1
|
Shariati A, Khezrpour A, Shariati F, Afkhami H, Yarahmadi A, Alavimanesh S, Kamrani S, Modarressi MH, Khani P. DNA vaccines as promising immuno-therapeutics against cancer: a new insight. Front Immunol 2025; 15:1498431. [PMID: 39872522 PMCID: PMC11769820 DOI: 10.3389/fimmu.2024.1498431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 12/11/2024] [Indexed: 01/30/2025] Open
Abstract
Cancer is one of the leading causes of mortality around the world and most of our conventional treatments are not efficient enough to combat this deadly disease. Harnessing the power of the immune system to target cancer cells is one of the most appealing methods for cancer therapy. Nucleotide-based cancer vaccines, especially deoxyribonucleic acid (DNA) cancer vaccines are viable novel cancer treatments that have recently garnered significant attention. DNA cancer vaccines are made of plasmid molecules that encode tumor-associated or tumor-specific antigens (TAAs or TSAs), and possibly some other immunomodulatory adjuvants such as pro-inflammatory interleukins. Following the internalization of plasmids into cells, their genes are expressed and the tumor antigens are loaded on major histocompatibility molecules to be presented to T-cells. After the T-cells have been activated, they will look for tumor antigens and destroy the tumor cells upon encountering them. As with any other treatment, there are pros and cons associated with using these vaccines. They are relatively safe, usually well-tolerated, stable, easily mass-produced, cost-effective, and easily stored and transported. They can induce a systemic immune response effective on both the primary tumor and metastases. The main disadvantage of DNA vaccines is their poor immunogenicity. Several approaches including structural modification, combination therapy with conventional and novel cancer treatments (such as chemotherapy, radiotherapy, and immune checkpoint blockade (ICB)), and the incorporation of adjuvants into the plasmid structure have been studied to enhance the vaccine's immunogenicity and improve the clinical outcome of cancer patients. In this review, we will discuss some of the most promising optimization strategies and examine some of the important trials regarding these vaccines.
Collapse
Affiliation(s)
- Alireza Shariati
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Arya Khezrpour
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Fatemeh Shariati
- Department of Genetics, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Sajad Alavimanesh
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sina Kamrani
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Hossein Modarressi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Pouria Khani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
2
|
Gül C, Gül A, Karakavuk T, Erkunt Alak S, Karakavuk M, Can H, Değirmenci Döşkaya A, Yavuz İ, Kaplan S, Erel Akbaba G, Şen Karaman D, Akbaba H, Efe Köseoğlu A, Ovayurt T, Yüksel Gürüz A, Ün C, Kantarcı AG, Döşkaya M. A novel DNA vaccine encoding the SRS13 protein administered by electroporation confers protection against chronic toxoplasmosis. Vaccine 2024; 42:126065. [PMID: 38880692 DOI: 10.1016/j.vaccine.2024.06.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/30/2024] [Accepted: 06/11/2024] [Indexed: 06/18/2024]
Abstract
Toxoplasma gondii is an obligate intracellular parasite that can infect a variety of mammals including humans and causes toxoplasmosis. Unfortunately, a protective and safe vaccine against toxoplasmosis hasn't been developed yet. In this study, we developed a DNA vaccine encoding the SRS13 protein and immunized BALB/c mice thrice with pVAX1-SRS13 through the intramuscular route (IM) or intradermally using an electroporation device (ID + EP). The immunogenicity of pVAX1-SRS13 was analyzed by ELISA, Western blot, cytokine ELISA, and flow cytometry. The protective efficacy of the pVAX1-SRS13 was investigated by challenging mice orally with T. gondii PRU strain tissue cysts. The results revealed that pVAX1-SRS13 administered through IM or ID + EP routes induced high level of anti-SRS13 IgG antibody responses (P = 0.0037 and P < 0.0001). The IFN-γ level elicited by the pVAX1-SRS13 (ID + EP) was significantly higher compared to the control group (P = 0.00159). In mice administered with pVAX1-SRS13 (ID + EP), CD8+ cells secreting IFN-γ was significantly higher compared to pVAX1-SRS13 (IM) (P = 0.0035) and the control group (P = 0.0068). Mice vaccinated with the SRS13 DNA vaccine did not induce significant IL-4 level. Moreover, a significant reduction in the number of tissue cysts and the load of T. gondii DNA was detected in brains of mice administered with pVAX1-SRS13 through ID + EP and IM routes compared to controls. In conclusion, the SRS13 DNA vaccine was found to be highly immunogenic and confers strong protection against chronic toxoplasmosis.
Collapse
Affiliation(s)
- Ceren Gül
- Ege University, Graduate School of Natural and Applied Sciences, Department of Biotechnology, İzmir, Türkiye; Ege University, Vaccine Development Application and Research Center, İzmir, Türkiye
| | - Aytül Gül
- Ege University, Vaccine Development Application and Research Center, İzmir, Türkiye; Ege University, Faculty of Engineering, Department of Bioengineering, İzmir, Türkiye
| | - Tuğba Karakavuk
- Ege University, Graduate School of Natural and Applied Sciences, Department of Biotechnology, İzmir, Türkiye; Ege University, Vaccine Development Application and Research Center, İzmir, Türkiye
| | - Sedef Erkunt Alak
- Ege University, Vaccine Development Application and Research Center, İzmir, Türkiye; Ege University, Faculty of Science, Department of Biology Molecular Biology Section, İzmir, Türkiye
| | - Muhammet Karakavuk
- Ege University, Vaccine Development Application and Research Center, İzmir, Türkiye; Ege University, Ödemiş Vocational School, İzmir, Türkiye; Ege University, Institute of Health Sciences, Department of Vaccine Studies, İzmir, Türkiye
| | - Hüseyin Can
- Ege University, Vaccine Development Application and Research Center, İzmir, Türkiye; Ege University, Faculty of Science, Department of Biology Molecular Biology Section, İzmir, Türkiye; Ege University, Institute of Health Sciences, Department of Vaccine Studies, İzmir, Türkiye
| | - Aysu Değirmenci Döşkaya
- Ege University, Vaccine Development Application and Research Center, İzmir, Türkiye; Ege University, Institute of Health Sciences, Department of Vaccine Studies, İzmir, Türkiye; Ege University, Faculty of Medicine, Department of Parasitology, İzmir, Türkiye
| | - İrem Yavuz
- Ege University, Vaccine Development Application and Research Center, İzmir, Türkiye; Ege University, Institute of Health Sciences, Department of Vaccine Studies, İzmir, Türkiye
| | - Seren Kaplan
- Ege University, Vaccine Development Application and Research Center, İzmir, Türkiye; Ege University, Institute of Health Sciences, Department of Vaccine Studies, İzmir, Türkiye
| | - Gülşah Erel Akbaba
- İzmir Katip Çelebi University, Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, İzmir, Türkiye
| | - Didem Şen Karaman
- İzmir Katip Çelebi University, Faculty of Engineering and Architecture, Department of Biomedical Engineering, İzmir, Türkiye
| | - Hasan Akbaba
- Ege University, Institute of Health Sciences, Department of Vaccine Studies, İzmir, Türkiye; Ege University, Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, İzmir, Türkiye
| | - Ahmet Efe Köseoğlu
- Ege University, Faculty of Science, Department of Biology Molecular Biology Section, İzmir, Türkiye; Duisburg-Essen University, Faculty of Chemistry, Department of Environmental Microbiology and Biotechnology, Essen, Germany
| | - Tolga Ovayurt
- İzmir Katip Çelebi University, Graduate School of Natural and Applied Sciences, Department of Biomedical Technology, İzmir, Türkiye
| | - Adnan Yüksel Gürüz
- Ege University, Vaccine Development Application and Research Center, İzmir, Türkiye; Ege University, Institute of Health Sciences, Department of Vaccine Studies, İzmir, Türkiye; Ege University, Faculty of Medicine, Department of Parasitology, İzmir, Türkiye
| | - Cemal Ün
- Ege University, Vaccine Development Application and Research Center, İzmir, Türkiye; Ege University, Faculty of Science, Department of Biology Molecular Biology Section, İzmir, Türkiye; Ege University, Institute of Health Sciences, Department of Vaccine Studies, İzmir, Türkiye
| | - Ayşe Gülten Kantarcı
- Ege University, Vaccine Development Application and Research Center, İzmir, Türkiye; Ege University, Institute of Health Sciences, Department of Vaccine Studies, İzmir, Türkiye; Ege University, Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, İzmir, Türkiye
| | - Mert Döşkaya
- Ege University, Vaccine Development Application and Research Center, İzmir, Türkiye; Ege University, Institute of Health Sciences, Department of Vaccine Studies, İzmir, Türkiye; Ege University, Faculty of Medicine, Department of Parasitology, İzmir, Türkiye
| |
Collapse
|
3
|
Carvalho BG, Nakayama A, Miwa H, Han SW, de la Torre LG, Di Carlo D, Lee J, Kim HJ, Khademhosseini A, de Barros NR. Gelatin methacryloyl granular scaffolds for localized mRNA delivery. AGGREGATE (HOBOKEN, N.J.) 2024; 5:e464. [PMID: 38800607 PMCID: PMC11126212 DOI: 10.1002/agt2.464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
mRNA therapy is the intracellular delivery of messenger RNA (mRNA) to produce desired therapeutic proteins. Developing strategies for local mRNA delivery is still required where direct intra-articular injections are inappropriate for targeting a specific tissue. The mRNA delivery efficiency depends on protecting nucleic acids against nuclease-mediated degradation and safe site-specific intracellular delivery. Herein, we report novel mRNA-releasing matrices based on RGD-moiety-rich gelatin methacryloyl (GelMA) microporous annealed particle (MAP) scaffolds. GelMA concentration in aerogel-based microgels (μgels) produced through a microfluidic process, MAP stiffnesses, and microporosity are crucial parameters for cell adhesion, spreading, and proliferation. After being loaded with mRNA complexes, MAP scaffolds composed of 10 % GelMA μgels display excellent cell viability with increasing cell infiltration, adhesion, proliferation, and gene transfer. The intracellular delivery is achieved by the sustained release of mRNA complexes from MAP scaffolds and cell adhesion on mRNA-releasing scaffolds. These findings highlight that hybrid systems can achieve efficient protein expression by delivering mRNA complexes, making them promising mRNA-releasing biomaterials for tissue engineering.
Collapse
Affiliation(s)
- Bruna Gregatti Carvalho
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), 13083-970, Brazil; Terasaki Institute for Biomedical Innovation (TIBI), 90064, USA
| | - Aya Nakayama
- Terasaki Institute for Biomedical Innovation (TIBI), 90064, USA
| | - Hiromi Miwa
- Department of Bioengineering, University of California at Los Angeles (UCLA), 90095, USA
| | - Sang Won Han
- Center for Cell Therapy and Molecular, Federal University of São Paulo (UNIFESP), 04044-010, Brazil
| | - Lucimara Gaziola de la Torre
- Department of Material and Bioprocess Engineering, School of Chemical Engineering, University of Campinas (UNICAMP), 13083-970, Brazil
| | - Dino Di Carlo
- Department of Bioengineering, University of California at Los Angeles (UCLA), 90095, USA
| | - Junmin Lee
- Department of Materials Science and Engineering, Pohang University of Science and Technology (POSTECH), 37673, Republic of Korea; Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Incheon 21983, Republic of Korea
| | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation (TIBI), 90064, USA; College of Pharmacy, Korea University, 30019, Republic of Korea; Vellore Institute of Technology (VIT), Vellore, 632014, India
| | | | | |
Collapse
|
4
|
Markarian NM, Abrahamyan L. AMDV Vaccine: Challenges and Perspectives. Viruses 2021; 13:v13091833. [PMID: 34578415 PMCID: PMC8472842 DOI: 10.3390/v13091833] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/09/2021] [Accepted: 09/09/2021] [Indexed: 12/13/2022] Open
Abstract
Aleutian mink disease virus (AMDV) is known to cause the most significant disease in the mink industry. It is globally widespread and manifested as a deadly plasmacytosis and hyperglobulinemia. So far, measures to control the viral spread have been limited to manual serological testing for AMDV-positive mink. Further, due to the persistent nature of this virus, attempts to eradicate Aleutian disease (AD) have largely failed. Therefore, effective strategies to control the viral spread are of crucial importance for wildlife protection. One potentially key tool in the fight against this disease is by the immunization of mink against AMDV. Throughout many years, several researchers have tried to develop AMDV vaccines and demonstrated varying degrees of protection in mink by those vaccines. Despite these attempts, there are currently no vaccines available against AMDV, allowing the continuation of the spread of Aleutian disease. Herein, we summarize previous AMDV immunization attempts in mink as well as other preventative measures with the purpose to shed light on future studies designing such a potentially crucial preventative tool against Aleutian disease.
Collapse
Affiliation(s)
- Nathan M. Markarian
- Faculty of Veterinary Medicine, Université de Montréal, Saint-Hyacinthe, QC J2S 2M2, Canada;
| | - Levon Abrahamyan
- Swine and Poultry Infectious Diseases Research Center (CRIPA), Research Group on Infectious Diseases of Production Animals (GREMIP), Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC J2S 2M2, Canada
- Correspondence:
| |
Collapse
|
5
|
Guibinga GH, Sahay B, Brown H, Cooch N, Chen J, Yan J, Reed C, Mishra M, Yung B, Pugh H, Schultheis K, Esquivel RN, Weiner DB, Humeau LH, Broderick KE, Smith TR. Protection against Borreliella burgdorferi infection mediated by a synthetically engineered DNA vaccine. Hum Vaccin Immunother 2020; 16:2114-2122. [PMID: 32783701 PMCID: PMC7553707 DOI: 10.1080/21645515.2020.1789408] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Lyme disease is the most common vector-borne disease in North America. The etiological agent is the spirochete Borreliella burgdorferi, transmitted to mammalian hosts by the Ixodes tick. In recent years there has been an increase in the number of cases of Lyme disease. Currently, there is no vaccine on the market for human use. We describe the development of a novel synthetically engineered DNA vaccine, pLD1 targeting the outer-surface protein A (OspA) of Borreliella burgdorferi. Immunization of C3 H/HeN mice with pLD1 elicits robust humoral and cellular immune responses that confer complete protection against a live Borreliella burgdorferi bacterial challenge. We also assessed intradermal (ID) delivery of pLD1 in Hartley guinea pigs, demonstrating the induction of robust and durable humoral immunity that lasts at least 1 year. We provide evidence of the potency of pLD1 by showing that antibodies targeting the OspA epitopes which have been associated with protection are prominently raised in the immunized guinea pigs. The described study provides the basis for the advancement of pDL1 as a potential vaccine for Lyme disease control.
Collapse
Affiliation(s)
- Ghiabe H. Guibinga
- Department of Research and Development, Inovio Pharmaceuticals, Plymouth Meeting, PA, USA
| | - Bikash Sahay
- College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Heather Brown
- Department of Research and Development, Inovio Pharmaceuticals, Plymouth Meeting, PA, USA
| | - Neil Cooch
- Department of Research and Development, Inovio Pharmaceuticals, Plymouth Meeting, PA, USA
| | - Jing Chen
- Department of Research and Development, Inovio Pharmaceuticals, Plymouth Meeting, PA, USA
| | - Jian Yan
- Department of Research and Development, Inovio Pharmaceuticals, Plymouth Meeting, PA, USA
| | - Charles Reed
- Department of Research and Development, Inovio Pharmaceuticals, Plymouth Meeting, PA, USA
| | - Meerambika Mishra
- College of Veterinary Medicine, University of Florida, Gainesville, FL, USA
| | - Bryan Yung
- Department of Research and Development, Inovio Pharmaceuticals, Plymouth Meeting, PA, USA
| | - Holly Pugh
- Department of Research and Development, Inovio Pharmaceuticals, Plymouth Meeting, PA, USA
| | - Katherine Schultheis
- Department of Research and Development, Inovio Pharmaceuticals, Plymouth Meeting, PA, USA
| | - Rianne N. Esquivel
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, USA
| | - David B. Weiner
- Vaccine and Immunotherapy Center, Wistar Institute, Philadelphia, PA, USA
| | - Laurent H. Humeau
- Department of Research and Development, Inovio Pharmaceuticals, Plymouth Meeting, PA, USA
| | - Kate E. Broderick
- Department of Research and Development, Inovio Pharmaceuticals, Plymouth Meeting, PA, USA
| | - Trevor R.F. Smith
- Department of Research and Development, Inovio Pharmaceuticals, Plymouth Meeting, PA, USA,CONTACT Trevor R.F. Smith Inovio Pharmaceuticals, San Diego, CA92121
| |
Collapse
|
6
|
Ejike UC, Chan CJ, Okechukwu PN, Lim RLH. New advances and potentials of fungal immunomodulatory proteins for therapeutic purposes. Crit Rev Biotechnol 2020; 40:1172-1190. [PMID: 32854547 DOI: 10.1080/07388551.2020.1808581] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Fungal immunomodulatory proteins (FIPs) are fascinating small and heat-stable bioactive proteins in a distinct protein family due to similarities in their structures and sequences. They are found in fungi, including the fruiting bodies producing fungi comprised of culinary and medicinal mushrooms. Structurally, most FIPs exist as homodimers; each subunit consisting of an N-terminal α-helix dimerization and a C-terminal fibronectin III domain. Increasing numbers of identified FIPs from either different or same fungal species clearly indicates the growing research interests into its medicinal properties which include immunomodulatory, anti-inflammation, anti-allergy, and anticancer. Most FIPs increased IFN-γ production in peripheral blood mononuclear cells, potentially exerting immunomodulatory and anti-inflammatory effects by inhibiting overproduction of T helper-2 (Th2) cytokines common in an allergy reaction. Recently, FIP from Ganoderma microsporum (FIP-gmi) was shown to promote neurite outgrowth for potential therapeutic applications in neuro-disorders. This review discussed FIPs' structural and protein characteristics, their recombinant protein production for functional studies, and the recent advances in their development and applications as pharmaceutics and functional foods.
Collapse
Affiliation(s)
| | - Chong Joo Chan
- Faculty of Applied Sciences, Department of Biotechnology, UCSI University, Kuala Lumpur, Malaysia
| | | | - Renee Lay Hong Lim
- Faculty of Applied Sciences, Department of Biotechnology, UCSI University, Kuala Lumpur, Malaysia
| |
Collapse
|
7
|
Guo J, Liu Q, Li Z, Guo H, Bai C, Wang F. miR-222-3p promotes osteosarcoma cell migration and invasion through targeting TIMP3. Onco Targets Ther 2018; 11:8643-8653. [PMID: 30584323 PMCID: PMC6284535 DOI: 10.2147/ott.s175745] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Abnormal expression of miRNAs has been reported in osteosarcoma (OS), and miR-222-3p levels have been found to be increased in the serum of OS patients. However, the exact role of miR-222-3p in OS remains unclear. In the present study, we aimed to identify the molecular mechanism underlying the role of miR-222-3p in the development of OS. METHODS We examined the expression level of miR-222-3p in OS tissues and OS cells using reverse-transcription quantitative PCR (RT-qPCR) analysis. MTT, colony formation, and transwell invasion assays were used to analyze the effects of miR-222-3p on the proliferation and invasion ability of OS cells. Luciferase reporter gene assays were used to confirm the target gene of miR-222-3p in OS cells. Tumor xenografts were then used to investigate the role of miR-222-3p in OS growth in vivo. RESULTS The data of the present study demonstrated that miR-222-3p levels were increased in OS tissues and OS cells. Downregulation of miR-222-3p significantly inhibited the proliferation, migration, and invasion of OS cells in vitro. Further analysis revealed that tissue inhibitors of metalloproteinases 3 (TIMP3) is one of the functional target genes of miR-222-3p, and inhibition of TIMP3 efficiently rescues the blocking of cell proliferation and invasion mediated by miR-222-3p inhibitor in OS cells. CONCLUSION Our findings constitute evidence that miR-222-3p promotes OS cell proliferation and invasion through targeting TIMP3 mRNA and provide novel insight into the mechanism underlying the development of OS.
Collapse
Affiliation(s)
- Jianping Guo
- Department of Orthopaedic Surgery, Affiliated Hospital of Beihua University, Jilin 132011, PR China,
| | - Quanxiang Liu
- Department of Orthopaedic Surgery, Affiliated Hospital of Beihua University, Jilin 132011, PR China,
| | - Zengxin Li
- Department of Orthopaedic Surgery, Affiliated Hospital of Beihua University, Jilin 132011, PR China,
| | - Haifeng Guo
- Department of Orthopaedic Surgery, Affiliated Hospital of Beihua University, Jilin 132011, PR China,
| | - Changshuang Bai
- Department of Orthopaedic Surgery, Affiliated Hospital of Beihua University, Jilin 132011, PR China,
| | - Fajia Wang
- Department of Orthopaedic Surgery, Affiliated Hospital of Beihua University, Jilin 132011, PR China,
| |
Collapse
|
8
|
A Novel Disulfide-Containing Polycationic Amphiphile: 1,28-Di[(cholest-5-en-3β-yl)disulfanyl]-4,25-dioxo-3,8,12,17,21,26-hexaazaoctacosane Tetrahydrochloride. MOLBANK 2018. [DOI: 10.3390/m981] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
9
|
Vijayachari P, Vedhagiri K, Mallilankaraman K, Mathur PP, Sardesai NY, Weiner DB, Ugen KE, Muthumani K. Immunogenicity of a novel enhanced consensus DNA vaccine encoding the leptospiral protein LipL45. Hum Vaccin Immunother 2016; 11:1945-53. [PMID: 26020621 DOI: 10.1080/21645515.2015.1047117] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Leptospirosis is a bacterial zoonotic disease caused by an infection with a spirochete belonging to the genus Leptospira. In animals, leptospirosis displays a wide range of pathologies, including fever, abortion, icterus, and uveitis. Conversely, infection in humans is associated with multi-organ injury, resulting in an increased rate of fatalities. Pathogenic leptospires are able to translocate through cell monolayers at a rate significantly greater than that of non-pathogenic leptospires. Thus, vaccine approaches have been focused on targeting bacterial motility, lipopolysaccharides (LPSs), lipoproteins, outer-membrane proteins (OMPs) and other potential virulence factors. Previous studies have indicated that leptospiral proteins elicit long-lasting immunological memory in infected humans. In the study reported here, the efficacy of a synthetic consensus DNA vaccine developed against the Leptospira membrane lipoprotein LipL45 was tested. After in vivo electroporation (EP) mediated intramuscular immunization with a synthetic LipL45 DNA vaccine (pLipL45) immunized mice developed a significant cellular response along with the development of anti-LipL45-specific antibodies. Specifically, the pLipL45 vaccine induced a significant Th1 type immune response, indicated by the higher production of IL-12 and IFN-γ cytokines. The results presented here are the first demonstration that a LipL45 based DNA immunogen has potential as a anti-Leptospira vaccine.
Collapse
Affiliation(s)
- P Vijayachari
- a Regional Medical Research Center; Indian Council of Medical Research ; Port Blair , Andaman & Nicobar Islands , India
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Zhu F, Liu X, Sun Z, Yu C, Liu L, Yang S, Li B, Wei K, Zhu R. Immune-Enhancing Effects of Taishan Pinus massoniana Pollen Polysaccharides on DNA Vaccine Expressing Bordetella avium ompA. Front Microbiol 2016; 7:66. [PMID: 26870023 PMCID: PMC4735580 DOI: 10.3389/fmicb.2016.00066] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 01/14/2016] [Indexed: 11/17/2022] Open
Abstract
Bordetella avium is the causative agent of bordetellosis, which remains to be the cause of severe losses in the turkey industry. Given the lack of vaccines that can provide good protection, developing a novel vaccine against B. avium infection is crucial. In this study, we constructed a eukaryotic expression plasmid, which expressed the outer membrane protein A (ompA) of B. avium, to prepare a B. avium recombinant ompA-DNA vaccine. Three concentrations (low, middle, and high) of Taishan Pinus massoniana pollen polysaccharides (TPPPS), a known immunomodulator, were used as adjuvants, and their immune conditioning effects on the developed DNA vaccine were examined. The pure ompA-DNA vaccine, Freund’s incomplete adjuvant ompA-DNA vaccine, and the empty plasmid served as the controls. The chickens in each group were separately inoculated with these vaccines three times at 1, 7, and 14 days old. Dynamic changes in antibody production, cytokine secretion, and lymphocyte count were then determined from 7 to 49 days after the first inoculation. Protective rates of the vaccines were also determined after the third inoculation. Results showed that the pure DNA vaccine obviously induced the production of antibodies, the secretion of cytokines, and the increase in CD4+ and CD8+ T lymphocyte counts in peripheral blood, as well as provided a protective rate of 50% to the B. avium-challenged chickens. The chickens inoculated with the TPPPS adjuvant ompA-DNA vaccine and Freund’s adjuvant ompA-DNA vaccine demonstrated higher levels of immune responses than those inoculated with pure ompA-DNA vaccine, whereas only the ompA-DNA vaccine with 200 mg/mL TPPPS completely protected the chickens against B. avium infection. These findings indicate that the B. avium ompA-DNA vaccine combined with TPPPS is a potentially effective B. avium vaccine.
Collapse
Affiliation(s)
- Fujie Zhu
- Laboratory of Animal Biological Products, College of Animal Science and Technology, Shandong Agricultural University Taian, China
| | - Xiao Liu
- Laboratory of Animal Biological Products, College of Animal Science and Technology, Shandong Agricultural University Taian, China
| | - Zhenhong Sun
- Analytic Laboratory, Institute of Preclinical Medicine, Taishan Medical College Taian, China
| | - Cuilian Yu
- Laboratory of Animal Biological Products, College of Animal Science and Technology, Shandong Agricultural University Taian, China
| | - Liping Liu
- Laboratory of Animal Biological Products, College of Animal Science and Technology, Shandong Agricultural University Taian, China
| | - Shifa Yang
- Laboratory of Animal Biological Products, College of Animal Science and Technology, Shandong Agricultural University Taian, China
| | - Bing Li
- Laboratory of Animal Biological Products, College of Animal Science and Technology, Shandong Agricultural University Taian, China
| | - Kai Wei
- Laboratory of Animal Biological Products, College of Animal Science and Technology, Shandong Agricultural University Taian, China
| | - Ruiliang Zhu
- Laboratory of Animal Biological Products, College of Animal Science and Technology, Shandong Agricultural University Taian, China
| |
Collapse
|
11
|
Tapping the Potential of DNA Delivery with Electroporation for Cancer Immunotherapy. Curr Top Microbiol Immunol 2015; 405:55-78. [PMID: 25682101 DOI: 10.1007/82_2015_431] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cancer is a worldwide leading cause of death, and current conventional therapies are limited. The search for alternative preventive or therapeutic solutions is critical if we are going to improve outcomes for patients. The potential for DNA vaccines in the treatment and prevention of cancer has gained great momentum since initial findings almost 2 decades ago that revealed that genetically engineered DNA can elicit an immune response. The combination of adjuvants and an effective delivery method such as electroporation is overcoming past setbacks for naked plasmid DNA (pDNA) as a potential preventive or therapeutic approach to cancer in large animals and humans. In this chapter, we aim to focus on the novel advances in recent years for DNA cancer vaccines, current preclinical data, and the importance of adjuvants and electroporation with emphasis on prostate, melanoma, and cervical cancer.
Collapse
|
12
|
Cho H, Lee HJ, Heo YK, Cho Y, Gwon YD, Kim MG, Park KH, Oh YK, Kim YB. Immunogenicity of a trivalent human papillomavirus L1 DNA-encapsidated, non-replicable baculovirus nanovaccine. PLoS One 2014; 9:e95961. [PMID: 24759938 PMCID: PMC3997520 DOI: 10.1371/journal.pone.0095961] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Accepted: 03/31/2014] [Indexed: 12/04/2022] Open
Abstract
Previously, we developed a non-replicating recombinant baculovirus coated with human endogenous retrovirus envelope protein (AcHERV) for enhanced cellular delivery of human papillomavirus (HPV) 16L1 DNA. Here, we report the immunogenicity of an AcHERV-based multivalent HPV nanovaccine in which the L1 segments of HPV 16, 18, and 58 genes were inserted into a single baculovirus genome of AcHERV. To test whether gene expression levels were affected by the order of HPV L1 gene insertion, we compared the efficacy of bivalent AcHERV vaccines with the HPV 16L1 gene inserted ahead of the 18L1 gene (AcHERV-HP16/18L1) with that of AcHERV with the HPV 18L1 gene inserted ahead of the 16L1 gene (AcHERV-HP18/16L1). Regardless of the order, the bivalent AcHERV DNA vaccines retained the immunogenicity of monovalent AcHERV-HP16L1 and AcHERV-HP18L1 DNA vaccines. Moreover, the immunogenicity of bivalent AcHERV-HP16/18L1 was not significantly different from that of AcHERV-HP18/16L1. In challenge tests, both bivalent vaccines provided complete protection against HPV 16 and 18 pseudotype viruses. Extending these results, we found that a trivalent AcHERV nanovaccine encoding HPV 16L1, 18L1, and 58L1 genes (AcHERV-HP16/18/58L1) provided high levels of humoral and cellular immunogenicity against all three subtypes. Moreover, mice immunized with the trivalent AcHERV-based nanovaccine were protected from challenge with HPV 16, 18, and 58 pseudotype viruses. These results suggest that trivalent AcHERV-HPV16/18/58L1 could serve as a potential prophylactic baculoviral nanovaccine against concurrent infection with HPV 16, 18, and 58.
Collapse
Affiliation(s)
- Hansam Cho
- Department of Bio-industrial Technologies, Konkuk University, Seoul, Republic of Korea
| | - Hee-Jung Lee
- Department of Bio-industrial Technologies, Konkuk University, Seoul, Republic of Korea
| | - Yoon-Ki Heo
- Department of Bio-industrial Technologies, Konkuk University, Seoul, Republic of Korea
| | - Yeondong Cho
- Department of Bio-industrial Technologies, Konkuk University, Seoul, Republic of Korea
| | - Yong-Dae Gwon
- Department of Bio-industrial Technologies, Konkuk University, Seoul, Republic of Korea
| | - Mi-Gyeong Kim
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Ki Hoon Park
- Department of Bio-industrial Technologies, Konkuk University, Seoul, Republic of Korea
| | - Yu-Kyoung Oh
- College of Pharmacy, Seoul National University, Seoul, Republic of Korea
- * E-mail: (YKO) or (YBK)
| | - Young Bong Kim
- Department of Bio-industrial Technologies, Konkuk University, Seoul, Republic of Korea
- * E-mail: (YKO) or (YBK)
| |
Collapse
|
13
|
Abstract
Vaccines to prevent HIV remain desperately needed, but a number of challenges, including retroviral integration, establishment of anatomic reservoir sites, high sequence diversity, and heavy envelope glycosylation. have precluded development of a highly effective vaccine. DNA vaccines have been utilized as candidate HIV vaccines because of their ability to generate cellular and humoral immune responses, the lack of anti-vector response allowing for repeat administration, and their ability to prime the response to viral-vectored vaccines. Because the HIV epidemic has disproportionately affected the developing world, the favorable thermostability profile and relative ease and low cost of manufacture of DNA vaccines offer additional advantages. In vivo electroporation (EP) has been utilized to improve immune responses to DNA vaccines as candidate HIV-1 vaccines in standalone or prime-boost regimens with both proteins and viral-vectored vaccines in several animal models and, more recently, in human clinical trials. This chapter describes the preclinical and clinical development of candidate DNA vaccines for HIV-1 delivered by EP, including challenges to bringing this technology to the developing world.
Collapse
Affiliation(s)
- Sandhya Vasan
- Department of Retrovirology, US Army Medical Component, Armed Forces Research Institute of Medical Sciences (AFRIMS), Bangkok, Thailand
| |
Collapse
|
14
|
Chen MH, Li WS, Lue YS, Chu CL, Pan IH, Ko CH, Chen DY, Lin CH, Lin SH, Chang CP, Lin CC. Clitocybe nuda Activates Dendritic Cells and Acts as a DNA Vaccine Adjuvant. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2013; 2013:761454. [PMID: 24058377 PMCID: PMC3766593 DOI: 10.1155/2013/761454] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 05/25/2013] [Accepted: 06/01/2013] [Indexed: 12/11/2022]
Abstract
This work represents the first evaluation of the effects of water extract of C. nuda (WE-CN), an edible mushroom, on murine bone marrow-derived dendritic cells (BMDCs) and the potential pathway through which the effects are mediated. Our experimental results show that WE-CN could induce phenotypic maturation of DCs, as shown by the increased expression of MHC and costimulatory molecules. In addition, it also induced the proinflammatory cytokines expression on DCs and enhanced both the proliferation and IFN- γ secretion of allogenic T cells. Therefore, since WE-CN did not induce maturation of DCs generated from mice with mutated TLR-4 or TLR-2, suggesting that TLR4 and TLR2 might function as membrane receptors for WE-CN. Moreover, the mechanism of action of WE-CN may be mediated by increased phosphorylation of ERK, p38, and JNK mitogen-activated protein kinase (MAPK) and increased NF- κ B p65 activity, which are important signaling molecules downstream of TLR-4 and TLR-2. Finally, coimmunization of mice with WE-CN and a HER-2/neu DNA vaccine induced a HER-2/neu-specific Th1 response that resulted in significant inhibition of HER-2/neu overexpressing mouse bladder tumor (MBT-2) growth. These data suggest that WE-CN induces DC maturation through TLR-4 and/or TLR-2 and that WE-CN can be used as an adjuvant in cancer vaccine immunotherapy.
Collapse
Affiliation(s)
- Mei-Hsing Chen
- Plant Pathology Division, Taiwan Agricultural Research Institute (TARI), Council of Agriculture (COA), Executive Yuan, Wufeng 413, Taiwan
| | - Wei-Sung Li
- Plant Pathology Division, Taiwan Agricultural Research Institute (TARI), Council of Agriculture (COA), Executive Yuan, Wufeng 413, Taiwan
| | - Yun-Sheng Lue
- Plant Pathology Division, Taiwan Agricultural Research Institute (TARI), Council of Agriculture (COA), Executive Yuan, Wufeng 413, Taiwan
| | - Ching-Liang Chu
- Graduate Institute of Immunology, National Taiwan University, Taipei 112, Taiwan
| | - I-Hong Pan
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu 300, Taiwan
| | - Ching-Huai Ko
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu 300, Taiwan
| | - Der-Yuan Chen
- Institute of Biomedical Science, National Chung-Hsing University, Taichung 402, Taiwan
- Faculty of Medicine, National Yang-Ming University, Taipei 112, Taiwan
- Division of Allergy, Immunology and Rheumatology, Taichung Veterans General Hospital, Taichung 407, Taiwan
| | - Ching-Hsiung Lin
- Division of Chest Medicine, Department of Internal Medicine, Changhua Christian Hospital, Changhua 500, Taiwan
| | - Sheng-Hao Lin
- Institute of Biomedical Science, National Chung-Hsing University, Taichung 402, Taiwan
- Division of Chest Medicine, Department of Internal Medicine, Changhua Christian Hospital, Changhua 500, Taiwan
| | - Chih-Peng Chang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Chi-Chen Lin
- Institute of Biomedical Science, National Chung-Hsing University, Taichung 402, Taiwan
- Department of Medical Research and Education, Taichung Veterans General Hospital, Taichung 407, Taiwan
| |
Collapse
|
15
|
Shiang YC, Ou CM, Chen SJ, Ou TY, Lin HJ, Huang CC, Chang HT. Highly efficient inhibition of human immunodeficiency virus type 1 reverse transcriptase by aptamers functionalized gold nanoparticles. NANOSCALE 2013; 5:2756-2764. [PMID: 23429884 DOI: 10.1039/c3nr33403a] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
We have developed aptamer (Apt)-conjugated gold nanoparticles (Apt-Au NPs, 13 nm in diameter) as highly effective inhibitors for human immunodeficiency virus type 1 reverse transcriptase (HIV-1 RT). Two Apts, RT1t49 (Aptpol) and ODN 93 (AptRH), which recognize the polymerase and RNase H regions of HIV-1 RT, are used to conjugate Au NPs to prepare Aptpol-Au NPs and AptRH-Au NPs, respectively. In addition to DNA sequence, the surface density of the aptamers on Au NPs (nApt-Au NPs; n is the number of aptamer molecules on each Au NP) and the linker length number (Tm; m is the base number of the deoxythymidine linker) between the aptamer and Au NPs play important roles in determining their inhibition activity. A HIV-lentiviral vector-based antiviral assay has been applied to determine the inhibitory effect of aptamers or Apt-Au NPs on the early stages of their replication cycle. The nuclease-stable G-quadruplex structure of 40AptRH-T45-Au NPs shows inhibitory efficiency in the retroviral replication cycle with a decreasing infectivity (40.2%).
Collapse
Affiliation(s)
- Yen-Chun Shiang
- Department of Chemistry, National Taiwan University, 1, Section 4, Roosevelt Road, Taipei 10617, Taiwan
| | | | | | | | | | | | | |
Collapse
|
16
|
Lee HJ, Hur YK, Cho YD, Kim MG, Lee HT, Oh YK, Kim YB. Immunogenicity of bivalent human papillomavirus DNA vaccine using human endogenous retrovirus envelope-coated baculoviral vectors in mice and pigs. PLoS One 2012; 7:e50296. [PMID: 23209698 PMCID: PMC3507738 DOI: 10.1371/journal.pone.0050296] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 10/18/2012] [Indexed: 11/19/2022] Open
Abstract
Human papillomavirus is known to be the major pathogen of cervical cancer. Here, we report the efficacy of a bivalent human papillomavirus type 16 and 18 DNA vaccine system following repeated dosing in mice and pigs using a recombinant baculovirus bearing human endogenous retrovirus envelope protein (AcHERV) as a vector. The intramuscular administration of AcHERV-based HPV16L1 and HPV18L1 DNA vaccines induced antigen-specific serum IgG, vaginal IgA, and neutralizing antibodies to levels comparable to those achieved using the commercially marketed vaccine Cervarix. Similar to Cervarix, AcHERV-based bivalent vaccinations completely blocked subsequent vaginal challenge with HPV type-specific pseudovirions. However, AcHERV-based bivalent vaccinations induced significantly higher cell-mediated immune responses than Cervarix, promoting 4.5- (HPV16L1) and 3.9-(HPV18L1) fold higher interferon-γ production in splenocytes upon stimulation with antigen type-specific pseudovirions. Repeated dosing did not affect the immunogenicity of AcHERV DNA vaccines. Three sequential immunizations with AcHERV-HP18L1 DNA vaccine followed by three repeated dosing with AcHERV-HP16L1 over 11 weeks induced an initial production of anti-HPV18L1 antibody followed by subsequent induction of anti-HPV16L1 antibody. Finally, AcHERV-based bivalent DNA vaccination induced antigen-specific serum IgG immune responses in pigs. These results support the further development of AcHERV as a bivalent human papillomavirus DNA vaccine system for use in preventing the viral infection as well as treating the infected women by inducing both humoral and cell-mediated immune responses. Moreover, the possibility of repeated dosing indicates the utility of AcHERV system for reusable vectors of other viral pathogen vaccines.
Collapse
Affiliation(s)
- Hee-Jung Lee
- College of Animal Bioscience & Technology, Konkuk University, Seoul, South Korea
| | - Yoon-Ki Hur
- College of Animal Bioscience & Technology, Konkuk University, Seoul, South Korea
| | - Youn-Dong Cho
- College of Animal Bioscience & Technology, Konkuk University, Seoul, South Korea
| | - Mi-Gyeong Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Hoon-Taek Lee
- College of Animal Bioscience & Technology, Konkuk University, Seoul, South Korea
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
- * E-mail: (Y-KO); (YBK)
| | - Young Bong Kim
- College of Animal Bioscience & Technology, Konkuk University, Seoul, South Korea
- * E-mail: (Y-KO); (YBK)
| |
Collapse
|
17
|
Barar J, Omidi Y. Translational Approaches towards Cancer Gene Therapy: Hurdles and Hopes. BIOIMPACTS : BI 2012; 2:127-43. [PMID: 23678451 DOI: 10.5681/bi.2012.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Revised: 09/02/2012] [Accepted: 09/11/2012] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Of the cancer gene therapy approaches, gene silencing, suicide/apoptosis inducing gene therapy, immunogene therapy and targeted gene therapy are deemed to sub-stantially control the biological consequences of genomic changes in cancerous cells. Thus, a large number of clinical trials have been conducted against various malignancies. In this review, we will discuss recent translational progresses of gene and cell therapy of cancer. METHODS Essential information on gene therapy of cancer were reviewed and discussed towards their clinical translations. RESULTS Gene transfer has been rigorously studied in vitro and in vivo, in which some of these gene therapy endeavours have been carried on towards translational investigations and clinical applications. About 65% of gene therapy trials are related to cancer therapy. Some of these trials have been combined with cell therapy to produce personalized medicines such as Sipuleucel-T (Provenge®, marketed by Dendreon, USA) for the treatment of asymptomatic/minimally symptomatic metastatic hormone-refractory prostate cancer. CONCLUSION Translational approach links two diverse boundaries of basic and clinical researches. For successful translation of geno-medicines into clinical applications, it is essential 1) to have the guidelines and standard operating procedures for development and application of the genomedicines specific to clinically relevant biomarker(s); 2) to conduct necessary animal experimental studies to show the "proof of concept" for the proposed genomedicines; 3) to perform an initial clinical investigation; and 4) to initiate extensive clinical trials to address all necessary requirements. In short, translational researches need to be refined to accelerate the geno-medicine development and clinical applications.
Collapse
Affiliation(s)
- Jaleh Barar
- Ovarian Cancer Research Center, Translational Research Center, University of Pennsylvania, Philadelphia, PA, USA
| | | |
Collapse
|
18
|
Abstract
Recently, there have been several advancements in material sciences and nanosciences. At the moment these new techniques are slowly entering into clinical settings in drug delivery and imaging. In this review, we will look more closely at the applications that are at the forefront of this translation and examine critical aspects that are involved in the process. Nanoparticles have been increasingly used in clinical settings for drug delivery over the past two decades. Lipid-based nanoparticles are front-runners, but other innovative strategies, such as small inorganic nanoparticles, are entering into the field, particularly for imaging applications. Lipid-based nanoparticles can be metabolized and consumed by the body and are regarded as safe for clinical use. They are usually large with hydrodynamic diameters of approximately 100-200 nm; however, phospholipid-containing particles such as microbubbles with diameters as low as 10 microm in size and micelles with diameters of 10-40 nm can also be used. Hollow liposomes with a large aqueous inner cavity can carry high payloads of drugs and imaging moieties, but are easily trapped by liver kupffer cells and can result in lower tissue penetration rates. New classes of particles with hydrodynamic diameters of < 10 nm, which are cleared by the kidneys, have recently been developed. These particles have been used primarily for imaging applications since they offer only small loading capacities for drugs. However, new strategies such as surface-coupled prodrugs have been developed to facilitate drug delivery in small nanoparticles. We describe different strategies for targeted delivery, imaging and controlled release, and discuss the ability of small inorganic particles as well as larger nanoparticles to be used broadly in human diagnostics and drug delivery.
Collapse
|
19
|
Lin F, Shen X, Kichaev G, Mendoza JM, Yang M, Armendi P, Yan J, Kobinger GP, Bello A, Khan AS, Broderick KE, Sardesai NY. Optimization of electroporation-enhanced intradermal delivery of DNA vaccine using a minimally invasive surface device. Hum Gene Ther Methods 2012; 23:157-68. [PMID: 22794496 DOI: 10.1089/hgtb.2011.209] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
In vivo electroporation (EP) is an efficient nonviral method for enhancing DNA vaccine delivery and immunogenicity in animals and humans. Intradermal delivery of DNA vaccines is an attractive strategy because of the immunocompetence of skin tissue. We have previously reported a minimally invasive surface intradermal EP (SEP) device for delivery of prophylactic DNA vaccines. Robust antibody responses were induced after vaccine delivery via surface EP in several tested animal models. Here we further investigated the optimal EP parameters for efficient delivery of DNA vaccines, with a specific emphasis on eliciting cellular immunity in addition to robust humoral responses. In a mouse model, using applied voltages of 10-100 V, transgene expression of green fluorescent protein and luciferase reporter genes increased significantly when voltages as low as 10 V were used as compared with DNA injection only. Tissue damage to skin was undetectable when voltages of 20 V and less were applied. However, inflammation and bruising became apparent at voltages above 40 V. Delivery of DNA vaccines encoding influenza virus H5 hemagglutinin (H5HA) and nucleoprotein (NP) of influenza H1N1 at applied voltages of 10-100 V elicited robust and sustained antibody responses. In addition, low-voltage (less than 20 V) EP elicited higher and more sustained cellular immune responses when compared with the higher voltage (above 20 V) EP groups after two immunizations. The data confirm that low-voltage EP, using the SEP device, is capable of efficient delivery of DNA vaccines into the skin, and establishes that these parameters are sufficient to elicit both robust and sustainable humoral as well as cellular immune responses without tissue damage. The SEP device, functioning within these parameters, may have important clinical applications for delivery of prophylactic DNA vaccines against diseases such as HIV infection, malaria, and tuberculosis that require both cellular and humoral immune responses for protection.
Collapse
Affiliation(s)
- Feng Lin
- Inovio Pharmaceuticals, Blue Bell, PA 19422, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Vaccines have shown promise for the prevention and treatment of solid tumors. Colorectal cancer and renal cell carcinoma are common malignancies that may be amenable to vaccine strategies. This review summarizes target antigens in colorectal and renal cell carcinoma, discusses some of the vaccine approaches in development, and details the results of pivotal phase III trials evaluating therapeutic vaccines in patients with advanced colorectal and renal cell carcinoma. Finally, some of the challenges with vaccine development for colorectal and renal cell carcinoma are described.
Collapse
Affiliation(s)
- Katherine Kabaker
- Division of Hematology & Oncology and Rush University Cancer Center, Rush University Medical Center, Chicago, IL, USA
| | | | | |
Collapse
|
21
|
An artificial PAP gene breaks self-tolerance and promotes tumor regression in the TRAMP model for prostate carcinoma. Mol Ther 2011; 20:555-64. [PMID: 22086231 DOI: 10.1038/mt.2011.241] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is the most commonly diagnosed type of cancer in men in western industrialized countries. As a public health burden, the need for the invention of new cost-saving PCa immunotherapies is apparent. In this study, we present a DNA vaccine encoding for the prostate-specific antigen prostatic acid phosphatase (PAP) linked to the J-domain and the SV40 enhancer sequence. The PAP DNA vaccine induced a strong PAP-specific cellular immune response after electroporation (EP)-based delivery in C57BL/6 mice. Splenocytes from mice immunized with PAP recognized the naturally processed PAP epitopes, indicating that vaccination with the PAP-J gene broke its self-tolerance against PAP. Remarkably, DNA vaccination with PAP-J inhibited tumor growth in the Transgenic Adenocarcinoma of the Mouse Prostate (TRAMP) mouse model that closely resembled human PCa. Therefore, this study highlights a novel cancer immunotherapy approach with the potential to control PCa in clinical settings.
Collapse
|
22
|
Harris F, Dennison SR, Singh J, Phoenix DA. On the selectivity and efficacy of defense peptides with respect to cancer cells. Med Res Rev 2011; 33:190-234. [PMID: 21922503 DOI: 10.1002/med.20252] [Citation(s) in RCA: 134] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Here, we review potential determinants of the anticancer efficacy of innate immune peptides (ACPs) for cancer cells. These determinants include membrane-based factors, such as receptors, phosphatidylserine, sialic acid residues, and sulfated glycans, and peptide-based factors, such as residue composition, sequence length, net charge, hydrophobic arc size, hydrophobicity, and amphiphilicity. Each of these factors may contribute to the anticancer action of ACPs, but no single factor(s) makes an overriding contribution to their overall selectivity and toxicity. Differences between the anticancer actions of ACPs seem to relate to different levels of interplay between these peptide and membrane-based factors.
Collapse
Affiliation(s)
- Frederick Harris
- School of Forensic and Investigative Sciences, University of Central Lancashire, Preston, Lancashire, United Kingdom
| | | | | | | |
Collapse
|
23
|
Yin J, Dai A, LeCureux J, Arango T, Kutzler MA, Yan J, Lewis MG, Khan A, Sardesai NY, Montefiore D, Ruprecht R, Weiner DB, Boyer JD. High antibody and cellular responses induced to HIV-1 clade C envelope following DNA vaccines delivered by electroporation. Vaccine 2011; 29:6763-70. [PMID: 21195801 PMCID: PMC10839813 DOI: 10.1016/j.vaccine.2010.12.055] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Clade C is the predominant HIV-1 strain infecting people in sub-Saharan Africa, India, and China and there is a critical need for a vaccine targeted to these areas. In this study we tested a DNA based vaccine that encodes the SIVgag, SIVpol and HIV-1 envelope clade C. METHODS Rhesus macaques were immunized by electroporation with the DNA plasmid encoding optimized SIVgag, SIVpol and an HIV-1 env clade C with or without the adjuvant RANTES. Animals were monitored for immune responses and challenged following the final immunization with 25 animal infectious doses (AID) of SHIV-1157ipd3N4. RESULTS We found that the vaccine induced high levels of antigen specific IFN-γ producing effector cells and the capacity for CD4+ and CD8+ to proliferate upon antigen stimulation. Importantly, we found that the vaccine induced antibody titers as high as 1/4000. These antibodies were capable of neutralizing tier 1 HIV-1 viruses. Finally, when macaques were challenged with SHIV, viral loads were controlled in vaccinated groups. CONCLUSION We conclude that immunization with a simian/human immunodeficiency virus DNA-based vaccine delivered by electroporation can induce cellular and humoral immune responses that are able to control viral replication.
Collapse
Affiliation(s)
- Jiangmei Yin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 422 Curie Blvd, Philadelphia PA, 19104
| | - Anlan Dai
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 422 Curie Blvd, Philadelphia PA, 19104
| | - Jonathan LeCureux
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 422 Curie Blvd, Philadelphia PA, 19104
| | - Tatiana Arango
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 422 Curie Blvd, Philadelphia PA, 19104
| | - Michele A. Kutzler
- Drexel University, College of Medicine, 245N 15 Street, Philadelphia PA, 19102
| | - Jian Yan
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 422 Curie Blvd, Philadelphia PA, 19104
| | | | - Amir Khan
- Inovio Pharmaceuticals, 1787 Sentry Parkway West, Blue Bell, PA 19422
| | | | | | - Ruth Ruprecht
- Dana-Farber Cancer Institute and Harvard Medical School, Boston MA, 02115
| | - David B. Weiner
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 422 Curie Blvd, Philadelphia PA, 19104
| | - Jean D. Boyer
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 422 Curie Blvd, Philadelphia PA, 19104
| |
Collapse
|
24
|
Cemazar M, Golzio M, Sersa G, Escoffre JM, Coer A, Vidic S, Teissie J. Hyaluronidase and collagenase increase the transfection efficiency of gene electrotransfer in various murine tumors. Hum Gene Ther 2011; 23:128-37. [PMID: 21797718 DOI: 10.1089/hum.2011.073] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
One of the applications of electroporation/electropulsation in biomedicine is gene electrotransfer, the wider use of which is hindered by low transfection efficiency in vivo compared with viral vectors. The aim of our study was to determine whether modulation of the extracellular matrix in solid tumors, using collagenase and hyaluronidase, could increase the transfection efficiency of gene electrotransfer in histologically different solid subcutaneous tumors in mice. Tumors were treated with enzymes before electrotransfer of plasmid DNA encoding either green fluorescent protein or luciferase. Transfection efficiency was determined 3, 9, and 15 days posttransfection. We demonstrated that pretreatment of tumors with a combination of enzymes significantly increased the transfection efficiency of electrotransfer in tumors with a high extracellular matrix area (LPB fibrosarcoma). In tumors with a smaller extracellular matrix area and less organized collagen lattice, the increase was not so pronounced (SA-1 fibrosarcoma and EAT carcinoma), whereas in B16 melanoma, in which only traces of collagen are present, pretreatment of tumors with hyaluronidase alone was more efficient than pretreatment with both enzymes. In conclusion, our results suggest that modification of the extracellular matrix could improve distribution of plasmid DNA in solid subcutaneous tumors, demonstrated by an increase in transfection efficiency, and thus have important clinical implications for electrogene therapy.
Collapse
Affiliation(s)
- Maja Cemazar
- Department of Experimental Oncology, Institute of Oncology Ljubljana , SI-1000 Ljubljana, Slovenia.
| | | | | | | | | | | | | |
Collapse
|
25
|
Lin CC, Yu YL, Shih CC, Liu KJ, Ou KL, Hong LZ, Chen JDC, Chu CL. A novel adjuvant Ling Zhi-8 enhances the efficacy of DNA cancer vaccine by activating dendritic cells. Cancer Immunol Immunother 2011; 60:1019-27. [PMID: 21499904 PMCID: PMC11029078 DOI: 10.1007/s00262-011-1016-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2011] [Accepted: 04/04/2011] [Indexed: 02/01/2023]
Abstract
DNA vaccine has been suggested to use in cancer therapy, but the efficacy remains to be improved. The immunostimulatory effect of a fungal immunomodulatory protein Ling Zhi-8 (LZ-8) isolated from Ganoderma lucidum has been reported. In this study, we tested the adjuvanticity of LZ-8 for HER-2/neu DNA vaccine against p185(neu) expressing tumor MBT-2 in mice. We found that recombinant LZ-8 stimulated mouse bone marrow-derived dendritic cells (DCs) via TLR4 and its stimulatory effect was not due to any microbe contaminant. In addition, LZ-8 enhanced the ability of DCs to induce antigen-specific T cell activation in vitro and in a subunit vaccine model in vivo. Surprisingly, LZ-8 cotreatment strongly improved the therapeutic effect of DNA vaccine against MBT-2 tumor in mice. This increase in antitumor activity was attributed to the enhancement of vaccine-induced Th1 and CTL responses. Consistent with the results from DCs, the promoting effect of LZ-8 on DNA vaccine was diminished when the MBT-2 tumor cells were grown in TLR4 mutant mice. Thus, we concluded that LZ-8 may be a promising adjuvant to enhance the efficacy of DNA vaccine by activating DCs via TLR4.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Animals
- Blotting, Western
- Cancer Vaccines/therapeutic use
- Dendritic Cells/immunology
- Disease Models, Animal
- Fungal Proteins/genetics
- Fungal Proteins/immunology
- Lymphocyte Activation
- Mice
- Mice, Inbred C3H
- Mice, Inbred C57BL
- RNA, Messenger/genetics
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/immunology
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes, Cytotoxic/immunology
- Transduction, Genetic
- Urinary Bladder Neoplasms/genetics
- Urinary Bladder Neoplasms/immunology
- Urinary Bladder Neoplasms/therapy
- Vaccines, DNA/therapeutic use
Collapse
Affiliation(s)
- Chi-Chen Lin
- Institute of Biomedical Sciences, National Chung Hsin University, Taichung, Taiwan.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Enhancing DNA immunization by targeting ASFV antigens to SLA-II bearing cells. Vaccine 2011; 29:5379-85. [PMID: 21679736 DOI: 10.1016/j.vaccine.2011.05.084] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 05/17/2011] [Accepted: 05/23/2011] [Indexed: 01/08/2023]
Abstract
One of the main criticisms to DNA vaccines is the poor immunogenicity that they confer on occasions, at least in large animals. Confirming this theory, immunization with plasmid DNA encoding two African swine fever virus genes in frame (pCMV-PQ), failed in inducing detectable immune responses in pigs, while it was successful in mice. Aiming to improve the immune responses induced in swine, a new plasmid was constructed, encoding the viral genes fused in frame with a single chain variable fragment of an antibody specific for a swine leukocyte antigen II (pCMV-APCH1PQ). Our results clearly demonstrate that targeting antigens to antigen professional cells exponentially enhanced the immune response induced in pigs, albeit that the DNA vaccine was not able to confer protection against lethal viral challenge. Indeed, a viremia exacerbation was observed in each of the pigs that received the pCMV-APCH1PQ plasmid, this correlating with the presence of non-neutralizing antibodies and antigen-specific SLA II-restricted T-cells. The implications of our discoveries for the development of future vaccines against African swine fever virus and other swine pathogens are discussed.
Collapse
|
27
|
Abstract
Pancreatic adenocarcinoma is the fourth leading cause of cancer death with an overall 5-year survival of less than 5%. As there is ample evidence that pancreatic adenocarcinomas elicit antitumor immune responses, identification of pancreatic cancer-associated antigens has spurred the development of vaccination-based strategies for treatment. While promising results have been observed in animal tumor models, most clinical studies have found only limited success. As most trials were performed in patients with advanced pancreatic cancer, the contribution of immune suppressor mechanisms should be taken into account. In this article, we detail recent work in tumor antigen vaccination and the recently identified mechanisms of immune suppression in pancreatic cancer. We offer our perspective on how to increase the clinical efficacy of vaccines for pancreatic cancer.
Collapse
Affiliation(s)
- Lindzy F Dodson
- Washington University School of Medicine, Department of Surgery, Saint Louis, MO 63110, USA.
| | | | | |
Collapse
|
28
|
Bolhassani A, Safaiyan S, Rafati S. Improvement of different vaccine delivery systems for cancer therapy. Mol Cancer 2011; 10:3. [PMID: 21211062 PMCID: PMC3024302 DOI: 10.1186/1476-4598-10-3] [Citation(s) in RCA: 163] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 01/07/2011] [Indexed: 12/18/2022] Open
Abstract
Cancer vaccines are the promising tools in the hands of the clinical oncologist. Many tumor-associated antigens are excellent targets for immune therapy and vaccine design. Optimally designed cancer vaccines should combine the best tumor antigens with the most effective immunotherapy agents and/or delivery strategies to achieve positive clinical results. Various vaccine delivery systems such as different routes of immunization and physical/chemical delivery methods have been used in cancer therapy with the goal to induce immunity against tumor-associated antigens. Two basic delivery approaches including physical delivery to achieve higher levels of antigen production and formulation with microparticles to target antigen-presenting cells (APCs) have demonstrated to be effective in animal models. New developments in vaccine delivery systems will improve the efficiency of clinical trials in the near future. Among them, nanoparticles (NPs) such as dendrimers, polymeric NPs, metallic NPs, magnetic NPs and quantum dots have emerged as effective vaccine adjuvants for infectious diseases and cancer therapy. Furthermore, cell-penetrating peptides (CPP) have been known as attractive carrier having applications in drug delivery, gene transfer and DNA vaccination. This review will focus on the utilization of different vaccine delivery systems for prevention or treatment of cancer. We will discuss their clinical applications and the future prospects for cancer vaccine development.
Collapse
Affiliation(s)
- Azam Bolhassani
- Molecular Immunology and Vaccine Research Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Shima Safaiyan
- Molecular Immunology and Vaccine Research Laboratory, Pasteur Institute of Iran, Tehran, Iran
| | - Sima Rafati
- Molecular Immunology and Vaccine Research Laboratory, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
29
|
Dolter KE, Evans CF, Ellefsen B, Song J, Boente-Carrera M, Vittorino R, Rosenberg TJ, Hannaman D, Vasan S. Immunogenicity, safety, biodistribution and persistence of ADVAX, a prophylactic DNA vaccine for HIV-1, delivered by in vivo electroporation. Vaccine 2011; 29:795-803. [DOI: 10.1016/j.vaccine.2010.11.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 10/20/2010] [Accepted: 11/03/2010] [Indexed: 12/11/2022]
|
30
|
Zupanic A, Corovic S, Miklavcic D, Pavlin M. Numerical optimization of gene electrotransfer into muscle tissue. Biomed Eng Online 2010; 9:66. [PMID: 21050435 PMCID: PMC2990758 DOI: 10.1186/1475-925x-9-66] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 11/04/2010] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Electroporation-based gene therapy and DNA vaccination are promising medical applications that depend on transfer of pDNA into target tissues with use of electric pulses. Gene electrotransfer efficiency depends on electrode configuration and electric pulse parameters, which determine the electric field distribution. Numerical modeling represents a fast and convenient method for optimization of gene electrotransfer parameters. We used numerical modeling, parameterization and numerical optimization to determine the optimum parameters for gene electrotransfer in muscle tissue. METHODS We built a 3D geometry of muscle tissue with two or six needle electrodes (two rows of three needle electrodes) inserted. We performed a parametric study and optimization based on a genetic algorithm to analyze the effects of distances between the electrodes, depth of insertion, orientation of electrodes with respect to muscle fibers and applied voltage on the electric field distribution. The quality of solutions were evaluated in terms of volumes of reversibly (desired) and irreversibly (undesired) electroporated muscle tissue and total electric current through the tissue. RESULTS Large volumes of reversibly electroporated muscle with relatively little damage can be achieved by using large distances between electrodes and large electrode insertion depths. Orienting the electrodes perpendicular to muscle fibers is significantly better than the parallel orientation for six needle electrodes, while for two electrodes the effect of orientation is not so pronounced. For each set of geometrical parameters, the window of optimal voltages is quite narrow, with lower voltages resulting in low volumes of reversibly electroporated tissue and higher voltages in high volumes of irreversibly electroporated tissue. Furthermore, we determined which applied voltages are needed to achieve the optimal field distribution for different distances between electrodes. CONCLUSION The presented numerical study of gene electrotransfer is the first that demonstrates optimization of parameters for gene electrotransfer on tissue level. Our method of modeling and optimization is generic and can be applied to different electrode configurations, pulsing protocols and different tissues. Such numerical models, together with knowledge of tissue properties can provide useful guidelines for researchers and physicians in selecting optimal parameters for in vivo gene electrotransfer, thus reducing the number of animals used in studies of gene therapy and DNA vaccination.
Collapse
Affiliation(s)
- Anze Zupanic
- University of Ljubljana, Faculty of Electrical Engineering, Trzaska cesta 25, SI-1000 Ljubljana, Slovenia
| | - Selma Corovic
- University of Ljubljana, Faculty of Electrical Engineering, Trzaska cesta 25, SI-1000 Ljubljana, Slovenia
| | - Damijan Miklavcic
- University of Ljubljana, Faculty of Electrical Engineering, Trzaska cesta 25, SI-1000 Ljubljana, Slovenia
| | - Mojca Pavlin
- University of Ljubljana, Faculty of Electrical Engineering, Trzaska cesta 25, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
31
|
Signori E, Iurescia S, Massi E, Fioretti D, Chiarella P, De Robertis M, Rinaldi M, Tonon G, Fazio VM. DNA vaccination strategies for anti-tumour effective gene therapy protocols. Cancer Immunol Immunother 2010; 59:1583-91. [PMID: 20390416 PMCID: PMC11030090 DOI: 10.1007/s00262-010-0853-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Accepted: 03/26/2010] [Indexed: 10/19/2022]
Abstract
After more than 15 years of experimentation, DNA vaccines have become a promising perspective for tumour diseases, and animal models are widely used to study the biological features of human cancer progression and to test the efficacy of vaccination protocols. In recent years, immunisation with naked plasmid DNA encoding tumour-associated antigens or tumour-specific antigens has revealed a number of advantages: antigen-specific DNA vaccination stimulates both cellular and humoral immune responses; multiple or multi-gene vectors encoding several antigens/determinants and immune-modulatory molecules can be delivered as single administration; DNA vaccination does not induce autoimmune disease in normal animals; DNA vaccines based on plasmid vectors can be produced and tested rapidly and economically. However, DNA vaccines have shown low immunogenicity when tested in human clinical trials, and compared with traditional vaccines, they induce weak immune responses. Therefore, the improvement of vaccine efficacy has become a critical goal in the development of effective DNA vaccination protocols for anti-tumour therapy. Several strategies are taken into account for improving the DNA vaccination efficacy, such as antigen optimisation, use of adjuvants and delivery systems like electroporation, co-expression of cytokines and co-stimulatory molecules in the same vector, different vaccination protocols. In this review we discuss how the combination of these approaches may contribute to the development of more effective DNA vaccination protocols for the therapy of lymphoma in a mouse model.
Collapse
Affiliation(s)
- Emanuela Signori
- CNR-Institute of Neurobiology and Molecular Medicine, Via Fosso del Cavaliere100, 00133 Rome, Italy
- Section of Molecular Medicine and Biotechnology, PRABB, Centre of Integrated Research, Università Campus Bio-Medico di Roma, Via A. del Portillo 21, 00128 Rome, Italy
| | - Sandra Iurescia
- CNR-Institute of Neurobiology and Molecular Medicine, Via Fosso del Cavaliere100, 00133 Rome, Italy
| | - Emanuela Massi
- CNR-Institute of Neurobiology and Molecular Medicine, Via Fosso del Cavaliere100, 00133 Rome, Italy
- Section of Molecular Medicine and Biotechnology, PRABB, Centre of Integrated Research, Università Campus Bio-Medico di Roma, Via A. del Portillo 21, 00128 Rome, Italy
| | - Daniela Fioretti
- CNR-Institute of Neurobiology and Molecular Medicine, Via Fosso del Cavaliere100, 00133 Rome, Italy
| | - Pieranna Chiarella
- CNR-Institute of Neurobiology and Molecular Medicine, Via Fosso del Cavaliere100, 00133 Rome, Italy
- Section of Molecular Medicine and Biotechnology, PRABB, Centre of Integrated Research, Università Campus Bio-Medico di Roma, Via A. del Portillo 21, 00128 Rome, Italy
| | - Mariangela De Robertis
- CNR-Institute of Neurobiology and Molecular Medicine, Via Fosso del Cavaliere100, 00133 Rome, Italy
- Section of Molecular Medicine and Biotechnology, PRABB, Centre of Integrated Research, Università Campus Bio-Medico di Roma, Via A. del Portillo 21, 00128 Rome, Italy
| | - Monica Rinaldi
- CNR-Institute of Neurobiology and Molecular Medicine, Via Fosso del Cavaliere100, 00133 Rome, Italy
| | - Giancarlo Tonon
- Bio-ker S.r.l., POLARIS, Località Piscinamanna, 09010 Pula, Cagliari Italy
| | - Vito Michele Fazio
- Section of Molecular Medicine and Biotechnology, PRABB, Centre of Integrated Research, Università Campus Bio-Medico di Roma, Via A. del Portillo 21, 00128 Rome, Italy
| |
Collapse
|
32
|
Ni J, Schirrmacher V, Fournier P. The hemagglutinin-neuraminidase gene of Newcastle Disease Virus: a powerful molecular adjuvant for DNA anti-tumor vaccination. Vaccine 2010; 28:6891-900. [PMID: 20709006 DOI: 10.1016/j.vaccine.2010.08.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Revised: 07/07/2010] [Accepted: 08/02/2010] [Indexed: 10/19/2022]
Abstract
Plasmid-encoded DNA vaccine is a novel and potentially powerful tool for cancer therapy. Since the strength of immune responses induced by DNA vaccine is usually rather low, a major goal in DNA vaccine development is to enhance vaccine-induced immunity. In this study, we investigated an approach based on the use of a viral surface protein with pleiotropic function as a potential immune enhancer. To this end, we prepared bicistronic DNA plasmids encoding the hemagglutinin-neuraminidase (HN) protein of Newcastle Disease Virus in addition to a tumor target antigen. We demonstrate a higher tumor antigen-specific T cell-mediated immune response and a lower humoral response upon vaccination with a bicistronic DNA plasmid with incorporated HN gene. In a prophylactic immunization tumor model with the surrogate tumor antigen beta-galactosidase (β-gal) and in a therapeutic immunization tumor model with the xenogeneic tumor antigen human Epithelial Cell Adhesion Molecule (hEpCAM), HN gene incorporation into the DNA vaccine led to better survival and tumor regression in mice. There was also cross protection in the therapeutic tumor model against a second challenge by the parental mouse mammary carcinoma cells in mice vaccinated with the bicistronic plasmids. This is the first report describing the HN protein as an immunomodulator for enhanced antigen-specific T cell responses via DNA plasmids. The results show that co-expression of HN with a tumor target antigen through bicistronic vectors ensures precise temporal and spatial co-delivery to direct anti-tumor immune responses preferentially towards Th1.
Collapse
Affiliation(s)
- Jing Ni
- Tumorimmunology Program, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | | | | |
Collapse
|
33
|
Carvalho JA, Azzoni AR, Prazeres DMF, Monteiro GA. Comparative analysis of antigen-targeting sequences used in DNA vaccines. Mol Biotechnol 2010; 44:204-12. [PMID: 20013075 DOI: 10.1007/s12033-009-9229-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Plasmid vectors can be optimized by including specific signals that promote antigen targeting to the major antigen presentation and processing pathways, increasing the immunogenicity and potency of DNA vaccines. A pVAX1-based backbone was used to encode the Green Fluorescence Protein (GFP) reporter gene fused either to ISG (Invariant Surface Glycoprotein) or to TSA (trans-sialidase) Trypanosoma brucei genes. The plasmids were further engineered to carry antigen-targeting sequences, which promote protein transport to the extracellular space (secretion signal), lysosomes (LAMP-1) and to the endoplasmic reticulum (adenovirus e1a). Transfection efficiency was not affected by differences in the size between each construct as no differences in the plasmid copy number per cell were found. This finding also suggests that the addition of both ISG gene and targeting sequences did not add sensitive regions prone to nuclease attack to the plasmid. Cells transfected with pVAX1GFP had a significant higher number of transcripts. This could be a result of lower mRNA stability and/or a lower transcription rate associated with the bigger transcripts. On the other hand, no differences were found between transcript levels of each ISG-GFP plasmids. Therefore, the addition of these targeting sequences does not affect the maturation/stability of the transcripts. Microscopy analysis showed differences in protein localization and fluorescent levels of cells transfected with pVAX1GFP and ISG constructs. Moreover, cells transfected with the lamp and secretory sequences presented a distinct distribution pattern when compared with ISG protein. Protein expression was quantified by flow cytometry. Higher cell fluorescence was observed in cells expressing the cytoplasmic fusion protein (ISG-GFP or TSA-GFP) compared with cells where the protein was transported to the lysosomal pathway. Protein transport to the endoplasmic reticulum does not lead to a decrease in the mean fluorescence values. The secretion signal was only effective when used in conjunction with TSA gene. Therefore, the characteristics of each protein (e.g., presence of transmembrane domains) might influence the efficacy of its cellular transport. This analysis constitutes a useful tool for the optimization of the design of DNA vaccines.
Collapse
Affiliation(s)
- Joana A Carvalho
- IBB-Institute for Biotechnology and Bioengineering, Centre for Biological and Chemical Engineering, Instituto Superior Técnico, Lisbon, Portugal
| | | | | | | |
Collapse
|
34
|
Hendrickson RC, Cicinnati VR, Albers A, Dworacki G, Gambotto A, Pagliano O, Tüting T, Mayordomo JI, Visus C, Appella E, Shabanowitz J, Hunt DF, DeLeo AB. Identification of a 17beta-hydroxysteroid dehydrogenase type 12 pseudogene as the source of a highly restricted BALB/c Meth A tumor rejection peptide. Cancer Immunol Immunother 2010; 59:113-24. [PMID: 19562340 PMCID: PMC2855844 DOI: 10.1007/s00262-009-0730-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2009] [Accepted: 06/03/2009] [Indexed: 12/23/2022]
Abstract
Mass spectrometric analysis identified the peptide recognized by a cytotoxic T lymphocyte (CTL) specific for the chemically induced BALB/c Meth A sarcoma as derived from a 17beta-hydroxysteroid dehydrogenase type 12 (Hsd17b12) pseudogene present in the BALB/c genome, but only expressed in Meth A sarcoma. The sequence of the peptide is TYDKIKTGL and corresponds to Hsd17b12(114-122) with threonine instead of isoleucine at codon 114 and is designated Hsd17b12(114T). Immunization of mice with an Hsd17b12(114T) peptide-pulsed dendritic cell-based vaccine or a non-viral plasmid construct expressing the Hsd17b12(114T) peptide protected the mice from lethal Meth A tumor challenge in tumor rejection assays. A Hsd17b12(114-122) peptide-pulsed vaccine was ineffective in inducing resistance in mice to Meth A sarcoma. These results confirm the immunogenicity of the identified tumor peptide, as well as demonstrate the efficacies of these vaccine vehicles. These findings suggest that the role of the human homolog of Hsd17b12, HSD17B12, as a potential human tumor antigen be explored.
Collapse
Affiliation(s)
| | - Vito R. Cicinnati
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Andreas Albers
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Grzegorz Dworacki
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Andrea Gambotto
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Ornella Pagliano
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Thomas Tüting
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Jose I. Mayordomo
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Carmen Visus
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| | - Ettore Appella
- Laboratory of Cell Biology, National Cancer Institute, Bethesda, MD 20892 USA
| | - Jeffrey Shabanowitz
- Department of Chemistry, University of Virginia, Charlottesville, VA, 22901 USA
| | - Donald F. Hunt
- Department of Chemistry, University of Virginia, Charlottesville, VA, 22901 USA
- Department of Pathology, University of Virginia, Charlottesville, VA, 22901 USA
| | - Albert B. DeLeo
- Division of Basic Research, Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Division of Basic Research, Hillman Cancer Center, Research Pavilion, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15213-2582 USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213 USA
| |
Collapse
|
35
|
The Place of the Electroporation-Based Antitumor Therapies in the Electrical Armamentarium against Cancer. IRREVERSIBLE ELECTROPORATION 2010. [DOI: 10.1007/978-3-642-05420-4_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
36
|
Livingston BD, Little SF, Luxembourg A, Ellefsen B, Hannaman D. Comparative performance of a licensed anthrax vaccine versus electroporation based delivery of a PA encoding DNA vaccine in rhesus macaques. Vaccine 2010; 28:1056-61. [DOI: 10.1016/j.vaccine.2009.10.111] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Revised: 10/16/2009] [Accepted: 10/19/2009] [Indexed: 11/27/2022]
|
37
|
Kang TH, Chung JY, Monie A, Pai SI, Hung CF, Wu TC. Enhancing DNA vaccine potency by co-administration of xenogenic MHC class-I DNA. Gene Ther 2009; 17:531-40. [PMID: 19940864 PMCID: PMC2851845 DOI: 10.1038/gt.2009.152] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Intramuscular administration of DNA vaccines can lead to the generation of antigen-specific immune responses through cross-priming mechanisms. We propose a strategy that is capable of leading to local inflammation and enhancing cross-priming, thus resulting in improved antigen-specific immune responses. Therefore, in the current study, we evaluated immunologic responses elicited through electroporation mediated intramuscular administration of a DNA vaccine encoding calreticulin (CRT) linked to HPV-16 E7 (CRT/E7) in combination with DNA expressing HLA-A2 as compared to CRT/E7 DNA vaccination alone. We found that the co-administration of a DNA vaccine in conjunction with a DNA encoding an xenogenic MHC molecule could significantly enhance the E7-specific CD8+ T cell immune responses as well an antitumor effects against an E7-expressing tumor, TC-1 in C57BL/6 tumor-bearing mice. Furthermore, a similar enhancement in E7-specific immune responses was observed by co-administration of CRT/E7 DNA with DNA encoding other types of xenogenic MHC class I molecules. This strategy was also applicable to another antigenic system, ovalbumin. Further characterization of the injection site revealed that co-administration of HLA-A2 DNA led to a significant increase in the number of infiltrating CD8+ T lymphocytes as well as CD11b/c+ antigen presenting cells. Furthermore, the E7-specific immune responses generated by intramuscular co-administration of CRT/E7 with HLA-A2 DNA were reduced in HLA-A2 transgenic mice. Thus, our data suggest that intramuscular co-administration of DNA encoding xenogenic MHC class I can further improve the antigen-specific immune responses as well as antitumor effects generated by DNA vaccines through enhancement of cross-priming mechanisms.
Collapse
Affiliation(s)
- T H Kang
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | | | | | | | | | | |
Collapse
|
38
|
Yin J, Dai A, Laddy DJ, Yan J, Arango T, Khan AS, Lewis MG, Andersen H, Kutzler MA, Draghia-Akli R, Weiner DB, Boyer JD. High dose of plasmid IL-15 inhibits immune responses in an influenza non-human primates immunogenicity model. Virology 2009; 393:49-55. [PMID: 19683780 PMCID: PMC4118595 DOI: 10.1016/j.virol.2009.07.017] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2009] [Revised: 06/12/2009] [Accepted: 07/10/2009] [Indexed: 01/08/2023]
Abstract
Interleukin (IL)-15, is a cytokine that is important for the maintenance of long-lasting, high-avidity T cell response to invading pathogens and has, therefore, been used in vaccine and therapeutic platforms as an adjuvant. In addition to pure protein delivery, plasmids encoding the IL-15 gene have been utilized. However, it is critical to determine the appropriate dose to maximize the adjuvanting effects. We immunized rhesus macaques with different doses of IL-15 expressing plasmid in an influenza non-human primate immunogenicity model. We found that co-immunization of rhesus macaques with a Flu DNA-based vaccine and low doses of plasmid encoding macaque IL-15 enhanced the production of IFN-gamma (0.5 mg) and the proliferation of CD4(+) and CD8(+) T cells, as well as T(CM) levels in proliferating CD8(+) T cells (0.25 mg). Whereas, high doses of IL-15 (4 mg) decrease the production of IFN-gamma and the proliferation of CD4(+) and CD8(+) T cells and T(CM) levels in the proliferating CD4(+) and CD8(+) T cells. In addition, the data of hemagglutination inhibition (HI) antibody titer suggest that although not significantly different, there appears to be a slight increase in antibodies at lower doses of IL-15. Importantly, however, the higher doses of IL-15 decrease the antibody levels significantly. This study demonstrates the importance of optimizing DNA-based cytokine adjuvants.
Collapse
Affiliation(s)
- Jiangmei Yin
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 505 SCL, 422 Curie Blvd. Philadelphia, PA 19104, USA
| | - Anlan Dai
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 505 SCL, 422 Curie Blvd. Philadelphia, PA 19104, USA
| | - Dominick J. Laddy
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 505 SCL, 422 Curie Blvd. Philadelphia, PA 19104, USA
| | - Jian Yan
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 505 SCL, 422 Curie Blvd. Philadelphia, PA 19104, USA
| | - Tatiana Arango
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 505 SCL, 422 Curie Blvd. Philadelphia, PA 19104, USA
| | - Amir S. Khan
- VGX Pharmaceuticals, Inc., The Woodlands, TX 77381, USA
| | | | | | | | | | - David B. Weiner
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 505 SCL, 422 Curie Blvd. Philadelphia, PA 19104, USA
| | - Jean D. Boyer
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, 505 SCL, 422 Curie Blvd. Philadelphia, PA 19104, USA
| |
Collapse
|
39
|
Electroporation of synthetic DNA antigens offers protection in nonhuman primates challenged with highly pathogenic avian influenza virus. J Virol 2009; 83:4624-30. [PMID: 19211745 DOI: 10.1128/jvi.02335-08] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Avian influenza highlights the need for novel vaccination techniques that would allow for the rapid design and production of safe and effective vaccines. An ideal platform would be capable of inducing both protective antibodies and potent cellular immune responses. These potential advantages of DNA vaccines remain unrealized due to a lack of efficacy in large animal studies and in human trials. Questions remain regarding the potential utility of cellular immune responses against influenza virus in primates. In this study, by construct optimization and in vivo electroporation of synthetic DNA-encoded antigens, we observed the induction of cross-reactive cellular and humoral immune responses individually capable of providing protection from influenza virus infection in the rhesus macaque. These studies advance the DNA vaccine field and provide a novel, more tolerable vaccine with broad immunogenicity to avian influenza virus. This approach appears important for further investigation, including studies with humans.
Collapse
|
40
|
Brown PA, Bodles-Brakhop AM, Pope MA, Draghia-Akli R. Gene therapy by electroporation for the treatment of chronic renal failure in companion animals. BMC Biotechnol 2009; 9:4. [PMID: 19149896 PMCID: PMC2663557 DOI: 10.1186/1472-6750-9-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2008] [Accepted: 01/16/2009] [Indexed: 12/17/2022] Open
Abstract
Background Growth hormone-releasing hormone (GHRH) plasmid-based therapy for the treatment of chronic renal failure and its complications was examined. Companion dogs (13.1 ± 0.8 years, 29.4 ± 5.01 kg) and cats (13.2 ± 0.9 years, 8.5 ± 0.37 kg) received a single 0.4 mg or 0.1 mg species-specific plasmid injection, respectively, intramuscularly followed by electroporation, and analyzed up to 75 days post-treatment; controls underwent electroporation without plasmid administration. Results Plasmid-treated animals showed an increase in body weight (dogs 22.5% and cats 3.2%) compared to control animals, and displayed improved quality of life parameters including significant increases in appetite, activity, mentation and exercise tolerance levels. Insulin-like growth factor I (IGF-I, the downstream effector of GHRH) levels were increased in the plasmid treated animals. Hematological parameters were also significantly improved. Protein metabolism changes were observed suggesting a shift from a catabolic to an anabolic state in the treated animals. Blood urea nitrogen and creatinine did not show any significant changes suggesting maintenance of kidney function whereas the control animal's renal function deteriorated. Treated animals survived longer than control animals with 70% of dogs and 80% of cats surviving until study day 75. Only 17% and 40% of the control dogs and cats, respectively, survived to day 75. Conclusion Improved quality of life, survival and general well-being indicate that further investigation is warranted, and show the potential of a plasmid-based therapy by electroporation in preventing and managing complications of renal insufficiency.
Collapse
Affiliation(s)
- Patricia A Brown
- VGX Animal Health, 2700 Research Forest Drive, Suite 180, The Woodlands, Texas 77381, USA.
| | | | | | | |
Collapse
|