1
|
Wang Y, Zhang X, Chen G, Shao M. Clinical research progress of telomerase targeted cancer immunotherapy: a literature review. Transl Cancer Res 2024; 13:3904-3921. [PMID: 39145070 PMCID: PMC11319969 DOI: 10.21037/tcr-24-196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/04/2024] [Indexed: 08/16/2024]
Abstract
Background and Objective Telomerase is activated or overexpressed in 85-90% of tumors, which maintains the length of telomere and has become an important anti-cancer target. Increasing clinical and preclinical data suggest that telomerase-targeted cancer immunotherapy could achieve effective killing of tumor cells in vivo. This article reviews the research progress of telomerase targeted cancer immunotherapy in clinical and pre-clinical trials, aiming to provide a reference for further clinical research and treatment of cancers. Methods We investigated the research progress of telomerase immunotherapy in the last 20 years from four electronic databases. Key Content and Findings Telomerase-targeted immunotherapies have been developed with the arising of a new era in immuno-oncology, including peptide vaccines, DNA vaccines, dendritic cells (DCs), adoptive cell transfer (ACT) therapies, antibodies, etc. Some of them have been approved for undergoing clinical trials by the Food and Drug Administration (FDA) for the treatment of various cancers, such as pancreatic cancer, non-small cell lung cancer, melanoma, leukaemia. Of all the treatment modalities, vaccines are the primary treatment methods, some of which have been even entered into phase III clinical trials. The main clinical application direction of telomerase vaccine is the combination with other drugs and treatment modalities, including combination with other vaccines targeting human telomerase reverse transcriptase (hTERT), traditional chemotherapy drugs and immunosuppressors. We also summarized the recent findings of immunotherapy targeting hTERT, focusing on various vaccines and the current status of associated clinical trials. We further discussed the advantages, disadvantages and potential developmental directions of various telomerase-targeted immunotherapies. Conclusions Telomerase-targeted cancer immunotherapy has promising prospects in improving patient survival expectancy. This review may provide data support and design ideas for all researchers and pharmaceutical enterprises in this field.
Collapse
Affiliation(s)
- Yu Wang
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Medicine, Huzhou University, Huzhou, China
| | - Xiaoying Zhang
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Medicine, Huzhou University, Huzhou, China
| | - Guangming Chen
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, School of Medicine, Huzhou University, Huzhou, China
| | - Mingzhe Shao
- Department of Vascular Surgery, Multidisciplinary Collaboration Group of Diabetic Foot, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| |
Collapse
|
2
|
Tao F, Ye Q, Chen Y, Luo L, Xu H, Xu J, Feng Z, Wang C, Li T, Wen Y, Hu Y, Dong H, Zhao X, Wu J. Antigen-loaded flagellate bacteria for enhanced adaptive immune response by intradermal injection. J Control Release 2023; 364:562-575. [PMID: 37926245 DOI: 10.1016/j.jconrel.2023.10.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/29/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Since the skin limits the distribution of intradermal vaccines, a large number of dendritic cells in the skin cannot be fully utilized to elicit a more effective immune response. Here, we loaded the antigen to the surface of the flagellate bacteria that was modified by cationic polymer, thus creating antigen-loaded flagellate bacteria (denoted as 'FB-Ag') to overcome the skin barrier and perform the active delivery of antigen in the skin. The FB-Ag showed fast speed (∼0.2 μm s-1) and strong dendritic cell activation capabilities in the skin model in vitro. In vivo, the FB-Ag promoted the spread of antigen in the skin through active movement, increased the contact between Intradermal dendritic cells and antigen, and effectively activated the internal dendritic cells in the skin. In a mouse of pulmonary metastatic melanoma and in mice bearing subcutaneous melanoma tumor, the FB-Ag effectively increased antigen-specific therapeutic efficacy and produced long-lasting immune memory. More importantly, the FB-Ag also enhanced the level of COVID-19 specific antibodies in the serum and the number of memory B cells in the spleen of mice. The movement of antigen-loaded flagellate bacteria to overcome intradermal constraints may enhance the activation of intradermal dendritic cells, providing new ideas for developing intradermal vaccines.
Collapse
Affiliation(s)
- Feng Tao
- Department of Andrology, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210093, China; State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing University, Nanjing 210093, China
| | - Qingsong Ye
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing University, Nanjing 210093, China
| | - Yimiao Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing University, Nanjing 210093, China
| | - Lifeng Luo
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing University, Nanjing 210093, China
| | - Haiheng Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing University, Nanjing 210093, China
| | - Jialong Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing University, Nanjing 210093, China
| | - Zhuo Feng
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing University, Nanjing 210093, China
| | - Chao Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing University, Nanjing 210093, China
| | - Tao Li
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing University, Nanjing 210093, China
| | - Yuxuan Wen
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing University, Nanjing 210093, China
| | - Yiqiao Hu
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing University, Nanjing 210093, China; Jiangsu Provincial Key Laboratory for Nano Technology, Nanjing University, Nanjing 210093, China
| | - Hong Dong
- State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing University, Nanjing 210093, China.
| | - Xiaozhi Zhao
- Department of Andrology, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210093, China.
| | - Jinhui Wu
- Department of Andrology, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing 210093, China; State Key Laboratory of Pharmaceutical Biotechnology, Medical School of Nanjing University, Nanjing University, Nanjing 210093, China; Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, China; Jiangsu Provincial Key Laboratory for Nano Technology, Nanjing University, Nanjing 210093, China.
| |
Collapse
|
3
|
LENG D, YAMADA S, CHIBA Y, YONEYAMA S, SAKAI Y, HIKONO H, MURAKAMI K. Co-administration of a plasmid encoding CD40 or CD63 enhances the immune responses to a DNA vaccine against bovine viral diarrhea virus in mice. J Vet Med Sci 2022; 84:1175-1184. [PMID: 35793950 PMCID: PMC9523294 DOI: 10.1292/jvms.22-0085] [Citation(s) in RCA: 1] [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: 03/01/2022] [Accepted: 06/26/2022] [Indexed: 11/30/2022] Open
Abstract
Bovine viral diarrhea virus (BVDV) causes substantial economic losses in the livestock industry worldwide. Plasmids encoding the BVDV E2 protein are potential DNA vaccines against BVDV, but their immunogenicity has been insufficient. Here, we investigated the adjuvant effect of CD40 and CD63 plasmids on the immune responses to a BVDV E2 DNA vaccine in mice. We constructed pUMVC4a-based plasmids encoding the BVDV E2 protein (pE2), mouse CD40 (pCD40), or mouse CD63 (pCD63). Protein expression by each plasmid was confirmed through Western blot analysis and immunofluorescence staining of cultured cell lines. BALB/c mice were immunized intradermally twice with pE2 in combination with, or without, pCD40 or pCD63, with 3 weeks between the two doses. pE2 with pCD40 induced significantly higher neutralizing antibody titers against BVDV than pE2 alone. pE2 with pCD63 induced significantly higher anti-E2 IgG2a antibody titers than pE2 alone. Furthermore, pE2 with pCD40 or pCD63 induced significantly increased lymphocyte proliferation and interferon (IFN)-γ production in response to BVDV, compared with E2 alone. These results suggest that a plasmid encoding CD40 or CD63 can be used as an adjuvant to enhance immune responses to DNA vaccines against BVDV.
Collapse
Affiliation(s)
- Dongze LENG
- Graduate School of Veterinary Sciences, Iwate University, Iwate, Japan
| | - Shinji YAMADA
- Graduate School of Veterinary Sciences, Iwate University, Iwate, Japan
| | - Yusuke CHIBA
- Graduate School of Veterinary Sciences, Iwate University, Iwate, Japan
| | - Syuji YONEYAMA
- Graduate School of Veterinary Sciences, Iwate University, Iwate, Japan
| | - Yusuke SAKAI
- Graduate School of Veterinary Sciences, Iwate University, Iwate, Japan
| | | | - Kenji MURAKAMI
- Graduate School of Veterinary Sciences, Iwate University, Iwate, Japan
| |
Collapse
|
4
|
Antigen-bearing outer membrane vesicles as tumour vaccines produced in situ by ingested genetically engineered bacteria. Nat Biomed Eng 2022; 6:898-909. [PMID: 35501399 DOI: 10.1038/s41551-022-00886-2] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 03/21/2022] [Indexed: 02/05/2023]
Abstract
The complex gastrointestinal environment and the intestinal epithelial barrier constrain the design and effectiveness of orally administered tumour vaccines. Here we show that outer membrane vesicles (OMVs) fused to a tumour antigen and produced in the intestine by ingested genetically engineered bacteria function as effective tumour vaccines in mice. We modified Escherichia coli to express, under the control of a promoter induced by the monosaccharide arabinose, a specific tumour antigen fused with the protein cytolysin A on the surface of OMVs released by the commensal bacteria. In mice, oral administration of arabinose and the genetically engineered E. coli led to the production of OMVs that crossed the intestinal epithelium into the lamina propria, where they stimulated dendritic cell maturation. In a mouse model of pulmonary metastatic melanoma and in mice bearing subcutaneous colon tumours, the antigen-bearing OMVs inhibited tumour growth and protected the animals against tumour re-challenge. The in situ production of OMVs by genetically modified commensal bacteria for the delivery of stimulatory molecules could be leveraged for the development of other oral vaccines and therapeutics.
Collapse
|
5
|
Yoo TJ. Anti-Inflammatory Gene Therapy Improves Spatial Memory Performance in a Mouse Model of Alzheimer's Disease. J Alzheimers Dis 2021; 85:1001-1008. [PMID: 34897091 PMCID: PMC8925118 DOI: 10.3233/jad-215270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The immune system plays a critical role in neurodegenerative processes involved in Alzheimer’s disease (AD). In this study, a gene-based immunotherapeutic method examined the effects of anti-inflammatory cellular immune response elements (CIREs) in the amyloid-β protein precursor (AβPP) mouse model. Bi-monthly intramuscular administration, beginning at either 4 or 6 months, and examined at 7.5 through 16 months, with plasmids encoding Interleukin (IL)-10, IL-4, TGF-β polynucleotides, or a combination thereof, into AβPP mice improved spatial memory performance. This work demonstrates an efficient gene therapy strategy to downregulate neuroinflammation, and possibly prevent or delay cognitive decline in AD.
Collapse
Affiliation(s)
- Tai June Yoo
- Korea Allergy Clinic, KangNam Gu, Seoul, South Korea.,University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
6
|
Mucciolo G, Roux C, Scagliotti A, Brugiapaglia S, Novelli F, Cappello P. The dark side of immunotherapy: pancreatic cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:491-520. [PMID: 35582441 PMCID: PMC8992483 DOI: 10.20517/cdr.2020.13] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/23/2020] [Accepted: 04/07/2020] [Indexed: 12/13/2022]
Abstract
Since the journal Science deemed cancer immunotherapy as the "breakthrough of the year" in 2014, there has been an explosion of clinical trials involving immunotherapeutic approaches that, in the last decade - thanks also to the renaissance of the immunosurveillance theory (renamed the three Es theory) - have been continuously and successfully developed. In the latest update of the development of the immuno-oncology drug pipeline, published last November by Nature Review Drug Discovery, it was clearly reported that the immunoactive drugs under study almost doubled in just two years. Of the different classes of passive and active immunotherapies, "cell therapy" is the fastest growing. The aim of this review is to discuss the preclinical and clinical studies that have focused on different immuno-oncology approaches applied to pancreatic cancer, which we assign to the "dark side" of immunotherapy, in the sense that it represents one of the solid tumors showing less response to this type of therapeutic strategy.
Collapse
Affiliation(s)
- Gianluca Mucciolo
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza di Torino, Turin 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10126, Italy
- The two authors contributed equally
| | - Cecilia Roux
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza di Torino, Turin 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10126, Italy
- The two authors contributed equally
| | - Alessandro Scagliotti
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza di Torino, Turin 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10126, Italy
| | - Silvia Brugiapaglia
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza di Torino, Turin 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10126, Italy
| | - Francesco Novelli
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza di Torino, Turin 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10126, Italy
- Molecular Biotechnology Center, University of Turin, Turin 10126, Italy
| | - Paola Cappello
- Center for Experimental Research and Medical Studies (CERMS), Città della Salute e della Scienza di Torino, Turin 10126, Italy
- Department of Molecular Biotechnology and Health Sciences, University of Turin, Turin 10126, Italy
- Molecular Biotechnology Center, University of Turin, Turin 10126, Italy
| |
Collapse
|
7
|
Tian Y, Liu Z, Tan H, Hou J, Wen X, Yang F, Cheng W. New Aspects of Ultrasound-Mediated Targeted Delivery and Therapy for Cancer. Int J Nanomedicine 2020; 15:401-418. [PMID: 32021187 PMCID: PMC6982438 DOI: 10.2147/ijn.s201208] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 12/02/2019] [Indexed: 12/11/2022] Open
Abstract
Ultrasound-mediated targeted delivery (UMTD), a novel delivery modality of therapeutic materials based on ultrasound, shows great potential in biomedical applications. By coupling ultrasound contrast agents with therapeutic materials, UMTD combines the advantages of ultrasound imaging and carrier, which benefit deep tissue penetration and high concentration aggregation. In this paper we introduced recent advances in ultrasound contrast agents and applications in tumor therapy. Ultrasound contrast agents were categorized by their functions, mainly including thermosensitive, pH-sensitive and photosensitive ultrasound contrast agents. The various applications of UMTD in tumor treatment were summarized as follows: drug therapy, transfection of anti-oncogene, RNA interference, vaccine immunotherapy, monoclonal antibody immunotherapy, adoptive cellular immunotherapy, cytokine immunotherapy, and so on. In the end, we elaborated on the current challenges and provided perspectives of UMTD for clinical applications.
Collapse
Affiliation(s)
- Yuhang Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin150080, People’s Republic of China
| | - Zhao Liu
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin150080, People’s Republic of China
| | - Haoyan Tan
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin150080, People’s Republic of China
| | - Jiahui Hou
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin150080, People’s Republic of China
| | - Xin Wen
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin150080, People’s Republic of China
| | - Fan Yang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin150080, People’s Republic of China
| | - Wen Cheng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin150080, People’s Republic of China
| |
Collapse
|
8
|
DNA vaccine based on conserved HA-peptides induces strong immune response and rapidly clears influenza virus infection from vaccinated pigs. PLoS One 2019; 14:e0222201. [PMID: 31553755 PMCID: PMC6760788 DOI: 10.1371/journal.pone.0222201] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 08/24/2019] [Indexed: 01/08/2023] Open
Abstract
Swine influenza virus (SIVs) infections cause a significant economic impact to the pork industry. Moreover, pigs may act as mixing vessel favoring genome reassortment of diverse influenza viruses. Such an example is the pandemic H1N1 (pH1N1) virus that appeared in 2009, harboring a combination of gene segments from avian, pig and human lineages, which rapidly reached pandemic proportions. In order to confront and prevent these possible emergences as well as antigenic drift phenomena, vaccination remains of vital importance. The present work aimed to evaluate a new DNA influenza vaccine based on distinct conserved HA-peptides fused with flagellin and applied together with Diluvac Forte as adjuvant using a needle-free device (IntraDermal Application of Liquids, IDAL®). Two experimental pig studies were performed to test DNA-vaccine efficacy against SIVs in pigs. In the first experiment, SIV-seronegative pigs were vaccinated with VC4-flagellin DNA and intranasally challenged with a pH1N1. In the second study, VC4-flagellin DNA vaccine was employed in SIV-seropositive animals and challenged intranasally with an H3N2 SIV-isolate. Both experiments demonstrated a reduction in the viral shedding after challenge, suggesting vaccine efficacy against both the H1 and H3 influenza virus subtypes. In addition, the results proved that maternally derived antibodies (MDA) did not constitute an obstacle to the vaccine approach used. Moreover, elevated titers in antibodies both against H1 and H3 proteins in serum and in bronchoalveolar lavage fluids (BALFs) was detected in the vaccinated animals along with a markedly increased mucosal IgA response. Additionally, vaccinated animals developed stronger neutralizing antibodies in BALFs and higher inhibiting hemagglutination titers in sera against both the pH1N1 and H3N2 influenza viruses compared to unvaccinated, challenged-pigs. It is proposed that the described DNA-vaccine formulation could potentially be used as a multivalent vaccine against SIV infections.
Collapse
|
9
|
Tondini E, Arakelian T, Oosterhuis K, Camps M, van Duikeren S, Han W, Arens R, Zondag G, van Bergen J, Ossendorp F. A poly-neoantigen DNA vaccine synergizes with PD-1 blockade to induce T cell-mediated tumor control. Oncoimmunology 2019; 8:1652539. [PMID: 31646082 PMCID: PMC6791440 DOI: 10.1080/2162402x.2019.1652539] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 07/23/2019] [Accepted: 07/31/2019] [Indexed: 12/20/2022] Open
Abstract
The combination of immune-stimulating strategies has the potency to improve immunotherapy of cancer. Vaccination against neoepitopes derived from patient tumor material can generate tumor-specific T cell immunity, which could reinforce the efficacy of checkpoint inhibitor therapies such as anti-PD-1 treatment. DNA vaccination is a versatile platform that allows the inclusion of multiple neoantigen-coding sequences in a single formulation and therefore represents an ideal platform for neoantigen vaccination. We developed an anti-tumor vaccine based on a synthetic DNA vector designed to contain multiple cancer-specific epitopes in tandem. The DNA vector encoded a fusion gene consisting of three neoepitopes derived from the mouse colorectal tumor MC38 and their natural flanking sequences as 40 amino acid stretches. In addition, we incorporated as reporter epitopes the helper and CTL epitope sequences of ovalbumin. The poly-neoantigen DNA vaccine elicited T cell responses to all three neoantigens and induced functional CD8 and CD4 T cell responses to the reporter antigen ovalbumin after intradermal injection in mice. The DNA vaccine was effective in preventing outgrowth of B16 melanoma expressing ovalbumin in a prophylactic setting. Moreover, the combination of therapeutic DNA vaccination and anti-PD-1 treatment was synergistic in controlling MC38 tumor growth whereas individual treatments did not succeed. These data demonstrate the potential of DNA vaccination to target multiple neoepitopes in a single formulation and highlight the cooperation between vaccine-based and checkpoint blockade immunotherapies for the successful eradication of established tumors.
Collapse
Affiliation(s)
- Elena Tondini
- Dept. Immunohematology and Blood transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Tsolere Arakelian
- Dept. Immunohematology and Blood transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Marcel Camps
- Dept. Immunohematology and Blood transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Suzanne van Duikeren
- Dept. Immunohematology and Blood transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | - Wanda Han
- Immunetune BV, Leiden, The Netherlands
| | - Ramon Arens
- Dept. Immunohematology and Blood transfusion, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Ferry Ossendorp
- Dept. Immunohematology and Blood transfusion, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
10
|
Wallis J, Shenton DP, Carlisle RC. Novel approaches for the design, delivery and administration of vaccine technologies. Clin Exp Immunol 2019; 196:189-204. [PMID: 30963549 PMCID: PMC6468175 DOI: 10.1111/cei.13287] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2019] [Indexed: 12/20/2022] Open
Abstract
It is easy to argue that vaccine development represents humankind's most important and successful endeavour, such is the impact that vaccination has had on human morbidity and mortality over the last 200 years. During this time the original method of Jenner and Pasteur, i.e. that of injecting live-attenuated or inactivated pathogens, has been developed and supplemented with a wide range of alternative approaches which are now in clinical use or under development. These next-generation technologies have been designed to produce a vaccine that has the effectiveness of the original live-attenuated and inactivated vaccines, but without the associated risks and limitations. Indeed, the method of development has undoubtedly moved away from Pasteur's three Is paradigm (isolate, inactivate, inject) towards an approach of rational design, made possible by improved knowledge of the pathogen-host interaction and the mechanisms of the immune system. These novel vaccines have explored methods for targeted delivery of antigenic material, as well as for the control of release profiles, so that dosing regimens can be matched to the time-lines of immune system stimulation and the realities of health-care delivery in dispersed populations. The methods by which vaccines are administered are also the subject of intense research in the hope that needle and syringe dosing, with all its associated issues regarding risk of injury, cross-infection and patient compliance, can be replaced. This review provides a detailed overview of new vaccine vectors as well as information pertaining to the novel delivery platforms under development.
Collapse
Affiliation(s)
- J. Wallis
- Institute of Biomedical EngineeringUniversity of OxfordOxfordUK
| | - D. P. Shenton
- Defence Science and Technology LaboratoryPorton DownUK
| | - R. C. Carlisle
- Institute of Biomedical EngineeringUniversity of OxfordOxfordUK
| |
Collapse
|
11
|
Sisteré-Oró M, Vergara-Alert J, Stratmann T, López-Serrano S, Pina-Pedrero S, Córdoba L, Pérez-Maillo M, Pleguezuelos P, Vidal E, Veljkovic V, Segalés J, Nielsen J, Fomsgaard A, Darji A. Conserved HA-peptide NG34 formulated in pCMV-CTLA4-Ig reduces viral shedding in pigs after a heterosubtypic influenza virus SwH3N2 challenge. PLoS One 2019; 14:e0212431. [PMID: 30822308 PMCID: PMC6396909 DOI: 10.1371/journal.pone.0212431] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 02/01/2019] [Indexed: 11/19/2022] Open
Abstract
Swine influenza viruses (SIVs), the causal agents of swine influenza, are not only important to control due to the economic losses in the swine industry, but also can be pandemic pathogens. Vaccination is one of the most relevant strategies to control and prevent influenza infection. Current human vaccines against influenza induce strain-specific immunity and annual update is required due to the virus antigenic shift phenomena. Previously, our group has reported the use of conserved hemagglutinin peptides (HA-peptides) derived from H1-influenza virus as a potential multivalent vaccine candidate. Immunization of swine with these HA-peptides elicited antibodies that recognized and neutralized heterologous influenza viruses in vitro and demonstrated strong hemagglutination-inhibiting activity. In the present work, we cloned one HA-peptide (named NG34) into a plasmid fused with cytotoxic T lymphocyte-associated antigen (CTLA4) which is a molecule that modifies T cell activation and with an adjuvant activity interfering with the adaptive immune response. The resulting plasmid, named pCMV-CTLA4-Ig-NG34, was administered twice to animals employing a needle-free delivery approach. Two studies were carried out to test the efficacy of pCMV-CTLA4-Ig-NG34 as a potential swine influenza vaccine, one in seronegative and another in seropositive pigs against SIV. The second one was aimed to evaluate whether pCMV-CTLA4-Ig-NG34 vaccination would overcome maternally derived antibodies (MDA). After immunization, all animals were intranasally challenged with an H3N2 influenza strain. A complete elimination or significant reduction in the viral shedding was observed within the first week after the challenge in the vaccinated animals from both studies. In addition, no challenged heterologous virus load was detected in the airways of vaccinated pigs. Overall, it is suggested that the pCMV-CTLA4-Ig-NG34 vaccine formulation could potentially be used as a multivalent vaccine against influenza viruses.
Collapse
Affiliation(s)
- Marta Sisteré-Oró
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Júlia Vergara-Alert
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Thomas Stratmann
- Department of Cell Biology, Physiology and Immunology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - Sergi López-Serrano
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Sonia Pina-Pedrero
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Lorena Córdoba
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Mónica Pérez-Maillo
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Patrícia Pleguezuelos
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Enric Vidal
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Veljko Veljkovic
- Centre for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, Belgrade, Serbia
| | - Joaquim Segalés
- UAB, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, UAB, Bellaterra (Cerdanyola del Vallès), Barcelona, Spain
| | - Jens Nielsen
- Virus Research and Development Laboratory, Department of Virus and Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen S, Denmark
| | - Anders Fomsgaard
- Virus Research and Development Laboratory, Department of Virus and Microbiological Special Diagnostics, Statens Serum Institut, Copenhagen S, Denmark
| | - Ayub Darji
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Campus de la Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- * E-mail:
| |
Collapse
|
12
|
Hegazy-Hassan W, Zepeda-Escobar JA, Ochoa-García L, Contreras-Ortíz JME, Tenorio-Borroto E, Barbabosa-Pliego A, Aparicio-Burgos JE, Oros-Pantoja R, Rivas-Santiago B, Díaz-Albiter H, Garg NJ, Vázquez-Chagoyán JC. TcVac1 vaccine delivery by intradermal electroporation enhances vaccine induced immune protection against Trypanosoma cruzi infection in mice. Vaccine 2018; 37:248-257. [PMID: 30497833 DOI: 10.1016/j.vaccine.2018.11.041] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 11/13/2018] [Accepted: 11/14/2018] [Indexed: 10/27/2022]
Abstract
The efforts for the development and testing of vaccines against Trypanosoma cruzi infection have increased during the past years. We have designed a TcVac series of vaccines composed of T. cruzi derived, GPI-anchored membrane antigens. The TcVac vaccines have been shown to elicit humoral and cellular mediated immune responses and provide significant (but not complete) control of experimental infection in mice and dogs. Herein, we aimed to test two immunization protocols for the delivery of DNA-prime/DNA-boost vaccine (TcVac1) composed of TcG2 and TcG4 antigens in a BALB/c mouse model. Mice were immunized with TcVac1 through intradermal/electroporation (IDE) or intramuscular (IM) routes, challenged with T. cruzi, and evaluated during acute phase of infection. The humoral immune response was evaluated through the assessment of anti-TcG2 and anti-TcG4 IgG subtypes by using an ELISA. Cellular immune response was assessed through a lymphocyte proliferation assay. Finally, clinical and morphopathological aspects were evaluated for all experimental animals. Our results demonstrated that when comparing TcVac1 IDE delivery vs IM delivery, the former induced significantly higher level of antigen-specific antibody response (IgG2a + IgG2b > IgG1) and lymphocyte proliferation, which expanded in response to challenge infection. Histological evaluation after challenge infection showed infiltration of inflammatory cells (macrophages and lymphocytes) in the heart and skeletal tissue of all infected mice. However, the largest increase in inflammatory infiltrate was observed in TcVac1_IDE/Tc mice when compared with TcVac1_IM/Tc or non-vaccinated/infected mice. The extent of tissue inflammatory infiltrate was directly associated with the control of tissue amastigote nests in vaccinated/infected (vs. non-vaccinated/infected) mice. Our results suggest that IDE delivery improves the protective efficacy of TcVac1 vaccine against T. cruzi infection in mice when compared with IM delivery of the vaccine.
Collapse
Affiliation(s)
- Wael Hegazy-Hassan
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Km 15.5 Carretera Panamericana Toluca-Atlacomulco, Toluca, Estado de México C.P. 50200, Mexico
| | - José Antonio Zepeda-Escobar
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Km 15.5 Carretera Panamericana Toluca-Atlacomulco, Toluca, Estado de México C.P. 50200, Mexico
| | - Laucel Ochoa-García
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Km 15.5 Carretera Panamericana Toluca-Atlacomulco, Toluca, Estado de México C.P. 50200, Mexico; Laboratorio Estatal de Salud Pública del Instituto de Salud del Estado de México, Independencia Oriente #1310 Colonia: Reforma y FFCC, CP. 50070 Toluca, Estado de México, Mexico
| | - J M Eloy Contreras-Ortíz
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Km 15.5 Carretera Panamericana Toluca-Atlacomulco, Toluca, Estado de México C.P. 50200, Mexico
| | - Esvieta Tenorio-Borroto
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Km 15.5 Carretera Panamericana Toluca-Atlacomulco, Toluca, Estado de México C.P. 50200, Mexico
| | - Alberto Barbabosa-Pliego
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Km 15.5 Carretera Panamericana Toluca-Atlacomulco, Toluca, Estado de México C.P. 50200, Mexico
| | - José Esteban Aparicio-Burgos
- Universidad Autónoma del Estado de Hidalgo, Escuela Superior de Apan, Carretera Apan-Calpulalpan, Km. 8, Chimalpa Tlalayote S/N, Colonia Chimalpa, Apan, C.P. 43920 Hidalgo, Mexico
| | - Rigoberto Oros-Pantoja
- Facultad de Medicina, Universidad Autónoma del Estado de México, Departamento de Neurociencias, Tollocan esq. Jesus Carranza S/N, Colonia Moderna de la Cruz, C.P. 50180 Estado de México, Toluca, Mexico
| | - Bruno Rivas-Santiago
- Unidad de Investigación Médica Zacatecas-IMSS, Interior de la Alameda, 45, Centro, C.P. 98000 Zacatecas, Mexico
| | - Héctor Díaz-Albiter
- Wellcome Trust Centre for Molecular Parasitology, University of Glasgow, University Place, Glasgow G12 8TA, United Kingdom; El Colegio de la Frontera Sur, Carretera Villahermosa-Reforma Km 15.5, Ranchería Guineo, sección II, CP 86280 Villahermosa, Tabasco, Mexico
| | - Nisha Jain Garg
- Departments of Microbiology & Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1070, United States; Departments of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77555-1070, United States
| | - Juan Carlos Vázquez-Chagoyán
- Centro de Investigación y Estudios Avanzados en Salud Animal, Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Km 15.5 Carretera Panamericana Toluca-Atlacomulco, Toluca, Estado de México C.P. 50200, Mexico.
| |
Collapse
|
13
|
Colluru VT, McNeel DG. B lymphocytes as direct antigen-presenting cells for anti-tumor DNA vaccines. Oncotarget 2018; 7:67901-67918. [PMID: 27661128 PMCID: PMC5356528 DOI: 10.18632/oncotarget.12178] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 09/16/2016] [Indexed: 01/21/2023] Open
Abstract
In spite of remarkable preclinical efficacy, DNA vaccination has demonstrated low immunogenicity in humans. While efforts have focused on increasing cross-presentation of DNA-encoded antigens, efforts to increase DNA vaccine immunogenicity by targeting direct presentation have remained mostly unexplored. In these studies, we compared the ability of different APCs to present antigen to T cells after simple co-culture with plasmid DNA. We found that human primary peripheral B lymphocytes, and not monocytes or in vitro derived dendritic cells (DCs), were able to efficiently encode antigen mRNA and expand cognate tumor antigen-specific CD8 T cells ex vivo. Similarly, murine B lymphocytes co-cultured with plasmid DNA, and not DCs, were able to prime antigen-specific T cells in vivo. Moreover, B lymphocyte-mediated presentation of plasmid antigen led to greater Th1-biased immunity and was sufficient to elicit an anti-tumor effect in vivo. Surprisingly, increasing plasmid presentation by B cells, and not cross presentation of peptides by DCs, further augmented traditional plasmid vaccination. Together, these data suggest that targeting plasmid DNA to B lymphocytes, for example through transfer of ex vivo plasmidloaded B cells, may be novel means to achieve greater T cell immunity from DNA vaccines.
Collapse
Affiliation(s)
- Viswa Teja Colluru
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Douglas G McNeel
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
14
|
Kim SY, Kang D, Choi HJ, Joo Y, Kim JH, Song JJ. Prime-boost immunization by both DNA vaccine and oncolytic adenovirus expressing GM-CSF and shRNA of TGF-β2 induces anti-tumor immune activation. Oncotarget 2017; 8:15858-15877. [PMID: 28178658 PMCID: PMC5362529 DOI: 10.18632/oncotarget.15008] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 12/31/2016] [Indexed: 12/21/2022] Open
Abstract
A successful DNA vaccine for the treatment of tumors should break established immune tolerance to tumor antigen. However, due to the relatively low immunogenicity of DNA vaccines, compared to other kinds of vaccines using live virus or protein, a recombinant viral vector was used to enhance humoral and cellular immunity. In the current study, we sought to develop a novel anti-cancer agent as a complex of DNA and oncolytic adenovirus for the treatment of malignant melanoma in the C57BL/6 mouse model. MART1, a human melanoma-specific tumor antigen, was used to induce an increased immune reaction, since a MART1-protective response is required to overcome immune tolerance to the melanoma antigen MelanA. Because GM-CSF is a potent inducer of anti-tumor immunity and TGF-β2 is involved in tumor survival and host immune suppression, mouse GM-CSF (mGM-CSF) and shRNA of mouse TGF-β2 (shmTGF-β2) genes were delivered together with MART1 via oncolytic adenovirus. MART1 plasmid was also used for antigen-priming. To compare the anti-tumor effect of oncolytic adenovirus expressing both mGM-CSF and shmTGF-β2 (AdGshT) with that of oncolytic adenovirus expressing mGM-CSF only (AdG), each virus was intratumorally injected into melanoma-bearing C57BL/6 mice. As a result, mice that received AdGshT showed delayed tumor growth than those that received AdG. Heterologous prime-boost immunization was combined with oncolytic AdGshT and MART1 expression to result in further delayed tumor growth. This regression is likely due to the following 4 combinations: MART1-derived mouse melanoma antigen-specific immune reaction, immune stimulation by mGM-CSF/shmTGF-β2, tumor growth inhibition by shmTGF-β2, and tumor cell-specific lysis via an oncolytic adenovirus. Immune activation was mainly induced by mature tumor-infiltrating dendritic cell (TIDC) and lowered regulatory T cells in tumor-infiltrating lymphocytes (TIL). Taken together, these findings demonstrate that human MART1 induces a mouse melanoma antigen-specific immune reaction. In addition, the results also indicate that combination therapy of MART1 plasmid, together with an oncolytic adenovirus expressing MART1, mGM-CSF, and shmTGF-β2, is a promising candidate for the treatment of malignant melanoma.
Collapse
Affiliation(s)
- So Young Kim
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea
| | - Dongxu Kang
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea.,Department of Oncology, Affiliated Hospital of Yanbian University, Yanji, Jilin Province, P.R. China
| | - Hye Jin Choi
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Yeonsoo Joo
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Joo-Hang Kim
- CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Jae J Song
- Institute for Cancer Research, Yonsei University College of Medicine, Seoul, Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
15
|
Zhu Z, Yu J, Niu Y, Sun S, Liu Y, Saxon A, Zhang K, Li W. Enhanced Prophylactic and Therapeutic Effects of Polylysine-Modified Ara h 2 DNA Vaccine in a Mouse Model of Peanut Allergy. Int Arch Allergy Immunol 2017; 171:241-250. [PMID: 28049187 DOI: 10.1159/000453264] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 11/08/2016] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The prevalence of food allergy has been increasing, but treatment is very limited. DNA vaccination has been recognized as a promising method for the treatment of allergic diseases; however, poor immunogenicity has hindered its application. METHODS BALB/c mice were intradermally injected with plasmid DNA encoding the peanut protein Ara h 2 (pAra h 2) or pAra h 2 pretreated with poly-L- lysine (PLL) before or after sensitization with Ara h 2 protein. Ara h 2-specific antibodies were measured by ELISA. CD207+ dendritic cells (DCs) and Treg cells in draining lymph nodes were analyzed by flow cytometry after DNA immunization, and cytokine production in splenocytes was also analyzed. RESULTS In the prophylactic study, pretreatment with pAra h 2 or PLL-pAra h 2 resulted in lower levels of Ara h 2-specific IgG1, IgG2a, and IgE after sensitization with Ara h 2 protein, and mice in the PLL-pAra h 2 group had a significantly lower level of antibodies than those in the pAra h 2 group. In the treatment study, intradermal injection with pAra h 2 or PLL-pAra h 2 after Ara h 2 protein sensitization significantly decreased the level of Ara h 2-specific antibodies, and PLL- pAra h 2 had stronger effects than pAra h 2. There were increased numbers of CD207+ DCs and Treg cells in the mice receiving intradermal injection with PLL-pAra h 2, and splenocytes from PLL-pAra h 2-treated mice secreted increased levels of IFN-γ and IL-10. CONCLUSIONS Modification of pAra h 2 with PLL improved its prophylactic and therapeutic effects in peanut-allergic mice.
Collapse
Affiliation(s)
- Zhenlai Zhu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Khademi F, Mostafaie A, Parvaneh S, Gholami Rad F, Mohammadi P, Bahrami G. Construction and characterization of monoclonal antibodies against the extracellular domain of B-lymphocyte antigen CD20 using DNA immunization method. Int Immunopharmacol 2016; 43:23-32. [PMID: 27939822 DOI: 10.1016/j.intimp.2016.11.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 10/28/2016] [Accepted: 11/29/2016] [Indexed: 12/19/2022]
Abstract
To date, several new anti-CD20 monoclonal antibodies (mAbs) have been developed for potential efficacies compared with familiar mAb rituximab. Despite the recent advances in development of anti-CD20 mAbs for the treatment of B cell malignancies, the efforts should be continued to develop novel antibodies with improved properties. However, the development of mAbs against CD20 as a multi-transmembrane protein is challenging due to the difficulty of providing a lipid environment that can maintain native epitopes. To overcome this limitation, we describe a simple and efficient DNA immunization strategy for the construction of a novel anti-CD20 mAb with improved anti-tumour properties. Using a DNA immunization strategy that includes intradermal (i.d.) immunization with naked plasmid DNA encoding the CD20 gene, we generated the hybridoma cell line D4, which secretes functional mAbs against an extracellular epitope of CD20. Immunocytochemistry analysis and a cell-based enzyme-linked immunosorbent assay using a Burkitt's lymphoma cell line showed that D4 mAbs are capable of binding to native extracellular epitopes of CD20. Moreover, the binding specificity of D4 mAbs was determined by western blot analysis. Cell proliferation was examined by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Apoptosis was detected by the annexin V/propidium iodide staining and dye exclusion assay. The results showed that D4 anti-CD20 mAbs produced by DNA immunization exhibit potent growth inhibitory activity and have superior direct B-cell cytotoxicity compared to rituximab. We propose that antibody-induced apoptosis is one of the mechanisms of cell growth inhibition. Taken together, the data reported here open the path to DNA-based immunization for generating pharmacologically active monoclonal antibodies against CD20. In addition, the data support future in vivo animal testing and subsequent procedures to produce a potential therapeutic mAb.
Collapse
Affiliation(s)
- Fatemeh Khademi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali Mostafaie
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Shahram Parvaneh
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farah Gholami Rad
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pantea Mohammadi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Gholamreza Bahrami
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran; School of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
17
|
Son HY, Jeon YH, Chung JK, Kim CW. In vivo monitoring of transfected DNA, gene expression kinetics, and cellular immune responses in mice immunized with a human NIS gene-expressing plasmid. Int J Immunopathol Pharmacol 2016; 29:612-625. [PMID: 27612483 PMCID: PMC5806837 DOI: 10.1177/0394632016659493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 05/30/2016] [Indexed: 11/16/2022] Open
Abstract
In assessing the effectiveness of DNA vaccines, it is important to monitor: (1) the kinetics of target gene expression in vivo; and (2) the movement of cells that become transfected with the plasmid DNA used in the immunization of a subject. In this study, we used, as a visual imaging marker, expression of the transfected human sodium/iodide symporter (hNIS) gene, which enhances intracellular radio-pertechnetate (TcO4-) accumulation. After intradermal (i.d.) and systemic injection of mice with pcDNA-hNIS and radioactive Technetium-99m (Tc-99m), respectively, whole-body images were obtained by nuclear scintigraphy. The migration of mice cells transfected with the hNIS gene was monitored over a 2-week period by gamma-radioactivity counting of isolated cell populations and was demonstrated in peripheral lymphoid tissues, especially in the draining lymph nodes (dLNs). Beginning at 24 h after DNA inoculation and continuing for the 2-week monitoring period, hNIS-expressing cells were observed specifically in the T-cell-rich zones of the paracortical area of the dLNs. Over the same time period, high levels of INF-γ-secreting CD8 T-cells were found in the dLNs of the pcDNA-hNIS immunized mice. Tumor growth was also significantly retarded in the mice that received hNIS DNA immunization followed by inoculation with CT26 colorectal adenocarcinoma cells that had been transfected with the rat NIS gene (rNIS), which is 93% homologous to the hNIS gene. In conclusion, mouse cells transfected with hNIS DNA after i.d. immunization were found to traffic to the dLNs, and hNIS gene expression in these cells continued for at least 2 weeks post immunization. Furthermore, sequential presentation of NIS DNA to T-cells by migratory antigen presenting cells could induce NIS DNA-specific Th1 immune responses and thus retard the growth of NIS-expressing tumors.
Collapse
Affiliation(s)
- Hye-Youn Son
- Department of Pathology, Tumor Immunity Medical Research Center and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yong-Hyun Jeon
- Department of Nuclear Medicine, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - June-Key Chung
- Department of Nuclear Medicine, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Chul-Woo Kim
- Department of Pathology, Tumor Immunity Medical Research Center and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
18
|
Dai G, Rady HF, Huang W, Shellito JE, Mason C, Ramsay AJ. Gene-based neonatal immune priming potentiates a mucosal adenoviral vaccine encoding mycobacterial Ag85B. Vaccine 2016; 34:6267-6275. [PMID: 27823900 DOI: 10.1016/j.vaccine.2016.10.065] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 10/01/2016] [Accepted: 10/24/2016] [Indexed: 12/01/2022]
Abstract
Tuberculosis remains a major public health hazard worldwide, with neonates and young infants potentially more susceptible to infection than adults. BCG, the only vaccine currently available, provides some protection against tuberculous meningitis in children but variable efficacy in adults, and is not safe to use in immune compromised individuals. A safe and effective vaccine that could be given early in life, and that could also potentiate subsequent booster immunization, would represent a significant advance. To test this proposition, we have generated gene-based vaccine vectors expressing Ag85B from Mycobacterium tuberculosis (Mtb) and designed experiments to test their immunogenicity and protective efficacy particularly when given in heterologous prime-boost combination, with the initial DNA vaccine component given soon after birth. Intradermal delivery of DNA vaccines elicited Th1-based immune responses against Ag85B in neonatal mice but did not protect them from subsequent aerosol challenge with virulent Mtb H37Rv. Recombinant adenovirus vectors encoding Ag85B, given via the intranasal route at six weeks of age, generated moderate immune responses and were poorly protective. However, neonatal DNA priming following by mucosal boosting with recombinant adenovirus generated strong immune responses, as evidenced by strong Ag85B-specific CD4+ and CD8+ T cell responses, both in the lung-associated lymph nodes and the spleen, by the quality of these responding cells (assessed by their capacity to secrete multiple antimicrobial factors), and by improved protection, as indicated by reduced bacterial burden in the lungs following pulmonary TB challenge. These results suggest that neonatal immunization with gene-based vaccines may create a favorable immunological environment that potentiates the pulmonary mucosal boosting effects of a subsequent heterologous vector vaccine encoding the same antigen. Our data indicate that immunization early in life with mycobacterial antigens in an appropriate vaccine setting can prime for protective immunity against Mtb.
Collapse
Affiliation(s)
- Guixiang Dai
- Department of Microbiology, Immunology & Parasitology, LSUHSC-New Orleans, LA 70112, USA; The Louisiana Vaccine Center, LSUHSC-New Orleans, LA 70112, USA
| | - Hamada F Rady
- Department of Microbiology, Immunology & Parasitology, LSUHSC-New Orleans, LA 70112, USA; The Louisiana Vaccine Center, LSUHSC-New Orleans, LA 70112, USA
| | - Weitao Huang
- Department of Microbiology, Immunology & Parasitology, LSUHSC-New Orleans, LA 70112, USA; The Louisiana Vaccine Center, LSUHSC-New Orleans, LA 70112, USA
| | - Judd E Shellito
- Internal Medicine, LSUHSC-New Orleans, LA 70112, USA; The Louisiana Vaccine Center, LSUHSC-New Orleans, LA 70112, USA
| | - Carol Mason
- Internal Medicine, LSUHSC-New Orleans, LA 70112, USA
| | - Alistair J Ramsay
- Department of Microbiology, Immunology & Parasitology, LSUHSC-New Orleans, LA 70112, USA; Internal Medicine, LSUHSC-New Orleans, LA 70112, USA; The Louisiana Vaccine Center, LSUHSC-New Orleans, LA 70112, USA.
| |
Collapse
|
19
|
Lee SH, Danishmalik SN, Sin JI. DNA vaccines, electroporation and their applications in cancer treatment. Hum Vaccin Immunother 2016; 11:1889-900. [PMID: 25984993 DOI: 10.1080/21645515.2015.1035502] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Numerous animal studies and recent clinical studies have shown that electroporation-delivered DNA vaccines can elicit robust Ag-specific CTL responses and reduce disease severity. However, cancer antigens are generally poorly immunogenic, requiring special conditions for immune response induction. To date, many different approaches have been used to elicit Ag-specific CTL and anti-neoplastic responses to DNA vaccines against cancer. In vivo electroporation is one example, whereas others include DNA manipulation, xenogeneic antigen use, immune stimulatory molecule and immune response regulator application, DNA prime-boost immunization strategy use and different DNA delivery methods. These strategies likely increase the immunogenicity of cancer DNA vaccines, thereby contributing to cancer eradication. However, cancer cells are heterogeneous and might become CTL-resistant. Thus, understanding the CTL resistance mechanism(s) employed by cancer cells is critical to develop counter-measures for this immune escape. In this review, the use of electroporation as a DNA delivery method, the strategies used to enhance the immune responses, the cancer antigens that have been tested, and the escape mechanism(s) used by tumor cells are discussed, with a focus on the progress of clinical trials using cancer DNA vaccines.
Collapse
Key Words
- AFP, α-fetoprotein
- APCs, antigen presenting cells
- CEA, carcinoembryonic antigen
- CTLA-4, cytotoxic T lymphocyte-associated antigen-4
- DCs, dendritic cells
- DNA vaccine
- EP, electroporation
- GITR, glucocorticoid-induced tumor necrosis factor receptor family-related gene
- HPV, human papillomavirus
- HSP, heat shock protein
- HSV, herpes simplex virus
- ID, intradermal
- IM, intramuscular
- MAGE, melanoma-associated antigen
- MART, melanoma antigen recognized by T cells
- PAP, prostatic acid phosphatase
- PD, programmed death
- PRAME, preferentially expressed antigen in melanoma
- PSA, prostate-specific antigen
- PSMA, prostate-specific membrane antigen
- WT1, Wilm's tumor
- anti-tumor immunity
- cancer
- hTERT, human telomerase reverse transcriptase
- tumor immune evasion
Collapse
Affiliation(s)
- Si-Hyeong Lee
- a BK21 Plus Graduate Program; Department of Microbiology ; School of Medicine; Kangwon National University ; Chuncheon , Gangwon-do , Korea
| | | | | |
Collapse
|
20
|
|
21
|
Kaurav M, Minz S, Sahu K, Kumar M, Madan J, Pandey RS. Nanoparticulate mediated transcutaneous immunization: Myth or reality. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 12:1063-1081. [PMID: 26767517 DOI: 10.1016/j.nano.2015.12.372] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 12/02/2015] [Accepted: 12/17/2015] [Indexed: 10/22/2022]
Abstract
UNLABELLED Transcutaneous immunization (TCI) is a promising route of vaccine delivery through skin due to many well documented advantages. The main obstacle in TCI is the skin's top dead layer i.e. stratum corneum which is difficult to penetrate. Efficiently delivery of antigen to the immune competent cells of epidermis or dermis in TCI might elicit an effective immune response. In this review, skin immunology with a particular focus on potential of immunological active receptors in influencing adaptive immune responses is highlighted. The challenges with TCI and methods to improve it using different adjuvants, chemical and physical approaches, delivery systems, and combination of above methods to further improve immune response following skin application of antigen are elaborately discussed. Nanoparticulate vaccine delivery systems with reference to their applications in TCI are classified according to their chronological development. Conclusively, clinical translations of above methods are also briefly reviewed. FROM THE CLINICAL EDITOR Transcutaneous immunization has been investigated by many as a promising route of vaccination. In this comprehensive review article, the authors described and discussed the existing knowledge and difficulties in this approach. Furthermore, ways of improving transcutaneous delivery were also reviewed.
Collapse
Affiliation(s)
- Monika Kaurav
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India.
| | - Sunita Minz
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India.
| | - Kantrol Sahu
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India.
| | - Manoj Kumar
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India.
| | | | - Ravi Shankar Pandey
- SLT Institute of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya, Bilaspur, India.
| |
Collapse
|
22
|
Abstract
DNA vaccination has emerged as an attractive immunotherapeutic approach against cancer
due to its simplicity, stability, and safety. Results from numerous clinical trials have
demonstrated that DNA vaccines are well tolerated by patients and do not trigger major
adverse effects. DNA vaccines are also very cost effective and can be administered
repeatedly for long-term protection. Despite all the practical advantages, DNA vaccines
face challenges in inducing potent antigen specific cellular immune responses as a result
of immune tolerance against endogenous self-antigens in tumors. Strategies to enhance
immunogenicity of DNA vaccines against self-antigens have been investigated including
encoding of xenogeneic versions of antigens, fusion of antigens to molecules that activate
T cells or trigger associative recognition, priming with DNA vectors followed by boosting
with viral vector, and utilization of immunomodulatory molecules. This review will focus
on discussing strategies that circumvent immune tolerance and provide updates on findings
from recent clinical trials.
Collapse
Key Words
- APCs, antigen presenting cells
- CEA, carcinoembryonic antigen
- CIN, cervical intraepithelial neoplasia
- CT antigens, cancer-testis antigens
- CTLs, cytotoxic lymphocytes
- DNA vaccines
- DOM, fragment c domain
- EP, electroporation
- GITR, glucocorticoid-induced tumor necrosis factor receptor family-related genes
- HER2, Her2/neu
- HSP70, heat shock protein 70
- IFNs, interferons
- IRF, interferon regulatory factor
- Id, idiotype
- MHC, major histocompatibility complex
- Mam-A, Mammaglobin-A
- NHP, non-human primate
- PAP, Prostatic acid phosphatase
- PMED, particle mediated epidermal delivery
- PSMA, prostate-specific membrane antigen
- SCT, single-chain trimer
- STING, stimulator of interferon genes
- TAAs, tumor-associated antigens
- TBK1, Tank-binding kinase 1
- TLRs, Toll-like receptors
- TT, tetanus toxin
- Trp2, tyrosinase related protein 2
- cellular immune response
- hTERT, human telomerase reverse transcriptase
- humoral immune response
- immune tolerance
- phTERT, optimized full-length hTERT
- tumor antigens
- vaccine delivery
Collapse
Affiliation(s)
- Benjamin Yang
- a Department of Pathology ; Johns Hopkins University ; Baltimore , MD USA
| | | | | | | | | |
Collapse
|
23
|
|
24
|
Nezam FS, Hosseini SM, Kheiri MT, Abdoli A, Memarnejadian A, Shenagari M, Gholami S, Sohani H, Rahmatollahi H, Jamali A. Suppressive Effects of Chronic Stress on Influenza Virus Protection after Vaccination with Plasmid DNA-Encoded Nucleoprotein. Neuroimmunomodulation 2015; 22:322-7. [PMID: 25765110 DOI: 10.1159/000371354] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 11/24/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Influenza is a highly infectious and acute respiratory disease caused by an infection of the host respiratory tract mucosa by the influenza virus. The use of DNA vaccines that express conserved genes such as nucleoprotein (NP) represents a new method of vaccination against influenza. In this study, the effect of chronic stress on the efficiency of this type of vaccine has been evaluated in a mouse model. METHODS The NP DNA vaccine was administered intradermally 3 times on days 0, 3 and 6 to stressed and nonstressed male BALB/c mice. Two weeks after the last immunization, half of these mice were challenged with A/Puerto Rico/8/34 (PR8) influenza virus and were weighed for 12 days, and their mortality rate was assessed during this period. The cellular immune response of the other half of the mice was evaluated by cytotoxicity assay. RESULTS The results indicate a significant reduction in the cytotoxic T-lymphocyte response of stressed mice in comparison with unstressed mice. Also, the percentage of weight loss and mortality after the challenge in stressed mice was significantly increased compared to the other group. CONCLUSION These results indicate that the NP DNA vaccine is not able to induce any effective cytotoxic T-lymphocyte response against influenza virus in stressed mice and cannot induce protective immunity against influenza infection in this group of mice.
Collapse
Affiliation(s)
- Fatemeh Sadat Nezam
- Department of Microbiology, Faculty of Biological Sciences, Shahid-Beheshti University, Tehran, Iran
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
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
|
26
|
Endmann A, Oswald D, Riede O, Talman EG, Vos RE, Schroff M, Kleuss C, Ruiters MHJ, Juhls C. Combination of MIDGE-Th1 DNA vaccines with the cationic lipid SAINT-18: studies on formulation, biodistribution and vector clearance. Vaccine 2014; 32:3460-7. [PMID: 24681271 DOI: 10.1016/j.vaccine.2014.03.048] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 03/05/2014] [Accepted: 03/13/2014] [Indexed: 11/29/2022]
Abstract
We have previously shown that the combination of MIDGE-Th1 DNA vectors with the cationic lipid SAINT-18 increases the immune response to the encoded antigen in mice. Here, we report on experiments to further optimize and characterize this approach. We evaluated different formulations of MIDGE-Th1 vectors with SAINT-18 by assessing their influence on the transfection efficiency in cell culture and on the immune response in mice. We found that high amounts of SAINT-18 in formulations with a w/w ratio MIDGE Th1/SAINT-18 of 1:4.8 are beneficial for cell transfection in vitro. In contrast, the formulation of HBsAg-encoding MIDGE-Th1 DNA vectors with the lowest amount of SAINT-18 (w/w ratio MIDGE Th1/SAINT-18 of 1:0.5) resulted in the highest serum IgG1 and IgG2a levels after intradermal immunization of mice. Consequently, latter formulation was selected for a comparative biodistribution study in rats. Following intradermal administration of both naked and formulated MIDGE-Th1 DNA, the vectors localized primarily at the site of injection. Vector DNA levels decreased substantially over the two months duration of the study. When administered in combination with SAINT-18, the vectors were found in significantly higher amounts in draining lymph nodes in comparison to administration of naked MIDGE-Th1 DNA. We propose that the high immune responses induced by MIDGE-Th1/SAINT-18 lipoplexes are mediated by enhanced transfection of cells in vivo, resulting in stronger antigen expression and presentation. Importantly, the combination of MIDGE-Th1 vectors with SAINT-18 was well tolerated in mice and rats and is expected to be safe in human clinical applications.
Collapse
Affiliation(s)
- Anne Endmann
- MOLOGEN AG, Fabeckstraße 30, 14195 Berlin, Germany.
| | | | - Oliver Riede
- MOLOGEN AG, Fabeckstraße 30, 14195 Berlin, Germany
| | - Eduard G Talman
- Synvolux Therapeutics B.V., L.J. Zielstraweg 1, 9713 GX Groningen, The Netherlands
| | - Roelien E Vos
- Synvolux Therapeutics B.V., L.J. Zielstraweg 1, 9713 GX Groningen, The Netherlands
| | | | | | - Marcel H J Ruiters
- Synvolux Therapeutics B.V., L.J. Zielstraweg 1, 9713 GX Groningen, The Netherlands; Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands
| | | |
Collapse
|
27
|
Pearton M, Pirri D, Kang SM, Compans RW, Birchall JC. Host responses in human skin after conventional intradermal injection or microneedle administration of virus-like-particle influenza vaccine. Adv Healthc Mater 2013; 2:1401-10. [PMID: 23564440 DOI: 10.1002/adhm.201300006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2012] [Indexed: 11/06/2022]
Abstract
Miniaturized microneedle devices are being developed for painlessly targeting vaccines to the immune cell populations in skin. As skin immunization studies are generally restricted to animal models however, where skin architecture and immunity is greatly different to human, surprisingly little is known about the local human response to intradermal (ID) vaccines. Here surgically excised human skin is used to explore for the first time the complex molecular and cellular host responses to a candidate influenza vaccine comprising nanoparticulate virus-like-particles (VLPs), administered via conventional hypodermic injection or reduced scale microneedles. Responses at the molecular level are determined by microarray analysis (47,296 discrete transcripts) and validated by quantitative PCR (96 genes). Cellular response is probed through monitoring migration of dendritic cells in viable skin tissue. Gene expression mapping, ontological analysis, and qPCR reveal up-regulation of a host of genes responsible for key immunomodulatory processes and host viral response, including cell recruitment, activation, migration, and T cell interaction following both ID and microneedle injection of VLPs; the response from the microneedles being more subtle. Significant morphological and migratory changes to skin dendritic cells are also apparent following microneedle VLP delivery. This is the first study displaying the global, multifaceted immunological events that occur at the site of vaccine deposition in human skin and will subsequently influence the degree and nature of innate and adaptive immune responses. An increased understanding of the detailed similarities and differences in response against antigen administered via different delivery modalities will inform the development of improved vaccines and vaccine delivery systems.
Collapse
Affiliation(s)
- Marc Pearton
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff, CF10 3NB, UK
| | | | | | | | | |
Collapse
|
28
|
Neuhaus V, Schwarz K, Klee A, Seehase S, Förster C, Pfennig O, Jonigk D, Fieguth HG, Koch W, Warnecke G, Yusibov V, Sewald K, Braun A. Functional testing of an inhalable nanoparticle based influenza vaccine using a human precision cut lung slice technique. PLoS One 2013; 8:e71728. [PMID: 23967238 PMCID: PMC3742667 DOI: 10.1371/journal.pone.0071728] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 07/02/2013] [Indexed: 12/12/2022] Open
Abstract
Annual outbreaks of influenza infections, caused by new influenza virus subtypes and high incidences of zoonosis, make seasonal influenza one of the most unpredictable and serious health threats worldwide. Currently available vaccines, though the main prevention strategy, can neither efficiently be adapted to new circulating virus subtypes nor provide high amounts to meet the global demand fast enough. New influenza vaccines quickly adapted to current virus strains are needed. In the present study we investigated the local toxicity and capacity of a new inhalable influenza vaccine to induce an antigen-specific recall response at the site of virus entry in human precision-cut lung slices (PCLS). This new vaccine combines recombinant H1N1 influenza hemagglutinin (HAC1), produced in tobacco plants, and a silica nanoparticle (NP)-based drug delivery system. We found no local cellular toxicity of the vaccine within applicable concentrations. However higher concentrations of NP (≥103 µg/ml) dose-dependently decreased viability of human PCLS. Furthermore NP, not the protein, provoked a dose-dependent induction of TNF-α and IL-1β, indicating adjuvant properties of silica. In contrast, we found an antigen-specific induction of the T cell proliferation and differentiation cytokine, IL-2, compared to baseline level (152±49 pg/mg vs. 22±5 pg/mg), which could not be seen for the NP alone. Additionally, treatment with 10 µg/ml HAC1 caused a 6-times higher secretion of IFN-γ compared to baseline (602±307 pg/mg vs. 97±51 pg/mg). This antigen-induced IFN-γ secretion was further boosted by the adjuvant effect of silica NP for the formulated vaccine to a 12-fold increase (97±51 pg/mg vs. 1226±535 pg/mg). Thus we were able to show that the plant-produced vaccine induced an adequate innate immune response and re-activated an established antigen-specific T cell response within a non-toxic range in human PCLS at the site of virus entry.
Collapse
Affiliation(s)
- Vanessa Neuhaus
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Katharina Schwarz
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Anna Klee
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Sophie Seehase
- Research Center Borstel, Leibniz Center for Medicine and Biosciences Airway Reserach Center North (ARCN), Borstel, Germany
- German Center for Lung Research (DZL), Hannover, Germany
| | | | - Olaf Pfennig
- KRH Klinikum Oststadt-Heidehaus, Hannover, Germany
| | - Danny Jonigk
- Institute for Pathology, Hanover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | | | - Wolfgang Koch
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Gregor Warnecke
- Division of Cardiac, Thoracic, Transplantation, and Vascular Surgery, Hanover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, Delaware, United States of America
| | - Katherina Sewald
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
| | - Armin Braun
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Institute of Immunology, Hanover Medical School, Hannover, Germany
- Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Centre for Lung Research (DZL), Hannover, Germany
- * E-mail:
| |
Collapse
|
29
|
Han KT, Sin JI. DNA vaccines targeting human papillomavirus-associated diseases: progresses in animal and clinical studies. Clin Exp Vaccine Res 2013; 2:106-14. [PMID: 23858401 PMCID: PMC3710918 DOI: 10.7774/cevr.2013.2.2.106] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 05/30/2013] [Accepted: 06/15/2013] [Indexed: 01/18/2023] Open
Abstract
Human papillomavirus (HPV) infection is a major cause of cervical cancer and its precancerous diseases. Cervical cancer is the second deadliest cancer killer among women worldwide. Moreover, HPV is also known to be a causative agent of oral, pharyngeal, anal and genital cancer. Recent application of HPV structural protein (L1)-targeted prophylactic vaccines (Gardasil® and Cervarix®) is expected to reduce the incidence of HPV infection and cervical cancer, and possibly other HPV-associated cancers. However, the benefit of the prophylactic vaccines for treating HPV-infected patients is unlikely, underscoring the importance of developing therapeutic vaccines against HPV infection. In this regard, numerous types of therapeutic vaccine approaches targeting the HPV regulatory proteins, E6 and E7, have been tested for their efficacy in animals and clinically. In this communication, we review HPV vaccine types, in particular DNA vaccines, their designs and delivery by electroporation and their immunologic and antitumor efficacy in animals and humans, along with the basics of HPV and its pathogenesis.
Collapse
Affiliation(s)
- Kyusun Torque Han
- Department of Microbiology, School of Medicine, Kangwon National University, Chuncheon, Korea
| | | |
Collapse
|
30
|
Koutsonanos DG, Compans RW, Skountzou I. Targeting the skin for microneedle delivery of influenza vaccine. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2013; 785:121-32. [PMID: 23456844 PMCID: PMC6525635 DOI: 10.1007/978-1-4614-6217-0_13] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Influenza infection represents a major socioeconomic burden worldwide. Skin represents a new target that has gained much attention in recent years for delivery of influenza vaccine as an alternative to the conventional intramuscular route of immunization. In this review we describe different microneedle vaccination approaches used in vivo, including metal and dissolving microneedle patches that have demonstrated promising results. Additionally we analyze the immunological basis for microneedle skin immunization and targeting of the skin's dense population of antigen presenting cells, their role, characterization, and function. Additionally we analyze the importance of inflammatory signaling in the skin after microneedle delivery.
Collapse
Affiliation(s)
- Dimitrios G. Koutsonanos
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322
| | - Richard W. Compans
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322
| | - Ioanna Skountzou
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322
| |
Collapse
|
31
|
Joseph SK, Sambanthamoorthy S, Dakshinamoorthy G, Munirathinam G, Ramaswamy K. Protective immune responses to biolistic DNA vaccination of Brugia malayi abundant larval transcript-2. Vaccine 2012; 30:6477-82. [PMID: 22885273 DOI: 10.1016/j.vaccine.2012.07.084] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Revised: 07/17/2012] [Accepted: 07/30/2012] [Indexed: 11/20/2022]
Abstract
Biolistic vaccination using gene gun is developed as a safer tool for delivery of DNA vaccines, a technique that combines high vaccine efficiency with lower antigen dosage and lower cost per vaccine dose. In this study, we compared the protective responses in mice after delivering the Brugia malayi abundant larval transcript-2 (BmALT-2) DNA vaccine using the conventional intradermal approach or with the needleless gene gun delivery approach. BmALT-2 is a leading vaccine candidate against B. malayi, a lymphatic filarial parasite of human. After optimizing the DNA dose and gene gun parameters for delivery into mouse skin, groups of mice were biolistically vaccinated with 5 μg of BmALT-2pVAX. Groups of mice vaccinated intradermally with 5 μg or 100 μg of BmALT-2pVAX was used for comparison of vaccine efficacy. Results demonstrated that gene gun vaccination with 5 μg of BmALT-2pVAX conferred significant protection against challenge infection that was comparable to the degree of protection conferred by intradermal vaccination with 100 μg of BmALT-2pVAX. This observation was further supported by an in vitro antibody dependent cellular cytotoxicity (ADCC) assay. Analysis of the immune response showed that the gene gun vaccination predominantly induced an IgG1 antibody response and significantly high Th2 cytokine response (IL-4) from spleen cells compared to intradermal BmALT-2 DNA delivery that induced predominantly an IgG2a and Th1 cytokine response (IFN-γ, IL-12 and TNF-α). These findings show that host protective responses could be achieved with 20 fold decrease in DNA dose using a gene gun and could prove to be an efficient delivery method in BmALT-2 DNA vaccination against lymphatic filariasis.
Collapse
Affiliation(s)
- S K Joseph
- Department of Biomedical Sciences, University of Illinois College of Medicine at Rockford, Rockford, IL, USA
| | | | | | | | | |
Collapse
|
32
|
Martínez D, Rodríguez N, Griñán T, Rondón T, Vázquez AM, Pérez R, Hernández AM. P3 mAb: An Immunogenic Anti-NeuGcGM3 Antibody with Unusual Immunoregulatory Properties. Front Immunol 2012; 3:94. [PMID: 22566972 PMCID: PMC3342266 DOI: 10.3389/fimmu.2012.00094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 04/11/2012] [Indexed: 11/23/2022] Open
Abstract
P3 is a murine IgM mAb that recognize N-glycosylated gangliosides, sulfatides, and antigens expressed in melanoma, breast, and lung human tumors. This antibody has the ability to trigger an IgG antibody response in the syngeneic BALB/c model, even when it is administered in the absence of adjuvant or carrier protein. The mechanism by which the P3 mAb, a self-immunoglobulin, induce this immune response in the absence of co-stimulatory or classical danger signals is still unknown. In the present paper we show that the high immunogenicity of P3 mAb depends not only on CD4 but also on CD8+ T cells, since the depletion of CD8+ or CD4+ T cells led to the loss of P3 mAb immunogenicity in the syngeneic model. Furthermore, the immunization with P3 mAb enhanced the recovery of the CD8+ T cell population in mice treated with an anti-CD8a antibody. Additionally, the immunization with P3 mAb restored the capacity of immunosuppressed mice to reject allogeneic tumors, a mechanism mediated by the action of CD8+ T cells. Finally, in mice with cyclophosphamide induced lymphopenia, the administration of P3 mAb accelerated the recovery of both CD4+ and CD8+ T cells. These results show new possibilities for B and CD8+ T cells interactions during the immune response elicited by a self-protein. Furthermore they point to P3 mAb as a potential interesting candidate for the treatment of immunosuppressed patients.
Collapse
Affiliation(s)
- Darel Martínez
- Tumor Immunology Direction, Center of Molecular Immunology Havana, Cuba
| | | | | | | | | | | | | |
Collapse
|
33
|
Chen HM, Wang PH, Aravindaram K, Chen YH, Yu HH, Yang WC, Yang NS. Shikonin enhances efficacy of a gene-based cancer vaccine via induction of RANTES. J Biomed Sci 2012; 19:42. [PMID: 22494696 PMCID: PMC3353861 DOI: 10.1186/1423-0127-19-42] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2012] [Accepted: 04/12/2012] [Indexed: 01/01/2023] Open
Abstract
Background Shikonin, a phytochemical purified from Lithospermum erythrorhizon, has been shown to confer diverse pharmacological activities, including accelerating granuloma formation, wound healing, anti-inflammation and others, and is explored for immune-modifier activities for vaccination in this study. Transdermal gene-based vaccine is an attractive approach for delivery of DNA transgenes encoding specific tumor antigens to host skin tissues. Skin dendritic cells (DCs), a potent antigen-presenting cell type, is known to play a critical role in transmitting and orchestrating tumor antigen-specific immunities against cancers. The present study hence employs these various components for experimentation. Method The mRNA and protein expression of RANTES were detected by RT-PCR and ELISA, respectively. The regional expression of RANTES and tissue damage in test skin were evaluated via immunohistochemistry assay. Fluorescein isothiocyanate sensitization assay was performed to trace the trafficking of DCs from the skin vaccination site to draining lymph nodes. Adjuvantic effect of shikonin on gene gun-delivered human gp100 (hgp100) DNA cancer vaccine was studied in a human gp100-transfected B16 (B16/hgp100) tumor model. Results Among various phytochemicals tested, shikonin induced the highest level of expression of RANTES in normal skin tissues. In comparison, mouse RANTES cDNA gene transfection induced a higher level of mRANTES expression for a longer period, but caused more extensive skin damage. Topical application of shikonin onto the immunization site before gene gun-mediated vaccination augmented the population of skin DCs migrating into the draining lymph nodes. A hgp100 cDNA gene vaccination regimen with shikonin pretreatment as an adjuvant in a B16/hgp100 tumor model increased cytotoxic T lymphocyte activities in splenocytes and lymph node cells on target tumor cells. Conclusion Together, our findings suggest that shikonin can effectively enhance anti-tumor potency of a gene-based cancer vaccine via the induction of RANTES expression at the skin immunization site.
Collapse
Affiliation(s)
- Hui-Ming Chen
- Department and Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
34
|
Xu L, Liu Y, Chen Z, Li W, Liu Y, Wang L, Liu Y, Wu X, Ji Y, Zhao Y, Ma L, Shao Y, Chen C. Surface-engineered gold nanorods: promising DNA vaccine adjuvant for HIV-1 treatment. NANO LETTERS 2012; 12:2003-12. [PMID: 22372996 DOI: 10.1021/nl300027p] [Citation(s) in RCA: 207] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
With the intense international response to the AIDS pandemic, HIV vaccines have been extensively investigated but have failed due to issues of safety or efficacy in humans. Adjuvants for HIV/AIDS vaccines are under intense research but a rational design approach is still lacking. Nanomaterials represent an obvious opportunity in this field due to their unique physicochemical properties. Gold nanostructures are being actively studied as a promising and versatile platform for biomedical application. Herein, we report novel surface-engineered gold nanorods (NRs) used as promising DNA vaccine adjuvant for HIV treatment. We have exploited the effects of surface chemistry on the adjuvant activity of the gold nanorod by placing three kinds of molecules, that is, cetyltrimethylammonium bromide (CTAB), poly(diallydimethylammonium chloride) (PDDAC), and polyethyleneimine (PEI) on the surface of the nanorod. These PDDAC- or PEI-modified Au NRs can significantly promote cellular and humoral immunity as well as T cell proliferation through activating antigen-presenting cells if compared to naked HIV-1 Env plasmid DNA treatment in vivo. These findings have shed light on the rational design of low-toxic nanomaterials as a versatile platform for vaccine nanoadjuvants/delivery systems.
Collapse
Affiliation(s)
- Ligeng Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing, 100190, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Elnekave M, Furmanov K, Hovav AH. Intradermal naked plasmid DNA immunization: mechanisms of action. Expert Rev Vaccines 2012; 10:1169-82. [PMID: 21854310 DOI: 10.1586/erv.11.66] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Plasmid DNA is a promising vaccine modality that is regularly examined in prime-boost immunization regimens. Recent advances in skin immunity increased our understanding of the sophisticated cutaneous immune network, which revived scientific interest in delivering vaccines to the skin. Intradermal administration of plasmid DNA via needle injection is a simple and inexpensive procedure that exposes the plasmid and its encoded antigen to the dermal immune surveillance system. This triggers unique mechanisms for eliciting local and systemic immunity that can confer protection against pathogens and tumors. Understanding the mechanisms of intradermal plasmid DNA immunization is essential for enhancing and modulating its immunogenicity. With regard to vaccination, this is of greater importance as this routine injection technique is highly desirable for worldwide immunization. This article will focus on the current understanding of the mechanisms involved in antigen expression and presentation during primary and secondary syringe and needle intradermal plasmid DNA immunization.
Collapse
Affiliation(s)
- Mazal Elnekave
- Institute of Dental Sciences, Hebrew University-Hadassah School of Dental Medicine, PO Box 122722, Jerusalem 91120, Israel
| | | | | |
Collapse
|
36
|
Fissolo N, Montalban X, Comabella M. DNA-based vaccines for multiple sclerosis: Current status and future directions. Clin Immunol 2012; 142:76-83. [DOI: 10.1016/j.clim.2010.11.011] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2010] [Revised: 11/16/2010] [Accepted: 11/17/2010] [Indexed: 01/23/2023]
|
37
|
Verheul RJ, Slütter B, Bal SM, Bouwstra JA, Jiskoot W, Hennink WE. Covalently stabilized trimethyl chitosan-hyaluronic acid nanoparticles for nasal and intradermal vaccination. J Control Release 2011; 156:46-52. [PMID: 21784113 DOI: 10.1016/j.jconrel.2011.07.014] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Revised: 07/06/2011] [Accepted: 07/09/2011] [Indexed: 02/01/2023]
Abstract
The physical stability of polyelectrolyte nanocomplexes composed of trimethyl chitosan (TMC) and hyaluronic acid (HA) is limited in physiological conditions. This may minimize the favorable adjuvant effects associated with particulate systems for nasal and intradermal immunization. Therefore, covalently stabilized nanoparticles loaded with ovalbumin (OVA) were prepared with thiolated TMC and thiolated HA via ionic gelation followed by spontaneous disulfide formation after incubation at pH 7.4 and 37°C. Also, maleimide PEG was coupled to the remaining thiol-moieties on the particles to shield their surface charge. OVA-loaded TMC/HA nanoparticles had a size of around 250-350nm, a positive zeta potential and OVA loading efficiencies up to 60%. Reacting the thiolated particles with maleimide PEG resulted in a slight reduction of zeta potential (from +7 to +4mV) and a minor increase in particle size. Stabilized TMC-S-S-HA particles (PEGylated or not) showed superior stability in saline solutions compared to non-stabilized particles (composed of nonthiolated polymers) but readily disintegrated upon incubation in a saline buffer containing 10mM dithiothreitol. In both the nasal and intradermal immunization study, OVA loaded stabilized TMC-S-S-HA particles demonstrated superior immunogenicity compared to non-stabilized particles (indicated by higher IgG titers). Intranasal, PEGylation completely abolished the beneficial effects of stabilization and it induced no enhanced immune responses against OVA after intradermal administration. In conclusion, stabilization of the TMC/HA particulate system greatly enhances the immunogenicity of OVA in nasal and intradermal vaccination.
Collapse
Affiliation(s)
- Rolf J Verheul
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | | | | | | | | | | |
Collapse
|
38
|
Koutsonanos DG, del Pilar Martin M, Zarnitsyn VG, Jacob J, Prausnitz MR, Compans RW, Skountzou I. Serological memory and long-term protection to novel H1N1 influenza virus after skin vaccination. J Infect Dis 2011; 204:582-91. [PMID: 21685355 DOI: 10.1093/infdis/jir094] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND A major goal in influenza vaccine development is induction of serological memory and cellular responses to confer long-term protection and limit virus spread after infection. Here, we investigate induction of long-lived immunity against the 2009 H1N1 virus after skin vaccination. METHODS BALB/c mice received a single dose of 5 μg inactivated A/California/04/09 virus via coated metal microneedles (MN) applied to skin or via subcutaneous injection. RESULTS MN or subcutaneous vaccination elicited similar serum IgG and hemagglutination inhibition titers and 100% protection against lethal viral challenge 6 weeks after vaccination. Six months after vaccination, the subcutaneous group exhibited a 60% decrease in functional antibody titers and extensive lung inflammation after challenge with 10 × LD(50) of homologous virus. In contrast, the MN group maintained high functional antibody titers and IFN-γ levels, inhibition of viral replication, and no signs of lung inflammation after challenge. MN vaccination conferred complete protection against lethal challenge, whereas subcutaneous vaccination induced only partial protection. These findings were further supported by high numbers of bone marrow plasma cells and spleen antibody-secreting cells detected in the MN group. CONCLUSIONS A single skin vaccination with MN induced potent long-lived immunity and improved protection against the 2009 H1N1 influenza virus, compared with subcutaneous injection.
Collapse
Affiliation(s)
- Dimitrios G Koutsonanos
- Department of Microbiology & Immunology and Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Lian B, Cheng A, Wang M, Zhu D, Luo Q, Jia R, Liu F, Han X, Chen X. Induction of immune responses in ducks with a DNA vaccine encoding duck plague virus glycoprotein C. Virol J 2011; 8:214. [PMID: 21569289 PMCID: PMC3115884 DOI: 10.1186/1743-422x-8-214] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2010] [Accepted: 05/10/2011] [Indexed: 02/07/2023] Open
Abstract
Background A DNA vaccine expressing glycoprotein C (gC) of duck plague virus (DPV) was evaluated for inducing immunity in ducks. The plasmid encoding gC of DPV was administered via intramuscular (IM) injection and gene gun bombardment. Results After immunization by both routes virus-specific serum antibody and T-cell responses developed. Vaccination of ducks by IM injection induced a stronger humoral, but weaker cell-mediated immune response. In contrast, a better cell-mediated immune response was achieved by using a gene gun to deliver DNA-coated gold beads to the epidermis with as little as 6 μg of DNA. Conclusions This demonstrated that both routes of DNA inoculation can be used for eliciting virus-specific immune responses. Although DNA vaccine containing DPV gC is effective in both intramuscular injection and gene gun bombardment, the latter could induce significantly higher cell-mediated responses against DPV.
Collapse
Affiliation(s)
- Bei Lian
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu city, Sichuan, 611130, China
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kunicher N, Tzur T, Amar D, Chaouat M, Yaacov B, Panet A. Characterization of factors that determine lentiviral vector tropism in skin tissue using an ex vivo model. J Gene Med 2011; 13:209-20. [DOI: 10.1002/jgm.1554] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
41
|
Luo M, Qu X, Pan R, Zhu D, Zhang Y, Wu J, Pan Z. The virus-induced signaling adaptor molecule enhances DNA-raised immune protection against H5N1 influenza virus infection in mice. Vaccine 2011; 29:2561-7. [PMID: 21303708 DOI: 10.1016/j.vaccine.2011.01.060] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2010] [Revised: 01/04/2011] [Accepted: 01/21/2011] [Indexed: 11/30/2022]
Abstract
As an adaptor molecule in the retinoic acid-inducible gene-I (RIG-I) signaling pathway, the virus-induced signaling adaptor (VISA) molecule activates NF-κB and IRF3 and thereby leads to the production of type I interferons (IFNs). To explore the potential of VISA as a genetic adjuvant for DNA vaccines, a eukaryotic expression plasmid, pVISA, was generated by cloning the VISA gene into the pVAX1vector. For comparison, the pTRIF plasmid was similarly constructed, encoding the known genetic adjuvant TRIF (TIR-domain-containing adapter-inducing interferon-β), an adapter in the Toll-like receptor (TLR) signaling pathway. Mice were immunized with the chimeric DNA vaccine pHA/NP(147-155), which encodes the HA (hemagglutinin) fused with NP (nucleoprotein) CTL epitope (NP(147-155)) of H5N1 influenza virus, either alone or in combination with pVISA or pTRIF. Antigen-specific immune responses were examined in immunized mice. Our results demonstrate that co-immunization of the pHA/NP(147-155) plasmid with the VISA adjuvant augmented DNA-raised cellular immune responses and provided protection against H5N1 influenza virus challenge in mice. In addition, our data suggest that VISA acts as a stronger adjuvant for DNA immunization than TRIF. We conclude that co-inoculation with a vector expressing the adaptor molecule VISA enhanced the protective immunity against H5N1 infection induced by pHA/NP(147-155) and that VISA could be developed as a novel genetic adjuvant for DNA vaccines.
Collapse
Affiliation(s)
- Mengcheng Luo
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Hubei Province, Wuhan 430072, PR China
| | | | | | | | | | | | | |
Collapse
|
42
|
Electroporation-Mediated DNA Vaccination. CLINICAL ASPECTS OF ELECTROPORATION 2011. [PMCID: PMC7122510 DOI: 10.1007/978-1-4419-8363-3_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
43
|
Microneedle-based transcutaneous immunisation in mice with N-trimethyl chitosan adjuvanted diphtheria toxoid formulations. Pharm Res 2010; 27:1837-47. [PMID: 20559701 PMCID: PMC2920068 DOI: 10.1007/s11095-010-0182-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Accepted: 05/28/2010] [Indexed: 01/15/2023]
Abstract
Purpose The purpose of this study was to gain insight into the delivery and immunogenicity of N-trimethyl chitosan (TMC) adjuvanted diphtheria toxoid (DT) formulations applied transcutaneously with microneedles. Methods Mice were vaccinated with DT-loaded TMC nanoparticles, a solution of TMC and DT (TMC/DT) or DT alone. The formulations were applied onto the skin before or after microneedle treatment with two different 300-µm-long microneedle arrays and also injected intradermally (ID). As a positive control, alum-adjuvanted DT (DT-alum) was injected subcutaneously (SC). Ex vivo confocal microscopy studies were performed with rhodamine-labelled TMC. Results Independent of the microneedle array used and the sequence of microneedle treatment and vaccine application, transcutaneous immunisation with the TMC/DT mixture elicited 8-fold higher IgG titres compared to the TMC nanoparticles or DT solution. The toxin-neutralising antibody titres from this group were similar to those elicited by SC DT-alum. After ID immunisation, both TMC-containing formulations induced enhanced titres compared to a DT solution. Confocal microscopy studies revealed that transport of the TMC nanoparticles across the microneedle conduits was limited compared to a TMC solution. Conclusions In conclusion, TMC has an adjuvant function in transcutaneous immunisation with microneedles, but only if applied in a solution. Electronic Supplementary Material The online version of this article (doi:10.1007/s11095-010-0182-y) contains supplementary material, which is available to authorized users.
Collapse
|
44
|
Induction of Specific CD8 T Cells against Intracellular Bacteria by CD8 T-Cell-Oriented Immunization Approaches. J Biomed Biotechnol 2010; 2010:764542. [PMID: 20508818 PMCID: PMC2875770 DOI: 10.1155/2010/764542] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 02/26/2010] [Accepted: 02/27/2010] [Indexed: 01/08/2023] Open
Abstract
For protection against intracellular bacteria such as Mycobacterium tuberculosis and Listeria monocytogenes, the cellular arm of adaptive immunity is necessary. A variety of immunization methods have been evaluated and are reported to induce specific CD8+ T cells against intracellular bacterial infection. Modified BCG vaccines have been examined to enhance CD8+ T-cell responses. Naked DNA vaccination is a promising strategy to induce CD8+ T cells. In addition to this strategy, live attenuated intracellular bacteria such as Shigella, Salmonella, and Listeria have been utilized as carriers of DNA vaccines in animal models. Vaccination with dendritic cells pulsed with antigenic peptides or the cells introduced antigen genes by virus vectors such as retroviruses is also a powerful strategy. Furthermore, vaccination with recombinant lentivirus has been attempted to induce specific CD8+ T cells. Combinations of these strategies (prime-boost immunization) have been studied for the efficient induction of intracellular bacteria-specific CD8+ T cells.
Collapse
|
45
|
Keeping the memory of influenza viruses. ACTA ACUST UNITED AC 2010; 58:e79-86. [DOI: 10.1016/j.patbio.2010.01.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2010] [Accepted: 01/26/2010] [Indexed: 01/08/2023]
|
46
|
DNA vaccines: developing new strategies against cancer. J Biomed Biotechnol 2010; 2010:174378. [PMID: 20368780 PMCID: PMC2846346 DOI: 10.1155/2010/174378] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2009] [Accepted: 02/05/2010] [Indexed: 12/14/2022] Open
Abstract
Due to their rapid and widespread development, DNA vaccines have entered into a variety of human clinical trials for vaccines against various diseases including cancer. Evidence that DNA vaccines are well tolerated and have an excellent safety profile proved to be of advantage as many clinical trials combines the first phase with the second, saving both time and money. It is clear from the results obtained in clinical trials that such DNA vaccines require much improvement in antigen expression and delivery methods to make them sufficiently effective in the clinic. Similarly, it is clear that additional strategies are required to activate effective immunity against poorly immunogenic tumor antigens. Engineering vaccine design for manipulating antigen presentation and processing pathways is one of the most important aspects that can be easily handled in the DNA vaccine technology. Several approaches have been investigated including DNA vaccine engineering, co-delivery of immunomodulatory molecules, safe routes of administration, prime-boost regimen and strategies to break the immunosuppressive networks mechanisms adopted by malignant cells to prevent immune cell function. Combined or single strategies to enhance the efficacy and immunogenicity of DNA vaccines are applied in completed and ongoing clinical trials, where the safety and tolerability of the DNA platform are substantiated.
In this review on DNA vaccines, salient aspects on this topic going from basic research to the clinic are evaluated. Some representative DNA cancer vaccine studies are also discussed.
Collapse
|
47
|
Xiang SD, Selomulya C, Ho J, Apostolopoulos V, Plebanski M. Delivery of DNA vaccines: an overview on the use of biodegradable polymeric and magnetic nanoparticles. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2010; 2:205-18. [DOI: 10.1002/wnan.88] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
48
|
Kim S, Li L, McMurtrey CP, Hildebrand WH, Weidanz JA, Gillanders WE, Diamond MS, Hansen TH. Single-chain HLA-A2 MHC trimers that incorporate an immundominant peptide elicit protective T cell immunity against lethal West Nile virus infection. THE JOURNAL OF IMMUNOLOGY 2010; 184:4423-30. [PMID: 20212098 DOI: 10.4049/jimmunol.0903955] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The generation of a robust CD8(+) T cell response is an ongoing challenge for the development of DNA vaccines. One problem encountered with classical DNA plasmid immunization is that peptides produced are noncovalently and transiently associated with MHC class I molecules and thus may not durably stimulate CD8(+) T cell responses. To address this and enhance the expression and presentation of the antigenic peptide/MHC complexes, we generated single-chain trimers (SCTs) composed of a single polypeptide chain with a linear composition of antigenic peptide, beta(2)-microglobulin, and H chain connected by flexible linkers. In this study, we test whether the preassembled nature of the SCT makes them effective for eliciting protective CD8(+) T cell responses against pathogens. A DNA plasmid was constructed encoding an SCT incorporating the human MHC class I molecule HLA-A2 and the immunodominant peptide SVG9 derived from the envelope protein of West Nile virus (WNV). HLA-A2 transgenic mice vaccinated with the DNA encoding the SVG9/HLA-A2 SCT generated a robust epitope-specific CD8(+) T cell response and showed enhanced survival rate and lower viral burden in the brain after lethal WNV challenge. Inclusion of a CD4(+) Th cell epitope within the SCT did not increase the frequency of SVG9-specific CD8(+) T cells, but did enhance protection against WNV challenge. Overall, these findings demonstrate that the SCT platform can induce protective CD8(+) T cell responses against lethal virus infection and may be paired with immunogens that elicit robust neutralizing Ab responses to generate vaccines that optimally activate all facets of adaptive immunity.
Collapse
Affiliation(s)
- Sojung Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Bal SM, Slütter B, van Riet E, Kruithof AC, Ding Z, Kersten GF, Jiskoot W, Bouwstra JA. Efficient induction of immune responses through intradermal vaccination with N-trimethyl chitosan containing antigen formulations. J Control Release 2010; 142:374-83. [DOI: 10.1016/j.jconrel.2009.11.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2009] [Revised: 11/11/2009] [Accepted: 11/15/2009] [Indexed: 10/20/2022]
|
50
|
Bae JY, Moon SH, Choi JA, Park JS, Hahn BS, Kim KY, Kim B, Song JY, Kwon DH, Lee SC, Kim JB, Yang JS. Recombinant DNA and Protein Vaccines for Foot-and-mouth Disease Induce Humoral and Cellular Immune Responses in Mice. Immune Netw 2009; 9:265-73. [PMID: 20157614 PMCID: PMC2816960 DOI: 10.4110/in.2009.9.6.265] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2009] [Revised: 11/23/2009] [Accepted: 12/18/2009] [Indexed: 12/01/2022] Open
Abstract
Background Foot-and-mouth disease virus (FMDV) is a small single-stranded RNA virus which belongs to the family Picornaviridae, genus Apthovirus. It is a principal cause of FMD which is highly contagious in livestock. In a wild type virus infection, infected animals usually elicit antibodies against structural and non-structural protein of FMDV. A structural protein, VP1, is involved in neutralization of virus particle, and has both B and T cell epitopes. A RNA-dependent RNA polymerase, 3D, is highly conserved among other serotypes and strongly immunogenic, therefore, we selected VP1 and 3D as vaccine targets. Methods VP1 and 3D genes were codon-optimized to enhance protein expression level and cloned into mammalian expression vector. To produce recombinant protein, VP1 and 3D genes were also cloned into pET vector. The VP1 and 3D DNA or proteins were co-immunized into 5 weeks old BALB/C mice. Results Antigen-specific serum antibody (Ab) responses were detected by Ab ELISA. Cellular immune response against VP1 and 3D was confirmed by ELISpot assay. Conclusion The results showed that all DNA- and protein-immunized groups induced cellular immune responses, suggesting that both DNA and recombinant protein vaccine administration efficiently induced Ag-specific humoral and cellular immune responses.
Collapse
Affiliation(s)
- Ji-Young Bae
- Department of Genetic Engineering, Faculty of Life Sciences and Technology, Sungkyunkwan University, Suwon 440-746, Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|