs, and proteins in the exosomes can serve as early diagnostic and prognostic markers for HCC. As exosomes are characterized by low immunogenicity and high stability in the tissues and circulation, they can be used to deliver the drugs in cancer therapies.
Collapse
Affiliation(s)
- Dongdong Xue
- Department of Hepatobiliary Surgery, General Hospital, Shijiazhuang, Hebei, P. R. China
| | - Jingzhao Han
- Department of Hepatobiliary Surgery, General Hospital, Shijiazhuang, Hebei, P. R. China.,Department of Graduate School, Hebei Medical University, Shijiazhuang, P. R. China
| | - Yifan Liu
- Department of Hepatobiliary Surgery, General Hospital, Shijiazhuang, Hebei, P. R. China.,Department of Graduate School, Hebei Medical University, Shijiazhuang, P. R. China
| | - Hongfang Tuo
- Department of Hepatobiliary Surgery, General Hospital, Shijiazhuang, Hebei, P. R. China
| | - Yanhui Peng
- Department of Hepatobiliary Surgery, General Hospital, Shijiazhuang, Hebei, P. R. China
| |
Collapse
|
212
|
Qi X, Chen S, He H, Wen W, Wang H. The role and potential application of extracellular vesicles in liver cancer. SCIENCE CHINA-LIFE SCIENCES 2021; 64:1281-1294. [PMID: 33847910 DOI: 10.1007/s11427-020-1905-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 02/10/2021] [Indexed: 12/13/2022]
Abstract
Liver cancer is one of the most common causes of cancer-related death worldwide and mainly includes hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA). Extracellular vesicles (EVs) are membrane-derived nanometer-sized vesicles that can be released by different cell types under normal and pathological conditions and thus play important roles in the transmission of biological information between cells. Increasing evidence suggests that liver cancer cell-derived EVs may help establish a favorable microenvironment to support the proliferation, invasion and metastasis of cancer cells. In this review, we summarized the role of EVs in the tumor microenvironment (TME) during the development and progression of liver cancer. As messenger carriers, EVs are loaded by various biomolecules, such as proteins, RNA, DNA, lipids and metabolites, making them potential liquid biopsy biomarkers for the diagnosis and prognosis of liver cancer. We also highlighted the progress of EVs as antigen carriers and EV-based therapeutics in preclinical studies of liver cancer.
Collapse
Affiliation(s)
- Xuewei Qi
- Cancer Research Center, The First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China
| | - Shuzhen Chen
- National Center for Liver Cancer, Second Military Medical University, Shanghai, 200438, China
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China
| | - Huisi He
- National Center for Liver Cancer, Second Military Medical University, Shanghai, 200438, China
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China
| | - Wen Wen
- National Center for Liver Cancer, Second Military Medical University, Shanghai, 200438, China.
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China.
| | - Hongyang Wang
- Cancer Research Center, The First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230026, China.
- National Center for Liver Cancer, Second Military Medical University, Shanghai, 200438, China.
- International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China.
| |
Collapse
|
213
|
Leng L, Dong X, Gao X, Ran N, Geng M, Zuo B, Wu Y, Li W, Yan H, Han G, Yin H. Exosome-mediated improvement in membrane integrity and muscle function in dystrophic mice. Mol Ther 2021; 29:1459-1470. [PMID: 33333294 PMCID: PMC8058444 DOI: 10.1016/j.ymthe.2020.12.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 10/28/2020] [Accepted: 12/10/2020] [Indexed: 12/17/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a devastating genetic disorder that leads to compromised cellular membranes, caused by the absence of membrane-bound dystrophin protein. Muscle membrane leakage results in disrupted intracellular homeostasis, protein degradation, and muscle wasting. Improving muscle membrane integrity may delay disease progression and extend the lifespan of DMD patients. Here, we demonstrate that exosomes, membranous extracellular vesicles, can elicit functional improvements in dystrophic mice by improving muscle membrane integrity. Systemic administration of exosomes from different sources induced phenotypic rescue and mitigated pathological progression in dystrophic mice without detectable toxicity. Improved membrane integrity conferred by exosomes inhibited intracellular calcium influx and calcium-dependent activation of calpain proteases, preventing the degradation of the destabilized dystrophin-associated protein complex. We show that exosomes, particularly myotube-derived exosomes, induced functional improvements and alleviated muscle deterioration by stabilizing damaged muscle membrane in dystrophic mice. Our findings suggest that exosomes may have therapeutic implications for DMD and other diseases with compromised membranes.
Collapse
MESH Headings
- Animals
- Calcium/metabolism
- Calpain/genetics
- Cell Membrane/genetics
- Cell Membrane/pathology
- Disease Models, Animal
- Dystrophin/genetics
- Exosomes/genetics
- Exosomes/metabolism
- Humans
- Mice
- Mice, Inbred mdx
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Peptide Hydrolases/genetics
Collapse
Affiliation(s)
- Ling Leng
- Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Xue Dong
- Department of Pharmacology and Tianjin Key Laboratory of Inflammation Biology, Tianjin Medical University, Tianjin 300070, China; Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xianjun Gao
- Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Ning Ran
- Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Mengyuan Geng
- Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Bingfeng Zuo
- Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Yingjie Wu
- Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin 300070, China
| | - Wei Li
- Department of Technology, Tianjin Ever Union Biotechnology, Tianjin 301900, China
| | - Hua Yan
- Department of Ophthalmology, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Gang Han
- School of Medical Laboratory, Tianjin Medical University, Guangdong Road, Tianjin 300203, China.
| | - HaiFang Yin
- Tianjin Key Laboratory of Cellular Homeostasis and Human Diseases, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Department of Cell Biology, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin 300070, China; School of Medical Laboratory, Tianjin Medical University, Guangdong Road, Tianjin 300203, China; Department of Neurology, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
214
|
Wang X, He L, Huang X, Zhang S, Cao W, Che F, Zhu Y, Dai J. Recent Progress of Exosomes in Multiple Myeloma: Pathogenesis, Diagnosis, Prognosis and Therapeutic Strategies. Cancers (Basel) 2021; 13:cancers13071635. [PMID: 33915822 PMCID: PMC8037106 DOI: 10.3390/cancers13071635] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/20/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary In the pathogenesis of multiple myeloma (MM), some exosomes act on different cells in the bone marrow microenvironment, which can create an environment conducive to the survival and growth of MM cells. In addition, due to the abnormal expression of cargos in the exosomes of MM patients, exosomes may help with the diagnosis and prognosis of MM. In contrast to traditional nanomaterials, exosomes exhibit very good safety, biocompatibility, stability and biodegradability, which shows their potential for delivering anti-cancer drugs and cancer vaccines. Given the research in recent decades, exosomes are becoming increasingly relevant to MM. Although exosomes have not been applied in the clinic for help with diagnosing, prognosticating or providing therapy for MM, they are very promising for clinical applications concerning MM, which will possibly materialize in the near future. Therefore, this review is worth reading for further understanding of the important roles of exosomes in MM.. Abstract Multiple myeloma (MM) is a hematological malignancy that is still incurable. The bone marrow microenvironment (BMM), with cellular and non-cellular components, can create a favorable environment for the survival, proliferation and migration of MM cells, which is the main reason for the failure of MM therapies. Many studies have demonstrated that exosomes play an important role in the tumor-supportive BMM. Exosomes are nanoscale vesicles that can be released by various cells. Some exosomes contribute to the pathogenesis and progression of MM. MM-derived exosomes act on different cells in the BMM, thereby creating an environment conducive to the survival and growth of MM cells. Owing to the important roles of exosomes in the BMM, targeting the secretion of exosomes may become an effective therapeutic strategy for MM. In addition, the abnormal expression of “cargos” in the exosomes of MM patients may be used to diagnose MM or used as part of a screen for the early prognoses of MM patients. Exosomes also have good biological properties, including safety, biocompatibility, stability and biodegradability. Therefore, the encapsulation of anti-cancer drugs in exosomes, along with surface modifications of exosomes with targeting molecules, are very promising strategies for cancer therapies—particularly for MM. In addition, DC-derived exosomes (DC-EXs) can express MHC-I, MHC-II and T cell costimulatory molecules. Therefore, DC-EXs may be used as a nanocarrier to deliver cancer vaccines in MM. This review summarizes the recent progress of exosome research regarding the pathogenesis of, diagnosis of, prognosis of and therapeutic strategies for MM.
Collapse
Affiliation(s)
- Xi Wang
- Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu 610000, China; (X.W.); (L.H.); (X.H.); (S.Z.); (F.C.)
| | - Lin He
- Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu 610000, China; (X.W.); (L.H.); (X.H.); (S.Z.); (F.C.)
| | - Xiaobing Huang
- Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu 610000, China; (X.W.); (L.H.); (X.H.); (S.Z.); (F.C.)
| | - Shasha Zhang
- Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu 610000, China; (X.W.); (L.H.); (X.H.); (S.Z.); (F.C.)
| | - Wanjun Cao
- School of Pharmacy, North Sichuan Medical College, Nanchong 637000, China;
| | - Feifei Che
- Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu 610000, China; (X.W.); (L.H.); (X.H.); (S.Z.); (F.C.)
| | - Yizhun Zhu
- State Key Laboratory of Quality Research in Chinese Medicine & School of Pharmacy, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, China
- Correspondence: (Y.Z.); (J.D.); Tel.: +86-15756317270 (J.D.)
| | - Jingying Dai
- Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology of China, No. 32, West Section 2, First Ring Road, Qingyang District, Chengdu 610000, China; (X.W.); (L.H.); (X.H.); (S.Z.); (F.C.)
- Correspondence: (Y.Z.); (J.D.); Tel.: +86-15756317270 (J.D.)
| |
Collapse
|
215
|
Ding J, Wang J, Chen J. Exosomes as therapeutic vehicles in liver diseases. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:735. [PMID: 33987433 PMCID: PMC8106083 DOI: 10.21037/atm-20-5422] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The diagnosis and treatment of various liver diseases have progressed greatly over the years, but clinical outcomes are still not satisfying. New research on the mechanisms and application thereof may effectuate positive changes. Exosomes are membrane-derived nanovesicles ranging in size from 40 to 160 nm and are released by a diversity of cells. They contain a variety of cargo, including lipids, proteins, coding RNAs, and noncoding RNAs. Recent studies have recognized exosomes as intercellular communication agents, which play important roles in physiological or biological processes in acute or chronic liver disorders by horizontal transferring of genetic bioinformation from donor cells to neighboring or distal target cells. In the hope that exosomes can potentially be used as vehicles for clinical intervention, this review aims to focus on the roles of exosomes and their cargo in the field of various liver disorders, including virus-related liver diseases, alcoholic liver diseases (ALD), nonalcoholic fatty liver diseases (NAFLD), and liver cancer. In addition, many studies have indicated that mesenchymal stem cell (MSC)-derived exosomes or engineered MSC-derived exosomes can also exert hepatoprotection, antioxidation, or enhance drug sensitivity on corresponding liver diseases with the advantage of low immunogenicity and high biocompatibility. Overall, exosomes are expected to serve as an important therapeutic tool for various liver diseases. However, there are still many problems that need to be resolved by further research and a greater body of evidence before exosomes are ready for clinical application.
Collapse
Affiliation(s)
- Jingyi Ding
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ju Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiajia Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
216
|
Shi Y, Du L, Lv D, Li Y, Zhang Z, Huang X, Tang H. Emerging role and therapeutic application of exosome in hepatitis virus infection and associated diseases. J Gastroenterol 2021; 56:336-349. [PMID: 33665710 PMCID: PMC8005397 DOI: 10.1007/s00535-021-01765-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 01/23/2021] [Indexed: 02/05/2023]
Abstract
Hepatitis viruses are chief pathogens of hepatitis and end-stage liver diseases. Their replication and related pathogenic process highly rely on the host micro-environment and multiple cellular elements, including exosomes. Representing with a sort of cell-derived vesicle structure, exosomes were considered to be dispensable cellular components, even wastes. Along with advancing investigation, a specific profile of exosome in driving hepatitis viruses' infection and hepatic disease progression is revealed. Exosomes greatly affect the pathogenesis of hepatitis viruses by mediating their replication and modulating the host immune responses. The characteristics of host exosomes are markedly changed after infection with hepatitis viruses. Exosomes released from hepatitis virus-infected cells can carry viral nucleic or protein components, thereby acting as an effective subterfuge for hepatitis viruses by participating in viral transportation and immune escape. On the contrary, immune cell-derived exosomes contribute toward the innate antiviral immune defense and virus eradication. There is growing evidence supporting the application of exosomal biomarkers for predicting disease progress or therapeutic outcome, while exosomal nanoshuttles are regarded as promising therapeutic options based on their delivery properties and immune compatibility. In this review, we summarize the biogenesis and secretion mechanism of exosomes, review the recent findings pertaining to the role of exosomes in the interplay between hepatitis viruses and innate immune responses, and conclude their potential in further therapeutic application.
Collapse
Affiliation(s)
- Ying Shi
- Center of Infectious Diseases, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan, China
- School of Medicine, University of Electronic Science and Technology of China, No. 4 Section 2, North Jianshe Road, Chengdu, 610054, Sichuan, China
- Department of Hepatobiliary Surgery and Cell Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, No. 32 Western Section 2, 1st Ring Rd., Chengdu, 610072, Sichuan, China
| | - Lingyao Du
- Center of Infectious Diseases, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, No. 17 People's South Road, Chengdu, 610041, Sichuan, China
| | - Duoduo Lv
- Center of Infectious Diseases, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan, China
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, No. 17 People's South Road, Chengdu, 610041, Sichuan, China
| | - Yan Li
- School of Medicine, University of Electronic Science and Technology of China, No. 4 Section 2, North Jianshe Road, Chengdu, 610054, Sichuan, China
- Department of Hepatobiliary Surgery and Cell Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, No. 32 Western Section 2, 1st Ring Rd., Chengdu, 610072, Sichuan, China
| | - Zilong Zhang
- School of Medicine, University of Electronic Science and Technology of China, No. 4 Section 2, North Jianshe Road, Chengdu, 610054, Sichuan, China
- Department of Hepatobiliary Surgery and Cell Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, No. 32 Western Section 2, 1st Ring Rd., Chengdu, 610072, Sichuan, China
| | - Xiaolun Huang
- School of Medicine, University of Electronic Science and Technology of China, No. 4 Section 2, North Jianshe Road, Chengdu, 610054, Sichuan, China
- Department of Hepatobiliary Surgery and Cell Transplantation Center, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, No. 32 Western Section 2, 1st Ring Rd., Chengdu, 610072, Sichuan, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, No. 37 Guoxue Alley, Wuhou District, Chengdu, 610041, Sichuan, China.
- Division of Infectious Diseases, State Key Laboratory of Biotherapy and Center of Infectious Diseases, West China Hospital, Sichuan University, No. 17 People's South Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
217
|
Kim J, Song Y, Park CH, Choi C. Platform technologies and human cell lines for the production of therapeutic exosomes. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2021; 2:3-17. [PMID: 39698504 PMCID: PMC11648496 DOI: 10.20517/evcna.2020.01] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/18/2021] [Accepted: 01/27/2021] [Indexed: 12/20/2024]
Abstract
Exosomes are extracellular vesicles secreted by most cell types and represent various biological properties depending on their producing cells. They are also known to be important mediators of intercellular communication. Recent data suggest that exosomes can mediate the therapeutic effects of their parental cells; hence, they have been in the spotlight as novel therapeutics. To develop and manufacture effective therapeutic exosomes, customized strategies are needed to use appropriate technologies for exosome engineering and to select suitable production cell lines. In this review, we provide an overview of currently available exosome engineering platform technologies for loading active pharmaceutical ingredient cargo and the types of human cells/cell lines that are being used as exosome-producing cells, particularly focusing on their characteristics, advantages, and disadvantages.
Collapse
Affiliation(s)
- Jiyoon Kim
- ILIAS Biologics Inc., Daejeon 34014, South Korea
- Authors contributed equally
| | - Yonghee Song
- ILIAS Biologics Inc., Daejeon 34014, South Korea
- Authors contributed equally
| | | | - Chulhee Choi
- ILIAS Biologics Inc., Daejeon 34014, South Korea
- Department of Bio and Brain Engineering, KAIST, Daejeon 34141, South Korea
| |
Collapse
|
218
|
Chen W, Mao Y, Liu C, Wu H, Chen S. Exosome in Hepatocellular Carcinoma: an update. J Cancer 2021; 12:2526-2536. [PMID: 33854614 PMCID: PMC8040701 DOI: 10.7150/jca.54566] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 02/18/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor in the digestive tract with limited therapeutic choices. Intercellular communication among cancer cells and their microenvironment is crucial to disease progression. Exosomes are extracellular vesicles secreted by multiple types of cells into the extracellular space, which contain a variety of active components of secretory cells, including lipids, proteins, RNA and DNA. This vesicle structure involves in the exchange of materials and information between cells and plays an important role in the development of many diseases. Studies have shown that exosomes participate in the communication between HCC cells and non-HCC cells and regulate the occurrence and development of hepatocellular carcinoma. Therefore, exosomes may be specific biomarkers for early diagnosis and metastasis of HCC, which are also potential targets for the treatment of HCC. This review summarizes the characteristic, types and biological functions of exosomes and discusses their research progress and application prospects in the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Wei Chen
- Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Yinqi Mao
- Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Chenbin Liu
- Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Han Wu
- Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Shuying Chen
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
219
|
Naseer M, Hadi S, Syed A, Safdari A, Tahan V. Exosomes: A new frontier under the spotlight for diagnosis and treatment of gastrointestinal diseases. World J Meta-Anal 2021; 9:12-28. [DOI: 10.13105/wjma.v9.i1.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 12/15/2020] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
|
220
|
Pi YN, Xia BR, Jin MZ, Jin WL, Lou G. Exosomes: Powerful weapon for cancer nano-immunoengineering. Biochem Pharmacol 2021; 186:114487. [PMID: 33647264 DOI: 10.1016/j.bcp.2021.114487] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/12/2021] [Accepted: 02/19/2021] [Indexed: 12/15/2022]
Abstract
Cancer immunotherapy (CIT) that targets the tumor immune microenvironment is regarded as a revolutionary advancement in the fight against cancer. The success and failure of CIT are due to the complexity of the immunosuppressive microenvironment. Cancer nanomedicine is a potential adjuvant therapeutic strategy for immune-based combination therapy. Exosomes are natural nanomaterials that play a pivotal role in mediating intercellular communications and package delivery in the tumor microenvironment. They affect the immune response or the effectiveness of immunotherapy. In particular, exosomal PD-L1 promotes cancer progression and resistance to immunotherapy. Exosomes possess high bioavailability, biological stability, targeting specificity, low toxicity, and immune characteristics, which indicate their potential for cancer therapy. They can be engineered to act as effective cancer therapeutic tools that activate anti-tumor immune response and start immune surveillance. In the current review, we introduce the role of exosomes in a tumor immune microenvironment, highlight the application of engineered exosomes to CIT, and discuss the challenges and prospects for clinical application.
Collapse
Affiliation(s)
- Ya-Nan Pi
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin 150086, PR China
| | - Bai-Rong Xia
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei, Anhui 230031, PR China
| | - Ming-Zhu Jin
- Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China.
| | - Ge Lou
- Department of Gynecology, Harbin Medical University Cancer Hospital, Harbin 150086, PR China.
| |
Collapse
|
221
|
Yin L, Zhou L, Xu R. Identification of Tumor Mutation Burden and Immune Infiltrates in Hepatocellular Carcinoma Based on Multi-Omics Analysis. Front Mol Biosci 2021; 7:599142. [PMID: 33681288 PMCID: PMC7928364 DOI: 10.3389/fmolb.2020.599142] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 12/21/2020] [Indexed: 12/16/2022] Open
Abstract
We aimed to explore the tumor mutational burden (TMB) and immune infiltration in HCC and investigate new biomarkers for immunotherapy. Transcriptome and gene mutation data were downloaded from the GDC portal, including 374 HCC samples and 50 matched normal samples. Furthermore, we divided the samples into high and low TMB groups, and analyzed the differential genes between them with GO, KEGG, and GSEA. Cibersort was used to assess the immune cell infiltration in the samples. Finally, univariate and multivariate Cox regression analyses were performed to identify differential genes related to TMB and immune infiltration, and a risk prediction model was constructed. We found 10 frequently mutated genes, including TP53, TTN, CTNNB1, MUC16, ALB, PCLO, MUC, APOB, RYR2, and ABCA. Pathway analysis indicated that these TMB-related differential genes were mainly enriched in PI3K-AKT. Cibersort analysis showed that memory B cells (p = 0.02), CD8+ T cells (p = 0.09), CD4+ memory activated T cells (p = 0.07), and neutrophils (p = 0.06) demonstrated a difference in immune infiltration between high and low TMB groups. On multivariate analysis, GABRA3 (p = 0.05), CECR7 (p < 0.001), TRIM16 (p = 0.003), and IL7R (p = 0.04) were associated with TMB and immune infiltration. The risk prediction model had an area under the curve (AUC) of 0.69, suggesting that patients with low risk had better survival outcomes. Our study demonstrated for the first time that CECR7, GABRA3, IL7R, and TRIM16L were associated with TMB and promoted antitumor immunity in HCC.
Collapse
Affiliation(s)
- Lu Yin
- Department of Pathology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liuzhi Zhou
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Rujun Xu
- Department of Pathology, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
222
|
Chen F, Wang Y, Gao J, Saeed M, Li T, Wang W, Yu H. Nanobiomaterial-based vaccination immunotherapy of cancer. Biomaterials 2021; 270:120709. [PMID: 33581608 DOI: 10.1016/j.biomaterials.2021.120709] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 01/27/2021] [Accepted: 01/31/2021] [Indexed: 12/15/2022]
Abstract
Cancer immunotherapies including cancer vaccines, immune checkpoint blockade or chimeric antigen receptor T cells have been exploited as the attractive treatment modalities in recent years. Among these approaches, cancer vaccines that designed to deliver tumor antigens and adjuvants to activate the antigen presenting cells (APCs) and induce antitumor immune responses, have shown significant efficacy in inhibiting tumor growth, preventing tumor relapse and metastasis. Despite the potential of cancer vaccination strategies, the therapeutic outcomes in preclinical trials are failed to promote their clinical translation, which is in part due to their inefficient vaccination cascade of five critical steps: antigen identification, antigen encapsulation, antigen delivery, antigen release and antigen presentation to T cells. In recent years, it has been demonstrated that various nanobiomaterials hold great potential to enhance cancer vaccination cascade and improve their antitumor performance and reduce the off-target effect. We summarize the cutting-edge advances of nanobiomaterials-based vaccination immunotherapy of cancer in this review. The various cancer nanovaccines including antigen peptide/adjuvant-based nanovaccines, nucleic acid-based nanovaccines as well as biomimetic nanobiomaterials-based nanovaccines are discussed in detail. We also provide some challenges and perspectives associated with the clinical translation of cancer nanovaccines.
Collapse
Affiliation(s)
- Fangmin Chen
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yingjie Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; Nano Science and Technology Institute, University of Science and Technology of China, Suzhou, 215123, China
| | - Jing Gao
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Madiha Saeed
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Tianliang Li
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Weiqi Wang
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Haijun Yu
- State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
223
|
Samuel M, Gabrielsson S. Personalized medicine and back-allogeneic exosomes for cancer immunotherapy. J Intern Med 2021; 289:138-146. [PMID: 31359504 DOI: 10.1111/joim.12963] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 04/17/2019] [Accepted: 05/13/2019] [Indexed: 12/12/2022]
Abstract
Extracellular vesicles, especially exosomes, have been explored for cancer immunotherapy. The initial studies made use of autologous B-cell or dendritic cell-derived exosomes, with the idea that MHC-peptide complexes on the exosomal surface would stimulate an MHC-restricted cancer-specific immune response. This was also verified in mouse systems, whilst the effects in human clinical systems were more modest. Several studies have explored the mechanisms for exosomal T-cell activation, and a picture emerges where the antigen-presenting cells, possibly both B cells and dendritic cells of the recipient, are needed to induce a potent T-cell response to exosomes. Therefore, the exosomes function more as an adjuvant-like delivery system of antigens, and we need to further understand the exact components that trigger the most broad and potent immune responses. Here, we describe the grounds for using allogeneic exosomes for cancer therapy, something that would greatly improve the feasibility of new exosome-based immunotherapeutic approaches to cure cancer.
Collapse
Affiliation(s)
- M Samuel
- From the, Division of Immunology and Allergy, Department of Medicine, Karolinska Institutet, Solna, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - S Gabrielsson
- From the, Division of Immunology and Allergy, Department of Medicine, Karolinska Institutet, Solna, Sweden.,Department of Clinical Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
224
|
Sinha D, Roy S, Saha P, Chatterjee N, Bishayee A. Trends in Research on Exosomes in Cancer Progression and Anticancer Therapy. Cancers (Basel) 2021; 13:cancers13020326. [PMID: 33477340 PMCID: PMC7829710 DOI: 10.3390/cancers13020326] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 01/09/2021] [Accepted: 01/14/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Intensive research in the field of cancer biology has discovered a unique mode of interplay between cells via extracellular bioactive vesicles called exosomes. Exosomes serve as intermediators among cells via their cargoes that, in turn, contribute in the progression of cancer. They are ubiquitously present in all body fluids as they are secreted from both normal and tumor cells. These minuscules exhibit multiple unique properties that facilitate their migration to distant locations and modulate the microenvironment for progression of cancer. This review summarizes the multifarious role of exosomes in various aspects of cancer research with its pros and cons. It discusses biogenesis of exosomes, their functional role in cancer metastasis, both protumorigenic and antitumorigenic, and also their applications in anticancer therapy. Abstract Exosomes, the endosome-derived bilayered extracellular nanovesicles with their contribution in many aspects of cancer biology, have become one of the prime foci of research. Exosomes derived from various cells carry cargoes similar to their originator cells and their mode of generation is different compared to other extracellular vesicles. This review has tried to cover all aspects of exosome biogenesis, including cargo, Rab-dependent and Rab-independent secretion of endosomes and exosomal internalization. The bioactive molecules of the tumor-derived exosomes, by virtue of their ubiquitous presence and small size, can migrate to distal parts and propagate oncogenic signaling and epigenetic regulation, modulate tumor microenvironment and facilitate immune escape, tumor progression and drug resistance responsible for cancer progression. Strategies improvised against tumor-derived exosomes include suppression of exosome uptake, modulation of exosomal cargo and removal of exosomes. Apart from the protumorigenic role, exosomal cargoes have been selectively manipulated for diagnosis, immune therapy, vaccine development, RNA therapy, stem cell therapy, drug delivery and reversal of chemoresistance against cancer. However, several challenges, including in-depth knowledge of exosome biogenesis and protein sorting, perfect and pure isolation of exosomes, large-scale production, better loading efficiency, and targeted delivery of exosomes, have to be confronted before the successful implementation of exosomes becomes possible for the diagnosis and therapy of cancer.
Collapse
Affiliation(s)
- Dona Sinha
- Department of Receptor Biology and Tumour Metastasis, Chittaranjan National Cancer Institute, Kolkata 700 026, India; (S.R.); (P.S.); (N.C.)
- Correspondence: or (D.S.); or (A.B.)
| | - Sraddhya Roy
- Department of Receptor Biology and Tumour Metastasis, Chittaranjan National Cancer Institute, Kolkata 700 026, India; (S.R.); (P.S.); (N.C.)
| | - Priyanka Saha
- Department of Receptor Biology and Tumour Metastasis, Chittaranjan National Cancer Institute, Kolkata 700 026, India; (S.R.); (P.S.); (N.C.)
| | - Nabanita Chatterjee
- Department of Receptor Biology and Tumour Metastasis, Chittaranjan National Cancer Institute, Kolkata 700 026, India; (S.R.); (P.S.); (N.C.)
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
- Correspondence: or (D.S.); or (A.B.)
| |
Collapse
|
225
|
Soluble and Exosome-Bound α-Galactosylceramide Mediate Preferential Proliferation of Educated NK Cells with Increased Anti-Tumor Capacity. Cancers (Basel) 2021; 13:cancers13020298. [PMID: 33467442 PMCID: PMC7830699 DOI: 10.3390/cancers13020298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/09/2021] [Accepted: 01/12/2021] [Indexed: 12/27/2022] Open
Abstract
Natural killer (NK) cells can kill target cells via the recognition of stress molecules and down-regulation of major histocompatibility complex class I (MHC-I). Some NK cells are educated to recognize and kill cells that have lost their MHC-I expression, e.g., tumor or virus-infected cells. A desired property of cancer immunotherapy is, therefore, to activate educated NK cells during anti-tumor responses in vivo. We here analyze NK cell responses to α-galactosylceramide (αGC), a potent activator of invariant NKT (iNKT) cells, or to exosomes loaded with αGC. In mouse strains which express different MHC-I alleles using an extended NK cell flow cytometry panel, we show that αGC induces a biased NK cell proliferation of educated NK cells. Importantly, iNKT cell-induced activation of NK cells selectively increased in vivo missing self-responses, leading to more effective rejection of tumor cells. Exosomes from antigen-presenting cells are attractive anti-cancer therapy tools as they may induce both innate and adaptive immune responses, thereby addressing the hurdle of tumor heterogeneity. Adding αGC to antigen-loaded dendritic-cell-derived exosomes also led to an increase in missing self-responses in addition to boosted T and B cell responses. This study manifests αGC as an attractive adjuvant in cancer immunotherapy, as it increases the functional capacity of educated NK cells and enhances the innate, missing self-based antitumor response.
Collapse
|
226
|
Roles of Bile-Derived Exosomes in Hepatobiliary Disease. BIOMED RESEARCH INTERNATIONAL 2021; 2021:8743409. [PMID: 33511212 PMCID: PMC7822672 DOI: 10.1155/2021/8743409] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 12/23/2020] [Accepted: 01/06/2021] [Indexed: 12/14/2022]
Abstract
Exosomes are vesicles with a diameter of 30-150 nm produced by living cells and secreted into the extracellular matrix. Exosomes mediate cellular communication by carrying active molecules, such as nucleic acids, proteins, and liposomes. Although exosomes are found in various body fluids, little is known about bile-derived exosomes. This review is the first to summarize the methods of bile storage and isolation of biliary exosomes, highlighting the roles of bile-derived exosomes, especially exosomal noncoding RNAs, in physiological and disease states and discussing their potential clinical applications.
Collapse
|
227
|
Exosomes in Immune Regulation. Noncoding RNA 2021; 7:ncrna7010004. [PMID: 33435564 PMCID: PMC7838779 DOI: 10.3390/ncrna7010004] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/26/2020] [Accepted: 01/04/2021] [Indexed: 02/08/2023] Open
Abstract
Exosomes, small extracellular vesicles mediate intercellular communication by transferring their cargo including DNA, RNA, proteins and lipids from cell to cell. Notably, in the immune system, they have protective functions. However in cancer, exosomes acquire new, immunosuppressive properties that cause the dysregulation of immune cells and immune escape of tumor cells supporting cancer progression and metastasis. Therefore, current investigations focus on the regulation of exosome levels for immunotherapeutic interventions. In this review, we discuss the role of exosomes in immunomodulation of lymphoid and myeloid cells, and their use as immune stimulatory agents to elicit specific cytotoxic responses against the tumor.
Collapse
|
228
|
Hong GQ, Cai D, Gong JP, Lai X. Innate immune cells and their interaction with T cells in hepatocellular carcinoma. Oncol Lett 2021; 21:57. [PMID: 33281968 PMCID: PMC7709558 DOI: 10.3892/ol.2020.12319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/08/2020] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor and is associated with necroinflammation driven by various immune cells, such as dendritic cells, macrophages and natural killer cells. Innate immune cells can directly affect HCC or regulate the T-cell responses that mediate HCC. In addition, innate immune cells and T cells are not isolated, which means the interaction between them is important in the HCC microenvironment. Considering the current unsatisfactory efficacy of immunotherapy in patients with HCC, understanding the relationship between innate immune cells and T cells is necessary. In the present review the roles and clinical value of innate immune cells that have been widely reported to be involved in HCC, including dendritic cells, macrophages (including kupffer cells), neutrophils, eosinophils, basophils and innate lymphoid cells and the crosstalk between the innate and adaptive immune responses in the antitumor process have been discussed. The present review will facilitate researchers in understanding the importance of innate immune cells in HCC and lead to innovative immunotherapy approaches for the treatment of HCC.
Collapse
Affiliation(s)
- Guo-Qing Hong
- Department of Hepatobiliary and Thyroid Breast Surgery, Tongnan District People's Hospital, Chongqing 402660, P.R. China
| | - Dong Cai
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Jian-Ping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Xing Lai
- Department of Hepatobiliary and Thyroid Breast Surgery, Tongnan District People's Hospital, Chongqing 402660, P.R. China
- Correspondence to: Dr Xing Lai, Department of Hepatobiliary and Thyroid Breast Surgery, Tongnan District People's Hospital, 271 Datong, Tongnan, Chongqing 402660, P.R. China, E-mail:
| |
Collapse
|
229
|
Xu J, Ji L, Liang Y, Wan Z, Zheng W, Song X, Gorshkov K, Sun Q, Lin H, Zheng X, Chen J, Jin RA, Liang X, Cai X. CircRNA-SORE mediates sorafenib resistance in hepatocellular carcinoma by stabilizing YBX1. Signal Transduct Target Ther 2020; 5:298. [PMID: 33361760 PMCID: PMC7762756 DOI: 10.1038/s41392-020-00375-5] [Citation(s) in RCA: 296] [Impact Index Per Article: 59.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 07/19/2020] [Accepted: 07/31/2020] [Indexed: 12/24/2022] Open
Abstract
Sorafenib is the first-line chemotherapeutic therapy for advanced hepatocellular carcinoma (HCC). However, sorafenib resistance significantly limits its therapeutic efficacy, and the mechanisms underlying resistance have not been fully clarified. Here we report that a circular RNA, circRNA-SORE (a circular RNA upregulated in sorafenib-resistant HCC cells), plays a significant role in sorafenib resistance in HCC. We found that circRNA-SORE is upregulated in sorafenib-resistant HCC cells and depletion of circRNA-SORE substantially increases the cell-killing ability of sorafenib. Further studies revealed that circRNA-SORE binds the master oncogenic protein YBX1 in the cytoplasm, which prevents YBX1 nuclear interaction with the E3 ubiquitin ligase PRP19 and thus blocks PRP19-mediated YBX1 degradation. Moreover, our in vitro and in vivo results suggest that circRNA-SORE is transported by exosomes to spread sorafenib resistance among HCC cells. Using different HCC mouse models, we demonstrated that silencing circRNA-SORE by injection of siRNA could substantially overcome sorafenib resistance. Our study provides a proof-of-concept demonstration for a potential strategy to overcome sorafenib resistance in HCC patients by targeting circRNA-SORE or YBX1.
Collapse
Affiliation(s)
- Junjie Xu
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, China.,Zhejiang University Cancer Center, 310016, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treantment Thechnology Research Center of Severe Hepatobiliary Disease, 310016, Hangzhou, China
| | - Lin Ji
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, China.,Zhejiang University Cancer Center, 310016, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treantment Thechnology Research Center of Severe Hepatobiliary Disease, 310016, Hangzhou, China
| | - Yuelong Liang
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, China.,Zhejiang University Cancer Center, 310016, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treantment Thechnology Research Center of Severe Hepatobiliary Disease, 310016, Hangzhou, China
| | - Zhe Wan
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, China.,Zhejiang University Cancer Center, 310016, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treantment Thechnology Research Center of Severe Hepatobiliary Disease, 310016, Hangzhou, China
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD, 20892, USA
| | - Xiaomin Song
- State Key Laboratory of Molecular Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, 200031, Shanghai, China
| | - Kirill Gorshkov
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD, 20892, USA
| | - Qiming Sun
- Department of Biochemistry, and Department of Cardiology of the Second Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Hui Lin
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, China.,Zhejiang University Cancer Center, 310016, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treantment Thechnology Research Center of Severe Hepatobiliary Disease, 310016, Hangzhou, China
| | - Xueyong Zheng
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, China.,Zhejiang University Cancer Center, 310016, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treantment Thechnology Research Center of Severe Hepatobiliary Disease, 310016, Hangzhou, China
| | - Jiang Chen
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, China.,Zhejiang University Cancer Center, 310016, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treantment Thechnology Research Center of Severe Hepatobiliary Disease, 310016, Hangzhou, China
| | - Ren-An Jin
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, China.,Zhejiang University Cancer Center, 310016, Hangzhou, China.,Zhejiang Minimal Invasive Diagnosis and Treantment Thechnology Research Center of Severe Hepatobiliary Disease, 310016, Hangzhou, China
| | - Xiao Liang
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, China. .,Zhejiang University Cancer Center, 310016, Hangzhou, China. .,Zhejiang Minimal Invasive Diagnosis and Treantment Thechnology Research Center of Severe Hepatobiliary Disease, 310016, Hangzhou, China.
| | - Xiujun Cai
- Key Laboratory of Laparoscopic Technology of Zhejiang Province, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Hangzhou, China. .,Zhejiang University Cancer Center, 310016, Hangzhou, China. .,Zhejiang Minimal Invasive Diagnosis and Treantment Thechnology Research Center of Severe Hepatobiliary Disease, 310016, Hangzhou, China.
| |
Collapse
|
230
|
Exosomes: Cell-Derived Nanoplatforms for the Delivery of Cancer Therapeutics. Int J Mol Sci 2020; 22:ijms22010014. [PMID: 33374978 PMCID: PMC7792591 DOI: 10.3390/ijms22010014] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/17/2020] [Accepted: 12/18/2020] [Indexed: 12/17/2022] Open
Abstract
Exosomes are cell-secreted nanovesicles that naturally contain biomolecular cargoes such as lipids, proteins, and nucleic acids. Exosomes mediate intercellular communication, enabling the transfer biological signals from the donor cells to the recipient cells. Recently, exosomes are emerging as promising drug delivery vehicles due to their strong stability in blood circulation, high biocompatibility, low immunogenicity, and natural targeting ability. In particular, exosomes derived from specific types of cells can carry endogenous signaling molecules with therapeutic potential for cancer treatment, thus presenting a significant impact on targeted drug delivery and therapy. Furthermore, exosomes can be engineered to display targeting moieties on their surface or to load additional therapeutic agents. Therefore, a comprehensive understanding of exosome biogenesis and the development of efficient exosome engineering techniques will provide new avenues to establish convincing clinical therapeutic strategies based on exosomes. This review focuses on the therapeutic applications of exosomes derived from various cells and the exosome engineering technologies that enable the accurate delivery of various types of cargoes to target cells for cancer therapy.
Collapse
|
231
|
Yong T, Li X, Wei Z, Gan L, Yang X. Extracellular vesicles-based drug delivery systems for cancer immunotherapy. J Control Release 2020; 328:562-574. [DOI: 10.1016/j.jconrel.2020.09.028] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022]
|
232
|
Yang B, Gao J, Pei Q, Xu H, Yu H. Engineering Prodrug Nanomedicine for Cancer Immunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2002365. [PMID: 33304763 PMCID: PMC7709995 DOI: 10.1002/advs.202002365] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/16/2020] [Indexed: 12/11/2022]
Abstract
Immunotherapy has shifted the clinical paradigm of cancer management. However, despite promising initial progress, immunotherapeutic approaches to cancer still suffer from relatively low response rates and the possibility of severe side effects, likely due to the low inherent immunogenicity of tumor cells, the immunosuppressive tumor microenvironment, and significant inter- and intratumoral heterogeneity. Recently, nanoformulations of prodrugs have been explored as a means to enhance cancer immunotherapy by simultaneously eliciting antitumor immune responses and reversing local immunosuppression. Prodrug nanomedicines, which integrate engineering advances in chemistry, oncoimmunology, and material science, are rationally designed through chemically modifying small molecule drugs, peptides, or antibodies to yield increased bioavailability and spatiotemporal control of drug release and activation at the target sites. Such strategies can help reduce adverse effects and enable codelivery of multiple immune modulators to yield synergistic cancer immunotherapy. In this review article, recent advances and translational challenges facing prodrug nanomedicines for cancer immunotherapy are overviewed. Last, key considerations are outlined for future efforts to advance prodrug nanomedicines aimed to improve antitumor immune responses and combat immune tolerogenic microenvironments.
Collapse
Affiliation(s)
- Bin Yang
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteTongji University School of MedicineTongji University Cancer CenterShanghai200072China
| | - Jing Gao
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteTongji University School of MedicineTongji University Cancer CenterShanghai200072China
| | - Qing Pei
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| | - Huixiong Xu
- Department of Medical UltrasoundShanghai Tenth People's HospitalUltrasound Research and Education InstituteTongji University School of MedicineTongji University Cancer CenterShanghai200072China
| | - Haijun Yu
- State Key Laboratory of Drug Research & Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of SciencesShanghai201203China
| |
Collapse
|
233
|
Li S, Yi M, Dong B, Jiao Y, Luo S, Wu K. The roles of exosomes in cancer drug resistance and its therapeutic application. Clin Transl Med 2020; 10:e257. [PMID: 33377643 PMCID: PMC7752167 DOI: 10.1002/ctm2.257] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/05/2020] [Accepted: 12/06/2020] [Indexed: 12/14/2022] Open
Abstract
Exosomes are a category of extracellular vesicles with a size ranging from 40 to 160 nm, which can be secreted by multiple cells in the tumor microenvironment. Exosomes serve as communicators in regulating biological functions and pathological processes, including drug response. Through transporting the cargo such as protein or nucleic acid, exosomes can modulate drug sensitivity via multiple mechanisms. Additionally, exosomes can be deployed as a delivery system to treat cancer due to their high-efficient loading capacity and tolerable toxicity. Recent studies have demonstrated the high efficacy of exosomes in cancer therapy. Herein, we conduct this review to summarize the mechanism of exosome-mediated drug resistance and the therapeutic potential of exosomes in cancer.
Collapse
Affiliation(s)
- Shiyu Li
- Department of OncologyTongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ming Yi
- Department of OncologyTongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Bing Dong
- Department of Molecular PathologyThe Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| | - Ying Jiao
- Department of OncologyTongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Suxia Luo
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| | - Kongming Wu
- Department of OncologyTongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer HospitalZhengzhouChina
| |
Collapse
|
234
|
Immune Regulation by Dendritic Cell Extracellular Vesicles in Cancer Immunotherapy and Vaccines. Cancers (Basel) 2020; 12:cancers12123558. [PMID: 33260499 PMCID: PMC7761478 DOI: 10.3390/cancers12123558] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/18/2020] [Accepted: 11/25/2020] [Indexed: 02/07/2023] Open
Abstract
Extracellular vesicles (EVs) play a crucial role in intercellular communication as vehicles for the transport of membrane and cytosolic proteins, lipids, and nucleic acids including different RNAs. Dendritic cells (DCs)-derived EVs (DEVs), albeit variably, express major histocompatibility complex (MHC)-peptide complexes and co-stimulatory molecules on their surface that enable the interaction with other immune cells such as CD8+ T cells, and other ligands that stimulate natural killer (NK) cells, thereby instructing tumor rejection, and counteracting immune-suppressive tumor microenvironment. Malignant cells oppose this effect by secreting EVs bearing a variety of molecules that block DCs function. For instance, tumor-derived EVs (TDEVs) can impair myeloid cell differentiation resulting in myeloid-derived suppressor cells (MDSCs) generation. Hence, the unique composition of EVs makes them suitable candidates for the development of new cancer treatment approaches including prophylactic vaccine targeting oncogenic pathogens, cancer vaccines, and cancer immunotherapeutics. We offer a perspective from both cell sides, DCs, and tumor cells, on how EVs regulate the antitumor immune response, and how this translates into promising therapeutic options by reviewing the latest advancement in DEV-based cancer therapeutics.
Collapse
|
235
|
Fu C, Zhou L, Mi QS, Jiang A. DC-Based Vaccines for Cancer Immunotherapy. Vaccines (Basel) 2020; 8:vaccines8040706. [PMID: 33255895 PMCID: PMC7712957 DOI: 10.3390/vaccines8040706] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/16/2020] [Accepted: 11/24/2020] [Indexed: 12/15/2022] Open
Abstract
As the sentinels of the immune system, dendritic cells (DCs) play a critical role in initiating and regulating antigen-specific immune responses. Cross-priming, a process that DCs activate CD8 T cells by cross-presenting exogenous antigens onto their MHCI (Major Histocompatibility Complex class I), plays a critical role in mediating CD8 T cell immunity as well as tolerance. Current DC vaccines have remained largely unsuccessful despite their ability to potentiate both effector and memory CD8 T cell responses. There are two major hurdles for the success of DC-based vaccines: tumor-mediated immunosuppression and the functional limitation of the commonly used monocyte-derived dendritic cells (MoDCs). Due to their resistance to tumor-mediated suppression as inert vesicles, DC-derived exosomes (DCexos) have garnered much interest as cell-free therapeutic agents. However, current DCexo clinical trials have shown limited clinical benefits and failed to generate antigen-specific T cell responses. Another exciting development is the use of naturally circulating DCs instead of in vitro cultured DCs, as clinical trials with both human blood cDC2s (type 2 conventional DCs) and plasmacytoid DCs (pDCs) have shown promising results. pDC vaccines were particularly encouraging, especially in light of promising data from a recent clinical trial using a human pDC cell line, despite pDCs being considered tolerogenic and playing a suppressive role in tumors. However, how pDCs generate anti-tumor CD8 T cell immunity remains poorly understood, thus hindering their clinical advance. Using a pDC-targeted vaccine model, we have recently reported that while pDC-targeted vaccines led to strong cross-priming and durable CD8 T cell immunity, cross-presenting pDCs required cDCs to achieve cross-priming in vivo by transferring antigens to cDCs. Antigen transfer from pDCs to bystander cDCs was mediated by pDC-derived exosomes (pDCexos), which similarly required cDCs for cross-priming of antigen-specific CD8 T cells. pDCexos thus represent a new addition in our arsenal of DC-based cancer vaccines that would potentially combine the advantage of pDCs and DCexos.
Collapse
Affiliation(s)
- Chunmei Fu
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI 48202, USA; (C.F.); (L.Z.); (Q.-S.M.)
| | - Li Zhou
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI 48202, USA; (C.F.); (L.Z.); (Q.-S.M.)
- Immunology Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI 48202, USA
| | - Qing-Sheng Mi
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI 48202, USA; (C.F.); (L.Z.); (Q.-S.M.)
- Immunology Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI 48202, USA
| | - Aimin Jiang
- Center for Cutaneous Biology and Immunology, Department of Dermatology, Henry Ford Health System, Detroit, MI 48202, USA; (C.F.); (L.Z.); (Q.-S.M.)
- Immunology Program, Henry Ford Cancer Institute, Henry Ford Health System, Detroit, MI 48202, USA
- Correspondence: ; Tel.: +1-716-400-2536
| |
Collapse
|
236
|
Nikfarjam S, Rezaie J, Kashanchi F, Jafari R. Dexosomes as a cell-free vaccine for cancer immunotherapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:258. [PMID: 33228747 PMCID: PMC7686678 DOI: 10.1186/s13046-020-01781-x] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/13/2020] [Indexed: 12/30/2022]
Abstract
Dendritic cells (DCs) secrete vast quantities of exosomes termed as dexosomes. Dexosomes are symmetric nanoscale heat-stable vesicles that consist of a lipid bilayer displaying a characteristic series of lipid and protein molecules. They include tetraspanins and all established proteins for presenting antigenic material such as the major histocompatibility complex class I/II (MHC I/II) and CD1a, b, c, d proteins and CD86 costimulatory molecule. Dexosomes contribute to antigen-specific cellular immune responses by incorporating the MHC proteins with antigen molecules and transferring the antigen-MHC complexes and other associated molecules to naïve DCs. A variety of ex vivo and in vivo studies demonstrated that antigen-loaded dexosomes were able to initiate potent antitumor immunity. Human dexosomes can be easily prepared using monocyte-derived DCs isolated by leukapheresis of peripheral blood and treated ex vivo by cytokines and other factors. The feasibility of implementing dexosomes as therapeutic antitumor vaccines has been verified in two phase I and one phase II clinical trials in malignant melanoma and non small cell lung carcinoma patients. These studies proved the safety of dexosome administration and showed that dexosome vaccines have the capacity to trigger both the adaptive (T lymphocytes) and the innate (natural killer cells) immune cell recalls. In the current review, we will focus on the perspective of utilizing dexosome vaccines in the context of cancer immunotherapy.
Collapse
Affiliation(s)
- Sepideh Nikfarjam
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, P.O. Box: 1138, Shafa St, Ershad Blvd., 57147, Urmia, Iran
| | - Fatah Kashanchi
- School of Systems Biology, Laboratory of Molecular Virology, George Mason University, Discovery Hall Room 182, 10900 University Blvd., VA, 20110, Manassas, USA.
| | - Reza Jafari
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, P.O. Box: 1138, Shafa St, Ershad Blvd., 57147, Urmia, Iran. .,Department of Immunology and Genetics, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
237
|
Atypical immunometabolism and metabolic reprogramming in liver cancer: Deciphering the role of gut microbiome. Adv Cancer Res 2020; 149:171-255. [PMID: 33579424 DOI: 10.1016/bs.acr.2020.10.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related mortality worldwide. Much recent research has delved into understanding the underlying molecular mechanisms of HCC pathogenesis, which has revealed to be heterogenous and complex. Two major hallmarks of HCC include: (i) a hijacked immunometabolism and (ii) a reprogramming in metabolic processes. We posit that the gut microbiota is a third component in an entanglement triangle contributing to HCC progression. Besides metagenomic studies highlighting the diagnostic potential in the gut microbiota profile, recent research is pinpointing the gut microbiota as an instigator, not just a mere bystander, in HCC. In this chapter, we discuss mechanistic insights on atypical immunometabolism and metabolic reprogramming in HCC, including the examination of tumor-associated macrophages and neutrophils, tumor-infiltrating lymphocytes (e.g., T-cell exhaustion, regulatory T-cells, natural killer T-cells), the Warburg effect, rewiring of the tricarboxylic acid cycle, and glutamine addiction. We further discuss the potential involvement of the gut microbiota in these characteristics of hepatocarcinogenesis. An immediate highlight is that microbiota metabolites (e.g., short chain fatty acids, secondary bile acids) can impair anti-tumor responses, which aggravates HCC. Lastly, we describe the rising 'new era' of immunotherapies (e.g., immune checkpoint inhibitors, adoptive T-cell transfer) and discuss for the potential incorporation of gut microbiota targeted therapeutics (e.g., probiotics, fecal microbiota transplantation) to alleviate HCC. Altogether, this chapter invigorates for continuous research to decipher the role of gut microbiome in HCC from its influence on immunometabolism and metabolic reprogramming.
Collapse
|
238
|
Immune-related microRNA signature for predicting prognosis and the immune microenvironment in hepatocellular carcinoma. Life Sci 2020; 265:118799. [PMID: 33220285 DOI: 10.1016/j.lfs.2020.118799] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 11/06/2020] [Accepted: 11/15/2020] [Indexed: 02/08/2023]
Abstract
AIMS As vital regulators of post-transcription gene expression, microRNAs are involved in the initiation and progression of hepatocellular carcinoma (HCC), including antitumor immune responses. We aimed to identify an immune-related microRNA signature and explore the influence of this signature on the prognosis and immunity of HCC. MATERIALS AND METHODS Differentially expressed immune-related microRNAs were identified between high- and low-immunity groups in the TCGA-HCC dataset. Then, Cox regression models were used to construct an immune-related microRNA signature. We assessed the prognostic value and clinical relevance of this signature. Furthermore, we analyzed the effect of the immune-related microRNA signature on immune cells and immune checkpoints. KEY FINDINGS We screened 41 differentially expressed immune-related microRNAs, of which 7 microRNAs were used to construct the immune signature. Survival analysis showed that high-risk patients had a shorter survival. The immune-related microRNA signature was an independent prognostic marker and was associated with the clinical stage. To understand the functional mechanism of the seven-microRNA signature, we predicted target genes of microRNAs. Enrichment analysis indicated that these target genes participated in immune responses. Moreover, we found that the microRNA signature was related to tumor-infiltrating immune cells. In high-risk patients, there was a higher expression of inhibitory immune checkpoints. On the contrary, the expression of stimulatory immune checkpoints was lower in high-risk patients. SIGNIFICANCE We successfully identified an immune-related microRNA signature that was strongly correlated with the prognosis and immune microenvironment of HCC. The immune-related microRNAs signature might have important implications for improving the clinical outcomes of HCC patients.
Collapse
|
239
|
Xu Z, Zeng S, Gong Z, Yan Y. Exosome-based immunotherapy: a promising approach for cancer treatment. Mol Cancer 2020; 19:160. [PMID: 33183286 PMCID: PMC7661275 DOI: 10.1186/s12943-020-01278-3] [Citation(s) in RCA: 328] [Impact Index Per Article: 65.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 11/03/2020] [Indexed: 02/08/2023] Open
Abstract
In the era of the rapid development of cancer immunotherapy, there is a high level of interest in the application of cell-released small vesicles that stimulate the immune system. As cell-derived nanovesicles, exosomes show great promise in cancer immunotherapy because of their immunogenicity and molecular transfer function. The cargoes carried on exosomes have been recently identified with improved technological advances and play functional roles in the regulation of immune responses. In particular, exosomes derived from tumor cells and immune cells exhibit unique composition profiles that are directly involved in anticancer immunotherapy. More importantly, exosomes can deliver their cargoes to targeted cells and thus influence the phenotype and immune-regulation functions of targeted cells. Accumulating evidence over the last decade has further revealed that exosomes can participate in multiple cellular processes contributing to cancer development and therapeutic effects, showing the dual characteristics of promoting and suppressing cancer. The potential of exosomes in the field of cancer immunotherapy is huge, and exosomes may become the most effective cancer vaccines, as well as targeted antigen/drug carriers. Understanding how exosomes can be utilized in immune therapy is important for controlling cancer progression; additionally, exosomes have implications for diagnostics and the development of novel therapeutic strategies. This review discusses the role of exosomes in immunotherapy as carriers to stimulate an anti-cancer immune response and as predictive markers for immune activation; furthermore, it summarizes the mechanism and clinical application prospects of exosome-based immunotherapy in human cancer.
Collapse
Affiliation(s)
- Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Shuangshuang Zeng
- Department of Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
| |
Collapse
|
240
|
Zhu L, Sun HT, Wang S, Huang SL, Zheng Y, Wang CQ, Hu BY, Qin W, Zou TT, Fu Y, Shen XT, Zhu WW, Geng Y, Lu L, Jia HL, Qin LX, Dong QZ. Isolation and characterization of exosomes for cancer research. J Hematol Oncol 2020; 13:152. [PMID: 33168028 PMCID: PMC7652679 DOI: 10.1186/s13045-020-00987-y] [Citation(s) in RCA: 310] [Impact Index Per Article: 62.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023] Open
Abstract
Exosomes are a subset of extracellular vesicles that carry specific combinations of proteins, nucleic acids, metabolites, and lipids. Mounting evidence suggests that exosomes participate in intercellular communication and act as important molecular vehicles in the regulation of numerous physiological and pathological processes, including cancer development. Exosomes are released by various cell types under both normal and pathological conditions, and they can be found in multiple bodily fluids. Moreover, exosomes carrying a wide variety of important macromolecules provide a window into altered cellular or tissue states. Their presence in biological fluids renders them an attractive, minimally invasive approach for liquid biopsies with potential biomarkers for cancer diagnosis, prediction, and surveillance. Due to their biocompatibility and low immunogenicity and cytotoxicity, exosomes have potential clinical applications in the development of innovative therapeutic approaches. Here, we summarize recent advances in various technologies for exosome isolation for cancer research. We outline the functions of exosomes in regulating tumor metastasis, drug resistance, and immune modulation in the context of cancer development. Finally, we discuss prospects and challenges for the clinical development of exosome-based liquid biopsies and therapeutics.
Collapse
Affiliation(s)
- Le Zhu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Hao-Ting Sun
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Shun Wang
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Sheng-Lin Huang
- Institutes of Biomedical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, China
- Fudan University Shanghai Cancer Center, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yan Zheng
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Chao-Qun Wang
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Bei-Yuan Hu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Wei Qin
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Tian-Tian Zou
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Yan Fu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Xiao-Tian Shen
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Wen-Wei Zhu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Yan Geng
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Lu Lu
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Hu-Liang Jia
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China
| | - Lun-Xiu Qin
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China.
- Institutes of Biomedical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, China.
| | - Qiong-Zhu Dong
- Department of General Surgery, Huashan Hospital, Cancer Metastasis Institute, Fudan University, 12 Urumqi Road (M), Shanghai, 200040, China.
- Institutes of Biomedical Sciences, Fudan University, 131 Dong An Road, Shanghai, 200032, China.
| |
Collapse
|
241
|
Xue VW, Chung JYF, Córdoba CAG, Cheung AHK, Kang W, Lam EWF, Leung KT, To KF, Lan HY, Tang PMK. Transforming Growth Factor-β: A Multifunctional Regulator of Cancer Immunity. Cancers (Basel) 2020. [PMID: 33114183 DOI: 10.3390/cancers12113099.pmid:33114183;pmcid:pmc7690808] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2023] Open
Abstract
Transforming growth factor-β (TGF-β) was originally identified as an anti-tumour cytokine. However, there is increasing evidence that it has important roles in the tumour microenvironment (TME) in facilitating cancer progression. TGF-β actively shapes the TME via modulating the host immunity. These actions are highly cell-type specific and complicated, involving both canonical and non-canonical pathways. In this review, we systemically update how TGF-β signalling acts as a checkpoint regulator for cancer immunomodulation. A better appreciation of the underlying pathogenic mechanisms at the molecular level can lead to the discovery of novel and more effective therapeutic strategies for cancer.
Collapse
Affiliation(s)
- Vivian Weiwen Xue
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Jeff Yat-Fai Chung
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Cristina Alexandra García Córdoba
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Alvin Ho-Kwan Cheung
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Eric W-F Lam
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London W12 0NN, UK
| | - Kam-Tong Leung
- Department of Paediatrics, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Patrick Ming-Kuen Tang
- Department of Anatomical and Cellular Pathology, State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
242
|
Transforming Growth Factor-β: A Multifunctional Regulator of Cancer Immunity. Cancers (Basel) 2020; 12:cancers12113099. [PMID: 33114183 PMCID: PMC7690808 DOI: 10.3390/cancers12113099] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/12/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Transforming growth factor beta (TGF-β) is a multifunctional cytokine that can restrict cancer onset but also promote cancer progression at late stages of cancer. The ability of TGF-β in producing diverse and sometimes opposing effects relies on its potential to control different cellular signalling and gene expression in distinct cell types, and environmental settings. The tumour promoting role of TGF-β is primarily mediated through its effects on the local tumour microenvironment (TME) of the cancer cells. In this review, we discuss the most recent research on the role and regulation of TGF-β, with a specific focus on its functions on promoting cancer progression through targeting different immune cells in the TME as well as its therapeutic perspectives. Abstract Transforming growth factor-β (TGF-β) was originally identified as an anti-tumour cytokine. However, there is increasing evidence that it has important roles in the tumour microenvironment (TME) in facilitating cancer progression. TGF-β actively shapes the TME via modulating the host immunity. These actions are highly cell-type specific and complicated, involving both canonical and non-canonical pathways. In this review, we systemically update how TGF-β signalling acts as a checkpoint regulator for cancer immunomodulation. A better appreciation of the underlying pathogenic mechanisms at the molecular level can lead to the discovery of novel and more effective therapeutic strategies for cancer.
Collapse
|
243
|
Kole C, Charalampakis N, Tsakatikas S, Vailas M, Moris D, Gkotsis E, Kykalos S, Karamouzis MV, Schizas D. Immunotherapy for Hepatocellular Carcinoma: A 2021 Update. Cancers (Basel) 2020; 12:2859. [PMID: 33020428 PMCID: PMC7600093 DOI: 10.3390/cancers12102859] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/26/2020] [Accepted: 09/30/2020] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of one of the most frequent liver cancers and the fourth leading cause of cancer-related mortality worldwide. Current treatment options such as surgery, neoadjuvant chemoradiotherapy, liver transplantation, and radiofrequency ablation will benefit only a very small percentage of patients. Immunotherapy is a novel treatment approach representing an effective and promising option against several types of cancer. The aim of our study is to present the currently ongoing clinical trials and to evaluate the efficacy of immunotherapy in HCC. In this paper, we demonstrate that combination of different immunotherapies or immunotherapy with other modalities results in better overall survival (OS) and progression-free survival (PFS) compared to single immunotherapy agent. Another objective of this paper is to demonstrate and highlight the importance of tumor microenvironment as a predictive and prognostic marker and its clinical implications in immunotherapy response.
Collapse
Affiliation(s)
- Christo Kole
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 115 27 Athens, Greece; (C.K.); (M.V.); (E.G.); (D.S.)
| | - Nikolaos Charalampakis
- Department of Medical Oncology, Metaxa Cancer Hospital, 185 37 Athens, Greece; (N.C.); (S.T.)
| | - Sergios Tsakatikas
- Department of Medical Oncology, Metaxa Cancer Hospital, 185 37 Athens, Greece; (N.C.); (S.T.)
| | - Michail Vailas
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 115 27 Athens, Greece; (C.K.); (M.V.); (E.G.); (D.S.)
| | - Dimitrios Moris
- Department of Surgery, Duke University School of Medicine, Durham, NC 27707, USA;
| | - Efthymios Gkotsis
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 115 27 Athens, Greece; (C.K.); (M.V.); (E.G.); (D.S.)
| | - Stylianos Kykalos
- Second Propedeutic Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 115 27 Athens, Greece;
| | - Michalis V. Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Dimitrios Schizas
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, 115 27 Athens, Greece; (C.K.); (M.V.); (E.G.); (D.S.)
| |
Collapse
|
244
|
Dehbarez FM, Nezafat N, Mahmoodi S. In Silico Design of a Novel Multi-Epitope Peptide Vaccine Against Hepatocellular Carcinoma. LETT DRUG DES DISCOV 2020. [DOI: 10.2174/1570180817999200502030038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:
Hepatocellular Carcinoma (HCC) is a prevalent cancer in the world. As
yet, there is no medication for complete treatment of HCC.
Objective:
There is a critical need to search for an innovative therapy for HCC. Recently, multiepitope
vaccines have been introduced as effective immunotherapy approach against HCC.
Methods:
In this research, several immunoinformatics methods were applied to create an original
multi-epitope vaccine against HCC consisting of CD8+ cytolytic T lymphocytes (CTLs) epitopes
selected from α- fetoprotein (AFP), glypican-3 (GPC3), aspartyl-β-hydroxylase (ASPH); CD4+
helper T lymphocytes (HTLs) epitopes from tetanus toxin fragment C (TTFC), and finally, two tandem
repeats of HSP70407-426 were used which stimulated strong innate and adaptive immune responses.
All the mentioned parts were connected together by relevant linkers.
Results:
According to physicochemical, structural, and immunological results, the designed
vaccine is stable, non-allergen, antigen; it also has a high-quality 3D structure, and numerous linear
and conformational B cell epitopes, whereby this vaccine may stimulate efficient humoral immunity.
Conclusion:
Center on the collected results, the designed vaccine potentially can induce cellular and
humoral immune responses in HCC cases; nonetheless, the efficiency of vaccine must be approved
within in vitro and in vivo immunological analyzes.
Collapse
Affiliation(s)
- Fatemeh Motamedi Dehbarez
- Department of Medical Biotechnology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Navid Nezafat
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Shirin Mahmoodi
- Department of Medical Biotechnology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| |
Collapse
|
245
|
Li M, Li S, Du C, Zhang Y, Li Y, Chu L, Han X, Galons H, Zhang Y, Sun H, Yu P. Exosomes from different cells: Characteristics, modifications, and therapeutic applications. Eur J Med Chem 2020; 207:112784. [PMID: 33007722 DOI: 10.1016/j.ejmech.2020.112784] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/19/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023]
Abstract
Exosomes are cystic vesicles secreted by living cells with a phospholipid bilayer membrane. Importantly, these vesicles could serve to carry lipids, proteins, genetic materials, and transmit biological information in vivo. The cell-specific proteins and genetic materials in exosomes are capable of reflecting their cell origin and physiological status. Based on the different tissues and cells (macrophage, dendritic cells, tumor cells, mesenchymal stem cells, various body fluids, and so on), exosomes exhibit different characteristics and functions. Furthermore, owing to their high delivery efficiency, biocompatibility, and multifunctional properties, exosomes are expected to become a new means of drug delivery, disease diagnosis, immunotherapy, and precise treatment. At the same time, in order to supplement or enhance the therapeutic applicability of exosomes, chemical or biological modifications can be used to broaden, change or improve their therapeutic capabilities. This review focuses on three aspects: the characteristics and original functions of exosomes secreted by different cells, the modification and transformation of exosomes, and the application of exosomes in different diseases.
Collapse
Affiliation(s)
- Mingyuan Li
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, 300457, China
| | - Shuangshuang Li
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, 300457, China
| | - Chunyang Du
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, 300457, China
| | - Yinan Zhang
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, 300457, China
| | - Yuan Li
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, 300457, China
| | - Liqiang Chu
- College of Chemical Engineering and Materials Science, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Xiao Han
- College of Chemical Engineering and Materials Science, Tianjin University of Science & Technology, Tianjin, 300457, China
| | - Hervé Galons
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, 300457, China
| | - Yongmin Zhang
- Institut Parisien de Chimie Moléculaire, UMR CNRS 8232, 4 Place Jussieu, 75005, Paris, France
| | - Hua Sun
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, 300457, China.
| | - Peng Yu
- College of Biotechnology, China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, Tianjin University of Science & Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, 300457, China.
| |
Collapse
|
246
|
Cabeza L, Perazzoli G, Peña M, Cepero A, Luque C, Melguizo C, Prados J. Cancer therapy based on extracellular vesicles as drug delivery vehicles. J Control Release 2020; 327:296-315. [PMID: 32814093 DOI: 10.1016/j.jconrel.2020.08.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/11/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) are lipid bilayer vesicles of nanometric size secreted by cells to communicate with other cells, either nearby or remotely. Their physicochemical properties make them a promising nanomedicine for drug transport and release in cancer therapy. In this review, we present the different types and biogenesis of EVs and highlight the importance of adequately selecting the cell of origin in cancer therapy. Furthermore, the main methodologies followed for the isolation of EVs and drug loading, as well as the modification and functionalization of these vesicles to generate EV-based nanocarriers are discussed. Finally, we review some of the main studies using drug-loaded exosomes in tumor therapy both in in vitro and in vivo models (even in resistant tumors). These investigations show promising results, achieving significant improvement in the antitumor effect of drugs in most cases. However, the number of clinical trials and patents based on these nanoformulations is still low, thus further research is still warranted in this area.
Collapse
Affiliation(s)
- Laura Cabeza
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| | - Gloria Perazzoli
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Mercedes Peña
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - Ana Cepero
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - Cristina Luque
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - Consolacion Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain.
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| |
Collapse
|
247
|
Wang Y, Xiang Y, Xin VW, Wang XW, Peng XC, Liu XQ, Wang D, Li N, Cheng JT, Lyv YN, Cui SZ, Ma Z, Zhang Q, Xin HW. Dendritic cell biology and its role in tumor immunotherapy. J Hematol Oncol 2020; 13:107. [PMID: 32746880 PMCID: PMC7397618 DOI: 10.1186/s13045-020-00939-6] [Citation(s) in RCA: 285] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 07/20/2020] [Indexed: 12/11/2022] Open
Abstract
As crucial antigen presenting cells, dendritic cells (DCs) play a vital role in tumor immunotherapy. Taking into account the many recent advances in DC biology, we discuss how DCs (1) recognize pathogenic antigens with pattern recognition receptors through specific phagocytosis and through non-specific micropinocytosis, (2) process antigens into small peptides with proper sizes and sequences, and (3) present MHC-peptides to CD4+ and CD8+ T cells to initiate immune responses against invading microbes and aberrant host cells. During anti-tumor immune responses, DC-derived exosomes were discovered to participate in antigen presentation. T cell microvillar dynamics and TCR conformational changes were demonstrated upon DC antigen presentation. Caspase-11-driven hyperactive DCs were recently reported to convert effectors into memory T cells. DCs were also reported to crosstalk with NK cells. Additionally, DCs are the most important sentinel cells for immune surveillance in the tumor microenvironment. Alongside DC biology, we review the latest developments for DC-based tumor immunotherapy in preclinical studies and clinical trials. Personalized DC vaccine-induced T cell immunity, which targets tumor-specific antigens, has been demonstrated to be a promising form of tumor immunotherapy in patients with melanoma. Importantly, allogeneic-IgG-loaded and HLA-restricted neoantigen DC vaccines were discovered to have robust anti-tumor effects in mice. Our comprehensive review of DC biology and its role in tumor immunotherapy aids in the understanding of DCs as the mentors of T cells and as novel tumor immunotherapy cells with immense potential.
Collapse
Affiliation(s)
- Yingying Wang
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095 China
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023 Hubei China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023 Hubei China
- Department of Gynaecology, Comprehensive Cancer Center, Hannover Medical School, 30625 Hannover, Germany
| | - Ying Xiang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023 Hubei China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023 Hubei China
| | | | - Xian-Wang Wang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023 Hubei China
- Department of Laboratory Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023 Hubei China
| | - Xiao-Chun Peng
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023 Hubei China
- Department of Pathophysiology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023 Hubei China
| | - Xiao-Qin Liu
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023 Hubei China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023 Hubei China
- Department of Medical Imaging, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023 Hubei China
| | - Dong Wang
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023 Hubei China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023 Hubei China
| | - Na Li
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei China
| | - Jun-Ting Cheng
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023 Hubei China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023 Hubei China
| | - Yan-Ning Lyv
- Institute for Infectious Diseases and Endemic Diseases Prevention and Control, Beijing Center for Diseases Prevention and Control, Beijing, 100013 China
| | - Shu-Zhong Cui
- State Key Laboratory of Respiratory Disease, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095 China
| | - Zhaowu Ma
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023 Hubei China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023 Hubei China
| | - Qing Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, 510275 China
- Institute of Sun Yat-sen University in Shenzhen, Shenzhen, China
| | - Hong-Wu Xin
- Laboratory of Oncology, Center for Molecular Medicine, School of Basic Medicine, Faculty of Medicine, Yangtze University, 1 Nanhuan Road, Jingzhou, 434023 Hubei China
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Faculty of Medicine, Yangtze University, Jingzhou, 434023 Hubei China
- People’s Hospital of Lianjiang, Lianjiang, 524400 Guangdong China
| |
Collapse
|
248
|
Shi X, Sun J, Li H, Lin H, Xie W, Li J, Tan W. Antitumor efficacy of interferon-γ-modified exosomal vaccine in prostate cancer. Prostate 2020; 80:811-823. [PMID: 32427375 DOI: 10.1002/pros.23996] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/26/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Exosomes secreted by tumor cells can be regarded as carriers of tumor-associated antigens and have potential value in tumor immunotherapy. The aim of this study was to evaluate the antitumor efficacy of a novel exosomal vaccine (interferon-γ [IFN-γ]-modified exosomal vaccine) in prostate cancer. METHODS Prostate cancer cell-derived exosomes were used to prepare the exosomal vaccine using our protein-anchoring technique. The immunogenicity and therapeutic efficacy of the exosomes was evaluated by measuring the effects of the exosomal vaccine on M1 macrophage differentiation, the ability of macrophages to engulf the exosomes, the production of antibodies against exosomes, and tumor angiogenesis and metastasis, and tumor growth. RESULTS The IFN-γ fusion protein was efficiently anchored on the surface of prostate cancer cell-derived exosomes and retained its bioactivity. The IFN-γ-exosomal vaccine increased the number of M1 macrophages, enhanced the ability of M1 macrophages to engulf RM-1 cell-derived exosomes, and induced the production of specific antibodies against exosomes. The exosomal vaccine downregulated the expression of vascular endothelial growth factor receptor 2 and attenuated the effect of exosomes in promoting tumor metastasis. The proportions of CD4+ , CD8+ , and IFN-γ+ CD8+ T cells in the exosomal vaccine group were the highest among the four groups. Interestingly, the IFN-γ-exosomal vaccine decreased the percentage of Tregs and downregulated the expression of programed death-ligand 1 and indoleamine 2, 3-dioxygenase 1 in the tumor environment. The exosomal vaccine significantly inhibited tumor growth and prolonged the survival time of mice with prostate cancer. The exosomal and tumor cell vaccines had a good synergistic effect in promoting tumor immunity. CONCLUSIONS The novel exosomal vaccine induced an immune response that cleared prostate cancer cell-derived exosomes, thereby eliminating the regulatory effect of the exosomes. This study may provide experimental evidence for the use of exosomes as a therapeutic tool or target in immunotherapy for human prostate cancer.
Collapse
Affiliation(s)
- Xiaojun Shi
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jie Sun
- Department of Health Management, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Haoran Li
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hao Lin
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weiwei Xie
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinlong Li
- Department of Institute of Biotherapy, Institute of Biotherapy, School of Biotechnology, Southern Medical University, Guangzhou, China
| | - Wanlong Tan
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
249
|
Wan Z, Dong Y, Wei M, Gao X, Yang G, Zhang J, Liu L. Exosomes in Tumor Immunotherapy: Mediator, Drug Carrier, and Prognostic Biomarker. ACTA ACUST UNITED AC 2020; 4:e2000061. [PMID: 32700829 DOI: 10.1002/adbi.202000061] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/31/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Zhuo Wan
- Department of Hematology Tangdu Hospital Fourth Military Medical University Xi'an 710038 P. R. China
| | - Yan Dong
- Department of Hematology Tangdu Hospital Fourth Military Medical University Xi'an 710038 P. R. China
| | - Mengying Wei
- State Key Laboratory of Cancer Biology Department of Biochemistry and Molecular Biology Fourth Military Medical University Xi'an 710032 P. R. China
| | - Xiaotong Gao
- Department of Hematology Tangdu Hospital Fourth Military Medical University Xi'an 710038 P. R. China
| | - Guodong Yang
- State Key Laboratory of Cancer Biology Department of Biochemistry and Molecular Biology Fourth Military Medical University Xi'an 710032 P. R. China
| | - Jian Zhang
- State Key Laboratory of Cancer Biology Department of Biochemistry and Molecular Biology Fourth Military Medical University Xi'an 710032 P. R. China
| | - Li Liu
- Department of Hematology Tangdu Hospital Fourth Military Medical University Xi'an 710038 P. R. China
| |
Collapse
|
250
|
Li C, Donninger H, Eaton J, Yaddanapudi K. Regulatory Role of Immune Cell-Derived Extracellular Vesicles in Cancer: The Message Is in the Envelope. Front Immunol 2020; 11:1525. [PMID: 32765528 PMCID: PMC7378739 DOI: 10.3389/fimmu.2020.01525] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 06/09/2020] [Indexed: 12/28/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogenous group of membrane-surrounded structures. Besides serving as a harbor for the unwanted material exocytosed by cells, EVs play a critical role in conveying intact protein, genetic, and lipid contents that are important for intercellular communication. EVs, broadly comprised of microvesicles and exosomes, are released to the extracellular environment from nearly all cells either via shedding from the plasma membrane or by originating from the endosomal system. Exosomes are 40–150 nm, endosome-derived small EVs (sEVs) that are released by cells into the extracellular environment. This review focuses on the biological properties of immune cell-derived sEVs, including composition and cellular targeting and mechanisms by which these immune cell-derived sEVs influence tumor immunity either by suppressing or promoting tumor growth, are discussed. The final section of this review discusses how the biological properties of immune cell-derived sEVs can be manipulated to improve their immunogenicity.
Collapse
Affiliation(s)
- Chi Li
- Experimental Therapeutics Group, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States.,Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Howard Donninger
- Experimental Therapeutics Group, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States.,Department of Medicine, University of Louisville, Louisville, KY, United States
| | - John Eaton
- Department of Medicine, University of Louisville, Louisville, KY, United States.,Immuno-Oncology Group, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States
| | - Kavitha Yaddanapudi
- Immuno-Oncology Group, James Graham Brown Cancer Center, University of Louisville, Louisville, KY, United States.,Division of Immunotherapy, Department of Surgery, University of Louisville, Louisville, KY, United States.,Department of Microbiology and Immunology, University of Louisville, Louisville, KY, United States
| |
Collapse
|