1
|
Sonar S, Das A, Kalele K, Subramaniyan V. Exosome-based cancer vaccine: a cell-free approach. Mol Biol Rep 2025; 52:421. [DOI: 10.1007/s11033-025-10519-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/15/2025] [Indexed: 05/04/2025]
|
2
|
Hopo MG, Mabrok M, Abu-Elala N, Yu Y. Navigating Fish Immunity: Focus on Mucosal Immunity and the Evolving Landscape of Mucosal Vaccines. BIOLOGY 2024; 13:980. [PMID: 39765647 PMCID: PMC11727089 DOI: 10.3390/biology13120980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 01/15/2025]
Abstract
The growing role of aquaculture in global food security has underscored the need for advanced immunological insights to protect fish health and boost productivity. As aquaculture's importance rises, understanding fish immunity is crucial for developing effective vaccination strategies. Fish possess a specialized immune system with unique mucosal structures that enable resilience in aquatic environments. This review examines critical advances in fish mucosal immunity, particularly focusing on mucosal vaccines that target infection at primary entry points, such as the gills, skin, and gastrointestinal tract. Mucosal vaccination has demonstrated a compelling capacity to stimulate localized and systemic immune responses, offering enhanced protection against waterborne pathogens. Additionally, this review addresses knowledge gaps from previous research on the global aquaculture vaccines market by offering a regional perspective on industry developments, recent trends, and innovative vaccine formulations. In doing so, it highlights the role of mucosal vaccines in overcoming the specific challenges of fish farming and supporting sustainable aquaculture. This synthesis of current methodologies, industry practices, and future directions contributes to a deeper understanding of fish immunology, ultimately informing strategies to achieve optimal disease management and bolster global aquaculture resilience.
Collapse
Affiliation(s)
- Mai G. Hopo
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China;
| | - Mahmoud Mabrok
- Department of Fish Diseases and Management, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt;
- Department of Microbiology and Parasitology, Faculty of Veterinary Medicine, King Salman International University, Ras Sudr 46612, Egypt
| | - Nermeen Abu-Elala
- Department of Aquatic Animal Medicine and Management, Faculty of Veterinary Medicine, Cairo University, Cairo 12211, Egypt
- Department of Animal Medicine, Faculty of Veterinary Medicine, King Salman International University, Ras Sudr 46612, Egypt
| | - Yongyao Yu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China;
- Hubei Hongshan Laboratory, Wuhan 430070, China
| |
Collapse
|
3
|
Song Y, Teng L, Chen Y, Dong CM. Glycopolypeptide Coordinated Nanovaccine: Fabrication, Characterization, and Antitumor Immune Response. CHEM & BIO ENGINEERING 2024; 1:633-646. [PMID: 39974696 PMCID: PMC11835264 DOI: 10.1021/cbe.4c00083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/07/2024] [Accepted: 06/08/2024] [Indexed: 02/21/2025]
Abstract
Cancer nanovaccine is a frontier immunotherapy strategy, in which the delivery carrier can protect antigen and adjuvant from degradation, increase blood circulation half-life, and improve antigen permeability and presentation, thus enhancing the security and potency of nanovaccine. To address the barriers of antigen delivery, we design and fabricate a kind of intracellular pH-sensitive glycopolypeptide coordinated nanovaccine (OVA-HPGM-Mn) with ∼30% loading capacity of ovalbumin (OVA). The nanovaccine OVA-HPGM-Mn could specifically deliver antigen to dendritic cells (DCs) and effectively escape from endolysosomes to cytoplasm after 6 h of incubation, while the blank counterpart HPGM-Mn acted as an adjuvant to promote DCs maturation and increase the percentage of maturated cells to 26.5% from 11.8% in vitro. Furthermore, the mannosylated polypeptide nanovaccine prolonged the retention time of OVA for 72 h to facilitate 29.5% DCs maturation in lymph nodes, activated 48.8% CD8+T cells in spleen, increased the CD8+/CD4+T cell ratio twice to 1.06, and upregulated the levels of pro-inflammatory cytokines including TNF-α, IFN-γ, and IL-6, thus inhibiting the tumor growth of ∼80%. Consequently, this work provides a versatile strategy for the fabrication of glycosylated polypeptide coordinated nanomaterials for antigen delivery and cancer immunotherapy.
Collapse
Affiliation(s)
- Yingying Song
- School of Chemistry and Chemical
Engineering, Frontiers Science Center for Transformative Molecules,
Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Lin Teng
- School of Chemistry and Chemical
Engineering, Frontiers Science Center for Transformative Molecules,
Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Yanzheng Chen
- School of Chemistry and Chemical
Engineering, Frontiers Science Center for Transformative Molecules,
Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Chang-Ming Dong
- School of Chemistry and Chemical
Engineering, Frontiers Science Center for Transformative Molecules,
Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| |
Collapse
|
4
|
Al-Hawary SIS, Jasim SA, Hjazi A, Oghenemaro EF, Kaur I, Kumar A, Al-Ani AM, Alwaily ER, Redhee AH, Mustafa YF. Nucleic acid-based vaccine for ovarian cancer cells; bench to bedside. Cell Biochem Funct 2024; 42:e3978. [PMID: 38515237 DOI: 10.1002/cbf.3978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 03/23/2024]
Abstract
Ovarian cancer continues to be a difficult medical issue that affects millions of individuals worldwide. Important platforms for cancer immunotherapy include checkpoint inhibitors, chimeric antigen receptor T cells, bispecific antibodies, cancer vaccines, and other cell-based treatments. To avoid numerous infectious illnesses, conventional vaccinations based on synthetic peptides, recombinant subunit vaccines, and live attenuated and inactivated pathogens are frequently utilized. Vaccine manufacturing processes, however, are not entirely safe and carry a significant danger of contaminating living microorganisms. As a result, the creation of substitute vaccinations is required for both viral and noninfectious illnesses, including cancer. Recently, there has been testing of nucleic acid vaccines, or NAVs, as a cancer therapeutic. Tumor antigens (TAs) are genetically encoded by DNA and mRNA vaccines, which the host uses to trigger immune responses against ovarian cancer cells that exhibit the TAs. Despite being straightforward, safe, and easy to produce, NAVs are not currently thought to be an ideal replacement for peptide vaccines. Some obstacles to this strategy include selecting the appropriate therapeutic agents (TAs), inadequate immunogenicity, and the immunosuppressive characteristic of ovarian cancer. We focus on strategies that have been employed to increase NAVs' effectiveness in the fight against ovarian cancer in this review.
Collapse
Affiliation(s)
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, Al-maarif University College, Anbar, Iraq
- Biotechnology Department, College of Applied Science, Fallujah University, Fallujah, Iraq
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Enwa Felix Oghenemaro
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Delta State University, Abraka, Nigeria
| | - Irwanjot Kaur
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka, India
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
| | - Abhinav Kumar
- Department of Nuclear and Renewable Energy, Ural Federal University Named after The First President of Russia, Yekaterinburg, Russia
| | | | - Enas R Alwaily
- Microbiology Research Group, College of Pharmacy, Al-Ayen University, Thi-Qar, Iraq
| | - Ahmed Huseen Redhee
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| |
Collapse
|
5
|
Sobhani-Nasab A, Banafshe HR, Atapour A, Khaksary Mahabady M, Akbari M, Daraei A, Mansoori Y, Moradi Hasan-Abad A. The use of nanoparticles in the treatment of infectious diseases and cancer, dental applications and tissue regeneration: a review. FRONTIERS IN MEDICAL TECHNOLOGY 2024; 5:1330007. [PMID: 38323112 PMCID: PMC10844477 DOI: 10.3389/fmedt.2023.1330007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 12/12/2023] [Indexed: 02/08/2024] Open
Abstract
The emergence of nanotechnology as a field of study can be traced back to the 1980s, at which point the means to artificially produce, control, and observe matter on a nanometer level was made viable. Recent advancements in technology have enabled us to extend our reach to the nanoscale, which has presented an unparalleled opportunity to directly target biomolecular interactions. As a result of these developments, there is a drive to arise intelligent nanostructures capable of overcoming the obstacles that have impeded the progress of conventional pharmacological methodologies. After four decades, the gradual amalgamation of bio- and nanotechnologies is initiating a revolution in the realm of disease detection, treatment, and monitoring, as well as unsolved medical predicaments. Although a significant portion of research in the field is still confined to laboratories, the initial application of nanotechnology as treatments, vaccines, pharmaceuticals, and diagnostic equipment has now obtained endorsement for commercialization and clinical practice. The current issue presents an overview of the latest progress in nanomedical strategies towards alleviating antibiotic resistance, diagnosing and treating cancer, addressing neurodegenerative disorders, and an array of applications, encompassing dentistry and tuberculosis treatment. The current investigation also scrutinizes the deployment of sophisticated smart nanostructured materials in fields of application such as regenerative medicine, as well as the management of targeted and sustained release of pharmaceuticals and therapeutic interventions. The aforementioned concept exhibits the potential for revolutionary advancements within the field of immunotherapy, as it introduces the utilization of implanted vaccine technology to consistently regulate and augment immune functions. Concurrently with the endeavor to attain the advantages of nanomedical intervention, it is essential to enhance the unceasing emphasis on nanotoxicological research and the regulation of nanomedications' safety. This initiative is crucial in achieving the advancement in medicine that currently lies within our reach.
Collapse
Affiliation(s)
- Ali Sobhani-Nasab
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamid Reza Banafshe
- Physiology Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Amir Atapour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahmood Khaksary Mahabady
- Anatomical Sciences Research Center, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Akbari
- Department of Surgery, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Abdolreza Daraei
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Yaser Mansoori
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Amin Moradi Hasan-Abad
- Autoimmune Diseases Research Center, Shahid Beheshti Hospital, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
6
|
Hashemi P, Mahmoodi S, Ghasemian A. An updated review on oral protein-based antigen vaccines efficiency and delivery approaches: a special attention to infectious diseases. Arch Microbiol 2023; 205:289. [PMID: 37468763 DOI: 10.1007/s00203-023-03629-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/04/2023] [Accepted: 07/09/2023] [Indexed: 07/21/2023]
Abstract
Various infectious agents affect human health via the oral entrance. The majority of pathogens lack approved vaccines. Oral vaccination is a convenient, safe and cost-effective approach with the potential of provoking mucosal and systemic immunity and maintaining individual satisfaction. However, vaccines should overcome the intricate environment of the gastrointestinal tract (GIT). Oral protein-based antigen vaccines (OPAVs) are easier to administer than injectable vaccines and do not require trained healthcare professionals. Additionally, the risk of needle-related injuries, pain, and discomfort is eliminated. However, OPAVs stability at environmental and GIT conditions should be considered to enhance their stability and facilitate their transport and storage. These vaccines elicit the local immunity, protecting GIT, genital tract and respiratory epithelial surfaces, where numerous pathogens penetrate the body. OPAVs can also be manipulated (such as using specific incorporated ligand and receptors) to elicit targeted immune response. However, low bioavailability of OPAVs necessitates development of proper protein carriers and formulations to enhance their stability and efficacy. There are several strategies to improve their efficacy or protective effects, such as incorporation of adjuvants, enzyme inhibitors, mucoadhesive or penetrating devices and permeation enhancers. Hence, efficient delivery of OPAVs into GIT require proper delivery systems mainly including smart target systems, probiotics, muco-adhesive carriers, lipid- and plant-based delivery systems and nano- and microparticles.
Collapse
Affiliation(s)
- Parisa Hashemi
- Department of Medical Biotechnology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Shirin Mahmoodi
- Department of Medical Biotechnology, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran.
| | - Abdolmajid Ghasemian
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran.
| |
Collapse
|
7
|
Rana I, Oh J, Baig J, Moon JH, Son S, Nam J. Nanocarriers for cancer nano-immunotherapy. Drug Deliv Transl Res 2023; 13:1936-1954. [PMID: 36190661 PMCID: PMC9528883 DOI: 10.1007/s13346-022-01241-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2022] [Indexed: 11/03/2022]
Abstract
The host immune system possesses an intrinsic ability to target and kill cancer cells in a specific and adaptable manner that can be further enhanced by cancer immunotherapy, which aims to train the immune system to boost the antitumor immune response. Several different categories of cancer immunotherapy have emerged as new standard cancer therapies in the clinic, including cancer vaccines, immune checkpoint inhibitors, adoptive T cell therapy, and oncolytic virus therapy. Despite the remarkable survival benefit for a subset of patients, the low response rate and immunotoxicity remain the major challenges for current cancer immunotherapy. Over the last few decades, nanomedicine has been intensively investigated with great enthusiasm, leading to marked advancements in nanoparticle platforms and nanoengineering technology. Advances in nanomedicine and immunotherapy have also led to the emergence of a nascent research field of nano-immunotherapy, which aims to realize the full therapeutic potential of immunotherapy with the aid of nanomedicine. In particular, nanocarriers present an exciting opportunity in immuno-oncology to boost the activity, increase specificity, decrease toxicity, and sustain the antitumor efficacy of immunological agents by potentiating immunostimulatory activity and favorably modulating pharmacological properties. This review discusses the potential of nanocarriers for cancer immunotherapy and introduces preclinical studies designed to improve clinical cancer immunotherapy modalities using nanocarrier-based engineering approaches. It also discusses the potential of nanocarriers to address the challenges currently faced by immuno-oncology as well as the challenges for their translation to clinical applications.
Collapse
Affiliation(s)
- Isra Rana
- College of Pharmacy, Chonnam National University, Gwangju, 61186, South Korea
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Jaeeun Oh
- Department of Biological Sciences, Inha University, Incheon, 22212, South Korea
| | - Juwon Baig
- Department of Biological Sciences, Inha University, Incheon, 22212, South Korea
| | - Jeong Hyun Moon
- Department of Biological Sciences, Inha University, Incheon, 22212, South Korea
| | - Sejin Son
- Department of Biological Sciences, Inha University, Incheon, 22212, South Korea.
- Department of Biological Sciences and Bioengineering, Inha University/Industry-Academia Interactive R&E Center for Bioprocess Innovation, Inha University, Incheon, South Korea.
| | - Jutaek Nam
- College of Pharmacy, Chonnam National University, Gwangju, 61186, South Korea.
| |
Collapse
|
8
|
Meng L, Teng Z, Yang S, Wang N, Guan Y, Chen X, Liu Y. Biomimetic nanoparticles for DC vaccination: a versatile approach to boost cancer immunotherapy. NANOSCALE 2023; 15:6432-6455. [PMID: 36916703 DOI: 10.1039/d2nr07071e] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Cancer immunotherapy, which harnesses the immune system to fight cancer, has begun to make a breakthrough in clinical applications. Dendritic cells (DCs) are the bridge linking innate and adaptive immunity and the trigger of tumor immune response. Considering the cumbersome process and poor efficacy of classic DC vaccines, there has been interest in transferring the field of in vitro-generated DC vaccines to nanovaccines. Conventional nanoparticles have insufficient targeting ability and are easily cleared by the reticuloendothelial system. Biological components have evolved very specific functions, which are difficult to fully reproduce with synthetic materials, making people interested in using the further understanding of biological systems to prepare nanoparticles with new and enhanced functions. Biomimetic nanoparticles are semi-biological or nature-derived delivery systems comprising one or more natural materials, which have a long circulation time in vivo and excellent performance of targeting DCs, and can mimic the antigen-presenting behavior of DCs. In this review, we introduce the classification, design, preparation, and challenges of different biomimetic nanoparticles, and discuss their application in activating DCs in vivo and stimulating T cell antitumor immunity. Incorporating biomimetic nanoparticles into cancer immunotherapy has shown outstanding advantages in precisely coaxing the immune system against cancer.
Collapse
Affiliation(s)
- Lingyang Meng
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - Zhuang Teng
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - Shuang Yang
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - Na Wang
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - YingHua Guan
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| | - Xiguang Chen
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
- Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266000, P.R. China
| | - Ya Liu
- College of Marine Life Science, Ocean University of China, Qingdao, 266003, P.R. China.
| |
Collapse
|
9
|
Bezbaruah R, Chavda VP, Nongrang L, Alom S, Deka K, Kalita T, Ali F, Bhattacharjee B, Vora L. Nanoparticle-Based Delivery Systems for Vaccines. Vaccines (Basel) 2022; 10:1946. [PMID: 36423041 PMCID: PMC9694785 DOI: 10.3390/vaccines10111946] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 11/08/2022] [Accepted: 11/09/2022] [Indexed: 11/19/2022] Open
Abstract
Vaccination is still the most cost-effective way to combat infectious illnesses. Conventional vaccinations may have low immunogenicity and, in most situations, only provide partial protection. A new class of nanoparticle-based vaccinations has shown considerable promise in addressing the majority of the shortcomings of traditional and subunit vaccines. This is due to recent breakthroughs in chemical and biological engineering, which allow for the exact regulation of nanoparticle size, shape, functionality, and surface characteristics, resulting in improved antigen presentation and robust immunogenicity. A blend of physicochemical, immunological, and toxicological experiments can be used to accurately characterize nanovaccines. This narrative review will provide an overview of the current scenario of the nanovaccine.
Collapse
Affiliation(s)
- Rajashri Bezbaruah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
| | - Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L. M. College of Pharmacy, Ahmedabad 380008, Gujarat, India
| | - Lawandashisha Nongrang
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
| | - Shahnaz Alom
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India
| | - Kangkan Deka
- Department of Pharmacognosy, NETES Institute of Pharmaceutical Science, Mirza, Guwahati 781125, Assam, India
| | - Tutumoni Kalita
- Department of Pharmaceutical Chemistry, Girijananda Chowdhury Institute of Pharmaceutical Sciences, Azara, Guwahati 781017, Assam, India
| | - Farak Ali
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India
- Department of Pharmaceutical Chemistry, Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India
| | - Bedanta Bhattacharjee
- Department of Pharmacology, Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India
| | | |
Collapse
|
10
|
Wang D, Liu J, Duan J, Ma Y, Gao H, Zhang Z, Liu J, Shi J, Zhang K. Photocontrolled Spatiotemporal Delivery of DNA Immunomodulators for Enhancing Membrane-Targeted Tumor Photodynamic Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2022; 14:44183-44198. [PMID: 36165393 DOI: 10.1021/acsami.2c12774] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Immunotherapy is emerging as a paradigm-shifting modality for treatment cancer. However, systemic administration of immunomodulators is usually accompanied by extra-tumor toxicity and adverse immune effects. Precise delivery of immunomodulators with a highly controllable system may provide a solution for this issue. Here, we developed a photocontrolled DNA nanomedicine for localized delivery of DNA immunomodulators to enhance membrane-targeted photodynamic immunotherapy. Specifically, the DNA nanomedicine is composed of long tandemly repeated functional DNA sequences (PDL1 aptamers and CpG) with a photosensitizer (TMPyP4) inserted into the DNA structure, providing high drug-loading capacity. By blocking the surface PDL1 aptamer with a pHLIP-modified cDNA, the DNA nanomedicine does not induce any obvious immune response and can be specifically delivered and anchored to the tumor membrane. Under localized irradiation, photodynamically generated reactive oxygen species (ROS) causes breakage of DNA sequences, which triggers the collapse of the nanostructure and release of internal DNA immunomodulators. Under localized illumination, photodynamically generated ROS can cause DNA sequence breaks, triggering the collapse of nanostructures and the release of internal DNA immunomodulators thus enhancing membrane-targeted photodynamic immunotherapy. We have demonstrated that the developed DNA nanomedicine can drive efficient immune responses in tumor tissue without perceptibly interfering off-tumor immunity, resulting in efficient antitumor treatment while mitigating systemic toxicity.
Collapse
Affiliation(s)
- Danyu Wang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Jingwen Liu
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Jie Duan
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Yanrui Ma
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Hua Gao
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Junjie Liu
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, State Key Laboratory of Esophageal Cancer Prevention & Treatment, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
11
|
Li P, Wang D, Hu J, Yang X. The role of imaging in targeted delivery of nanomedicine for cancer therapy. Adv Drug Deliv Rev 2022; 189:114447. [PMID: 35863515 DOI: 10.1016/j.addr.2022.114447] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 05/27/2022] [Accepted: 07/06/2022] [Indexed: 01/24/2023]
Abstract
Nanomedicines overcome the pharmacokinetic limitations of traditional drug formulations and have promising prospect in cancer treatment. However, nanomedicine delivery in vivo is still facing challenges from the complex physiological environment. For the purpose of effective tumor therapy, they should be designed to guarantee the five features principle, including long blood circulation, efficient tumor accumulation, deep matrix penetration, enhanced cell internalization and accurate drug release. To ensure the excellent performance of the designed nanomedicine, it would be better to monitor the drug delivery process as well as the therapeutic effects by real-time imaging. In this review, we summarize strategies in developing nanomedicines for efficiently meeting the five features of drug delivery, and the role of several imaging modalities (fluorescent imaging (FL), magnetic resonance imaging (MRI), computed tomography (CT), photoacoustic imaging (PAI), positron emission tomography (PET), and electron microscopy) in tracing drug delivery and therapeutic effect in vivo based on five features principle.
Collapse
Affiliation(s)
- Puze Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Dongdong Wang
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jun Hu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics of Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China.
| |
Collapse
|
12
|
Yang W, Frickenstein AN, Sheth V, Holden A, Mettenbrink EM, Wang L, Woodward AA, Joo BS, Butterfield SK, Donahue ND, Green DE, Thomas AG, Harcourt T, Young H, Tang M, Malik ZA, Harrison RG, Mukherjee P, DeAngelis PL, Wilhelm S. Controlling Nanoparticle Uptake in Innate Immune Cells with Heparosan Polysaccharides. NANO LETTERS 2022; 22:7119-7128. [PMID: 36048773 PMCID: PMC9486251 DOI: 10.1021/acs.nanolett.2c02226] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
We used heparosan (HEP) polysaccharides for controlling nanoparticle delivery to innate immune cells. Our results show that HEP-coated nanoparticles were endocytosed in a time-dependent manner by innate immune cells via both clathrin-mediated and macropinocytosis pathways. Upon endocytosis, we observed HEP-coated nanoparticles in intracellular vesicles and the cytoplasm, demonstrating the potential for nanoparticle escape from intracellular vesicles. Competition with other glycosaminoglycan types inhibited the endocytosis of HEP-coated nanoparticles only partially. We further found that nanoparticle uptake into innate immune cells can be controlled by more than 3 orders of magnitude via systematically varying the HEP surface density. Our results suggest a substantial potential for HEP-coated nanoparticles to target innate immune cells for efficient intracellular delivery, including into the cytoplasm. This HEP nanoparticle surface engineering technology may be broadly used to develop efficient nanoscale devices for drug and gene delivery as well as possibly for gene editing and immuno-engineering applications.
Collapse
Affiliation(s)
- Wen Yang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Alex N. Frickenstein
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Vinit Sheth
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Alyssa Holden
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Evan M. Mettenbrink
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Lin Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Alexis A. Woodward
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Bryan S. Joo
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Sarah K. Butterfield
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Nathan D. Donahue
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Dixy E. Green
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
| | - Abigail G. Thomas
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Tekena Harcourt
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Hamilton Young
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Mulan Tang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Zain A. Malik
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Roger G. Harrison
- School of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
| | - Priyabrata Mukherjee
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
| | - Paul L. DeAngelis
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, 73019, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, 73104, USA
- Institute for Biomedical Engineering, Science, and Technology (IBEST), University of Oklahoma, Norman, Oklahoma, 73019, USA
| |
Collapse
|
13
|
Fang X, Lan H, Jin K, Gong D, Qian J. Nanovaccines for Cancer Prevention and Immunotherapy: An Update Review. Cancers (Basel) 2022; 14:3842. [PMID: 36010836 PMCID: PMC9405528 DOI: 10.3390/cancers14163842] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 07/21/2022] [Accepted: 07/24/2022] [Indexed: 11/23/2022] Open
Abstract
Cancer immunotherapy has received more and more attention from cancer researchers over the past few decades. Various methods such as cell therapy, immune checkpoint blockers, and cancer vaccines alone or in combination therapies have achieved relatively satisfactory results in cancer therapy. Among these immunotherapy-based methods, cancer vaccines alone have not yet had the necessary efficacy in the clinic. Therefore, nanomaterials have increased the efficacy and ef-fectiveness of cancer vaccines by increasing their half-life and durability, promoting tumor mi-croenvironment (TME) reprogramming, and enhancing their anti-tumor immunity with minimal toxicity. In this review, according to the latest studies, the structure and different types of nanovaccines, the mechanisms of these vaccines in cancer treatment, as well as the advantages and disadvantages of these nanovaccines are discussed.
Collapse
Affiliation(s)
- Xingliang Fang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Shaoxing University, Shaoxing 312000, China
| | - Huanrong Lan
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hosptial, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hosptial, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Daojun Gong
- Department of Gastrointestinal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua 321000, China
| | - Jun Qian
- Department of Colorectal Surgery, Xinchang People’s Hospital, Affiliated Xinchang Hosptial, Wenzhou Medical University, Xinchang 312500, China
| |
Collapse
|
14
|
Davodabadi F, Sarhadi M, Arabpour J, Sargazi S, Rahdar A, Díez-Pascual AM. Breast cancer vaccines: New insights into immunomodulatory and nano-therapeutic approaches. J Control Release 2022; 349:844-875. [PMID: 35908621 DOI: 10.1016/j.jconrel.2022.07.036] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/23/2022] [Accepted: 07/25/2022] [Indexed: 10/16/2022]
Abstract
Breast cancer (BC) is known to be a highly heterogeneous disease that is clinically subdivided into four primary molecular subtypes, each having distinct morphology and clinical implications. These subtypes are principally defined by hormone receptors and other proteins involved (or not involved) in BC development. BC therapeutic vaccines [including peptide-based vaccines, protein-based vaccines, nucleic acid-based vaccines (DNA/RNA vaccines), bacterial/viral-based vaccines, and different immune cell-based vaccines] have emerged as an appealing class of cancer immunotherapeutics when used alone or combined with other immunotherapies. Employing the immune system to eliminate BC cells is a novel therapeutic modality. The benefit of active immunotherapies is that they develop protection against neoplastic tissue and readjust the immune system to an anti-tumor monitoring state. Such immunovaccines have not yet shown effectiveness for BC treatment in clinical trials. In recent years, nanomedicines have opened new windows to increase the effectiveness of vaccinations to treat BC. In this context, some nanoplatforms have been designed to efficiently deliver molecular, cellular, or subcellular vaccines to BC cells, increasing the efficacy and persistence of anti-tumor immunity while minimizing undesirable side effects. Immunostimulatory nano-adjuvants, liposomal-based vaccines, polymeric vaccines, virus-like particles, lipid/calcium/phosphate nanoparticles, chitosan-derived nanostructures, porous silicon microparticles, and selenium nanoparticles are among the newly designed nanostructures that have been used to facilitate antigen internalization and presentation by antigen-presenting cells, increase antigen stability, enhance vaccine antigenicity and remedial effectivity, promote antigen escape from the endosome, improve cytotoxic T lymphocyte responses, and produce humoral immune responses in BC cells. Here, we summarized the existing subtypes of BC and shed light on immunomodulatory and nano-therapeutic strategies for BC vaccination. Finally, we reviewed ongoing clinical trials on BC vaccination and highlighted near-term opportunities for moving forward.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran
| | - Mohammad Sarhadi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan 9816743463, Iran
| | - Javad Arabpour
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saman Sargazi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan 9816743463, Iran.
| | - Abbas Rahdar
- Department of Physics, University of Zabol, Zabol 98613-35856, Iran.
| | - Ana M Díez-Pascual
- Universidad de Alcalá, Facultad de Ciencias, Departamento de Química Analítica, Química Física e Ingeniería Química, Ctra. Madrid-Barcelona, Km. 33.6, 28805 Alcalá de Henares, Madrid, Spain.
| |
Collapse
|
15
|
Yu M, Zeng W, Ouyang Y, Liang S, Yi Y, Hao H, Yu J, Liu Y, Nie Y, Wang T, Deng Y, Wu M. ATP-exhausted nanocomplexes for intratumoral metabolic intervention and photoimmunotherapy. Biomaterials 2022; 284:121503. [DOI: 10.1016/j.biomaterials.2022.121503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 11/02/2022]
|
16
|
Li Q, Liu Y, Huang Z, Guo Y, Li Q. Triggering Immune System With Nanomaterials for Cancer Immunotherapy. Front Bioeng Biotechnol 2022; 10:878524. [PMID: 35497343 PMCID: PMC9046726 DOI: 10.3389/fbioe.2022.878524] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/30/2022] [Indexed: 12/29/2022] Open
Abstract
Cancer is a major cause of incidence rate and mortality worldwide. In recent years, cancer immunotherapy has made great progress in the preclinical and clinical treatment of advanced malignant tumors. However, cancer patients will have transient cancer suppression reaction and serious immune related adverse reactions when receiving immunotherapy. In recent years, nanoparticle-based immunotherapy, which can accurately deliver immunogens, activate antigen presenting cells (APCs) and effector cells, provides a new insight to solve the above problems. In this review, we discuss the research progress of nanomaterials in immunotherapy including nanoparticle-based delivery systems, nanoparticle-based photothermal and photodynamic immunotherapy, nanovaccines, nanoparticle-based T cell cancer immunotherapy and nanoparticle-based bacteria cancer immunotherapy. We also put forward the current challenges and prospects of immunomodulatory therapy.
Collapse
Affiliation(s)
| | | | | | - Yajie Guo
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Qingjiao Li
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
17
|
Wang TT, Xia YY, Gao JQ, Xu DH, Han M. Recent Progress in the Design and Medical Application of In Situ Self-Assembled Polypeptide Materials. Pharmaceutics 2021; 13:753. [PMID: 34069645 PMCID: PMC8160760 DOI: 10.3390/pharmaceutics13050753] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 05/05/2021] [Accepted: 05/11/2021] [Indexed: 12/20/2022] Open
Abstract
Inspired by molecular self-assembly, which is ubiquitous in natural environments and biological systems, self-assembled peptides have become a research hotspot in the biomedical field due to their inherent biocompatibility and biodegradability, properties that are afforded by the amide linkages forming the peptide backbone. This review summarizes the biological advantages, principles, and design strategies of self-assembled polypeptide systems. We then focus on the latest advances in in situ self-assembly of polypeptides in medical applications, such as oncotherapy, materials science, regenerative medicine, and drug delivery, and then briefly discuss their potential challenges in clinical treatment.
Collapse
Affiliation(s)
- Tian-Tian Wang
- Department of Pharmacy, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China;
| | - Yi-Yi Xia
- Institution of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (Y.-Y.X.); (J.-Q.G.)
| | - Jian-Qing Gao
- Institution of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (Y.-Y.X.); (J.-Q.G.)
| | - Dong-Hang Xu
- Department of Pharmacy, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China;
| | - Min Han
- Institution of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China; (Y.-Y.X.); (J.-Q.G.)
| |
Collapse
|
18
|
Zheng B, Peng W, Gan L, Guo M, Wang S, Zhang XD, Ming D. Sendai virus-based immunoadjuvant in hydrogel vaccine intensity-modulated dendritic cells activation for suppressing tumorigenesis. Bioact Mater 2021; 6:3879-3891. [PMID: 33937591 PMCID: PMC8076650 DOI: 10.1016/j.bioactmat.2021.04.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 03/30/2021] [Accepted: 04/01/2021] [Indexed: 12/19/2022] Open
Abstract
The conventional immunoadjuvants in vaccine have weak effect on stimulating antigen presentation and activating anti-tumor immunity. Unexpectedly, we discovered that non-pathogenic Sendai virus (SeV) could activate antigen-presenting cells (APCs) represented by dendritic cells (DCs). Here, we designed an injectable SeV-based hydrogel vaccine (SHV) to execute multi-channel recruitment and stimulation of DCs for boosting the specific immune response against tumors. After the release of the NIR-triggered antigens from tumor cells, dendritic cells around the vaccine efficiently transport the antigens to lymph nodes and present them to T lymphocytes, thereby inducing systemic anti-tumor immune memory. Our findings demonstrated that the SHV with excellent universality, convenience and flexibility has achieved better immune protection effects in inhibiting the occurrence of melanoma and breast cancer. In conclusion, the SHV system might serve as the next generation of personalized anti-tumor vaccines with enhanced features over standard vaccination regimens, and represented an alternative way to suppress tumorigenesis. SeV served as immuneadjuvant can activate APCs through TLR7/8 and TLR3 pathways. Non-pathogenic SeV in the injectable hydrogel vaccine recruit and activate DCs. Tumor cells acted as an “antigen library” to release all antigens by NIR-trigger. Fragmented DNA from tumor cells after photothermal damage activated STING pathway. The synergy effect of SHV and aOX40 greatly enhanced anti-tumor immune memory.
Collapse
Affiliation(s)
- Bin Zheng
- Academy of Medical Engineering and Translational Medicine, Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
- Corresponding author.
| | - Wenchang Peng
- Academy of Medical Engineering and Translational Medicine, Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Lin Gan
- School of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin, 300072, China
| | - Mingming Guo
- Academy of Medical Engineering and Translational Medicine, Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Shuchao Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Xiao-Dong Zhang
- Academy of Medical Engineering and Translational Medicine, Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China
- School of Precision Instrument and Opto-electronics Engineering, Tianjin University, Tianjin, 300072, China
- Corresponding author. Academy of Medical Engineering and Translational Medicine, Tianjin Key Laboratory of Brain Science and Neural Engineering, Tianjin University, 92 Weijin Road, Nankai District, Tianjin, 300072, China.
| |
Collapse
|
19
|
Wang Z, Ren C, Shang Y, Yang C, Guo Q, Chu L, Liu J. Co-assembled Supramolecular Nanofibers With Tunable Surface Properties for Efficient Vaccine Delivery. Front Chem 2020; 8:500. [PMID: 32850613 PMCID: PMC7396696 DOI: 10.3389/fchem.2020.00500] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 05/14/2020] [Indexed: 12/30/2022] Open
Abstract
The utilization of nanotechnology to deliver vaccines and modulate immunity has shown great potential in cancer therapy. Peptide-based supramolecular hydrogels as novel vaccine adjuvants have been found to effectively improve the immune response and tumor curative effect. In this study, we designed a set of reduction-responsive self-assembled peptide precursors (Fbp-GDFDFDYD(E, S, or K)-ss-ERGD), which can be reduced by glutathione (GSH) into Fbp-GDFDFDYD(E, S or K)-SH for forming of hydrogel with different surface properties (E-gel, S-gel, and K-gel, respectively). Using the same method, co-assembled hydrogel vaccines (E-vac, S-vac, and K-vac, respectively) can also be prepared by mixing different precursors with antigens before GSH reduction. Through TEM observation of the nanostructure, we found that all the co-assembled hydrogels, especially K-vac, possessed much denser and more unified nanofiber networks as compared with antigen-free hydrogels, which were very suitable for antigen storage and vaccine delivery. Although the three peptides adopted similar β-sheet secondary structures, the mechanical properties of their resulted co-assembled hydrogel vaccines were obviously different. Compared to E-vac, S-vac had a much weaker mechanical property, while K-vac had a much higher. In vivo experiments, co-assembled hydrogel vaccines, especially K-vac, also promoted antibody production and anti-tumor immune responses more significantly than the other two vaccines. Our results demonstrated that co-assembled hydrogels formed by peptides and antigens co-assembly could act as effective vaccine delivery systems for boosting antibody production, and different immune effects can be acquired by tuning the surface properties of the involved self-assembling peptides.
Collapse
Affiliation(s)
- Zhongyan Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Chunhua Ren
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Yuna Shang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Cuihong Yang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Qingxiang Guo
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Liping Chu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Jianfeng Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| |
Collapse
|
20
|
Wang Z, Shang Y, Tan Z, Li X, Li G, Ren C, Wang F, Yang Z, Liu J. A supramolecular protein chaperone for vaccine delivery. Theranostics 2020; 10:657-670. [PMID: 31903143 PMCID: PMC6929975 DOI: 10.7150/thno.39132] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 09/26/2019] [Indexed: 12/11/2022] Open
Abstract
Rationale: Nanomaterials capable of specifically interacting with proteins are very important for protein storage and vaccine delivery. Supramolecular hydrogels based on peptides have emerged as promising vaccine adjuvants because of their good compatibility, ease of antigen incorporation and display, and efficiency in activating immune responses. Methods: We synthesized a self-assembling peptide (Fbp-GDFDFDYDK(γE)2-NH2, Comp. 1 ) serving as a supramolecular protein chaperone for protein antigen delivery. The gelation was triggered by simply mixing Comp. 1 and proteins. The vaccine adjuvant potential of Comp. 1 was demonstrated by using two protein antigens, ovalbumin (OVA) and hepatitis B surface antigen (HBsAg). Results: The peptide derivative Comp. 1 exhibited high protein binding capacity. Upon contacting proteins, Comp. 1 rapidly formed coassembled nanofibers/hydrogels with the proteins, which greatly delayed the release of protein antigens. Our supramolecular protein chaperone significantly stimulated specific antibody titers by assisting protein delivery to antigen-presenting cells, promoting dendritic cell (DC) maturation, prolonging antigen accumulation and retention in the lymph nodes, and eliciting the secretion of cytokines. Most importantly, our supramolecular protein chaperone strongly stimulated the cellular immune response and significantly retarded tumor growth. Conclusion: Our study demonstrated the great potential of the supramolecular protein chaperone in protein storage and delivery, vaccine production and tumor immunotherapy.
Collapse
Affiliation(s)
- Zhongyan Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, P.R. China
| | - Yuna Shang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, P. R. China
| | - Zhaoqi Tan
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, P. R. China
| | - Xiaoyan Li
- Analysis Center, Nanjing Medical University, Nanjing, Jiangsu 210029, P. R. China
| | - Guoliang Li
- Lab of Functional and Biomedical Nanomaterials, College of Materials Science and Engineering, Qingdao University of Science and Technology, Qingdao, 266042, P. R. China
| | - Chunhua Ren
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, P.R. China
| | - Fuqiang Wang
- Analysis Center, Nanjing Medical University, Nanjing, Jiangsu 210029, P. R. China
| | - Zhimou Yang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, P. R. China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, P. R. China
| | - Jianfeng Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, P.R. China
- Lab of Functional and Biomedical Nanomaterials, College of Materials Science and Engineering, Qingdao University of Science and Technology, Qingdao, 266042, P. R. China
| |
Collapse
|
21
|
Zhang Y, Lin S, Wang XY, Zhu G. Nanovaccines for cancer immunotherapy. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2019; 11:e1559. [PMID: 31172659 PMCID: PMC7040494 DOI: 10.1002/wnan.1559] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 03/27/2019] [Indexed: 12/17/2022]
Abstract
The past few decades have witnessed the booming field of cancer immunotherapy. Cancer therapeutic vaccines, either alone or in combination with other immunotherapies such as adoptive cell therapy or immune checkpoint blockade therapy, are an attractive class of cancer immunotherapeutics. However, cancer vaccines have thus far shown suboptimal efficacy in the clinic. Nanomedicines offer unique opportunities to improve the efficacy of these vaccines. A variety of nanoplatforms have been investigated to deliver molecular or cellular or subcellular vaccines to target lymphoid tissues and cells, thereby promoting the potency and durability of anti-tumor immunity while reducing adverse side effects. In this article, we reviewed the key parameters and features of nanovaccines for cancer immunotherapy; we highlighted recent advances in the development of cancer nanovaccines based on synthetic nanocarriers, biogenic nanocarriers, as well as semi-biogenic nanocarriers; and we summarized newly emerging types of nanovaccines, such as those based on stimulator of interferon genes agonists, cancer neoantigens, mRNA vaccines, as well as artificial antigen-presenting cells. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Rehabilitation Medicine, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Center for Pharmaceutical Engineering and Sciences, Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
| | - Shuibin Lin
- Department of Rehabilitation Medicine, Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, Virginia
- Institute of Molecular Medicine, Virginia Commonwealth University, Richmond, Virginia
- Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| | - Guizhi Zhu
- Center for Pharmaceutical Engineering and Sciences, Department of Pharmaceutics, School of Pharmacy, Virginia Commonwealth University, Richmond, Virginia
- The Developmental Therapeutics Program, Massey Cancer Center, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|