1
|
Zhang X, Goedegebuure SP, Chen MY, Mishra R, Zhang F, Yu YY, Singhal K, Li L, Gao F, Myers NB, Vickery T, Hundal J, McLellan MD, Sturmoski MA, Kim SW, Chen I, Davidson JT, Sankpal NV, Myles S, Suresh R, Ma CX, Foluso A, Wang-Gillam A, Davies S, Hagemann IS, Mardis ER, Griffith O, Griffith M, Miller CA, Hansen TH, Fleming TP, Schreiber RD, Gillanders WE. Neoantigen DNA vaccines are safe, feasible, and induce neoantigen-specific immune responses in triple-negative breast cancer patients. Genome Med 2024; 16:131. [PMID: 39538331 PMCID: PMC11562513 DOI: 10.1186/s13073-024-01388-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 09/20/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Neoantigen vaccines can induce or enhance highly specific antitumor immune responses with minimal risk of autoimmunity. We have developed a neoantigen DNA vaccine platform capable of efficiently presenting both HLA class I and II epitopes and performed a phase 1 clinical trial in triple-negative breast cancer patients with persistent disease on surgical pathology following neoadjuvant chemotherapy, a patient population at high risk of disease recurrence. METHODS Expressed somatic mutations were identified by tumor/normal exome sequencing and tumor RNA sequencing. The pVACtools software suite of neoantigen prediction algorithms was used to identify and prioritize cancer neoantigens and facilitate vaccine design for manufacture in an academic GMP facility. Neoantigen DNA vaccines were administered via electroporation in the adjuvant setting (i.e., following surgical removal of the primary tumor and completion of standard of care therapy). Vaccines were monitored for safety and immune responses via ELISpot, intracellular cytokine production via flow cytometry, and TCR sequencing. RESULTS Eighteen subjects received three doses of a neoantigen DNA vaccine encoding on average 11 neoantigens per patient (range 4-20). The vaccinations were well tolerated with relatively few adverse events. Neoantigen-specific T cell responses were induced in 14/18 patients as measured by ELISpot and flow cytometry. At a median follow-up of 36 months, recurrence-free survival was 87.5% (95% CI: 72.7-100%) in the cohort of vaccinated patients. CONCLUSION Our study demonstrates neoantigen DNA vaccines are safe, feasible, and capable of inducing neoantigen-specific immune responses. CLINICAL TRIAL REGISTRATION NUMBER NCT02348320.
Collapse
Affiliation(s)
- Xiuli Zhang
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - S Peter Goedegebuure
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Michael Y Chen
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Rashmi Mishra
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Felicia Zhang
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Yik Yeung Yu
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kartik Singhal
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Lijin Li
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Feng Gao
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Nancy B Myers
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Tammi Vickery
- Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jasreet Hundal
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Michael D McLellan
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Mark A Sturmoski
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Samuel W Kim
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ina Chen
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Jesse T Davidson
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Narendra V Sankpal
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Stephanie Myles
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Rama Suresh
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Cynthia X Ma
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Ademuyiwa Foluso
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Andrea Wang-Gillam
- Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Sherri Davies
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ian S Hagemann
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Elaine R Mardis
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
- Current Affiliation: Department of Pediatrics, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
| | - Obi Griffith
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Malachi Griffith
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Christopher A Miller
- McDonnell Genome Institute, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ted H Hansen
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
| | - Timothy P Fleming
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - Robert D Schreiber
- Bursky Center for Human Immunology & Immunotherapy Programs, Washington University School of Medicine, Saint Louis, MO, USA
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA
| | - William E Gillanders
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA.
- The Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
2
|
Kozak M, Hu J. DNA Vaccines: Their Formulations, Engineering and Delivery. Vaccines (Basel) 2024; 12:71. [PMID: 38250884 PMCID: PMC10820593 DOI: 10.3390/vaccines12010071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/02/2024] [Accepted: 01/08/2024] [Indexed: 01/23/2024] Open
Abstract
The concept of DNA vaccination was introduced in the early 1990s. Since then, advancements in the augmentation of the immunogenicity of DNA vaccines have brought this technology to the market, especially in veterinary medicine, to prevent many diseases. Along with the successful COVID mRNA vaccines, the first DNA vaccine for human use, the Indian ZyCovD vaccine against SARS-CoV-2, was approved in 2021. In the current review, we first give an overview of the DNA vaccine focusing on the science, including adjuvants and delivery methods. We then cover some of the emerging science in the field of DNA vaccines, notably efforts to optimize delivery systems, better engineer delivery apparatuses, identify optimal delivery sites, personalize cancer immunotherapy through DNA vaccination, enhance adjuvant science through gene adjuvants, enhance off-target and heritable immunity through epigenetic modification, and predict epitopes with bioinformatic approaches. We also discuss the major limitations of DNA vaccines and we aim to address many theoretical concerns.
Collapse
Affiliation(s)
- Michael Kozak
- The Jake Gittlen Laboratories for Cancer Research, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
- The Department of Pathology and Laboratory Medicine, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| | - Jiafen Hu
- The Jake Gittlen Laboratories for Cancer Research, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
- The Department of Pathology and Laboratory Medicine, College of Medicine, Pennsylvania State University, Hershey, PA 17033, USA
| |
Collapse
|
3
|
de Lima MR, Leandro ACCS, de Souza AL, Barradas MM, Roma EH, Fernandes ATG, Galdino-Silva G, Carvalho JKMR, Marchevsky RS, Coelho JMCO, Gonçalves EDC, VandeBerg JL, Silva CL, Bonecini-Almeida MDG. Safety and Immunogenicity of an In Vivo Muscle Electroporation Delivery System for DNA- hsp65 Tuberculosis Vaccine in Cynomolgus Monkeys. Vaccines (Basel) 2023; 11:1863. [PMID: 38140266 PMCID: PMC10747856 DOI: 10.3390/vaccines11121863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023] Open
Abstract
A Bacille Calmette-Guérin (BCG) is still the only licensed vaccine for the prevention of tuberculosis, providing limited protection against Mycobacterium tuberculosis infection in adulthood. New advances in the delivery of DNA vaccines by electroporation have been made in the past decade. We evaluated the safety and immunogenicity of the DNA-hsp65 vaccine administered by intramuscular electroporation (EP) in cynomolgus macaques. Animals received three doses of DNA-hsp65 at 30-day intervals. We demonstrated that intramuscular electroporated DNA-hsp65 vaccine immunization of cynomolgus macaques was safe, and there were no vaccine-related effects on hematological, renal, or hepatic profiles, compared to the pre-vaccination parameters. No tuberculin skin test conversion nor lung X-ray alteration was identified. Further, low and transient peripheral cellular immune response and cytokine expression were observed, primarily after the third dose of the DNA-hsp65 vaccine. Electroporated DNA-hsp65 vaccination is safe but provides limited enhancement of peripheral cellular immune responses. Preclinical vaccine trials with DNA-hsp65 delivered via EP may include a combination of plasmid cytokine adjuvant and/or protein prime-boost regimen, to help the induction of a stronger cellular immune response.
Collapse
Affiliation(s)
- Monique Ribeiro de Lima
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
| | - Ana Cristina C. S. Leandro
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
- Division of Human Genetics, South Texas Diabetes and Obesity Institute, The University of Texas Rio Grande Valley, Brownsville, TX 78520, USA;
| | - Andreia Lamoglia de Souza
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
| | - Marcio Mantuano Barradas
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
| | - Eric Henrique Roma
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
| | - Ana Teresa Gomes Fernandes
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
| | - Gabrielle Galdino-Silva
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
| | - Joyce Katiuccia M. Ramos Carvalho
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
| | - Renato Sergio Marchevsky
- Laboratory of Neurovirulence, Instituto de Biotecnologia em Imunobiológicos, Biomanguinhos, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil;
| | - Janice M. C. Oliveira Coelho
- Laboratory of Pathology, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil;
| | | | - John L. VandeBerg
- Division of Human Genetics, South Texas Diabetes and Obesity Institute, The University of Texas Rio Grande Valley, Brownsville, TX 78520, USA;
| | - Celio Lopes Silva
- Farmacore Biotecnologia Ltda, Ribeirão Preto 14056-680, SP, Brazil; (E.D.C.G.); (C.L.S.)
- Laboratory for Research and Development of Immunobiologicals, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Maria da Gloria Bonecini-Almeida
- Laboratory of Immunology and Immunogenetic in Infectious Diseases, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro 21040-360, RJ, Brazil; (M.R.d.L.); (A.C.C.S.L.); (A.L.d.S.); (M.M.B.); (E.H.R.); (A.T.G.F.); (G.G.-S.); (J.K.M.R.C.)
| |
Collapse
|
4
|
Dong L, Feng M, Qiao Y, Liu C, Zhou Y, Xing S, Zhang K, Cai Z, Wu H, Wu J, Yu X, Zhang H, Kong W. Preclinical safety and Biodistribution in mice following single dose intramuscular inoculation of tumor DNA vaccine by electroporation. Hum Gene Ther 2022; 33:757-764. [DOI: 10.1089/hum.2022.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Ling Dong
- Jilin University, 12510, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Changchun, Jilin, China
| | - Mengfan Feng
- Jilin University, 12510, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Changchun, Jilin, China
| | - Yaru Qiao
- Jilin University, 12510, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Changchun, Jilin, China
| | - Chenlu Liu
- Jilin University, 12510, Department of Biobank, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yi Zhou
- Jilin University, 12510, Changchun, China
| | - Shanshan Xing
- Jilin University, 12510, Changchun, Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, China
| | - Ke Zhang
- Jilin University, 12510, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Changchun, Jilin, China
| | - Zongyu Cai
- Jilin University, 12510, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Changchun, Jilin, China
| | - Hui Wu
- Jilin University, 12510, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Changchun, Jilin, China
| | - Jiaxin Wu
- Jilin University, 12510, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Changchun, Jilin, China
| | - Xianghui Yu
- Jilin University, 12510, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, School of Life Sciences, Jilin University, Changchun, Changchun, Jilin, China, 130012
- Jilin University, 12510, Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, School of Life Sciences, Jilin University, Changchun, Changchun, Jilin, China, 130012
| | - Haihong Zhang
- Jilin University, 12510, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Changchun, Jilin, China
| | - Wei Kong
- Jilin University, 12510, National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Changchun, Jilin, China
- Jilin University, 12510, Key Laboratory for Molecular Enzymology and Engineering, the Ministry of Education, School of Life Sciences, Changchun, China
| |
Collapse
|
5
|
Tang J, Li M, Zhao C, Shen D, Liu L, Zhang X, Wei L. Therapeutic DNA Vaccines against HPV-Related Malignancies: Promising Leads from Clinical Trials. Viruses 2022; 14:v14020239. [PMID: 35215833 PMCID: PMC8874761 DOI: 10.3390/v14020239] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/19/2022] [Accepted: 01/22/2022] [Indexed: 01/27/2023] Open
Abstract
In 2014 and 2021, two nucleic-acid vaccine candidates named MAV E2 and VGX-3100 completed phase III clinical trials in Mexico and U.S., respectively, for patients with human papillomavirus (HPV)-related, high-grade squamous intraepithelial lesions (HSIL). These well-tolerated but still unlicensed vaccines encode distinct HPV antigens (E2 versus E6+E7) to elicit cell-mediated immune responses; their clinical efficacy, as measured by HSIL regression or cure, was modest when compared with placebo or surgery (conization), but both proved highly effective in clearing HPV infection, which should help further optimize strategies for enhancing vaccine immunogenicity, toward an ultimate goal of preventing malignancies in millions of patients who are living with persistent, oncogenic HPV infection but are not expected to benefit from current, prophylactic vaccines. The major roadblocks to a highly efficacious and practical product remain challenging and can be classified into five categories: (i) getting the vaccines into the right cells for efficient expression and presentation of HPV antigens (fusion proteins or epitopes); (ii) having adequate coverage of oncogenic HPV types, beyond the current focus on HPV-16 and -18; (iii) directing immune protection to various epithelial niches, especially anogenital mucosa and upper aerodigestive tract where HPV-transformed cells wreak havoc; (iv) establishing the time window and vaccination regimen, including dosage, interval and even combination therapy, for achieving maximum efficacy; and (v) validating therapeutic efficacy in patients with poor prognosis because of advanced, recurrent or non-resectable malignancies. Overall, the room for improvements is still large enough that continuing efforts for research and development will very likely extend into the next decade.
Collapse
Affiliation(s)
- Jianming Tang
- Aeonvital Biomedical Research Institute, Beijing 102208, China; (L.L.); (X.Z.)
- Correspondence: or
| | - Mingzhu Li
- Department of Gynecology and Obstetrics, Peking University People’s Hospital, Beijing 100033, China; (M.L.); (C.Z.); (D.S.); (L.W.)
| | - Chao Zhao
- Department of Gynecology and Obstetrics, Peking University People’s Hospital, Beijing 100033, China; (M.L.); (C.Z.); (D.S.); (L.W.)
| | - Danhua Shen
- Department of Gynecology and Obstetrics, Peking University People’s Hospital, Beijing 100033, China; (M.L.); (C.Z.); (D.S.); (L.W.)
| | - Lei Liu
- Aeonvital Biomedical Research Institute, Beijing 102208, China; (L.L.); (X.Z.)
| | - Xiujun Zhang
- Aeonvital Biomedical Research Institute, Beijing 102208, China; (L.L.); (X.Z.)
| | - Lihui Wei
- Department of Gynecology and Obstetrics, Peking University People’s Hospital, Beijing 100033, China; (M.L.); (C.Z.); (D.S.); (L.W.)
| |
Collapse
|
6
|
Ribeiro SP, De Moura Mattaraia VG, Almeida RR, Valentine EJG, Sales NS, Ferreira LCS, Sa-Rocha LC, Jacintho LC, Santana VC, Sidney J, Sette A, Rosa DS, Kalil J, Cunha-Neto E. A promiscuous T cell epitope-based HIV vaccine providing redundant population coverage of the HLA class II elicits broad, polyfunctional T cell responses in nonhuman primates. Vaccine 2021; 40:239-246. [PMID: 34961636 DOI: 10.1016/j.vaccine.2021.11.076] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/07/2021] [Accepted: 11/24/2021] [Indexed: 11/15/2022]
Abstract
Over the last few decades, several emerging or reemerging viral diseases with no readily available vaccines have ravaged the world. A platform to fastly generate vaccines inducing potent and durable neutralizing antibody and T cell responses is sorely needed. Bioinformatically identified epitope-based vaccines can focus on immunodominant T cell epitopes and induce more potent immune responses than a whole antigen vaccine and may be deployed more rapidly and less costly than whole-gene vaccines. Increasing evidence has shown the importance of the CD4+ T cell response in protection against HIV and other viral infections. The previously described DNA vaccine HIVBr18 encodes 18 conserved, promiscuous epitopes binding to multiple HLA-DR-binding HIV epitopes amply recognized by HIV-1-infected patients. HIVBr18 elicited broad, polyfunctional, and durable CD4+and CD8+ T cell responses in BALB/c and mice transgenic to HLA class II alleles, showing cross-species promiscuity. To fully delineate the promiscuity of the HLA class II vaccine epitopes, we assessed their binding to 34 human class II (HLA-DR, DQ, and -DP) molecules, and immunized nonhuman primates. Results ascertained redundant 100% coverage of the human population for multiple peptides. We then immunized Rhesus macaques with HIVBr18 under in vivo electroporation. The immunization induced strong, predominantly polyfunctional CD4+ T cell responses in all animals to 13 out of the 18 epitopes; T cells from each animal recognized 7-11 epitopes. Our results provide a preliminary proof of concept that immunization with a vaccine encoding epitopes with high and redundant coverage of the human population can elicit potent T cell responses to multiple epitopes, across species and MHC barriers. This approach may facilitate the rapid deployment of immunogens eliciting cellular immunity against emerging infectious diseases, such as COVID-19.
Collapse
Affiliation(s)
- Susan Pereira Ribeiro
- Emory University, Atlanta, USA; Laboratory of Clinical Immunology and Allergy-LIM60/University of Sao Paulo School of Medicine, São Paulo, Brazil; Institute for Investigation in Immunology - iii-INCT, São Paulo, Brazil; Laboratory of Immunology, Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil
| | | | - Rafael Ribeiro Almeida
- Laboratory of Clinical Immunology and Allergy-LIM60/University of Sao Paulo School of Medicine, São Paulo, Brazil; Institute for Investigation in Immunology - iii-INCT, São Paulo, Brazil; Laboratory of Immunology, Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil
| | | | - Natiely Silva Sales
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | - Luís Carlos S Ferreira
- Department of Microbiology, Institute of Biomedical Sciences, University of Sao Paulo, São Paulo, Brazil
| | | | - Lucas Cauê Jacintho
- Laboratory of Clinical Immunology and Allergy-LIM60/University of Sao Paulo School of Medicine, São Paulo, Brazil; Institute for Investigation in Immunology - iii-INCT, São Paulo, Brazil
| | - Vinicius Canato Santana
- Laboratory of Clinical Immunology and Allergy-LIM60/University of Sao Paulo School of Medicine, São Paulo, Brazil; Institute for Investigation in Immunology - iii-INCT, São Paulo, Brazil; Laboratory of Immunology, Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil
| | - John Sidney
- La Jolla Institute for Immunology (LJI), LA Jolla, CA, USA
| | | | - Daniela Santoro Rosa
- Institute for Investigation in Immunology - iii-INCT, São Paulo, Brazil; Department of Microbiology, Immunology and Parasitology, Federal University of São Paulo (UNIFESP/EPM), São Paulo, Brazil
| | - Jorge Kalil
- Laboratory of Clinical Immunology and Allergy-LIM60/University of Sao Paulo School of Medicine, São Paulo, Brazil; Institute for Investigation in Immunology - iii-INCT, São Paulo, Brazil; Laboratory of Immunology, Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratory of Clinical Immunology and Allergy-LIM60/University of Sao Paulo School of Medicine, São Paulo, Brazil; Institute for Investigation in Immunology - iii-INCT, São Paulo, Brazil; Laboratory of Immunology, Heart Institute, University of São Paulo School of Medicine, São Paulo, Brazil.
| |
Collapse
|
7
|
Arora S, Sharma D, Layek B, Singh J. A Review of Brain-Targeted Nonviral Gene-Based Therapies for the Treatment of Alzheimer's Disease. Mol Pharm 2021; 18:4237-4255. [PMID: 34705472 DOI: 10.1021/acs.molpharmaceut.1c00611] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diseases of the central nervous system (CNS) are difficult to treat owing to the complexity of the brain and the presence of a natural blood-brain-barrier (BBB). Alzheimer's disease (AD) is one of the major progressive and currently incurable neurodegenerative disorders of the CNS, which accounts for 60-80% of cases of dementia. The pathophysiology of AD involves the accumulation of amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain. Additionally, synaptic loss and imbalance of neuronal signaling molecules are characterized as important markers of AD. Existing treatments of AD help in the management of its symptoms and aim toward the maintenance of cognitive functions, behavior, and attenuation of gradual memory loss. Over the past decade, nonviral gene therapy has attracted increasing interest due to its various advantages over its viral counterparts. Moreover, advancements in nonviral gene technology have led to their increasing contributions in clinical trials. However, brain-targeted nonviral gene delivery vectors come across various extracellular and intracellular barriers, limiting their ability to transfer the therapeutic gene into the target cells. Chief barriers to nonviral gene therapy have been discussed briefly in this review. We have also highlighted the rapid advancement of several nonviral gene therapies for AD, which are broadly categorized into physical and chemical methods. These methods aim to modulate Aβ, beta-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1), apolipoprotein E, or neurotrophic factors' expression in the CNS. Overall, this review discusses challenges and recent advancements of nonviral gene therapy for AD.
Collapse
Affiliation(s)
- Sanjay Arora
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Divya Sharma
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Buddhadev Layek
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| | - Jagdish Singh
- Department of Pharmaceutical Sciences, School of Pharmacy, College of Health Professions, North Dakota State University, Fargo, North Dakota 58105, United States
| |
Collapse
|
8
|
Fomsgaard A, Liu MA. The Key Role of Nucleic Acid Vaccines for One Health. Viruses 2021; 13:258. [PMID: 33567520 PMCID: PMC7916035 DOI: 10.3390/v13020258] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 01/07/2023] Open
Abstract
The ongoing SARS-CoV-2 pandemic has highlighted both the importance of One Health, i.e., the interactions and transmission of pathogens between animals and humans, and the potential power of gene-based vaccines, specifically nucleic acid vaccines. This review will highlight key aspects of the development of plasmid DNA Nucleic Acid (NA) vaccines, which have been licensed for several veterinary uses, and tested for a number of human diseases, and will explain how an understanding of their immunological and real-world attributes are important for their efficacy, and how they helped pave the way for mRNA vaccines. The review highlights how combining efforts for vaccine development for both animals and humans is crucial for advancing new technologies and for combatting emerging diseases.
Collapse
Affiliation(s)
- Anders Fomsgaard
- Department of Virology and Microbiological Special Diagnostic, Statens Serum Institut, 5 Artillerivej, DK-2300 Copenhagen, Denmark
| | - Margaret A. Liu
- ProTherImmune, 3656 Happy Valley Road, Lafayette, CA 94549, USA
| |
Collapse
|
9
|
Le Guen YT, Le Gall T, Midoux P, Guégan P, Braun S, Montier T. Gene transfer to skeletal muscle using hydrodynamic limb vein injection: current applications, hurdles and possible optimizations. J Gene Med 2020; 22:e3150. [PMID: 31785130 DOI: 10.1002/jgm.3150] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 11/06/2022] Open
Abstract
Hydrodynamic limb vein injection is an in vivo locoregional gene delivery method. It consists of administrating a large volume of solution containing nucleic acid constructs in a limb with both blood inflow and outflow temporarily blocked using a tourniquet. The fast, high pressure delivery allows the musculature of the whole limb to be reached. The skeletal muscle is a tissue of choice for a variety of gene transfer applications, including gene therapy for Duchenne muscular dystrophy or other myopathies, as well as for the production of antibodies or other proteins with broad therapeutic effects. Hydrodynamic limb vein delivery has been evaluated with success in a large range of animal models. It has also proven to be safe and well-tolerated in muscular dystrophy patients, thus supporting its translation to the clinic. However, some possible limitations may occur at different steps of the delivery process. Here, we have highlighted the interests, bottlenecks and potential improvements that could further optimize non-viral gene transfer following hydrodynamic limb vein injection.
Collapse
Affiliation(s)
| | - Tony Le Gall
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200, Brest, France
| | - Patrick Midoux
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Université d'Orléans, France
| | - Philippe Guégan
- Laboratoire de chimie des polymères, Sorbonne Université, CNRS UMR 8232, UPMC Paris 06, F-75005, Paris, France
| | - Serge Braun
- AFM Telethon, 1 rue de l'Internationale, BP59, 91002 Evry, France
| | - Tristan Montier
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200, Brest, France.,Service de Génétique Médicale et Biologie de la Reproduction, Centre de référence des maladies rares 'Maladies neuromusculaires', CHRU de Brest, F-29200, Brest, France
| |
Collapse
|
10
|
Non-human papillomaviruses for gene delivery in vitro and in vivo. PLoS One 2018; 13:e0198996. [PMID: 29912929 PMCID: PMC6005490 DOI: 10.1371/journal.pone.0198996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/18/2018] [Indexed: 12/17/2022] Open
Abstract
Papillomavirus capsids are known to have the ability to package DNA plasmids and deliver them both in vitro and in vivo. Of all known papillomavirus types, human papillomaviruses (HPVs) are by far the most intensely studied. Although HPVs work well as gene transfer vectors, their use is limited as most individuals are exposed to this virus either through a HPV vaccination or natural infection. To circumvent these constraints, we produced pseudovirions (PsVs) of ten non-human papillomavirus types and tested their transduction efficiencies in vitro. PsVs based on Macaca fascicularis papillomavirus-11 and Puma concolor papillomavirus-1 were further tested in vivo. Intramuscular transduction by PsVs led to months-long expression of a reporter plasmid, indicating that PsVs have potential as gene delivery vectors.
Collapse
|
11
|
Wang ZL, Zhou W, Xiong ZA, Yu TH, Wu LM, Li CX, Yao CG, Wu YT, Hua YY. Irreversible electroporation-mediated shRNA knockdown of the HPV18 E6 gene suppresses cervical cancer growth in vitro and in vivo. Oncol Lett 2017; 14:1943-1949. [PMID: 28781638 PMCID: PMC5530149 DOI: 10.3892/ol.2017.6405] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 01/12/2017] [Indexed: 12/25/2022] Open
Abstract
Irreversible electroporation (IRE) is a physical, non-thermal cancer therapy, which leads to cell death via permanent membrane permeability. This differs from reversible electroporation (RE), which is used to transfer macromolecules into target cells via transient membrane permeability. Given the electrical impedance of the electric field, RE co-exists outside the central zone of IRE ablation. In the present study, the feasibility of using IRE at a therapeutic dose to mediate short hairpin RNA (shRNA) knockdown of human papillomavirus (HPV)18 E6 in HeLa cervical cancer cells in vitro and in vivo was investigated. Experimental results indicated that the HeLa cells survived the combined treatment with IRE and shRNA plasmid transfection. Additionally, residual tumor tissue in a nude mouse model demonstrated green fluorescence. Subsequent studies showed that the combined treatment inhibited the growth of HeLa cells and tumors. Western blotting analysis showed marked changes in the growth-associated proteins between the combined treatment group and the control. It was concluded that a therapeutic dose of IRE was able to mediate the transfection of HPV18 E6 shRNA into HeLa cervical cancer cells in vitro and in vivo. This combined treatment strategy has promising implications in cancer treatment for the ablation of tumors, and in eliminating microscopic residual tumor tissue.
Collapse
Affiliation(s)
- Zhi-Liang Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Wei Zhou
- Department of Obstetrics, Chongqing Health Center for Women and Children, Chongqing 400013, P.R. China
| | - Zheng-Ai Xiong
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Teng-Hua Yu
- Department of Breast Surgery, Jiangxi Cancer Hospital, Nanchang 330029, P.R. China
| | - Li-Mei Wu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Cheng-Xiang Li
- State Key Laboratory of Power Transmission Equipment and System Security and New Technology, Chongqing University, Chongqing 400044, P.R. China
| | - Cheng-Guo Yao
- State Key Laboratory of Power Transmission Equipment and System Security and New Technology, Chongqing University, Chongqing 400044, P.R. China
| | - Yu-Tong Wu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| | - Yuan-Yuan Hua
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, P.R. China
| |
Collapse
|
12
|
Huang X, Zhu Q, Huang X, Yang L, Song Y, Zhu P, Zhou P. In vivo electroporation in DNA-VLP prime-boost preferentially enhances HIV-1 envelope-specific IgG2a, neutralizing antibody and CD8 T cell responses. Vaccine 2017; 35:2042-2051. [DOI: 10.1016/j.vaccine.2017.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 01/24/2017] [Accepted: 03/03/2017] [Indexed: 01/14/2023]
|
13
|
Pamornpathomkul B, Wongkajornsilp A, Laiwattanapaisal W, Rojanarata T, Opanasopit P, Ngawhirunpat T. A combined approach of hollow microneedles and nanocarriers for skin immunization with plasmid DNA encoding ovalbumin. Int J Nanomedicine 2017; 12:885-898. [PMID: 28184159 PMCID: PMC5291464 DOI: 10.2147/ijn.s125945] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The aim of this study was to investigate the use of different types of microneedles (MNs) and nanocarriers for in vitro skin permeation and in vivo immunization of plasmid DNA encoding ovalbumin (pOVA). In vitro skin permeation studies indicated that hollow MNs had a superior enhancing effect on skin permeation compared with solid MN patches, electroporation (EP) patches, the combination of MN and EP patches, and untreated skin. Upon using hollow MNs combined with nanocarriers for pOVA delivery, the skin permeation was higher than for the delivery of naked pOVA, as evidenced by the increased amount of pOVA in Franz diffusion cells and immunoglobulin G (IgG) antibody responses. When the hollow MNs were used for the delivery of nanocarrier:pOVA complexes into the skin of mice, they induced a stronger IgG immune response than conventional subcutaneous (SC) injections. In addition, immunization of mice with the hollow MNs did not induce signs of skin infection or pinpoint bleeding. Accordingly, the hollow MNs combined with a nanocarrier delivery system is a promising approach for delivering pOVA complexes to the skin for promoting successful immunization.
Collapse
Affiliation(s)
- Boonnada Pamornpathomkul
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Pharmaceutical Development of Green Innovations Group, Silpakorn University, Nakhon Pathom
| | - Adisak Wongkajornsilp
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok
| | - Wanida Laiwattanapaisal
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Theerasak Rojanarata
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Pharmaceutical Development of Green Innovations Group, Silpakorn University, Nakhon Pathom
| | - Praneet Opanasopit
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Pharmaceutical Development of Green Innovations Group, Silpakorn University, Nakhon Pathom
| | - Tanasait Ngawhirunpat
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Pharmaceutical Development of Green Innovations Group, Silpakorn University, Nakhon Pathom
| |
Collapse
|
14
|
Abstract
Nucleic acid vaccines are a next-generation branch of vaccines which offer major benefits over their conventional protein, bacteria, or viral-based counterparts. However, to be effective in large mammals and humans, an enhancing delivery technology is required. Electroporation is a physical technique which results in improved delivery of large molecules through the cell membrane. In the case of plasmid DNA and mRNA, electroporation enhances both the uptake and expression of the delivered nucleic acids. The muscle is an attractive tissue for nucleic acid vaccination in a clinical setting due to the accessibility and abundance of the target tissue. Historical clinical studies of electroporation in the muscle have demonstrated the procedure to be generally well tolerated in patients. Previous studies have determined that optimized electroporation parameters (such as electrical field intensity, pulse length, pulse width and drug product formulation) majorly impact the efficiency of nucleic acid delivery. We provide an overview of DNA/RNA vaccination in the muscle of mice. Our results suggest that the technique is safe and effective and is highly applicable to a research setting as well as scalable to larger animals and humans.
Collapse
Affiliation(s)
- Kate E Broderick
- Inovio Pharmaceuticals, 660 West Germantown Pike, Suite 110, Plymouth Meeting, PA, 19462, USA.
| | - Laurent M Humeau
- Inovio Pharmaceuticals, 660 West Germantown Pike, Suite 110, Plymouth Meeting, PA, 19462, USA
| |
Collapse
|
15
|
Gordy JT, Luo K, Zhang H, Biragyn A, Markham RB. Fusion of the dendritic cell-targeting chemokine MIP3α to melanoma antigen Gp100 in a therapeutic DNA vaccine significantly enhances immunogenicity and survival in a mouse melanoma model. J Immunother Cancer 2016; 4:96. [PMID: 28018602 PMCID: PMC5168589 DOI: 10.1186/s40425-016-0189-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 11/07/2016] [Indexed: 01/17/2023] Open
Abstract
Background Although therapeutic cancer vaccines have been mostly disappointing in the clinic, the advent of novel immunotherapies and the future promise of neoantigen-based therapies have created the need for new vaccine modalities that can easily adapt to current and future developments in cancer immunotherapy. One such novel platform is a DNA vaccine fusing the chemokine Macrophage Inflammatory Protein-3α (MIP-3α) to an antigen, here melanoma antigen gp100. Previous published work has indicated that MIP-3α targets nascent peptides to immature dendritic cells, leading to processing by class I and II MHC pathways. This platform has shown enhanced efficacy in prophylactic melanoma and therapeutic lymphoma model systems. Methods The B16F10 melanoma syngeneic mouse model system was utilized, with a standard therapeutic protocol: challenge with lethal dose of B16F10 cells (5 × 104) on day 0 and then vaccinate by intramuscular electroporation with 50 μg plasmid on days three, 10, and 17. Efficacy was assessed by analysis of tumor burden, tumor growth, and mouse survival, using the statistical tests ANOVA, mixed effects regression, and log-rank, respectively. Immunogenicity was assessed by ELISA and flow cytometric methods, including intracellular cytokine staining to assess vaccine-specific T-cell responses, all tested by ANOVA. Results We demonstrate that the addition of MIP3α to gp100 significantly enhances systemic anti-gp100 immunological parameters. Further, chemokine-fusion vaccine therapy significantly reduces tumor burden, slows tumor growth, and enhances mouse overall survival compared to antigen-only, irrelevant-antigen, and mock vaccines, with efficacy mediated by both CD4+ and CD8+ effector T cells. Antigen-only, irrelevant-antigen, and chemokine-fusion vaccines elicit significantly higher and similar CD4+ and CD8+ tumor-infiltrating lymphocyte (TIL) levels compared to mock vaccine. However, vaccine-specific CD8+ TILs are significantly higher in the chemokine-fusion vaccine group, indicating that the critical step induced by the fusion vaccine construct is the enhancement of vaccine-specific T-cell effectors. Conclusions The current study shows that fusion of MIP3α to melanoma antigen gp100 enhances the immunogenicity and efficacy of a DNA vaccine in a therapeutic B16F10 mouse melanoma model. This study analyzes an adaptable and easily produced MIP3α-antigen modular vaccine platform that could lend itself to a variety of functionalities, including combination treatments and neoantigen vaccination in the pursuit of personalized cancer therapy. Electronic supplementary material The online version of this article (doi:10.1186/s40425-016-0189-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- James T Gordy
- The Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205 USA
| | - Kun Luo
- The Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205 USA
| | - Hong Zhang
- The Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205 USA
| | - Arya Biragyn
- Immunoregulation Section, Laboratory of Molecular Biology and Immunology, National Institute on Aging, National Institutes of Health, 251 Bayview, Blvd, Suite 100, Baltimore, MD 21224 USA
| | - Richard B Markham
- The Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205 USA
| |
Collapse
|
16
|
Borggren M, Nielsen J, Karlsson I, Dalgaard TS, Trebbien R, Williams JA, Fomsgaard A. A polyvalent influenza DNA vaccine applied by needle-free intradermal delivery induces cross-reactive humoral and cellular immune responses in pigs. Vaccine 2016; 34:3634-40. [PMID: 27211039 PMCID: PMC4940207 DOI: 10.1016/j.vaccine.2016.05.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/09/2016] [Accepted: 05/12/2016] [Indexed: 11/25/2022]
Abstract
BACKGROUND Pigs are natural hosts for influenza A viruses, and the infection is widely prevalent in swine herds throughout the world. Current commercial influenza vaccines for pigs induce a narrow immune response and are not very effective against antigenically diverse viruses. To control influenza in pigs, the development of more effective swine influenza vaccines inducing broader cross-protective immune responses is needed. Previously, we have shown that a polyvalent influenza DNA vaccine using vectors containing antibiotic resistance genes induced a broadly protective immune response in pigs and ferrets using intradermal injection followed by electroporation. However, this vaccination approach is not practical in large swine herds, and DNA vaccine vectors containing antibiotic resistance genes are undesirable. OBJECTIVES To investigate the immunogenicity of an optimized version of our preceding polyvalent DNA vaccine, characterized by a next-generation expression vector without antibiotic resistance markers and delivered by a convenient needle-free intradermal application approach. METHODS The humoral and cellular immune responses induced by three different doses of the optimized DNA vaccine were evaluated in groups of five to six pigs. The DNA vaccine consisted of six selected influenza genes of pandemic origin, including internally expressed matrix and nucleoprotein and externally expressed hemagglutinin and neuraminidase. RESULTS Needle-free vaccination of growing pigs with the optimized DNA vaccine resulted in specific, dose-dependent immunity down to the lowest dose (200μg DNA/vaccination). Both the antibody-mediated and the recall lymphocyte immune responses demonstrated high reactivity against vaccine-specific strains and cross-reactivity to vaccine-heterologous strains. CONCLUSION The results suggest that polyvalent DNA influenza vaccination may provide a strong tool for broad protection against swine influenza strains threatening animal as well as public health. In addition, the needle-free administration technique used for this DNA vaccine will provide an easy and practical approach for the large-scale vaccination of swine.
Collapse
Affiliation(s)
- Marie Borggren
- Virus Research and Development Laboratory, Department of Microbiological Diagnostics and Virology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark.
| | - Jens Nielsen
- Virus Research and Development Laboratory, Department of Microbiological Diagnostics and Virology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Ingrid Karlsson
- Virus Research and Development Laboratory, Department of Microbiological Diagnostics and Virology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - Tina S Dalgaard
- Immunology and Microbiology Laboratory, Department of Animal Science, Aarhus University, Blichers Alle 20, 8830 Tjele, Denmark
| | - Ramona Trebbien
- National Influenza Center Denmark, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark
| | - James A Williams
- Nature Technology Corporation, 4701 Innovation Dr, Lincoln, NE 68521, USA
| | - Anders Fomsgaard
- Virus Research and Development Laboratory, Department of Microbiological Diagnostics and Virology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen S, Denmark; Infectious Disease Research Unit, Clinical Institute, University of Southern Denmark, Sdr. Boulevard 29, DK-5000 Odense C, Denmark
| |
Collapse
|
17
|
Amante DH, Smith TRF, Mendoza JM, Schultheis K, McCoy JR, Khan AS, Sardesai NY, Broderick KE. Skin Transfection Patterns and Expression Kinetics of Electroporation-Enhanced Plasmid Delivery Using the CELLECTRA-3P, a Portable Next-Generation Dermal Electroporation Device. Hum Gene Ther Methods 2016. [PMID: 26222896 DOI: 10.1089/hgtb.2015.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The CELLECTRA-3P dermal electroporation device (Inovio Pharmaceuticals, Plymouth Meeting, PA) has been evaluated in the clinic and shown to enhance the delivery of an influenza DNA vaccine. To understand the mechanism by which this device aids in enhancing the host immune response to DNA vaccines we investigated the expression kinetics and localization of a reporter plasmid (pGFP) delivered via the CELLECTRA-3P. Histological analysis revealed green fluorescent protein (GFP) expression as early as 1 hr posttreatment in the epidermal and dermal layers, and as early as 2 hr posttreatment in the subdermal layers. Immunofluorescence techniques identified keratinocytes, fibrocytes, dendritic-like cells, adipocytes, and myocytes as the principal cell populations transfected. We proceeded to demonstrate elicitation of robust host immune responses after plasmid DNA (pDNA) vaccination. In guinea pigs equivalent humoral (antibody binding titers) immune responses were observed between protocols using either CELLECTRA-3P or intramuscular electroporation to deliver the DNA vaccine. In nonhuman primates, robust interferon-γ enzyme-linked immunospot and protective levels of hemagglutination inhibition titers after pDNA vaccination were observed in groups treated with the CELLECTRA-3P. In conclusion, these findings may assist in the future to design efficient, tolerable DNA vaccination strategies for the clinic.
Collapse
Affiliation(s)
| | | | | | | | - Jay R McCoy
- Inovio Pharmaceuticals , Plymouth Meeting, Pennsylvania
| | - Amir S Khan
- Inovio Pharmaceuticals , Plymouth Meeting, Pennsylvania
| | | | | |
Collapse
|
18
|
Bagley K, Xu R, Ota-Setlik A, Egan M, Schwartz J, Fouts T. The catalytic A1 domains of cholera toxin and heat-labile enterotoxin are potent DNA adjuvants that evoke mixed Th1/Th17 cellular immune responses. Hum Vaccin Immunother 2016; 11:2228-40. [PMID: 26042527 PMCID: PMC4635876 DOI: 10.1080/21645515.2015.1026498] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
DNA encoded adjuvants are well known for increasing the magnitude of cellular and/or humoral immune responses directed against vaccine antigens. DNA adjuvants can also tune immune responses directed against vaccine antigens to better protect against infection of the target organism. Two potent DNA adjuvants that have unique abilities to tune immune responses are the catalytic A1 domains of Cholera Toxin (CTA1) and Heat-Labile Enterotoxin (LTA1). Here, we have characterized the adjuvant activities of CTA1 and LTA1 using HIV and SIV genes as model antigens. Both of these adjuvants enhanced the magnitude of antigen-specific cellular immune responses on par with those induced by the well-characterized cytokine adjuvants IL-12 and GM-CSF. CTA1 and LTA1 preferentially enhanced cellular responses to the intracellular antigen SIVmac239-gag over those for the secreted HIVBaL-gp120 antigen. IL-12, GM-CSF and electroporation did the opposite suggesting differences in the mechanisms of actions of these diverse adjuvants. Combinations of CTA1 or LTA1 with IL-12 or GM-CSF generated additive and better balanced cellular responses to both of these antigens. Consistent with observations made with the holotoxin and the CTA1-DD adjuvant, CTA1 and LTA1 evoked mixed Th1/Th17 cellular immune responses. Together, these results show that CTA1 and LTA1 are potent DNA vaccine adjuvants that favor the intracellular antigen gag over the secreted antigen gp120 and evoke mixed Th1/Th17 responses against both of these antigens. The results also indicate that achieving a balanced immune response to multiple intracellular and extracellular antigens delivered via DNA vaccination may require combining adjuvants that have different and complementary mechanisms of action.
Collapse
|
19
|
Borggren M, Nielsen J, Bragstad K, Karlsson I, Krog JS, Williams JA, Fomsgaard A. Vector optimization and needle-free intradermal application of a broadly protective polyvalent influenza A DNA vaccine for pigs and humans. Hum Vaccin Immunother 2016; 11:1983-90. [PMID: 25746201 DOI: 10.1080/21645515.2015.1011987] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The threat posed by the 2009 pandemic H1N1 virus emphasized the need for new influenza A virus vaccines inducing a broad cross-protective immune response for use in both humans and pigs. An effective and broad influenza vaccine for pigs would greatly benefit the pork industry and contribute to public health by diminishing the risk of emerging highly pathogenic reassortants. Current inactivated protein vaccines against swine influenza produce only short-lived immunity and have no efficacy against heterologous strains. DNA vaccines are a potential alternative with advantages such as the induction of cellular and humoral immunity, inherent safety and rapid production time. We have previously developed a DNA vaccine encoding selected influenza proteins of pandemic origin and demonstrated broad protective immune responses in ferrets and pigs. In this study, we evaluated our DNA vaccine expressed by next-generation vectors. These new vectors can improve gene expression, but they are also efficiently produced on large scales and comply with regulatory guidelines by avoiding antibiotic resistance genes. In addition, a new needle-free delivery of the vaccine, convenient for mass vaccinations, was compared with intradermal needle injection followed by electroporation. We report that when our DNA vaccine is expressed by the new vectors and delivered to the skin with the needle-free device in the rabbit model, it can elicit an antibody response with the same titers as a conventional vector with intradermal electroporation. The needle-free delivery is already in use for traditional protein vaccines in pigs but should be considered as a practical alternative for the mass administration of broadly protective influenza DNA vaccines.
Collapse
Key Words
- BSA, bovine serum albumin
- DK, Denmark
- DNA vaccine
- DNA, DeoxyriboNucleic Acid
- ELISA, Enzyme Linked Immunosorbent Assay
- EP, electroporation
- FCS, fetal calf serum
- HA, hemagglutinin
- HAI, hemagglutination inhibition assay
- HAU, hemagglutination units
- HI, hemagglutination inhibition
- IDAL®, IntraDermal Application of Liquids®
- IgG, immunoglobulin G
- M, matrix protein
- MDCK cells, Madin-Darby Canine Kidney epithelial cells
- NA, neuraminidase
- NP, nucleoprotein
- NTC8385-VA1
- NTC9385R
- NZW, New Zealand White
- PBS, phosphate buffered saline
- RDE, receptor destroying enzyme
- SEM, standard error mean
- TMB, tetramethylbenzidine
- US, the United States
- WHO, world health organization
- bp, base pair
- i.d., intra-dermal
- influenza
- needle-free
- polyvalent
- tPA, tissue plasminogen activator
Collapse
Affiliation(s)
- Marie Borggren
- a Virus Research and Development Laboratory ; Department of Microbiological Diagnostic and Virology; Statens Serum Institut ; Copenhagen , Denmark
| | | | | | | | | | | | | |
Collapse
|
20
|
McCoy JR, Mendoza JM, Spik KW, Badger C, Gomez AF, Schmaljohn CS, Sardesai NY, Broderick KE. A multi-head intradermal electroporation device allows for tailored and increased dose DNA vaccine delivery to the skin. Hum Vaccin Immunother 2015; 11:746-54. [PMID: 25839221 DOI: 10.4161/21645515.2014.978223] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The identification of an effective and tolerable delivery method is a necessity for the success of DNA vaccines in the clinic. This article describes the development and validation of a multi-headed intradermal electroporation device which would be applicable for delivering multiple DNA vaccine plasmids simultaneously but spatially separated. Reporter gene plasmids expressing green and red fluorescent proteins were used to demonstrate the impact of spatial separation on DNA delivery to increase the number of transfected cells and avoid interference through visible expression patterns. To investigate the impact of plasmid interference on immunogenicity, a disease target was investigated where issues with multi-valent vaccines had been previously described. DNA-based Hantaan and Puumala virus vaccines were delivered separately or as a combination and the effect of multi-valence was determined by appropriate assays. While a negative impact was observed for both antigenic vaccines when delivered together, these effects were mitigated when the vaccine was delivered using the multi-head device. We also demonstrate how the multi-head device facilitates higher dose delivery to the skin resulting in improved immune responses. This new multi-head platform device is an efficient, tolerable and non-invasive method to deliver multiple plasmid DNA constructs simultaneously allowing the tailoring of delivery sites for combination vaccines. Additionally, this device would allow the delivery of multi-plasmid vaccine formulations without risk of impacted immune responses through interference. Such a low-cost, easy to use device platform for the delivery of multi-agent DNA vaccines would have direct applications by the military and healthcare sectors for mass vaccination purposes.
Collapse
Affiliation(s)
- Jay R McCoy
- a Inovio Pharmaceuticals Inc. ; Blue Bell , PA USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Ramirez LA, Arango T, Boyer J. Therapeutic and prophylactic DNA vaccines for HIV-1. Expert Opin Biol Ther 2015; 13:563-73. [PMID: 23477730 DOI: 10.1517/14712598.2013.758709] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION DNA vaccines have moved into clinical trials in several fields and their success will be important for licensure of this vaccine modality. An effective vaccine for HIV-1 remains elusive and the development of one is troubled by safety and efficacy issues. Additionally, the ability for an HIV-1 vaccine to induce both the cellular and humoral arms of the immune system is needed. DNA vaccines not only offer a safe approach for the development of an HIV-1 vaccine but they have also been shown to elicit both arms of the immune system. AREAS COVERED This review explores how DNA vaccine design including the regimen, genetic adjuvants used, targeting, and mode of delivery continues to undergo improvements, thereby providing a potential option for an immunogenic vaccine for HIV-1. EXPERT OPINION Continued improvements in delivery technology, in particular electroporation, and the use of prime-boost vaccine strategies will aid in boosting the immunogenicity of DNA vaccines. Basic immunology research will also help discover new potential adjuvant targets that can be combined with DNA vaccination, such as inhibitors of inhibitory receptors.
Collapse
Affiliation(s)
- Lorenzo Antonio Ramirez
- University of Pennsylvania, Pathology, Stellar Chance Labs, 422 Curie Blvd, Philadelphia, PA 19104, USA.
| | | | | |
Collapse
|
22
|
Grunwald T, Ulbert S. Improvement of DNA vaccination by adjuvants and sophisticated delivery devices: vaccine-platforms for the battle against infectious diseases. Clin Exp Vaccine Res 2015; 4:1-10. [PMID: 25648133 PMCID: PMC4313101 DOI: 10.7774/cevr.2015.4.1.1] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 11/30/2014] [Accepted: 12/31/2014] [Indexed: 01/24/2023] Open
Abstract
Advantages of DNA vaccination against infectious diseases over more classical immunization methods include the possibilities for rapid manufacture, fast adaptation to newly emerging pathogens and high stability at ambient temperatures. In addition, upon DNA immunization the antigen is produced by the cells of the vaccinated individual, which leads to activation of both cellular and humoral immune responses due to antigen presentation via MHC I and MHC II molecules. However, so far DNA vaccines have shown most efficient immunogenicity mainly in small rodent models, whereas in larger animals including humans there is still the need to improve effectiveness. This is mostly due to inefficient delivery of the DNA plasmid into cells and nuclei. Here, we discuss technologies used to overcome this problem, including physical means such as in vivo electroporation and co-administration of adjuvants. Several of these methods have already entered clinical testing in humans.
Collapse
Affiliation(s)
- Thomas Grunwald
- Department of Immunology, Fraunhofer-Institute for Cell Therapy and Immunology, Leipzig, Germany
| | - Sebastian Ulbert
- Department of Immunology, Fraunhofer-Institute for Cell Therapy and Immunology, Leipzig, Germany
| |
Collapse
|
23
|
Hu Y, Xu B, Xu J, Shou D, Liu E, Gao J, Liang W, Huang Y. Microneedle-assisted dendritic cell-targeted nanoparticles for transcutaneous DNA immunization. Polym Chem 2015. [DOI: 10.1039/c4py01394h] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Transcutaneous DNA immunization with microneedle-assisted dendritic cell-targeted nanoparticles is an attractive strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Ying Hu
- Zhejiang Pharmaceutical College
- Ningbo
- China
- College of Pharmaceutical Sciences
- Zhejiang University
| | - Beihua Xu
- Zhejiang Pharmaceutical College
- Ningbo
- China
| | - Jiaojiao Xu
- Zhejiang Pharmaceutical College
- Ningbo
- China
- Department of Medicine
- Wenzhou Medical University
| | - Dan Shou
- Department of Medicine
- Zhejiang Academy of Traditional Chinese Medicine
- Hangzhou
- China
| | - Ergang Liu
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| | - Jianqing Gao
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou
- China
| | - Wenquan Liang
- College of Pharmaceutical Sciences
- Zhejiang University
- Hangzhou
- China
| | - Yongzhuo Huang
- Shanghai Institute of Materia Medica
- Chinese Academy of Sciences
- Shanghai 201203
- China
| |
Collapse
|
24
|
Jiang X, Dalebout TJ, Lukashevich IS, Bredenbeek PJ, Franco D. Molecular and immunological characterization of a DNA-launched yellow fever virus 17D infectious clone. J Gen Virol 2014; 96:804-814. [PMID: 25516543 DOI: 10.1099/jgv.0.000026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Yellow fever virus (YFV)-17D is an empirically developed, highly effective live-attenuated vaccine that has been administered to human beings for almost a century. YFV-17D has stood as a paradigm for a successful viral vaccine, and has been exploited as a potential virus vector for the development of recombinant vaccines against other diseases. In this study, a DNA-launched YFV-17D construct (pBeloBAC-FLYF) was explored as a new modality to the standard vaccine to combine the commendable features of both DNA vaccine and live-attenuated viral vaccine. The DNA-launched YFV-17D construct was characterized extensively both in cell culture and in mice. High titres of YFV-17D were generated upon transfection of the DNA into cells, whereas a mutant with deletion in the capsid-coding region (pBeloBAC-YF/ΔC) was restricted to a single round of infection, with no release of progeny virus. Homologous prime-boost immunization of AAD mice with both pBeloBAC-FLYF and pBeloBAC-YF/ΔC elicited specific dose-dependent cellular immune response against YFV-17D. Vaccination of A129 mice with pBeloBAC-FLYF resulted in the induction of YFV-specific neutralizing antibodies in all vaccinated subjects. These promising results underlined the potential of the DNA-launched YFV both as an alternative to standard YFV-17D vaccination and as a vaccine platform for the development of DNA-based recombinant YFV vaccines.
Collapse
Affiliation(s)
- Xiaohong Jiang
- Department of Medical Microbiology, Center of Infectious Diseases, Leiden University Medical Center, P. O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Tim J Dalebout
- Department of Medical Microbiology, Center of Infectious Diseases, Leiden University Medical Center, P. O. Box 9600, 2300 RC Leiden, The Netherlands
| | - Igor S Lukashevich
- Department of Pharmacology and Toxicology, School of Medicine, Center for Predictive Medicine for Biodefense and Emerging Infectious Diseases, NIH Regional Bio-containment Laboratory, University of Louisville, KY, USA
| | - Peter J Bredenbeek
- Department of Medical Microbiology, Center of Infectious Diseases, Leiden University Medical Center, P. O. Box 9600, 2300 RC Leiden, The Netherlands
| | - David Franco
- Aaron Diamond AIDS Research Center, Rockefeller University, 455 First Avenue, New York, NY 10016, USA
| |
Collapse
|
25
|
Broderick KE, Humeau LM. Electroporation-enhanced delivery of nucleic acid vaccines. Expert Rev Vaccines 2014; 14:195-204. [PMID: 25487734 DOI: 10.1586/14760584.2015.990890] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The naked delivery of nucleic acid vaccines is notoriously inefficient, and an enabling delivery technology is required to direct efficiently these constructs intracellularly. A delivery technology capable of enhancing nucleic acid uptake in both cells in tissues and in culture is electroporation (EP). EP is a physical delivery mechanism that increases the permeability of mammalian cell membranes and allows the trafficking of large macromolecules into the cell. EP has now been used extensively in the clinic and been shown to be an effective method to increase both the uptake of the construct and the breadth and magnitude of the resulting immune responses. Excitingly, 2014 saw the announcement of the first EP-enhanced DNA vaccine Phase II trial demonstrating clinical efficacy. This review seeks to introduce the reader to EP as a technology to enhance the delivery of DNA and RNA vaccines and highlight several published clinical trials using this delivery modality.
Collapse
Affiliation(s)
- Kate E Broderick
- Inovio Pharmaceuticals Inc., 660 West Germantown Pike, Suite 110, Plymouth Meeting, PA 19462, USA
| | | |
Collapse
|
26
|
McCoy JR, Mendoza JM, Spik KW, Badger C, Gomez AF, Schmaljohn CS, Sardesai NY, Broderick KE. A multi-head intradermal electroporation device allows for tailored and increased dose DNA vaccine delivery to the skin. Hum Vaccin Immunother 2014; 10:3039-47. [PMID: 25483486 PMCID: PMC5443063 DOI: 10.4161/hv.29671] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 06/09/2014] [Accepted: 06/20/2014] [Indexed: 11/19/2022] Open
Abstract
The identification of an effective and tolerable delivery method is a necessity for the success of DNA vaccines in the clinic. This manuscript describes the development and validation of a multi-headed intradermal electroporation device which would be applicable for delivering multiple DNA vaccine plasmids simultaneously but spatially separated. Reporter gene plasmids expressing green and red fluorescent proteins were used to demonstrate the impact of spatial separation on DNA delivery to increase the number of transfected cells and avoid interference through visible expression patterns. To investigate the impact of plasmid interference on immunogenicity, a disease target was investigated where issues with multi-valent vaccines had been previously described. DNA-based Hantaan and Puumala virus vaccines were delivered separately or as a combination and the effect of multi-valence was determined by appropriate assays. While a negative impact was observed for both antigenic vaccines when delivered together, these effects were mitigated when the vaccine was delivered using the multi-head device. We also demonstrate how the multi-head device facilitates higher dose delivery to the skin resulting in improved immune responses. This new multi-head platform device is an efficient, tolerable and non-invasive method to deliver multiple plasmid DNA constructs simultaneously allowing the tailoring of delivery sites for combination vaccines. Additionally, this device would allow the delivery of multi-plasmid vaccine formulations without risk of impacted immune responses through interference. Such a low-cost, easy to use device platform for the delivery of multi-agent DNA vaccines would have direct applications by the military and healthcare sectors for mass vaccination purposes.
Collapse
Affiliation(s)
- Jay R McCoy
- Inovio Pharmaceuticals Inc.; Plymouth Meeting, PA USA
| | | | - Kristin W Spik
- United States Army Medical Research Institute of Infectious Diseases; Fort Detrick, MD USA
| | - Catherine Badger
- United States Army Medical Research Institute of Infectious Diseases; Fort Detrick, MD USA
| | - Alan F Gomez
- Inovio Pharmaceuticals Inc.; Plymouth Meeting, PA USA
| | - Connie S Schmaljohn
- United States Army Medical Research Institute of Infectious Diseases; Fort Detrick, MD USA
| | | | | |
Collapse
|
27
|
Klein R, Templeton DM, Schwenk M. Applications of immunochemistry in human health: advances in vaccinology and antibody design (IUPAC Technical Report). PURE APPL CHEM 2014. [DOI: 10.1515/pac-2013-1028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
This report discusses the history and mechanisms of vaccination of humans as well as the engineering of therapeutic antibodies. Deeper understanding of the molecular interactions involved in both acquired and innate immunity is allowing sophistication in design of modified and even synthetic vaccines. Recombinant DNA technologies are facilitating development of DNA-based vaccines, for example, with the recognition that unmethylated CpG sequences in plasmid DNA will target Toll-like receptors on antigen-presenting cells. Formulations of DNA vaccines with increased immunogenicity include engineering into plasmids with “genetic adjuvant” capability, incorporation into polymeric or magnetic nanoparticles, and formulation with cationic polymers and other polymeric and non-polymeric coatings. Newer methods of delivery, such as particle bombardment, DNA tattooing, electroporation, and magnetic delivery, are also improving the effectiveness of DNA vaccines. RNA-based vaccines and reverse vaccinology based on gene sequencing and bioinformatic approaches are also considered. Structural vaccinology is an approach in which the detailed molecular structure of viral epitopes is used to design synthetic antigenic peptides. Virus-like particles are being designed for vaccine deliveries that are based on structures of viral capsid proteins and other synthetic lipopeptide building blocks. A new generation of adjuvants is being developed to further enhance immunogenicity, based on squalene and other oil–water emulsions, saponins, muramyl dipeptide, immunostimulatory oligonucleotides, Toll-like receptor ligands, and lymphotoxins. Finally, current trends in engineering of therapeutic antibodies including improvements of antigen-binding properties, pharmacokinetic and pharmaceutical properties, and reduction of immunogenicity are discussed. Taken together, understanding the chemistry of vaccine design, delivery and immunostimulation, and knowledge of the techniques of antibody design are allowing targeted development for the treatment of chronic disorders characterized by continuing activation of the immune system, such as autoimmune disorders, cancer, or allergies that have long been refractory to conventional approaches.
Collapse
|
28
|
Sha S, Xing XN, Cao YP. Active immunotherapy facilitates Aβ plaque removal following through microglial activation without obvious T cells infiltrating the CNS. J Neuroimmunol 2014; 274:62-70. [DOI: 10.1016/j.jneuroim.2014.06.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 06/13/2014] [Accepted: 06/17/2014] [Indexed: 12/30/2022]
|
29
|
Short noncoding DNA fragments improve the immune potency of electroporation-mediated HBV DNA vaccination. Gene Ther 2014; 21:703-8. [DOI: 10.1038/gt.2014.44] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2013] [Revised: 03/11/2014] [Accepted: 04/01/2014] [Indexed: 12/11/2022]
|
30
|
Felber BK, Valentin A, Rosati M, Bergamaschi C, Pavlakis GN. HIV DNA Vaccine: Stepwise Improvements Make a Difference. Vaccines (Basel) 2014; 2:354-79. [PMID: 26344623 PMCID: PMC4494255 DOI: 10.3390/vaccines2020354] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 04/11/2014] [Accepted: 04/18/2014] [Indexed: 12/15/2022] Open
Abstract
Inefficient DNA delivery methods and low expression of plasmid DNA have been major obstacles for the use of plasmid DNA as vaccine for HIV/AIDS. This review describes successful efforts to improve DNA vaccine methodology over the past ~30 years. DNA vaccination, either alone or in combination with other methods, has the potential to be a rapid, safe, and effective vaccine platform against AIDS. Recent clinical trials suggest the feasibility of its translation to the clinic.
Collapse
Affiliation(s)
- Barbara K Felber
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, P.O. Box B, Frederick, MD 21702, USA.
| | - Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, P.O. Box B, Frederick, MD 21702, USA.
| | - Margherita Rosati
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, P.O. Box B, Frederick, MD 21702, USA.
| | - Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, P.O. Box B, Frederick, MD 21702, USA.
| | - George N Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute at Frederick, P.O. Box B, Frederick, MD 21702, USA.
| |
Collapse
|
31
|
Grunwald T, Tenbusch M, Schulte R, Raue K, Wolf H, Hannaman D, de Swart RL, Überla K, Stahl-Hennig C. Novel vaccine regimen elicits strong airway immune responses and control of respiratory syncytial virus in nonhuman primates. J Virol 2014; 88:3997-4007. [PMID: 24453366 PMCID: PMC3993754 DOI: 10.1128/jvi.02736-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Accepted: 01/11/2014] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Induction of long-lasting immunity against viral respiratory tract infections remains an elusive goal. Using a nonhuman primate model of human respiratory syncytial virus (hRSV) infection, we compared mucosal and systemic immune responses induced by different DNA delivery approaches to a novel parenteral DNA prime-tonsillar adenoviral vector booster immunization regimen. Intramuscular (i.m.) electroporation (EP) of a DNA vaccine encoding the fusion protein of hRSV induced stronger systemic immune responses than intradermal EP, tattoo immunization, and conventional i.m. DNA injection. A single EP i.m., followed by two atraumatic tonsillar immunizations with the adenoviral vector, elicited strong systemic immune responses, an unique persistent CD4(+) and CD8(+) T cell response in the lower respiratory tract and protection from intranasal hRSV challenge. Thus, parenteral DNA priming followed by booster immunization targeted to a mucosal inductive site constitutes an effective vaccine regimen for eliciting protective immune responses at mucosal effector sites. IMPORTANCE The human respiratory syncytial virus (hRSV) is the most common cause of severe respiratory tract disease in infancy and leads to substantial morbidity and morality in the elderly. In this study, we compared the immunogenicity and efficacy of several gene-based immunization protocols in rhesus macaques. Thereby, we found that the combination of an initially parenterally delivered DNA vaccine with a subsequent atraumatic tonsillar adenoviral vector immunization results in a strong systemic immune response accompanied by an exceptional high T-cell response in the mucosa. Strikingly, these animals were protected against a RSV challenge infection controlling the viral replication indicated by a 1,000-fold-lower viral load in the lower respiratory tract. Since mucosal cellular responses of this strength had not been described in earlier RSV vaccine studies, this heterologous DNA prime-tonsillar boost vaccine strategy is very promising and should be pursued for further preclinical and clinical testing.
Collapse
Affiliation(s)
- Thomas Grunwald
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Germany
| | - Matthias Tenbusch
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Germany
| | - Reiner Schulte
- Unit of Infection Models, German Primate Center, Göttingen, Germany
| | - Katharina Raue
- Unit of Infection Models, German Primate Center, Göttingen, Germany
| | - Hans Wolf
- Institute for Medical Microbiology and Hygiene, Regensburg, Germany
| | - Drew Hannaman
- Ichor Medical Systems, Inc., San Diego, California, USA
| | - Rik L. de Swart
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Klaus Überla
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Germany
| | | |
Collapse
|
32
|
Jalah R, Kulkarni V, Patel V, Rosati M, Alicea C, Bear J, Yu L, Guan Y, Shen X, Tomaras GD, LaBranche C, Montefiori DC, Prattipati R, Pinter A, Bess J, Lifson JD, Reed SG, Sardesai NY, Venzon DJ, Valentin A, Pavlakis GN, Felber BK. DNA and protein co-immunization improves the magnitude and longevity of humoral immune responses in macaques. PLoS One 2014; 9:e91550. [PMID: 24626482 PMCID: PMC3953433 DOI: 10.1371/journal.pone.0091550] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 02/11/2014] [Indexed: 11/25/2022] Open
Abstract
We tested the concept of combining DNA with protein to improve anti-HIV Env systemic and mucosal humoral immune responses. Rhesus macaques were vaccinated with DNA, DNA&protein co-immunization or DNA prime followed by protein boost, and the magnitude and mucosal dissemination of the antibody responses were monitored in both plasma and mucosal secretions. We achieved induction of robust humoral responses by optimized DNA vaccination delivered by in vivo electroporation. These responses were greatly increased upon administration of a protein boost. Importantly, a co-immunization regimen of DNA&protein injected in the same muscle at the same time induced the highest systemic binding and neutralizing antibodies to homologous or heterologous Env as well as the highest Env-specific IgG in saliva. Inclusion of protein in the vaccine resulted in more immunized animals with Env-specific IgG in rectal fluids. Inclusion of DNA in the vaccine significantly increased the longevity of systemic humoral immune responses, whereas protein immunization, either as the only vaccine component or as boost after DNA prime, was followed by a great decline of humoral immune responses overtime. We conclude that DNA&protein co-delivery in a simple vaccine regimen combines the strength of each vaccine component, resulting in improved magnitude, extended longevity and increased mucosal dissemination of the induced antibodies in immunized rhesus macaques.
Collapse
Affiliation(s)
- Rashmi Jalah
- Human Retrovirus Pathogenesis Section, National Cancer Institute, Frederick, Maryland, United States of America
| | - Viraj Kulkarni
- Human Retrovirus Pathogenesis Section, National Cancer Institute, Frederick, Maryland, United States of America
| | - Vainav Patel
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Margherita Rosati
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Candido Alicea
- Human Retrovirus Pathogenesis Section, National Cancer Institute, Frederick, Maryland, United States of America
| | - Jenifer Bear
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Lei Yu
- Institute of Human Virology, Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Yongjun Guan
- Institute of Human Virology, Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Xiaoying Shen
- Duke Human Vaccine Institute and Departments of Surgery and Immunology, Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, United States of America
| | - Georgia D. Tomaras
- Duke Human Vaccine Institute and Departments of Surgery and Immunology, Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, United States of America
| | - Celia LaBranche
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - David C. Montefiori
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Rajasekhar Prattipati
- Public Health Research Institute, University of Medicine and Dentistry of New Jersey, Newark, New Jersey, United States of America
| | - Abraham Pinter
- Public Health Research Institute, University of Medicine and Dentistry of New Jersey, Newark, New Jersey, United States of America
| | - Julian Bess
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Steven G. Reed
- Infectious Disease Research Institute, Seattle, Washington, United States of America
| | | | - David J. Venzon
- Biostatistics and Data Management Section, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Antonio Valentin
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - George N. Pavlakis
- Human Retrovirus Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, United States of America
| | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section, National Cancer Institute, Frederick, Maryland, United States of America
| |
Collapse
|
33
|
Luo K, Zhang H, Zavala F, Biragyn A, Espinosa DA, Markham RB. Fusion of antigen to a dendritic cell targeting chemokine combined with adjuvant yields a malaria DNA vaccine with enhanced protective capabilities. PLoS One 2014; 9:e90413. [PMID: 24599116 PMCID: PMC3943962 DOI: 10.1371/journal.pone.0090413] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 01/30/2014] [Indexed: 12/31/2022] Open
Abstract
Although sterilizing immunity to malaria can be elicited by irradiated sporozoite vaccination, no clinically practical subunit vaccine has been shown to be capable of preventing the approximately 600,000 annual deaths attributed to this infection. DNA vaccines offer several potential advantages for a disease that primarily affects the developing world, but new approaches are needed to improve the immunogenicity of these vaccines. By using a novel, lipid-based adjuvant, Vaxfectin, to attract immune cells to the immunization site, in combination with an antigen-chemokine DNA construct designed to target antigen to immature dendritic cells, we elicited a humoral immune response that provided sterilizing immunity to malaria challenge in a mouse model system. The chemokine, MIP3αCCL20, did not significantly enhance the cellular infiltrate or levels of cytokine or chemokine expression at the immunization site but acted with Vaxfectin to reduce liver stage malaria infection by orders of magnitude compared to vaccine constructs lacking the chemokine component. The levels of protection achieved were equivalent to those observed with irradiated sporozoites, a candidate vaccine undergoing development for further large scale clinical trial. Only vaccination with the combined regimen of adjuvant and chemokine provided 80–100% protection against the development of bloodstream infection. Treating the immunization process as requiring the independent steps of 1) attracting antigen-presenting cells to the site of immunization and 2) specifically directing vaccine antigen to the immature dendritic cells that initiate the adaptive immune response may provide a rational strategy for the development of a clinically applicable malaria DNA vaccine.
Collapse
Affiliation(s)
- Kun Luo
- The Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Hong Zhang
- The Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Fidel Zavala
- The Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Arya Biragyn
- Immunoregulation Section, Laboratory of Molecular Biology and Immunology, National Institute on Aging, Baltimore, Maryland, United States of America
| | - Diego A. Espinosa
- The Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Richard B. Markham
- The Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
34
|
van Drunen Littel-van den Hurk S, Hannaman D. Electroporation for DNA immunization: clinical application. Expert Rev Vaccines 2014; 9:503-17. [DOI: 10.1586/erv.10.42] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
35
|
Abstract
DNA vaccines are a next generation branch of vaccines which offer major benefits over their conventional counterparts. However, to be effective in large mammals and humans, an enhancing delivery technology is required. Electroporation is a physical technique which results in improved delivery of large molecules through the cell membrane. In the case of plasmid DNA, electroporation enhances both the uptake and expression of the delivered DNA. The skin is an attractive tissue for DNA vaccination in a clinical setting due to the accessibility of the target, the ease of monitoring, and most importantly the immunocompetent nature of the dermis. Electroporation in the skin has the benefit of being minimally invasive and generally well tolerated. Previous studies have determined that optimized electroporation parameters (such as electrical field intensity, pulse length, pulse width, and plasmid formulation) majorly impact the efficiency of DNA delivery to the skin. We provide an overview of DNA vaccination in skin and muscle. In addition, we detail a protocol for the successful intradermal electroporation of plasmid DNA to guinea pig skin, an excellent dermatological animal model. The work detailed here suggests that the technique is safe and effective and could be highly applicable to a clinical setting.
Collapse
Affiliation(s)
- Kate E Broderick
- Inovio Pharmaceuticals, 1787 Sentry Parkway West, Blue Bell PA, 19422, USA,
| | | | | |
Collapse
|
36
|
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
|
37
|
Chiarella P, Signori E. Intramuscular DNA vaccination protocols mediated by electric fields. Methods Mol Biol 2014; 1121:315-24. [PMID: 24510835 DOI: 10.1007/978-1-4614-9632-8_28] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Vaccination is historically one of the most important methods for preventing infectious diseases in humans and animals. New insights in the biology of the immune system allow a more rational design of vaccines, and new vaccination strategies are emerging. DNA vaccines have been proposed as a promising approach for introducing foreign antigens into the host for inducing protective immunity against infectious and cancer diseases. Nevertheless, because of their poor immunogenicity, plasmid DNA vaccination strategies need further implementations. Recent data suggest electrotransfer as a useful tool to improve DNA-based vaccination protocols, being able to stimulate both the humoral and cellular immune responses. In preclinical trials, gene electrotransfer is successfully used in prime-boost combination protocols and its tolerability and safety has been demonstrated also in Phase I clinical trials. In this chapter, we report a short comment supporting electrotransfer as an effective strategy to improve DNA-based vaccination protocols and describe the vaccination procedures by plasmid DNA in combination with electrotransfer and hyaluronidase pretreatment in use in our laboratory.
Collapse
Affiliation(s)
- Pieranna Chiarella
- Laboratory of Molecular Pathology and Experimental Oncology, CNR-IFT, Rome, Italy
| | | |
Collapse
|
38
|
Electroporation as a method to induce myofiber regeneration and increase the engraftment of myogenic cells in skeletal muscles of primates. J Neuropathol Exp Neurol 2013; 72:723-34. [PMID: 23860026 DOI: 10.1097/nen.0b013e31829bac22] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Engraftment of intramuscularly transplanted myogenic cells in mice can be optimized after induction of massive myofiber damage that triggers myofiber regeneration and recruitment of grafted cells; this generally involves either myotoxin injection or cryodamage. There are no effective methods to produce a similar process in the muscles of large mammals such as primates. In this study, we tested the use of intramuscular electroporation for this purpose in 11 macaques. The test sites were 1 cm of skeletal muscle. Each site was treated with 3 penetrations of a 2-needle electrode with 1 cm spacing, applying 3 pulses of 400 V/cm, for a duration of 5 milliseconds and a delay of 200 milliseconds during each penetration. Transplantation of β-galactosidase-labeled myoblasts was done in electroporated and nonelectroporated sites. Electroporation induced massive myofiber necrosis that was followed by efficient muscle regeneration. Myoblast engraftment was substantially increased in electroporated compared with nonelectroporated sites. This suggests that electroporation may be a useful tool to study muscle regeneration in primates and other large mammals and as a method for increasing the engraftment of myoblasts and other myogenic cells in intramuscular transplantation.
Collapse
|
39
|
Mendoza JM, Amante DH, Kichaev G, Knott CL, Kiosses WB, Smith TRF, Sardesai NY, Broderick KE. Elucidating the Kinetics of Expression and Immune Cell Infiltration Resulting from Plasmid Gene Delivery Enhanced by Surface Dermal Electroporation. Vaccines (Basel) 2013; 1:384-97. [PMID: 26344120 PMCID: PMC4494224 DOI: 10.3390/vaccines1030384] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 08/13/2013] [Accepted: 08/21/2013] [Indexed: 11/16/2022] Open
Abstract
The skin is an attractive tissue for vaccination in a clinical setting due to the accessibility of the target, the ease of monitoring and most importantly the immune competent nature of the dermal tissue. While skin electroporation offers an exciting and novel future methodology for the delivery of DNA vaccines in the clinic, little is known about the actual mechanism of the approach and the elucidation of the resulting immune responses. To further understand the mechanism of this platform, the expression kinetics and localization of a reporter plasmid delivered via a surface dermal electroporation (SEP) device as well as the effect that this treatment would have on the resident immune cells in that tissue was investigated. Initially a time course (day 0 to day 21) of enhanced gene delivery with electroporation (EP) was performed to observe the localization of green fluorescent protein (GFP) expression and the kinetics of its appearance as well as clearance. Using gross imaging, GFP expression was not detected on the surface of the skin until 8 h post treatment. However, histological analysis by fluorescent microscopy revealed GFP positive cells as early as 1 h after plasmid delivery and electroporation. Peak GFP expression was observed at 24 h and the expression was maintained in skin for up to seven days. Using an antibody specific for a keratinocyte cell surface marker, reporter gene positive keratinocytes in the epidermis were identified. H&E staining of treated skin sections demonstrated an influx of monocytes and granulocytes at the EP site starting at 4 h and persisting up to day 14 post treatment. Immunological staining revealed a significant migration of lymphocytic cells to the EP site, congregating around cells expressing the delivered antigen. In conclusion, this study provides insights into the expression kinetics following EP enhanced DNA delivery targeting the dermal space. These findings may have implications in the future to design efficient DNA vaccination strategies for the clinic.
Collapse
Affiliation(s)
- Janess M Mendoza
- Inovio Pharmaceuticals Inc., 1787 Sentry Parkway West, Building 18, Suite 400, Blue Bell, PA 19422, USA
| | - Dinah H Amante
- Inovio Pharmaceuticals Inc., 1787 Sentry Parkway West, Building 18, Suite 400, Blue Bell, PA 19422, USA
| | - Gleb Kichaev
- Inovio Pharmaceuticals Inc., 1787 Sentry Parkway West, Building 18, Suite 400, Blue Bell, PA 19422, USA
| | - Christine L Knott
- Inovio Pharmaceuticals Inc., 1787 Sentry Parkway West, Building 18, Suite 400, Blue Bell, PA 19422, USA
| | - William B Kiosses
- The Scripps Research Institute, Core Microscopy Facility, 10550 North Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Trevor R F Smith
- Inovio Pharmaceuticals Inc., 1787 Sentry Parkway West, Building 18, Suite 400, Blue Bell, PA 19422, USA
| | - Niranjan Y Sardesai
- Inovio Pharmaceuticals Inc., 1787 Sentry Parkway West, Building 18, Suite 400, Blue Bell, PA 19422, USA
| | - Kate E Broderick
- Inovio Pharmaceuticals Inc., 1787 Sentry Parkway West, Building 18, Suite 400, Blue Bell, PA 19422, USA.
| |
Collapse
|
40
|
Lindsay RW, Ouellette I, Arendt HE, Martinez J, DeStefano J, Lopez M, Pavlakis GN, Chiuchiolo MJ, Parks CL, King CR. SIV antigen-specific effects on immune responses induced by vaccination with DNA electroporation and plasmid IL-12. Vaccine 2013; 31:4749-58. [PMID: 23954384 DOI: 10.1016/j.vaccine.2013.08.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 06/28/2013] [Accepted: 08/05/2013] [Indexed: 10/26/2022]
Abstract
Molecular adjuvants are important for augmenting or modulating immune responses induced by DNA vaccination. Promising results have been obtained using IL-12 expression plasmids in a variety of disease models including the SIV model of HIV infection. We used a mouse model to evaluate plasmid IL-12 (pIL-12) in a DNA prime, recombinant adenovirus serotype 5 (rAd5) boost regimen specifically to evaluate the effect of IL-12 expression on cellular and humoral immunity induced against both SIVmac239 Gag and Env antigens. Priming with electroporated (EP) DNA+pIL-12 resulted in a 2-4-fold enhanced frequency of Gag-specific CD4 T cells which was maintained through the end of the study irrespective of the pIL-12 dose, while memory Env-specific CD4+T cells were maintained only at the low dose of pIL-12. There was little positive effect of pIL-12 on the humoral response to Env, and in fact, high dose pIL-12 dramatically reduced SIV Env-specific IgG. Additionally, both doses of pIL-12 diminished the frequency of CD8 T-cells after DNA prime, although a rAd5 boost recovered CD8 responses regardless of the pIL-12 dose. In this prime-boost regimen, we have shown that a high dose pIL-12 can systemically reduce Env-specific humoral responses and CD4T cell frequency, but not Gag-specific CD4+ T cells. These data indicate that it is important to independently characterize individual SIV or HIV antigen immunogenicity in multi-antigenic vaccines as a function of adjuvant dose.
Collapse
Affiliation(s)
- Ross W Lindsay
- International AIDS Vaccine Initiative, 140 58th Street, Brooklyn, NY 11220, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Burgain A, Rochard A, Trollet C, Mazuet C, Popoff MR, Escriou V, Scherman D, Bigey P. DNA electroporation in rabbits as a method for generation of high-titer neutralizing antisera: examples of the botulinum toxins types A, B, and E. Hum Vaccin Immunother 2013; 9:2147-56. [PMID: 23877030 DOI: 10.4161/hv.25192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Raising high titer antibodies in animals is usually performed by protein immunization, which requires the long and sometimes difficult step of production of the recombinant protein. DNA immunization is an alternative to recombinant proteins, only requiring the building of an eukaryotic expression plasmid. Thanks to efficient DNA delivery techniques such as in vivo electroporation, DNA vaccination has proven useful the last few years. In this work, we have shown that it is possible to raise very high antibody titers in rabbit by DNA electroporation of an antigen encoding plasmid in the skeletal muscle with the right set of electrodes and rabbit strain. In a model of botulinum toxins types A and E, the neutralizing titers obtained after three treatments were high enough to fit the European Pharmacopeia, while it did not for type B toxin. Furthermore, the raised antibodies have high avidity and are suitable for in vitro and in vivo immunodetection of proteins.
Collapse
Affiliation(s)
- Aurore Burgain
- Université Paris Descartes; Paris, France; ENSCP Chimie ParisTech; Paris, France; CNRS UMR8151; Paris, France; Inserm U1022; Paris, France
| | - Alice Rochard
- Université Paris Descartes; Paris, France; ENSCP Chimie ParisTech; Paris, France; CNRS UMR8151; Paris, France; Inserm U1022; Paris, France
| | - Capucine Trollet
- Université Paris Descartes; Paris, France; ENSCP Chimie ParisTech; Paris, France; CNRS UMR8151; Paris, France; Inserm U1022; Paris, France
| | | | | | - Virginie Escriou
- Université Paris Descartes; Paris, France; ENSCP Chimie ParisTech; Paris, France; CNRS UMR8151; Paris, France; Inserm U1022; Paris, France
| | - Daniel Scherman
- Université Paris Descartes; Paris, France; ENSCP Chimie ParisTech; Paris, France; CNRS UMR8151; Paris, France; Inserm U1022; Paris, France
| | - Pascal Bigey
- Université Paris Descartes; Paris, France; ENSCP Chimie ParisTech; Paris, France; CNRS UMR8151; Paris, France; Inserm U1022; Paris, France
| |
Collapse
|
42
|
Optimization of HIV-1 Envelope DNA Vaccine Candidates within Three Different Animal Models, Guinea Pigs, Rabbits and Cynomolgus Macaques. Vaccines (Basel) 2013; 1:305-27. [PMID: 26344115 PMCID: PMC4494233 DOI: 10.3390/vaccines1030305] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Revised: 07/05/2013] [Accepted: 07/10/2013] [Indexed: 11/17/2022] Open
Abstract
HIV-1 DNA vaccines have many advantageous features. Evaluation of HIV-1 vaccine candidates often starts in small animal models before macaque and human trials. Here, we selected and optimized DNA vaccine candidates through systematic testing in rabbits for the induction of broadly neutralizing antibodies (bNAb). We compared three different animal models: guinea pigs, rabbits and cynomolgus macaques. Envelope genes from the prototype isolate HIV-1 Bx08 and two elite neutralizers were included. Codon-optimized genes, encoded secreted gp140 or membrane bound gp150, were modified for expression of stabilized soluble trimer gene products, and delivered individually or mixed. Specific IgG after repeated i.d. inoculations with electroporation confirmed in vivo expression and immunogenicity. Evaluations of rabbits and guinea pigs displayed similar results. The superior DNA construct in rabbits was a trivalent mix of non-modified codon-optimized gp140 envelope genes. Despite NAb responses with some potency and breadth in guinea pigs and rabbits, the DNA vaccinated macaques displayed less bNAb activity. It was concluded that a trivalent mix of non-modified gp140 genes from rationally selected clinical isolates was, in this study, the best option to induce high and broad NAb in the rabbit model, but this optimization does not directly translate into similar responses in cynomolgus macaques.
Collapse
|
43
|
Kulkarni V, Rosati M, Bear J, Pilkington GR, Jalah R, Bergamaschi C, Singh AK, Alicea C, Chowdhury B, Zhang GM, Kim EY, Wolinsky SM, Huang W, Guan Y, LaBranche C, Montefiori DC, Broderick KE, Sardesai NY, Valentin A, Felber BK, Pavlakis GN. Comparison of intradermal and intramuscular delivery followed by in vivo electroporation of SIV Env DNA in macaques. Hum Vaccin Immunother 2013; 9:2081-94. [PMID: 23811579 DOI: 10.4161/hv.25473] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A panel of SIVmac251 transmitted Env sequences were tested for expression, function and immunogenicity in mice and macaques. The immunogenicity of a DNA vaccine cocktail expressing SIVmac239 and three transmitted SIVmac251 Env sequences was evaluated upon intradermal or intramuscular injection followed by in vivo electroporation in macaques using sequential vaccination of gp160, gp120 and gp140 expressing DNAs. Both intradermal and intramuscular vaccination regimens using the gp160 expression plasmids induced robust humoral immune responses, which further improved using the gp120 expressing DNAs. The responses showed durability of binding and neutralizing antibody titers and high avidity for>1 y. The intradermal DNA delivery regimen induced higher cross-reactive responses able to neutralize the heterologous tier 1B-like SIVsmE660_CG7V. Analysis of cellular immune responses showed induction of Env-specific memory responses and cytotoxic granzyme B(+) T cells in both vaccine groups, although the magnitude of the responses were ~10x higher in the intramuscular/electroporation group. The cellular responses induced by both regimens were long lasting and could be detected ~1 y after the last vaccination. These data show that both DNA delivery methods are able to induce robust and durable immune responses in macaques.
Collapse
Affiliation(s)
- Viraj Kulkarni
- Human Retrovirus Pathogenesis Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Margherita Rosati
- Human Retrovirus Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Jenifer Bear
- Human Retrovirus Pathogenesis Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Guy R Pilkington
- Human Retrovirus Pathogenesis Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Rashmi Jalah
- Human Retrovirus Pathogenesis Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Ashish K Singh
- Human Retrovirus Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Candido Alicea
- Human Retrovirus Pathogenesis Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Bhabadeb Chowdhury
- Human Retrovirus Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Gen-Mu Zhang
- Human Retrovirus Pathogenesis Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA; Human Retrovirus Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Eun-Young Kim
- Division of Infectious Diseases; The Feinberg School of Medicine; Northwestern University; Chicago, IL USA
| | - Steven M Wolinsky
- Division of Infectious Diseases; The Feinberg School of Medicine; Northwestern University; Chicago, IL USA
| | - Wensheng Huang
- Institute of Human Virology; Department of Microbiology and Immunology; University of Maryland School of Medicine; Baltimore, MD USA
| | - Yongjun Guan
- Institute of Human Virology; Department of Microbiology and Immunology; University of Maryland School of Medicine; Baltimore, MD USA
| | - Celia LaBranche
- Department of Surgery; Laboratory for AIDS Vaccine Research and Development; Duke University Medical Center; Durham, NC USA
| | - David C Montefiori
- Department of Surgery; Laboratory for AIDS Vaccine Research and Development; Duke University Medical Center; Durham, NC USA
| | | | | | - Antonio Valentin
- Human Retrovirus Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - Barbara K Felber
- Human Retrovirus Pathogenesis Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| | - George N Pavlakis
- Human Retrovirus Section; Vaccine Branch; Center for Cancer Research; National Cancer Institute; Frederick, MD USA
| |
Collapse
|
44
|
O'Hagan DT, Ott GS, Nest GV, Rappuoli R, Giudice GD. The history of MF59(®) adjuvant: a phoenix that arose from the ashes. Expert Rev Vaccines 2013; 12:13-30. [PMID: 23256736 DOI: 10.1586/erv.12.140] [Citation(s) in RCA: 237] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The first clinical trial of an MF59(®)-adjuvanted influenza vaccine (Novartis) was conducted 20 years ago in 1992. The product that emerged (Fluad(®), Novartis) was licensed first in Italy in 1997 and is now licensed worldwide in 30 countries. US licensure is expected in the coming years. By contrast, many alternative adjuvanted vaccines have failed to progress. The key decisions that allowed MF59 to succeed in such a challenging environment are highlighted here and the lessons that were learned along the way are discussed. MF59 was connected to vaccines that did not succeed and was perceived as a 'failure' before it was a success. Importantly, it never failed for safety reasons and was always well tolerated. Even when safety issues have emerged for alternative adjuvants, careful analysis of the substantial safety database for MF59 have shown that there are no significant concerns with widespread use, even in more 'sensitive' populations.
Collapse
Affiliation(s)
- Derek T O'Hagan
- Novartis Vaccines and Diagnostics, Cambridge, MA 02139, USA.
| | | | | | | | | |
Collapse
|
45
|
Assessment of delivery parameters with the multi-electrode array for development of a DNA vaccine against Bacillus anthracis. Bioelectrochemistry 2013; 94:1-6. [PMID: 23727769 DOI: 10.1016/j.bioelechem.2013.04.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2012] [Revised: 04/18/2013] [Accepted: 04/19/2013] [Indexed: 11/20/2022]
Abstract
Gene electrotransfer (GET) enhances delivery of DNA vaccines by increasing both gene expression and immune responses. Our lab has developed the multi-electrode array (MEA) for DNA delivery to skin. The MEA was used at constant pulse duration (150 ms) and frequency (6.67 Hz). In this study, delivery parameters including applied voltage (5-45 V), amount of plasmid (100-300 μg), and number of treatments (2-3) were evaluated for delivery of a DNA vaccine. Mice were intradermally injected with plasmid expressing Bacillus anthracis protective antigen with or without GET and αPA serum titers measured. Within this experiment no significant differences were noted in antibody levels from varying dose or treatment number. However, significant differences were measured from applied voltages of 25 and 35 V. These voltages generated antibody levels between 20,000 and 25,000. Serum from animals vaccinated with these conditions also resulted in toxin neutralization in 40-60% of animals. Visual damage was noted at MEA conditions of 40 V. No damage was noted either visually or histologically from conditions of 35 V or below. These results reflect the importance of establishing appropriate electrical parameters and the potential for the MEA in non-invasive DNA vaccination against B. anthracis.
Collapse
|
46
|
Kwak K, Jiang R, Jagu S, Wang JW, Wang C, Christensen ND, Roden RBS. Multivalent human papillomavirus l1 DNA vaccination utilizing electroporation. PLoS One 2013; 8:e60507. [PMID: 23536912 PMCID: PMC3607584 DOI: 10.1371/journal.pone.0060507] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 02/27/2013] [Indexed: 11/19/2022] Open
Abstract
Objectives Naked DNA vaccines can be manufactured simply and are stable at ambient temperature, but require improved delivery technologies to boost immunogenicity. Here we explore in vivo electroporation for multivalent codon-optimized human papillomavirus (HPV) L1 and L2 DNA vaccination. Methods Balb/c mice were vaccinated three times at two week intervals with a fusion protein comprising L2 residues ∼11−88 of 8 different HPV types (11−88×8) or its DNA expression vector, DNA constructs expressing L1 only or L1+L2 of a single HPV type, or as a mixture of several high-risk HPV types and administered utilizing electroporation, i.m. injection or gene gun. Serum was collected two weeks and 3 months after the last vaccination. Sera from immunized mice were tested for in-vitro neutralization titer, and protective efficacy upon passive transfer to naive mice and vaginal HPV challenge. Heterotypic interactions between L1 proteins of HPV6, HPV16 and HPV18 in 293TT cells were tested by co-precipitation using type-specific monoclonal antibodies. Results Electroporation with L2 multimer DNA did not elicit detectable antibody titer, whereas DNA expressing L1 or L1+L2 induced L1-specific, type-restricted neutralizing antibodies, with titers approaching those induced by Gardasil. Co-expression of L2 neither augmented L1-specific responses nor induced L2-specific antibodies. Delivery of HPV L1 DNA via in vivo electroporation produces a stronger antibody response compared to i.m. injection or i.d. ballistic delivery via gene gun. Reduced neutralizing antibody titers were observed for certain types when vaccinating with a mixture of L1 (or L1+L2) vectors of multiple HPV types, likely resulting from heterotypic L1 interactions observed in co-immunoprecipitation studies. High titers were restored by vaccinating with individual constructs at different sites, or partially recovered by co-expression of L2, such that durable protective antibody titers were achieved for each type. Discussion Multivalent vaccination via in vivo electroporation requires spatial separation of individual type L1 DNA vaccines.
Collapse
MESH Headings
- Alphapapillomavirus/classification
- Alphapapillomavirus/genetics
- Alphapapillomavirus/immunology
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Antibody Specificity
- Capsid Proteins/genetics
- Capsid Proteins/immunology
- Cell Line
- Electroporation
- Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18
- Humans
- Mice
- Papillomavirus Infections/prevention & control
- Papillomavirus Vaccines/administration & dosage
- Papillomavirus Vaccines/immunology
- Vaccination
- Vaccines, DNA/administration & dosage
- Vaccines, DNA/immunology
Collapse
Affiliation(s)
- Kihyuck Kwak
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Rosie Jiang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Subhashini Jagu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Joshua W. Wang
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Chenguang Wang
- Department of Biostatistics, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Neil D. Christensen
- Departments of Pathology, Microbiology and Immunology, Penn State University, Hershey, Pennsylvania, United States of America
| | - Richard B. S. Roden
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
47
|
Langer B, Renner M, Scherer J, Schüle S, Cichutek K. Safety assessment of biolistic DNA vaccination. Methods Mol Biol 2013; 940:371-388. [PMID: 23104355 DOI: 10.1007/978-1-62703-110-3_27] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
DNA-based vector systems have been widely studied as new modalities for the prevention and treatment of human diseases. As for all other medicinal products, safety is an important aspect in the evaluation of such products. In this chapter we reflect on the basic safety issues which have been raised with respect to preventive and therapeutic DNA vaccines, including insertional mutagenesis in case of chromosomal integration, possible formation of anti-DNA antibodies, induction of autoimmune responses and/or immunological tolerance. In addition, local reactions at the site of administration and adverse effects resulting from plasmid DNA spread to nontarget tissues are discussed. Most importantly, however, the benefit-risk profile of a medicinal product is crucial for a decision on providing marketing authorization or not. A product has an acceptable benefit-risk profile if the benefits of the product outweigh its risks for the treated patient.
Collapse
Affiliation(s)
- Barbara Langer
- Division of Medical Biotechnology, Paul-Ehrlich-Institut, Langen, Germany
| | | | | | | | | |
Collapse
|
48
|
Daemi A, Bolhassani A, Rafati S, Zahedifard F, Hosseinzadeh S, Doustdari F. Different domains of glycoprotein 96 influence HPV16 E7 DNA vaccine potency via electroporation mediated delivery in tumor mice model. Immunol Lett 2012; 148:117-25. [PMID: 23085605 DOI: 10.1016/j.imlet.2012.10.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 09/20/2012] [Accepted: 10/06/2012] [Indexed: 11/26/2022]
Abstract
DNA vaccines have emerged as a promising approach for generating antigen-specific immunotherapy. However, due to their low immunogenicity, there is a need to enhance DNA-based vaccine potency. Two main strategies to increase DNA-based vaccine potency are the employment of immuno-adjuvants such as heat shock proteins (HSPs) and a method of improving the delivery of naked plasmid DNA by electroporation. In the current study, we evaluated the effects of linkage of human papillomavirus (HPV) type 16 E7 as a model antigen to N-terminal and C-terminal of glycoprotein 96 (NT-/CT-gp96) on the potency of E7-specific immunity generated by DNA vaccines. We found that subcutaneous DNA injection with E7-CT (gp96) followed by electroporation generates the significant E7-specific IFN-γ immune responses as well as the best protective effects in vaccinated mice as compared to E7 or E7-NT (gp96) DNA vaccines. Therefore, our data indicate that subcutaneous administration of E7 DNA linked to CT (gp96) fragment followed by electroporation can significantly enhance the potency of DNA vaccines. Indeed, the structural domains of immuno-chaperones show the potential of generating effective immune responses against different clinical disorders such as cancer. Altogether, our results show that comparable regions of gp96 (N-/C-terminal fragments of gp96) may have qualitatively different immunological effects in vaccine design.
Collapse
Affiliation(s)
- Amin Daemi
- Molecular Immunology and Vaccine Research Lab., Pasteur Institute of Iran, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
49
|
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
|
50
|
Sha S, Xing XN, Guo WS, Li Y, Zong LX, Guo R, Cao YP. In vivo electroporation of a new gene vaccine encoding ten repeats of Aβ3-10 prevents brain Aβ deposition and delays cognitive impairment in young Tg-APPswe/PSEN1dE9 mice. Neurochem Res 2012; 37:1534-44. [PMID: 22437434 DOI: 10.1007/s11064-012-0748-7] [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] [Received: 12/01/2011] [Revised: 02/24/2012] [Accepted: 03/04/2012] [Indexed: 01/01/2023]
Abstract
Active immunization holds great promise for the treatment of Alzheimer's disease but the infiltration of T-lymphocytes and associated meningoencephalitis observed in clinical trials needs to be overcome. To avoid this toxicity, previous studies have used synthetic truncated derivatives of Aβ to promote humoral immunity. In this study, we developed a novel vaccine [p(Aβ3-10)10-MT] that expresses ten repeats of Aβ3-10 with melatonin (MT) as an adjuvant, and administered it intramuscularly in three-month-old Tg-APPswe/PSEN1dE9 (Tg) mice by in vivo electroporation. The p(Aβ3-10)10-MT vaccine induced high titers of anti-Aβ antibodies, which in turn reduced Aβ deposits in the mouse brains and decreased cognitive impairment. Immunoglobulin isotyping revealed a predominantly IgG1 response, indicating a Th2 anti-inflammatory response. Ex vivo cultured splenocytes exhibited a low IFN-γ and high IL-4 response. Immunohistochemical analysis revealed that glial cell activation was also attenuated. These results indicate that p(Aβ3-10)10-MT may potentially be an effective vaccine to reduce accumulated Aβ and attenuate cognitive deficits.
Collapse
Affiliation(s)
- Sha Sha
- Department of Neurology, First Affiliated Hospital of China Medical University, No. 155 Nanjing North Street, Heping District, Shenyang, 110001, Liao Ning Province, China.
| | | | | | | | | | | | | |
Collapse
|