1
|
Zheng W, Li S, Shi Z, Su K, Ding Y, Zhang L, Tang Q, Han J, Zhao H, Wang F, Zhang H, Hong Z. Recombinant ferritin-based nanoparticles as neoantigen carriers significantly inhibit tumor growth and metastasis. J Nanobiotechnology 2024; 22:562. [PMID: 39272180 PMCID: PMC11401311 DOI: 10.1186/s12951-024-02837-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 09/05/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Tumor neoantigen peptide-based vaccines, systemic immunotherapies that enhance antitumor immunity by activating and expanding antigen-specific T cells, have achieved remarkable results in the treatment of a variety of solid tumors. However, how to effectively deliver neoantigens to induce robust antitumor immune responses remains a major obstacle. RESULTS Here, we developed a safe and effective neoantigen peptide delivery system (neoantigen-ferritin nanoparticles, neoantigen-FNs) that successfully achieved effective lymph node targeting and induced robust antitumor immune responses. The genetically engineered self-assembled particles neoantigen-FNs with a size of 12 nm were obtained by fusing a neoantigen with optimized ferritin, which rapidly drainage to and continuously accumulate in lymph nodes. The neoantigen-FNs vaccine induced a greater quantity and quality of antigen-specific CD8+ T cells and resulted in significant growth control of multiple tumors, dramatic inhibition of melanoma metastasis and regression of established tumors. In addition, no obvious toxic side effects were detected in the various models, indicating the high safety of optimized ferritin as a vaccine carrier. CONCLUSIONS Homogeneous and safe neoantigen-FNs could be a very promising system for neoantigen peptide delivery because of their ability to efficiently drainage to lymph nodes and induce efficient antitumor immune responses.
Collapse
Affiliation(s)
- Wei Zheng
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Shixiong Li
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Zhongliang Shi
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Kailing Su
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Yu Ding
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Luyue Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Qian Tang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Jiani Han
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Han Zhao
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Fengwei Wang
- School of Medicine, Nankai University, Tianjin, 300071, PR China
- People's Hospital of Tianjin, Tianjin, 300180, PR China
| | - Hongru Zhang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
- Nankai International Advanced Research Institute (SHENZHEN FUTIAN), Shenzhen, 518045, PR China.
| | - Zhangyong Hong
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Protein Sciences, Cancer Biology Center, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
- Nankai International Advanced Research Institute (SHENZHEN FUTIAN), Shenzhen, 518045, PR China.
| |
Collapse
|
2
|
Cao S, Ma D, Ji S, Zhou M, Zhu S. Self-Assembled Ferritin Nanoparticles for Delivery of Antigens and Development of Vaccines: From Structure and Property to Applications. Molecules 2024; 29:4221. [PMID: 39275069 PMCID: PMC11397193 DOI: 10.3390/molecules29174221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/31/2024] [Accepted: 09/04/2024] [Indexed: 09/16/2024] Open
Abstract
Ferritin, an iron storage protein, is ubiquitously distributed across diverse life forms, fulfilling crucial roles encompassing iron retention, conversion, orchestration of cellular iron metabolism, and safeguarding cells against oxidative harm. Noteworthy attributes of ferritin include its innate amenability to facile modification, scalable mass production, as well as exceptional stability and safety. In addition, ferritin boasts unique physicochemical properties, including pH responsiveness, resilience to elevated temperatures, and resistance to a myriad of denaturing agents. Therefore, ferritin serves as the substrate for creating nanomaterials typified by uniform particle dimensions and exceptional biocompatibility. Comprising 24 subunits, each ferritin nanocage demonstrates self-assembly capabilities, culminating in the formation of nanostructures akin to intricate cages. Recent years have witnessed the ascendance of ferritin-based self-assembled nanoparticles, owing to their distinctive physicochemical traits, which confer substantial advantages and wide-ranging applications within the biomedical domain. Ferritin is highly appealing as a carrier for delivering drug molecules and antigen proteins due to its distinctive structural and biochemical properties. This review aims to highlight recent advances in the use of self-assembled ferritin as a novel carrier for antigen delivery and vaccine development, discussing the molecular mechanisms underlying its action, and presenting it as a promising and effective strategy for the future of vaccine development.
Collapse
Affiliation(s)
- Shinuo Cao
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225306, China;
| | - Dongxue Ma
- Department of Veterinary Medicine, Agriculture College of Yanbian University, Yanji 133000, China; (D.M.); (S.J.)
| | - Shengwei Ji
- Department of Veterinary Medicine, Agriculture College of Yanbian University, Yanji 133000, China; (D.M.); (S.J.)
| | - Mo Zhou
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225306, China;
| | - Shanyuan Zhu
- Jiangsu Key Laboratory for High-Tech Research and Development of Veterinary Biopharmaceuticals, Engineering Technology Research Center for Modern Animal Science and Novel Veterinary Pharmaceutic Development, Jiangsu Agri-Animal Husbandry Vocational College, Taizhou 225306, China;
| |
Collapse
|
3
|
Pan X, Han T, Zhao Z, Wang X, Fang X. Emerging Nanotechnology in Preclinical Pancreatic Cancer Immunotherapy: Driving Towards Clinical Applications. Int J Nanomedicine 2024; 19:6619-6641. [PMID: 38975321 PMCID: PMC11227336 DOI: 10.2147/ijn.s466459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 06/16/2024] [Indexed: 07/09/2024] Open
Abstract
The high malignant degree and poor prognosis of pancreatic cancer (PC) pose severe challenges to the basic research and clinical translation of next-generation therapies. The rise of immunotherapy has improved the treatment of a variety of solid tumors, while the application in PC is highly restricted by the challenge of immunosuppressive tumor microenvironment. The latest progress of nanotechnology as drug delivery platform and immune adjuvant has improved drug delivery in a variety of disease backgrounds and enhanced tumor therapy based on immunotherapy. Based on the immune loop of PC and the status quo of clinical immunotherapy of tumors, this article discussed and critically analyzed the key transformation difficulties of immunotherapy adaptation to the treatment of PC, and then proposed the rational design strategies of new nanocarriers for drug delivery and immune regulation, especially the design of combined immunotherapy. This review also put forward prospective views on future research directions, so as to provide information for the new means of clinical treatment of PC combined with the next generation of nanotechnology and immunotherapy.
Collapse
Affiliation(s)
- Xuan Pan
- Department of Hepato-Biliary-Pancreatic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, People’s Republic of China
| | - Ting Han
- Department of Gastroenterology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, People’s Republic of China
| | - Zixuan Zhao
- The Translational Research Institute for Neurological Disorders of Department of Neurosurgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, People’s Republic of China
- The Institute of Brain Science, Wannan Medical College, Wuhu, 241000, People’s Republic of China
| | - Xiaoming Wang
- Department of Hepato-Biliary-Pancreatic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, People’s Republic of China
| | - Xiaosan Fang
- Department of Hepato-Biliary-Pancreatic Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241000, People’s Republic of China
| |
Collapse
|
4
|
Xu M, Wei S, Duan L, Ji Y, Han X, Sun Q, Weng L. The recent advancements in protein nanoparticles for immunotherapy. NANOSCALE 2024; 16:11825-11848. [PMID: 38814163 DOI: 10.1039/d4nr00537f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
In recent years, the advancement of nanoparticle-based immunotherapy has introduced an innovative strategy for combatting diseases. Compared with other types of nanoparticles, protein nanoparticles have obtained substantial attention owing to their remarkable biocompatibility, biodegradability, ease of modification, and finely designed spatial structures. Nature provides several protein nanoparticle platforms, including viral capsids, ferritin, and albumin, which hold significant potential for disease treatment. These naturally occurring protein nanoparticles not only serve as effective drug delivery platforms but also augment antigen delivery and targeting capabilities through techniques like genetic modification and covalent conjugation. Motivated by nature's originality and driven by progress in computational methodologies, scientists have crafted numerous protein nanoparticles with intricate assembly structures, showing significant potential in the development of multivalent vaccines. Consequently, both naturally occurring and de novo designed protein nanoparticles are anticipated to enhance the effectiveness of immunotherapy. This review consolidates the advancements in protein nanoparticles for immunotherapy across diseases including cancer and other diseases like influenza, pneumonia, and hepatitis.
Collapse
Affiliation(s)
- Miaomiao Xu
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| | - Siyuan Wei
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| | - Lifan Duan
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| | - Yifan Ji
- Portland Institute, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Xiaofan Han
- Portland Institute, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Qipeng Sun
- Portland Institute, Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Lixing Weng
- State Key Laboratory of Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| |
Collapse
|
5
|
Hao X, Yuan F, Yao X. Advances in virus-like particle-based SARS-CoV-2 vaccines. Front Cell Infect Microbiol 2024; 14:1406091. [PMID: 38988812 PMCID: PMC11233461 DOI: 10.3389/fcimb.2024.1406091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024] Open
Abstract
The Coronavirus Disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has incurred devastating human and economic losses. Vaccination remains the most effective approach for controlling the COVID-19 pandemic. Nonetheless, the sustained evolution of SARS-CoV-2 variants has provoked concerns among the scientific community regarding the development of next-generation COVID-19 vaccines. Among these, given their safety, immunogenicity, and flexibility to display varied and native epitopes, virus-like particle (VLP)-based vaccines represent one of the most promising next-generation vaccines. In this review, we summarize the advantages and characteristics of VLP platforms, strategies for antigen display, and current clinical trial progress of SARS-CoV-2 vaccines based on VLP platforms. Importantly, the experience and lessons learned from the development of SARS-CoV-2 VLP vaccines provide insights into the development of strategies based on VLP vaccines to prevent future coronavirus pandemics and other epidemics.
Collapse
Affiliation(s)
- Xiaoting Hao
- Department of Teaching Administration, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Feifei Yuan
- Department of Reproductive Medicine, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| | - Xuan Yao
- Department of Neurology, Xiangyang No.1 People’s Hospital, Hubei University of Medicine, Xiangyang, China
| |
Collapse
|
6
|
Sobczak JM, Barkovska I, Balke I, Rothen DA, Mohsen MO, Skrastina D, Ogrina A, Martina B, Jansons J, Bogans J, Vogel M, Bachmann MF, Zeltins A. Identifying Key Drivers of Efficient B Cell Responses: On the Role of T Help, Antigen-Organization, and Toll-like Receptor Stimulation for Generating a Neutralizing Anti-Dengue Virus Response. Vaccines (Basel) 2024; 12:661. [PMID: 38932390 PMCID: PMC11209419 DOI: 10.3390/vaccines12060661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024] Open
Abstract
T help (Th), stimulation of toll-like receptors (pathogen-associated molecular patterns, PAMPs), and antigen organization and repetitiveness (pathogen-associated structural patterns, PASPs) were shown numerous times to be important in driving B-cell and antibody responses. In this study, we dissected the individual contributions of these parameters using newly developed "Immune-tag" technology. As model antigens, we used eGFP and the third domain of the dengue virus 1 envelope protein (DV1 EDIII), the major target of virus-neutralizing antibodies. The respective proteins were expressed alone or genetically fused to the N-terminal fragment of the cucumber mosaic virus (CMV) capsid protein-nCMV, rendering the antigens oligomeric. In a step-by-step manner, RNA was attached as a PAMP, and/or a universal Th-cell epitope was genetically added for additional Th. Finally, a PASP was added to the constructs by displaying the antigens highly organized and repetitively on the surface of CMV-derived virus-like particles (CuMV VLPs). Sera from immunized mice demonstrated that each component contributed stepwise to the immunogenicity of both proteins. All components combined in the CuMV VLP platform induced by far the highest antibody responses. In addition, the DV1 EDIII induced high levels of DENV-1-neutralizing antibodies only if displayed on VLPs. Thus, combining multiple cues typically associated with viruses results in optimal antibody responses.
Collapse
Affiliation(s)
- Jan M. Sobczak
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Irena Barkovska
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Ina Balke
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Dominik A. Rothen
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Mona O. Mohsen
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Dace Skrastina
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Anete Ogrina
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Byron Martina
- Artemis Bioservices, 2629 JD Delft, The Netherlands;
- Protinhi Therapeutics, 6534 AT Nijmegen, The Netherlands
| | - Juris Jansons
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Janis Bogans
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| | - Monique Vogel
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
| | - Martin F. Bachmann
- Department of Immunology, University Clinic of Rheumatology and Immunology, Inselspital, CH-3010 Bern, Switzerland; (D.A.R.); (M.O.M.); (M.V.); (M.F.B.)
- Department of BioMedical Research, University of Bern, CH-3008 Bern, Switzerland
- Nuffield Department of Medicine, The Jenner Institute, University of Oxford, Oxford OX3 7BN, UK
| | - Andris Zeltins
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (I.B.); (I.B.); (D.S.); (A.O.); (J.J.); (J.B.); (A.Z.)
| |
Collapse
|
7
|
Fan Z, Iqbal H, Ni J, Khan NU, Irshad S, Razzaq A, Alfaifi MY, Elbehairi SEI, Shati AA, Zhou J, Cheng H. Rationalized landscape on protein-based cancer nanomedicine: Recent progress and challenges. Int J Pharm X 2024; 7:100238. [PMID: 38511068 PMCID: PMC10951516 DOI: 10.1016/j.ijpx.2024.100238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/22/2024] Open
Abstract
The clinical advancement of protein-based nanomedicine has revolutionized medical professionals' perspectives on cancer therapy. Protein-based nanoparticles have been exploited as attractive vehicles for cancer nanomedicine due to their unique properties derived from naturally biomacromolecules with superior biocompatibility and pharmaceutical features. Furthermore, the successful translation of Abraxane™ (paclitaxel-based albumin nanoparticles) into clinical application opened a new avenue for protein-based cancer nanomedicine. In this mini-review article, we demonstrate the rational design and recent progress of protein-based nanoparticles along with their applications in cancer diagnosis and therapy from recent literature. The current challenges and hurdles that hinder clinical application of protein-based nanoparticles are highlighted. Finally, future perspectives for translating protein-based nanoparticles into clinic are identified.
Collapse
Affiliation(s)
- Zhechen Fan
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Haroon Iqbal
- Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China
| | - Jiang Ni
- Department of Pharmacy, Affiliated Hospital of Jiangnan University, Wuxi 214000, China
| | - Naveed Ullah Khan
- Department of Pharmacy, Zhejiang University of Technology, Hangzhou 310000, China
| | - Shahla Irshad
- Department of Allied Health Sciences, Faculty of Health and Medical Sciences, Mirpur University of Science and Technology (MUST), Mirpur, Azad Jammu and Kashmir 10250, Pakistan
| | - Anam Razzaq
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Mohammad Y. Alfaifi
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | | | - Ali A. Shati
- King Khalid University, Faculty of Science, Biology Department, Abha 9004, Saudi Arabia
| | - Jianping Zhou
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| | - Hao Cheng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
8
|
Sarkar Lotfabadi A, Abadi B, Rezaei N. Biomimetic nanotechnology for cancer immunotherapy: State of the art and future perspective. Int J Pharm 2024; 654:123923. [PMID: 38403091 DOI: 10.1016/j.ijpharm.2024.123923] [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: 12/10/2023] [Revised: 02/11/2024] [Accepted: 02/18/2024] [Indexed: 02/27/2024]
Abstract
Cancer continues to be a significant worldwide cause of mortality. This underscores the urgent need for novel strategies to complement and overcome the limitations of conventional therapies, such as imprecise targeting and drug resistance. Cancer Immunotherapy utilizes the body's immune system to target malignant cells, reducing harm to healthy tissue. Nevertheless, the efficacy of immunotherapy exhibits variation across individuals and has the potential to induce autoimmune responses. Biomimetic nanoparticles (bNPs) have transformative potential in cancer immunotherapy, promising improved accurate targeting, immune system activation, and resistance mechanisms, while also reducing the occurrence of systemic autoimmune side effects. This integration offers opportunities for personalized medicine and better therapeutic outcomes. Despite considerable potential, bNPs face barriers like insufficient targeting, restricted biological stability, and interactions within the tumor microenvironment. The resolution of these concerns is crucial in order to expedite the integration of bNPs from the research setting into clinical therapeutic uses. In addition, optimizing manufacturing processes and reducing bNP-related costs are essential for practical implementation. The present research introduces comprehensive classifications of bNPs as well as recent achievements in their application in cancer immunotherapies, emphasizing the need to address barriers for swift clinical integration.
Collapse
Affiliation(s)
- Alireza Sarkar Lotfabadi
- Department of Cellular and Molecular Biology, Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Banafshe Abadi
- Herbal and Traditional Medicines Research Center, Kerman University of Medical Sciences, Kerman, Iran; Brain Cancer Research Core (BCRC), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Zhang W, Wang H, Wu T, Gao X, Shang Y, Zhang Z, Liu X, Li Y. A SARS-CoV-2 Nanobody Displayed on the Surface of Human Ferritin with High Neutralization Activity. Int J Nanomedicine 2024; 19:2429-2440. [PMID: 38476285 PMCID: PMC10929646 DOI: 10.2147/ijn.s450829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Purpose COVID-19 is rampant throughout the world, which has caused great damage to human lives and seriously hindered the development of the global economy. Aiming at the treatment of SARS-CoV-2, in this study, we proposed a novel fenobody strategy based on ferritin (Fe) self-assembly technology. Methods The neutralizing nanobody H11-D4 of SARS-CoV-2 fused to the C-terminus of end-modified human ferritin was expressed in E. coli and silkworm baculovirus expression systems. A large number of nanoparticles were successfully self-assembled in silkworms, while relatively few nanoparticles can be observed in the treated products from E. coli by electron microscopy. Subsequently, the fenobody's expression level and neutralizing activity were then evaluated. Results The results showed that the IC50 of H11-D4 and fenobody Fe-H11-D4 expressed in E. coli were 171.1 nmol L-1 and 20.87 nmol L-1, respectively. However, the IC50 of Fe-HD11-D4 expressed in silkworms was 1.46 nmol L-1 showing better neutralization activity. Conclusion Therefore, fenobodies can be well self-assembled in silkworm baculovirus expression system, and ferritin self-assembly technology can effectively improve nanobody neutralization activity.
Collapse
Affiliation(s)
- Wenrong Zhang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
- College of Life Sciences, Capital Normal University, Beijing, People’s Republic of China
| | - Haining Wang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Tong Wu
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Xintao Gao
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Yuting Shang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Zhifang Zhang
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Xingjian Liu
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| | - Yinü Li
- National Key Laboratory of Agricultural Microbiology, Biotechnology Research Institute, Chinese Academy of Agricultural Sciences, Beijing, People’s Republic of China
| |
Collapse
|
10
|
Kim M, Yoon HJ, Lee C, Lee M, Park RW, Lee B, Park EJ, Kim S. Immune Checkpoint-Blocking Nanocages Cross the Blood-Brain Barrier and Impede Brain Tumor Growth. ACS Biomater Sci Eng 2024; 10:575-587. [PMID: 38150627 PMCID: PMC10777349 DOI: 10.1021/acsbiomaterials.3c01200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 12/29/2023]
Abstract
Glioblastoma (GBM) is the deadliest tumor of the central nervous system, with a median survival of less than 15 months. Despite many trials, immune checkpoint-blocking (ICB) therapies using monoclonal antibodies against the PD-1/PD-L1 axis have demonstrated only limited benefits for GBM patients. Currently, the main hurdles in brain tumor therapy include limited drug delivery across the blood-brain barrier (BBB) and the profoundly immune-suppressive microenvironment of GBM. Thus, there is an urgent need for new therapeutics that can cross the BBB and target brain tumors to modulate the immune microenvironment. To this end, we developed an ICB strategy based on the BBB-permeable, 24-subunit human ferritin heavy chain, modifying the ferritin surface with 24 copies of PD-L1-blocking peptides to create ferritin-based ICB nanocages. The PD-L1pep ferritin nanocages first demonstrated their tumor-targeting and antitumor activities in an allograft colon cancer model. Next, we found that these PD-L1pep ferritin nanocages efficiently penetrated the BBB and targeted brain tumors through specific interactions with PD-L1, significantly inhibiting tumor growth in an orthotopic intracranial tumor model. The addition of PD-L1pep ferritin nanocages to triple in vitro cocultures of T cells, GBM cells, and glial cells significantly inhibited PD-1/PD-L1 interactions and restored T-cell activity. Collectively, these findings indicate that ferritin nanocages displaying PD-L1-blocking peptides can overcome the primary hurdle of brain tumor therapy and are, therefore, promising candidates for treating GBM.
Collapse
Affiliation(s)
- Minseong Kim
- Department
of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
- BK21
Plus KNU Biomedical Convergence Program, Department of Biomedical
Science, School of Medicine, Kyungpook National
University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic
of Korea
- CMRI,
School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
| | - Hee Jung Yoon
- Immuno-Oncology
Branch, Division of Cancer Biomedical Science, Graduate School of
Cancer Science and Policy, National Cancer
Center, Goyang 10408, Republic
of Korea
| | - Chanju Lee
- Immuno-Oncology
Branch, Division of Cancer Biomedical Science, Graduate School of
Cancer Science and Policy, National Cancer
Center, Goyang 10408, Republic
of Korea
| | - Minah Lee
- Department
of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
- BK21
Plus KNU Biomedical Convergence Program, Department of Biomedical
Science, School of Medicine, Kyungpook National
University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic
of Korea
- CMRI,
School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
| | - Rang-Woon Park
- Department
of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
- BK21
Plus KNU Biomedical Convergence Program, Department of Biomedical
Science, School of Medicine, Kyungpook National
University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic
of Korea
- CMRI,
School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
| | - Byungheon Lee
- Department
of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
- BK21
Plus KNU Biomedical Convergence Program, Department of Biomedical
Science, School of Medicine, Kyungpook National
University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic
of Korea
- CMRI,
School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
| | - Eun Jung Park
- Immuno-Oncology
Branch, Division of Cancer Biomedical Science, Graduate School of
Cancer Science and Policy, National Cancer
Center, Goyang 10408, Republic
of Korea
| | - Soyoun Kim
- Department
of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
- BK21
Plus KNU Biomedical Convergence Program, Department of Biomedical
Science, School of Medicine, Kyungpook National
University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic
of Korea
- CMRI,
School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu 41944, Republic of Korea
| |
Collapse
|
11
|
Nagar N, Naidu G, Mishra A, Poluri KM. Protein-Based Nanocarriers and Nanotherapeutics for Infection and Inflammation. J Pharmacol Exp Ther 2024; 388:91-109. [PMID: 37699711 DOI: 10.1124/jpet.123.001673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 08/04/2023] [Accepted: 08/21/2023] [Indexed: 09/14/2023] Open
Abstract
Infectious and inflammatory diseases are one of the leading causes of death globally. The status quo has become more prominent with the onset of the coronavirus disease 2019 (COVID-19) pandemic. To combat these potential crises, proteins have been proven as highly efficacious drugs, drug targets, and biomarkers. On the other hand, advancements in nanotechnology have aided efficient and sustained drug delivery due to their nano-dimension-acquired advantages. Combining both strategies together, the protein nanoplatforms are equipped with the advantageous intrinsic properties of proteins as well as nanoformulations, eloquently changing the field of nanomedicine. Proteins can act as carriers, therapeutics, diagnostics, and theranostics in their nanoform as fusion proteins or as composites with other organic/inorganic materials. Protein-based nanoplatforms have been extensively explored to target the major infectious and inflammatory diseases of clinical concern. The current review comprehensively deliberated proteins as nanocarriers for drugs and nanotherapeutics for inflammatory and infectious agents, with special emphasis on cancer and viral diseases. A plethora of proteins from diverse organisms have aided in the synthesis of protein-based nanoformulations. The current study specifically presented the proteins of human and pathogenic origin to dwell upon the field of protein nanotechnology, emphasizing their pharmacological advantages. Further, the successful clinical translation and current bottlenecks of the protein-based nanoformulations associated with the infection-inflammation paradigm have also been discussed comprehensively. SIGNIFICANCE STATEMENT: This review discusses the plethora of promising protein-based nanocarriers and nanotherapeutics explored for infectious and inflammatory ailments, with particular emphasis on protein nanoparticles of human and pathogenic origin with reference to the advantages, ADME (absorption, distribution, metabolism, and excretion parameters), and current bottlenecks in development of protein-based nanotherapeutic interventions.
Collapse
Affiliation(s)
- Nupur Nagar
- Department of Biosciences and Bioengineering (N.N., G.N., K.M.P.) and Centre for Nanotechnology (K.M.P.), Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India; and Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India (A.M.)
| | - Goutami Naidu
- Department of Biosciences and Bioengineering (N.N., G.N., K.M.P.) and Centre for Nanotechnology (K.M.P.), Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India; and Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India (A.M.)
| | - Amit Mishra
- Department of Biosciences and Bioengineering (N.N., G.N., K.M.P.) and Centre for Nanotechnology (K.M.P.), Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India; and Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India (A.M.)
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering (N.N., G.N., K.M.P.) and Centre for Nanotechnology (K.M.P.), Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India; and Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India (A.M.)
| |
Collapse
|
12
|
Hu J, Sha X, Li Y, Wu J, Ma J, Zhang Y, Yang R. Multifaceted Applications of Ferritin Nanocages in Delivering Metal Ions, Bioactive Compounds, and Enzymes: A Comprehensive Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:19903-19919. [PMID: 37955969 DOI: 10.1021/acs.jafc.3c05510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Ferritin, a distinctive iron-storage protein, possesses a unique cage-like nanoscale structure that enables it to encapsulate and deliver a wide range of biomolecules. Recent advances prove that ferritin can serve as an efficient 8 nm diameter carrier for various bioinorganic nutrients, such as minerals, bioactive polyphenols, and enzymes. This review offers a comprehensive summary of ferritin's structural features from different sources and emphasizes its functions in iron supplementation, calcium delivery, single- and coencapsulation of polyphenols, and enzyme package. Additionally, the influence of innovative food processing technologies, including manothermosonication, pulsed electric field, and atmospheric cold plasma, on the structure and function of ferritin are examined. Furthermore, the limitations and prospects of ferritin in food and nutritional applications are discussed. The exploration of ferritin as a multifunctional protein with the capacity to load various biomolecules is crucial to fully harnessing its potential in food applications.
Collapse
Affiliation(s)
- Jiangnan Hu
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Xinmei Sha
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Yue Li
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Jincan Wu
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Junrui Ma
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Yuyu Zhang
- Key Laboratory of Geriatric Nutrition and Health (Beijing Technology and Business University), Ministry of Education, Beijing 100048, China
| | - Rui Yang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science & Technology, Tianjin 300457, China
| |
Collapse
|
13
|
Yang YC, Zhu Y, Sun SJ, Zhao CJ, Bai Y, Wang J, Ma LT. ROS regulation in gliomas: implications for treatment strategies. Front Immunol 2023; 14:1259797. [PMID: 38130720 PMCID: PMC10733468 DOI: 10.3389/fimmu.2023.1259797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 10/30/2023] [Indexed: 12/23/2023] Open
Abstract
Gliomas are one of the most common primary malignant tumours of the central nervous system (CNS), of which glioblastomas (GBMs) are the most common and destructive type. The glioma tumour microenvironment (TME) has unique characteristics, such as hypoxia, the blood-brain barrier (BBB), reactive oxygen species (ROS) and tumour neovascularization. Therefore, the traditional treatment effect is limited. As cellular oxidative metabolites, ROS not only promote the occurrence and development of gliomas but also affect immune cells in the immune microenvironment. In contrast, either too high or too low ROS levels are detrimental to the survival of glioma cells, which indicates the threshold of ROS. Therefore, an in-depth understanding of the mechanisms of ROS production and scavenging, the threshold of ROS, and the role of ROS in the glioma TME can provide new methods and strategies for glioma treatment. Current methods to increase ROS include photodynamic therapy (PDT), sonodynamic therapy (SDT), and chemodynamic therapy (CDT), etc., and methods to eliminate ROS include the ingestion of antioxidants. Increasing/scavenging ROS is potentially applicable treatment, and further studies will help to provide more effective strategies for glioma treatment.
Collapse
Affiliation(s)
- Yu-Chen Yang
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi’an, China
| | - Yu Zhu
- College of Health, Dongguan Polytechnic, Dongguan, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Si-Jia Sun
- Department of Postgraduate Work, Xi’an Medical University, Xi’an, China
| | - Can-Jun Zhao
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi’an, China
| | - Yang Bai
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Jin Wang
- Department of Radiation Protection Medicine, Faculty of Preventive Medicine, Air Force Medical University (Fourth Military Medical University), Xi’an, China
- Shaanxi Key Laboratory of Free Radical and Medicine, Xi’an, China
| | - Li-Tian Ma
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi’an, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Tumor Diagnosis and Treatment in Shaanxi Province, Xi’an, China
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi’an, China
| |
Collapse
|
14
|
Zhao T, Cai Y, Jiang Y, He X, Wei Y, Yu Y, Tian X. Vaccine adjuvants: mechanisms and platforms. Signal Transduct Target Ther 2023; 8:283. [PMID: 37468460 PMCID: PMC10356842 DOI: 10.1038/s41392-023-01557-7] [Citation(s) in RCA: 105] [Impact Index Per Article: 105.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/19/2023] [Accepted: 06/27/2023] [Indexed: 07/21/2023] Open
Abstract
Adjuvants are indispensable components of vaccines. Despite being widely used in vaccines, their action mechanisms are not yet clear. With a greater understanding of the mechanisms by which the innate immune response controls the antigen-specific response, the adjuvants' action mechanisms are beginning to be elucidated. Adjuvants can be categorized as immunostimulants and delivery systems. Immunostimulants are danger signal molecules that lead to the maturation and activation of antigen-presenting cells (APCs) by targeting Toll-like receptors (TLRs) and other pattern recognition receptors (PRRs) to promote the production of antigen signals and co-stimulatory signals, which in turn enhance the adaptive immune responses. On the other hand, delivery systems are carrier materials that facilitate antigen presentation by prolonging the bioavailability of the loaded antigens, as well as targeting antigens to lymph nodes or APCs. The adjuvants' action mechanisms are systematically summarized at the beginning of this review. This is followed by an introduction of the mechanisms, properties, and progress of classical vaccine adjuvants. Furthermore, since some of the adjuvants under investigation exhibit greater immune activation potency than classical adjuvants, which could compensate for the deficiencies of classical adjuvants, a summary of the adjuvant platforms under investigation is subsequently presented. Notably, we highlight the different action mechanisms and immunological properties of these adjuvant platforms, which will provide a wide range of options for the rational design of different vaccines. On this basis, this review points out the development prospects of vaccine adjuvants and the problems that should be paid attention to in the future.
Collapse
Affiliation(s)
- Tingmei Zhao
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yulong Cai
- Division of Biliary Tract Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yujie Jiang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
| | - Yifan Yu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaohe Tian
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, 610041, Sichuan, People's Republic of China.
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
15
|
João J, Prazeres DMF. Manufacturing of non-viral protein nanocages for biotechnological and biomedical applications. Front Bioeng Biotechnol 2023; 11:1200729. [PMID: 37520292 PMCID: PMC10374429 DOI: 10.3389/fbioe.2023.1200729] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/05/2023] [Indexed: 08/01/2023] Open
Abstract
Protein nanocages are highly ordered nanometer scale architectures, which are typically formed by homo- or hetero-self-assembly of multiple monomers into symmetric structures of different size and shape. The intrinsic characteristics of protein nanocages make them very attractive and promising as a biological nanomaterial. These include, among others, a high surface/volume ratio, multi-functionality, ease to modify or manipulate genetically or chemically, high stability, mono-dispersity, and biocompatibility. Since the beginning of the investigation into protein nanocages, several applications were conceived in a variety of areas such as drug delivery, vaccine development, bioimaging, biomineralization, nanomaterial synthesis and biocatalysis. The ability to generate large amounts of pure and well-folded protein assemblies is one of the keys to transform nanocages into clinically valuable products and move biomedical applications forward. This calls for the development of more efficient biomanufacturing processes and for the setting up of analytical techniques adequate for the quality control and characterization of the biological function and structure of nanocages. This review concisely covers and overviews the progress made since the emergence of protein nanocages as a new, next-generation class of biologics. A brief outline of non-viral protein nanocages is followed by a presentation of their main applications in the areas of bioengineering, biotechnology, and biomedicine. Afterwards, we focus on a description of the current processes used in the manufacturing of protein nanocages with particular emphasis on the most relevant aspects of production and purification. The state-of-the-art on current characterization techniques is then described and future alternative or complementary approaches in development are also discussed. Finally, a critical analysis of the limitations and drawbacks of the current manufacturing strategies is presented, alongside with the identification of the major challenges and bottlenecks.
Collapse
Affiliation(s)
- Jorge João
- iBB–Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB–Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| | - Duarte Miguel F. Prazeres
- iBB–Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- Associate Laboratory i4HB–Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
16
|
Roy M, Roy A, Rustagi S, Pandey N. An Overview of Nanomaterial Applications in Pharmacology. BIOMED RESEARCH INTERNATIONAL 2023; 2023:4838043. [PMID: 37388336 PMCID: PMC10307208 DOI: 10.1155/2023/4838043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/06/2023] [Accepted: 06/13/2023] [Indexed: 07/01/2023]
Abstract
Nanotechnology has become one of the most extensive fields of research. Nanoparticles (NPs) form the base for nanotechnology. Recently, nanomaterials (NMs) are widely used due to flexible chemical, biological, and physical characteristics with improved efficacy in comparison to bulk counterparts. The significance of each class of NMs is enhanced by identifying their properties. Day by day, there is an emergence of various applications of NMs, but the toxic effects associated with them cannot be avoided. NMs demonstrate therapeutic abilities by enhancing the drug delivery system, diagnosis, and therapeutic effects of numerous agents, but determining the benefits of NMs over other clinical applications (disease-specific) or substances is an ongoing investigation. This review is aimed at defining NMs and NPs and their types, synthesis, and pharmaceutical, biomedical, and clinical applications.
Collapse
Affiliation(s)
- Madhura Roy
- Centre for Translational and Clinical Research, School of Chemical and Life Sciences, Jamia Hamdard, India
| | - Arpita Roy
- Department of Biotechnology, Sharda School of Engineering & Technology, Sharda University, Greater Noida, India
| | - Sarvesh Rustagi
- School of Applied and Life sciences, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Neha Pandey
- Department of Biotechnology, Graphic Era Deemed to Be University, Dehradun, Uttarakhand, India
| |
Collapse
|
17
|
Chen F, Huang Y, Huang Z, Jiang T, Yang Z, Zeng J, Jin A, Zuo H, Huang CZ, Mao C. DNA-scaffolded multivalent vaccine against SARS-CoV-2. Acta Biomater 2023; 164:387-396. [PMID: 37088158 PMCID: PMC10122553 DOI: 10.1016/j.actbio.2023.04.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 03/12/2023] [Accepted: 04/13/2023] [Indexed: 04/25/2023]
Abstract
Short peptides are poor immunogens. One way to increase their immune responses is by arraying immunogens in multivalency. Simple and efficient scaffolds for spatial controlling the inter-antigen distance and enhancing immune activation are required. Here, we report a molecular vaccine design principle that maximally drives potent SARS-CoV-2 RBD subunit vaccine on DNA duplex to induce robust and efficacious immune responses in vivo. We expect that the DNA-peptide epitope platform represents a facile and generalizable strategy to enhance the immune response. STATEMENT OF SIGNIFICANCE: DNA scaffolds offer a biocompatible and convenient platform for arraying immunogens in multivalency antigenic peptides, and spatially control the inter-antigen distance. This can effectively enhance immune response. Peptide (instead of entire protein) vaccines are highly attractive. However, short peptides are poor immunogens. Our DNA scaffolded multivalent peptide immunogen system induced robust and efficacious immune response in vivo as demonstrated by the antigenic peptide against SAR-CoV-2. The present strategy could be readily generalized and adapted to prepare multivalent vaccines against other viruses or disease. Particularly, the different antigens could be integrated into one single vaccine and lead to super-vaccines that can protect the host from multiple different viruses or multiple variants of the same virus.
Collapse
Affiliation(s)
- Fangfang Chen
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Yuhan Huang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhengyu Huang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Tingting Jiang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zailin Yang
- Department of Hematology-Oncology, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Jie Zeng
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Aishun Jin
- Department of Immunology, College of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Hua Zuo
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Cheng Zhi Huang
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China.
| | - Chengde Mao
- Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China; Department of Chemistry, Purdue University, West Lafayette 47907, IN, USA.
| |
Collapse
|
18
|
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: 8] [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
|
19
|
Zhu Y, Zhu Y, Cao T, Liu X, Liu X, Yan Y, Shi Y, Wang JC. Ferritin-based nanomedicine for disease treatment. MEDICAL REVIEW (2021) 2023; 3:49-74. [PMID: 37724111 PMCID: PMC10471093 DOI: 10.1515/mr-2023-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Accepted: 02/01/2023] [Indexed: 09/20/2023]
Abstract
Ferritin is an endogenous protein which is self-assembled by 24 subunits into a highly uniform nanocage structure. Due to the drug-encapsulating ability in the hollow inner cavity and abundant modification sites on the outer surface, ferritin nanocage has been demonstrated great potential to become a multi-functional nanomedicine platform. Its good biocompatibility, low toxicity and immunogenicity, intrinsic tumor-targeting ability, high stability, low cost and massive production, together make ferritin nanocage stand out from other nanocarriers. In this review, we summarized ferritin-based nanomedicine in field of disease diagnosis, treatment and prevention. The different types of drugs to be loaded in ferritin, as well as drug-loading methods were classified. The strategies for site-specific and non-specific functional modification of ferritin were investigated, then the application of ferritin for disease imaging, drug delivery and vaccine development were discussed. Finally, the challenges restricting the clinical translation of ferritin-based nanomedicines were analyzed.
Collapse
Affiliation(s)
- Yuanjun Zhu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yuefeng Zhu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Tianmiao Cao
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiaoyu Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiaoyan Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yi Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Yujie Shi
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jian-Cheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
- Laboratory of Innovative Formulations and Pharmaceutical Excipients, Ningbo Institute of Marine Medicine, Peking University, Ningbo, Zhejiang Province, China
| |
Collapse
|
20
|
Yang R, Ma J, Hu J, Sun H, Han Y, Meng D, Wang Z, Cheng L. Formation of ferritin-agaro oligosaccharide-epigallocatechin gallate nanoparticle induced by CHAPS and partitioned by the ferritin shell with enhanced delivery efficiency. Food Hydrocoll 2022. [DOI: 10.1016/j.foodhyd.2022.108396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
21
|
Mamuti M, Chen W, Jiang X. Nanotechnology‐Assisted Immunoengineering for Cancer Vaccines. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
- Muhetaerjiang Mamuti
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science and Engineering College of Chemistry and Chemical Engineering Jiangsu Key Laboratory for Nanotechnology Nanjing University Nanjing China
| | - Weizhi Chen
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science and Engineering College of Chemistry and Chemical Engineering Jiangsu Key Laboratory for Nanotechnology Nanjing University Nanjing China
| | - Xiqun Jiang
- MOE Key Laboratory of High Performance Polymer Materials and Technology, and Department of Polymer Science and Engineering College of Chemistry and Chemical Engineering Jiangsu Key Laboratory for Nanotechnology Nanjing University Nanjing China
| |
Collapse
|
22
|
Files MA, Kristjansson KM, Rudra JS, Endsley JJ. Nanomaterials-based vaccines to target intracellular bacterial pathogens. Front Microbiol 2022; 13:1040105. [PMID: 36466676 PMCID: PMC9715960 DOI: 10.3389/fmicb.2022.1040105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 11/02/2022] [Indexed: 11/21/2022] Open
Abstract
Development of novel immunization approaches to combat a growing list of emerging and ancient infectious agents is a global health priority. Intensive efforts over the last several decades have identified alternative approaches to improve upon traditional vaccines that are based on live, attenuated agents, or formulations of inactivated agents with adjuvants. Rapid advances in RNA-based and other delivery systems for immunization have recently revolutionized the potential to protect populations from viral pathogens, such as SARS-CoV-2. Similar efforts to combat bacterial pathogens, especially species with an intracellular niche, have lagged significantly. In the past decade, advances in nanotechnology have yielded a variety of new antigen/adjuvant carrier systems for use in vaccine development against infectious viruses and bacteria. The tunable properties of nanomaterial-based vaccines allow for balancing immunogenicity and safety which is a key hurdle in traditional antigen and adjuvant formulations. In this review, we discuss several novel nanoparticle-based vaccine platforms that show promise for use against intracellular bacteria as demonstrated by the feasibility of construction, enhanced antigen presentation, induction of cell mediated and humoral immune responses, and improved survival outcomes in in vivo models.
Collapse
Affiliation(s)
- Megan A. Files
- Department of Microbiology and Immunology, Galveston, TX, United States
- Institute of Translational Science, University of Texas Medical Branch, Galveston, TX, United States
- Department of Medicine, School of Medicine, Seattle, WA, United States
| | - Kadin M. Kristjansson
- Department of Chemistry, Smith College, Northampton MA, United States
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, United States
| | - Jai S. Rudra
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, MO, United States
| | - Janice J. Endsley
- Department of Microbiology and Immunology, Galveston, TX, United States
| |
Collapse
|
23
|
Aljabali AAA, Rezigue M, Alsharedeh RH, Obeid MA, Mishra V, Serrano-Aroca Á, Tambuwala MM. Protein-Based Drug Delivery Nanomedicine Platforms: Recent Developments. Pharm Nanotechnol 2022; 10:257-267. [PMID: 35980061 DOI: 10.2174/2211738510666220817120307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/08/2022] [Accepted: 04/26/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND Naturally occurring protein cages, both viral and non-viral assemblies, have been developed for various pharmaceutical applications. Protein cages are ideal platforms as they are compatible, biodegradable, bioavailable, and amenable to chemical and genetic modification to impart new functionalities for selective targeting or tracking of proteins. The ferritin/ apoferritin protein cage, plant-derived viral capsids, the small Heat shock protein, albumin, soy and whey protein, collagen, and gelatin have all been exploited and characterized as drugdelivery vehicles. Protein cages come in many shapes and types with unique features such as unmatched uniformity, size, and conjugations. OBJECTIVES The recent strategic development of drug delivery will be covered in this review, emphasizing polymer-based, specifically protein-based, drug delivery nanomedicine platforms. The potential and drawbacks of each kind of protein-based drug-delivery system will also be highlighted. METHODS Research examining the usability of nanomaterials in the pharmaceutical and medical sectors were identified by employing bibliographic databases and web search engines. RESULTS Rings, tubes, and cages are unique protein structures that occur in the biological environment and might serve as building blocks for nanomachines. Furthermore, numerous virions can undergo reversible structural conformational changes that open or close gated pores, allowing customizable accessibility to their core and ideal delivery vehicles. CONCLUSION Protein cages' biocompatibility and their ability to be precisely engineered indicate they have significant potential in drug delivery and intracellular administration.
Collapse
Affiliation(s)
- Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid 21163 - P.O. BOX 566, Jordan
| | - Meriem Rezigue
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid 21163 - P.O. BOX 566, Jordan
| | - Rawan H Alsharedeh
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid 21163 - P.O. BOX 566, Jordan
| | - Mohammad A Obeid
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid 21163 - P.O. BOX 566, Jordan
| | - Vijay Mishra
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara 144411, India
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia, San Vicente Mártir, 46001 Valencia, Spain
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, England, UK
| |
Collapse
|
24
|
Sudarev VV, Dolotova SM, Bukhalovich SM, Bazhenov SV, Ryzhykau YL, Uversky VN, Bondarev NA, Osipov SD, Mikhailov AE, Kuklina DD, Murugova TN, Manukhov IV, Rogachev AV, Gordeliy VI, Gushchin IY, Kuklin AI, Vlasov AV. Ferritin self-assembly, structure, function, and biotechnological applications. Int J Biol Macromol 2022; 224:319-343. [DOI: 10.1016/j.ijbiomac.2022.10.126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Revised: 09/28/2022] [Accepted: 10/13/2022] [Indexed: 11/05/2022]
|
25
|
Expression and Evaluation of a Novel PPRV Nanoparticle Antigen Based on Ferritin Self-Assembling Technology. Pharmaceutics 2022; 14:pharmaceutics14091902. [PMID: 36145650 PMCID: PMC9500948 DOI: 10.3390/pharmaceutics14091902] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Peste des Petits Ruminants (PPR) is a highly pathogenic disease that is classified as a World Organization for Animal Health (OIE)-listed disease. PPRV mainly infects small ruminants such as goats and sheep. In view of the global and high pathogenicity of PPRV, in this study, we proposed a novel nanoparticle vaccine strategy based on ferritin (Fe) self-assembly technology. Using Helicobacter pylori (H. pylori) ferritin as an antigen delivery vector, a PPRV hemagglutinin (H) protein was fused with ferritin and then expressed and purified in both Escherichia coli (E. coli) and silkworm baculovirus expression systems. Subsequently, the nanoparticle antigens’ expression level, immunogenicity and protective immune response were evaluated. Our results showed that the PPRV hemagglutinin–ferritin (H-Fe) protein was self-assembled in silkworms, while it was difficult to observe the correctly folded nanoparticle in E. coli. Meanwhile, the expression level of the H-Fe protein was higher than that of the H protein alone. Furthermore, the immunogenicity and protective immune response of H-Fe nanoparticle antigens expressed by silkworms were improved compared with the H antigen alone. Particularly, the protective immune response of H-Fe antigens expressed in E. coli did not change, as opposed to the H antigen, which was probably due to the incomplete nanoparticle structure in E. coli. This study indicated that the use of ferritin nanoparticles as antigen delivery carriers could increase the expression of antigen proteins and improve the immunogenicity and immune effect of antigens.
Collapse
|
26
|
Kim SA, Kim S, Kim GB, Goo J, Kim N, Lee Y, Nam GH, Lim S, Kim T, Chang KH, Lee TG, Kim IS, Lee EJ. A Multivalent Vaccine Based on Ferritin Nanocage Elicits Potent Protective Immune Responses against SARS-CoV-2 Mutations. Int J Mol Sci 2022; 23:6123. [PMID: 35682801 PMCID: PMC9181758 DOI: 10.3390/ijms23116123] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/27/2022] [Accepted: 05/27/2022] [Indexed: 02/05/2023] Open
Abstract
The SARS-CoV-2 pandemic has created a global public crisis and heavily affected personal lives, healthcare systems, and global economies. Virus variants are continuously emerging, and, thus, the pandemic has been ongoing for over two years. Vaccines were rapidly developed based on the original SARS-CoV-2 (Wuhan-Hu-1) to build immunity against the coronavirus disease. However, they had a very low effect on the virus' variants due to their low cross-reactivity. In this study, a multivalent SARS-CoV-2 vaccine was developed using ferritin nanocages, which display the spike protein from the Wuhan-Hu-1, B.1.351, or B.1.429 SARS-CoV-2 on their surfaces. We show that the mixture of three SARS-CoV-2 spike-protein-displaying nanocages elicits CD4+ and CD8+ T cells and B-cell immunity successfully in vivo. Furthermore, they generate a more consistent antibody response against the B.1.351 and B.1.429 variants than a monovalent vaccine. This leads us to believe that the proposed ferritin-nanocage-based multivalent vaccine platform will provide strong protection against emerging SARS-CoV-2 variants of concern (VOCs).
Collapse
Affiliation(s)
- Seong A. Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea;
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02456, Korea;
| | - Seohyun Kim
- Department of Research and Development, ShiftBio, Seoul 02751, Korea; (S.K.); (G.B.K.); (G.-H.N.)
| | - Gi Beom Kim
- Department of Research and Development, ShiftBio, Seoul 02751, Korea; (S.K.); (G.B.K.); (G.-H.N.)
| | - Jiyoung Goo
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02456, Korea;
- KHU-KIST Department of Converging Science and Technology, Kyunghee University, Seoul 02447, Korea
| | - Nayeon Kim
- Department of Chemical Engineering, Kyungpook National University, Daegu 41566, Korea; (N.K.); (Y.L.)
| | - Yeram Lee
- Department of Chemical Engineering, Kyungpook National University, Daegu 41566, Korea; (N.K.); (Y.L.)
| | - Gi-Hoon Nam
- Department of Research and Development, ShiftBio, Seoul 02751, Korea; (S.K.); (G.B.K.); (G.-H.N.)
| | - Seungho Lim
- R&D Department Drug Development Division, LabGenomics Corporation, Gyeonggi-do 13488, Korea; (S.L.); (T.K.); (K.H.C.); (T.G.L.)
| | - Taeerk Kim
- R&D Department Drug Development Division, LabGenomics Corporation, Gyeonggi-do 13488, Korea; (S.L.); (T.K.); (K.H.C.); (T.G.L.)
| | - Ki Hwan Chang
- R&D Department Drug Development Division, LabGenomics Corporation, Gyeonggi-do 13488, Korea; (S.L.); (T.K.); (K.H.C.); (T.G.L.)
| | - Tae Gyu Lee
- R&D Department Drug Development Division, LabGenomics Corporation, Gyeonggi-do 13488, Korea; (S.L.); (T.K.); (K.H.C.); (T.G.L.)
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea;
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul 02456, Korea;
- Department of Research and Development, ShiftBio, Seoul 02751, Korea; (S.K.); (G.B.K.); (G.-H.N.)
| | - Eun Jung Lee
- Department of Chemical Engineering, Kyungpook National University, Daegu 41566, Korea; (N.K.); (Y.L.)
| |
Collapse
|
27
|
Xu X, Tian K, Lou X, Du Y. Potential of Ferritin-Based Platforms for Tumor Immunotherapy. Molecules 2022; 27:2716. [PMID: 35566065 PMCID: PMC9104857 DOI: 10.3390/molecules27092716] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/14/2022] [Accepted: 04/20/2022] [Indexed: 02/04/2023] Open
Abstract
Ferritin is an iron storage protein that plays a key role in iron homeostasis and cellular antioxidant activity. Ferritin has many advantages as a tumor immunotherapy platform, including a small particle size that allows for penetration into tumor-draining lymph nodes or tumor tissue, a unique structure consisting of 24 self-assembled subunits, cavities that can encapsulate drugs, natural targeting functions, and a modifiable outer surface. In this review, we summarize related research applying ferritin as a tumor immune vaccine or a nanocarrier for immunomodulator drugs based on different targeting mechanisms (including dendritic cells, tumor-associated macrophages, tumor-associated fibroblasts, and tumor cells). In addition, a ferritin-based tumor vaccine expected to protect against a wide range of coronaviruses by targeting multiple variants of SARS-CoV-2 has entered phase I clinical trials, and its efficacy is described in this review. Although ferritin is already on the road to transformation, there are still many difficulties to overcome. Therefore, three barriers (drug loading, modification sites, and animal models) are also discussed in this paper. Notwithstanding, the ferritin-based nanoplatform has great potential for tumor immunotherapy, with greater possibility of clinical transformation.
Collapse
Affiliation(s)
- Xiaoling Xu
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (X.X.); (K.T.)
| | - Kewei Tian
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou 310015, China; (X.X.); (K.T.)
| | - Xuefang Lou
- School of Medicine, Zhejiang University City College, Hangzhou 310015, China
| | - Yongzhong Du
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
28
|
Zhao X, Yang J, Zhang J, Wang X, Chen L, Zhang C, Shen Z. Inhibitory Effect of Aptamer-Carbon Dot Nanomaterial-siRNA Complex on the Metastasis of Hepatocellular Carcinoma Cells by Interfering with FMRP. Eur J Pharm Biopharm 2022; 174:47-55. [PMID: 35364257 DOI: 10.1016/j.ejpb.2022.03.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 03/23/2022] [Accepted: 03/27/2022] [Indexed: 11/04/2022]
Abstract
Using small interfering RNA (siRNA) for the specific gene-silencing has been a novel therapeutic method for the treatment of incurable diseases such as malignancies. However, it remains a challenge whether siRNA can be safely and effectively delivered into target cells. Therefore, we synthesized fluorescent carbon dots (CDs) as a gene vector at the siRNA delivery system that induced efficient gene knockdown in vitro while binding aptamer AS1411 to resolve the difficulty in cell targeting. We found that CDs with adequate biocompatibility can improve the efficiency of cellular uptake of siRNA. CLSM and FCM results showed that CDs were mainly localized in the cytoplasm and emitted bright green fluorescence. In addition, the CD/siRNA delivery system mediated by the aptamer AS1411 effectively silenced the expression of Fragile X mental retardation protein (FMRP) and successfully inhibited the migration and invasive propensity of hepatocellular carcinoma (HCC) cells. In summary, we have synthesized a valuable siRNA delivery vector enabling not only bioimaging but also effective downregulation of gene expression, which is indicative of an efficient potential for gene delivery and therapy.
Collapse
Affiliation(s)
- Xiaoliang Zhao
- Research Center for Molecular Oncology and Functional Nucleic Acids, School of Laboratory Medicine, and School of Basic Medicine, Xinxiang Medical University, Xinxiang, 453003, China; these authors contributed equally to this work
| | - Jie Yang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China; Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China; these authors contributed equally to this work
| | - Jing Zhang
- Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China
| | - Xiangyun Wang
- Research Center for Molecular Oncology and Functional Nucleic Acids, School of Laboratory Medicine, and School of Basic Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Lulu Chen
- Research Center for Molecular Oncology and Functional Nucleic Acids, School of Laboratory Medicine, and School of Basic Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Chao Zhang
- Research Center for Molecular Oncology and Functional Nucleic Acids, School of Laboratory Medicine, and School of Basic Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Zhifa Shen
- Research Center for Molecular Oncology and Functional Nucleic Acids, School of Laboratory Medicine, and School of Basic Medicine, Xinxiang Medical University, Xinxiang, 453003, China; Key Laboratory of Laboratory Medicine, Ministry of Education of China, and Zhejiang Provincial Key Laboratory of Medical Genetics, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
29
|
Self-assembled BPIV3 nanoparticles can induce comprehensive immune responses and protection against BPIV3 challenge by inducing dendritic cell maturation in mice. Vet Microbiol 2022; 268:109415. [DOI: 10.1016/j.vetmic.2022.109415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 01/24/2023]
|
30
|
Côté-Cyr M, Zottig X, Gauthier L, Archambault D, Bourgault S. Self-Assembly of Flagellin into Immunostimulatory Ring-like Nanostructures as an Antigen Delivery System. ACS Biomater Sci Eng 2022; 8:694-707. [PMID: 35080372 DOI: 10.1021/acsbiomaterials.1c01332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Proteinaceous nanoparticles represent attractive antigen carriers for vaccination as their size and repetitive antigen displays that mimic most viral particles enable efficient immune processing. However, these nanocarriers are often unable to stimulate efficiently the innate immune system, requiring coadministration with adjuvants to promote long-lasting protective immunity. The protein flagellin, which constitutes the primary constituent of the bacterial flagellum, has been widely evaluated as an antigen carrier due to its intrinsic adjuvant properties involving activation of the innate immune receptor Toll-like receptor 5 (TLR5). Although flagellin is known for its ability to self-assemble into micron-scale length nanotubes, few studies have evaluated the potential usage of flagellin-based nanostructures as immunostimulatory antigen carriers. In this study, we reported for the first time a strategy to guide the self-assembly of a flagellin protein from Bacillus subtilis, Hag, into lower aspect ratio nanoparticles by hindering non-covalent interactions responsible for its elongation into nanotubes. We observed that addition of an antigenic sequence derived from the influenza A virus (3M2e) at the C-terminus of this flagellin, as opposed to positioning the epitope into mid-sequence, precluded filament elongation and resulted in low aspect ratio ring-like nanostructures upon salting-out-induced self-assembly. These nanostructures displayed the antigen at their surface and shared morphological and structural characteristics with flagellin nanotubes, with a diameter of approximately 12 nm, and an α-helix-rich secondary structure. Flagellin ring-like nanostructures were efficiently internalized by antigen-presenting cells, and avidly activated the TLR5 in vitro as well as the innate and adaptive immune responses. Intranasal immunization of mice with these nanostructures resulted in the potentiation of the antigen-specific antibody response and protection against a lethal infection with the influenza A virus, illustrating the potential of these intrinsically immunostimulatory nanostructures as antigen carriers.
Collapse
Affiliation(s)
- Mélanie Côté-Cyr
- Chemistry Department, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec G1V 0A6, Canada.,Department of Biological Sciences, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
| | - Ximena Zottig
- Chemistry Department, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec G1V 0A6, Canada.,Department of Biological Sciences, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
| | - Laurie Gauthier
- Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec G1V 0A6, Canada.,Department of Biological Sciences, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
| | - Denis Archambault
- Department of Biological Sciences, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
| | - Steve Bourgault
- Chemistry Department, Université du Québec à Montréal, C.P. 8888, Succursale Centre-Ville, Montreal H3C 3P8, Canada.,Quebec Network for Research on Protein Function, Engineering and Applications (PROTEO), Québec G1V 0A6, Canada.,The Swine and Poultry Infectious Diseases Research Centre (CRIPA), Saint-Hyacinthe J2S 2M2, Canada
| |
Collapse
|
31
|
Li Y, Sun J, Li J, Liu K, Zhang H. Engineered protein nanodrug as an emerging therapeutic tool. NANO RESEARCH 2022; 15:5161-5172. [PMID: 35281219 PMCID: PMC8900963 DOI: 10.1007/s12274-022-4103-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/20/2021] [Accepted: 12/25/2021] [Indexed: 05/05/2023]
Abstract
Functional proteins are the most versatile macromolecules. They can be obtained by extraction from natural sources or by genetic engineering technologies. The outstanding selectivity, specificity, binding activity, and biocompatibility endow engineered proteins with outstanding performance for disease therapy. Nevertheless, their stability is dramatically impaired in blood circulation, hindering clinical translations. Thus, many strategies have been developed to improve the stability, efficacy, bioavailability, and productivity of therapeutic proteins for clinical applications. In this review, we summarize the recent progress in the fabrication and application of therapeutic proteins. We first introduce various strategies for improving therapeutic efficacy via bioengineering and nanoassembly. Furthermore, we highlight their diverse applications as growth factors, nanovaccines, antibody-based drugs, bioimaging molecules, and cytokine receptor antagonists. Finally, a summary and perspective for the future development of therapeutic proteins are presented.
Collapse
Affiliation(s)
- Yuanxin Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 China
- University of Science and Technology of China, Hefei, 230026 China
| | - Jing Sun
- Institute of Organic Chemistry, University of Ulm, Albert-Einstein-Allee 11, Ulm, 89081 Germany
| | - Jingjing Li
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 China
| | - Kai Liu
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 China
- University of Science and Technology of China, Hefei, 230026 China
- Department of Chemistry, Tsinghua University, Beijing, 100084 China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022 China
- University of Science and Technology of China, Hefei, 230026 China
- Department of Chemistry, Tsinghua University, Beijing, 100084 China
| |
Collapse
|
32
|
Mainini F, Bonizzi A, Sevieri M, Sitia L, Truffi M, Corsi F, Mazzucchelli S. Protein-Based Nanoparticles for the Imaging and Treatment of Solid Tumors: The Case of Ferritin Nanocages, a Narrative Review. Pharmaceutics 2021; 13:pharmaceutics13122000. [PMID: 34959283 PMCID: PMC8708614 DOI: 10.3390/pharmaceutics13122000] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022] Open
Abstract
Protein nanocages have been studied extensively, due to their unique architecture, exceptional biocompatibility and highly customization capabilities. In particular, ferritin nanocages (FNs) have been employed for the delivery of a vast array of molecules, ranging from chemotherapeutics to imaging agents, among others. One of the main favorable characteristics of FNs is their intrinsic targeting efficiency toward the Transferrin Receptor 1, which is overexpressed in many tumors. Furthermore, genetic manipulation can be employed to introduce novel variants that are able to improve the loading capacity, targeting capabilities and bio-availability of this versatile drug delivery system. In this review, we discuss the main characteristics of FN and the most recent applications of this promising nanotechnology in the field of oncology with a particular emphasis on the imaging and treatment of solid tumors.
Collapse
Affiliation(s)
- Francesco Mainini
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
| | - Arianna Bonizzi
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
| | - Marta Sevieri
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
| | - Leopoldo Sitia
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
| | - Marta Truffi
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy;
| | - Fabio Corsi
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
- Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy;
- Correspondence: (F.C.); (S.M.)
| | - Serena Mazzucchelli
- Dipartimento di Scienze Biomediche e Cliniche “L. Sacco”, Università di Milano, 20157 Milano, Italy; (F.M.); (A.B.); (M.S.); (L.S.)
- Correspondence: (F.C.); (S.M.)
| |
Collapse
|
33
|
Rodrigues MQ, Alves PM, Roldão A. Functionalizing Ferritin Nanoparticles for Vaccine Development. Pharmaceutics 2021; 13:1621. [PMID: 34683914 PMCID: PMC8540537 DOI: 10.3390/pharmaceutics13101621] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/24/2022] Open
Abstract
In the last decade, the interest in ferritin-based vaccines has been increasing due to their safety and immunogenicity. Candidates against a wide range of pathogens are now on Phase I clinical trials namely for influenza, Epstein-Barr, and SARS-CoV-2 viruses. Manufacturing challenges related to particle heterogeneity, improper folding of fused antigens, and antigen interference with intersubunit interactions still need to be overcome. In addition, protocols need to be standardized so that the production bioprocess becomes reproducible, allowing ferritin-based therapeutics to become readily available. In this review, the building blocks that enable the formulation of ferritin-based vaccines at an experimental stage, including design, production, and purification are presented. Novel bioengineering strategies of functionalizing ferritin nanoparticles based on modular assembly, allowing the challenges associated with genetic fusion to be circumvented, are discussed. Distinct up/down-stream approaches to produce ferritin-based vaccines and their impact on production yield and vaccine efficacy are compared. Finally, ferritin nanoparticles currently used in vaccine development and clinical trials are summarized.
Collapse
Affiliation(s)
- Margarida Q. Rodrigues
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (M.Q.R.); (P.M.A.)
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - Paula M. Alves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (M.Q.R.); (P.M.A.)
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| | - António Roldão
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal; (M.Q.R.); (P.M.A.)
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República, 2780-157 Oeiras, Portugal
| |
Collapse
|
34
|
Zhang Z, Qiu C, Li X, McClements DJ, Jiao A, Wang J, Jin Z. Advances in research on interactions between polyphenols and biology-based nano-delivery systems and their applications in improving the bioavailability of polyphenols. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
35
|
Choi Y, Nam GH, Kim GB, Kim S, Kim YK, Kim SA, Kim HJ, Lee EJ, Kim IS. Nanocages displaying SIRP gamma clusters combined with prophagocytic stimulus of phagocytes potentiate anti-tumor immunity. Cancer Gene Ther 2021; 28:960-970. [PMID: 34349240 DOI: 10.1038/s41417-021-00372-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 06/10/2021] [Accepted: 07/20/2021] [Indexed: 02/07/2023]
Abstract
Antigen-presenting cells (APCs), including macrophages and dendritic cells (DCs), play a crucial role in bridging innate and adaptive immunity; thereby, innate immune checkpoint blockade-based therapy is an attractive approach for the induction of sustainable tumor-specific immunity. The interaction between the cluster of differentiation 47 (CD47) on tumor and signal-regulatory protein alpha (SIRPα) on phagocytic cells inhibits the phagocytic function of APCs, acting as a "don't eat me" signal. Accordingly, CD47 blockade is known to increase tumor cell phagocytosis, eliciting tumor-specific CD8+ T-cell immunity. Here, we introduced a nature-derived nanocage to deliver SIRPγ for blocking of antiphagocytic signaling through binding to CD47 and combined it with prophagocytic stimuli using a metabolic reprogramming reagent for APCs (CpG-oligodeoxynucleotides). Upon delivering the clustered SIRPγ variant, the nanocage showed enhanced CD47 binding profiles on tumor cells, thereby promoting active engulfment by phagocytes. Moreover, combination with CpG potentiated the prophagocytic ability, leading to the establishment of antitumorigenic surroundings. This combination treatment could competently inhibit tumor growth by invigorating APCs and CD8+ T-cells in TMEs in B16F10 orthotopic tumor models, known to be resistant to CD47-targeting therapeutics. Collectively, enhanced delivery of an innate immune checkpoint antagonist with metabolic modulation stimuli of immune cells could be a promising strategy for arousing immune responses against cancer.
Collapse
Affiliation(s)
- Yoonjeong Choi
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Gi-Hoon Nam
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea.,Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Gi Beom Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Seohyun Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Yoon Kyoung Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Seong A Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Ha-Jeong Kim
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Eun Jung Lee
- Department of Chemical Engineering, School of Applied Chemical Engineering, Kyungpook National University, Daegu, Republic of Korea.
| | - In-San Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea. .,Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Van de Steen A, Khalife R, Colant N, Mustafa Khan H, Deveikis M, Charalambous S, Robinson CM, Dabas R, Esteban Serna S, Catana DA, Pildish K, Kalinovskiy V, Gustafsson K, Frank S. Bioengineering bacterial encapsulin nanocompartments as targeted drug delivery system. Synth Syst Biotechnol 2021; 6:231-241. [PMID: 34541345 PMCID: PMC8435816 DOI: 10.1016/j.synbio.2021.09.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/25/2021] [Accepted: 09/01/2021] [Indexed: 11/21/2022] Open
Abstract
The development of Drug Delivery Systems (DDS) has led to increasingly efficient therapies for the treatment and detection of various diseases. DDS use a range of nanoscale delivery platforms produced from polymeric of inorganic materials, such as micelles, and metal and polymeric nanoparticles, but their variant chemical composition make alterations to their size, shape, or structures inherently complex. Genetically encoded protein nanocages are highly promising DDS candidates because of their modular composition, ease of recombinant production in a range of hosts, control over assembly and loading of cargo molecules and biodegradability. One example of naturally occurring nanocompartments are encapsulins, recently discovered bacterial organelles that have been shown to be reprogrammable as nanobioreactors and vaccine candidates. Here we report the design and application of a targeted DDS platform based on the Thermotoga maritima encapsulin reprogrammed to display an antibody mimic protein called Designed Ankyrin repeat protein (DARPin) on the outer surface and to encapsulate a cytotoxic payload. The DARPin9.29 chosen in this study specifically binds to human epidermal growth factor receptor 2 (HER2) on breast cancer cells, as demonstrated in an in vitro cell culture model. The encapsulin-based DDS is assembled in one step in vivo by co-expressing the encapsulin-DARPin9.29 fusion protein with an engineered flavin-binding protein mini-singlet oxygen generator (MiniSOG), from a single plasmid in Escherichia coli. Purified encapsulin-DARPin_miniSOG nanocompartments bind specifically to HER2 positive breast cancer cells and trigger apoptosis, indicating that the system is functional and specific. The DDS is modular and has the potential to form the basis of a multi-receptor targeted system by utilising the DARPin screening libraries, allowing use of new DARPins of known specificities, and through the proven flexibility of the encapsulin cargo loading mechanism, allowing selection of cargo proteins of choice.
Collapse
Key Words
- Annexin V-FITC, Annexin V-Fluorescein IsoThiocyanate Conjugate
- Cytotoxic protein
- DARPin
- DARPin9.29, Designed Ankyrin Repeat Protein 9.29
- DDS, Drug Delivery System
- Drug delivery system
- EPR, Enhanced Permeability and Retention effect
- Encapsulin
- HER2, Human Epidermal growth factor Receptor 2
- His6, Hexahistidine
- MSCs, Mesenchymal Stem Cells
- NPs, NanoParticles
- SK-BR-3, Sloan-Kettering Breast cancer cell line/HER2-overexpressing human breast cancer cell line
- STII, StrepII-tag, an eight-residue peptide sequence (Trp-Ser-His-Pro-Gln-Phe-Glu-Lys) with intrinsic affinity toward streptavidin that can be fused to recombinant protein in various fashions
- T. maritima, Thermotoga maritima
- VLPs, Virus-Like Particle
- iGEM, international Genetically Engineered Machine
- iLOV, improved Light, Oxygen or Voltage-sensing flavoprotein
- mScarlet, a bright monomeric red fluorescent protein
- miniSOG, mini-Singlet Oxygen Generator
- rTurboGFP, recombinant Turbo Green Fluorescent Protein
Collapse
Affiliation(s)
| | - Rana Khalife
- Department of Biochemical Engineering, University College London, UK
| | - Noelle Colant
- Department of Biochemical Engineering, University College London, UK
| | | | - Matas Deveikis
- Department of Biochemical Engineering, University College London, UK
- UCL iGEM Student Team 2019, UK
| | - Saverio Charalambous
- Department of Biochemical Engineering, University College London, UK
- UCL iGEM Student Team 2019, UK
| | - Clare M. Robinson
- Natural Sciences, University College London, UK
- UCL iGEM Student Team 2019, UK
| | - Rupali Dabas
- Natural Sciences, University College London, UK
- UCL iGEM Student Team 2019, UK
| | - Sofia Esteban Serna
- Division of Biosciences, University College London, UK
- UCL iGEM Student Team 2019, UK
| | - Diana A. Catana
- Division of Biosciences, University College London, UK
- UCL iGEM Student Team 2019, UK
| | - Konstantin Pildish
- Division of Biosciences, University College London, UK
- UCL iGEM Student Team 2019, UK
| | - Vladimir Kalinovskiy
- Division of Biosciences, University College London, UK
- UCL iGEM Student Team 2019, UK
| | - Kenth Gustafsson
- Department of Biochemical Engineering, University College London, UK
| | - Stefanie Frank
- Department of Biochemical Engineering, University College London, UK
| |
Collapse
|
37
|
Liu ZH, Xu HL, Han GW, Tao LN, Lu Y, Zheng SY, Fang WH, He F. Self-Assembling Nanovaccine Enhances Protective Efficacy Against CSFV in Pigs. Front Immunol 2021; 12:689187. [PMID: 34367147 PMCID: PMC8334734 DOI: 10.3389/fimmu.2021.689187] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/01/2021] [Indexed: 01/01/2023] Open
Abstract
Classical swine fever virus (CSFV) is a highly contagious pathogen, which pose continuous threat to the swine industry. Though most attenuated vaccines are effective, they fail to serologically distinguish between infected and vaccinated animals, hindering CSFV eradication. Beneficially, nanoparticles (NPs)-based vaccines resemble natural viruses in size and antigen structure, and offer an alternative tool to circumvent these limitations. Using self-assembling NPs as multimerization platforms provides a safe and immunogenic tool against infectious diseases. This study presented a novel strategy to display CSFV E2 glycoprotein on the surface of genetically engineered self-assembling NPs. Eukaryotic E2-fused protein (SP-E2-mi3) could self-assemble into uniform NPs as indicated in transmission electron microscope (TEM) and dynamic light scattering (DLS). SP-E2-mi3 NPs showed high stability at room temperature. This NP-based immunization resulted in enhanced antigen uptake and up-regulated production of immunostimulatory cytokines in antigen presenting cells (APCs). Moreover, the protective efficacy of SP-E2-mi3 NPs was evaluated in pigs. SP-E2-mi3 NPs significantly improved both humoral and cellular immunity, especially as indicated by the elevated CSFV-specific IFN-γ cellular immunity and >10-fold neutralizing antibodies as compared to monomeric E2. These observations were consistent to in vivo protection against CSFV lethal virus challenge in prime-boost immunization schedule. Further results revealed single dose of 10 μg of SP-E2-mi3 NPs provided considerable clinical protection against lethal virus challenge. In conclusion, these findings demonstrated that this NP-based technology has potential to enhance the potency of subunit vaccine, paving ways for nanovaccine development.
Collapse
Affiliation(s)
- Ze-Hui Liu
- Institute of Preventive Veterinary Sciences & College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Hui-Ling Xu
- Institute of Preventive Veterinary Sciences & College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Guang-Wei Han
- Institute of Preventive Veterinary Sciences & College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Li-Na Tao
- Institute of Preventive Veterinary Sciences & College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Ying Lu
- Institute of Preventive Veterinary Sciences & College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Su-Ya Zheng
- Institute of Preventive Veterinary Sciences & College of Animal Sciences, Zhejiang University, Hangzhou, China
| | - Wei-Huan Fang
- Institute of Preventive Veterinary Sciences & College of Animal Sciences, Zhejiang University, Hangzhou, China.,Department of Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, China
| | - Fang He
- Institute of Preventive Veterinary Sciences & College of Animal Sciences, Zhejiang University, Hangzhou, China.,Department of Veterinary Medicine, Zhejiang Provincial Key Laboratory of Preventive Veterinary Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
38
|
Qu Y, Zhang B, Wang Y, Yin S, Pederick JL, Bruning JB, Sun Y, Middelberg A, Bi J. Immunogenicity study of engineered ferritins with C- and N-terminus insertion of Epstein-Barr nuclear antigen 1 epitope. Vaccine 2021; 39:4830-4841. [PMID: 34284876 DOI: 10.1016/j.vaccine.2021.07.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 12/25/2022]
Abstract
Human ferritin heavy chain, an example of a protein nanoparticle, has recently been used as a vaccine delivery platform. Human ferritin has advantages of uniform architecture, robust thermal and chemical stabilities, and good biocompatibility and biodegradation. There is however a lack of understanding about the relationship between insertion sites in ferritin (N-terminus and C-terminus) and the corresponding humoral and cell-mediated immune responses. To bridge this gap, we utilized an Epstein-Barr Nuclear Antigen 1 (EBNA1) epitope as a model to produce engineered ferritin-based vaccines E1F1 (N-terminus insertion) and F1E1 (C-terminus insertion) for the prevention of Epstein-Barr virus (EBV) infections. X-ray crystallography confirmed the relative positions of the N-terminus insertion and C-terminus insertion. For N-terminus insertion, the epitopes were located on the exterior surface of ferritin, while for C-terminus insertion, the epitopes were inside the ferritin cage. Based on the results of antigen-specific antibody titers from in-vivo tests, we found that there was no obvious difference on humoral immune responses between N-terminus and C-terminus insertion. We also evaluated splenocyte proliferation and memory lymphocyte T cell differentiation. Both results suggested C-terminus insertion produced a stronger proliferative response and cell-mediated immune response than N-terminus insertion. C-terminus insertion of EBNA1 epitope was also processed more efficiently by dendritic cells (DCs) than N-terminus insertion. This provides new insight into the relationship between the insertion site and immunogenicity of ferritin nanoparticle vaccines.
Collapse
Affiliation(s)
- Yiran Qu
- School of Chemical Engineering and Advanced Materials, Faculty of Engineering, Computer and Mathematical Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Bingyang Zhang
- School of Chemical Engineering and Advanced Materials, Faculty of Engineering, Computer and Mathematical Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Yingli Wang
- Shanxi University of Chinese Medicine, Shanxi, China
| | - Shuang Yin
- School of Chemical Engineering and Advanced Materials, Faculty of Engineering, Computer and Mathematical Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Jordan L Pederick
- Institute for Photonics and Advanced Sensing, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia; Department of Molecular and Cellular Biology, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - John B Bruning
- Institute for Photonics and Advanced Sensing, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia; Department of Molecular and Cellular Biology, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Yan Sun
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Anton Middelberg
- Division of Research and Innovation, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Jingxiu Bi
- School of Chemical Engineering and Advanced Materials, Faculty of Engineering, Computer and Mathematical Sciences, The University of Adelaide, Adelaide, South Australia 5005, Australia.
| |
Collapse
|
39
|
Choi H, Eom S, Kim HU, Bae Y, Jung HS, Kang S. Load and Display: Engineering Encapsulin as a Modular Nanoplatform for Protein-Cargo Encapsulation and Protein-Ligand Decoration Using Split Intein and SpyTag/SpyCatcher. Biomacromolecules 2021; 22:3028-3039. [PMID: 34142815 DOI: 10.1021/acs.biomac.1c00481] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Protein cage nanoparticles have a unique spherical hollow structure that provides a modifiable interior space and an exterior surface. For full application, it is desirable to utilize both the interior space and the exterior surface simultaneously with two different functionalities in a well-combined way. Here, we genetically engineered encapsulin protein cage nanoparticles (Encap) as modular nanoplatforms by introducing a split-C-intein (IntC) fragment and SpyTag into the interior and exterior surfaces, respectively. A complementary split-N-intein (IntN) was fused to various protein cargoes, such as NanoLuc luciferase (Nluc), enhanced green fluorescent protein (eGFP), and Nluc-miniSOG, individually, which led to their successful encapsulation into Encaps to form Cargo@Encap through split intein-mediated protein ligation during protein coexpression and cage assembly in bacteria. Conversely, the SpyCatcher protein was fused to various protein ligands, such as a glutathione binder (GST-SC), dimerizing ligands (FKBP12-SC and FRB-SC), and a cancer-targeting affibody (SC-EGFRAfb); subsequently, they were displayed on Cargo@Encaps through SpyTag/SpyCatcher ligation to form Cargo@Encap/Ligands in a mix-and-match manner. Nluc@Encap/glutathione-S-transferase (GST) was effectively immobilized on glutathione (GSH)-coated solid supports exhibiting repetitive and long-term usage of the encapsulated luciferases. We also established luciferase-embedded layer-by-layer (LbL) nanostructures by alternately depositing Nluc@Encap/FKBP12 and Nluc@Encap/FRB in the presence of rapamycin and applied enhanced green fluorescent protein (eGFP)@Encap/EGFRAfb as a target-specific fluorescent imaging probe to visualize specific cancer cells selectively. Modular functionalization of the interior space and the exterior surface of a protein cage nanoparticle may offer the opportunity to develop new protein-based nanostructured devices and nanomedical tools.
Collapse
Affiliation(s)
- Hyukjun Choi
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Soomin Eom
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Han-Ul Kim
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, 1, Kangwondaehak-gil, Chuncheon-si 24341, Gangwon-do, Korea
| | - Yoonji Bae
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| | - Hyun Suk Jung
- Department of Biochemistry, College of Natural Sciences, Kangwon National University, 1, Kangwondaehak-gil, Chuncheon-si 24341, Gangwon-do, Korea
| | - Sebyung Kang
- Department of Biological Sciences, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Korea
| |
Collapse
|
40
|
Lu L, Duong VT, Shalash AO, Skwarczynski M, Toth I. Chemical Conjugation Strategies for the Development of Protein-Based Subunit Nanovaccines. Vaccines (Basel) 2021; 9:563. [PMID: 34071482 PMCID: PMC8228360 DOI: 10.3390/vaccines9060563] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
The production of subunit nanovaccines relies heavily on the development of a vaccine delivery system that is safe and efficient at delivering antigens to the target site. Nanoparticles have been extensively investigated for vaccine delivery over the years, as they often possess self-adjuvanting properties. The conjugation of antigens to nanoparticles by covalent bonds ensures co-delivery of these components to the same subset of immune cells in order to trigger the desired immune responses. Herein, we review covalent conjugation strategies for grafting protein or peptide antigens onto other molecules or nanoparticles to obtain subunit nanovaccines. We also discuss the advantages of chemical conjugation in developing these vaccines.
Collapse
Affiliation(s)
| | | | | | - Mariusz Skwarczynski
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (L.L.); (V.T.D.); (A.O.S.)
| | - Istvan Toth
- School of Chemistry and Molecular Biosciences, The University of Queensland, St Lucia, QLD 4072, Australia; (L.L.); (V.T.D.); (A.O.S.)
| |
Collapse
|
41
|
Qu Y, Wang L, Yin S, Zhang B, Jiao Y, Sun Y, Middelberg A, Bi J. Stability of Engineered Ferritin Nanovaccines Investigated by Combined Molecular Simulation and Experiments. J Phys Chem B 2021; 125:3830-3842. [PMID: 33825471 DOI: 10.1021/acs.jpcb.1c00276] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Human ferritin is regarded as an attractive and promising vaccine platform because of its uniform structure, good plasticity, and desirable thermal and chemical stabilities. Besides, it is biocompatible and presumed safe when used as a vaccine carrier. However, there is a lack of knowledge of how different antigen insertion sites on the ferritin nanocage impact the resulting protein stability and performance. To address this question, we selected Epstein-Barr nuclear antigen 1 as a model epitope and fused it at the DNA level with different insertion sites, namely, the N- and C-termini of ferritin, to engineer proteins E1F1 and F1E1, respectively. Protein properties including hydrophobicity and thermal, pH, and chemical stability were investigated both by molecular dynamics (MD) simulation and by experiments. Both methods demonstrate that the insertion site plays an important role in protein properties. The C-terminus insertion (F1E1) leads to a less hydrophobic surface and more tolerance to the external influence of high temperature, pH, and high concentration of chemical denaturants compared to N-terminus insertion (E1F1). Simulated protein hydrophobicity and thermal stability by MD were in high accordance with experimental results. Thus, MD simulation can be used as a valuable tool to engineer nanovaccine candidates, cutting down costs by reducing the experimental effort and accelerating vaccine design.
Collapse
Affiliation(s)
- Yiran Qu
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Lijie Wang
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Shuang Yin
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Bingyang Zhang
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Yan Jiao
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Yan Sun
- Department of Biochemical Engineering and Key Laboratory of Systems Bioengineering of the Ministry of Education, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
| | - Anton Middelberg
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Jingxiu Bi
- School of Chemical Engineering and Advanced Materials, The University of Adelaide, Adelaide, South Australia 5005, Australia
| |
Collapse
|
42
|
The Versatile Manipulations of Self-Assembled Proteins in Vaccine Design. Int J Mol Sci 2021; 22:ijms22041934. [PMID: 33669238 PMCID: PMC7919822 DOI: 10.3390/ijms22041934] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/06/2021] [Accepted: 02/11/2021] [Indexed: 12/16/2022] Open
Abstract
Protein assemblies provide unique structural features which make them useful as carrier molecules in biomedical and chemical science. Protein assemblies can accommodate a variety of organic, inorganic and biological molecules such as small proteins and peptides and have been used in development of subunit vaccines via display parts of viral pathogens or antigens. Such subunit vaccines are much safer than traditional vaccines based on inactivated pathogens which are more likely to produce side-effects. Therefore, to tackle a pandemic and rapidly produce safer and more effective subunit vaccines based on protein assemblies, it is necessary to understand the basic structural features which drive protein self-assembly and functionalization of portions of pathogens. This review highlights recent developments and future perspectives in production of non-viral protein assemblies with essential structural features of subunit vaccines.
Collapse
|
43
|
Yang R, Zhu L, Meng D, Wang Q, Zhou K, Wang Z, Zhou Z. Proteins from leguminous plants: from structure, property to the function in encapsulation/binding and delivery of bioactive compounds. Crit Rev Food Sci Nutr 2021; 62:5203-5223. [PMID: 33569994 DOI: 10.1080/10408398.2021.1883545] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Leguminous proteins are important nutritional components in leguminous plants, and they have different structures and functions depending on their sources. Due to their specific structures and physicochemical properties, leguminous proteins have received much attention in food and nutritional applications, and they can be applied as various carriers for binding/encapsulation and delivery of food bioactive compounds. In this review, we systematically summarize the different structures and functional properties of several leguminous proteins which can be classified as ferritin, trypsin inhibitor, β-conglycinin, glycinin, and various leguminous proteins isolates. Moreover, we review the development of leguminous proteins as carriers of food bioactive compounds, and emphasize the functions of leguminous protein-based binding/encapsulation and delivery in overcoming the low bioavailability, instability and low absorption efficiency of food bioactive compounds. The limitations and challenges of the utilization of leguminous proteins as carriers of food bioactive compounds are also discussed. Possible approaches to resolve the limitations of applying leguminous proteins such as instability of proteins and poor absorption of bioactive compounds are recommended.
Collapse
Affiliation(s)
- Rui Yang
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, P. R. China
| | - Lei Zhu
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, P. R. China
| | - Demei Meng
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, P. R. China
| | - Qiaoe Wang
- Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing, P. R. China
| | - Kai Zhou
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, P. R. China
| | - Zhiwei Wang
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, P. R. China
| | - Zhongkai Zhou
- Key Laboratory of Food Nutrition and Safety, Ministry of Education, Tianjin Key Laboratory of Food Nutrition and Safety, Tianjin University of Science & Technology, Tianjin, P. R. China
| |
Collapse
|
44
|
Jeon IS, Yoo JD, Gurung S, Kim M, Lee C, Park EJ, Park RW, Lee B, Kim S. Anticancer nanocage platforms for combined immunotherapy designed to harness immune checkpoints and deliver anticancer drugs. Biomaterials 2021; 270:120685. [PMID: 33524811 DOI: 10.1016/j.biomaterials.2021.120685] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 12/27/2020] [Accepted: 01/13/2021] [Indexed: 12/15/2022]
Abstract
The interaction of programmed cell death 1 ligand 1 (PD-L1) with its receptor, programmed cell death 1 (PD-1), inhibits T cell responses. Monoclonal antibodies that block this interaction have been shown effective as immunotherapy. However, only a subset of cancers exhibits a durable response to PD-1/PD-L1 blockade. Moreover, antibody-based immune checkpoint blockade is costly and is occasionally accompanied by systemic side effects. To overcome these limitations of antibody-based immune checkpoint blockade, an immune checkpoint-blocking ferritin nanocage displaying 24 PD-L1 binding peptides (PD-L1pep1) on its surface was designed and constructed. These ferritin nanocages displaying PD-L1pep1 (PpNF) specifically bind to PD-L1 expressed on cancer cells or to purified PD-L1 with a ~30 nM binding affinity. The addition of PpNF to co-cultures of T cells and cancer cells inhibited PD-1/PD-L1 interactions and restored T cell activities. In a mouse model of syngeneic colon cancer, PpNF specifically targeted tumors and showed antitumor activity. Moreover, PpNF nanocages encapsulating the chemotherapeutic drug doxorubicin had more potent antitumor activity than a monoclonal antibody against PD-L1. These results demonstrate that ferritin nanocages displaying surface PD-L1pep1 can be efficiently applied for immunotherapy, especially when encapsulating small chemotherapeutic drugs. These nanocages may have promise as an immunotherapeutic nanomedicine against various solid tumors.
Collapse
Affiliation(s)
- In Seon Jeon
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; CMRI, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Jae Do Yoo
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; CMRI, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Smriti Gurung
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Minseong Kim
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; CMRI, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Chanju Lee
- Cancer Immunology Branch, Division of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Eun Jung Park
- Cancer Immunology Branch, Division of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Rang-Woon Park
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; CMRI, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Byungheon Lee
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; CMRI, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea
| | - Soyoun Kim
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea; CMRI, School of Medicine, Kyungpook National University, 680 Gukchaebosang-ro, Jung-gu, Daegu, 41944, Republic of Korea.
| |
Collapse
|
45
|
Yang R, Tian J, Liu Y, Zhu L, Sun J, Meng D, Wang Z, Wang C, Zhou Z, Chen L. Interaction mechanism of ferritin protein with chlorogenic acid and iron ion: The structure, iron redox, and polymerization evaluation. Food Chem 2021; 349:129144. [PMID: 33540218 DOI: 10.1016/j.foodchem.2021.129144] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 01/15/2021] [Accepted: 01/17/2021] [Indexed: 10/22/2022]
Abstract
Ferritin is an iron-containing protein and functions in the maintenance of iron balance in organisms. Currently the interaction among ferritin, ion iron, and food bioactive compounds is still unclear. In this study, the mechanism underlying the interaction of ferritin, ion iron, and chlorogenic acid was investigated, as well as the effect of chlorogenic acid on the physicochemical properties of ferritin. The results showed that chlorogenic acid could interact with Fe(III) to form chlorogenic acid-Fe(III) complexes, which then bonded with ferritin via hydrogen bonds in the ferritin-chlorogenic acid-Fe(III) complexes. The chlorogenic acid showed a high efficiency in Fe(II) chelation and hydroxyl radical (•OH) capture, and could promote iron oxidation and iron release induced by ferritin. Chlorogenic acid could also effectively reduce the polymerization extent of ferritin induced by Fe(III) and Fe(II). This study elucidates the interactions of multiple components in foodstuffs by using a protein-metal-polyphenol model.
Collapse
Affiliation(s)
- Rui Yang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Technology, Tianjin University of Science & Technology, Tianjin 300457, China.
| | - Jing Tian
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Technology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Yuqian Liu
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Technology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Lei Zhu
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Technology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Jixuan Sun
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Technology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Demei Meng
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Technology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Zhiwei Wang
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Technology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Chengtao Wang
- Beijing Engineering and Technology Research Center of Food Additives, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Zhongkai Zhou
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Technology, Tianjin University of Science & Technology, Tianjin 300457, China
| | - Lingyun Chen
- Department of Agricultural, Food & Nutritional Sciences, University of Alberta, Edmonton AB, T6G 2P5, Canada.
| |
Collapse
|
46
|
Zhao Z, Chen X, Chen Y, Li H, Fang K, Chen H, Li X, Qian P. A Self-Assembling Ferritin Nanoplatform for Designing Classical Swine Fever Vaccine: Elicitation of Potent Neutralizing Antibody. Vaccines (Basel) 2021; 9:45. [PMID: 33451123 PMCID: PMC7828615 DOI: 10.3390/vaccines9010045] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 01/29/2023] Open
Abstract
Protein-based self-assembling nanoplatforms exhibit superior immunogenicity compared with soluble antigens. Here, we present a comprehensive vaccine strategy for displaying classical swine fever virus (CSFV) E2 glycoprotein on the surface of ferritin (fe) nanocages. An E2-specific blocking antibody assay showed that the blocking rates in pE2-fe/Gel02 (84.3%) and a half-dose cohort of E2-fe/Gel02 (81.9%) were significantly higher (p < 0.05) than that in a ferritin-free cohort of pE2/Gel02 (62.7%) at 21 days post immunization (dpi) in vivo. Furthermore, quantitation of neutralizing potency revealed that a highly significant difference (p < 0.001) was observed between the pE2-fe/Gel02 cohort (1:32, equivalent to live-attenuated strain C at 1:32) and the pE2/Gel02 cohort (1:4) at 21 dpi. Moreover, the innate immune cytokines of IL-4 and IFN-γ activated by the half-dose (20 μg) cohort of E2-fe/Gel02 were equivalent to those elicited by the full dose (40 μg) of purified E2 in the pE2/Gel02 cohort at most time points. In conclusion, we successfully obtained an antigen-displaying E2-ferritin nanoplatform and confirmed high ferritin-assisted humoral and cellular immunities. Our results provided a novel paradigm of self-assembling nanovaccine development for the defense and elimination of potentially pandemic infectious viral pathogens.
Collapse
Affiliation(s)
- Zekai Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (Z.Z.); (X.C.); (Y.C.); (H.L.); (K.F.); (H.C.); (X.L.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xinghua Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (Z.Z.); (X.C.); (Y.C.); (H.L.); (K.F.); (H.C.); (X.L.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yibao Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (Z.Z.); (X.C.); (Y.C.); (H.L.); (K.F.); (H.C.); (X.L.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Hui Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (Z.Z.); (X.C.); (Y.C.); (H.L.); (K.F.); (H.C.); (X.L.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Kui Fang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (Z.Z.); (X.C.); (Y.C.); (H.L.); (K.F.); (H.C.); (X.L.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (Z.Z.); (X.C.); (Y.C.); (H.L.); (K.F.); (H.C.); (X.L.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiangmin Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (Z.Z.); (X.C.); (Y.C.); (H.L.); (K.F.); (H.C.); (X.L.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Ping Qian
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China; (Z.Z.); (X.C.); (Y.C.); (H.L.); (K.F.); (H.C.); (X.L.)
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
47
|
Wang J, Li Y, Nie G. Multifunctional biomolecule nanostructures for cancer therapy. NATURE REVIEWS. MATERIALS 2021; 6:766-783. [PMID: 34026278 PMCID: PMC8132739 DOI: 10.1038/s41578-021-00315-x] [Citation(s) in RCA: 202] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/06/2021] [Indexed: 05/08/2023]
Abstract
Biomolecule-based nanostructures are inherently multifunctional and harbour diverse biological activities, which can be explored for cancer nanomedicine. The supramolecular properties of biomolecules can be precisely programmed for the design of smart drug delivery vehicles, enabling efficient transport in vivo, targeted drug delivery and combinatorial therapy within a single design. In this Review, we discuss biomolecule-based nanostructures, including polysaccharides, nucleic acids, peptides and proteins, and highlight their enormous design space for multifunctional nanomedicines. We identify key challenges in cancer nanomedicine that can be addressed by biomolecule-based nanostructures and survey the distinct biological activities, programmability and in vivo behaviour of biomolecule-based nanostructures. Finally, we discuss challenges in the rational design, characterization and fabrication of biomolecule-based nanostructures, and identify obstacles that need to be overcome to enable clinical translation.
Collapse
Affiliation(s)
- Jing Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, China, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Yiye Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, China, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, China, Beijing, China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, China
- GBA Research Innovation Institute for Nanotechnology, Guangdong, China
- Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
48
|
Yoo JD, Bae SM, Seo J, Jeon IS, Vadevoo SMP, Kim SY, Kim IS, Lee B, Kim S. Designed ferritin nanocages displaying trimeric TRAIL and tumor-targeting peptides confer superior anti-tumor efficacy. Sci Rep 2020; 10:19997. [PMID: 33203916 PMCID: PMC7672110 DOI: 10.1038/s41598-020-77095-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 11/05/2020] [Indexed: 12/15/2022] Open
Abstract
TRAIL is considered a promising target for cancer therapy because it mediates activation of the extrinsic apoptosis pathway in a tumor-specific manner by binding to and trimerizing its functional receptors, DR4 or DR5. Although recombinant human TRAIL has shown high potency and specificity for killing cancer cells in preclinical studies, it has failed in multiple clinical trials for several reasons, including a very short half-life mainly caused by instability of the monomeric form of TRAIL and rapid renal clearance of the off-targeted TRAIL. To overcome such obstacles, we developed a TRAIL-active trimer nanocage (TRAIL-ATNC) that presents the TRAIL ligand in its trimer-like conformation by connecting it to a triple helix sequence that links to the threefold axis of the ferritin nanocage. We also ligated the tumor-targeting peptide, IL4rP, to TRAIL-ATNC to enhance tumor targeting. The developed TRAIL-ATNCIL4rP showed enhanced agonistic activity compared with monomeric TRAIL. The in vivo serum half-life of TRAIL-ATNCIL4rP was ~ 16-times longer than that of native TRAIL. As a consequence of these properties, TRAIL-ATNCIL4rP exhibited efficacy as an anti-tumor agent in vivo against xenograft breast cancer as well as orthotopic pancreatic cancer models, highlighting the promise of this system for development as novel therapeutics against cancer.
Collapse
Affiliation(s)
- Jae Do Yoo
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Sang Mun Bae
- PrismCDX, Inc., 593-16, Dongtangiheung-ro, Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Junyoung Seo
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - In Seon Jeon
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Sri Murugan Poongkavithai Vadevoo
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Sang-Yeob Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 138-736, Republic of Korea
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
| | - In-San Kim
- Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Byungheon Lee
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea
| | - Soyoun Kim
- Department of Biochemistry and Cell Biology, Cell and Matrix Research Institute, School of Medicine, Kyungpook National University, Daegu, 41944, Republic of Korea.
| |
Collapse
|
49
|
Ma H, Li X, Li J, Zhao Z, Zhang H, Hao G, Chen H, Qian P. Immunization with a recombinant fusion of porcine reproductive and respiratory syndrome virus modified GP5 and ferritin elicits enhanced protective immunity in pigs. Virology 2020; 552:112-120. [PMID: 33152628 DOI: 10.1016/j.virol.2020.10.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 10/09/2020] [Accepted: 10/23/2020] [Indexed: 01/19/2023]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) has caused huge economic losses in the swine industry worldwide. Live and inactivated vaccines have only been partially successful in generating protective immune responses. The PRRS virus (PRRSV) glycoprotein 5 (GP5) is a major viral antigenic target and is thus suitable for development of genetically engineered PRRSV vaccines. Here, a modified GP5 and ferritin were fused and expressed using a baculovirus system to generate a GP5m-ferritin nanoparticle vaccine. We demonstrated that the GP5m-ferritin vaccine elicited higher serum antibody titers in pigs than inactivated PRRSV. Moreover, immunization with GP5m-Ft promoted a Th1-dominant cellular immune response and enhanced specific T-lymphocyte immune responses. GP5m-ferritin-vaccinated pigs had significantly lower mean rectal temperatures, respiratory scores, viremia, and macroscopic and microscopic lung lesion scores post-challenge compared with unvaccinated pigs. These results indicated that GP5m-ferritin subunit vaccines can elicit specific protective immune responses and represent promising vaccine candidates.
Collapse
Affiliation(s)
- Hui Ma
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China; College of Food and Bioengineering, Henan University of Animal Husbandry and Economy, Zhengzhou, 450000, Henan, China
| | - Xiangmin Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China; Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Jianglong Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
| | - Zekai Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
| | - Huawei Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
| | - Genxi Hao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China; Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China
| | - Ping Qian
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, Hubei, China; Key Laboratory of Preventive Veterinary Medicine in Hubei Province, The Cooperative Innovation Center for Sustainable Pig Production, Wuhan, Hubei, 430070, China; Laboratory of Animal Virology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, Hubei, China.
| |
Collapse
|
50
|
Li S, Qiao Y, Xu Y, Li P, Nie J, Zhao Q, Chai W, Shi Y, Kong W, Shan Y. Identification of Linear Peptide Immunogens with Verified Broad-spectrum Immunogenicity from the Conserved Regions within the Hemagglutinin Stem Domain of H1N1 Influenza Virus. Immunol Invest 2020; 51:411-424. [PMID: 33078652 DOI: 10.1080/08820139.2020.1834579] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background: Influenza A viruses (IAVs) induce acute respiratory disease and cause severe epidemics and pandemics. Since IAVs exhibit antigenic variation and genome reassortment, the development of broad-spectrum influenza vaccines is crucial. The stem of the hemagglutinin (HA) is highly conserved across IAV strains and thus has been explored in broad-spectrum influenza vaccine studies. The present study aimed to identify viral epitopes capable of eliciting effective host immune responses, which can be explored for the development of broad-spectrum non-strain specific prophylactic options against IAV.Methods: In this study, a series of conserved linear sequences from the HA stem of IAV (H1N1) was recognized by sequence alignment and B/T-cell epitope prediction after being chemically coupled to the Keyhole Limpet Hemocyanin (KLH) protein. The predicted linear epitopes were identified by enzyme-linked immunosorbent assay (ELISA) after animal immunization and then fused with ferritin carriers.Results: Three predicted linear epitopes with relatively strong immunogenicity, P3, P6 and P8 were fused with ferritin carriers P3F, P6F and P8F, respectively to further improve their immunogenicity. Antibody titre of the sera of mice immunized with the recombinant immunogens revealed the elicitation of specific antibody-binding activities by the identified sequences. While hemagglutinin-inhibition activities were not detected in the antisera, neutralizing antibodies against the H1 and H3 virus subtypes were detected by the microneutralization assay.Conclusion: The linear epitopes fused with ferritin identified in this study can lay the foundation for future advancements in development of broad-spectrum subunit vaccine against IAV (H1N1), and give rise to the potential future applicability of ferritin-based antigen delivery nanoplatforms.
Collapse
Affiliation(s)
- Shuang Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Yongbo Qiao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Yan Xu
- Laboratory of Molecular Immunology, State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Pengju Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Jiaojiao Nie
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Qi Zhao
- Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, China
| | - Wen Chai
- Changchun Institute of Biological Products Co., Ltd, Changchun, Jilin, China
| | - Yuhua Shi
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China.,Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin, China
| | - Yaming Shan
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China.,Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, China.,Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Jilin University, Changchun, Jilin, China
| |
Collapse
|