1
|
Um‐e‐Kalsoom, Wang S, Qu J, Liu L. Innovative optical imaging strategies for monitoring immunotherapy in the tumor microenvironments. Cancer Med 2024; 13:e70155. [PMID: 39387259 PMCID: PMC11465031 DOI: 10.1002/cam4.70155] [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: 01/13/2024] [Revised: 08/01/2024] [Accepted: 08/16/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND The tumor microenvironment (TME) plays a critical role in cancer progression and response to immunotherapy. Immunotherapy targeting the immune system has emerged as a promising treatment modality, but challenges in understanding the TME limit its efficacy. Optical imaging strategies offer noninvasive, real-time insights into the interactions between immune cells and the TME. OBJECTIVE This review assesses the progress of optical imaging technologies in monitoring immunotherapy within the TME and explores their potential applications in clinical trials and personalized cancer treatment. METHODS This is a comprehensive literature review based on the advances in optical imaging modalities including fluorescence imaging (FLI), bioluminescence imaging (BLI), and photoacoustic imaging (PAI). These modalities were analyzed for their capacity to provide high-resolution, real-time imaging of immune cell dynamics, tumor vasculature, and other critical components of the TME. RESULTS Optical imaging techniques have shown significant potential in tracking immune cell infiltration, assessing immune checkpoint inhibitors, and visualizing drug delivery within the TME. Technologies like FLI and BLI are pivotal in tracking immune responses in preclinical models, while PAI provides functional imaging with deeper tissue penetration. The integration of these modalities with immunotherapy holds promise for improving treatment monitoring and outcomes. CONCLUSION Optical imaging is a powerful tool for understanding the complexities of the TME and optimizing immunotherapy. Further advancements in imaging technologies, combined with nanomaterial-based approaches, could pave the way for enhanced diagnostic accuracy and therapeutic efficacy in cancer treatment.
Collapse
Affiliation(s)
- Um‐e‐Kalsoom
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Shiqi Wang
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Junle Qu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| | - Liwei Liu
- Key Laboratory of Optoelectronic Devices and Systems of Guangdong Province and Ministry of Education, College of Physics and Optoelectronic EngineeringShenzhen UniversityShenzhenChina
| |
Collapse
|
2
|
Priwitaningrum DL, Pednekar K, Gabriël AV, Varela-Moreira AA, Le Gac S, Vellekoop I, Storm G, Hennink WE, Prakash J. Evaluation of paclitaxel-loaded polymeric nanoparticles in 3D tumor model: impact of tumor stroma on penetration and efficacy. Drug Deliv Transl Res 2023; 13:1470-1483. [PMID: 36853438 PMCID: PMC10102101 DOI: 10.1007/s13346-023-01310-1] [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] [Accepted: 02/02/2023] [Indexed: 03/01/2023]
Abstract
Since tumor stroma poses as a barrier to achieve efficacy of nanomedicines, it is essential to evaluate nano-chemotherapeutics in stroma-mimicking 3D models that reliably predict their behavior regarding these hurdles limiting efficacy. In this study, we evaluated the effect of paclitaxel-loaded polymeric micelles (PTX-PMCs) and polymeric nanoparticles (PTX-PNPs) in a tumor stroma-mimicking 3D in vitro model. PTX-PMCs (77 nm) based on a amphiphilic block copolymer of mPEG-b-p(HPMAm-Bz) and PTX-PNPs (159 nm) based on poly(lactic-co-glycolic acid) were prepared, which had an encapsulation efficiency (EE%) of 81 ± 15% and 45 ± 8%, respectively. 3D homospheroids of mouse 4T1 breast cancer cells and heterospheroids of NIH3T3 fibroblasts and 4T1 (5:1 ratio) were prepared and characterized with high content two-photon microscopy and immunostaining. Data showed an induction of epithelial-mesenchymal transition (α-SMA) in both homo- and heterospheroids, while ECM (collagen) deposition only in heterospheroids. Two-photon imaging revealed that both fluorescently labeled PMCs and PNPs penetrated into the core of homospheroids and only PMCs penetrated into heterospheroids. Furthermore, PTX-PMCs, PTX-PNPs, and free PTX induced cytotoxicity in tumor cells and fibroblasts grown as monolayer, but these effects were substantially reduced in 3D models, in particular in heterospheroids. Gene expression analysis showed that heterospheroids had a significant increase of drug resistance markers (Bcl2, Abgc2) compared to 2D or 3D monocultures. Altogether, this study shows that the efficacy of nanotherapeutics is challenged by stroma-induced poor penetration and development of resistant phenotype. Therefore, this tumor stroma-mimicking 3D model can provide an excellent platform to study penetration and effects of nanotherapeutics before in vivo studies.
Collapse
Affiliation(s)
- Dwi L Priwitaningrum
- Engineered Therapeutics, Department of Advanced Organ Bioengineering and Therapeutics, TechMed Centre, Faculty of Science and Technology, University of Twente, Drienerlolaan 5, 7500AE, Enschede, The Netherlands
- Department of Pharmaceutics, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Kunal Pednekar
- Engineered Therapeutics, Department of Advanced Organ Bioengineering and Therapeutics, TechMed Centre, Faculty of Science and Technology, University of Twente, Drienerlolaan 5, 7500AE, Enschede, The Netherlands
| | - Alexandros V Gabriël
- Engineered Therapeutics, Department of Advanced Organ Bioengineering and Therapeutics, TechMed Centre, Faculty of Science and Technology, University of Twente, Drienerlolaan 5, 7500AE, Enschede, The Netherlands
| | - Aida A Varela-Moreira
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Severine Le Gac
- Applied Microfluidics for BioEngineering Research, Faculty of Electrical Engineering, Mathematics and Computer Science, MESA+ Institute for Nanotechnology, TechMed Centre, University of Twente, Enschede, The Netherlands
| | - Ivo Vellekoop
- Biomedical Photonic Imaging, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Jai Prakash
- Engineered Therapeutics, Department of Advanced Organ Bioengineering and Therapeutics, TechMed Centre, Faculty of Science and Technology, University of Twente, Drienerlolaan 5, 7500AE, Enschede, The Netherlands.
| |
Collapse
|
3
|
Upconversion nanoparticle platform for efficient dendritic cell antigen delivery and simultaneous tracking. Mikrochim Acta 2022; 189:368. [PMID: 36057018 PMCID: PMC9440881 DOI: 10.1007/s00604-022-05441-z] [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: 05/03/2022] [Accepted: 07/31/2022] [Indexed: 11/10/2022]
Abstract
Upconversion nanoparticles (UCNPs) represent a group of NPs that can convert near-infrared (NIR) light into ultraviolet and visible light, thus possess deep tissue penetration power with less background fluorescence noise interference, and do not induce damage to biological tissues. Due to their unique optical properties and possibility for surface modification, UCNPs can be exploited for concomitant antigen delivery into dendritic cells (DCs) and monitoring by molecular imaging. In this study, we focus on the development of a nano-delivery platform targeting DCs for immunotherapy and simultaneous imaging. OVA 254–267 (OVA24) peptide antigen, harboring a CD8 T cell epitope, and Pam3CysSerLys4 (Pam3CSK4) adjuvant were chemically linked to the surface of UCNPs by amide condensation to stimulate DC maturation and antigen presentation. The OVA24-Pam3CSK4-UCNPs were thoroughly characterized and showed a homogeneous morphology and surface electronegativity, which promoted a good dispersion of the NPs. In vitro experiments demonstrated that OVA24-Pam3CSK4-UCNPs induced a strong immune response, including DC maturation, T cell activation, and proliferation, as well as interferon gamma (IFN-γ) production. In vivo, highly sensitive upconversion luminescence (UCL) imaging of OVA24-Pam3CSK4-UCNPs allowed tracking of UCNPs from the periphery to lymph nodes. In summary, OVA24-Pam3CSK4-UCNPs represent an effective tool for DC-based immunotherapy.
Collapse
|
4
|
Li X, Pan C, Sun P, Peng Z, Feng E, Wu J, Wang H, Zhu L. Orthogonal modular biosynthesis of nanoscale conjugate vaccines for vaccination against infection. NANO RESEARCH 2022; 15:1645-1653. [PMID: 34405037 PMCID: PMC8359766 DOI: 10.1007/s12274-021-3713-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/17/2021] [Accepted: 06/24/2021] [Indexed: 05/21/2023]
Abstract
UNLABELLED Conjugate vaccines represent one of the most effective means for controlling the occurrence of bacterial diseases. Although nanotechnology has been greatly applied in the field of vaccines, it is seldom used for conjugate vaccine research because it is very difficult to connect polysaccharides and nanocarriers. In this work, an orthogonal and modular biosynthesis method was used to produce nanoconjugate vaccines using the SpyTag/SpyCatcher system. When SpyTag/SpyCatcher system is combined with protein glycosylation technology, bacterial O-polysaccharide obtained from Shigela flexneri 2a can be conjugated onto the surfaces of different virus-like particles (VLPs) in a biocompatible and controlled manner. After confirming the excellent lymph node targeting and humoral immune activation abilities, these nanoconjugate vaccines further induced efficient prophylactic effects against infection in a mouse model. These results demonstrated that natural polysaccharide antigens can be easily connected to VLPs to prepare highly efficient nanoconjugate vaccines. To the best of the researchers' knowledge, this is the first time VLP-based nanoconjugate vaccines are produced efficiently, and this strategy could be applied to develop various pathogenic nanoconjugate vaccines. ELECTRONIC SUPPLEMENTARY MATERIAL Supplementary material (Figs. S1-S9) is available in the online version of this article at 10.1007/s12274-021-3713-4.
Collapse
Affiliation(s)
- Xin Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Chao Pan
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Peng Sun
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Zhehui Peng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Erling Feng
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Jun Wu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Hengliang Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| | - Li Zhu
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, Beijing, 100071 China
| |
Collapse
|
5
|
Deng S, Gigliobianco MR, Mijit E, Minicucci M, Cortese M, Campisi B, Voinovich D, Battistelli M, Salucci S, Gobbi P, Lupidi G, Zambito G, Mezzanotte L, Censi R, Di Martino P. Dually Cross-Linked Core-Shell Structure Nanohydrogel with Redox-Responsive Degradability for Intracellular Delivery. Pharmaceutics 2021; 13:pharmaceutics13122048. [PMID: 34959330 PMCID: PMC8708258 DOI: 10.3390/pharmaceutics13122048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/20/2021] [Accepted: 11/25/2021] [Indexed: 11/16/2022] Open
Abstract
A redox-responsive nanocarrier is a promising strategy for the intracellular drug release because it protects the payload, prevents its undesirable leakage during extracellular transport, and favors site-specific drug delivery. In this study, we developed a novel redox responsive core-shell structure nanohydrogel prepared by a water in oil nanoemulsion method using two biocompatible synthetic polymers: vinyl sulfonated poly(N-(2-hydroxypropyl) methacrylamide mono/dilactate)-polyethylene glycol-poly(N-(2-hydroxypropyl) methacrylamide mono/dilactate) triblock copolymer, and thiolated hyaluronic acid. The influence on the nanohydrogel particle size and distribution of formulation parameters was investigated by a three-level full factorial design to optimize the preparation conditions. The surface and core-shell morphology of the nanohydrogel were observed by scanning electron microscope, transmission electron microscopy, and further confirmed by Fourier transform infrared spectroscopy and Raman spectroscopy from the standpoint of chemical composition. The redox-responsive biodegradability of the nanohydrogel in reducing environments was determined using glutathione as reducing agent. A nanohydrogel with particle size around 250 nm and polydispersity index around 0.1 is characterized by a thermosensitive shell which jellifies at body temperature and crosslinks at the interface of a redox-responsive hyaluronic acid core via the Michael addition reaction. The nanohydrogel showed good encapsulation efficiency for model macromolecules of different molecular weight (93% for cytochrome C, 47% for horseradish peroxidase, and 90% for bovine serum albumin), capacity to retain the peroxidase-like enzymatic activity (around 90%) of cytochrome C and horseradish peroxidase, and specific redox-responsive release behavior. Additionally, the nanohydrogel exhibited excellent cytocompatibility and internalization efficiency into macrophages. Therefore, the developed core-shell structure nanohydrogel can be considered a promising tool for the potential intracellular delivery of different pharmaceutical applications, including for cancer therapy.
Collapse
Affiliation(s)
- Siyuan Deng
- School of Pharmacy, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy; (S.D.); (M.C.); (G.L.)
| | | | - Emin Mijit
- Physics Division, School of Science and Technology, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy; (E.M.); (M.M.)
| | - Marco Minicucci
- Physics Division, School of Science and Technology, University of Camerino, Via Madonna delle Carceri 9, 62032 Camerino, Italy; (E.M.); (M.M.)
| | - Manuela Cortese
- School of Pharmacy, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy; (S.D.); (M.C.); (G.L.)
| | - Barbara Campisi
- Department of Economic, Business, Mathematic and Statistical Sciences, University of Trieste, 34127 Trieste, Italy;
| | - Dario Voinovich
- Department of Chemical and Pharmaceutical Science, University of Trieste, P. le Europa 1, 34127 Trieste, Italy;
| | - Michela Battistelli
- Institute of Morphological Sciences, University of Urbino, Via Ca’ le Suore 2, 61029 Urbino, Italy; (M.B.); (P.G.)
| | - Sara Salucci
- Cellular Signalling Laboratory, Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, 40126 Bologna, Italy;
| | - Pietro Gobbi
- Institute of Morphological Sciences, University of Urbino, Via Ca’ le Suore 2, 61029 Urbino, Italy; (M.B.); (P.G.)
| | - Giulio Lupidi
- School of Pharmacy, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy; (S.D.); (M.C.); (G.L.)
| | - Giorgia Zambito
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (G.Z.); (L.M.)
| | - Laura Mezzanotte
- Department of Radiology and Nuclear Medicine, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands; (G.Z.); (L.M.)
| | - Roberta Censi
- School of Pharmacy, University of Camerino, Via S. Agostino 1, 62032 Camerino, Italy; (S.D.); (M.C.); (G.L.)
- Correspondence: ; Tel.: +39-0737-40-2231
| | - Piera Di Martino
- Dipartimento di Farmacia, Università “G. D’Annunzio” Chieti e Pescara, Via dei Vestini, 1, 66100 Chieti, Italy;
| |
Collapse
|
6
|
Haseda Y, Munakata L, Kimura C, Kinugasa-Katayama Y, Mori Y, Suzuki R, Aoshi T. Development of combination adjuvant for efficient T cell and antibody response induction against protein antigen. PLoS One 2021; 16:e0254628. [PMID: 34339430 PMCID: PMC8328330 DOI: 10.1371/journal.pone.0254628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 07/01/2021] [Indexed: 01/01/2023] Open
Abstract
Most current clinical vaccines work primarily by inducing the production of neutralizing antibodies against pathogens. Vaccine adjuvants that efficiently induce T cell responses to protein antigens need to be developed. In this study, we developed a new combination adjuvant consisting of 1,2-dioleoyl-3-trimethylammonium propane (DOTAP), D35, and an aluminum salt. Among the various combinations tested, the DOTAP/D35/aluminum salt adjuvant induced strong T cell and antibody responses against the model protein antigen with a single immunization. Adjuvant component and model antigen interaction studies in vitro also revealed that the strong mutual interactions among protein antigens and other components were one of the important factors for this efficient immune induction by the novel combination adjuvant. In addition, in vivo imaging of the antigen distribution suggested that the DOTAP component in the combination adjuvant formulation elicited transient antigen accumulation at the draining lymph nodes, possibly by antigen uptake DC migration. These results indicate the potential of the new combination adjuvant as a promising vaccine adjuvant candidate to treat infectious diseases and cancers.
Collapse
Affiliation(s)
- Yasunari Haseda
- Vaccine Dynamics Project, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Lisa Munakata
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University, Itabashi-ku, Tokyo, Japan
| | - Chiyo Kimura
- Department of Cellular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Yumi Kinugasa-Katayama
- Department of Cellular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
| | - Yasuko Mori
- Division of Clinical Virology, Center for Infectious Diseases, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryo Suzuki
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University, Itabashi-ku, Tokyo, Japan
| | - Taiki Aoshi
- Department of Cellular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, Japan
- * E-mail:
| |
Collapse
|
7
|
Cid R, Bolívar J. Platforms for Production of Protein-Based Vaccines: From Classical to Next-Generation Strategies. Biomolecules 2021; 11:1072. [PMID: 34439738 PMCID: PMC8394948 DOI: 10.3390/biom11081072] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 07/16/2021] [Accepted: 07/17/2021] [Indexed: 12/12/2022] Open
Abstract
To date, vaccination has become one of the most effective strategies to control and reduce infectious diseases, preventing millions of deaths worldwide. The earliest vaccines were developed as live-attenuated or inactivated pathogens, and, although they still represent the most extended human vaccine types, they also face some issues, such as the potential to revert to a pathogenic form of live-attenuated formulations or the weaker immune response associated with inactivated vaccines. Advances in genetic engineering have enabled improvements in vaccine design and strategies, such as recombinant subunit vaccines, have emerged, expanding the number of diseases that can be prevented. Moreover, antigen display systems such as VLPs or those designed by nanotechnology have improved the efficacy of subunit vaccines. Platforms for the production of recombinant vaccines have also evolved from the first hosts, Escherichia coli and Saccharomyces cerevisiae, to insect or mammalian cells. Traditional bacterial and yeast systems have been improved by engineering and new systems based on plants or insect larvae have emerged as alternative, low-cost platforms. Vaccine development is still time-consuming and costly, and alternative systems that can offer cost-effective and faster processes are demanding to address infectious diseases that still do not have a treatment and to face possible future pandemics.
Collapse
Affiliation(s)
- Raquel Cid
- ADL Bionatur Solutions S.A., Av. del Desarrollo Tecnológico 11, 11591 Jerez de la Frontera, Spain
| | - Jorge Bolívar
- Department of Biomedicine, Biotechnology and Public Health-Biochemistry and Molecular Biology, Campus Universitario de Puerto Real, University of Cadiz, 11510 Puerto Real, Spain
| |
Collapse
|
8
|
Seyfoori A, Shokrollahi Barough M, Mokarram P, Ahmadi M, Mehrbod P, Sheidary A, Madrakian T, Kiumarsi M, Walsh T, McAlinden KD, Ghosh CC, Sharma P, Zeki AA, Ghavami S, Akbari M. Emerging Advances of Nanotechnology in Drug and Vaccine Delivery against Viral Associated Respiratory Infectious Diseases (VARID). Int J Mol Sci 2021; 22:6937. [PMID: 34203268 PMCID: PMC8269337 DOI: 10.3390/ijms22136937] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/19/2021] [Accepted: 06/19/2021] [Indexed: 12/12/2022] Open
Abstract
Viral-associated respiratory infectious diseases are one of the most prominent subsets of respiratory failures, known as viral respiratory infections (VRI). VRIs are proceeded by an infection caused by viruses infecting the respiratory system. For the past 100 years, viral associated respiratory epidemics have been the most common cause of infectious disease worldwide. Due to several drawbacks of the current anti-viral treatments, such as drug resistance generation and non-targeting of viral proteins, the development of novel nanotherapeutic or nano-vaccine strategies can be considered essential. Due to their specific physical and biological properties, nanoparticles hold promising opportunities for both anti-viral treatments and vaccines against viral infections. Besides the specific physiological properties of the respiratory system, there is a significant demand for utilizing nano-designs in the production of vaccines or antiviral agents for airway-localized administration. SARS-CoV-2, as an immediate example of respiratory viruses, is an enveloped, positive-sense, single-stranded RNA virus belonging to the coronaviridae family. COVID-19 can lead to acute respiratory distress syndrome, similarly to other members of the coronaviridae. Hence, reviewing the current and past emerging nanotechnology-based medications on similar respiratory viral diseases can identify pathways towards generating novel SARS-CoV-2 nanotherapeutics and/or nano-vaccines.
Collapse
Affiliation(s)
- Amir Seyfoori
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (T.W.)
- Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran
| | - Mahdieh Shokrollahi Barough
- Department of Immunology, Iran University of Medical Sciences, Tehran 1449614535, Iran;
- ATMP Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran 1517964311, Iran
| | - Pooneh Mokarram
- Department of Clinical Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran;
- Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Mazaher Ahmadi
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (M.A.); (T.M.)
| | - Parvaneh Mehrbod
- Influenza and Respiratory Viruses Department, Pasteur Institute of IRAN, Tehran 1316943551, Iran;
| | - Alireza Sheidary
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14155-6451, Iran;
| | - Tayyebeh Madrakian
- Department of Analytical Chemistry, Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6517838695, Iran; (M.A.); (T.M.)
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 14155-6451, Iran;
| | - Mohammad Kiumarsi
- Department of Human Anatomy and Cell Science, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
| | - Tavia Walsh
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (T.W.)
| | - Kielan D. McAlinden
- Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Launceston, TAS 7248, Australia;
| | - Chandra C. Ghosh
- Roger Williams Medical Center, Immuno-Oncology Institute (Ix2), Providence, RI 02908, USA;
| | - Pawan Sharma
- Center for Translational Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, Jane & Leonard Korman Respiratory Institute, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Amir A. Zeki
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, U.C. Davis Lung Center, Davis School of Medicine, University of California, Davis, CA 95817, USA;
- Veterans Affairs Medical Center, Mather, CA 95817, USA
| | - Saeid Ghavami
- Autophagy Research Center, Health Policy Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Department of Human Anatomy and Cell Science, Rady College of Medicine, Max Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Mohsen Akbari
- Laboratory for Innovations in Micro Engineering (LiME), Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2, Canada; (A.S.); (T.W.)
- Biotechnology Center, Silesian University of Technology, Akademicka 2A, 44-100 Gliwice, Poland
- Center for Advanced Materials and Related Technologies, University of Victoria, Victoria, BC V8P 5C2, Canada
| |
Collapse
|
9
|
Lee ALZ, Yang C, Gao S, Wang Y, Hedrick JL, Yang YY. Biodegradable Cationic Polycarbonates as Vaccine Adjuvants. ACS APPLIED MATERIALS & INTERFACES 2020; 12:52285-52297. [PMID: 33179910 DOI: 10.1021/acsami.0c09649] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In this study, biodegradable cationic polycarbonate and polylactide block copolymers were synthesized and successfully used as novel vaccine adjuvants to provide enhanced anticancer immunity. The polymers formed nanoparticles with the model vaccine, ovalbumin (OVA), and the immunostimulant toll-like receptor 3 agonist poly(I:C) (a synthetic analog of the double-stranded RNA). Higher uptake of poly(I:C) by the bone marrow-derived dendritic cells and macrophages and OVA by dendritic cells was observed when delivered using the polymer adjuvant. In vivo experiments showed that these nanoparticles remained longer in the subcutaneous injection site as compared to OVA alone and led to higher production of anti-OVA specific antibodies with prolonged immunostimulation. When OVA was combined with poly(I:C) that was either co-entrapped in the same particles or as separate particles, a comparable level of anti-OVA IgG1 antibodies and interleukin-6 (IL-6) was produced in mouse blood plasma, and a similar level of cytotoxic T lymphocyte (CTL) response in mice was stimulated as compared to OVA/Alum particles. Furthermore, tumor rejection in the mice that were vaccinated for 9 months with the formulations containing the polymer adjuvant was stronger than the other treatment groups without the polymer. Notably, the cationic polycarbonates were not associated with any adverse in vivo effects. Thus, these biodegradable polymers may be promising substitutes for aluminum-based adjuvants in vaccine formulations.
Collapse
Affiliation(s)
- Ashlynn L Z Lee
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Chuan Yang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Shujun Gao
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
- NanoBio Lab, 31 Biopolis Way, #09-01 The Nanos, Singapore 138669, Singapore
| | - Yanming Wang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - James L Hedrick
- IBM Almaden Research Center, 650 Harry Road, San Jose, California 95120 United States
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| |
Collapse
|
10
|
Priwitaningrum DL, Jentsch J, Bansal R, Rahimian S, Storm G, Hennink WE, Prakash J. Apoptosis-inducing peptide loaded in PLGA nanoparticles induces anti-tumor effects in vivo. Int J Pharm 2020; 585:119535. [PMID: 32534162 DOI: 10.1016/j.ijpharm.2020.119535] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 06/04/2020] [Accepted: 06/06/2020] [Indexed: 01/17/2023]
Abstract
Induction of apoptosis in tumor cells specifically within the complex tumor microenvironment is highly desirable to kill them efficiently and to enhance the effects of chemotherapy. Second mitochondria-derived activator of caspase (Smac) is a key pro-apoptotic pathway which can be activated with a Smac mimetic peptide. However, in vivo application of peptides is hampered by several limitations such as poor pharmacokinetics, rapid elimination, enzymatic degradation, and insufficient intracellular delivery. In this study, we developed a nanosystem to deliver a Smac peptide to tumor by passive targeting. We first synthesized a chimeric peptide that consists of the 8-mer Smac peptide and a 14-mer cell penetrating peptide (CPP) and then encapsulated the Smac-CPP into polymeric nanoparticles (Smac-CPP-NPs). In vitro, Smac-CPP-NPs were rapidly internalized by 4T1 mammary tumor cells and subsequently released Smac-CPP into the cells, as shown with fluorescence microscopy. Furthermore, Smac-CPP-NPs induced apoptosis in tumor cells, as confirmed with cell viability and caspase 3/7 assays. Interestingly, combination of Smac-CPP-NPs with doxorubicin (dox), a clinically used cytostatic drug, showed combined effects in vitro in 4T1 cells. The effect was significantly better than that of SMAC-CPP-NPs alone as well as empty nanoparticles and dox. In vivo, co-treatment with Smac-CPP-NPs and free dox reduced the tumor growth to 85%. Furthermore, the combination of Smac-CPP-NPs and free dox showed reduced proliferating tumor cells (Ki-67 staining) and increased apoptotic cells (cleaved caspase-3 staining) in tumors. In conclusion, the present study demonstrates that the intracellular delivery of Smac-mimetic peptide using nanoparticle system can be an interesting strategy to attenuate the tumor growth and to potentiate the therapeutic efficacy of chemotherapy in vivo.
Collapse
Affiliation(s)
- Dwi L Priwitaningrum
- Targeted Therapeutics and Nanomedicine, Department of Biomaterials Science and Technology, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Faculty of Pharmacy, University of Sumatera Utara, Medan, Indonesia
| | - Julian Jentsch
- Targeted Therapeutics and Nanomedicine, Department of Biomaterials Science and Technology, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Ruchi Bansal
- Targeted Therapeutics and Nanomedicine, Department of Biomaterials Science and Technology, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Sima Rahimian
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Wim E Hennink
- Department of Pharmaceutics, Faculty of Pharmacy, University of Sumatera Utara, Medan, Indonesia
| | - Jai Prakash
- Targeted Therapeutics and Nanomedicine, Department of Biomaterials Science and Technology, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands.
| |
Collapse
|
11
|
Oliveira TL, Bacelo KL, Forster KM, Ilha V, Rodrigues OE, Hartwig DD. DNA nanovaccines prepared using LemA antigen protect Golden Syrian hamsters against Leptospira lethal infection. Mem Inst Oswaldo Cruz 2020; 115:e190396. [PMID: 32321154 PMCID: PMC7164400 DOI: 10.1590/0074-02760190396] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 03/03/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Nanoparticles (NPs) are viable candidates as carriers of exogenous materials into cells via transfection and can be used in the DNA vaccination strategy against leptospirosis. OBJECTIVES We evaluated the efficiency of halloysite clay nanotubes (HNTs) and amine-functionalised multi-walled carbon nanotubes (NH2-MWCNTs) in facilitating recombinant LemA antigen (rLemA) expression and protecting Golden Syrian hamsters (Mesocricetus auratus) against Leptospira interrogans lethal infection. METHODS An indirect immunofluorescent technique was used to investigate the potency of HNTs and NH2-MWCNTs in enhancing the transfection and expression efficiency of the DNA vaccine in Chinese hamster ovary (CHO) cells. Hamsters were immunised with two doses of vaccines HNT-pTARGET/lemA, NH2-MWCNTs-pTARGET/lemA, pTARGET/lemA, and empty pTARGET (control), and the efficacy was determined in terms of humoral immune response and protection against a lethal challenge. FINDINGS rLemA DNA vaccines carried by NPs were able to transfect CHO cells effectively, inducing IgG immune response in hamsters (p < 0.05), and did not exhibit cytotoxic effects. Furthermore, 83.3% of the hamsters immunised with NH2-MWCNTs-pTARGET/lemA were protected against the lethal challenge (p < 0.01), and 66.7% of hamsters immunised with HNT-pTARGET/lemA survived (p < 0.05). MAIN CONCLUSIONS NH2-MWCNTs and HNTs can act as antigen carriers for mammalian cells and are suitable for DNA nanovaccine delivery.
Collapse
Affiliation(s)
- Thaís Larré Oliveira
- Universidade Federal de Pelotas, Centro de Desenvolvimento Tecnológico, Programa de Pós-Graduação em Biotecnologia, Núcleo de Biotecnologia, Pelotas, RS, Brasil
| | - Kátia Leston Bacelo
- Universidade Federal de Pelotas, Centro de Desenvolvimento Tecnológico, Programa de Pós-Graduação em Biotecnologia, Núcleo de Biotecnologia, Pelotas, RS, Brasil
| | - Karine Maciel Forster
- Universidade Federal de Pelotas, Centro de Desenvolvimento Tecnológico, Programa de Pós-Graduação em Biotecnologia, Núcleo de Biotecnologia, Pelotas, RS, Brasil
| | - Vinicius Ilha
- Universidade Federal de Santa Maria, Departamento de Química, Santa Maria, RS, Brasil
| | | | - Daiane D Hartwig
- Universidade Federal de Pelotas, Centro de Desenvolvimento Tecnológico, Programa de Pós-Graduação em Biotecnologia, Núcleo de Biotecnologia, Pelotas, RS, Brasil.,Universidade Federal de Pelotas, Instituto de Biologia, Departamento de Microbiologia e Parasitologia, Pelotas, RS, Brasil
| |
Collapse
|
12
|
Cancer Immunoimaging with Smart Nanoparticles. Trends Biotechnol 2020; 38:388-403. [DOI: 10.1016/j.tibtech.2019.11.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 10/30/2019] [Accepted: 11/05/2019] [Indexed: 12/31/2022]
|
13
|
Shields CW, Wang LLW, Evans MA, Mitragotri S. Materials for Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1901633. [PMID: 31250498 DOI: 10.1002/adma.201901633] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/17/2019] [Indexed: 05/20/2023]
Abstract
Breakthroughs in materials engineering have accelerated the progress of immunotherapy in preclinical studies. The interplay of chemistry and materials has resulted in improved loading, targeting, and release of immunomodulatory agents. An overview of the materials that are used to enable or improve the success of immunotherapies in preclinical studies is presented, from immunosuppressive to proinflammatory strategies, with particular emphasis on technologies poised for clinical translation. The materials are organized based on their characteristic length scale, whereby the enabling feature of each technology is organized by the structure of that material. For example, the mechanisms by which i) nanoscale materials can improve targeting and infiltration of immunomodulatory payloads into tissues and cells, ii) microscale materials can facilitate cell-mediated transport and serve as artificial antigen-presenting cells, and iii) macroscale materials can form the basis of artificial microenvironments to promote cell infiltration and reprogramming are discussed. As a step toward establishing a set of design rules for future immunotherapies, materials that intrinsically activate or suppress the immune system are reviewed. Finally, a brief outlook on the trajectory of these systems and how they may be improved to address unsolved challenges in cancer, infectious diseases, and autoimmunity is presented.
Collapse
Affiliation(s)
- C Wyatt Shields
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Lily Li-Wen Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Michael A Evans
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
14
|
Schmidt ST, Pedersen GK, Christensen D. Rational Design and In Vivo Characterization of Vaccine Adjuvants. ILAR J 2019; 59:309-322. [PMID: 30624655 DOI: 10.1093/ilar/ily018] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 09/05/2018] [Indexed: 12/14/2022] Open
Abstract
Many different adjuvants are currently being developed for subunit vaccines against a number of pathogens and diseases. Rational design is increasingly used to develop novel vaccine adjuvants, which requires extensive knowledge of, for example, the desired immune responses, target antigen-presenting cell subsets, their localization, and expression of relevant pattern-recognition receptors. The adjuvant mechanism of action and efficacy are usually evaluated in animal models, where mice are by far the most used. In this review, we present methods for assessing adjuvant efficacy and function in animal models: (1) whole-body biodistribution evaluated by using fluorescently and radioactively labeled vaccine components; (2) association and activation of immune cell subsets at the injection site, in the draining lymph node, and the spleen; (4) adaptive immune responses, such as cytotoxic T-lymphocytes, various T-helper cell subsets, and antibody responses, which may be quantitatively evaluated using ELISA, ELISPOT, and immunoplex assays and qualitatively evaluated using flow cytometric and single cell sequencing assays; and (5) effector responses, for example, antigen-specific cytotoxic potential of CD8+ T cells and antibody neutralization assays. While the vaccine-induced immune responses in mice often correlate with the responses induced in humans, there are instances where immune responses detected in mice are not translated to the human situation. We discuss some examples of correlation and discrepancy between mouse and human immune responses and how to understand them.
Collapse
Affiliation(s)
- Signe Tandrup Schmidt
- Statens Serum Institut, Center for Vaccine Research, Department of Infectious Disease Immunology, Copenhagen S, Denmark
| | - Gabriel Kristian Pedersen
- Statens Serum Institut, Center for Vaccine Research, Department of Infectious Disease Immunology, Copenhagen S, Denmark
| | - Dennis Christensen
- Statens Serum Institut, Center for Vaccine Research, Department of Infectious Disease Immunology, Copenhagen S, Denmark
| |
Collapse
|
15
|
Enhanced anti-tumor immunotherapy by dissolving microneedle patch loaded ovalbumin. PLoS One 2019; 14:e0220382. [PMID: 31386690 PMCID: PMC6684091 DOI: 10.1371/journal.pone.0220382] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 07/15/2019] [Indexed: 01/04/2023] Open
Abstract
The skin is a very suitable organ for the induction of immune responses to vaccine antigens. Antigen delivery systems to the skin by needle and syringe directly deposit the antigen into the epidermal-dermal compartment, one of the most immunocompetent sites due to the presence of professional antigen-presenting cells aimed at the induction of antigen-specific T cells. In this study, we analyzed the amount of ovalbumin as an antigen delivered to the skin by a microneedle. When ovalbumin protein as an antigen was delivered to the skin of mice using a dissolving microneedle, it induced an immune response through the enhanced proliferation and cytokines production by the splenocytes and lymph nodes. Also, it effectively increased the ovalbumin-specific CD8+ T cell and CD4+ T cell population and induced an ovalbumin-specific CTL response against the graft of ovalbumin-expressing EG7 tumor cells in the immunized mice. Also, we identified the inhibition of tumor growth and prevention of tumor formation in the context of the therapeutic and prophylactic vaccine, respectively through EG-7 tumor mouse model. Finally, these data show the potential of patches as attractive antigen delivery vehicles.
Collapse
|
16
|
Zhang L, Wu S, Qin Y, Fan F, Zhang Z, Huang C, Ji W, Lu L, Wang C, Sun H, Leng X, Kong D, Zhu D. Targeted Codelivery of an Antigen and Dual Agonists by Hybrid Nanoparticles for Enhanced Cancer Immunotherapy. NANO LETTERS 2019; 19:4237-4249. [PMID: 30868883 DOI: 10.1021/acs.nanolett.9b00030] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Among approaches of current cancer immunotherapy, a dendritic cell (DC)-targeted vaccine based on nanotechnology could be a promising way to efficiently induce potent immune responses. To enhance DC targeting and vaccine efficiency, we included imiquimod (IMQ), a toll-like receptor 7/8 (TLR 7/8) agonist, and monophosphoryl lipid A (MPLA), a TLR4 agonist, to synthesize lipid-polymer hybrid nanoparticles using PCL-PEG-PCL and DOTAP (IMNPs) as well as DSPE-PEG-mannose (MAN-IMNPS). The spatiotemporal delivery of MPLA (within the outer lipid layer) to extracellular TLR4 and IMQ (in the hydrophobic core of NPs) to intracellular TLR7/8 can activate DCs synergistically to improve vaccine efficacy. Ovalbumin (OVA) as a model antigen was readily absorbed by positively charged DOTAP and showed a quick release in vitro. Our results demonstrated that this novel nanovaccine enhanced cellular uptake, cytokine production, and maturation of DCs. Compared with the quick metabolism of free OVA-agonists, the depot effect of OVA-IMNPs was observed, whereas MAN-OVA-IMNPs promoted trafficking to secondary lymphoid organs. After immunization with a subcutaneous injection, the nanovaccine, especially MAN-OVA-IMNPs, induced more antigen-specific CD8+ T cells, greater lymphocyte activation, stronger cross-presentation, and more generation of memory T cells, antibody, IFN-γ, and granzyme B. Prophylactic vaccination of MAN-OVA-IMNPs significantly delayed tumor development and prolonged the survival in mice. The therapeutic tumor challenge indicated that MAN-OVA-IMNPs prohibited tumor progression more efficiently than other formulations, and the combination with an immune checkpoint blockade further enhanced antitumor effects. Hence, the DC-targeted vaccine codelivery with IMQ and MPLA adjuvants by hybrid cationic nanoparticles in a spatiotemporal manner is a promising multifunctional antigen delivery system in cancer immunotherapy.
Collapse
Affiliation(s)
- Linhua Zhang
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| | - Shengjie Wu
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| | - Yu Qin
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| | - Fan Fan
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| | - Zhiming Zhang
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| | - Chenlu Huang
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| | - Weihang Ji
- Department of Biomedical Engineering , University of Minnesota , 7-116 Hasselmo Hall, 312 Church Street SE , Minneapolis , Minnesota 55455 , United States
| | - Lu Lu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine , Tianjin 300192 , China
| | - Chun Wang
- Department of Biomedical Engineering , University of Minnesota , 7-116 Hasselmo Hall, 312 Church Street SE , Minneapolis , Minnesota 55455 , United States
| | - Hongfan Sun
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| | - Xigang Leng
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| | - Deling Kong
- The Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences , Nankai University , Tianjin 300071 , China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute , Xuzhou Medical University , Xuzhou 221004 , Jiangsu , China
| | - Dunwan Zhu
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , China
| |
Collapse
|
17
|
Song H, Yang P, Huang P, Zhang C, Kong D, Wang W. Injectable polypeptide hydrogel-based co-delivery of vaccine and immune checkpoint inhibitors improves tumor immunotherapy. Theranostics 2019; 9:2299-2314. [PMID: 31149045 PMCID: PMC6531311 DOI: 10.7150/thno.30577] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/24/2019] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy, an attractive option for cancer treatment, necessitates the direct stimulation of immune cells in vivo and the simultaneous effective inhibition of immunosuppressive tumor microenvironments. Methods: In the present study, we developed an injectable PEG-b-poly(L-alanine) hydrogel for co-delivery of a tumor vaccine and dual immune checkpoint inhibitors to increase tumor immunotherapy efficacy. Tumor cell lysates, granulocyte-macrophage colony stimulating factor (GM-CSF), and immune checkpoint inhibitors (anti-CTLA-4/PD-1 antibody) were readily encapsulated in the porous hydrogel during the spontaneous self-assembly of polypeptide in aqueous solution. Results: Sustained release of tumor antigens and GM-CSF persistently recruited and activated dendritic cells (DCs) and induced a strong T-cell response in vivo, which was further enhanced by the immune checkpoint therapy. The hydrogel vaccine also upregulated the production of IgG and the secretion of cytokines including IFN-γ, IL-4, and TNF-α. Importantly, the hydrogel-based combination therapy had superior immunotherapy effects against melanoma and 4T-1 tumor in comparison with the vaccine alone or in addition with a single immune checkpoint blockade. In studying the underlying mechanism, we found that the hydrogel-based combinatorial immunotherapy not only significantly increased the activated effector CD8+ T cells within the spleens and tumors of vaccinated mice, but also reduced the ratio of Tregs. Conclusion: Our findings indicate that the polypeptide hydrogel can be used as an effective sustained delivery platform for vaccines and immune checkpoint inhibitors, providing an advanced combinatorial immunotherapy approach for cancer treatment.
Collapse
|
18
|
Kelly HG, Kent SJ, Wheatley AK. Immunological basis for enhanced immunity of nanoparticle vaccines. Expert Rev Vaccines 2019; 18:269-280. [PMID: 30707635 DOI: 10.1080/14760584.2019.1578216] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Immunization has been a remarkably successful public health intervention; however, new approaches to vaccine design are essential to counter existing and emerging infectious diseases which have defied traditional vaccination efforts to date. Nanoparticles (ordered structures with dimensions in the range of 1-1000 nm) have great potential to supplement traditional vaccines based upon pathogen subunits, or killed or attenuated microorganisms, as exemplified by the successful licensure of virus-like particle vaccines for human papillomavirus and hepatitis B. However, the immunological mechanisms that underpin the potent immunity of nanoparticle vaccines are poorly defined. AREAS COVERED Here, we review the immunity of nanoparticle immunization. The display of antigen in a repetitive, ordered array mimics the surface of a pathogen, as does their nanoscale size. These properties facilitate enhanced innate immune activation, improved drainage and retention in lymph nodes, stronger engagement with B cell receptors, and augmented T cell help in driving B cell activation. EXPERT OPINION In the near future, increasingly complex nanoparticle vaccines displaying multiple antigens and/or co-delivered adjuvants will reach clinical trials. An improved mechanistic understanding of nanoparticle vaccination will ultimately facilitate the rational design of improved vaccines for human health.
Collapse
Affiliation(s)
- Hannah G Kelly
- a Department of Microbiology and Immunology , University of Melbourne, at The Peter Doherty Institute for Infection and Immunity , Melbourne , Australia.,b ARC Centre for Excellence in Convergent Bio-Nano Science and Technology , University of Melbourne , Parkville , Australia
| | - Stephen J Kent
- a Department of Microbiology and Immunology , University of Melbourne, at The Peter Doherty Institute for Infection and Immunity , Melbourne , Australia.,b ARC Centre for Excellence in Convergent Bio-Nano Science and Technology , University of Melbourne , Parkville , Australia.,c Melbourne Sexual Health Centre and Department of Infectious Diseases , Alfred Hospital and Central Clinical School, Monash University , Melbourne , Australia
| | - Adam K Wheatley
- a Department of Microbiology and Immunology , University of Melbourne, at The Peter Doherty Institute for Infection and Immunity , Melbourne , Australia.,b ARC Centre for Excellence in Convergent Bio-Nano Science and Technology , University of Melbourne , Parkville , Australia
| |
Collapse
|
19
|
Lee ES, Shin JM, Son S, Ko H, Um W, Song SH, Lee JA, Park JH. Recent Advances in Polymeric Nanomedicines for Cancer Immunotherapy. Adv Healthc Mater 2019; 8:e1801320. [PMID: 30666822 DOI: 10.1002/adhm.201801320] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 12/08/2018] [Indexed: 12/20/2022]
Abstract
Immunotherapy has emerged as a promising approach to treat cancer, since it facilitates eradication of cancer by enhancing innate and/or adaptive immunity without using cytotoxic drugs. Of the immunotherapeutic approaches, significant clinical potentials are shown in cancer vaccination, immune checkpoint therapy, and adoptive cell transfer. Nevertheless, conventional immunotherapies often involve immune-related adverse effects, such as liver dysfunction, hypophysitis, type I diabetes, and neuropathy. In an attempt to address these issues, polymeric nanomedicines are extensively investigated in recent years. In this review, recent advances in polymeric nanomedicines for cancer immunotherapy are highlighted and thoroughly discussed in terms of 1) antigen presentation, 2) activation of antigen-presenting cells and T cells, and 3) promotion of effector cells. Also, the future perspectives to develop ideal nanomedicines for cancer immunotherapy are provided.
Collapse
Affiliation(s)
- Eun Sook Lee
- Department of Health Sciences and Technology; SAIHST; Sungkyunkwan University; Suwon 16419 Republic of Korea
| | - Jung Min Shin
- School of Chemical Engineering; College of Engineering; Sungkyunkwan University; Suwon 16419 Republic of Korea
| | - Soyoung Son
- Department of Health Sciences and Technology; SAIHST; Sungkyunkwan University; Suwon 16419 Republic of Korea
| | - Hyewon Ko
- Department of Health Sciences and Technology; SAIHST; Sungkyunkwan University; Suwon 16419 Republic of Korea
| | - Wooram Um
- Department of Health Sciences and Technology; SAIHST; Sungkyunkwan University; Suwon 16419 Republic of Korea
| | - Seok Ho Song
- School of Chemical Engineering; College of Engineering; Sungkyunkwan University; Suwon 16419 Republic of Korea
| | - Jae Ah Lee
- School of Chemical Engineering; College of Engineering; Sungkyunkwan University; Suwon 16419 Republic of Korea
| | - Jae Hyung Park
- Department of Health Sciences and Technology; SAIHST; Sungkyunkwan University; Suwon 16419 Republic of Korea
- School of Chemical Engineering; College of Engineering; Sungkyunkwan University; Suwon 16419 Republic of Korea
| |
Collapse
|
20
|
Dong X, Liang J, Yang A, Qian Z, Kong D, Lv F. A Visible Codelivery Nanovaccine of Antigen and Adjuvant with Self-Carrier for Cancer Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2019; 11:4876-4888. [PMID: 30628437 DOI: 10.1021/acsami.8b20364] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Codelivery nanovaccines of antigens and adjuvants have achieved positive therapy for cancer immunotherapy. The insufficient immunogenicity of these vaccines leads to the difficulty of eliciting robust immune effects for immune clearance due to the inadequate loading efficiency, complex preparation processes, low safety concerns, and weak immune responses. Herein, a visible codelivery nanovaccine of an antigen and adjuvant based on self-cross-linked antigen nanoparticles (ovalbumin nanoparticles (ONPs)) combined with the adjuvant (CpG) for cancer immunotherapy was prepared using antigens themselves as carriers. ONPs not only provide sufficient antigens for continuous simulation of the immune response with high antigen loading efficiency but also serve as natural carriers of CpG. In vitro and in vivo experiments proved that ONPs-CpG can elicit a robust immune response including DC maturity, T cell activation, and IFN-γ production. ONPs-CpG induced strong tumor-specific immunity and exhibited remarkable antitumor immunotherapy effects in vivo using mouse models of lymphoma. Furthermore, to perform the precise vaccine delivery, the dual fluorescent codelivery nanovaccine was monitored in real time in vivo by the visible imaging method. With regard to migration tracking, fluorescence imaging allowed for both high resolution and sensitivity of visible detection based on the fluorescence of ONPs and CpG. The multifunctional nanovaccine could function as a robust platform for cancer immunotherapy and a visible system for antigen-adjuvant tracking.
Collapse
Affiliation(s)
- Xia Dong
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , PR China
| | - Jie Liang
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , PR China
| | - Afeng Yang
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , PR China
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy, West China Hospital, and Collaborative Innovation Center of Biotherapy , Sichuan University , Chengdu 610041 , Sichuan , PR China
| | - Deling Kong
- Tianjin Key Laboratory of Biomedical Materials , Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , PR China
| | | |
Collapse
|
21
|
Al-Halifa S, Gauthier L, Arpin D, Bourgault S, Archambault D. Nanoparticle-Based Vaccines Against Respiratory Viruses. Front Immunol 2019; 10:22. [PMID: 30733717 PMCID: PMC6353795 DOI: 10.3389/fimmu.2019.00022] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/07/2019] [Indexed: 12/29/2022] Open
Abstract
The respiratory mucosa is the primary portal of entry for numerous viruses such as the respiratory syncytial virus, the influenza virus and the parainfluenza virus. These pathogens initially infect the upper respiratory tract and then reach the lower respiratory tract, leading to diseases. Vaccination is an affordable way to control the pathogenicity of viruses and constitutes the strategy of choice to fight against infections, including those leading to pulmonary diseases. Conventional vaccines based on live-attenuated pathogens present a risk of reversion to pathogenic virulence while inactivated pathogen vaccines often lead to a weak immune response. Subunit vaccines were developed to overcome these issues. However, these vaccines may suffer from a limited immunogenicity and, in most cases, the protection induced is only partial. A new generation of vaccines based on nanoparticles has shown great potential to address most of the limitations of conventional and subunit vaccines. This is due to recent advances in chemical and biological engineering, which allow the design of nanoparticles with a precise control over the size, shape, functionality and surface properties, leading to enhanced antigen presentation and strong immunogenicity. This short review provides an overview of the advantages associated with the use of nanoparticles as vaccine delivery platforms to immunize against respiratory viruses and highlights relevant examples demonstrating their potential as safe, effective and affordable vaccines.
Collapse
Affiliation(s)
- Soultan Al-Halifa
- Département de Chimie, Université du Québec à Montréal, Montreal, QC, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Quebec, QC, Canada
| | - Laurie Gauthier
- Département de Chimie, Université du Québec à Montréal, Montreal, QC, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Quebec, QC, Canada
- Département des Sciences Biologiques, Université du Québec à Montréal, Montreal, QC, Canada
- Faculté de Médecine Vétérinaire, Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Université de Montréal, St-Hyacinthe, QC, Canada
| | - Dominic Arpin
- Département de Chimie, Université du Québec à Montréal, Montreal, QC, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Quebec, QC, Canada
- Département des Sciences Biologiques, Université du Québec à Montréal, Montreal, QC, Canada
- Faculté de Médecine Vétérinaire, Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Université de Montréal, St-Hyacinthe, QC, Canada
| | - Steve Bourgault
- Département de Chimie, Université du Québec à Montréal, Montreal, QC, Canada
- Quebec Network for Research on Protein Function, Engineering and Applications, PROTEO, Quebec, QC, Canada
- Faculté de Médecine Vétérinaire, Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Université de Montréal, St-Hyacinthe, QC, Canada
| | - Denis Archambault
- Département des Sciences Biologiques, Université du Québec à Montréal, Montreal, QC, Canada
- Faculté de Médecine Vétérinaire, Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Université de Montréal, St-Hyacinthe, QC, Canada
| |
Collapse
|
22
|
Hajizade A, Salmanian AH, Amani J, Ebrahimi F, Arpanaei A. EspA-loaded mesoporous silica nanoparticles can efficiently protect animal model against enterohaemorrhagic E. coli O157: H7. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 46:S1067-S1075. [PMID: 30638077 DOI: 10.1080/21691401.2018.1529676] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In the present study, the application of mesoporous silica nanoparticles (MSNPs) loaded with recombinant EspA protein, an immunogen of enterohaemorrhagic E. coli, was investigated in the case of BALB/c mice immunization against the bacterium. MSNPs of 96.9 ± 15.9 nm in diameter were synthesized using template removing method. The immunization of mice was carried out orally and subcutaneously. Significant immune responses to the antigen were observed for the immunized mice when rEspA-loaded MSNPs were administered in both routes in comparison to that of the antigen formulated using a well-known adjuvant, i.e. Freund's. According to the titretitre of serum IL-4, the most potent humoral responses were observed when the mice were immunized subcutaneously with antigen-loaded MSNPs (244, 36 and 14 ng/dL of IL-4 in the serum of mice immunized subcutaneously or orally by antigen-loaded MSNPs, and subcutaneously by Freund's adjuvant formulated-antigen, respectively). However, the difference in serum IgG and serum IgA was not significant in mice subcutaneously immunized with antigen-loaded MSNPs and mice immunized with Freund's adjuvant formulated-antigen. Finally, the immunized mice were challenged orally by enterohaemorrhagic E. coli cells. The amount of bacterial shedding was significantly reduced in faecesfaeces of the animals immunized by antigen-loaded MSNPs in both subcutaneous and oral routes.
Collapse
Affiliation(s)
- Abbas Hajizade
- a Applied Biotechnology Research Centre , Baqiyatallah University of Medical Sciences , Tehran , Iran
| | - Ali Hatef Salmanian
- b Agriculture Biotechnology Department , National Institute for Genetic Engineering and Biotechnology , Tehran , Iran
| | - Jafar Amani
- c Applied Microbiology Research Center, Systems Biology and Poisonings Institute , Baqiyatallah University of Medical Sciences , Tehran , Iran
| | - Firouz Ebrahimi
- d Biology Research Centre , Imam Hossein University , Tehran , Iran
| | - Ayyoob Arpanaei
- e Department of Industrial and Environmental Biotechnology , National Institute of Genetic Engineering and Biotechnology , Tehran , Iran
| |
Collapse
|
23
|
Kleinovink JW, Mezzanotte L, Zambito G, Fransen MF, Cruz LJ, Verbeek JS, Chan A, Ossendorp F, Löwik C. A Dual-Color Bioluminescence Reporter Mouse for Simultaneous in vivo Imaging of T Cell Localization and Function. Front Immunol 2019; 9:3097. [PMID: 30671062 PMCID: PMC6333049 DOI: 10.3389/fimmu.2018.03097] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 12/13/2018] [Indexed: 12/16/2022] Open
Abstract
Non-invasive imaging technologies to visualize the location and functionality of T cells are of great value in immunology. Here, we describe the design and generation of a transgenic mouse in which all T cells constitutively express green-emitting click-beetle luciferase (CBG99) while expression of the red-emitting firefly luciferase (PpyRE9) is induced by Nuclear Factor of Activated T cells (NFAT) such as during T cell activation, which allows multicolor bioluminescence imaging of T cell location and function. This dual-luciferase mouse, which we named TbiLuc, showed high constitutive luciferase expression in lymphoid organs such as lymph nodes and the spleen. Ex vivo purified CD8+ and CD4+ T cells both constitutively expressed luciferase, whereas B cells showed no detectable signal. We cross-bred TbiLuc mice to T cell receptor-transgenic OT-I mice to obtain luciferase-expressing naïve CD8+ T cells with defined antigen-specificity. TbiLuc*OT-I T cells showed a fully antigen-specific induction of the T cell activation-dependent luciferase. In vaccinated mice, we visualized T cell localization and activation in vaccine-draining lymph nodes with high sensitivity using two distinct luciferase substrates, D-luciferin and CycLuc1, of which the latter specifically reacts with the PpyRE9 enzyme. This dual-luciferase T cell reporter mouse can be applied in many experimental models studying the location and functional state of T cells.
Collapse
Affiliation(s)
- Jan Willem Kleinovink
- Department of Immunohematology and Blood Transfusion, Tumor Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Laura Mezzanotte
- Department of Radiology and Nuclear Medicine, Optical Molecular Imaging, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Molecular Genetics, Erasmus Medical Center Rotterdam, Netherlands
| | - Giorgia Zambito
- Department of Radiology and Nuclear Medicine, Optical Molecular Imaging, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Molecular Genetics, Erasmus Medical Center Rotterdam, Netherlands.,Medres, Cologne, Germany
| | - Marieke F Fransen
- Department of Immunohematology and Blood Transfusion, Tumor Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Luis J Cruz
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - J Sjef Verbeek
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Alan Chan
- Percuros B.V., Enschede, Netherlands
| | - Ferry Ossendorp
- Department of Immunohematology and Blood Transfusion, Tumor Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Clemens Löwik
- Department of Radiology and Nuclear Medicine, Optical Molecular Imaging, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Molecular Genetics, Erasmus Medical Center Rotterdam, Netherlands.,Department of Oncology, CHUV Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
24
|
Recent advances in applying nanotechnologies for cancer immunotherapy. J Control Release 2018; 288:239-263. [PMID: 30223043 DOI: 10.1016/j.jconrel.2018.09.010] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/11/2018] [Accepted: 09/12/2018] [Indexed: 12/13/2022]
Abstract
Cancer immunotherapy aimed at boosting cancer-specific immunoresponses to eradicate tumor cells has evolved as a new treatment modality. Nanoparticles incorporating antigens and immunomodulatory agents can activate immune cells and modulate the tumor microenvironment to enhance anti-tumor immunity. The nanotechnology approach has been demonstrated to be superior to standard formulations in in-vivo settings. In this article, we focus on recent advances made within the last 5 years in nanoparticle-based cancer immunotherapy, including peptide- and nucleic acid-based nanovaccines, nanomedicines containing an immunoadjuvant to activate anti-tumor immunity, nanoparticle delivery of immune checkpoint inhibitors and the combination of the above approaches. Encouraging results and new emerging nanotechnologies in drug delivery promise the continuous growth of this field and ultimately clinical translation of enhanced immunotherapy of cancer.
Collapse
|
25
|
Post-PEGylated and crosslinked polymeric ssRNA nanocomplexes as adjuvants targeting lymph nodes with increased cytolytic T cell inducing properties. J Control Release 2018; 284:73-83. [DOI: 10.1016/j.jconrel.2018.06.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/04/2018] [Accepted: 06/05/2018] [Indexed: 01/04/2023]
|
26
|
Dong X, Liang J, Yang A, Wang C, Kong D, Lv F. In Vivo Imaging Tracking and Immune Responses to Nanovaccines Involving Combined Antigen Nanoparticles with a Programmed Delivery. ACS APPLIED MATERIALS & INTERFACES 2018; 10:21861-21875. [PMID: 29901978 DOI: 10.1021/acsami.8b04867] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Combined nanovaccine can generate robust and persistent antigen-specific immune responses. A combined nanovaccine was developed based on antigen-loaded genipin-cross-linked-polyethyleneimine-antigen nanoparticles and in vivo multispectral fluorescence imaging tracked the antigen delivery of combined nanovaccine. The inner layer antigen nanoparticles carried abundant antigens by self-cross-linking for persistent immune response, whereas the outer antigen on the surface of antigen nanoparticles provided the initial antigen exposure. The delivery of combined nanovaccine was tracked dynamically and objectively by the separation of inner genipin cross-linked antigen nanoparticle and the outer fluorescent antigen. The immune responses of the combined nanovaccine were evaluated including antigen-specific CD4+ and CD8+ T-cell responses, IgG antibody level, immunological memory, and CD8+ cytotoxic T lymphocyte responses. The results indicated that the inner and outer antigens of combined vaccine can be tracked in real time with a programmed delivery by the dual fluorescence imaging. The programmed delivery of the inner and outer antigens induced strong immune responses with a combination of a quick delivery and a persistent delivery. With adequate antigen exposure, the dendritic cells were effectively activated and matured, and following T cells were further activated for immune response. Compared with a single nanoparticle formulation, the combined nanovaccine exactly elicited a stronger antigen-specific immune response.
Collapse
Affiliation(s)
- Xia Dong
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering , Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , P. R. China
| | - Jie Liang
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering , Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , P. R. China
| | - Afeng Yang
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering , Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , P. R. China
| | - Chun Wang
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering , Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , P. R. China
- Department of Biomedical Engineering , University of Minnesota , Minneapolis , Minnesota 55455 , United States
| | - Deling Kong
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering , Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , P. R. China
| | - Feng Lv
- Tianjin Key Laboratory of Biomedical Materials, Institute of Biomedical Engineering , Chinese Academy of Medical Sciences & Peking Union Medical College , Tianjin 300192 , P. R. China
| |
Collapse
|
27
|
Gao M, Peng Y, Jiang L, Qiu L. Effective intracellular delivery and Th1 immune response induced by ovalbumin loaded in pH-responsive polyphosphazene polymersomes. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1609-1618. [DOI: 10.1016/j.nano.2018.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 03/13/2018] [Accepted: 04/02/2018] [Indexed: 10/17/2022]
|
28
|
Dong X, Sun Z, Liang J, Wang H, Zhu D, Leng X, Wang C, Kong D, Lv F. A visible fluorescent nanovaccine based on functional genipin crosslinked ovalbumin protein nanoparticles. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1087-1098. [DOI: 10.1016/j.nano.2018.02.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/29/2018] [Accepted: 02/10/2018] [Indexed: 01/11/2023]
|
29
|
Qiu L, Valente M, Dolen Y, Jäger E, Beest MT, Zheng L, Figdor CG, Verdoes M. Endolysosomal-Escape Nanovaccines through Adjuvant-Induced Tumor Antigen Assembly for Enhanced Effector CD8 + T Cell Activation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1703539. [PMID: 29493121 DOI: 10.1002/smll.201703539] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/16/2018] [Indexed: 06/08/2023]
Abstract
The activation of tumor-specific effector immune cells is key for successful immunotherapy and vaccination is a powerful strategy to induce such adaptive immune responses. However, the generation of effective anticancer vaccines is challenging. To overcome these challenges, a novel straight-forward strategy of adjuvant-induced tumor antigen assembly to generate nanovaccines with superior antigen/adjuvant loading efficiency is developed. To protect nanovaccines in circulation and to introduce additional functionalities, a biocompatible polyphenol coating is installed. The resulting functionalizable nanovaccines are equipped with a pH (low) insertion peptide (pHLIP) to facilitate endolysosomal escape and to promote cytoplasmic localization, with the aim to enhance cross-presentation of the antigen by dendritic cells to effectively activate CD8+ T cell. The results demonstrate that pHLIP-functionalized model nanovaccine can induce endolysosomal escape and enhance CD8+ T cell activation both in vitro and in vivo. Furthermore, based on the adjuvant-induced antigen assembly, nanovaccines of the clinically relevant tumor-associated antigen NY-ESO-1 are generated and show excellent capacity to elicit NY-ESO-1-specific CD8+ T cell activation, demonstrating a high potential of this functionalizable nanovaccine formulation strategy for clinical applications.
Collapse
Affiliation(s)
- Liping Qiu
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
- Department of Tumor Immunology, Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Michael Valente
- Department of Tumor Immunology, Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Yusuf Dolen
- Department of Tumor Immunology, Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Eliezer Jäger
- Department of Tumor Immunology, Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
- Institute of Macromolecular Chemistry v.v.i., Academy of Sciences of the Czech Republic, Heyrovsky Sq. 2, 162 06, Prague 6, Czech Republic
| | - Martin Ter Beest
- Department of Tumor Immunology, Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Liyan Zheng
- Molecular Science and Biomedicine Laboratory, State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Carl G Figdor
- Department of Tumor Immunology, Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| | - Martijn Verdoes
- Department of Tumor Immunology, Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|
30
|
Song H, Huang P, Niu J, Shi G, Zhang C, Kong D, Wang W. Injectable polypeptide hydrogel for dual-delivery of antigen and TLR3 agonist to modulate dendritic cells in vivo and enhance potent cytotoxic T-lymphocyte response against melanoma. Biomaterials 2018; 159:119-129. [DOI: 10.1016/j.biomaterials.2018.01.004] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 10/13/2017] [Accepted: 01/01/2018] [Indexed: 12/27/2022]
|
31
|
Riehl M, Harms M, Lucas H, Ebensen T, Guzmán CA, Mäder K. Dual dye in-vivo imaging of differentially charged PLGA carriers reveals antigen-depot effect, leading to improved immune responses in preclinical models. Eur J Pharm Sci 2018; 117:88-97. [PMID: 29408551 DOI: 10.1016/j.ejps.2018.01.040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/23/2018] [Accepted: 01/25/2018] [Indexed: 12/17/2022]
Abstract
The present in-vivo study investigated the behavior and performance of differently charged poly(lactic‑co‑glycolic) acid microparticles (PLGA MP) as vaccination platform. For this purpose, particles loaded with ovalbumin (OVA) as model antigen were subcutaneously (s.c.) injected in SKH1 mice. The utilized SKH1 hairless mice exhibit a fully operative immune system and allow parallel imaging investigations due to the lack of hair. Usage of this species enabled the combination of two investigations within a single study protocol, namely noninvasive in-vivo imaging and immune responses directed towards the antigen. All treatments were well tolerated, no safety drop-outs occurred. The fate of the model antigen OVA as well as the PLGA particles was monitored using a dual dye approach (CF660C & DiR) by multispectral fluorescence imaging (msFI). A depot effect for the OVA antigen adsorbed to the MP surface could be observed for the positively charged MPs. The immune response against OVA was then analyzed. OVA alone did not induce an immune response, whereas the positively charged as well as the neutral MP induced a strong and consistent humoral immune response with a clear favor of IgG1 over IgG2a subclass antibodies. In contrast, negatively charged MP were not able to induce measurable antibody responses. Cellular immune response was weak and inconsistent for all treated groups, which verifies previous in-vitro results conducted with the herein described microparticulate antigen platform. In conclusion, the characterization of the in-vivo performance yielded valuable information about antigen and carrier fate after application. The presented adjuvant platform is capable of inducing strong TH2 dominated immune responses characterized by enhanced IgG1 subclass titers which are critical for vaccines aimed at promoting induction of neutralizing antibodies.
Collapse
Affiliation(s)
- Markus Riehl
- Institute of Pharmacy, Faculty I of Natural Sciences, Martin-Luther-University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle/Saale, Germany; Department Pharmaceutical Technologies, Merck KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Meike Harms
- Department Pharmaceutical Technologies, Merck KGaA, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Henrike Lucas
- Institute of Pharmacy, Faculty I of Natural Sciences, Martin-Luther-University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle/Saale, Germany
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Inhoffenstr 7, 38124 Braunschweig, Germany
| | - Carlos A Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Inhoffenstr 7, 38124 Braunschweig, Germany
| | - Karsten Mäder
- Institute of Pharmacy, Faculty I of Natural Sciences, Martin-Luther-University Halle-Wittenberg, Wolfgang-Langenbeck-Str. 4, 06120 Halle/Saale, Germany.
| |
Collapse
|
32
|
Zanganeh S, Spitler R, Hutter G, Ho JQ, Pauliah M, Mahmoudi M. Tumor-associated macrophages, nanomedicine and imaging: the axis of success in the future of cancer immunotherapy. Immunotherapy 2017; 9:819-835. [DOI: 10.2217/imt-2017-0041] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The success of any given cancer immunotherapy relies on several key factors. In particular, success hinges on the ability to stimulate the immune system in a controlled and precise fashion, select the best treatment options and appropriate therapeutic agents, and use highly effective tools to accurately and efficiently assess the outcome of the immunotherapeutic intervention. Furthermore, a deep understanding and effective utilization of tumor-associated macrophages (TAMs), nanomedicine and biomedical imaging must be harmonized to improve treatment efficacy. Additionally, a keen appreciation of the dynamic interplay that occurs between immune cells and the tumor microenvironment (TME) is also essential. New advances toward the modulation of the immune TME have led to many novel translational research approaches focusing on the targeting of TAMs, enhanced drug and nucleic acid delivery, and the development of theranostic probes and nanoparticles for clinical trials. In this review, we discuss the key cogitations that influence TME, TAM modulations and immunotherapy in solid tumors as well as the methods and resources of tracking the tumor response. The vast array of current nanomedicine technologies can be readily modified to modulate immune function, target specific cell types, deliver therapeutic payloads and be monitored using several different imaging modalities. This allows for the development of more effective treatments, which can be specifically designed for particular types of cancer or on an individual basis. Our current capacities have allowed for greater use of theranostic probes and multimodal imaging strategies that have led to better image contrast, real-time imaging capabilities leveraging targeting moieties, tracer kinetics and enabling more detailed response profiles at the cellular and molecular levels. These novel capabilities along with new discoveries in cancer biology should drive innovation for improved biomarkers for efficient and individualized cancer therapy.
Collapse
Affiliation(s)
- Saeid Zanganeh
- Department of Radiology, Memorial Sloan Kettering, New York, NY 10065, USA
| | - Ryan Spitler
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Gregor Hutter
- Department of Neurosurgery, Stanford University, Stanford, CA 94304, USA
| | - Jim Q Ho
- Department of Biology, Middlebury College, Middlebury, VT 05753, USA
| | - Mohan Pauliah
- Department of Radiology, Memorial Sloan Kettering, New York, NY 10065, USA
| | - Morteza Mahmoudi
- Department of Nanotechnology, Nanotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, 14155–6451, Iran
- Department of Anesthesiology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
33
|
Nanoparticles for tumor immunotherapy. Eur J Pharm Biopharm 2017; 115:243-256. [DOI: 10.1016/j.ejpb.2017.03.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Revised: 03/01/2017] [Accepted: 03/17/2017] [Indexed: 12/21/2022]
|
34
|
Rad-Malekshahi M, Fransen MF, Krawczyk M, Mansourian M, Bourajjaj M, Chen J, Ossendorp F, Hennink WE, Mastrobattista E, Amidi M. Self-Assembling Peptide Epitopes as Novel Platform for Anticancer Vaccination. Mol Pharm 2017; 14:1482-1493. [PMID: 28088862 PMCID: PMC5415879 DOI: 10.1021/acs.molpharmaceut.6b01003] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 12/29/2016] [Accepted: 01/14/2017] [Indexed: 12/17/2022]
Abstract
The aim of the present study was to improve the immunogenicity of peptide epitope vaccines using novel nanocarriers based on self-assembling materials. Several studies demonstrated that peptide antigens in nanoparticulate form induce stronger immune responses than their soluble forms. However, several issues such as poor loading and risk of inducing T cell anergy due to premature release of antigenic epitopes have challenged the clinical success of such systems. In the present study, we developed two vaccine delivery systems by appending a self-assembling peptide (Ac-AAVVLLLW-COOH) or a thermosensitive polymer poly(N-isopropylacrylamide (pNIPAm) to the N-terminus of different peptide antigens (OVA250-264, HPV-E743-57) to generate self-assembling peptide epitopes (SAPEs). The obtained results showed that the SAPEs were able to form nanostructures with a diameter from 20 to 200 nm. The SAPEs adjuvanted with CpG induced and expanded antigen-specific CD8+ T cells in mice. Furthermore, tumor-bearing mice vaccinated with SAPEs harboring the HPV E743-57 peptide showed a delayed tumor growth and an increased survival compared to sham-treated mice. In conclusion, self-assembling peptide based systems increase the immunogenicity of peptide epitope vaccines and therefore warrants further development toward clinical use.
Collapse
Affiliation(s)
- Mazda Rad-Malekshahi
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Department
of Pharmaceutical Biomaterials and Medical Biomaterials Research Center,
Faculty of Pharmacy, Tehran University of
Medical Sciences, Tehran, Iran
| | - Marieke F. Fransen
- Department
of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Małgorzata Krawczyk
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Mercedeh Mansourian
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Meriem Bourajjaj
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jian Chen
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Ferry Ossendorp
- Department
of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Wim E. Hennink
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Enrico Mastrobattista
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Maryam Amidi
- Department
of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
35
|
Zhang C, Shi G, Zhang J, Song H, Niu J, Shi S, Huang P, Wang Y, Wang W, Li C, Kong D. Targeted antigen delivery to dendritic cell via functionalized alginate nanoparticles for cancer immunotherapy. J Control Release 2017; 256:170-181. [PMID: 28414151 DOI: 10.1016/j.jconrel.2017.04.020] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 03/17/2017] [Accepted: 04/12/2017] [Indexed: 01/05/2023]
Abstract
The purpose of the present study was to identify an "easy-to-adopt" strategy to enhance immune responses using functionalized alginate (ALG) nanoparticles (MAN-ALG/ALG=OVA NPs), which were prepared by CaCl2 cross-linking of two different types of ALG. The mannose (MAN) modified ALG (MAN-ALG) was used for dendritic cell targeting. The other component, composed of ovalbumin (OVA), a model antigen, is conjugated to ALG (ALG=OVA) via pH sensitive Schiff base bond. Grafting of alginate was demonstrated by FT-IR and 1H NMR, while the morphological structure, particle size, Zeta potential of MAN-ALG/ALG=OVA NPs were measured using TEM and DLS. The OVA releasing behavior of MAN-ALG/ALG=OVA NPs was determined as a function of pH. Antigen uptake was examined by flow cytometry and confocal laser scanning microscopy in vitro using mouse bone marrow dendritic cells (BMDCs). The results showed that MAN-ALG/ALG=OVA NPs facilitated antigen uptake of BMDCs and cytosolic release of the antigen. Significant up-regulation of cytokine secretion and expression levels of the surface co-stimulatory molecules were also observed in MAN-ALG/ALG=OVA NPs-treated BMDCs, compared to free OVA. In vivo bio-distribution study using Cy7 (a near-infrared fluorescence dye) labeled MAN-ALG/ALG=OVA NPs showed efficient in vivo trafficking of the nanoparticles from the injection site to the draining lymph nodes. Moreover, MAN-ALG/ALG=OVA NPs were found to enhance cross-presentation of OVA to B3Z T cell hybridoma in vitro. Subcutaneous administration of MAN-ALG/ALG=OVA NPs also induced major cytotoxic T lymphocytes (CTL) response and inhibition of E.G7 tumor growth in C57BL/6 mice. In summary, we report here that the MAN-ALG/ALG=OVA NPs have the potential as a potent nanovaccine for cancer immunotherapy.
Collapse
Affiliation(s)
- Chuangnian Zhang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China; Key Laboratory of Functional Polymer Materials, Ministry of Education, Nankai University, Tianjin 300071, China
| | - Gaona Shi
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Ju Zhang
- Basic Nursing T&R Section, School of Nursing, Qingdao University, Qingdao, Shandong Province 26000, China
| | - Huijuan Song
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Jinfeng Niu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, 300353, China
| | - Shengbin Shi
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Pingsheng Huang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Yanming Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, 300353, China
| | - Weiwei Wang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China
| | - Chen Li
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China.
| | - Deling Kong
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science & Peking Union Medical College, Tianjin 300192, China; Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, Tianjin 300071, China.
| |
Collapse
|
36
|
Zhao Z, Powers K, Hu Y, Raleigh M, Pentel P, Zhang C. Engineering of a hybrid nanoparticle-based nicotine nanovaccine as a next-generation immunotherapeutic strategy against nicotine addiction: A focus on hapten density. Biomaterials 2017; 123:107-117. [PMID: 28167389 DOI: 10.1016/j.biomaterials.2017.01.038] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Revised: 01/26/2017] [Accepted: 01/27/2017] [Indexed: 12/21/2022]
Abstract
Although vaccination is a promising way to combat nicotine addiction, most traditional hapten-protein conjugate nicotine vaccines only show limited efficacy due to their poor recognition and uptake by immune cells. This study aimed to develop a hybrid nanoparticle-based nicotine vaccine with improved efficacy. The focus was to study the impact of hapten density on the immunological efficacy of the proposed hybrid nanovaccine. It was shown that the nanovaccine nanoparticles were taken up by the dendritic cells more efficiently than the conjugate vaccine, regardless of the hapten density on the nanoparticles. At a similar hapten density, the nanovaccine induced a significantly stronger immune response against nicotine than the conjugate vaccine in mice. Moreover, the high- and medium-density nanovaccines resulted in significantly higher anti-nicotine antibody titers than their low-density counterpart. Specifically, the high-density nanovaccine exhibited better immunogenic efficacy, resulting in higher anti-nicotine antibody titers and lower anti-carrier protein antibody titers than the medium- and low-density versions. The high-density nanovaccine also had the best ability to retain nicotine in serum and to block nicotine from entering the brain. These results suggest that the hybrid nanoparticle-based nicotine vaccine can elicit strong immunogenicity by modulating the hapten density, thereby providing a promising next-generation immunotherapeutic strategy against nicotine addiction.
Collapse
Affiliation(s)
- Zongmin Zhao
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA 24061, United States
| | - Kristen Powers
- Department of Biological Science, Virginia Tech, Blacksburg, VA 24061, United States
| | - Yun Hu
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA 24061, United States
| | - Michael Raleigh
- Minneapolis Medical Research Foundation, Minneapolis, MN 55404, United States
| | - Paul Pentel
- Minneapolis Medical Research Foundation, Minneapolis, MN 55404, United States
| | - Chenming Zhang
- Department of Biological Systems Engineering, Virginia Tech, Blacksburg, VA 24061, United States.
| |
Collapse
|
37
|
Kemal E, Abelha TF, Urbano L, Peters R, Owen DM, Howes P, Green M, Dailey LA. Bright, near infrared emitting PLGA–PEG dye-doped CN-PPV nanoparticles for imaging applications. RSC Adv 2017. [DOI: 10.1039/c6ra25004a] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In this publication, we describe the synthesis of near-IR emitting conjugated polymer nanoparticles with an engineered surface, and their use in biological imaging.
Collapse
Affiliation(s)
- Evren Kemal
- King's College London
- Department of Physics
- London
- UK
| | | | - Laura Urbano
- King's College London
- Institute of Pharmaceutical Science
- London
- UK
| | - Ruby Peters
- King's College London
- Department of Physics
- London
- UK
| | | | - P. Howes
- King's College London
- Department of Physics
- London
- UK
| | - Mark Green
- King's College London
- Department of Physics
- London
- UK
| | - Lea Ann Dailey
- King's College London
- Institute of Pharmaceutical Science
- London
- UK
| |
Collapse
|
38
|
Shi GN, Zhang CN, Xu R, Niu JF, Song HJ, Zhang XY, Wang WW, Wang YM, Li C, Wei XQ, Kong DL. Enhanced antitumor immunity by targeting dendritic cells with tumor cell lysate-loaded chitosan nanoparticles vaccine. Biomaterials 2017; 113:191-202. [DOI: 10.1016/j.biomaterials.2016.10.047] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 10/27/2016] [Accepted: 10/28/2016] [Indexed: 12/27/2022]
|
39
|
Abstract
This review focuses on summarizing the existing work about nanomaterial-based cancer immunotherapy in detail.
Collapse
Affiliation(s)
- Lijia Luo
- Key Laboratory of Magnetic Materials and Devices
- CAS & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
- Ningbo
| | - Rui Shu
- University of Chinese Academy of Sciences
- Beijing 100049
- China
- Key Laboratory of Marine Materials and Related Technology
- CAS & Ningbo Institute of Materials Technology and Engineering
| | - Aiguo Wu
- Key Laboratory of Magnetic Materials and Devices
- CAS & Key Laboratory of Additive Manufacturing Materials of Zhejiang Province, & Division of Functional Materials and Nanodevices
- Ningbo Institute of Materials Technology and Engineering
- Chinese Academy of Sciences
- Ningbo
| |
Collapse
|
40
|
Ma Q, Lei K, Ding J, Yu L, Ding J. Design, synthesis and ring-opening polymerization of a new iodinated carbonate monomer: a universal route towards ultrahigh radiopaque aliphatic polycarbonates. Polym Chem 2017. [DOI: 10.1039/c7py01411b] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
A universal route towards ultrahigh radiopaque aliphatic polycarbonates was developed based on a new iodinated carbonate monomer.
Collapse
Affiliation(s)
- Qian Ma
- State Key Laboratory of Molecular Engineering of Polymers
- Department of Macromolecular Science
- Fudan University
- Shanghai 200433
- China
| | - Kewen Lei
- State Key Laboratory of Molecular Engineering of Polymers
- Department of Macromolecular Science
- Fudan University
- Shanghai 200433
- China
| | - Jian Ding
- State Key Laboratory of Molecular Engineering of Polymers
- Department of Macromolecular Science
- Fudan University
- Shanghai 200433
- China
| | - Lin Yu
- State Key Laboratory of Molecular Engineering of Polymers
- Department of Macromolecular Science
- Fudan University
- Shanghai 200433
- China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers
- Department of Macromolecular Science
- Fudan University
- Shanghai 200433
- China
| |
Collapse
|
41
|
Lee SY, Cho HJ. Dopamine-conjugated poly(lactic-co-glycolic acid) nanoparticles for protein delivery to macrophages. J Colloid Interface Sci 2016; 490:391-400. [PMID: 27914338 DOI: 10.1016/j.jcis.2016.11.078] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 11/22/2016] [Accepted: 11/22/2016] [Indexed: 11/19/2022]
Abstract
Poly(lactic-co-glycolic acid)-dopamine (PLGA-D)-based nanoparticles (NPs) were developed for the delivery of protein to macrophages. PLGA-D was synthesized via amide bond formation between the amine group of D and the carboxylic acid group of PLGA. Bovine serum albumin (BSA, model protein) was encapsulated in PLGA NPs and PLGA-D NPs, which had an approximately 200nm mean diameter, <0.2 polydispersity index, and negative zeta potential value. There was no increment in the mean diameters of BSA-loaded NPs after 24h of incubation in biological fluid-simulated media (i.e., aqueous buffer and serum media). The primary, secondary, and tertiary structures of BSA released from the NPs were studied by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), circular dichroism, and fluorescence spectrophotometry; the structural stability of BSA was preserved during its encapsulation in the NPs and release from the NPs. PLGA/BSA NPs and PLGA-D/BSA NPs did not induce serious cytotoxicity in RAW 264.7 cells (mouse macrophage cell line) in an established concentration range. In RAW 264.7 cells, the intracellular accumulation of PLGA-D NPs was 2-fold higher than that of PLGA NPs. All of these findings indicated that PLGA-D NPs are a promising system for delivering proteins to macrophages.
Collapse
Affiliation(s)
- Song Yi Lee
- College of Pharmacy, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea
| | - Hyun-Jong Cho
- College of Pharmacy, Kangwon National University, Chuncheon, Gangwon 24341, Republic of Korea.
| |
Collapse
|
42
|
Chesson CB, Ekpo-Otu S, Endsley JJ, Rudra JS. Biomaterials-Based Vaccination Strategies for the Induction of CD8 +T Cell Responses. ACS Biomater Sci Eng 2016; 3:126-143. [PMID: 33450791 DOI: 10.1021/acsbiomaterials.6b00412] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Natural and synthetic biomaterials are increasingly being used for the development of vaccines and immunotherapies as alternatives to traditional live-attenuated formulations due to their improved safety profiles and no risk of reversion to virulence. Polymeric materials in particular enjoy attention due to the ease of fabrication, control over physicochemical properties, and their wide range of immunogenicity. While the majority of studies focus on inducing protective antibody responses, in recent years, materials-based strategies for the delivery of antigens and immunomodulators to improve CD8+T cell immunity against infectious and non-infectious diseases have gained momentum. Notably, platforms based on polymeric nanoparticles, liposomes, micelles, virus-like particles, self-assembling peptides and peptidomimetics, and multilayer thin films show considerable promise in preclinical studies. In this Review, we first introduce the concepts of CD8+T cell activation, effector and memory functions, and cytotoxic activity, followed by vaccine design for eliciting robust and protective long-lived CD8+T cell immunity. We then discuss different materials-based vaccines developed in the past decade to elicit CD8+T cell responses based on molecular composition or fabrication methods and conclude with a summary and glimpse at the future trends in this area.
Collapse
Affiliation(s)
- Charles B Chesson
- Department of Pharmacology & Toxicology, ‡Department of Microbiology & Immunology, and §Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Shaunte Ekpo-Otu
- Department of Pharmacology & Toxicology, Department of Microbiology & Immunology, and §Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Janice J Endsley
- Department of Pharmacology & Toxicology, Department of Microbiology & Immunology, and Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jai S Rudra
- Department of Pharmacology & Toxicology, Department of Microbiology & Immunology, and Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
43
|
Lyalina T, Zubareva A, Varlamov V, Svirshchevskaya E. Cross-presentation of lactoferrin encapsulated into chitosan-based nanoparticles. Nanobiomedicine (Rij) 2016; 3:1849543516667355. [PMID: 29942386 PMCID: PMC5998264 DOI: 10.1177/1849543516667355] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 08/11/2016] [Indexed: 12/20/2022] Open
Abstract
Induction of CD8+ cytotoxic T-cell response is essential for the protection from intracellular pathogens. It requires major histocompatibility complex class I processing of newly synthesized proteins transported from the cytosolic pathway. Presentation of mature soluble proteins occurs via a cross-presentation (CP) pathway that is much less efficient in the activation of cytotoxic response. Encapsulation of proteins into polymeric nanoparticles (NPs) can modulate the efficacy of antigen CP. In this article, a model antigen lactoferrin (L) was encapsulated into polysaccharide NPs with different physicochemical properties (size, charge, and hydrophobicity) and used as an immunogen. CD8+ or CD4+ associated IgG2a or IgG1 subclasses of L-specific antibodies, respectively, served as a measure of CD8+ versus CD4+ T-cell activation. Among five types of NPs produced, only succinylchitosan–galactomannan (LSG) and succinylchitosan–PEG-chitosan (LSPC) NPs induced a significant IgG2a response. IgG1 production was comparable in all but hydrophobic succinyl-dodecyl-chitosan (LSD) NPs, where it was only marginal. Confocal studies demonstrated that galactomannan-equipped LSG-NPs induced vacuolar type of CP, while positively charged LSPC-NPs were transported mostly via the cytosolic CP pathway.
Collapse
Affiliation(s)
- Tatiana Lyalina
- Enzyme engineering laboratory, Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation
| | - Anastasia Zubareva
- Enzyme engineering laboratory, Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation
| | - Valery Varlamov
- Enzyme engineering laboratory, Institute of Bioengineering, Research Center of Biotechnology of the Russian Academy of Sciences, Moscow, Russian Federation
| | - Elena Svirshchevskaya
- Immunology Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russian Federation
| |
Collapse
|
44
|
Overview on experimental models of interactions between nanoparticles and the immune system. Biomed Pharmacother 2016; 83:1365-1378. [DOI: 10.1016/j.biopha.2016.08.060] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 08/14/2016] [Accepted: 08/24/2016] [Indexed: 01/05/2023] Open
|
45
|
Wen R, Umeano AC, Francis L, Sharma N, Tundup S, Dhar S. Mitochondrion: A Promising Target for Nanoparticle-Based Vaccine Delivery Systems. Vaccines (Basel) 2016; 4:E18. [PMID: 27258316 PMCID: PMC4931635 DOI: 10.3390/vaccines4020018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 03/31/2016] [Accepted: 04/08/2016] [Indexed: 02/07/2023] Open
Abstract
Vaccination is one of the most popular technologies in disease prevention and eradication. It is promising to improve immunization efficiency by using vectors and/or adjuvant delivery systems. Nanoparticle (NP)-based delivery systems have attracted increasing interest due to enhancement of antigen uptake via prevention of vaccine degradation in the biological environment and the intrinsic immune-stimulatory properties of the materials. Mitochondria play paramount roles in cell life and death and are promising targets for vaccine delivery systems to effectively induce immune responses. In this review, we focus on NPs-based delivery systems with surfaces that can be manipulated by using mitochondria targeting moieties for intervention in health and disease.
Collapse
Affiliation(s)
- Ru Wen
- NanoTherapeutics Research Laboratory, Department of Chemistry, University of Georgia, Athens, GA 30602, USA.
| | - Afoma C Umeano
- NanoTherapeutics Research Laboratory, Department of Chemistry, University of Georgia, Athens, GA 30602, USA.
| | - Lily Francis
- NanoTherapeutics Research Laboratory, Department of Chemistry, University of Georgia, Athens, GA 30602, USA.
| | - Nivita Sharma
- NanoTherapeutics Research Laboratory, Department of Chemistry, University of Georgia, Athens, GA 30602, USA.
| | - Smanla Tundup
- School of Medicine, Department of Pulmonary and Critical Care, University of Virginia, Charlottesville, WV 22908, USA.
| | - Shanta Dhar
- NanoTherapeutics Research Laboratory, Department of Chemistry, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
46
|
Vartak A, Sucheck SJ. Recent Advances in Subunit Vaccine Carriers. Vaccines (Basel) 2016; 4:vaccines4020012. [PMID: 27104575 PMCID: PMC4931629 DOI: 10.3390/vaccines4020012] [Citation(s) in RCA: 190] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 03/28/2016] [Accepted: 04/01/2016] [Indexed: 12/11/2022] Open
Abstract
The lower immunogenicity of synthetic subunit antigens, compared to live attenuated vaccines, is being addressed with improved vaccine carriers. Recent reports indicate that the physio-chemical properties of these carriers can be altered to achieve optimal antigen presentation, endosomal escape, particle bio-distribution, and cellular trafficking. The carriers can be modified with various antigens and ligands for dendritic cells targeting. They can also be modified with adjuvants, either covalently or entrapped in the matrix, to improve cellular and humoral immune responses against the antigen. As a result, these multi-functional carrier systems are being explored for use in active immunotherapy against cancer and infectious diseases. Advancing technology, improved analytical methods, and use of computational methodology have also contributed to the development of subunit vaccine carriers. This review details recent breakthroughs in the design of nano-particulate vaccine carriers, including liposomes, polymeric nanoparticles, and inorganic nanoparticles.
Collapse
Affiliation(s)
- Abhishek Vartak
- Department of Chemistry and Biochemistry, The University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA.
| | - Steven J Sucheck
- Department of Chemistry and Biochemistry, The University of Toledo, 2801 West Bancroft Street, Toledo, OH 43606, USA.
| |
Collapse
|
47
|
Diaminosulfide based polymer microparticles as cancer vaccine delivery systems. J Control Release 2015; 220:682-90. [PMID: 26359124 DOI: 10.1016/j.jconrel.2015.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Revised: 09/01/2015] [Accepted: 09/04/2015] [Indexed: 01/18/2023]
Abstract
The aim of the research presented here was to determine the characteristics and immunostimulatory capacity, in vivo, of antigen and adjuvant co-loaded into microparticles made from a novel diaminosulfide polymer, poly(4,4'-trimethylenedipiperdyl sulfide) (PNSN), and to assess their potential as cancer vaccine vectors. PNSN microparticles co-loaded with the antigen, ovalbumin (OVA), and adjuvant, CpG 1826, (PNSN(OVA + CpG)) were fabricated and characterized for size (1.64 μm diameter; PDI=0.62), charge (-23.1 ± 0.3), and loading efficiencies of antigen (7.32 μg/mg particles) and adjuvant (0.95 μg/mg particles). The ability of PNSN(OVA + CpG) to stimulate cellular and humoral immune responses in vivo was compared with other PNSN microparticle formulations as well as with poly(lactic-co-glycolic acid)(PLGA)-based microparticles, co-loaded with OVA and CpG (PLGA(OVA + CpG)), an adenovirus encoding OVA (Ad5-OVA), and OVA delivered with incomplete Freund's adjuvant (IFA(OVA)). In vivo OVA-specific IgG1 responses, after subcutaneous prime/boosts in mice, were similar when PNSN(OVA + CpG) and PLGA(OVA + CpG) were compared and the presence of CpG 1826 within the PNSN microparticles demonstrated significantly improved responses when compared to PNSN microparticles loaded with OVA alone (PNSN(OVA)), plus or minus soluble CpG 1826. Cellular immune responses to all particle-based vaccine formulations ranged from being negligible to modest with PNSN(OVA + CpG) generating the greatest responses, displaying significantly increased levels of OVA-specific CD8+ T lymphocytes compared to controls and IFA(OVA) treated mice. Finally, it was shown that of all vaccination formulations tested PNSN(OVA + CpG) was the most protective against subsequent challenge with an OVA-expressing tumor cell line, E.G7. Thus, microparticles made from poly(diaminosulfide)-based macromolecules possess promising potential as vaccine vectors and, as demonstrated here, may have impact as cancer vaccines in particular.
Collapse
|
48
|
Li X, Yang W, Zou Y, Meng F, Deng C, Zhong Z. Efficacious delivery of protein drugs to prostate cancer cells by PSMA-targeted pH-responsive chimaeric polymersomes. J Control Release 2015; 220:704-14. [PMID: 26348387 DOI: 10.1016/j.jconrel.2015.08.058] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Revised: 08/22/2015] [Accepted: 08/31/2015] [Indexed: 12/31/2022]
Abstract
Protein drugs as one of the most potent biotherapeutics have a tremendous potential in cancer therapy. Their application is, nevertheless, restricted by absence of efficacious, biocompatible, and cancer-targeting nanosystems. In this paper, we report that 2-[3-[5-amino-1-carboxypentyl]-ureido]-pentanedioic acid (Acupa)-decorated pH-responsive chimaeric polymersomes (Acupa-CPs) efficiently deliver therapeutic proteins into prostate cancer cells. Acupa-CPs had a unimodal distribution with average sizes ranging from 157-175 nm depending on amounts of Acupa. They displayed highly efficient loading of both model proteins, bovine serum albumin (BSA) and cytochrome C (CC), affording high protein loading contents of 9.1-24.5 wt.%. The in vitro release results showed that protein release was markedly accelerated at mildly acidic pH due to the hydrolysis of acetal bonds in the vesicular membrane. CLSM and MTT studies demonstrated that CC-loaded Acupa10-CPs mediated efficient delivery of protein drugs into PSMA positive LNCaP cells leading to pronounced antitumor effect, in contrast to their non-targeting counterparts and free CC. Remarkably, granzyme B (GrB)-loaded Acupa10-CPs caused effective apoptosis of LNCaP cells with a low half-maximal inhibitory concentration (IC50) of 1.6 nM. Flow cytometry and CLSM studies using MitoCapture™ revealed obvious depletion of mitochondria membrane potential in LNCaP cells treated with GrB-loaded Acupa10-CPs. The preliminary in vivo experiments showed that Acupa-CPs had a long circulation time with an elimination phase half-life of 3.3h in nude mice. PSMA-targeted, pH-responsive, and chimaeric polymersomes have appeared as efficient protein nanocarriers for targeted prostate cancer therapy.
Collapse
Affiliation(s)
- Xiang Li
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, PR China
| | - Weijing Yang
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, PR China
| | - Yan Zou
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, PR China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, PR China.
| | - Chao Deng
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, PR China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, and Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
49
|
Sánchez Vallecillo MF, Minguito de la Escalera MM, Aguirre MV, Ullio Gamboa GV, Palma SD, González-Cintado L, Chiodetti AL, Soldano G, Morón G, Allemandi DA, Ardavín C, Pistoresi-Palencia MC, Maletto BA. A liquid crystal of ascorbyl palmitate, used as vaccine platform, provides sustained release of antigen and has intrinsic pro-inflammatory and adjuvant activities which are dependent on MyD88 adaptor protein. J Control Release 2015; 214:12-22. [DOI: 10.1016/j.jconrel.2015.07.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 06/25/2015] [Accepted: 07/02/2015] [Indexed: 12/22/2022]
|
50
|
Fan Y, Moon JJ. Nanoparticle Drug Delivery Systems Designed to Improve Cancer Vaccines and Immunotherapy. Vaccines (Basel) 2015; 3:662-85. [PMID: 26350600 PMCID: PMC4586472 DOI: 10.3390/vaccines3030662] [Citation(s) in RCA: 181] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 08/19/2015] [Accepted: 08/20/2015] [Indexed: 12/20/2022] Open
Abstract
Recent studies have demonstrated great therapeutic potential of educating and unleashing our own immune system for cancer treatment. However, there are still major challenges in cancer immunotherapy, including poor immunogenicity of cancer vaccines, off-target side effects of immunotherapeutics, as well as suboptimal outcomes of adoptive T cell transfer-based therapies. Nanomaterials with defined physico-biochemical properties are versatile drug delivery platforms that may address these key technical challenges facing cancer vaccines and immunotherapy. Nanoparticle systems have been shown to improve targeted delivery of tumor antigens and therapeutics against immune checkpoint molecules, amplify immune activation via the use of new stimuli-responsive or immunostimulatory materials, and augment the efficacy of adoptive cell therapies. Here, we review the current state-of-the-art in nanoparticle-based strategies designed to potentiate cancer immunotherapies, including cancer vaccines with subunit antigens (e.g., oncoproteins, mutated neo-antigens, DNA and mRNA antigens) and whole-cell tumor antigens, dendritic cell-based vaccines, artificial antigen-presenting cells, and immunotherapeutics based on immunogenic cell death, immune checkpoint blockade, and adoptive T-cell therapy.
Collapse
Affiliation(s)
- Yuchen Fan
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA.
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI 48109, USA.
- Biointerfaces Institute, University of Michigan, Ann Arbor, MI 48109, USA.
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|