1
|
Chung YH, Steinmetz NF. Metastatic Melanoma Treatment and Prophylaxis with S100A9-Targeting Cowpea Mosaic Virus Nanoparticles. Methods Mol Biol 2025; 2902:13-36. [PMID: 40029594 DOI: 10.1007/978-1-0716-4402-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Metastatic cancer continues to be the main cause of cancer-related death, and new therapies must be continuously researched to eradicate these cancers. Immunotherapy aims to stimulate the patient's own immune system to recognize and eliminate tumors and metastatic sites. A particular powerful approach is the use of immunostimulatory agents to reprogram the tumor microenvironment from "cold" to "hot" to prime systemic antitumor immunity. Plant viruses have been investigated for this purpose because their repetitive coat protein structures with encapsidated nucleic acids render them potent immunostimulatory agents. In particular, the cowpea mosaic virus (CPMV) has been found to be a potent anticancer agent when injected intratumorally. However, metastatic cancers cannot be injected in situ, and therefore a systemically administered CPMV prevention and treatment option that is targeted to S100A9 was developed. S100A9 is an immunostimulatory protein that regulates metastatic cancer seeding and growth, thereby making it an attractive target for both prevention and treatment. Protocols for the production and characterization of S100A9-targeted CPMV nanoparticles are described and in vivo experiments that can be carried out to assess the efficacy of the S100A9-targeted CPMV nanoparticles in the prevention and treatment of metastatic melanoma to the lung are detailed. Finally, instructions for flow cytometry analysis of the innate immune cell composition within the lungs following S100A9-targeted CPMV administration are provided.
Collapse
Affiliation(s)
- Young Hun Chung
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Nicole F Steinmetz
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA.
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA, USA.
- Department of Radiology, University of California, San Diego, La Jolla, CA, USA.
- Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, CA, USA.
- Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
2
|
Jung E, Chung YH, Steinmetz NF. TLR Agonists Delivered by Plant Virus and Bacteriophage Nanoparticles for Cancer Immunotherapy. Bioconjug Chem 2023; 34:1596-1605. [PMID: 37611278 PMCID: PMC10538388 DOI: 10.1021/acs.bioconjchem.3c00271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Toll-like receptors (TLRs) are promising targets in cancer immunotherapy due to their role in activating the immune system; therefore, various small-molecule TLR agonists have been tested in clinical applications. However, the clinical use of TLR agonists is hindered by their non-specific side effects and poor pharmacokinetics. To overcome these limitations, we used plant virus nanoparticles (VNPs) and bacteriophage virus-like particles (VLPs) as drug delivery systems. We conjugated TLR3 or TLR7 agonists to cowpea mosaic virus (CPMV) VNPs, cowpea chlorotic mottle virus (CCMV) VNPs, and bacteriophage Qβ VLPs. The conjugation of TLR7 agonist, 2-methoxyethoxy-8-oxo-9-(4-carboxybenzyl)adenine (1V209), resulted in the potent activation of immune cells and promoted the production of pro-inflammatory cytokine interleukin 6. We found that 1V209 conjugated to CPMV, CCMV, and Qβ reduced tumor growth in vivo and prolonged the survival of mice compared to those treated with free 1V209 or a simple admixture of 1V209 and viral particles. Nucleic acid-based TLR3 agonist, polyinosinic acid with polycytidylic acid (poly(I:C)), was also delivered by CPMV VNPs, resulting in enhanced mice survival. All our data suggest that coupling and co-delivery are required to enhance the anti-tumor efficacy of TLR agonists and simple mixing of the VLPs with the agonists does not confer a survival benefit. The delivery of 1V209 or poly(I:C) conjugated to VNPs/VLPs probably enhances their efficacy due to the multivalent presentation, prolongation of tumor residence time, and targeting of the innate immune cells mediated by the VNP/VLP carrier.
Collapse
Affiliation(s)
- Eunkyeong Jung
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Dr., La Jolla, California 92093, United States
| | - Young Hun Chung
- Department of Bioengineering, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92093, United States
- Moores Cancer Center, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92093, United States
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California, San Diego, 9500 Gilman Dr., La Jolla, California 92093, United States
- Department of Bioengineering, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92093, United States
- Moores Cancer Center, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92093, United States
- Department of Radiology, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92093, United States
- Institute for Materials Discovery and Design, University of California San Diego, 9500 Gilman Dr., La Jolla, California 92093, United States
| |
Collapse
|
3
|
Noureddine A, Maestas-Olguin A, Tang L, Corman-Hijar JI, Olewine M, Krawchuck JA, Tsala Ebode J, Edeh C, Dang C, Negrete OA, Watt J, Howard T, Coker EN, Guo J, Brinker CJ. Future of Mesoporous Silica Nanoparticles in Nanomedicine: Protocol for Reproducible Synthesis, Characterization, Lipid Coating, and Loading of Therapeutics (Chemotherapeutic, Proteins, siRNA and mRNA). ACS NANO 2023; 17:16308-16325. [PMID: 37643407 DOI: 10.1021/acsnano.3c07621] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Owing to their uniform and tunable particle size, pore size, and shape, along with their modular surface chemistry and biocompatibility, mesoporous silica nanoparticles (MSNs) have found extensive applications as nanocarriers to deliver therapeutic, diagnostic and combined "theranostic" cargos to cells and tissues. Although thoroughly investigated, MSN have garnered FDA approval for only one MSN system via oral administration. One possible reason is that there is no recognized, reproducible, and widely adopted MSN synthetic protocol, meaning not all MSNs are created equal in the laboratory nor in the eyes of the FDA. This manuscript provides the sol-gel and MSN research communities a reproducible, fully characterized synthetic protocol to synthesize MSNs and corresponding lipid-coated MSN delivery vehicles with predetermined particle size, pore size, and drug loading and release characteristics. By carefully articulating the step-by-step synthetic procedures and highlighting critical points and troubleshooting, augmented with videos and schematics, this Article will help researchers entering this rapidly expanding field to yield reliable results.
Collapse
Affiliation(s)
- Achraf Noureddine
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Angelea Maestas-Olguin
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Lien Tang
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
- University of New Mexico School of Medicine, Albuquerque, New Mexico 87131, United States
| | - Jim I Corman-Hijar
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
- Biomedical Engineering Department, Pontifical Catholic University of Peru, San Miguel 15088, Peru
| | - Marian Olewine
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Jacob A Krawchuck
- Center for Integrated Nanotechnologies, Sandia National Laboratories, Albuquerque, New Mexico 87123, United States
| | - Johanna Tsala Ebode
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Chuzube Edeh
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Caleb Dang
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Oscar A Negrete
- Systems Biology Department, Sandia National Laboratories, Livermore, California 94550 United States
| | - John Watt
- Center for Integrated Nanotechnologies, Los Alamos National Laboratory, Los Alamos, New Mexico 87545, United States
| | - Tamara Howard
- Department of Cell Biology & Physiology, UNM School of Medicine, Albuquerque, New Mexico 87131, United States
| | - Eric N Coker
- Electronic, Optical, and Nano Materials, Sandia National Laboratories, Albuquerque, New Mexico 87185, United States
| | - Jimin Guo
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
- College of Materials Sciences and Engineering, Beijing University of Chemical Technology, Beijing, 100029 China
| | - C Jeffrey Brinker
- Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131, United States
| |
Collapse
|
4
|
Charbonneau M, Harper K, Brochu-Gaudreau K, Perreault A, Roy LO, Lucien F, Tian S, Fortin D, Dubois CM. The development of a rapid patient-derived xenograft model to predict chemotherapeutic drug sensitivity/resistance in malignant glial tumors. Neuro Oncol 2023; 25:1605-1616. [PMID: 36821432 PMCID: PMC10479744 DOI: 10.1093/neuonc/noad047] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND High-grade gliomas (HGG) are aggressive brain tumors associated with short median patient survival and limited response to therapies, driving the need to develop tools to improve patient outcomes. Patient-derived xenograft (PDX) models, such as mouse PDX, have emerged as potential Avatar platforms for personalized oncology approaches, but the difficulty for some human grafts to grow successfully and the long time required for mice to develop tumors preclude their use for HGG. METHODS We used a rapid and efficient ex-ovo chicken embryo chorioallantoic membrane (CAM) culture system to evaluate the efficacy of oncologic drug options for HGG patients. RESULTS Implantation of fresh glioma tissue fragments from 59 of 60 patients, that include difficult-to-grow IDH-mutated samples, successfully established CAM tumor xenografts within 7 days, with a tumor take rate of 98.3%. These xenografts faithfully recapitulate the histological and molecular characteristics of the primary tumor, and the ability of individual fragments to form tumors was predictive of poor patient prognosis. Treatment of drug-sensitive or drug-resistant xenografts indicates that the CAM-glioma assay enables testing tumor sensitivity to temozolomide and carboplatin at doses consistent with those administered to patients. In a proof-of-concept study involving 14 HGG patients, we observed a correlation of 100% between the CAM xenograft response to temozolomide or carboplatin and the clinical response of patients. CONCLUSION The CAM-glioma model is a fast and reliable assay that has the potential to serve as a complementary model to drug discovery and a real-time Avatar platform to predict the best treatment for HGG patients.
Collapse
Affiliation(s)
- Martine Charbonneau
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Kelly Harper
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Karine Brochu-Gaudreau
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Alexis Perreault
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Laurent-Olivier Roy
- Department of Surgery, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | | | - Shulan Tian
- Division of Biomedical Statistics and Informatics, Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - David Fortin
- Department of Surgery, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| | - Claire M Dubois
- Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, QC J1H 5N4, Canada
| |
Collapse
|
5
|
Jung E, Chung YH, Mao C, Fiering SN, Steinmetz NF. The Potency of Cowpea Mosaic Virus Particles for Cancer In Situ Vaccination Is Unaffected by the Specific Encapsidated Viral RNA. Mol Pharm 2023; 20:3589-3597. [PMID: 37294891 PMCID: PMC10530639 DOI: 10.1021/acs.molpharmaceut.3c00214] [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] [Indexed: 06/11/2023]
Abstract
Plant virus nanoparticles can be used as drug carriers, imaging reagents, vaccine carriers, and immune adjuvants in the formulation of intratumoral in situ cancer vaccines. One example is the cowpea mosaic virus (CPMV), a nonenveloped virus with a bipartite positive-strand RNA genome with each RNA packaged separately into identical protein capsids. Based on differences in their densities, the components carrying RNA-1 (6 kb) denoted as the bottom (B) component or carrying RNA-2 (3.5 kb) denoted as the middle (M) component can be separated from each other and from a top (T) component, which is devoid of any RNA. Previous preclinical mouse studies and canine cancer trials used mixed populations of CPMV (containing B, M, and T components), so it is unclear whether the particle types differ in their efficacies. It is known that the CPMV RNA genome contributes to immunostimulation by activation of TLR7. To determine whether the two RNA genomes that have different sizes and unrelated sequences cause different immune stimulation, we compared the therapeutic efficacies of B and M components and unfractionated CPMV in vitro and in mouse cancer models. We found that separated B and M particles behaved similarly to the mixed CPMV, activating innate immune cells to induce the secretion of pro-inflammatory cytokines such as IFNα, IFNγ, IL-6, and IL-12, while inhibiting immunosuppressive cytokines such as TGF-β and IL-10. In murine models of melanoma and colon cancer, the mixed and separated CPMV particles all significantly reduced tumor growth and prolonged survival with no significant difference. This shows that the specific RNA genomes similarly stimulate the immune system even though B particles have 40% more RNA than M particles; each CPMV particle type can be used as an effective adjuvant against cancer with the same efficacy as native mixed CPMV. From a translational point of view, the use of either B or M component vs the mixed CPMV formulation offers the advantage that separated B or M alone is noninfectious toward plants and thus provides agronomic safety.
Collapse
Affiliation(s)
- Eunkyeong Jung
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Young Hun Chung
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
| | - Chenkai Mao
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, New Hampshire 03755, United States
| | - Steven N Fiering
- Department of Microbiology and Immunology, Dartmouth Geisel School of Medicine, Hanover, New Hampshire 03755, United States
- Dartmouth Cancer Center, Dartmouth Geisel School of Medicine, Hanover, New Hampshire 03755, United States
| | - Nicole F Steinmetz
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
- Department of Bioengineering, University of California San Diego, La Jolla, California 92093, United States
- Department of Radiology, University of California San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, University of California San Diego, La Jolla, California 92093, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California 92093, United States
- Institute for Materials Design and Discovery, University of California San Diego, La Jolla, California 92093, United States
- Center for Engineering in Cancer, Institute for Engineering in Medicine, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
6
|
Ghani MA, Bangar A, Yang Y, Jung E, Sauceda C, Mandt T, Shukla S, Webster NJG, Steinmetz NF, Newton IG. Treatment of Hepatocellular Carcinoma by Multimodal In Situ Vaccination Using Cryoablation and a Plant Virus Immunostimulant. J Vasc Interv Radiol 2023; 34:1247-1257.e8. [PMID: 36997021 PMCID: PMC10829876 DOI: 10.1016/j.jvir.2023.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/08/2023] [Accepted: 03/21/2023] [Indexed: 03/30/2023] Open
Abstract
PURPOSE To test the hypothesis that cryoablation combined with intratumoral immunomodulating nanoparticles from cowpea mosaic virus (CPMV) as an in situ vaccination approach induces systemic antitumoral immunity in a murine model of hepatocellular carcinoma (HCC). MATERIALS AND METHODS Mice with bilateral, subcutaneous RIL-175 cell-derived HCCs were randomized to 4 groups: (a) phosphate-buffered saline (control), (b) cryoablation only (Cryo), (c) CPMV-treated only (CPMV), and (d) cryoablation plus CPMV-treated (Cryo + CPMV) (N = 11-14 per group). Intratumoral CPMV was administered every 3 days for 4 doses, with cryoablation performed on the third day. Contralateral tumors were monitored. Tumor growth and systemic chemokine/cytokine levels were measured. A subset of tumors and spleens were harvested for immunohistochemistry (IHC) and flow cytometry. One- or 2-way analysis of variance was performed for statistical comparisons. A P value of <.05 was used as the threshold for statistical significance. RESULTS At 2 weeks after treatment, the Cryo and CPMV groups, alone or combined, outperformed the control group in the treated tumor; however, the Cryo + CPMV group showed the strongest reduction and lowest variance (1.6-fold ± 0.9 vs 6.3-fold ± 0.5, P < .0001). For the untreated tumor, only Cryo + CPMV significantly reduced tumor growth compared with control (9.2-fold ± 0.9 vs 17.8-fold ± 2.1, P = .01). The Cryo + CPMV group exhibited a transient increase in interleukin-10 and persistently decreased CXCL1. Flow cytometry revealed natural killer cell enrichment in the untreated tumor and increased PD-1 expression in the spleen. Tumor-infiltrating lymphocytes increased in Cryo + CPMV-treated tumors by IHC. CONCLUSIONS Cryoablation and intratumoral CPMV, alone or combined, demonstrated potent efficacy against treated HCC tumors; however, only cryoablation combined with CPMV slowed the growth of untreated tumors, consistent with an abscopal effect.
Collapse
Affiliation(s)
- Mansur A Ghani
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Amandip Bangar
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Yunpeng Yang
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Eunkyeong Jung
- Department of NanoEngineering, University of California San Diego, La Jolla, California
| | - Consuelo Sauceda
- Department of Pharmacology, University of California San Diego, La Jolla, California; Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California
| | - Tyler Mandt
- Department of Radiology, University of California San Diego, La Jolla, California
| | - Sourabh Shukla
- Department of NanoEngineering, University of California San Diego, La Jolla, California
| | - Nicholas J G Webster
- Division of Endocrinology and Metabolism, Department of Medicine, University of California San Diego, La Jolla, California; Moores Cancer Center, University of California San Diego, La Jolla, California; VA San Diego Healthcare System, San Diego, California
| | - Nicole F Steinmetz
- Department of Radiology, University of California San Diego, La Jolla, California; Department of NanoEngineering, University of California San Diego, La Jolla, California; Moores Cancer Center, University of California San Diego, La Jolla, California; Department of Bioengineering, University of California San Diego, La Jolla, California; Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, California; Institute for Materials Discovery and Design, University of California San Diego, La Jolla, California
| | - Isabel G Newton
- Department of Radiology, University of California San Diego, La Jolla, California; VA San Diego Healthcare System, San Diego, California.
| |
Collapse
|
7
|
Tan JS, Jaffar Ali MNB, Gan BK, Tan WS. Next-generation viral nanoparticles for targeted delivery of therapeutics: Fundamentals, methods, biomedical applications, and challenges. Expert Opin Drug Deliv 2023; 20:955-978. [PMID: 37339432 DOI: 10.1080/17425247.2023.2228202] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 06/19/2023] [Indexed: 06/22/2023]
Abstract
INTRODUCTION Viral nanoparticles (VNPs) are virus-based nanocarriers that have been studied extensively and intensively for biomedical applications. However, their clinical translation is relatively low compared to the predominating lipid-based nanoparticles. Therefore, this article describes the fundamentals, challenges, and solutions of the VNP-based platform, which will leverage the development of next-generation VNPs. AREAS COVERED Different types of VNPs and their biomedical applications are reviewed comprehensively. Strategies and approaches for cargo loading and targeted delivery of VNPs are examined thoroughly. The latest developments in controlled release of cargoes from VNPs and their mechanisms are highlighted too. The challenges faced by VNPs in biomedical applications are identified, and solutions are provided to overcome them. EXPERT OPINION In the development of next-generation VNPs for gene therapy, bioimaging and therapeutic deliveries, focus must be given to reduce their immunogenicity, and increase their stability in the circulatory system. Modular virus-like particles (VLPs) which are produced separately from their cargoes or ligands before all the components are coupled can speed up clinical trials and commercialization. In addition, removal of contaminants from VNPs, cargo delivery across the blood brain barrier (BBB), and targeting of VNPs to organelles intracellularly are challenges that will preoccupy researchers in this decade.
Collapse
Affiliation(s)
- Jia Sen Tan
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Muhamad Norizwan Bin Jaffar Ali
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Bee Koon Gan
- Department of Biological Science, Faculty of Science, National University of Singapore, Singapore
| | - Wen Siang Tan
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
8
|
Yuan B, Liu Y, Lv M, Sui Y, Hou S, Yang T, Belhadj Z, Zhou Y, Chang N, Ren Y, Sun C. Virus-like particle-based nanocarriers as an emerging platform for drug delivery. J Drug Target 2023; 31:433-455. [PMID: 36940208 DOI: 10.1080/1061186x.2023.2193358] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
New nanocarrier technologies are emerging, and they have great potential for improving drug delivery, targeting efficiency, and bioavailability. Virus-like particles (VLPs) are natural nanoparticles from animal and plant viruses and bacteriophages. Hence, VLPs present several great advantages, such as morphological uniformity, biocompatibility, reduced toxicity, and easy functionalisation. VLPs can deliver many active ingredients to the target tissue and have great potential as a nanocarrier to overcome the limitations associated with other nanoparticles. This review will focus primarily on the construction and applications of VLPs, particularly as a novel nanocarrier to deliver active ingredients. Herein, the main methods for the construction, purification, and characterisation of VLPs, as well as various VLP-based materials used in delivery systems are summarised. The biological distribution of VLPs in drug delivery, phagocyte-mediated clearance, and toxicity are also discussed.
Collapse
Affiliation(s)
| | - Yang Liu
- School of Pharmaceutical Sciences, Zhengzhou University, No.100, Kexue Avenue, Zhengzhou 450001, China
| | - Meilin Lv
- Harbin Medical University-Daqing, Daqing 163319, China
| | - Yilei Sui
- Harbin Medical University-Daqing, Daqing 163319, China
| | - Shenghua Hou
- Harbin Medical University-Daqing, Daqing 163319, China
| | - Tinghui Yang
- Harbin Medical University-Daqing, Daqing 163319, China
| | - Zakia Belhadj
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yulong Zhou
- College of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Naidan Chang
- Harbin Medical University-Daqing, Daqing 163319, China
| | - Yachao Ren
- Harbin Medical University-Daqing, Daqing 163319, China.,School of Chemistry and Chemical Engineering, Tianjin University of Technology, tianjin, 300000, China
| | | |
Collapse
|
9
|
Pouresmaeil M, Dall'Ara M, Salvato M, Turri V, Ratti C. Cauliflower mosaic virus: Virus-host interactions and its uses in biotechnology and medicine. Virology 2023; 580:112-119. [PMID: 36812696 DOI: 10.1016/j.virol.2023.02.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023]
Abstract
Cauliflower mosaic virus (CaMV) was the first discovered plant virus with genomic DNA that uses reverse transcriptase for replication. The CaMV 35S promoter is a constitutive promoter and thus, an attractive driver of gene expression in plant biotechnology. It is used in most transgenic crops to activate foreign genes which have been artificially inserted into the host plant. In the last century, producing food for the world's population while preserving the environment and human health is the main topic of agriculture. The damage caused by viral diseases has a significant negative economic impact on agriculture, and disease control is based on two strategies: immunization and prevention to contain virus spread, so correct identification of plant viruses is important for disease management. Here, we discuss CaMV from different aspects: taxonomy, structure and genome, host plants and symptoms, transmission and pathogenicity, prevention, control and application in biotechnology as well as in medicine. Also, we calculated the CAI index for three ORFs IV, V, and VI of the CaMV virus in host plants, the results of which can be used in the discussion of gene transfer or antibody production to identify the CaMV.
Collapse
Affiliation(s)
- Mahin Pouresmaeil
- Department of Biotechnology, Faculty of Agriculture, Azarbijan Shahid Madani University, Tabriz, Iran.
| | - Mattia Dall'Ara
- Department of Agricultural and Food Sciences, School of Agriculture and Veterinary Medicine, University of Bologna, 40127, Bologna, Italy
| | - Maria Salvato
- University of Maryland, Department of Veterinary Medicine, College Park, MD, 20742, USA
| | - Valentina Turri
- Healthcare Direction, Istituto Scientifico Romagnolo per Lo Studio e La Cura Dei Tumori, IRCCS, 47014, Meldola, FC, Italy
| | - Claudio Ratti
- Department of Agricultural and Food Sciences, School of Agriculture and Veterinary Medicine, University of Bologna, 40127, Bologna, Italy
| |
Collapse
|
10
|
Fischer D, Fluegen G, Garcia P, Ghaffari-Tabrizi-Wizsy N, Gribaldo L, Huang RYJ, Rasche V, Ribatti D, Rousset X, Pinto MT, Viallet J, Wang Y, Schneider-Stock R. The CAM Model-Q&A with Experts. Cancers (Basel) 2022; 15:cancers15010191. [PMID: 36612187 PMCID: PMC9818221 DOI: 10.3390/cancers15010191] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/20/2022] [Accepted: 12/24/2022] [Indexed: 12/30/2022] Open
Abstract
The chick chorioallantoic membrane (CAM), as an extraembryonic tissue layer generated by the fusion of the chorion with the vascularized allantoic membrane, is easily accessible for manipulation. Indeed, grafting tumor cells on the CAM lets xenografts/ovografts develop in a few days for further investigations. Thus, the CAM model represents an alternative test system that is a simple, fast, and low-cost tool to study tumor growth, drug response, or angiogenesis in vivo. Recently, a new era for the CAM model in immune-oncology-based drug discovery has been opened up. Although there are many advantages offering extraordinary and unique applications in cancer research, it has also disadvantages and limitations. This review will discuss the pros and cons with experts in the field.
Collapse
Affiliation(s)
- Dagmar Fischer
- Division of Pharmaceutical Technology, Department of Chemistry and Pharmacy, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany
| | - Georg Fluegen
- Department of General, Visceral, Thoracic and Pediatric Surgery (A), Medical Faculty, Heinrich-Heine-University, University Hospital Duesseldorf, 40225 Duesseldorf, Germany
| | - Paul Garcia
- Institute for Advanced Biosciences, Research Center Université Grenoble Alpes (UGA)/Inserm U 1209/CNRS 5309, 38700 La Tronche, France
- R&D Department, Inovotion, 38700 La Tronche, France
| | - Nassim Ghaffari-Tabrizi-Wizsy
- SFL Chicken CAM Lab, Department of Immunology, Otto Loewi Research Center, Medical University of Graz, 8010 Graz, Austria
| | - Laura Gribaldo
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy
| | - Ruby Yun-Ju Huang
- School of Medicine, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei 10051, Taiwan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Volker Rasche
- Department of Internal Medicine II, Ulm University Medical Center, 89073 Ulm, Germany
| | - Domenico Ribatti
- Department of Translational Biomedicine and Neurosciences, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | | | - Marta Texeira Pinto
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- Ipatimup—Instituto de Patologia e Imunologia Molecular da Universidade do Porto, 4200-135 Porto, Portugal
| | - Jean Viallet
- R&D Department, Inovotion, 38700 La Tronche, France
| | - Yan Wang
- R&D Department, Inovotion, 38700 La Tronche, France
| | - Regine Schneider-Stock
- Experimental Tumorpathology, Institute of Pathology, Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, 91054 Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), Universitätsklinikum Erlangen, FAU Erlangen-Nürnberg, 94054 Erlangen, Germany
- Correspondence: ; Tel.: +49-9131-8526-069
| |
Collapse
|
11
|
Butler K, Brinker CJ, Leong HS. Bridging the In Vitro to In Vivo gap: Using the Chick Embryo Model to Accelerate Nanoparticle Validation and Qualification for In Vivo studies. ACS NANO 2022; 16:19626-19650. [PMID: 36453753 PMCID: PMC9799072 DOI: 10.1021/acsnano.2c03990] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 10/17/2022] [Indexed: 06/17/2023]
Abstract
We postulate that nanoparticles (NPs) for use in therapeutic applications have largely not realized their clinical potential due to an overall inability to use in vitro results to predict NP performance in vivo. The avian embryo and associated chorioallantoic membrane (CAM) has emerged as an in vivo preclinical model that bridges the gap between in vitro and in vivo, enabling rapid screening of NP behavior under physiologically relevant conditions and providing a rapid, accessible, economical, and more ethical means of qualifying nanoparticles for in vivo use. The CAM is highly vascularized and mimics the diverging/converging vasculature of the liver, spleen, and lungs that serve as nanoparticle traps. Intravital imaging of fluorescently labeled NPs injected into the CAM vasculature enables immediate assessment and quantification of nano-bio interactions at the individual NP scale in any tissue of interest that is perfused with a microvasculature. In this review, we highlight how utilization of the avian embryo and its CAM as a preclinical model can be used to understand NP stability in blood and tissues, extravasation, biocompatibility, and NP distribution over time, thereby serving to identify a subset of NPs with the requisite stability and performance to introduce into rodent models and enabling the development of structure-property relationships and NP optimization without the sacrifice of large populations of mice or other rodents. We then review how the chicken embryo and CAM model systems have been used to accelerate the development of NP delivery and imaging agents by allowing direct visualization of targeted (active) and nontargeted (passive) NP binding, internalization, and cargo delivery to individual cells (of relevance for the treatment of leukemia and metastatic cancer) and cellular ensembles (e.g., cancer xenografts of interest for treatment or imaging of cancer tumors). We conclude by showcasing emerging techniques for the utilization of the CAM in future nano-bio studies.
Collapse
Affiliation(s)
- Kimberly
S. Butler
- Molecular
and Microbiology, Sandia National Laboratories, Albuquerque, New Mexico 87123, United States
| | - C. Jeffrey Brinker
- Department
of Chemical and Biological Engineering and the Comprehensive Cancer
Center, The University of New Mexico, Albuquerque, New Mexico 87131, United States
| | - Hon Sing Leong
- Department
of Medical Biophysics, Faculty of Medicine, University of Toronto, Toronto M5G 1L7, Canada
- Biological
Sciences Platform, Sunnybrook Hospital, Toronto M4N 3M5, Canada
| |
Collapse
|
12
|
Longitudinal bioluminescence imaging to monitor breast tumor growth and treatment response using the chick chorioallantoic membrane model. Sci Rep 2022; 12:17192. [PMID: 36229503 PMCID: PMC9562337 DOI: 10.1038/s41598-022-20854-9] [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: 06/20/2022] [Accepted: 09/20/2022] [Indexed: 01/05/2023] Open
Abstract
The development of successful treatment regimens for breast cancer requires strong pre-clinical data generated in physiologically relevant pre-clinical models. Here we report the development of the chick embryo chorioallantoic membrane (CAM) model to study tumor growth and angiogenesis using breast cancer cell lines. MDA-MB-231 and MCF7 tumor cell lines were engrafted onto the chick embryo CAM to study tumor growth and treatment response. Tumor growth was evaluated through bioluminescence imaging and a significant increase in tumor size and vascularization was found over a 9-day period. We then evaluated the impact of anti-angiogenic drugs, axitinib and bevacizumab, on tumor growth and angiogenesis. Drug treatment significantly reduced tumor vascularization and size. Overall, our findings demonstrate that the chick embryo CAM is a clinically relevant model to monitor therapeutic response in breast cancer and can be used as a platform for drug screening to evaluate not only gross changes in tumor burden but physiological processes such as angiogenesis.
Collapse
|
13
|
Miebach L, Berner J, Bekeschus S. In ovo model in cancer research and tumor immunology. Front Immunol 2022; 13:1006064. [PMID: 36248802 PMCID: PMC9556724 DOI: 10.3389/fimmu.2022.1006064] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/13/2022] [Indexed: 11/15/2022] Open
Abstract
Considering cancer not only as malignant cells on their own but as a complex disease in which tumor cells interact and communicate with their microenvironment has motivated the establishment of clinically relevant 3D models in past years. Technological advances gave rise to novel bioengineered models, improved organoid systems, and microfabrication approaches, increasing scientific importance in preclinical research. Notwithstanding, mammalian in vivo models remain closest to mimic the patient’s situation but are limited by cost, time, and ethical constraints. Herein, the in ovo model bridges the gap as an advanced model for basic and translational cancer research without the need for ethical approval. With the avian embryo being a naturally immunodeficient host, tumor cells and primary tissues can be engrafted on the vascularized chorioallantoic membrane (CAM) with high efficiencies regardless of species-specific restrictions. The extraembryonic membranes are connected to the embryo through a continuous circulatory system, readily accessible for manipulation or longitudinal monitoring of tumor growth, metastasis, angiogenesis, and matrix remodeling. However, its applicability in immunoncological research is largely underexplored. Dual engrafting of malignant and immune cells could provide a platform to study tumor-immune cell interactions in a complex, heterogenic and dynamic microenvironment with high reproducibility. With some caveats to keep in mind, versatile methods for in and ex ovo monitoring of cellular and molecular dynamics already established in ovo are applicable alike. In this view, the present review aims to emphasize and discuss opportunities and limitations of the chicken embryo model for pre-clinical research in cancer and cancer immunology.
Collapse
Affiliation(s)
- Lea Miebach
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
- Department of General, Thoracic, Vascular, and Visceral Surgery, Greifswald University Medical Center, Greifswald, Germany
| | - Julia Berner
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
- Department of General, Thoracic, Vascular, and Visceral Surgery, Greifswald University Medical Center, Greifswald, Germany
- Department of Oral and Maxillofacial Surgery, Plastic Surgery, Greifswald University Medical Center, Greifswald, Germany
| | - Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
- Department of General, Thoracic, Vascular, and Visceral Surgery, Greifswald University Medical Center, Greifswald, Germany
- *Correspondence: Sander Bekeschus,
| |
Collapse
|
14
|
Hesemans E, Buttiens K, Manshian BB, Soenen SJ. The Role of Optical Imaging in Translational Nanomedicine. J Funct Biomater 2022; 13:137. [PMID: 36135572 PMCID: PMC9502568 DOI: 10.3390/jfb13030137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/23/2022] [Accepted: 08/25/2022] [Indexed: 11/18/2022] Open
Abstract
Nanomedicines have been a major research focus in the past two decades and are increasingly emerging in a broad range of clinical applications. However, a proper understanding of their biodistribution is required to further progress the field of nanomedicine. For this, imaging methods to monitor the delivery and therapeutic efficacy of nanoparticles are urgently needed. At present, optical imaging is the most common method used to study the biodistribution of nanomaterials, where the unique properties of nanomaterials and advances in optical imaging can jointly result in novel methods for optimal monitoring of nanomaterials in preclinical animal models. This review article aims to give an introduction to nanomedicines and their translational impact to highlight the potential of optical imaging to study the biodistribution of nanoparticles and to monitor the delivery and therapeutic efficacy at the preclinical level. After introducing both domains, the review focuses on different techniques that can be used to overcome some intrinsic limitations of optical imaging and how this can specifically benefit nanoparticle studies. Finally, we point out some important key features of nanoparticles that currently hinder their full potential in the clinic and how the advances in optical imaging can help to provide us with the information needed to further boost the clinical translation and expand the field of nanomedicines.
Collapse
Affiliation(s)
- Evelien Hesemans
- NanoHealth and Optical Imaging Group, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Kiana Buttiens
- NanoHealth and Optical Imaging Group, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
| | - Bella B. Manshian
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
- Leuven Cancer Institute, 3000 Leuven, Belgium
| | - Stefaan J. Soenen
- NanoHealth and Optical Imaging Group, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Herestraat 49, B3000 Leuven, Belgium
- Leuven Cancer Institute, 3000 Leuven, Belgium
| |
Collapse
|
15
|
Mapanao AK, Che PP, Sarogni P, Sminia P, Giovannetti E, Voliani V. Tumor grafted - chick chorioallantoic membrane as an alternative model for biological cancer research and conventional/nanomaterial-based theranostics evaluation. Expert Opin Drug Metab Toxicol 2021; 17:947-968. [PMID: 33565346 DOI: 10.1080/17425255.2021.1879047] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/18/2021] [Indexed: 02/09/2023]
Abstract
Introduction: Advancements in cancer management and treatment are associated with strong preclinical research data, in which reliable cancer models are demanded. Indeed, inconsistent preclinical findings and stringent regulations following the 3Rs principle of reduction, refinement, and replacement of conventional animal models currently pose challenges in the development and translation of efficient technologies. The chick embryo chorioallantoic membrane (CAM) is a system for the evaluation of treatment effects on the vasculature, therefore suitable for studies on angiogenesis. Apart from vascular effects, the model is now increasingly employed as a preclinical cancer model following tumor-grafting procedures.Areas covered: The broad application of CAM tumor model is highlighted along with the methods for analyzing the neoplasm and vascular system. The presented and cited investigations focus on cancer biology and treatment, encompassing both conventional and emerging nanomaterial-based modalities.Expert opinion: The CAM tumor model finds increased significance given the influences of angiogenesis and the tumor microenvironment in cancer behavior, then providing a qualified miniature system for oncological research. Ultimately, the establishment and increased employment of such a model may resolve some of the limitations present in the standard preclinical tumor models, thereby redefining the preclinical research workflow.
Collapse
Affiliation(s)
- Ana Katrina Mapanao
- Center for Nanotechnology Innovation@NEST, Istituto Italiano Di Tecnologia, Pisa, Italy
- NEST-Scuola Normale Superiore, Pisa, Italy
| | - Pei Pei Che
- Department of Radiation Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, The Netherlands
| | - Patrizia Sarogni
- Center for Nanotechnology Innovation@NEST, Istituto Italiano Di Tecnologia, Pisa, Italy
| | - Peter Sminia
- Department of Radiation Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center, Amsterdam, The Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Cancer Center Amsterdam, The Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, Fondazione Pisana per La Scienza, Pisa, Italy
| | - Valerio Voliani
- Center for Nanotechnology Innovation@NEST, Istituto Italiano Di Tecnologia, Pisa, Italy
| |
Collapse
|
16
|
Park J, Wen AM, Gao H, Shin MD, Simon DI, Wang Y, Steinmetz NF. Designing S100A9-Targeted Plant Virus Nanoparticles to Target Deep Vein Thrombosis. Biomacromolecules 2021; 22:2582-2594. [PMID: 34060817 DOI: 10.1021/acs.biomac.1c00303] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Thromboembolic conditions are a leading cause of death worldwide, and deep vein thrombosis (DVT), or occlusive venous clot formation, is a critical and rising problem that contributes to damage of vital organs, long-term complications, and life-threatening conditions such as pulmonary embolism. Early diagnosis and treatment are correlated to better prognosis. However, current technologies in these areas, such as ultrasonography for diagnostics and anticoagulants for treatment, are limited in terms of their accuracy and therapeutic windows. In this work, we investigated targeting myeloid related protein 14 (MRP-14, also known as S100A9) using plant virus-based nanoparticle carriers as a means to achieve tissue specificity aiding prognosis and therapeutic intervention. We used a combinatorial peptide library screen to identify peptide ligands that bind MRP-14. Candidates were selected and formulated as nanoparticles by using cowpea mosaic virus (CPMV) and tobacco mosaic virus (TMV). Intravascular delivery of our MRP-14-targeted nanoparticles in a murine model of DVT resulted in enhanced accumulation in the thrombi and reduced thrombus size, suggesting application of nanoparticles for molecular targeting of MRP-14 could be a promising direction for improving DVT diagnostics, therapeutics, and therefore prognosis.
Collapse
Affiliation(s)
- Jooneon Park
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Amy M Wen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Huiyun Gao
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Matthew D Shin
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, United States
| | - Daniel I Simon
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Yunmei Wang
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, United States.,Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States.,Department of Radiology, University of California, San Diego, La Jolla, California 92093, United States.,Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, California 92093, United States.,Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, United States.,Institute for Materials Discovery and Design, University of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
17
|
Ademi H, Shinde DA, Gassmann M, Gerst D, Chaachouay H, Vogel J, Gorr TA. Targeting neovascularization and respiration of tumor grafts grown on chick embryo chorioallantoic membranes. PLoS One 2021; 16:e0251765. [PMID: 33999935 PMCID: PMC8128225 DOI: 10.1371/journal.pone.0251765] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 05/02/2021] [Indexed: 12/25/2022] Open
Abstract
Since growing tumors stimulate angiogenesis, via vascular endothelial growth factor (VEGF), angiogenesis inhibitors (AIs, blockers of the VEGF signaling pathway) have been introduced to cancer therapy. However, AIs often yielded only modest and short-lived gains in cancer patients and more invasive tumor phenotypes in animal models. Combining anti-VEGF strategies with lactate uptake blockers may boost both efficacy and safety of AIs. We assessed this hypothesis by using the ex ovo chorioallantoic membrane (CAM) assay. We show that AI-based monotherapy (Avastin®, AVA) increases tumor hypoxia in human CAM cancer cell xenografts and cell spread in human as well as canine CAM cancer cell xenografts. In contrast, combining AVA treatment with lactate importer MCT1 inhibitors (α-cyano-4-hydroxycinnamic acid (CHC) or AZD3965 (AZD)) reduced both tumor growth and cell dissemination of human and canine explants. Moreover, combining AVA+AZD diminished blood perfusion and tumor hypoxia in human explants. Thus, the ex ovo CAM assay as an easy, fast and cheap experimental setup is useful for pre-clinical cancer research. Moreover, as an animal-free experimental setup the CAM assay can reduce the high number of laboratory animals used in pre-clinical cancer research.
Collapse
Affiliation(s)
- Hyrije Ademi
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Center for Clinical Studies at the Vetsuisse Faculty of the University of Zurich, Zurich, Switzerland
| | - Dheeraj A. Shinde
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Max Gassmann
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Zurich Centre for Integrative Human Physiology (ZIHP), University of Zurich, Zurich, Switzerland
| | - Daniela Gerst
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Hassan Chaachouay
- Division of Radiation Oncology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- Bioactives, Health & Environment Laboratory, Epigenetics, Health & Environment Unit, Faculty of Science and Techniques, Moulay Ismail University, Errachidia, Morocco
| | - Johannes Vogel
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Thomas A. Gorr
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
18
|
Nkanga CI, Steinmetz NF. The pharmacology of plant virus nanoparticles. Virology 2021; 556:39-61. [PMID: 33545555 PMCID: PMC7974633 DOI: 10.1016/j.virol.2021.01.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/14/2022]
Abstract
The application of nanoparticles for medical purposes has made enormous strides in providing new solutions to health problems. The observation that plant virus-based nanoparticles (VNPs) can be repurposed and engineered as smart bio-vehicles for targeted drug delivery and imaging has launched extensive research for improving the therapeutic and diagnostic management of various diseases. There is evidence that VNPs are promising high value nanocarriers with potential for translational development. This is mainly due to their unique features, encompassing structural uniformity, ease of manufacture and functionalization by means of expression, chemical biology and self-assembly. While the development pipeline is moving rapidly, with many reports focusing on engineering and manufacturing aspects to tailor the properties and efficacy of VNPs, fewer studies have focused on gaining insights into the nanotoxicity of this novel platform nanotechnology. Herein, we discuss the pharmacology of VNPs as a function of formulation and route of administration. VNPs are reviewed in the context of their application as therapeutic adjuvants or nanocarrier excipients to initiate, enhance, attenuate or impede the formulation's toxicity. The summary of the data however also underlines the need for meticulous VNP structure-nanotoxicity studies to improve our understanding of their in vivo fates and pharmacological profiles to pave the way for translation of VNP-based formulations into the clinical setting.
Collapse
Affiliation(s)
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA, 92039, United States; Department of Bioengineering, Department of Radiology, Center for NanoImmunoEngineering, Moores Cancer Center, Institute for Materials Discovery and Design, University of California-San Diego, La Jolla, CA, 92039, United States.
| |
Collapse
|
19
|
Chariou PL, Beiss V, Ma Y, Steinmetz NF. In situ vaccine application of inactivated CPMV nanoparticles for cancer immunotherapy. MATERIALS ADVANCES 2021; 2:1644-1656. [PMID: 34368764 PMCID: PMC8323807 DOI: 10.1039/d0ma00752h] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 01/26/2021] [Indexed: 05/24/2023]
Abstract
Cowpea mosaic virus (CPMV) is currently in the development pipeline for multiple biomedical applications, including cancer immunotherapy. In particular the application of CPMV as in situ vaccine has shown promise; here the plant viral nanoparticle is used as an adjuvant and is injected directly into a tumor to reverse immunosuppression and prime systemic anti-tumor immunity. Efficacy of this CPMV-based cancer immunotherapy has been demonstrated in multiple tumor mouse models and canine cancer patients. However, while CPMV is non-infectious to mammals, it is infectious to legumes and therefore, from a safety perspective, it is desired to develop non-infectious CPMV formulations. Non-infectious virus-like particles of CPMV devoid of nucleic acids have been produced; nevertheless, efficacy of such empty CPMV nanoparticles does not match efficacy of nucleic acid-laden CPMV. The multivalent capsid activates the innate immune system through pathogen pattern recognition receptors (PRRs) such as toll-like receptors (TLRs); the RNA cargo provides additional signaling through TLR-7/8, which boosts the efficacy of this adjuvant. Therefore, in this study, we set out to develop RNA-laden, but non-infectious CPMV. We report inactivation of CPMV using UV light and chemical inactivation using β-propiolactone (βPL) or formalin. 7.5 J cm-2 UV, 50 mM βPL or 1 mM formalin was determined to be sufficient to inactivate CPMV and prevented plant infection. We compared the immunogenicity of native CPMV and inactivated CPMV formulations in vitro and in vivo using RAW-Blue™ reporter cells and a murine syngeneic, orthotropic melanoma model (using B16F10 cells and C57BL6 mice). While the in vitro assay indicated activation of the RAW-Blue™ reporter cells by formaldehyde and UV-inactivated CPMV at levels comparable to native CPMV; βPL-inactivated CPMV appeared to have diminished activity. Tumor mouse model experiments indicate potent efficacy of the chemically-inactivated CPMV (UV-treated CPMV was not tested) leading to tumor regression and increased survival; efficacy was somewhat reduced when compared to CPMV, however these samples outperformed the empty CPMV nanoparticles. These results will facilitate the translational development of safe and potent CPMV-based cancer immunotherapies.
Collapse
Affiliation(s)
- Paul L. Chariou
- Department of Bioengineering, University of California-San DiegoLa JollaCA 92039USA
| | - Veronique Beiss
- Department of NanoEngineering, University of California-San DiegoLa JollaCA 92039USA
| | - Yifeng Ma
- Department of NanoEngineering, University of California-San DiegoLa JollaCA 92039USA
| | - Nicole F. Steinmetz
- Department of Bioengineering, University of California-San DiegoLa JollaCA 92039USA
- Department of NanoEngineering, University of California-San DiegoLa JollaCA 92039USA
- Department of Radiology, University of California-San DiegoLa JollaCA 92039USA
- Moores Cancer Center, University of California-San DiegoLa JollaCA 92039USA
- Center for Nano-ImmunoEngineering, University of California-San DiegoLa JollaCA 92039USA
- Institute for Materials Discovery and Design, University of California-San DiegoLa JollaCA 92039USA
| |
Collapse
|
20
|
de la Torre P, Paris JL, Fernández-de la Torre M, Vallet-Regí M, Flores AI. Endostatin Genetically Engineered Placental Mesenchymal Stromal Cells Carrying Doxorubicin-Loaded Mesoporous Silica Nanoparticles for Combined Chemo- and Antiangiogenic Therapy. Pharmaceutics 2021; 13:244. [PMID: 33578733 PMCID: PMC7916487 DOI: 10.3390/pharmaceutics13020244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 02/05/2023] Open
Abstract
Combination therapies constitute a powerful tool for cancer treatment. By combining drugs with different mechanisms of action, the limitations of each individual agent can be overcome, while increasing therapeutic benefit. Here, we propose employing tumor-migrating decidua-derived mesenchymal stromal cells as therapeutic agents combining antiangiogenic therapy and chemotherapy. First, a plasmid encoding the antiangiogenic protein endostatin was transfected into these cells by nucleofection, confirming its expression by ELISA and its biological effect in an ex ovo chick embryo model. Second, doxorubicin-loaded mesoporous silica nanoparticles were introduced into the cells, which would act as vehicles for the drug being released. The effect of the drug was evaluated in a coculture in vitro model with mammary cancer cells. Third, the combination of endostatin transfection and doxorubicin-nanoparticle loading was carried out with the decidua mesenchymal stromal cells. This final cell platform was shown to retain its tumor-migration capacity in vitro, and the combined in vitro therapeutic efficacy was confirmed through a 3D spheroid coculture model using both cancer and endothelial cells. The results presented here show great potential for the development of combination therapies based on genetically-engineered cells that can simultaneously act as cellular vehicles for drug-loaded nanoparticles.
Collapse
Affiliation(s)
- Paz de la Torre
- Grupo de Medicina Regenerativa, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Avda. Cordoba s/n 28041, 28041 Madrid, Spain;
| | - Juan L. Paris
- Departamento de Química en Ciencias Farmacéuticas (Unidad Docente de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Miguel Fernández-de la Torre
- Grupo de Enfermedades Raras, Mitocondriales y Neuromusculares, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Avda. Cordoba s/n 28041, 28041 Madrid, Spain;
| | - María Vallet-Regí
- Departamento de Química en Ciencias Farmacéuticas (Unidad Docente de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - Ana I. Flores
- Grupo de Medicina Regenerativa, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Avda. Cordoba s/n 28041, 28041 Madrid, Spain;
| |
Collapse
|
21
|
Intravital Optical Imaging to Monitor Anti-Tumor Immunological Response in Preclinical Models. Bioanalysis 2021. [DOI: 10.1007/978-3-030-78338-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
22
|
Duval KEA, Wagner RJ, Beiss V, Fiering SN, Steinmetz NF, Hoopes PJ. Cowpea Mosaic Virus Nanoparticle Enhancement of Hypofractionated Radiation in a B16 Murine Melanoma Model. Front Oncol 2020; 10:594614. [PMID: 33392089 PMCID: PMC7773968 DOI: 10.3389/fonc.2020.594614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/02/2020] [Indexed: 01/06/2023] Open
Abstract
INTRODUCTION Virus and virus-like nanoparticles (VNPs) have been used for a variety of preclinical treatments, including in situ anti-cancer vaccination. The Cowpea mosaic virus (CPMV) is a VNP that has shown the ability to stimulate an anti-cancer immune response. The hypothesis of this study is two-fold: that intratumoral CPMV enhances the immunogenetic and cytotoxic response of hypofractionated radiation (15 Gy or 3 x 8 Gy), and that the effect differs between fraction regimens in the murine B16 flank melanoma model. METHODS CPMV nanoparticles were delivered intratumorally, 100 μg/tumor to B16 murine melanoma flank tumors alone, and in combination with either 15 Gy or 3 x 8 Gy (3 consecutive days). Tumors were assessed for immune and cytotoxic gene and protein expression, and cytotoxic T cell infiltration 4 days post treatment. Treatment based tumor control was assessed by a 3-fold tumor growth assay. RESULTS Both CPMV and radiation alone demonstrated the activation of a number of important immune and cytotoxic genes including natural killer cell and T cell mediated cytotoxicity pathways. However, the combination treatment activated greater expression than either treatment alone. CPMV combined with a single dose of 15 Gy demonstrated greater immune and cytotoxic gene expression, protein expression, CD8+ T cell infiltration activity, and greater tumor growth delay compared to 3 x 8 Gy with CPMV. CONCLUSION CPMV presents a unique and promising hypofractionated radiation adjuvant that leads to increased anti-tumor cytotoxic and immune signaling, especially with respect to the immune mediated cytotoxicity, immune signaling, and toll-like receptor signaling pathways. This improvement was greater with a single dose than with a fractionated dose.
Collapse
Affiliation(s)
- Kayla E. A. Duval
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - Robert J. Wagner
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
| | - Veronique Beiss
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, United States
| | - Steven N. Fiering
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| | - Nicole F. Steinmetz
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, United States
- Department of Bioengineering, University of California San Diego, La Jolla, CA, United States
- Department of Radiology, University of California San Diego, La Jolla, CA, United States
- Moores Cancer Center, University of California San Diego, La Jolla, CA, United States
- Center for Nano-ImmunoEngineering, University of California San Diego, La Jolla, CA, United States
| | - P. Jack Hoopes
- Geisel School of Medicine, Dartmouth College, Hanover, NH, United States
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
- Norris Cotton Cancer Center, Dartmouth-Hitchcock Medical Center, Lebanon, NH, United States
| |
Collapse
|
23
|
Shukla S, Hu H, Cai H, Chan SK, Boone CE, Beiss V, Chariou PL, Steinmetz NF. Plant Viruses and Bacteriophage-Based Reagents for Diagnosis and Therapy. Annu Rev Virol 2020; 7:559-587. [PMID: 32991265 PMCID: PMC8018517 DOI: 10.1146/annurev-virology-010720-052252] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Viral nanotechnology exploits the prefabricated nanostructures of viruses, which are already abundant in nature. With well-defined molecular architectures, viral nanocarriers offer unprecedented opportunities for precise structural and functional manipulation using genetic engineering and/or bio-orthogonal chemistries. In this manner, they can be loaded with diverse molecular payloads for targeted delivery. Mammalian viruses are already established in the clinic for gene therapy and immunotherapy, and inactivated viruses or virus-like particles have long been used as vaccines. More recently, plant viruses and bacteriophages have been developed as nanocarriers for diagnostic imaging, vaccine and drug delivery, and combined diagnosis/therapy (theranostics). The first wave of these novel virus-based tools has completed clinical development and is poised to make an impact on clinical practice.
Collapse
Affiliation(s)
- Sourabh Shukla
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, USA
| | - He Hu
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Hui Cai
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Soo-Khim Chan
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Christine E Boone
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Veronique Beiss
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Paul L Chariou
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, USA
| | - Nicole F Steinmetz
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, USA
- Department of Radiology, University of California, San Diego, La Jolla, California 92093, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, USA
- Moores Cancer Center and Center for Nano-ImmunoEngineering, University of California, San Diego, La Jolla, California 92093, USA;
| |
Collapse
|
24
|
Liu ZL, Clausen JR, Wagner JL, Butler KS, Bolintineanu DS, Lechman JB, Rao RR, Aidun CK. Heterogeneous partition of cellular blood-borne nanoparticles through microvascular bifurcations. Phys Rev E 2020; 102:013310. [PMID: 32795082 DOI: 10.1103/physreve.102.013310] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/18/2020] [Indexed: 06/11/2023]
Abstract
Blood flowing through microvascular bifurcations has been an active research topic for many decades, while the partitioning pattern of nanoscale solutes in the blood remains relatively unexplored. Here we demonstrate a multiscale computational framework for direct numerical simulation of the nanoparticle (NP) partitioning through physiologically relevant vascular bifurcations in the presence of red blood cells (RBCs). The computational framework is established by embedding a particulate suspension inflow-outflow boundary condition into a multiscale blood flow solver. The computational framework is verified by recovering a tubular blood flow without a bifurcation and validated against the experimental measurement of an intravital bifurcation flow. The classic Zweifach-Fung (ZF) effect is shown to be well captured by the method. Moreover, we observe that NPs exhibit a ZF-like heterogeneous partition in response to the heterogeneous partition of the RBC phase. The NP partitioning prioritizes the high-flow-rate daughter branch except for extreme (large or small) suspension flow partition ratios under which the complete phase separation tends to occur. By analyzing the flow field and the particle trajectories, we show that the ZF-like heterogeneity in the NP partition can be explained by the RBC-entrainment effect caused by the deviation of the flow separatrix preceded by the tank treading of RBCs near the bifurcation junction. The recovery of homogeneity in the NP partition under extreme flow partition ratios is due to the plasma skimming of NPs in the cell-free layer. These findings, based on the multiscale computational framework, provide biophysical insights to the heterogeneous distribution of NPs in microvascular beds that are observed pathophysiologically.
Collapse
Affiliation(s)
- Zixiang L Liu
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Jonathan R Clausen
- Thermal and Fluid Processes, Sandia National Laboratories, Albuquerque, New Mexico 87185, USA
| | - Justin L Wagner
- Aerosciences Department, Sandia National Laboratories, Albuquerque, New Mexico 87185, USA
| | - Kimberly S Butler
- Molecular and Microbiology, Sandia National Laboratories, Albuquerque, New Mexico 87185, USA
| | - Dan S Bolintineanu
- Fluid and Reactive Processes, Sandia National Laboratories, Albuquerque, New Mexico 87185, USA
| | - Jeremy B Lechman
- Fluid and Reactive Processes, Sandia National Laboratories, Albuquerque, New Mexico 87185, USA
| | - Rekha R Rao
- Fluid and Reactive Processes, Sandia National Laboratories, Albuquerque, New Mexico 87185, USA
| | - Cyrus K Aidun
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| |
Collapse
|
25
|
Chung YH, Cai H, Steinmetz NF. Viral nanoparticles for drug delivery, imaging, immunotherapy, and theranostic applications. Adv Drug Deliv Rev 2020; 156:214-235. [PMID: 32603813 PMCID: PMC7320870 DOI: 10.1016/j.addr.2020.06.024] [Citation(s) in RCA: 242] [Impact Index Per Article: 48.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/19/2020] [Accepted: 06/21/2020] [Indexed: 02/06/2023]
Abstract
Viral nanoparticles (VNPs) encompass a diverse array of naturally occurring nanomaterials derived from plant viruses, bacteriophages, and mammalian viruses. The application and development of VNPs and their genome-free versions, the virus-like particles (VLPs), for nanomedicine is a rapidly growing. VLPs can encapsulate a wide range of active ingredients as well as be genetically or chemically conjugated to targeting ligands to achieve tissue specificity. VLPs are manufactured through scalable fermentation or molecular farming, and the materials are biocompatible and biodegradable. These properties have led to a wide range of applications, including cancer therapies, immunotherapies, vaccines, antimicrobial therapies, cardiovascular therapies, gene therapies, as well as imaging and theranostics. The use of VLPs as drug delivery agents is evolving, and sufficient research must continuously be undertaken to translate these therapies to the clinic. This review highlights some of the novel research efforts currently underway in the VNP drug delivery field in achieving this greater goal.
Collapse
Affiliation(s)
- Young Hun Chung
- Department of Bioengineering, University of California-San Diego, La Jolla, CA 92093, United States
| | - Hui Cai
- Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92093, United States
| | - Nicole F Steinmetz
- Department of Bioengineering, University of California-San Diego, La Jolla, CA 92093, United States; Department of NanoEngineering, University of California-San Diego, La Jolla, CA 92093, United States; Department of Radiology, University of California-San Diego, La Jolla, CA 92093, United States; Moores Cancer Center, University of California-San Diego, La Jolla, CA 92093, United States; Center for Nano-ImmunoEngineering, University of California-San Diego, La Jolla, CA 92093, United States.
| |
Collapse
|
26
|
Patel D, Zode SS, Bansal AK. Formulation aspects of intravenous nanosuspensions. Int J Pharm 2020; 586:119555. [PMID: 32562654 DOI: 10.1016/j.ijpharm.2020.119555] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/23/2020] [Accepted: 06/14/2020] [Indexed: 01/04/2023]
Abstract
Intravenous (IV) route is preferred for rapid onset of action, avoiding first pass metabolism and achieving site specific delivery. Development of IV formulations for poorly water soluble drugs poses significant challenges. Formulation approaches like salt formation, co-solvents, surfactants and inclusion complexation using cyclodextrins are used for solubilisation. However, these approaches are not applicable universally and have limitations in extent of solubilisation, hypersensitivity, toxicity and application to only specific type of molecules. IV nanosuspension have been attracting attention as a viable strategy for development of IV formulations of poorly water-soluble drugs. Nanosuspension consists of nanocrystals of poorly water soluble drug suspended in aqueous media and stabilized using minimal concentration of stabilizers. Recent years have witnessed their potential in formulations for toxicological studies and clinical trials. However various challenges are associated with the translational development of IV nanosuspensions. Therefore, the objective of the current review is to provide a holistic view of formulation development and desired properties of IV nanosuspensions. It will also focus on advancements in characterization tools, manufacturing techniques and post-production processing. Challenges associated with translational development and regulatory aspects of IV nanosuspension will be addressed. Additionally, their role in preclinical evaluation and special applications like targeting will also be discussed with the help of case studies. The applications of IV nanosuspensions shall expand as their applications move from preclinical phase to commercialization.
Collapse
Affiliation(s)
- Dipeekakumari Patel
- National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Sandeep S Zode
- National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab 160062, India
| | - Arvind K Bansal
- National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab 160062, India.
| |
Collapse
|
27
|
Rahman HS, Othman HH, Hammadi NI, Yeap SK, Amin KM, Abdul Samad N, Alitheen NB. Novel Drug Delivery Systems for Loading of Natural Plant Extracts and Their Biomedical Applications. Int J Nanomedicine 2020; 15:2439-2483. [PMID: 32346289 PMCID: PMC7169473 DOI: 10.2147/ijn.s227805] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 10/10/2019] [Indexed: 12/18/2022] Open
Abstract
Many types of research have distinctly addressed the efficacy of natural plant metabolites used for human consumption both in cell culture and preclinical animal model systems. However, these in vitro and in vivo effects have not been able to be translated for clinical use because of several factors such as inefficient systemic delivery and bioavailability of promising agents that significantly contribute to this disconnection. Over the past decades, extraordinary advances have been made successfully on the development of novel drug delivery systems for encapsulation of plant active metabolites including organic, inorganic and hybrid nanoparticles. The advanced formulas are confirmed to have extraordinary benefits over conventional and previously used systems in the manner of solubility, bioavailability, toxicity, pharmacological activity, stability, distribution, sustained delivery, and both physical and chemical degradation. The current review highlights the development of novel nanocarrier for plant active compounds, their method of preparation, type of active ingredients, and their biomedical applications.
Collapse
Affiliation(s)
- Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Sulaimani, Sulaymaniyah46001, Republic of Iraq
- Department of Medical Laboratory Sciences, College of Health Sciences, Komar University of Science and Technology, Sulaymaniyah, Republic of Iraq
| | - Hemn Hassan Othman
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Sulaimani, Sulaymaniyah46001, Republic of Iraq
| | - Nahidah Ibrahim Hammadi
- Department of Histology, College of Veterinary Medicine, University of Al-Anbar, Ramadi, Republic of Iraq
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, Sepang, Malaysia
| | - Kawa Mohammad Amin
- Department of Microbiology, College of Medicine, University of Sulaimani, Sulaymaniyah46001, Republic of Iraq
| | - Nozlena Abdul Samad
- Integrative Medicine Cluster, Institut Perubatan dan Pergigian Termaju (IPPT), Sains@BERTAM, Universiti Sains Malaysia, Kepala Batas13200, Pulau Pinang, Malaysia
| | - Noorjahan Banu Alitheen
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Bio-Molecular Sciences, Universiti Putra Malaysia, Selangor, Malaysia
| |
Collapse
|
28
|
Huang Q, Cohen MA, Alsina FC, Devlin G, Garrett A, McKey J, Havlik P, Rakhilin N, Wang E, Xiang K, Mathews P, Wang L, Bock C, Ruthig V, Wang Y, Negrete M, Wong CW, Murthy PKL, Zhang S, Daniel AR, Kirsch DG, Kang Y, Capel B, Asokan A, Silver DL, Jaenisch R, Shen X. Intravital imaging of mouse embryos. Science 2020; 368:181-186. [PMID: 32273467 PMCID: PMC7646360 DOI: 10.1126/science.aba0210] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 03/16/2020] [Indexed: 12/11/2022]
Abstract
Embryonic development is a complex process that is unamenable to direct observation. In this study, we implanted a window to the mouse uterus to visualize the developing embryo from embryonic day 9.5 to birth. This removable intravital window allowed manipulation and high-resolution imaging. In live mouse embryos, we observed transient neurotransmission and early vascularization of neural crest cell (NCC)-derived perivascular cells in the brain, autophagy in the retina, viral gene delivery, and chemical diffusion through the placenta. We combined the imaging window with in utero electroporation to label and track cell division and movement within embryos and observed that clusters of mouse NCC-derived cells expanded in interspecies chimeras, whereas adjacent human donor NCC-derived cells shrank. This technique can be combined with various tissue manipulation and microscopy methods to study the processes of development at unprecedented spatiotemporal resolution.
Collapse
Affiliation(s)
- Qiang Huang
- Department of Pediatric Surgery, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Malkiel A Cohen
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Fernando C Alsina
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA
| | - Garth Devlin
- Department of Surgery, School of Medicine, Duke University, Durham, NC, USA
| | - Aliesha Garrett
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Jennifer McKey
- Department of Cell Biology, School of Medicine, Duke University, Durham, NC, USA
| | - Patrick Havlik
- Department of Surgery, School of Medicine, Duke University, Durham, NC, USA
| | - Nikolai Rakhilin
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Ergang Wang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Kun Xiang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Parker Mathews
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC, USA
| | - Lihua Wang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Cheryl Bock
- Duke Cancer Institute, School of Medicine, Duke University, Durham, NC, USA
| | - Victor Ruthig
- Department of Cell Biology, School of Medicine, Duke University, Durham, NC, USA
| | - Yi Wang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Marcos Negrete
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Chi Wut Wong
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Preetish K L Murthy
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA
| | - Shupei Zhang
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Andrea R Daniel
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
| | - David G Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, NC, USA
- Department of Pharmacology & Cancer Biology, Duke University Medical Center, Durham, NC, USA
| | - Yubin Kang
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC, USA
| | - Blanche Capel
- Department of Cell Biology, School of Medicine, Duke University, Durham, NC, USA
| | - Aravind Asokan
- Department of Surgery, School of Medicine, Duke University, Durham, NC, USA
| | - Debra L Silver
- Department of Molecular Genetics and Microbiology, School of Medicine, Duke University, Durham, NC, USA
| | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
29
|
Sharma A, Ishak N, Swee-Hin T, Pramanik M. High resolution, label-free photoacoustic imaging of live chicken embryo developing in bioengineered eggshell. JOURNAL OF BIOPHOTONICS 2020; 13:e201960108. [PMID: 31908159 DOI: 10.1002/jbio.201960108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/19/2019] [Accepted: 12/23/2019] [Indexed: 05/09/2023]
Abstract
Chicken embryos have been proven to be an attractive vertebrate model for biomedical research. They have helped in making significant contributions for advancements in various fields like developmental biology, cancer research and cardiovascular studies. However, a non-invasive, label-free method of imaging live chicken embryo at high resolution still needs to be developed and optimized. In this work, we have shown the potential of photoacoustic tomography (PAT) for imaging live chicken embryos cultured in bioengineered eggshells. Laser pulses at wavelengths of 532 and 740 nm were used for attaining cross-sectional images of chicken embryos at different developmental stages. Cross-sections along different depths were imaged to gain knowledge of the relative depth of different vessels and organs. Due to high optical absorption of vasculature and embryonic eye, images with good optical contrast could be acquired using this method. We have thus reported a label-free method of performing cross-sectional imaging of chicken embryos at high resolution demonstrating the capacity of PAT as a promising tool for avian embryo imaging.
Collapse
Affiliation(s)
- Arunima Sharma
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Noreen Ishak
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Teoh Swee-Hin
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Manojit Pramanik
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| |
Collapse
|
30
|
Kanwar N, Carmine-Simmen K, Nair R, Wang C, Moghadas-Jafari S, Blaser H, Tran-Thanh D, Wang D, Wang P, Wang J, Pasculescu A, Datti A, Mak T, Lewis JD, Done SJ. Amplification of a calcium channel subunit CACNG4 increases breast cancer metastasis. EBioMedicine 2020; 52:102646. [PMID: 32062352 PMCID: PMC7016384 DOI: 10.1016/j.ebiom.2020.102646] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/20/2019] [Accepted: 01/15/2020] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Previously, we found that amplification of chromosome 17q24.1-24.2 is associated with lymph node metastasis, tumour size, and lymphovascular invasion in invasive ductal carcinoma. A gene within this amplicon, CACNG4, an L-type voltage-gated calcium channel gamma subunit, is elevated in breast cancers with poor prognosis. Calcium homeostasis is achieved by maintaining low intracellular calcium levels. Altering calcium influx/efflux mechanisms allows tumour cells to maintain homeostasis despite high serum calcium levels often associated with advanced cancer (hypercalcemia) and aberrant calcium signaling. METHODS In vitro 2-D and 3-D assays, and intracellular calcium influx assays were utilized to measure tumourigenic activity in response to altered CANCG4 levels and calcium channel blockers. A chick-CAM model and mouse model for metastasis confirmed these results in vivo. FINDINGS CACNG4 alters cell motility in vitro, induces malignant transformation in 3-dimensional culture, and increases lung-specific metastasis in vivo. CACNG4 functions by closing the channel pore, inhibiting calcium influx, and altering calcium signaling events involving key survival and metastatic pathway genes (AKT2, HDAC3, RASA1 and PKCζ). INTERPRETATION CACNG4 may promote homeostasis, thus increasing the survival and metastatic ability of tumour cells in breast cancer. Our findings suggest an underlying pathway for tumour growth and dissemination regulated by CACNG4 that is significant with respect to developing treatments that target these channels in tumours with aberrant calcium signaling. FUNDING Canadian Breast Cancer Foundation, Ontario; Canadian Institutes of Health Research.
Collapse
Affiliation(s)
- Nisha Kanwar
- The Campbell Family for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada; Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | | | - Ranju Nair
- The Campbell Family for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Chunjie Wang
- Department of Pathology and Laboratory Medicine, Saskatoon City Hospital, Saskatoon, SK S7K 0M7, Canada
| | - Soode Moghadas-Jafari
- The Campbell Family for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Heiko Blaser
- The Campbell Family for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Danh Tran-Thanh
- Department of Pathology, Centre Hospitalier de l'Université de Montréal, Montréal, QC H2W 1T8, Canada
| | - Dongyu Wang
- Department of Medical Biophysics, Faculty of Medicine, University of Toronto, ON M5S 1A1, Canada
| | - Peiqi Wang
- The Campbell Family for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada
| | - Jenny Wang
- Samuel Lunenfeld Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Adrian Pasculescu
- Samuel Lunenfeld Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada
| | - Alessandro Datti
- Samuel Lunenfeld Research Institute, Mt. Sinai Hospital, Toronto, ON M5G 1X5, Canada; Department of Agricultural, Food, and Environmental Sciences, University of Perugia, Perugia, Italy
| | - Tak Mak
- The Campbell Family for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada; Department of Medical Biophysics, Faculty of Medicine, University of Toronto, ON M5S 1A1, Canada
| | - John D Lewis
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2E1, Canada
| | - Susan J Done
- The Campbell Family for Breast Cancer Research, Princess Margaret Cancer Centre, Toronto, ON M5G 2M9, Canada; Department of Laboratory Medicine and Pathobiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada; Department of Medical Biophysics, Faculty of Medicine, University of Toronto, ON M5S 1A1, Canada; Laboratory Medicine Program, Department of Pathology, University Health Network, Toronto General Hospital, 200 Elizabeth Street, 11th floor, Toronto, ON M5G 2C4, Canada.
| |
Collapse
|
31
|
Albakri MM, Veliz FA, Fiering SN, Steinmetz NF, Sieg SF. Endosomal toll-like receptors play a key role in activation of primary human monocytes by cowpea mosaic virus. Immunology 2020; 159:183-192. [PMID: 31630392 PMCID: PMC6954739 DOI: 10.1111/imm.13135] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/20/2019] [Accepted: 10/14/2019] [Indexed: 12/30/2022] Open
Abstract
The plant virus, cowpea mosaic virus (CPMV), has demonstrated a remarkable capacity to induce anti-tumour immune responses following direct administration into solid tumours. The molecular pathways that account for these effects and the capacity of CPMV to activate human cells are not well defined. Here, we examine the ability of CPMV particles to activate human monocytes, dendritic cells (DCs) and macrophages. Monocytes in peripheral blood mononuclear cell cultures and purified CD14+ monocytes were readily activated by CPMV in vitro, leading to induction of HLA-DR, CD86, PD-L1, IL-15R and CXCL10 expression. Monocytes released chemokines, CXCL10, MIP-1α and MIP-1β into cell culture supernatants after incubation with CPMV. DC subsets (pDC and mDC) and monocyte-derived macrophages also demonstrated evidence of activation after incubation with CPMV. Inhibitors of spleen tyrosine kinase (SYK), endocytosis or endocytic acidification impaired the capacity of CPMV to activate monocytes. Furthermore, CPMV activation of monocytes was partially blocked by a TLR7/8 antagonist. These data demonstrate that CPMV activates human monocytes in a manner dependent on SYK signalling, endosomal acidification and with an important contribution from TLR7/8 recognition.
Collapse
Affiliation(s)
- Marwah M. Albakri
- Department of PathologySchool of MedicineCase Western Reserve UniversityClevelandOHUSA
- Department of Medical Laboratory TechnologyCollege of Applied Medical SciencesTaibah UniversityMedinaSaudi Arabia
| | - Frank A. Veliz
- Department of Biomedical EngineeringSchool of MedicineCase Western Reserve UniversityClevelandOHUSA
| | - Steven N. Fiering
- Department of Microbiology and ImmunologyGeisel School of Medicine at DartmouthNorris Cotton Cancer CenterLebanonNHUSA
| | - Nicole F. Steinmetz
- Department of NanoEngineeringUniversity of California San DiegoLa JollaCAUSA
- Department of RadiologyUniversity of California San DiegoLa JollaCAUSA
- Department of BioengineeringUniversity of California San DiegoLa JollaCAUSA
- Moores Cancer CenterUniversity of California San DiegoLa JollaCAUSA
| | - Scott F. Sieg
- Division of Infectious Diseases and HIV MedicineSchool of MedicineCase Western Reserve UniversityClevelandOHUSA
| |
Collapse
|
32
|
Paris JL, Villaverde G, Gómez-Graña S, Vallet-Regí M. Nanoparticles for multimodal antivascular therapeutics: Dual drug release, photothermal and photodynamic therapy. Acta Biomater 2020; 101:459-468. [PMID: 31706040 PMCID: PMC7616912 DOI: 10.1016/j.actbio.2019.11.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 10/24/2019] [Accepted: 11/01/2019] [Indexed: 12/13/2022]
Abstract
The poor delivery of nanoparticles to target cancer cells hinders their success in the clinical setting. In this work, an alternative target readily available for circulating nanoparticles has been selected to eliminate the need for nanoparticle penetration in the tissue: the tumor blood vessels. A tumor endothelium-targeted nanoparticle (employing an RGD-containing peptide) capable of co-delivering two anti-vascular drugs (one anti-angiogenic drug and one vascular disruption agent) is here presented. Furthermore, the nanodevice presents two additional anti-vascular capabilities upon activation by Near-Infrared light: provoking local hyperthermia (by gold nanorods in the system) and generating toxic reactive oxygen species (by the presence of a photosensitizer). RGD-targeting is shown to increase uptake by HUVEC cells, and while the nanoparticles are shown not to be toxic for these cells, upon Near-Infrared irradiation their almost complete killing is achieved. The combination of all four therapeutic modalities is then evaluated in an ex ovo fibrosarcoma xenograft model, which shows a significant reduction in the number of blood vessels irrigating the xenografts when the nanoparticles are present, as well as the destruction of the existing blood vessels upon irradiation. These results suggest that the combination of different anti-vascular therapeutic strategies in a single nanocarrier appears promising and should be further explored in the future. STATEMENT OF SIGNIFICANCE MVR2019: The combination of antivascular drugs with different mechanisms of action (such as antiangiogenic drugs and vascular disruption agents) has been recently proposed as a promising approach to maximize the therapeutic potential of anti-vascular therapeutics. Given the capacity of nanoparticles to co-deliver different drugs in optimizable ratios, nanomedicine appears to have a huge potential for the development of this kind of multimodal antivascular. To showcase this, an multimodal anti-vascular nanodevice for cancer therapy is here presented. This tumor endothelium-targeted nanosystem is capable of co-delivering two anti-vascular drugs (anti-angiogenic and vascular disruption agent) while also providing two additional therapeutic modalities that can be activated by Near-Infrared light: provoking local hyperthermia (photothermal therapy) and generating toxic reactive oxygen species (photodynamic therapy).
Collapse
Affiliation(s)
- Juan L Paris
- Dpto. Química en Ciencias Farmacéuticas (Unidad Docente de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| | - Gonzalo Villaverde
- Dpto. Química en Ciencias Farmacéuticas (Unidad Docente de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Sergio Gómez-Graña
- Dpto. Química en Ciencias Farmacéuticas (Unidad Docente de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - María Vallet-Regí
- Dpto. Química en Ciencias Farmacéuticas (Unidad Docente de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| |
Collapse
|
33
|
Wu J, Wu H, Nakagawa S, Gao J. Virus-derived materials: bury the hatchet with old foes. Biomater Sci 2020; 8:1058-1072. [DOI: 10.1039/c9bm01383k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Viruses, with special architecture and unique biological nature, can be utilized for various biomedical applications.
Collapse
Affiliation(s)
- Jiahe Wu
- Institute of Pharmaceutics
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou 310058
- China
| | - Honghui Wu
- Institute of Pharmaceutics
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou 310058
- China
| | - Shinsaku Nakagawa
- Department of Pharmaceutics
- Graduate School of Pharmaceutical Sciences
- Osaka University
- Suita
- Japan
| | - Jianqing Gao
- Institute of Pharmaceutics
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou 310058
- China
| |
Collapse
|
34
|
Schneider-Stock R, Ribatti D. The CAM Assay as an Alternative In Vivo Model for Drug Testing. Handb Exp Pharmacol 2020; 265:303-323. [PMID: 32776283 DOI: 10.1007/164_2020_375] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the last decade, the chicken chorioallantoic membrane (CAM) assay has been re-discovered in cancer research to study the molecular mechanisms of anti-cancer drug effects. Literature about the CAM assay as an alternative in vivo cancer xenograft model according to the 3R principles has exploded in the last 3 years. Following a summary of the basic knowledge about the chicken embryo, we compare advantages and disadvantages with the classical mouse xenograft model, exemplify established and innovative imaging techniques that are used in the CAM model, and give examples of its successful utilization for studying major hallmarks of cancer such as angiogenesis, proliferation, invasion, and metastasis.
Collapse
Affiliation(s)
- Regine Schneider-Stock
- Experimental Tumorpathology, Institute of Pathology, University Hospital, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| |
Collapse
|
35
|
Leong HS, Butler KS, Brinker CJ, Azzawi M, Conlan S, Dufés C, Owen A, Rannard S, Scott C, Chen C, Dobrovolskaia MA, Kozlov SV, Prina-Mello A, Schmid R, Wick P, Caputo F, Boisseau P, Crist RM, McNeil SE, Fadeel B, Tran L, Hansen SF, Hartmann NB, Clausen LPW, Skjolding LM, Baun A, Ågerstrand M, Gu Z, Lamprou DA, Hoskins C, Huang L, Song W, Cao H, Liu X, Jandt KD, Jiang W, Kim BYS, Wheeler KE, Chetwynd AJ, Lynch I, Moghimi SM, Nel A, Xia T, Weiss PS, Sarmento B, das Neves J, Santos HA, Santos L, Mitragotri S, Little S, Peer D, Amiji MM, Alonso MJ, Petri-Fink A, Balog S, Lee A, Drasler B, Rothen-Rutishauser B, Wilhelm S, Acar H, Harrison RG, Mao C, Mukherjee P, Ramesh R, McNally LR, Busatto S, Wolfram J, Bergese P, Ferrari M, Fang RH, Zhang L, Zheng J, Peng C, Du B, Yu M, Charron DM, Zheng G, Pastore C. On the issue of transparency and reproducibility in nanomedicine. NATURE NANOTECHNOLOGY 2019; 14:629-635. [PMID: 31270452 PMCID: PMC6939883 DOI: 10.1038/s41565-019-0496-9] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Affiliation(s)
- Hon S Leong
- Department of Urology, Mayo Clinic, Rochester, MN, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Kimberly S Butler
- Department of Nanobiology, Sandia National Laboratories, Albuquerque, NM, USA
| | - C Jeffrey Brinker
- Center for Micro-Engineered Materials, University of New Mexico Albuquerque, Albuquerque, NM, USA
- Departments of Chemical and Biological Engineering, University of New Mexico, Albuquerque, NM, USA
- Department of Molecular Genetics and Microbiology, University of New Mexico, Albuquerque, NM, USA
- UNM Comprehensive Cancer Center, University of New Mexico, Albuquerque, NM, USA
| | - May Azzawi
- Cardiovascular Research Group, School of Healthcare Science, Manchester Metropolitan University, Manchester, UK
- British Society for Nanomedicine
| | - Steve Conlan
- British Society for Nanomedicine
- Institute of Life Science, Swansea University Medical School, Swansea University, Swansea, UK
| | - Christine Dufés
- British Society for Nanomedicine
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - Andrew Owen
- British Society for Nanomedicine
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Steve Rannard
- British Society for Nanomedicine
- Department of Chemistry, School of Physical Sciences, University of Liverpool, Liverpool, UK
| | - Chris Scott
- British Society for Nanomedicine
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, Belfast, UK
| | - Chunying Chen
- National Center for Nanoscience and Technology of China, Beijing, China
| | - Marina A Dobrovolskaia
- Cancer Research Technology Program, Nanotechnology Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Laboratory of Animal Sciences Program, Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Serguei V Kozlov
- Laboratory of Animal Sciences Program, Center for Advanced Preclinical Research, Frederick National Laboratory for Cancer Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Adriele Prina-Mello
- Trinity Translational Medicine Institute, Department of Clinical Medicine, Trinity College Dublin, Dublin, Ireland
- Laboratory for Biological Characterisation of Advanced Materials, Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
- Nanomedicine Group, Advanced Materials and Bioengineering Research (AMBER) centre, Centre for Research on Adaptive Nanostructures and Nanodevices, Trinity College Dublin, Dublin, Ireland
| | | | - Peter Wick
- Empa - Swiss Federal Laboratories for Materials Science and Technology, St Gallen, Switzerland
| | - Fanny Caputo
- University Grenoble Alpes, CEA, LETI, Grenoble, Switzerland
| | | | - Rachael M Crist
- Cancer Research Technology Program, Nanotechnology Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Scott E McNeil
- Cancer Research Technology Program, Nanotechnology Characterization Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Bengt Fadeel
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lang Tran
- Institute of Occupational Medicine, Edinburgh, UK
| | - Steffen Foss Hansen
- Department of Environmental Engineering, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Nanna B Hartmann
- Department of Environmental Engineering, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lauge P W Clausen
- Department of Environmental Engineering, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lars M Skjolding
- Department of Environmental Engineering, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Anders Baun
- Department of Environmental Engineering, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Marlene Ågerstrand
- Department of Environmental Science and Analytical Chemistry (ACES), Stockholm University, Stockholm, Sweden
| | - Zhen Gu
- Department of Bioengineering, California Nanosystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Clare Hoskins
- Institute of Science and Technology in Medicine, Keele University, Keele, UK
| | - Leaf Huang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - Wantong Song
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, China
| | - Huiliang Cao
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
- Otto Schott Institute of Materials Research, Friedrich Schiller University Jena, Jena, Germany
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, China
| | - Klaus D Jandt
- Otto Schott Institute of Materials Research, Friedrich Schiller University Jena, Jena, Germany
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer, Houston, TX, USA
| | - Korin E Wheeler
- Department of Chemistry and Biochemistry, Santa Clara University, Santa Clara, CA, USA
| | - Andrew J Chetwynd
- School of Geography Earth and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Iseult Lynch
- School of Geography Earth and Environmental Sciences, University of Birmingham, Birmingham, UK
| | - Seyed Moein Moghimi
- School of Pharmacy, Newcastle University, Newcastle upon Tyne, UK
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| | - André Nel
- Division of NanoMedicine, Department of Medicine, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Tian Xia
- Division of NanoMedicine, Department of Medicine, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Paul S Weiss
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Materials Science and Engineering, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - José das Neves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Hélder A Santos
- Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Luis Santos
- Dosage Form Design and Development, MedImmune, LLC, Gaithersburg, MD, USA
| | - Samir Mitragotri
- Wyss Institute of Biologically Inspired Engineering, Harvard University, Cambridge, MA, USA
| | - Steve Little
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dan Peer
- George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Mansoor M Amiji
- School of Pharmacy, Northeastern University, Boston, MA, USA
| | - Maria José Alonso
- CIMUS Research Institute, Universidade de Santiago de Compostela, Santiago de Compostela, Spain
| | - Alke Petri-Fink
- Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
| | - Sandor Balog
- Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
| | - Aaron Lee
- Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
| | - Barbara Drasler
- Adolphe Merkle Institute, University of Fribourg, Fribourg, Switzerland
| | | | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
- Stephenson Cancer Center, Oklahoma City, OK, USA
| | - Handan Acar
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
- Stephenson Cancer Center, Oklahoma City, OK, USA
| | - Roger G Harrison
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
- Stephenson Cancer Center, Oklahoma City, OK, USA
- School of Chemical, Biological and Materials Engineering, University of Oklahoma, Norman, OK, USA
| | - Chuanbin Mao
- Stephenson Cancer Center, Oklahoma City, OK, USA
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, OK, USA
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang, China
| | - Priyabrata Mukherjee
- Stephenson Cancer Center, Oklahoma City, OK, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rajagopal Ramesh
- Stephenson Cancer Center, Oklahoma City, OK, USA
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Lacey R McNally
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC, USA
- Department of Bioengineering, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Sara Busatto
- Department of Transplantation Medicine, Mayo Clinic, Jacksonville, FL, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, USA
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- CSGI, Research Center for Colloids and Nanoscience, Florence, Italy
| | - Joy Wolfram
- Department of Transplantation Medicine, Mayo Clinic, Jacksonville, FL, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Jacksonville, FL, USA
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
| | - Paolo Bergese
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
- CSGI, Research Center for Colloids and Nanoscience, Florence, Italy
| | - Mauro Ferrari
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA
- Department of Medicine, Weill Cornell Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ronnie H Fang
- Department of NanoEngineering, Chemical Engineering Program, University of California, San Diego, La Jolla, CA, USA
| | - Liangfang Zhang
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Jie Zheng
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, USA
| | - Chuanqi Peng
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, USA
| | - Bujie Du
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, USA
| | - Mengxiao Yu
- Department of Chemistry and Biochemistry, The University of Texas at Dallas, Richardson, TX, USA
| | - Danielle M Charron
- Institute of Biomaterials and Biomedical Engineering, University of Toronto Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Gang Zheng
- Department of Medical Biophysics, University of Toronto Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | | |
Collapse
|
36
|
Hu H, Masarapu H, Gu Y, Zhang Y, Yu X, Steinmetz NF. Physalis Mottle Virus-like Nanoparticles for Targeted Cancer Imaging. ACS APPLIED MATERIALS & INTERFACES 2019; 11:18213-18223. [PMID: 31074602 PMCID: PMC7060085 DOI: 10.1021/acsami.9b03956] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
One of the greatest challenges in nanomedicine is the low efficiency with which nanoparticles are delivered to lesions such as tumors in vivo. Here, we show that Physalis mottle virus (PhMV)-like nanoparticles can be developed as bimodal contrast agents to achieve long circulation, specific targeting capability, and efficient delivery to tumors in vivo. The self-assembling coat protein nanostructure offers various opportunities to modify the internal and external surfaces separately. After loading the internal cavity of the particles with the fluorescent dye Cy5.5 and paramagnetic Gd(III) complexes, we modified the outer surface by PEGylation and conjugation with targeting peptides. Using this combined approach, we were able to monitor a human prostate tumor model for up to 10 days by near-infrared fluorescence and magnetic resonance imaging, with up to 6% of the injection dose remaining. Our results show that PhMV-like nanoparticles provide a promising and innovative platform for the development of next-generation diagnostic and therapeutic agents.
Collapse
Affiliation(s)
- He Hu
- Department of NanoEngineering, University of California—San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Hema Masarapu
- Department of Virology, Sri Venkateswara University, Tirupati 517502, Andhra Pradesh, India
| | - Yuning Gu
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Yifan Zhang
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Xin Yu
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Nicole F. Steinmetz
- Department of NanoEngineering, University of California—San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Moores Cancer Center, University of California—San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Radiology, University of California—San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Bioengineering, University of California—San Diego, 9500 Gilman Drive, La Jolla, California 92093, United States
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| |
Collapse
|
37
|
Beatty PH, Lewis JD. Cowpea mosaic virus nanoparticles for cancer imaging and therapy. Adv Drug Deliv Rev 2019; 145:130-144. [PMID: 31004625 DOI: 10.1016/j.addr.2019.04.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 12/07/2018] [Accepted: 04/15/2019] [Indexed: 12/12/2022]
Abstract
Nanoparticle platforms are particularly attractive for theranostic applications due to their capacity for multifunctionality and multivalency. Some of the most promising nano-scale scaffold systems have been co-opted from nature including plant viruses such as cowpea mosaic virus (CPMV). The use of plant viruses like CPMV as viral nanoparticles is advantageous for many reasons; they are non-infectious and nontoxic to humans and safe for use in intravital imaging and drug delivery. The CPMV capsid icosahedral shape allows for enhanced multifunctional group display and the ability to carry specific cargoes. The native tropism of CPMV for cell-surface displayed vimentin and the enhanced permeability and retention effect allow them to preferentially extravasate from tumor neovasculature and efficiently penetrate tumors. Furthermore, CPMVs can be engineered via several straightforward chemistries to display targeting and imaging moieties on external, addressable residues and they can be loaded internally with therapeutic drug cargoes. These qualities make them highly effective as biocompatible platforms for tumor targeting, intravital imaging and cancer therapy.
Collapse
|
38
|
Said SS, Yin H, Elfarnawany M, Nong Z, O'Neil C, Leong H, Lacefield JC, Mequanint K, Pickering JG. Fortifying Angiogenesis in Ischemic Muscle with FGF9-Loaded Electrospun Poly(Ester Amide) Fibers. Adv Healthc Mater 2019; 8:e1801294. [PMID: 30785239 DOI: 10.1002/adhm.201801294] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 02/03/2019] [Indexed: 12/24/2022]
Abstract
Delivery of angiogenic growth factors lessens ischemia in preclinical models but has demonstrated little benefit in patients with peripheral vascular disease. Augmenting the wrapping of nascent microvessels by mural cells constitutes an alternative strategy to regenerating a functional microvasculature, particularly if integrated with a sustained delivery platform. Herein, electrospun poly(ester amide) (PEA) nanofiber mats are fabricated for delivering a mural cell-targeting factor, fibroblast growth factor 9 (FGF9). Proof-of-principle is established by placing FGF9/FGF2-loaded PEA fiber mats on the chick chorioallantoic membrane and identifying enhanced angiogenesis by 3D power Doppler micro-ultrasound imaging. To assess the delivery system in ischemic muscle, FGF9-loaded PEA fiber mats are implanted onto the surface of the tibialis anterior muscle of mice with hindlimb ischemia. The system supplies FGF9 into the tibialis anterior muscle and yields a neo-microvascular network with enhanced mural cell coverage up to 28 days after injury. The regenerating muscle that receives FGF9 display near-normal sized myofibers and reduced interstitial fibrosis. Moreover, the mice demonstrate improved locomotion. These findings of locally released FGF9 from PEA nanofibers raise prospects for a microvascular remodeling approach to improve muscle health in peripheral vascular disease.
Collapse
Affiliation(s)
- Somiraa S. Said
- School of Biomedical EngineeringWestern University London Ontario N6A 5B9 Canada
| | - Hao Yin
- Robarts Research InstituteWestern University London Ontario N6A 5B7 Canada
| | - Mai Elfarnawany
- Department of OtolaryngologyWestern University London Ontario N6A 5W9 Canada
| | - Zengxuan Nong
- Robarts Research InstituteWestern University London Ontario N6A 5B7 Canada
| | - Caroline O'Neil
- Robarts Research InstituteWestern University London Ontario N6A 5B7 Canada
| | - Hon Leong
- Department of Surgery (Urology)Schulich School of Medicine and DentistryWestern University London Ontario N6A 5C1 Canada
| | - James C. Lacefield
- School of Biomedical EngineeringDepartment of Electrical and Computer EngineeringDepartment of Medical BiophysicsWestern University London Ontario N6A 5B9 Canada
| | - Kibret Mequanint
- School of Biomedical EngineeringDepartment of Chemical and Biochemical EngineeringWestern University London Ontario N6A 5B9 Canada
| | - J. Geoffrey Pickering
- Robarts Research InstituteDepartment of Medicine (Cardiology)Department of BiochemistryDepartment of Medical BiophysicsSchulich School of Medicine and DentistryWestern UniversityLondon Health Sciences Centre 339 Windermere Rd London Ontario N6A 5A5 Canada
| |
Collapse
|
39
|
Paris JL, Lafuente-Gómez N, Cabañas MV, Román J, Peña J, Vallet-Regí M. Fabrication of a nanoparticle-containing 3D porous bone scaffold with proangiogenic and antibacterial properties. Acta Biomater 2019; 86:441-449. [PMID: 30654210 PMCID: PMC6667335 DOI: 10.1016/j.actbio.2019.01.013] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/09/2019] [Accepted: 01/10/2019] [Indexed: 12/19/2022]
Abstract
3D porous scaffolds based on agarose and nanocrystalline apatite, two structural components that act as a temporary mineralized extracellular matrix, were prepared by the GELPOR3D method. This shaping technology allows the introduction of thermally-labile molecules within the scaffolds during the fabrication procedure. An angiogenic protein, Vascular Endothelial Growth Factor, and an antibiotic, cephalexin, loaded in mesoporous silica nanoparticles, were included to design multifunctional scaffolds for bone reconstruction. The dual release of both molecules showed a marked increase in the number of blood vessels on embryonic day 14 in chicken embryos grown ex ovo, while, at the same time providing an antibiotic local concentration capable of inhibiting Staphylococcus aureus bacterial growth. In this sense, different release patterns, monitored by UV-spectroscopy, could be tailored as a function of the cephalexin loading strategy, either releasing all the loaded cephalexin in the first 4 h or less than 50% after 24 h. The scaffold surface was characterized by a high hydrophilicity, with contact angles between 50° and 63°, which enabled the adhesion and proliferation of preosteoblastic cells. STATEMENT OF SIGNIFICANCE: The localized delivery of bioactive molecules has attracted significant attention due to the potential for dose reduction as well as reduced side effects compared to systemic delivery. In this article multifunctional 3D porous scaffolds with a designed porosity have been fabricated. The method also enables the controlled loading of an antibiotic drug and an angiogenic protein into the scaffold. These scaffolds, whose composition resembles the extracellular matrix are suitable for the adhesion of preosteoblast cells, exhibit a sustained cephalexin delivery adequate for inhibiting bacterial growth as well as release the proangiogenic molecule which induces blood vessel formation in chicken embryos grown ex ovo.
Collapse
Affiliation(s)
- Juan L Paris
- Dpto. Química en Ciencias Farmacéuticas (Unidad de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Nuria Lafuente-Gómez
- Dpto. Química en Ciencias Farmacéuticas (Unidad de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain
| | - M Victoria Cabañas
- Dpto. Química en Ciencias Farmacéuticas (Unidad de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain.
| | - Jesús Román
- Dpto. Química en Ciencias Farmacéuticas (Unidad de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain
| | - Juan Peña
- Dpto. Química en Ciencias Farmacéuticas (Unidad de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain
| | - María Vallet-Regí
- Dpto. Química en Ciencias Farmacéuticas (Unidad de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain.
| |
Collapse
|
40
|
Röder J, Dickmeis C, Commandeur U. Small, Smaller, Nano: New Applications for Potato Virus X in Nanotechnology. FRONTIERS IN PLANT SCIENCE 2019; 10:158. [PMID: 30838013 PMCID: PMC6390637 DOI: 10.3389/fpls.2019.00158] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 01/29/2019] [Indexed: 05/08/2023]
Abstract
Nanotechnology is an expanding interdisciplinary field concerning the development and application of nanostructured materials derived from inorganic compounds or organic polymers and peptides. Among these latter materials, proteinaceous plant virus nanoparticles have emerged as a key platform for the introduction of tailored functionalities by genetic engineering and conjugation chemistry. Tobacco mosaic virus and Cowpea mosaic virus have already been developed for bioimaging, vaccination and electronics applications, but the flexible and filamentous Potato virus X (PVX) has received comparatively little attention. The filamentous structure of PVX particles allows them to carry large payloads, which are advantageous for applications such as biomedical imaging in which multi-functional scaffolds with a high aspect ratio are required. In this context, PVX achieves superior tumor homing and retention properties compared to spherical nanoparticles. Because PVX is a protein-based nanoparticle, its unique functional properties are combined with enhanced biocompatibility, making it much more suitable for biomedical applications than synthetic nanomaterials. Moreover, PVX nanoparticles have very low toxicity in vivo, and superior pharmacokinetic profiles. This review focuses on the production of PVX nanoparticles engineered using chemical and/or biological techniques, and describes current and future opportunities and challenges for the application of PVX nanoparticles in medicine, diagnostics, materials science, and biocatalysis.
Collapse
Affiliation(s)
| | | | - Ulrich Commandeur
- Institute for Molecular Biotechnology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
41
|
Le DHT, Méndez-López E, Wang C, Commandeur U, Aranda MA, Steinmetz NF. Biodistribution of Filamentous Plant Virus Nanoparticles: Pepino Mosaic Virus versus Potato Virus X. Biomacromolecules 2019; 20:469-477. [PMID: 30516960 PMCID: PMC6485256 DOI: 10.1021/acs.biomac.8b01365] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Nanoparticles with high aspect ratios have favorable attributes for drug delivery and bioimaging applications based on their enhanced tissue penetration and tumor homing properties. Here, we investigated a novel filamentous viral nanoparticle (VNP) based on the Pepino mosaic virus (PepMV), a relative of the established platform Potato virus X (PVX). We studied the chemical reactivity of PepMV, produced fluorescent versions of PepMV and PVX, and then evaluated their biodistribution in mouse tumor models. We found that PepMV can be conjugated to various small chemical modifiers including fluorescent probes via the amine groups of surface-exposed lysine residues, yielding VNPs carrying payloads of up to 1600 modifiers per particle. Although PepMV and PVX share similarities in particle size and shape, PepMV achieved enhanced tumor homing and less nonspecific tissue distribution compared to PVX in mouse models of triple negative breast cancer and ovarian cancer. In conclusion, PepMV provides a novel tool for nanomedical research but more research is needed to fully exploit the potential of plant VNPs for health applications.
Collapse
Affiliation(s)
- Duc H. T. Le
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| | - Eduardo Méndez-López
- Centro de Edafología y Biología Aplicada del Segura (CEBAS)-CSIC, Campus Universitario de Espinardo, 30100 Murcia, Spain
| | - Chao Wang
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, United States
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| | - Ulrich Commandeur
- Department of Molecular Biology, RWTH-Aachen University, Aachen 52064, Germany
| | - Miguel A. Aranda
- Centro de Edafología y Biología Aplicada del Segura (CEBAS)-CSIC, Campus Universitario de Espinardo, 30100 Murcia, Spain
| | - Nicole F. Steinmetz
- Department of NanoEngineering, University of California, San Diego, La Jolla, California 92093, United States
- Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, United States
- Department of Radiology, University of California, San Diego, La Jolla, California 92093, United States
- Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
- Department of Biomedical Engineering, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, United States
| |
Collapse
|
42
|
Virus-like nanoparticles: emerging tools for targeted cancer diagnostics and therapeutics. Ther Deliv 2018; 8:1019-1021. [PMID: 29125065 DOI: 10.4155/tde-2017-0098] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
43
|
Lam P, Lin R, Steinmetz NF. Delivery of mitoxantrone using a plant virus-based nanoparticle for the treatment of glioblastomas. J Mater Chem B 2018; 6:5888-5895. [PMID: 30923616 PMCID: PMC6433411 DOI: 10.1039/c8tb01191e] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Mitoxatrone (MTO), an antineoplastic chemotherapeutic, has potent activity against the most common and agressive type of primary brain tumor, glioblastoma multiforme (GBM). However, its poor penetration through the blood brain barrier, and cardiotoxic side effects from systemic delivery limit its effectiveness for clinical treatment. To address these limitations, we utilize a plant virus-based nanoparticle, cowpea mosaic virus (CPMV), to deliver MTO to treat GBM. In this work, we loaded MTO into the interior cavity of CPMV (CPMV-MTO) through diffusion through its pores. We report the uptake of CPMV-MTO in glioma cells and demonstrate its cytotoxic effects in vitro as a solo therapy, and in combination with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). These results reveal the potential for this plant virus-based nanoparticle platform for the treatment of GBM.
Collapse
Affiliation(s)
- Patricia Lam
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Richard Lin
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Nicole F. Steinmetz
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of NanoEngineering, University of California, San Diego, La Jolla, CA 92093, USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
44
|
Patel R, Czapar AE, Fiering S, Oleinick NL, Steinmetz NF. Radiation Therapy Combined with Cowpea Mosaic Virus Nanoparticle in Situ Vaccination Initiates Immune-Mediated Tumor Regression. ACS OMEGA 2018; 3:3702-3707. [PMID: 29732445 PMCID: PMC5928490 DOI: 10.1021/acsomega.8b00227] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 03/21/2018] [Indexed: 05/17/2023]
Abstract
Epithelial ovarian cancer is a deadly gynecologic malignancy because of its late detection, usually after local and distant metastatic spread. These cancers develop resistance to traditional chemotherapeutic agents; therefore, the development of next-generation immunotherapeutic approaches may have a significant promise in improving outcomes. A novel immunotherapeutic approach utilizing combination radiation therapy (RT) with immunostimulatory cowpea mosaic virus (CPMV) was tested in a preclinical syngeneic mouse model of ovarian carcinoma. ID8-Defb29/Vegf tumors were generated in C57BL/6 mice. Compared to placebo-treated control tumors or those treated with a single agent RT or CPMV, the combination treatment resulted in a significantly improved tumor growth delay (p < 0.05). Additionally, immunohistochemical profiling of tumor samples after treatment with CPMV demonstrated an increase in tumor infiltrating lymphocytes (TILs). These results suggest that utilizing CPMV particles in combination with RT can turn an immunologically "cold" tumor (with low number of TILs) into an immunologically "hot" tumor. This novel combination treatment approach of RT and CPMV demonstrated the ability to control tumor growth in a preclinical ID8 ovarian cancer model, showing promise as an in situ tumor vaccine and warrants further testing.
Collapse
Affiliation(s)
- Ravi Patel
- Department
of Radiation Oncology, Department of Pathology, Department of Biomedical Engineering, Department of Radiology, Department of Materials
Science and Engineering, Department of Macromolecular Science and Engineering, and Case Comprehensive
Cancer Center, Division of General Medical Sciences-Oncology, Case Western Reserve University, 109000 Euclid Av., Cleveland, Ohio 44106, United States
| | - Anna E. Czapar
- Department
of Radiation Oncology, Department of Pathology, Department of Biomedical Engineering, Department of Radiology, Department of Materials
Science and Engineering, Department of Macromolecular Science and Engineering, and Case Comprehensive
Cancer Center, Division of General Medical Sciences-Oncology, Case Western Reserve University, 109000 Euclid Av., Cleveland, Ohio 44106, United States
| | - Steven Fiering
- Department
of Microbiology and Immunology, Geisel School
of Medicine at Dartmouth, 1 Rope Ferry Rd., Hanover, New Hampshire 03755, United States
| | - Nancy L. Oleinick
- Department
of Radiation Oncology, Department of Pathology, Department of Biomedical Engineering, Department of Radiology, Department of Materials
Science and Engineering, Department of Macromolecular Science and Engineering, and Case Comprehensive
Cancer Center, Division of General Medical Sciences-Oncology, Case Western Reserve University, 109000 Euclid Av., Cleveland, Ohio 44106, United States
| | - Nicole F. Steinmetz
- Department
of Radiation Oncology, Department of Pathology, Department of Biomedical Engineering, Department of Radiology, Department of Materials
Science and Engineering, Department of Macromolecular Science and Engineering, and Case Comprehensive
Cancer Center, Division of General Medical Sciences-Oncology, Case Western Reserve University, 109000 Euclid Av., Cleveland, Ohio 44106, United States
- E-mail: (N.F.S.)
| |
Collapse
|
45
|
Choi B, Kim H, Choi H, Kang S. Protein Cage Nanoparticles as Delivery Nanoplatforms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:27-43. [DOI: 10.1007/978-981-13-0445-3_2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
46
|
Veliz FA, Ma Y, Molugu SK, Tiu BDB, Stewart PL, French RH, Steinmetz NF. Photon Management through Virus-Programmed Supramolecular Arrays. ACTA ACUST UNITED AC 2017; 1:e1700088. [PMID: 32646196 DOI: 10.1002/adbi.201700088] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/20/2017] [Indexed: 11/06/2022]
Abstract
Photon extraction and capture efficiency is a complex function of the material's composition, its molecular structure at the nanoscale, and the overall organization spanning multiple length scales. The architecture of the material defines the performance; nanostructured features within the materials enhance the energy efficiency. Photon capturing materials are largely produced through lithographic, top-down, manufacturing schemes; however, there are limits to the smallest dimension achievable using this technology. To overcome these technological barriers, a bottom-up nanomanufacturing is pursued. Inspired by the self-programmed assembly of virus arrays in host cells resulting in iridescence of infected organisms, virus-programmed, nanostructured arrays are studied to pave the way for new design principles in photon management and biology-inspired materials science. Using the nanoparticles formed by plant viruses in combination with charged polymers (dendrimers), a bottom-up approach is illustrated to prepare a family of broadband, low-angular dependent antireflection mesoscale layered materials for potential application as photon management coatings. Measurement and theory demonstrate antireflectance and phototrapping properties of the virus-programmed assembly. This opens up new bioengineering principles for the nanomanufacture of coatings and films for use in LED lighting and photovoltaics.
Collapse
Affiliation(s)
- Frank A Veliz
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA
| | - Yingfang Ma
- Department of Materials Science and Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA
| | - Sudheer K Molugu
- Department of Pharmacology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA.,Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA
| | - Brylee David B Tiu
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA.,Department of Macromolecular Science and Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA
| | - Phoebe L Stewart
- Department of Pharmacology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA.,Cleveland Center for Membrane and Structural Biology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA
| | - Roger H French
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA.,Department of Materials Science and Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA.,Department of Macromolecular Science and Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA.,Department of Physics, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA
| | - Nicole F Steinmetz
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA.,Department of Materials Science and Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA.,Department of Macromolecular Science and Engineering, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA.,Department of Radiology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA.,Division of General Medical Sciences-Oncology, Case Western Reserve University, 10900 Euclid Ave., Cleveland, OH, 44106, USA
| |
Collapse
|
47
|
Cho CF, Yu L, Nsiama TK, Kadam AN, Raturi A, Shukla S, Amadei GA, Steinmetz NF, Luyt LG, Lewis JD. Viral nanoparticles decorated with novel EGFL7 ligands enable intravital imaging of tumor neovasculature. NANOSCALE 2017; 9:12096-12109. [PMID: 28799610 PMCID: PMC5770569 DOI: 10.1039/c7nr02558k] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Angiogenesis is a dynamic process fundamental to the development of solid tumors. Epidermal growth factor-like domain 7 (EGFL7) is a protein whose expression is restricted to endothelial cells undergoing active remodeling that has emerged as a key mediator of this process. EGFL7 expression is associated with poor outcome in several cancers, making it a promising target for imaging or therapeutic strategies. Here, EGFL7 is explored as a molecular target for active neovascularization. Using a combinatorial peptide screening approach, we describe the discovery and characterization of a novel high affinity EGFL7-binding peptide, E7p72, that specifically targets human endothelial cells. Viral nanoparticles decorated with E7p72 peptides specifically target tumor-associated neovasculature with high specificity as assessed by intravital imaging. This work highlights the value of EGFL7 as a target for angiogenic vessels and opens the door for novel targeted therapeutic approaches.
Collapse
Affiliation(s)
- Choi-Fong Cho
- Translational Prostate Cancer Research Group, University of Alberta, 5-142C Katz Group Building, 114th St and 87th Ave, Edmonton, AB T6G 2E1, Canada.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Lee KL, Murray AA, Le DHT, Sheen MR, Shukla S, Commandeur U, Fiering S, Steinmetz NF. Combination of Plant Virus Nanoparticle-Based in Situ Vaccination with Chemotherapy Potentiates Antitumor Response. NANO LETTERS 2017; 17. [PMID: 28650644 PMCID: PMC5623935 DOI: 10.1021/acs.nanolett.7b00107] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Immunotherapeutics are gaining more traction in the armamentarium used to combat cancer. Specifically, in situ vaccination strategies have gained interest because of their ability to alter the tumor microenvironment to an antitumor state. Herein, we investigate whether flexuous plant virus-based nanoparticles formed by the potato virus X (PVX) can be used as an immunotherapeutic for in situ vaccine monotherapy. We further developed dual chemo-immunotherapeutics by incorporating doxorubicin (DOX) into PVX yielding a dual-functional nanoparticle (PVX-DOX) or by coadministration of the two therapeutic regimes, PVX immunotherapy and DOX chemotherapy (PVX+DOX). In the context of B16F10 melanoma, PVX was able to elicit delayed tumor progression when administered as an intratumoral in situ vaccine. Furthermore, the coadministration of DOX via PVX+DOX enhanced the response of the PVX monotherapy through increased survival, which was also represented in the enhanced antitumor cytokine/chemokine profile stimulated by PVX+DOX when compared to PVX or DOX alone. Importantly, coadministered PVX+DOX was better for in situ vaccination than PVX loaded with DOX (PVX-DOX). Whereas the nanomedicine field strives to design multifunctional nanoparticles that integrate several functions and therapeutic regimens into a single nanoparticle, our data suggest a paradigm shift; some therapeutics may need to be administered separately to synergize and achieve the most potent therapeutic outcome. Altogether, our studies show that development of plant viral nanoparticles for in situ vaccines for treatment is a possibility, and dual mechanistic therapeutics can increase efficacy. Nonetheless, combining immunotherapeutics with cytolytic chemotherapy requires detailed investigation to inform optimal integration of cytolytic and immunotherapies and maximize synergy and efficacy.
Collapse
Affiliation(s)
- Karin L. Lee
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Abner A. Murray
- Department of Microbiology and Molecular Biology, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Duc H. T. Le
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Mee Rie Sheen
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755, United States
| | - Sourabh Shukla
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
| | - Ulrich Commandeur
- Department of Molecular Biotechnology, RWTH-Aachen University, 52064 Aachen, Germany
| | - Steven Fiering
- Department of Microbiology and Immunology, The Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755, United States
- Norris Cotton Cancer Center, Lebanon, New Hampshire 03756, United States
| | - Nicole F. Steinmetz
- Department of Biomedical Engineering, Case Western Reserve University Schools of Medicine and Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
- Department of Radiology, Case Western Reserve University School of Medicine, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
- Department of Materials Science and Engineering, Case Western Reserve University School of Engineering, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
- Department of Macromolecular Science and Engineering, Case Western Reserve University School of Engineering, 10900 Euclid Ave., Cleveland, Ohio 44106, United States
- Division of General Medical Sciences-Oncology, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, Ohio 44106, United States
- Corresponding Author:
| |
Collapse
|
49
|
Steele JFC, Peyret H, Saunders K, Castells‐Graells R, Marsian J, Meshcheriakova Y, Lomonossoff GP. Synthetic plant virology for nanobiotechnology and nanomedicine. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9:e1447. [PMID: 28078770 PMCID: PMC5484280 DOI: 10.1002/wnan.1447] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/12/2016] [Accepted: 11/23/2016] [Indexed: 12/12/2022]
Abstract
Nanotechnology is a rapidly expanding field seeking to utilize nano-scale structures for a wide range of applications. Biologically derived nanostructures, such as viruses and virus-like particles (VLPs), provide excellent platforms for functionalization due to their physical and chemical properties. Plant viruses, and VLPs derived from them, have been used extensively in biotechnology. They have been characterized in detail over several decades and have desirable properties including high yields, robustness, and ease of purification. Through modifications to viral surfaces, either interior or exterior, plant-virus-derived nanoparticles have been shown to support a range of functions of potential interest to medicine and nano-technology. In this review we highlight recent and influential achievements in the use of plant virus particles as vehicles for diverse functions: from delivery of anticancer compounds, to targeted bioimaging, vaccine production to nanowire formation. WIREs Nanomed Nanobiotechnol 2017, 9:e1447. doi: 10.1002/wnan.1447 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
| | - Hadrien Peyret
- Department of Biology ChemistryJohn Innes CentreNorwichUK
| | - Keith Saunders
- Department of Biology ChemistryJohn Innes CentreNorwichUK
| | | | | | | | | |
Collapse
|
50
|
Hypoxia-induced mobilization of NHE6 to the plasma membrane triggers endosome hyperacidification and chemoresistance. Nat Commun 2017. [PMID: 28635961 PMCID: PMC5482059 DOI: 10.1038/ncomms15884] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The pH-dependent partitioning of chemotherapeutic drugs is a fundamental yet understudied drug distribution mechanism that may underlie the low success rates of current approaches to counter multidrug resistance (MDR). This mechanism is influenced by the hypoxic tumour microenvironment and results in selective trapping of weakly basic drugs into acidified compartments such as the extracellular environment. Here we report that hypoxia not only leads to acidification of the tumour microenvironment but also induces endosome hyperacidification. The acidity of the vesicular lumen, together with the alkaline pH of the cytoplasm, gives rise to a strong intracellular pH gradient that drives intravesicular drug trapping and chemoresistance. Endosome hyperacidification is due to the relocalization of the Na+/H+ exchanger isoform 6 (NHE6) from endosomes to the plasma membrane, an event that involves binding of NHE6 to the activated protein kinase C-receptor for activated C kinase 1 complex. These findings reveal a novel mechanism of hypoxia-induced MDR that involves the aberrant intracellular distribution of NHE6.
Collapse
|