1
|
Oelkrug C. Analysis of physical and biological delivery systems for DNA cancer vaccines and their translation to clinical development. Clin Exp Vaccine Res 2024; 13:73-82. [PMID: 38752006 PMCID: PMC11091436 DOI: 10.7774/cevr.2024.13.2.73] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/17/2023] [Accepted: 03/30/2024] [Indexed: 05/18/2024] Open
Abstract
DNA cancer vaccines as an approach in tumor immunotherapy are still being investigated in preclinical and clinical settings. Nevertheless, only a small number of clinical studies have been published so far and are still active. The investigated vaccines show a relatively stable expression in in-vitro transfected cells and may be favorable for developing an immunologic memory in patients. Therefore, DNA vaccines could be suitable as a prophylactic or therapeutic approach against cancer. Due to the low efficiency of these vaccines, the administration technique plays an important role in the vaccine design and its efficacy. These DNA cancer vaccine delivery systems include physical, biological, and non-biological techniques. Although the pre-clinical studies show promising results in the application of the different delivery systems, further studies in clinical trials have not yet been successfully proven.
Collapse
|
2
|
Yin Y, Yang J, Gao G, Zhou H, Chi B, Yang HY, Li J, Wang Y. Enhancing cell-scale performance via sustained release of the varicella-zoster virus antigen from a microneedle patch under simulated microgravity. Biomater Sci 2024; 12:763-775. [PMID: 38164004 DOI: 10.1039/d3bm01440a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
The immune system of astronauts might become weakened in the microgravity environment in space, and the dormant varicella-zoster virus (VZV) in the body might be reactivated, seriously affecting their work and safety. For working in orbit for the long term, there is currently no efficient and durable delivery system of general vaccines in a microgravity environment. Accordingly, based on the previous foundation, we designed, modified, and synthesized a biodegradable and biocompatible copolymer, polyethylene glycol-polysulfamethazine carbonate urethane (PEG-PSCU) that could be mainly adopted to fabricate a novel sustained-release microneedle (S-R MN) patch. Compared with conventional biodegradable microneedles, this S-R MN patch could not only efficiently encapsulate protein vaccines (varicella-zoster virus glycoprotein E, VZV gE) but also further prolong the release time of VZV gE in a simulated microgravity (SMG) environment. Eventually, we verified the activation of dendritic cells by VZV gE released from the S-R MN patch in an SMG environment and the positive bioeffect of activated dendritic cells on lymphocytes using an in vitro lymph node model. This study is of great significance for the exploration of long-term specific immune responses to the VZV in an SMG environment.
Collapse
Affiliation(s)
- Yue Yin
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Junyuan Yang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Huaijuan Zhou
- Advanced Research Institute of Multidisciplinary Sciences, Beijing Institute of Technology, Beijing 100081, China
| | - Bowen Chi
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China.
| | - Hong Yu Yang
- College of Materials Science and Engineering, Jilin Institute of Chemical Technology, Jilin City 132022, Jilin Province, China.
| | - Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Yilong Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China.
| |
Collapse
|
3
|
Peng S, Tu HF, Cheng M, Hu MH, Tsai HL, Tsai YC, Koenig C, Brayton C, Wang H, Chang YN, Arend RC, Levinson K, Roden RBS, Wu TC, Hung CF. Immune responses, therapeutic anti-tumor effects, and tolerability upon therapeutic HPV16/18 E6/E7 DNA vaccination via needle-free biojector. mBio 2023; 14:e0212123. [PMID: 37791765 PMCID: PMC10653862 DOI: 10.1128/mbio.02121-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 08/15/2023] [Indexed: 10/05/2023] Open
Abstract
IMPORTANCE Respectively, HPV16 and HPV18 cause 50% and 20% of cervical cancer cases globally. Viral proteins E6 and E7 are obligate drivers of oncogenic transformation. We recently developed a candidate therapeutic DNA vaccine, pBI-11, that targets HPV16 and HPV18 E6 and E7. Single-site intramuscular delivery of pBI-11 via a needle elicited therapeutic anti-tumor effects in mice and is now being tested in high-risk human papillomavirus+ head and neck cancer patients (NCT05799144). Needle-free biojectors such as the Tropis device show promise due to ease of administration, high patient acceptability, and the possibility of improved delivery. For example, vaccination of patients with the ZyCoV-D DNA vaccine using the Tropis device is effective against COVID19, well tolerated, and licensed. Here we show that split-dose, multi-site administration and intradermal delivery via the Tropis biojector increase the delivery of pBI-11 DNA vaccine, enhance HPV antigen-specific CD8+ T-cell responses, and improve anti-tumor therapeutic effects, suggesting its translational potential to treat HPV16/18 infection and disease.
Collapse
Affiliation(s)
- Shiwen Peng
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hsin-Fang Tu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michelle Cheng
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ming-Hung Hu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hua-Ling Tsai
- Department of Oncology Biostatistics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ya-Chea Tsai
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chelsea Koenig
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Cory Brayton
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hao Wang
- Department of Oncology Biostatistics, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Rebecca C. Arend
- Department of Obstetrics and Gynecology, O’Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Kimberly Levinson
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Richard B. S. Roden
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
| | - T. C. Wu
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Chien-Fu Hung
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Obstetrics and Gynecology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
4
|
Zhao H, Li P, Bian L, Zhang W, Jiang C, Chen Y, Kong W, Zhang Y. Immune Response of Inactivated Rabies Vaccine Inoculated via Intraperitoneal, Intramuscular, Subcutaneous and Needle-Free Injection Technology-Based Intradermal Routes in Mice. Int J Mol Sci 2023; 24:13587. [PMID: 37686393 PMCID: PMC10488038 DOI: 10.3390/ijms241713587] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Inoculation routes may significantly affect vaccine performance due to the local microenvironment, antigen localization and presentation, and, therefore, final immune responses. In this study, we conducted a head-to-head comparison of immune response and safety of inactivated rabies vaccine inoculated via intraperitoneal (IP), intramuscular (IM), subcutaneous (SC) and needle-free injection technology-based intradermal (ID) routes in ICR mice. Immune response was assessed in terms of antigen-specific antibodies, antibody subtypes and neutralizing antibodies for up to 28 weeks. A live rabies virus challenge was also carried out to evaluate vaccine potency. The dynamics of inflammatory cell infiltration at the skin and muscle levels were determined via histopathological examination. The kinetics and distribution of a model antigen were also determined by using in vivo fluorescence imaging. Evidence is presented that the vaccine inoculated via the ID route resulted in the highest antigen-specific antibody and neutralizing antibody titers among all administration routes, while IP and IM routes were comparable, followed by the SC route. Antibody subtype analysis shows that the IP route elicited a Th1-biased immune response, while SC and IM administration elicited a prominent Th2-type immune response. Unexpectedly, the ID route leads to a balanced Th1 and Th2 immune response. In addition, the ID route conferred effective protection against lethal challenge with 40 LD50 of the rabies CVS strain, which was followed by IP and IM routes. Moreover, a one-third dose of the vaccine inoculated via the ID route provided comparable or higher efficacy to a full dose of the vaccine via the other three routes. The superior performance of ID inoculation over other routes is related to longer local retention at injection sites and higher lymphatic drainage. Histopathology examination reveals a transient inflammatory cell infiltration at ID and IM injection sites which peaked at 48 h and 24 h, respectively, after immunization, with all side effects disappearing within one week. These results suggest that needle-free injection technology-based ID inoculation is a promising strategy for rabies vaccination in regard to safety and efficacy.
Collapse
Affiliation(s)
- Huiting Zhao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Peixuan Li
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Lijun Bian
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Wen Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Chunlai Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
- NMPA Key Laboratory of Humanized Animal Models for Evaluation of Vaccines and Cell Therapy Products, Jilin University, Changchun 130012, China
| | - Yan Chen
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
- NMPA Key Laboratory of Humanized Animal Models for Evaluation of Vaccines and Cell Therapy Products, Jilin University, Changchun 130012, China
| | - Wei Kong
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
- NMPA Key Laboratory of Humanized Animal Models for Evaluation of Vaccines and Cell Therapy Products, Jilin University, Changchun 130012, China
| | - Yong Zhang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, Changchun 130012, China
- NMPA Key Laboratory of Humanized Animal Models for Evaluation of Vaccines and Cell Therapy Products, Jilin University, Changchun 130012, China
| |
Collapse
|
5
|
Prins MLM, Prins C, de Vries JJC, Visser LG, Roukens AHE. Establishing immunogenicity and safety of needle-free intradermal delivery by nanoporous ceramic skin patch of mRNA SARS-CoV-2 vaccine as a revaccination strategy in healthy volunteers. Virus Res 2023; 334:199175. [PMID: 37473964 PMCID: PMC10392605 DOI: 10.1016/j.virusres.2023.199175] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/22/2023]
Abstract
INTRODUCTION Nanoporous microneedle arrays (npMNA) are being developed as skin patches for vaccine delivery. As alternative for needle-based immunisation, they may potentially result in higher vaccine acceptance, which is important for future mass vaccination campaigns to control outbreaks, such as COVID-19, and for public vaccination in general. In this study we investigated the safety and immunogenicity of needle-free intradermal delivery of a fractional third or fourth dose of mRNA-1273 vaccine by npMNA. METHODS This study was an open-label, randomised-controlled, proof-of-concept study. Healthy adults were eligible if they had received a primary immunisation series against SARS-CoV-2 with two doses of mRNA-1273 (Moderna) or BNT162b2 (Pfizer-BioNTech) mRNA vaccine. A history of a COVID-19 infection or booster vaccination with mRNA-1273 or BNT162b2 was allowed if it occurred at least three months before inclusion. Participants were randomised in a 1:1 ratio to receive 20 µg mRNA-1273 vaccine, either through npMNA patch applied on the skin (ID-patch group), or through intramuscular (IM) injection (IM-control group). Primary outcomes were reactogenicity up to two weeks after vaccination, and fold-increase of SARS-CoV-2 spike S1-specific IgG antibodies 14 days post-vaccination. RESULTS In April 2022, 20 participants were enroled. The geometric mean concentration (GMC) did not increase in the ID-patch group after vaccination, in contrast to the IM-control group (GMC was 1,006 BAU/mL (95% CI 599-1,689), 3,855 (2,800-5,306), and 3,513 (2,554-4,833) at day 1, 15 and 29, respectively). In addition, SARS-CoV-2-specific T cell responses were lower after ID vaccination through npMNA. CONCLUSION Needle-free delivery of 20 µg mRNA-1273 vaccine by npMNA failed to induce antibody and T cell responses. As this is a potentially very useful vaccination method, it is important to determine which adjustments are needed to make this npMNA successful. CLINICAL TRIAL REGISTRY (ON CLINICALTRIAL.GOV): NCT05315362.
Collapse
Affiliation(s)
- Manon L M Prins
- Department of Infectious Diseases, Leiden University Medical Centre, C5-P Albinusdreef 2, Leiden, ZA 2333, the Netherlands.
| | - Corine Prins
- Department of Infectious Diseases, Leiden University Medical Centre, C5-P Albinusdreef 2, Leiden, ZA 2333, the Netherlands
| | - Jutte J C de Vries
- Department of Medical Microbiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Leo G Visser
- Department of Infectious Diseases, Leiden University Medical Centre, C5-P Albinusdreef 2, Leiden, ZA 2333, the Netherlands
| | - Anna H E Roukens
- Department of Infectious Diseases, Leiden University Medical Centre, C5-P Albinusdreef 2, Leiden, ZA 2333, the Netherlands
| |
Collapse
|
6
|
He A, Li X, Dai Z, Li Q, Zhang Y, Ding M, Wen ZF, Mou Y, Dong H. Nanovaccine-based strategies for lymph node targeted delivery and imaging in tumor immunotherapy. J Nanobiotechnology 2023; 21:236. [PMID: 37482608 PMCID: PMC10364424 DOI: 10.1186/s12951-023-01989-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/08/2023] [Indexed: 07/25/2023] Open
Abstract
Therapeutic tumor vaccines have attracted considerable attention in the past decade; they can induce tumor regression, eradicate minimal residual disease, establish lasting immune memory and avoid non-specific and adverse side effects. However, the challenge in the field of therapeutic tumor vaccines is ensuring the delivery of immune components to the lymph nodes (LNs) to activate immune cells. The clinical response rate of traditional therapeutic tumor vaccines falls short of expectations due to inadequate lymph node delivery. With the rapid development of nanotechnology, a large number of nanoplatform-based LN-targeting nanovaccines have been exploited for optimizing tumor immunotherapies. In addition, some nanovaccines possess non-invasive visualization performance, which is benefit for understanding the kinetics of nanovaccine exposure in LNs. Herein, we present the parameters of nanoplatforms, such as size, surface modification, shape, and deformability, which affect the LN-targeting functions of nanovaccines. The recent advances in nanoplatforms with different components promoting LN-targeting are also summarized. Furthermore, emerging LNs-targeting nanoplatform-mediated imaging strategies to both improve targeting performance and enhance the quality of LN imaging are discussed. Finally, we summarize the prospects and challenges of nanoplatform-based LN-targeting and /or imaging strategies, which optimize the clinical efficacy of nanovaccines in tumor immunotherapies.
Collapse
Affiliation(s)
- Ao He
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Xiaoye Li
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Zhuo Dai
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Qiang Li
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Yu Zhang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Meng Ding
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China
| | - Zhi-Fa Wen
- Department of Clinical Laboratory, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, 210004, China.
| | - Yongbin Mou
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China.
| | - Heng Dong
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, 30 Zhongyang Road, Nanjing, 210008, China.
| |
Collapse
|
7
|
Xia Y, Fu S, Ma Q, Liu Y, Zhang N. Application of Nano-Delivery Systems in Lymph Nodes for Tumor Immunotherapy. NANO-MICRO LETTERS 2023; 15:145. [PMID: 37269391 PMCID: PMC10239433 DOI: 10.1007/s40820-023-01125-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/07/2023] [Indexed: 06/05/2023]
Abstract
Immunotherapy has become a promising research "hotspot" in cancer treatment. "Soldier" immune cells are not uniform throughout the body; they accumulate mostly in the immune organs such as the spleen and lymph nodes (LNs), etc. The unique structure of LNs provides the microenvironment suitable for the survival, activation, and proliferation of multiple types of immune cells. LNs play an important role in both the initiation of adaptive immunity and the generation of durable anti-tumor responses. Antigens taken up by antigen-presenting cells in peripheral tissues need to migrate with lymphatic fluid to LNs to activate the lymphocytes therein. Meanwhile, the accumulation and retaining of many immune functional compounds in LNs enhance their efficacy significantly. Therefore, LNs have become a key target for tumor immunotherapy. Unfortunately, the nonspecific distribution of the immune drugs in vivo greatly limits the activation and proliferation of immune cells, which leads to unsatisfactory anti-tumor effects. The efficient nano-delivery system to LNs is an effective strategy to maximize the efficacy of immune drugs. Nano-delivery systems have shown beneficial in improving biodistribution and enhancing accumulation in lymphoid tissues, exhibiting powerful and promising prospects for achieving effective delivery to LNs. Herein, the physiological structure and the delivery barriers of LNs were summarized and the factors affecting LNs accumulation were discussed thoroughly. Moreover, developments in nano-delivery systems were reviewed and the transformation prospects of LNs targeting nanocarriers were summarized and discussed.
Collapse
Affiliation(s)
- Yiming Xia
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Shunli Fu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Qingping Ma
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China
| | - Yongjun Liu
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.
| | - Na Zhang
- Department of Pharmaceutics, Key Laboratory of Chemical Biology (Ministry of Education), NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 Wenhua Xi Road, Jinan, 250012, Shandong, People's Republic of China.
| |
Collapse
|
8
|
Skin-Based Vaccination: A Systematic Mapping Review of the Types of Vaccines and Methods Used and Immunity and Protection Elicited in Pigs. Vaccines (Basel) 2023; 11:vaccines11020450. [PMID: 36851328 PMCID: PMC9962282 DOI: 10.3390/vaccines11020450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
The advantages of skin-based vaccination include induction of strong immunity, dose-sparing, and ease of administration. Several technologies for skin-based immunisation in humans are being developed to maximise these key advantages. This route is more conventionally used in veterinary medicine. Skin-based vaccination of pigs is of high relevance due to their anatomical, physiological, and immunological similarities to humans, as well as being a source of zoonotic diseases and their livestock value. We conducted a systematic mapping review, focusing on vaccine-induced immunity and safety after the skin immunisation of pigs. Veterinary vaccines, specifically anti-viral vaccines, predominated in the literature. The safe and potent skin administration to pigs of adjuvanted vaccines, particularly emulsions, are frequently documented. Multiple methods of skin immunisation exist; however, there is a lack of consistent terminology and accurate descriptions of the route and device. Antibody responses, compared to other immune correlates, are most frequently reported. There is a lack of research on the underlying mechanisms of action and breadth of responses. Nevertheless, encouraging results, both in safety and immunogenicity, were observed after skin vaccination that were often comparable to or superior the intramuscular route. Further research in this area will underlie the development of enhanced skin vaccine strategies for pigs, other animals and humans.
Collapse
|
9
|
Yang N, Jin X, Zhu C, Gao F, Weng Z, Du X, Feng G. Subunit vaccines for Acinetobacter baumannii. Front Immunol 2023; 13:1088130. [PMID: 36713441 PMCID: PMC9878323 DOI: 10.3389/fimmu.2022.1088130] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 12/22/2022] [Indexed: 01/15/2023] Open
Abstract
Acinetobacter baumannii is a gram-negative bacterium and a crucial opportunistic pathogen in hospitals. A. baumannii infection has become a challenging problem in clinical practice due to the increasing number of multidrug-resistant strains and their prevalence worldwide. Vaccines are effective tools to prevent and control A. baumannii infection. Many researchers are studying subunit vaccines against A. baumannii. Subunit vaccines have the advantages of high purity, safety, and stability, ease of production, and highly targeted induced immune responses. To date, no A. baumannii subunit vaccine candidate has entered clinical trials. This may be related to the easy degradation of subunit vaccines in vivo and weak immunogenicity. Using adjuvants or delivery vehicles to prepare subunit vaccines can slow down degradation and improve immunogenicity. The common immunization routes include intramuscular injection, subcutaneous injection, intraperitoneal injection and mucosal vaccination. The appropriate immunization method can also enhance the immune effect of subunit vaccines. Therefore, selecting an appropriate adjuvant and immunization method is essential for subunit vaccine research. This review summarizes the past exploration of A. baumannii subunit vaccines, hoping to guide current and future research on these vaccines.
Collapse
Affiliation(s)
- Ning Yang
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Jin
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chenghua Zhu
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fenglin Gao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zheqi Weng
- The Second Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xingran Du
- Department of Infectious Disease, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China,*Correspondence: Xingran Du, ; Ganzhu Feng,
| | - Ganzhu Feng
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China,*Correspondence: Xingran Du, ; Ganzhu Feng,
| |
Collapse
|
10
|
Roozen GVT, Prins MLM, van Binnendijk R, den Hartog G, Kuiper VP, Prins C, Janse JJ, Kruithof AC, Feltkamp MCW, Kuijer M, Rosendaal FR, Roestenberg M, Visser LG, Roukens AHE. Safety and Immunogenicity of Intradermal Fractional Dose Administration of the mRNA-1273 Vaccine: A Proof-of-Concept Study. Ann Intern Med 2022; 175:1771-1774. [PMID: 36279543 PMCID: PMC9593280 DOI: 10.7326/m22-2089] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Geert V T Roozen
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Margaretha L M Prins
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Rob van Binnendijk
- Department of Immune Surveillance, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Gerco den Hartog
- Department of Immune Surveillance, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Vincent P Kuiper
- Department of Parasitology, Leiden University Medical Center, Leiden, the Netherlands
| | - Corine Prins
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Jacqueline J Janse
- Department of Parasitology, Leiden University Medical Center, Leiden, the Netherlands
| | - Annelieke C Kruithof
- Department of Infectious Diseases, Leiden University Medical Center, and Centre for Human Drug Research, Leiden, the Netherlands
| | - Mariet C W Feltkamp
- Department of Medical Microbiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Marjan Kuijer
- Department of Immune Surveillance, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Frits R Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Meta Roestenberg
- Department of Infectious Diseases and Department of Parasitology, Leiden University Medical Center, Leiden, the Netherlands
| | - Leo G Visser
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Anna H E Roukens
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
11
|
Chopra A, Gupta A. Skin as an immune organ and the site of biomimetic, non-invasive vaccination. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
12
|
Mrsny RJ. Does an Intradermal Vaccination for Monkeypox Make Sense? AAPS J 2022; 24:104. [PMID: 36195806 PMCID: PMC9531852 DOI: 10.1208/s12248-022-00754-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 09/19/2022] [Indexed: 11/07/2022] Open
Abstract
Mankind has recently had to deal a series of virus-mediated pandemics, resulting in extensive morbidity and mortality rates that have severely strained healthcare systems. While dealing with viral infections as a healthcare concern is not new, our exceptionally mobile society has added to the critical challenge of limiting pathogen spread of a highly transmissible virus prior to the generation, testing, and distribution of safe and effective vaccines. The tremendous global effort put forth to address the recent pandemic induced by SARS-CoV-2 infection has highlighted many of the strengths and weaknesses of how vaccines are identified, tested, and used to provide protection. These uncertainties are exacerbated by the lack of clear and consistent messaging that can occur when the processes of research, development, and clinical testing that normally requires years of study and consideration are compressed into a few months. In this commentary, I will provide some background on the intramuscular (IM), subcutaneous (SC), and intradermal (ID) administration routes used for injectable vaccines and some information on potential immunological outcomes. With this background, I will address the recent FDA decision to allow an approved vaccine against monkeypox virus to be administered by ID, as well as its initial approval route via SC, injection as a dose-sparing strategy to maximize immunization numbers using current stockpiles.
Collapse
Affiliation(s)
- Randall J. Mrsny
- grid.7340.00000 0001 2162 1699Department of Life Sciences and Centre for Technology Innovation, University of Bath, Bath, BA2 7AY UK
| |
Collapse
|
13
|
Intradermal Allergen Immunotherapy for Allergic Rhinitis: Current Evidence. J Pers Med 2022; 12:jpm12081341. [PMID: 36013290 PMCID: PMC9409804 DOI: 10.3390/jpm12081341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/11/2022] [Accepted: 08/17/2022] [Indexed: 12/02/2022] Open
Abstract
Allergic rhinitis (AR) is an immunoglobulin E (IgE)-mediated inflammatory disease that is induced by allergen introduction to the nasal mucosa, which triggers an inflammatory response. The current treatments for AR include allergen avoidance and pharmacotherapy; however, allergen-specific immunotherapy (AIT) is the only treatment that can be employed to modify immunologic responses and to achieve a cure for allergic diseases. The current standard routes of AIT administration are the subcutaneous and sublingual routes. Alternatively, the dermis contains a high density of dermal dendritic cells that act as antigen-presenting cells, so intradermal administration may confer added advantages and increase the efficacy of AIT. Moreover, intradermal immunotherapy (IDIT) may facilitate a reduction in the allergen dosage and a shortening of the treatment duration. The aim of this review was to search and evaluate the current evidence specific to IDIT, including its modified formulations, such as allergoids and peptides. The results of this review reveal conflicting evidence that suggests that the overall benefit of IDIT remains unclear. As such, further clinical trials are needed to establish the clinical utility of IDIT, and to determine the optimal treatment-related protocols.
Collapse
|
14
|
Intradermal delivery of mRNA using cryomicroneedles. Acta Biomater 2022; 148:133-141. [PMID: 35697200 DOI: 10.1016/j.actbio.2022.06.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 05/30/2022] [Accepted: 06/07/2022] [Indexed: 11/21/2022]
Abstract
Microneedles can realize the intradermal and transdermal delivery of drugs. However, most conventional microneedles made of metal, polymer and ceramics are unsuitable for the delivery of mRNA drugs that are fragile and temperature-sensitive. This study explores the usage of cryomicroneedles (CryoMNs) for the intradermal delivery of mRNA molecules. Taking luciferase mRNA as an example, we first optimize the formulation of CryoMNs to maximize mRNA stability. Later, in the mouse model, we compare the delivery efficiency with the conventional subcutaneous injection for both the luciferase mRNA and COVID-19 Comirnaty mRNA vaccines, where CryoMNs delivered mRNA vaccines successfully induce specific B-cell antibody, neutralizing activity and T-cell responses. STATEMENT OF SIGNIFICANCE: mRNA vaccines are fragile and temperature-sensitive, so they are mainly delivered by intramuscular injection that often causes pain and requires clinical expertise to immunize patients. Microneedles permit convenient, fast and safe vaccination. However, existing microneedle platforms are ineffective to protect the integrity of mRNA vaccines in fabrication, storage, and administration. This work utilizes cryomicroneedles (CryoMNs) technology to intradermally deliver mRNA. In the mouse model, CryoMNs are compared with the subcutaneous injection for the delivery efficiency of both the luciferase mRNA and COVID-19 Comirnaty mRNA vaccines, where CryoMNs delivered mRNA vaccines successfully produce specific B-cell antibodies, T-cell responses, and neutralizing activity. This work is expected to provide a new delivery strategy for the emerging mRNA therapeutics.
Collapse
|
15
|
Meng X, Zhu Z, Ahmed N, Ma Q, Wang Q, Deng B, Chen Q, Lu Y, Yang P. Dermal Microvascular Units in Domestic Pigs (Sus scrofa domestica): Role as Transdermal Passive Immune Channels. Front Vet Sci 2022; 9:891286. [PMID: 35548054 PMCID: PMC9083201 DOI: 10.3389/fvets.2022.891286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 03/30/2022] [Indexed: 12/14/2022] Open
Abstract
The dermal microvascular unit (DMU) is a perivascular functional unit in the dermis. It is composed of microvascular and capillary lymphatics surrounded by immune cells. In this study, jet needle-free injection system was used to injected biocompatible carbon nanoparticles into the cervical skin of domestic pigs (Sus scrofa domestica) and assessed the morphological distribution of DMUs by hematoxylin erythrosine staining, immunohistochemistry (IHC), and transmission electron microscopy (TEM), and TEM was also used to observe the ultrastructural changes of DMUs after jet needle-free injection. Following our study, we identified DMUs in the dermis stratum papillare and similar structures in the dermis stratum reticulare, but the aggregation of CD68+ and CD1a+ cells in the dermis stratum papillare of DMUs by IHC confirmed that DMUs act as reservoirs of dermal immune cells, while similar structures in the dermis stratum reticulare should not be considered as DMUs. Ultrastructure of DMUs was revealed by TEM. Marvelous changes were found following xenobiotics attack, including the rearrangement of endothelial cells and pericytes, and the reactivity of immune cells. Novel interstitial cell telocyte (TC) was also identified around the microvasculature, which may have been previously known as the veil cell. Our results successfully identified the distribution of DMUs in the skin of domestic pigs, which might act as reservoirs of immune cells in the skin and play a role in immune surveillance and immune defense.
Collapse
Affiliation(s)
- Xiangfei Meng
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhaoxuan Zhu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Nisar Ahmed
- Department of Veterinary Anatomy and Histology, Faculty of Veterinary and Animal Sciences, Lasbela University of Agriculture, Water & Marine Sciences (LUAWMS), Uthal, Pakistan
| | - Qianhui Ma
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Qi Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Bihua Deng
- National Research Center of Engineering and Technology for Veterinary Biologicals, Institute of Veterinary Immunology and Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Qiusheng Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yu Lu
- National Research Center of Engineering and Technology for Veterinary Biologicals, Institute of Veterinary Immunology and Engineering, Jiangsu Academy of Agricultural Sciences, Nanjing, China
- Yu Lu
| | - Ping Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- *Correspondence: Ping Yang
| |
Collapse
|
16
|
Labouta HI, Langer R, Cullis PR, Merkel OM, Prausnitz MR, Gomaa Y, Nogueira SS, Kumeria T. Role of drug delivery technologies in the success of COVID-19 vaccines: a perspective. Drug Deliv Transl Res 2022; 12:2581-2588. [PMID: 35290656 PMCID: PMC8923087 DOI: 10.1007/s13346-022-01146-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/28/2022] [Indexed: 12/15/2022]
Abstract
The triumphant success of mRNA vaccines is a testimony to the important role drug delivery technologies have played in protecting billions of people against SARS-CoV-2 (or the Corona Virus Disease 2019; COVID-19). Several lipid nanoparticle (LNP) mRNA vaccines were developed and have been instrumental in preventing the disease by boosting the immune system against the pathogen, SARS-CoV-2. These vaccines have been built on decades of scientific research in drug delivery of mRNA, vaccines, and other biologicals. In this manuscript, several leading and emerging scientists in the field of drug delivery share their perspective on the role of drug delivery technologies in developing safe and efficacious vaccines, in a roundtable discussion. The authors also discussed their viewpoint on the current challenges, and the key research questions that should drive this important area of research.
Collapse
Affiliation(s)
- Hagar I. Labouta
- grid.21613.370000 0004 1936 9609College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5 Canada ,grid.21613.370000 0004 1936 9609Biomedical Engineering, University of Manitoba, Winnipeg, MB Canada ,grid.460198.20000 0004 4685 0561Children’s Hospital Research Institute of Manitoba, Winnipeg, MB R3E 3P4 Canada ,grid.7155.60000 0001 2260 6941Faculty of Pharmacy, Alexandria University, Alexandria, 21521 Egypt
| | - Robert Langer
- grid.116068.80000 0001 2341 2786David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139 USA ,grid.116068.80000 0001 2341 2786Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
| | - Pieter R. Cullis
- grid.17091.3e0000 0001 2288 9830Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Olivia M. Merkel
- grid.5252.00000 0004 1936 973XDepartment of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians-University, 81337 Munich, Germany
| | - Mark R. Prausnitz
- grid.213917.f0000 0001 2097 4943School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332 USA
| | - Yasmine Gomaa
- grid.213917.f0000 0001 2097 4943School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA 30332 USA
| | - Sara S. Nogueira
- grid.1005.40000 0004 4902 0432School of Materials Science and Engineering, University of New South Wales, Kensington, Sydney, NSW 2052 Australia ,grid.1005.40000 0004 4902 0432Australian Centre for NanoMedicine, University of New South Wales, Kensington, Sydney, NSW 2052 Australia
| | - Tushar Kumeria
- grid.1005.40000 0004 4902 0432School of Materials Science and Engineering, University of New South Wales, Kensington, Sydney, NSW 2052 Australia ,grid.1005.40000 0004 4902 0432Australian Centre for NanoMedicine, University of New South Wales, Kensington, Sydney, NSW 2052 Australia
| |
Collapse
|
17
|
Ding Y, Li Z, Jaklenec A, Hu Q. Vaccine delivery systems toward lymph nodes. Adv Drug Deliv Rev 2021; 179:113914. [PMID: 34363861 PMCID: PMC9418125 DOI: 10.1016/j.addr.2021.113914] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/05/2021] [Accepted: 07/31/2021] [Indexed: 12/15/2022]
Abstract
Strategies of improving vaccine targeting ability toward lymph nodes have been attracting considerable interest in recent years, though there are remaining delivery barriers based on the inherent properties of lymphatic systems and limited administration routes of vaccination. Recently, emerging vaccine delivery systems using various materials as carriers are widely developed to achieve efficient lymph node targeting and improve vaccine-triggered adaptive immune response. In this review, to further optimize the vaccine targeting ability for future research, the design principles of lymph node targeting vaccine delivery based on the anatomy of lymph nodes and vaccine administration routes are first summarized. Then different designs of lymph node targeting vaccine delivery systems, including vaccine delivery systems in clinical applications, are carefully surveyed. Also, the challenges and opportunities of current delivery systems for vaccines are concluded in the end.
Collapse
Affiliation(s)
- Yingyue Ding
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, United States,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Zhaoting Li
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, United States,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, United States
| | - Ana Jaklenec
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, United States
| | - Quanyin Hu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, United States; Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, United States.
| |
Collapse
|
18
|
Microneedle-Mediated Vaccination: Innovation and Translation. Adv Drug Deliv Rev 2021; 179:113919. [PMID: 34375682 DOI: 10.1016/j.addr.2021.113919] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022]
Abstract
Vaccine administration by subcutaneous or intramuscular injection is the most commonly prescribed route for inoculation, however, it is often associated with some deficiencies such as low compliance, high professionalism, and risk of infection. Therefore, the application of microneedles for vaccine delivery has gained widespread interests in the past few years due to its high compliance, minimal invasiveness, and convenience. This review focuses on recent advances in the development and application of microneedles for vaccination based on different delivery strategies, and introduces the current status of microneedle-mediated vaccination in clinical translation. The prospects for its application including opportunities and challenges are further discussed.
Collapse
|
19
|
Choudhury SM, Ma X, Dang W, Li Y, Zheng H. Recent Development of Ruminant Vaccine Against Viral Diseases. Front Vet Sci 2021; 8:697194. [PMID: 34805327 PMCID: PMC8595237 DOI: 10.3389/fvets.2021.697194] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 10/04/2021] [Indexed: 01/21/2023] Open
Abstract
Pathogens of viral origin produce a large variety of infectious diseases in livestock. It is essential to establish the best practices in animal care and an efficient way to stop and prevent infectious diseases that impact animal husbandry. So far, the greatest way to combat the disease is to adopt a vaccine policy. In the fight against infectious diseases, vaccines are very popular. Vaccination's fundamental concept is to utilize particular antigens, either endogenous or exogenous to induce immunity against the antigens or cells. In light of how past emerging and reemerging infectious diseases and pandemics were handled, examining the vaccination methods and technological platforms utilized for the animals may provide some useful insights. New vaccine manufacturing methods have evolved because of developments in technology and medicine and our broad knowledge of immunology, molecular biology, microbiology, and biochemistry, among other basic science disciplines. Genetic engineering, proteomics, and other advanced technologies have aided in implementing novel vaccine theories, resulting in the discovery of new ruminant vaccines and the improvement of existing ones. Subunit vaccines, recombinant vaccines, DNA vaccines, and vectored vaccines are increasingly gaining scientific and public attention as the next generation of vaccines and are being seen as viable replacements to conventional vaccines. The current review looks at the effects and implications of recent ruminant vaccine advances in terms of evolving microbiology, immunology, and molecular biology.
Collapse
Affiliation(s)
- Sk Mohiuddin Choudhury
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - XuSheng Ma
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Wen Dang
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - YuanYuan Li
- Gansu Agricultural University, Lanzhou, China
| | - HaiXue Zheng
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
20
|
Lee SI, Jeong CG, Ul Salam Mattoo S, Nazki S, Prasad Aganja R, Kim SC, Khatun A, Oh Y, Noh SH, Lee SM, Kim WI. Protective immunity induced by concurrent intradermal injection of porcine circovirus type 2 and Mycoplasma hyopneumoniae inactivated vaccines in pigs. Vaccine 2021; 39:6691-6699. [PMID: 34538524 DOI: 10.1016/j.vaccine.2021.07.043] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 05/25/2021] [Accepted: 07/16/2021] [Indexed: 11/15/2022]
Abstract
Vaccines against porcine circovirus type 2 (PCV2) and Mycoplasma hyopneumoniae (Mhp) are routinely used by intramuscular injection. However, since intramuscular vaccination causes stress and increases the risk of cross-contamination among pigs, research on intradermal vaccination is currently being actively conducted. This study was designed to evaluate the efficacy of intradermally administered inactivated vaccines against PCV2 and Mhp in pigs. Three-week-old specific pathogen-free pigs were divided into three groups (5 pigs per group). Pigs in the two groups were intradermally vaccinated with the PCV2 or Mhp vaccine using a needle-free injector. Pigs in the third group were kept as nonvaccinated controls. At 21 days post-vaccination, pigs in one of these vaccinated groups and the nonvaccinated group were intranasally challenged with PCV2b and Mhp, while the other vaccinated group pigs were maintained as vaccine controls. Vaccine efficacy was evaluated by observing weight gain, pathogen load, pathological changes, and humoral or cellular immune responses. As a result, vaccinated pigs revealed significantly higher body weight gain, with lower clinical scores. Vaccinated pigs also showed higher antibody responses but lower PCV2b or Mhp loads in sera, nasal swabs, or lungs than nonvaccinated pigs. Intriguingly, vaccinated pigs upregulated cytotoxic T cells (CTLs), helper T type 1 cells (Th1 cells), and helper T type 17 cells (Th17 cells) after immunization and showed significantly higher levels of CTLs, Th1 and Th17 cells at 14 days post-challenge than nonvaccinated and challenged pigs. This study demonstrated that protective immune responses against PCV2 and Mhp could be efficiently induced in pigs using a relatively small volume of intradermal vaccines, probably due to effective antigen delivery to antigen-presenting cells in the dermis.
Collapse
Affiliation(s)
- Sim-In Lee
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea.
| | - Chang-Gi Jeong
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea.
| | | | - Salik Nazki
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea; The Pirbright Institute, Ash Road, Pirbright-GU24 0NF, Woking, United Kingdom.
| | - Ram Prasad Aganja
- Division of Biotechnology, Jeonbuk National University, Iksan, Republic of Korea.
| | - Seung-Chai Kim
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea.
| | - Amina Khatun
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea; Department of Pathology, Faculty of Animal Science and Veterinary Medicine, Sher-e-Bangla Agricultural University, Sher-e-Bangla Nagar, Dhaka 1207, Bangladesh.
| | - Yeonsu Oh
- Department of Veterinary Pathology, College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Republic of Korea.
| | - Sang-Hyun Noh
- MSD Animal Health Korea Ltd., Seoul 04637, Republic of Korea.
| | - Sang-Myeong Lee
- Division of Biotechnology, Jeonbuk National University, Iksan, Republic of Korea.
| | - Won-Il Kim
- College of Veterinary Medicine, Jeonbuk National University, Iksan, Republic of Korea.
| |
Collapse
|
21
|
Che Y, Yang Y, Suo J, Chen C, Wang X. Intratumoral Injection of a Human Papillomavirus Therapeutic Vaccine-Induced Strong Anti-TC-1-Grafted Tumor Activity in Mice. Cancer Manag Res 2021; 13:7339-7354. [PMID: 34584459 PMCID: PMC8464315 DOI: 10.2147/cmar.s329471] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 09/08/2021] [Indexed: 12/27/2022] Open
Abstract
Purpose The route of administration of a therapeutic tumor vaccine is a critical factor in inducing antitumor activity. In this study, we explored the effects of three vaccination routes (subcutaneous, peritumoral, and intratumoral injection) on antitumor activity induced by a human papillomavirus (HPV) therapeutic vaccine containing HPV16 E7 peptide combined with the adjuvant CpG ODN in established TC-1 grafted tumors. Methods We used flow cytometry to evaluate splenic and tumor-infiltrating immune cells. We also assessed transcriptional changes in a sequence of immune-related genes in tumors of different treatment groups using quantitative real-time polymerase chain reaction. Immunohistochemistry was used to determine the expression of molecules related to tumor infiltrating immune cells, angiogenesis, and cancer-associated fibroblasts in tumor tissues. Results Our results suggested that intratumoral and peritumoral vaccination generated enhanced antitumor activity compared to subcutaneous delivery. In particular, intratumoral vaccination elicited a stronger antitumor effect, with two of the six treated mice being nearly tumor-free at day 28. Three vaccination routes induced increases in splenic CD4+ and/or CD8+ T lymphocytes, and marked decreases in immunosuppressive cells. Peritumoral vaccination increased the tumor-infiltrating CD8+T cells in tumors, while intratumoral vaccination enhanced the tumor-infiltrating CD4+ and CD8+ T lymphocytes, as well as decreased the tumor-infiltrating of immunosuppressive cells, which may result in stronger inhibition of tumor growth and prolonged survival in mice bearing tumors. Furthermore, compared to the subcutaneous route, intratumoral vaccination led to a significant increase in antitumor cytokines and chemokines. In addition, our data showed marked downregulation of MMP-2, MMP-9, VEGF, CD31, and α-SMA in the intratumoral vaccination group, which might contribute to the suppression of tumor invasion, angiogenesis, and metastasis. Conclusion Overall, intratumoral vaccination is superior to subcutaneous delivery and has the potential to inhibit tumor growth by improving the tumor microenvironment.
Collapse
Affiliation(s)
- Yuxin Che
- Department of Microbiology and Parasitology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yang Yang
- Department of Microbiology and Parasitology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Jinguo Suo
- Department of Microbiology and Parasitology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Chang Chen
- Department of Dermatology, The First Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xuelian Wang
- Department of Microbiology and Parasitology, College of Basic Medical Science, China Medical University, Shenyang, Liaoning, People's Republic of China
| |
Collapse
|
22
|
Jiang Y, Li X, Yu L, Tong W, Chen P, Wang S, Zhao K, Tan X, Gao F, Yu H, Li G, Li L, Zhang Y, van den Born E, Zhou Y, Tong G. Immune efficacy of a candidate porcine reproductive and respiratory syndrome vaccine rHN-NP49 administered by a Needle-free intradermal delivery system in comparison with intramuscular injection. Vaccine 2021; 39:5557-5562. [PMID: 34412921 DOI: 10.1016/j.vaccine.2021.08.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/04/2021] [Accepted: 08/08/2021] [Indexed: 11/25/2022]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is one of the major drivers of economic loss in the swine industry worldwide. In commercial pig production, vaccination is the first option in an attempt to control infectious diseases. Pigs are therefore often immunized with different vaccines, and almost all of them are delivered via the intramuscular (IM) route. However, the IM injection may result in physical damage, stress reactions, and is labor demanding. An alternative route is urgently needed to reduce the disadvantages of conventional vaccination. In this study, a needle-free intradermal (ID) delivery system was evaluated for delivering a live PRRS vaccine as compared with the traditional needle-syringe method. Fifty-two 4-week-old piglets were divided into six groups: piglets in groups A-C were immunized using ID delivery system with 104, 105 and 106 TCID50 of PRRS candidate vaccine strain rHN-NP49, respectively; piglets in group D were immunized IM with 105 TCID50 of rHN-NP49; and group E and F were used as challenge and control groups, respectively. At 28 days post vaccination, piglets in group A to E were challenged with a lethal dose of highly-pathogenic PRRSV. Similar results were found in viremia and antibody response among the ID and IM groups during the immunization stage. After challenge, similar results were found in average body weight gain, viral shedding, serum viral load, and clinical score among the immunization groups, with a higher protection ratio in the ID group compared with IM group with the same immunization dose. These results demonstrated that the ID delivery system could provide similar or even better protection compared with IM route, and could be an effective route for PRRS vaccination.
Collapse
Affiliation(s)
- Yifeng Jiang
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Xianbin Li
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Lingxue Yu
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Wu Tong
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Pengfei Chen
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Shuaiyong Wang
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Kuan Zhao
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Xiangmei Tan
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Fei Gao
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Hai Yu
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Guoxin Li
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Liwei Li
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | - Yujiao Zhang
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China
| | | | - Yanjun Zhou
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China.
| | - Guangzhi Tong
- Department of Swine Infectious Diseases, Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, PR China; Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Disease and Zoonosis Yangzhou University, Yangzhou 225009, PR China.
| |
Collapse
|
23
|
Joshi D, Gala RP, Uddin MN, D'Souza MJ. Novel ablative laser mediated transdermal immunization for microparticulate measles vaccine. Int J Pharm 2021; 606:120882. [PMID: 34298102 DOI: 10.1016/j.ijpharm.2021.120882] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 07/03/2021] [Accepted: 07/11/2021] [Indexed: 12/20/2022]
Abstract
With the need for safe and efficacious vaccines which could be administered via non-invasive procedure, alternatives to traditional injectables vaccines are sought after. The present study aimed to develop the microparticulate formulation of measles vaccine and explore the feasibility of transdermal delivery via ablative laser mediated skin microporation. Transdermal route offers several advantages including painless immunization and ease of administration. We propose to use P.L.E.A.S.E. ablative laser for transdermal immunization of the microparticulate measles vaccine. This laser emits energy at 2940 µm, enabling cold ablation. This creates the micropores of defined size for delivery of vaccines into the skin. We compared the efficacy of transdermal immunization using the particulate formulation of the vaccine to that of traditional subcutaneous immunization using soluble and particulate vaccine. The microparticles were formulated using the biocompatible and biodegradable bovine serum albumin (BSA)-based polymer matrix. These vaccine microparticles were non-cytotoxic to the antigen presenting cells (APCs) and could effectively stimulate the innate immune response, confirmed by release of nitric oxide (NO) from the Griess's assay. The APCs when exposed to vaccine microparticles also showed a significantly higher expression of antigen-presenting molecules, MHC I and MHC II, and their co-stimulatory molecules, CD80 and CD40 as compared to the blank microparticles. The microparticulate measles vaccine was evaluated in vivo in the murine model. We compared the serum IgG and IgM levels in the mice receiving the vaccine subcutaneously and transdermally post-immunization. The results revealed that transdermal immunization with microparticulate vaccine is as efficient as the traditional subcutaneous administration.
Collapse
Affiliation(s)
- Devyani Joshi
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, United States
| | - Rikhav P Gala
- Fraunhofer USA, Center Mid-Atlantic, Biotechnology Division, Newark, DE 19702, United States
| | - Mohammad N Uddin
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, United States
| | - Martin J D'Souza
- Center for Drug Delivery Research, Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, United States.
| |
Collapse
|
24
|
Rouphael NG, Lai L, Tandon S, McCullough MP, Kong Y, Kabbani S, Natrajan MS, Xu Y, Zhu Y, Wang D, O'Shea J, Sherman A, Yu T, Henry S, McAllister D, Stadlbauer D, Khurana S, Golding H, Krammer F, Mulligan MJ, Prausnitz MR. Immunologic mechanisms of seasonal influenza vaccination administered by microneedle patch from a randomized phase I trial. NPJ Vaccines 2021; 6:89. [PMID: 34262052 PMCID: PMC8280206 DOI: 10.1038/s41541-021-00353-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 06/11/2021] [Indexed: 12/02/2022] Open
Abstract
In a phase 1 randomized, single-center clinical trial, inactivated influenza virus vaccine delivered through dissolvable microneedle patches (MNPs) was found to be safe and immunogenic. Here, we compare the humoral and cellular immunologic responses in a subset of participants receiving influenza vaccination by MNP to the intramuscular (IM) route of administration. We collected serum, plasma, and peripheral blood mononuclear cells in 22 participants up to 180 days post-vaccination. Hemagglutination inhibition (HAI) titers and antibody avidity were similar after MNP and IM vaccination, even though MNP vaccination used a lower antigen dose. MNPs generated higher neuraminidase inhibition (NAI) titers for all three influenza virus vaccine strains tested and triggered a larger percentage of circulating T follicular helper cells (CD4 + CXCR5 + CXCR3 + ICOS + PD-1+) compared to the IM route. Our study indicates that inactivated influenza virus vaccination by MNP produces humoral and cellular immune response that are similar or greater than IM vaccination.
Collapse
Affiliation(s)
- Nadine G Rouphael
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia.
| | - Lilin Lai
- Emory Vaccine Center, Department of Pediatrics, School of Medicine, Emory University, Atlanta, Georgia
| | - Sonia Tandon
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia.,Laney Graduate School, Emory University, Atlanta, Georgia
| | - Michele Paine McCullough
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Yunchuan Kong
- Laney Graduate School, Emory University, Atlanta, Georgia
| | - Sarah Kabbani
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Muktha S Natrajan
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Yongxian Xu
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Yerun Zhu
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Dongli Wang
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Jesse O'Shea
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Amy Sherman
- Hope Clinic of the Emory Vaccine Center, Division of Infectious Diseases, Department of Medicine, School of Medicine, Emory University, Atlanta, Georgia
| | - Tianwei Yu
- Laney Graduate School, Emory University, Atlanta, Georgia
| | | | | | - Daniel Stadlbauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Surender Khurana
- Division of Viral Products Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA
| | - Hana Golding
- Division of Viral Products Center for Biologics Evaluation and Research, FDA, Silver Spring, MD, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mark J Mulligan
- New York University Langone Medical Center, Alexandria Center for Life Sciences, New York, NY, USA
| | - Mark R Prausnitz
- Micron Biomedical, Inc., Atlanta, Georgia.,School of Chemical & Biomolecular Engineering, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|
25
|
Mammucari M, Paolucci T, Russo D, Maggiori E, Di Marzo R, Migliore A, Massafra U, Ronconi G, Ferrara PE, Gori F, Bifarini B, Brauneis S, Vellucci R, Mediati RD, Violo B, Natoli S, Pediliggieri C, Di Campli C, Collina MC. A Call to Action by the Italian Mesotherapy Society on Scientific Research. Drug Des Devel Ther 2021; 15:3041-3047. [PMID: 34285471 PMCID: PMC8285234 DOI: 10.2147/dddt.s321215] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 06/22/2021] [Indexed: 01/22/2023] Open
Abstract
Mesotherapy (local intradermal therapy, LIT) is a technique used to slowly spread drugs in tissues underlying the site of injection to prolong the pharmacological effect with respect to intramuscular injection. Recommendations for proper medical use of this technique have been made for pain medicine and rehabilitation, chronic venous disease, sport medicine, musculoskeletal disorders, several dermatological conditions, skin ageing, and immune-prophylaxis. Although mesotherapy is considered a valid technique, unresolved questions remain, which should be answered to standardize methodology and dosing regimen as well as to define the right indications in clinical practice. New randomized controlled trials are needed to test single products (dose, frequency of administration, efficacy and safety). Even infiltration of substances for dermo-cosmetic purposes must be guided by safety and efficacy tests before being proposed by mesotherapy. In this article, we put forth a preclinical and clinical research plan and a health technology assessment as a call to action by doctors, researchers and scientific societies to aid national health authorities in considering mesotherapy for prevention, treatment and rehabilitation paths.
Collapse
Affiliation(s)
| | - Teresa Paolucci
- Unit of Physical Medicine and Rehabilitation, G. D’ Annunzio University of Chieti-Pescara, Department of Oral Medical Science and Biotechnology (DSMOB), Chieti, Italy
| | | | | | | | - Alberto Migliore
- Unit of Rheumatology, San Pietro Fatebenefratelli Hospital, Rome, Italy
| | - Umberto Massafra
- Unit of Rheumatology, San Pietro Fatebenefratelli Hospital, Rome, Italy
| | - Gianpaolo Ronconi
- Physical Medicine and Rehabilitation Unit, IRCCS, Catholic University of Sacred Heart, Rome, Italy
| | - Paola Emilia Ferrara
- Physical Medicine and Rehabilitation Unit, IRCCS, Catholic University of Sacred Heart, Rome, Italy
| | - Fabio Gori
- Section of Anaesthesia, Intensive Care, and Pain Medicine, University Hospital Santa Maria Della Misericordia, Perugia, Italy
| | - Barbara Bifarini
- Section of Anaesthesia, Intensive Care, and Pain Medicine, University Hospital Santa Maria Della Misericordia, Perugia, Italy
| | - Stefano Brauneis
- Pain Centre “Enzo BorzomatI”, University Hospital of Rome “Policlinico Umberto I”, Rome, Italy
| | - Renato Vellucci
- Anaesthesiology Department, University Hospital Careggi, Florence, Italy
| | | | - Bartolomeo Violo
- Pain Therapy Unit, S. Spirito Nuovo Regina Margherita Hospital, Rome, Italy
| | - Silvia Natoli
- Department of Clinical Science and Translational Medicine, Tor Vergata University, Rome, Italy
| | - Carmen Pediliggieri
- Department of Vascular Surgery, Limb Salvage and Diabetic Foot, IDI-IRCCS, Rome, Italy
| | - Cristiana Di Campli
- Department of Vascular Surgery, Limb Salvage and Diabetic Foot, IDI-IRCCS, Rome, Italy
| | - Maria Chiara Collina
- Department of Vascular Surgery, Limb Salvage and Diabetic Foot, IDI-IRCCS, Rome, Italy
| |
Collapse
|
26
|
Migliore A, Gigliucci G, Di Marzo R, Russo D, Mammucari M. Intradermal Vaccination: A Potential Tool in the Battle Against the COVID-19 Pandemic? Risk Manag Healthc Policy 2021; 14:2079-2087. [PMID: 34045909 PMCID: PMC8144901 DOI: 10.2147/rmhp.s309707] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 04/26/2021] [Indexed: 12/20/2022] Open
Abstract
This narrative review is the final output of an initiative of the SIM (Italian Society of Mesotherapy). A narrative review of scientific literature on the efficacy of fractional intradermal vaccination in comparison with full doses has been conducted for the following pathogens: influenza virus, rabies virus, poliovirus (PV), hepatitis B virus (HBV), hepatitis A virus (HAV), diphtheria-tetanus-pertussis bacterias (DTP), human papillomavirus (HPV), Japanese encephalitis virus (JE), meningococcus, varicella zoster virus (VZV) and yellow fever virus. The findings suggest that the use of the intradermal route represents a valid strategy in terms of efficacy and efficiency for influenza, rabies and HBV vaccines. Some systematic reviews on influenza vaccines suggest the absence of a substantial difference between immunogenicity induced by a fractional ID dose of up to 20% and the IM dose in healthy adults, elderly, immunocompromised patients and children. Clinical studies of remaining vaccines against other pathogens (HAV, DTP bacterias, JE, meningococcal disease, VZV, and yellow fever virus) are scarce, but promising. In the context of a COVID-19 vaccine shortage, countries should investigate if a fractional dosing scheme may help to save doses and achieve herd immunity quickly. SIM urges the scientific community and health authorities to investigate the potentiality of fractionate intradermal administration in anti-COVID-19 vaccination. ![]()
Point your SmartPhone at the code above. If you have a QR code reader the video abstract will appear. Or use: https://youtu.be/xyVoP0mH6sQ
Collapse
Affiliation(s)
- Alberto Migliore
- Department of Internal Medicine, Unit of Rheumatology, San Pietro Fatebenefratelli Hospital, Rome, Italy
| | - Gianfranco Gigliucci
- Department of Internal Medicine, Unit of Rheumatology, San Pietro Fatebenefratelli Hospital, Rome, Italy
| | | | | | | |
Collapse
|
27
|
Tripp CH, Voit H, An A, Seidl-Philipp M, Krapf J, Sigl S, Romani N, Del Frari B, Stoitzner P. Laser-assisted epicutaneous immunization to target human skin dendritic cells. Exp Dermatol 2021; 30:1279-1289. [PMID: 33797121 DOI: 10.1111/exd.14346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/10/2021] [Accepted: 03/23/2021] [Indexed: 01/11/2023]
Abstract
Dendritic cells (DC) are promising targets for immunotherapy of cancer. Clinically, immunization against cancer antigens by means of the most potent antigen-presenting cells, that is DC, remains an important treatment option in combination with the modern immune checkpoint approaches. Instead of adoptively transferring in vitro monocyte-derived DC, they can also be loaded in situ by antibody-mediated targeting of antigen. Conventionally, these vaccines are delivered by classical intradermal injections. Here, we tested an alternative approach, namely laser-assisted epicutaneous immunization. With an infrared laser ("Precise Laser Epidermal System"/P.L.E.A.S.E.® Laser System), we created micropores in human skin and applied monoclonal antibodies (mAbs) against C-type lectins, for example DEC-205/CD205 and Langerin/CD207. Optimal parameters for formation of pores in epidermis and dermis were determined. We could induce pores of defined depths without enhanced apoptosis around them. Antibodies applied epicutaneously to the laser-porated skin could be detected both in Langerhans cells (LC) in situ in the epidermis and in migratory skin DC subsets from short term human skin explant culture, demonstrating uptake and transport of Langerin and DEC-205 mAbs. Efficacy of targeting was similar between the different laser treatments and pore depths. Thus, laser-assisted epicutaneous immunization may be a valuable alternative to intradermal injection, yet the loading efficacy of DC needs to be further improved.
Collapse
Affiliation(s)
- Christoph H Tripp
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Hermann Voit
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Angela An
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Magdalena Seidl-Philipp
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Johanna Krapf
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Stephan Sigl
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Nikolaus Romani
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Barbara Del Frari
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Patrizia Stoitzner
- Department of Dermatology, Venereology & Allergology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
28
|
Leboux RJT, Schipper P, van Capel TMM, Kong L, van der Maaden K, Kros A, Jiskoot W, de Jong EC, Bouwstra JA. Antigen Uptake After Intradermal Microinjection Depends on Antigen Nature and Formulation, but Not on Injection Depth. FRONTIERS IN ALLERGY 2021; 2:642788. [PMID: 35386985 PMCID: PMC8974696 DOI: 10.3389/falgy.2021.642788] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022] Open
Abstract
The skin is an attractive alternative administration route for allergy vaccination, as the skin is rich in dendritic cells (DCs) and is easily accessible. In the skin multiple subsets of DCs with distinct roles reside at different depths. In this study antigen (=allergen for allergy) formulations were injected in ex vivo human skin in a depth-controlled manner by using a hollow microneedle injection system. Biopsies were harvested at the injection site, which were then cultured for 72 h. Subsequently, the crawled-out cells were collected from the medium and analyzed with flow cytometry. Intradermal administration of ovalbumin (OVA, model antigen) solution at various depths in the skin did not affect the migration and maturation of DCs. OVA was taken up efficiently by the DCs, and this was not affected by the injection depth. In contrast, Bet v 1, the major allergen in birch pollen allergy, was barely taken up by dermal DCs (dDCs). Antigens were more efficiently taken up by CD14+ dDCs than CD1a+ dDCs, which in turn were more efficient at taken up antigen than Langerhans cells. Subsequently, both OVA and Bet v 1 were formulated in cationic and anionic liposomes, which altered antigen uptake drastically following intradermal microinjection. While OVA uptake was reduced by formulation in liposomes, Bet v 1 uptake in dDCs was increased by encapsulation in both cationic and anionic liposomes. This highlights the potential use of liposomes as adjuvant in intradermal allergy vaccine delivery. In conclusion, we observed that antigen uptake after intradermal injection was not affected by injection depth, but varied between different antigens and formulation.
Collapse
Affiliation(s)
- Romain J. T. Leboux
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Pim Schipper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Toni M. M. van Capel
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, Netherlands
| | - Lily Kong
- Division of Supramolecular Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
- Tongji School of Pharmacy, HuaZhong University of Science and Technology, Wuhan, China
| | - Koen van der Maaden
- Tumor Immunology Group, Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
- TECO Development GmbH, Rheinbach, Germany
| | - Alexander Kros
- Division of Supramolecular Chemistry, Leiden Institute of Chemistry, Leiden University, Leiden, Netherlands
| | - Wim Jiskoot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Esther C. de Jong
- Department of Experimental Immunology, Academic Medical Center, Amsterdam, Netherlands
- Esther C. de Jong
| | - Joke A. Bouwstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
- *Correspondence: Joke A. Bouwstra
| |
Collapse
|
29
|
Ho W, Gao M, Li F, Li Z, Zhang X, Xu X. Next-Generation Vaccines: Nanoparticle-Mediated DNA and mRNA Delivery. Adv Healthc Mater 2021; 10:e2001812. [PMID: 33458958 PMCID: PMC7995055 DOI: 10.1002/adhm.202001812] [Citation(s) in RCA: 130] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/06/2020] [Indexed: 01/07/2023]
Abstract
Nucleic acid vaccines are a method of immunization aiming to elicit immune responses akin to live attenuated vaccines. In this method, DNA or messenger RNA (mRNA) sequences are delivered to the body to generate proteins, which mimic disease antigens to stimulate the immune response. Advantages of nucleic acid vaccines include stimulation of both cell-mediated and humoral immunity, ease of design, rapid adaptability to changing pathogen strains, and customizable multiantigen vaccines. To combat the SARS-CoV-2 pandemic, and many other diseases, nucleic acid vaccines appear to be a promising method. However, aid is needed in delivering the fragile DNA/mRNA payload. Many delivery strategies have been developed to elicit effective immune stimulation, yet no nucleic acid vaccine has been FDA-approved for human use. Nanoparticles (NPs) are one of the top candidates to mediate successful DNA/mRNA vaccine delivery due to their unique properties, including unlimited possibilities for formulations, protective capacity, simultaneous loading, and delivery potential of multiple DNA/mRNA vaccines. This review will summarize the many varieties of novel NP formulations for DNA and mRNA vaccine delivery as well as give the reader a brief synopsis of NP vaccine clinical trials. Finally, the future perspectives and challenges for NP-mediated nucleic acid vaccines will be explored.
Collapse
Affiliation(s)
- William Ho
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Mingzhu Gao
- Engineering Research Center of Cell & Therapeutic AntibodyMinistry of Educationand School of PharmacyShanghai Jiao Tong University800 Dongchuan RoadShanghai200240P. R. China
| | - Fengqiao Li
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Zhongyu Li
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
| | - Xue‐Qing Zhang
- Engineering Research Center of Cell & Therapeutic AntibodyMinistry of Educationand School of PharmacyShanghai Jiao Tong University800 Dongchuan RoadShanghai200240P. R. China
| | - Xiaoyang Xu
- Department of Chemical and Materials EngineeringNew Jersey Institute of TechnologyNewarkNJ07102USA
- Department of Biomedical EngineeringNew Jersey Institute of Technology323 Dr Martin Luther King Jr BlvdNewarkNJ07102USA
| |
Collapse
|
30
|
Mercuri M, Fernandez Rivas D. Challenges and opportunities for small volumes delivery into the skin. BIOMICROFLUIDICS 2021; 15:011301. [PMID: 33532017 PMCID: PMC7826167 DOI: 10.1063/5.0030163] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 01/09/2021] [Indexed: 05/04/2023]
Abstract
Each individual's skin has its own features, such as strength, elasticity, or permeability to drugs, which limits the effectiveness of one-size-fits-all approaches typically found in medical treatments. Therefore, understanding the transport mechanisms of substances across the skin is instrumental for the development of novel minimal invasive transdermal therapies. However, the large difference between transport timescales and length scales of disparate molecules needed for medical therapies makes it difficult to address fundamental questions. Thus, this lack of fundamental knowledge has limited the efficacy of bioengineering equipment and medical treatments. In this article, we provide an overview of the most important microfluidics-related transport phenomena through the skin and versatile tools to study them. Moreover, we provide a summary of challenges and opportunities faced by advanced transdermal delivery methods, such as needle-free jet injectors, microneedles, and tattooing, which could pave the way to the implementation of better therapies and new methods.
Collapse
Affiliation(s)
- Magalí Mercuri
- Instituto de Nanociencia y Nanotecnología (CNEA-CONICET), Av. Gral. Paz 1499, 1650 San Martín, Buenos Aires, Argentina
| | - David Fernandez Rivas
- Mesoscale Chemical Systems Group, MESA+ Institute, TechMed Centre and Faculty of Science and Technology, University of Twente, P.O. Box 217, 7500 AE Enschede, The Netherlands
| |
Collapse
|
31
|
A first in human clinical trial assessing the safety and immunogenicity of transcutaneously delivered enterotoxigenic Escherichia coli fimbrial tip adhesin with heat-labile enterotoxin with mutation R192G. Vaccine 2020; 38:7040-7048. [DOI: 10.1016/j.vaccine.2020.09.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/04/2020] [Accepted: 09/08/2020] [Indexed: 01/09/2023]
|
32
|
Preclinical optimization of an enterotoxigenic Escherichia coli adjuvanted subunit vaccine using response surface design of experiments. NPJ Vaccines 2020; 5:83. [PMID: 32983577 PMCID: PMC7486917 DOI: 10.1038/s41541-020-00228-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 08/10/2020] [Indexed: 12/19/2022] Open
Abstract
Enterotoxigenic E. coli (ETEC) is a leading cause of moderate-to-severe diarrhoea. ETEC colonizes the intestine through fimbrial tip adhesin colonization factors and produces heat-stable and/or heat-labile (LT) toxins, stimulating fluid and electrolyte release leading to watery diarrhoea. We reported that a vaccine containing recombinant colonization factor antigen (CfaEB) targeting fimbrial tip adhesin of the colonization factor antigen I (CFA/I) and an attenuated LT toxoid (dmLT) elicited mucosal and systemic immune responses against both targets. Additionally, the toll-like receptor 4 ligand second-generation lipid adjuvant (TLR4-SLA) induced a potent mucosal response, dependent on adjuvant formulation. However, a combination of vaccine components at their respective individual optimal doses may not achieve the optimal immune profile. We studied a subunit ETEC vaccine prototype in mice using a response surface design of experiments (DoE), consisting of 64 vaccine dose-combinations of CfaEB, dmLT and SLA in four formulations (aqueous, aluminium oxyhydroxide, squalene-in-water stable nanoemulsion [SE] or liposomes containing the saponin Quillaja saponaria-21 [LSQ]). Nine readouts focusing on antibody functionality and plasma cell response were selected to profile the immune response of parenterally administered ETEC vaccine prototype. The data were integrated in a model to identify the optimal dosage of each vaccine component and best formulation. Compared to maximal doses used in mouse models (10 µg CfaEB, 1 µg dmLT and 5 µg SLA), a reduction in the vaccine components up to 37%, 60% and 88% for CfaEB, dmLT and SLA, respectively, maintained or even maximized immune responses, with SE and LSQ the best formulations. The DoE approach can help determine the best vaccine composition with a limited number of experiments and may accelerate development of multi-antigen/component ETEC vaccines.
Collapse
|
33
|
Agarwal Y, Beatty C, Ho S, Thurlow L, Das A, Kelly S, Castronova I, Salunke R, Biradar S, Yeshi T, Richardson A, Bility M. Development of humanized mouse and rat models with full-thickness human skin and autologous immune cells. Sci Rep 2020; 10:14598. [PMID: 32884084 PMCID: PMC7471691 DOI: 10.1038/s41598-020-71548-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 08/13/2020] [Indexed: 12/12/2022] Open
Abstract
The human skin is a significant barrier for protection against pathogen transmission. Rodent models used to investigate human-specific pathogens that target the skin are generated by introducing human skin grafts to immunocompromised rodent strains. Infection-induced immunopathogenesis has been separately studied in humanized rodent models developed with human lymphoid tissue and hematopoietic stem cell transplants. Successful co-engraftment of human skin, autologous lymphoid tissues, and autologous immune cells in a rodent model has not yet been achieved, though it could provide a means of studying the human immune response to infection in the human skin. Here, we introduce the human Skin and Immune System (hSIS)-humanized NOD-scid IL2Rγnull (NSG) mouse and Sprague–Dawley-Rag2tm2hera Il2rγtm1hera (SRG) rat models, co-engrafted with human full-thickness fetal skin, autologous fetal lymphoid tissues, and autologous fetal liver-derived hematopoietic stem cells. hSIS-humanized rodents demonstrate the development of human full-thickness skin, along with autologous lymphoid tissues, and autologous immune cells. These models also support human skin infection following intradermal inoculation with community-associated methicillin-resistant Staphylococcus aureus. The co-engraftment of these human skin and immune system components into a single humanized rodent model could provide a platform for studying human skin infections.
Collapse
Affiliation(s)
- Yash Agarwal
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Cole Beatty
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Sara Ho
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Lance Thurlow
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, USA
| | - Antu Das
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Samantha Kelly
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Isabella Castronova
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Rajeev Salunke
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | - Shivkumar Biradar
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA
| | | | - Anthony Richardson
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, USA
| | - Moses Bility
- Department of Infectious Diseases and Microbiology, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
34
|
Schnyder JL, De Pijper CA, Garcia Garrido HM, Daams JG, Goorhuis A, Stijnis C, Schaumburg F, Grobusch MP. Fractional dose of intradermal compared to intramuscular and subcutaneous vaccination - A systematic review and meta-analysis. Travel Med Infect Dis 2020; 37:101868. [PMID: 32898704 PMCID: PMC7474844 DOI: 10.1016/j.tmaid.2020.101868] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Vaccine supply shortages are of global concern. We hypothesise that intradermal (ID) immunisation as an alternative to standard routes might augment vaccine supply utilisation without loss of vaccine immunogenicity and efficacy. METHODS We conducted a systematic review and meta-analysis searching Medline, Embase and Web of Science databases. Studies were included if: licensed, currently available vaccines were used; fractional dose of ID was compared to IM or SC immunisation; primary immunisation schedules were evaluated; immunogenicity, safety data and/or cost were reported. We calculated risk differences (RD). Studies were included in meta-analysis if: a pre-defined immune correlate of protection was assessed; WHO-recommend schedules and antigen doses were used in the control group; the same schedule was applied to both ID and control groups (PROSPERO registration no. CRD42020151725). RESULTS The primary search yielded 5,873 articles, of which 156 articles were included; covering 12 vaccines. Non-inferiority of immunogenicity with 20-60% of antigen used with ID vaccines was demonstrated for influenza (H1N1: RD -0·01; 95% CI -0·02, 0·01; I2 = 55%, H2N3: RD 0·00; 95% CI -0·01, 0·01; I2 = 0%, B: RD -0·00; 95% CI -0·02, 0·01; I2 = 72%), rabies (RD 0·00; 95% CI -0·02, 0·02; I2 = 0%), and hepatitis B vaccines (RD -0·01; 95% CI -0·04, 0·02; I2 = 20%). Clinical trials on the remaining vaccines yielded promising results, but are scarce. CONCLUSIONS There is potential for inoculum/antigen dose-reduction by using ID immunisation as compared to standard routes of administration for some vaccines (e.g. influenza, rabies). When suitable, vaccine trials should include an ID arm.
Collapse
Affiliation(s)
- Jenny L Schnyder
- Centre for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, Amsterdam Infection & Immunity, Amsterdam Public Health, University of Amsterdam, Meibergdreef 9, 1100 DD, Amsterdam, Netherlands
| | - Cornelis A De Pijper
- Centre for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, Amsterdam Infection & Immunity, Amsterdam Public Health, University of Amsterdam, Meibergdreef 9, 1100 DD, Amsterdam, Netherlands
| | - Hannah M Garcia Garrido
- Centre for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, Amsterdam Infection & Immunity, Amsterdam Public Health, University of Amsterdam, Meibergdreef 9, 1100 DD, Amsterdam, Netherlands
| | - Joost G Daams
- Medical Library, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, Netherlands
| | - Abraham Goorhuis
- Centre for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, Amsterdam Infection & Immunity, Amsterdam Public Health, University of Amsterdam, Meibergdreef 9, 1100 DD, Amsterdam, Netherlands
| | - Cornelis Stijnis
- Centre for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, Amsterdam Infection & Immunity, Amsterdam Public Health, University of Amsterdam, Meibergdreef 9, 1100 DD, Amsterdam, Netherlands
| | - Frieder Schaumburg
- Institute of Medical Microbiology, University Hospital Münster, Domagkstraße 10, 48149, Münster, Germany
| | - Martin P Grobusch
- Centre for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, Amsterdam Infection & Immunity, Amsterdam Public Health, University of Amsterdam, Meibergdreef 9, 1100 DD, Amsterdam, Netherlands.
| |
Collapse
|
35
|
Akhtar N, Singh V, Yusuf M, Khan RA. Non-invasive drug delivery technology: development and current status of transdermal drug delivery devices, techniques and biomedical applications. ACTA ACUST UNITED AC 2020; 65:243-272. [PMID: 31926064 DOI: 10.1515/bmt-2019-0019] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 08/30/2019] [Indexed: 12/25/2022]
Abstract
Pay-load deliveries across the skin barrier to the systemic circulation have been one of the most challenging delivery options. Necessitated requirements of the skin and facilitated skin layer cross-over delivery attempts have resulted in development of different non-invasive, non-oral methods, devices and systems which have been standardized, concurrently used and are in continuous upgrade and improvements. Iontophoresis, electroporation, sonophoresis, magnetophoresis, dermal patches, nanocarriers, needled and needle-less shots, and injectors are among some of the methods of transdermal delivery. The current review covers the current state of the art, merits and shortcomings of the systems, devices and transdermal delivery patches, including drugs' and other payloads' passage facilitation techniques, permeation and absorption feasibility studies, as well as physicochemical properties affecting the delivery through different transdermal modes along with examples of drugs, vaccines, genes and other payloads.
Collapse
Affiliation(s)
- Naseem Akhtar
- Department of Pharmaceutics, College of Pharmacy,Buraydah Colleges, PO Box 31717, Qassim 51418, Saudi Arabia
| | - Varsha Singh
- Manav Rachna International University (MRIU) and Manav Rachna International Institute of Research and Study (MRIIRS), Faridabad, HR 121 001, India
| | - Mohammad Yusuf
- College of Pharmacy, University of Taif, Taif Al-Haweiah, Taif, Saudi Arabia.https://orcid.org/0000-0003- 1417-7774
| | - Riaz A Khan
- Manav Rachna International University (MRIU) and Manav Rachna International Institute of Research and Study (MRIIRS), Faridabad, HR 121 001, India.,Department of Medicinal Chemistry, College of Pharmacy, Qassim University, Qassim 51452, Saudi Arabia
| |
Collapse
|
36
|
Gutiérrez RL, Porter CK, Jarell A, Alcala A, Riddle MS, Turiansky GW. A grading system for local skin reactions developed for clinical trials of an intradermal and transcutaneous ETEC vaccine. Vaccine 2020; 38:3773-3779. [PMID: 32253098 DOI: 10.1016/j.vaccine.2020.02.079] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 02/24/2020] [Accepted: 02/26/2020] [Indexed: 11/15/2022]
Abstract
BACKGROUND Trials assessing the safety of novel vaccine candidates are essential in the evaluation and development of candidate vaccines. Immunogenicity and dose-sparing features of vaccination approaches which target skin and associated tissues have garnered increased interest; for enteric vaccines, cutaneous vaccination has been of particular interest. Cutaneous vaccine site reactions are among the most common and visible vaccine related adverse events (AEs) when skin routes are used. Regulatory guidelines governing classification of severity focus on functional impact but are insufficient to characterize a spectrum of skin reaction and allow for comparisons of routes, doses and products with similar local cutaneous AEs. OBJECTIVES Our group developed a grading scale to evaluate and compare cutaneous vaccine site reactions ahead of early-phase clinical trials of intradermal (ID) and transcutaneous immunization (TCI) with enterotoxigenic E.coli (ETEC) vaccine candidates (adhesin-based vaccine co-administered with LTR192G). We reviewed existing methods for characterizing the appearance and severity of local vaccine site reactions following TCI and ID vaccination and devised a standardized vaccine site appearance grading scale (VSAGS) for use in the clinical development of novel ETEC vaccine candidates which focused on pathophysiologic manifestation of skin findings. RESULTS Available data from published reports revealed erythematous papules and pruritus were the most common local AEs associated with TCI. Frequency of reactions varied notably across studies as did TCI vaccination methodologies and products. ID vaccination commonly results in erythema and induration at the vaccine site as well as pigmentation changes. There was no published methodology to characterize the spectrum of dermatologic findings. CONCLUSION ID and TCI vaccination are associated with a largely predictable range of cutaneous AEs. A grading scale focused on the appearance of cutaneous changes was useful in comparing cutaneous AEs. A standardized grading scale will facilitate documentation and comparison of cutaneous AEs.
Collapse
Affiliation(s)
- Ramiro L Gutiérrez
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, United States.
| | - Chad K Porter
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, United States.
| | - Abel Jarell
- Dermatology Department, Walter Reed National Military Medical Center, Bethesda, MD, United States
| | - Ashley Alcala
- Enteric Diseases Department, Naval Medical Research Center, Silver Spring, MD, United States
| | - Mark S Riddle
- Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - George W Turiansky
- Uniformed Services University of the Health Sciences, Bethesda, MD, United States.
| |
Collapse
|
37
|
A new approach for therapeutic vaccination against chronic HBV infections. Vaccine 2020; 38:3105-3120. [DOI: 10.1016/j.vaccine.2020.02.063] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 02/14/2020] [Accepted: 02/20/2020] [Indexed: 12/11/2022]
|
38
|
Kashiwagi S. Laser adjuvant for vaccination. FASEB J 2020; 34:3485-3500. [PMID: 31994227 DOI: 10.1096/fj.201902164r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 01/09/2020] [Accepted: 01/09/2020] [Indexed: 12/14/2022]
Abstract
The use of an immunologic adjuvant to augment the immune response is essential for modern vaccines which are relatively ineffective on their own. In the past decade, researchers have been consistently reporting that skin treatment with a physical parameter, namely laser light, augments the immune response to vaccine and functions as an immunologic adjuvant. This "laser adjuvant" has numerous advantages over the conventional chemical or biological agents; it is free from cold chain storage, hypodermic needles, biohazardous sharp waste, irreversible formulation with vaccine antigen, undesirable biodistribution in vital organs, or unknown long-term toxicity. Since vaccine formulations are given to healthy populations, these characteristics render the "laser adjuvant" significant advantages for clinical use and open a new developmental path for a safe and effective vaccine. In addition, laser technology has been used in the clinic for more than three decades and is therefore technically matured and has been proved to be safe. Currently, four classes of laser adjuvant have been reported; ultrashort pulsed, non-pulsed, non-ablative fractional, and ablative fractional lasers. Since each class of the laser adjuvant shows a distinct mechanism of action, a proper choice is necessary to craft an effective vaccine formulation toward a desired clinical benefit for a clinical vaccine to maximize protection. In addition, data also suggest that further improvement in the efficacy is possible when a laser adjuvant is combined with chemical or biological adjuvant(s). To realize these goals, further efforts to uncover the molecular mechanisms of action of the laser adjuvants is warranted. This review provides a summary and comments of the recent updates in the laser adjuvant technology.
Collapse
Affiliation(s)
- Satoshi Kashiwagi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| |
Collapse
|
39
|
Joubert IA, Kovacs D, Scheiblhofer S, Winter P, Korotchenko E, Strandt H, Weiss R. Mast cells and γδ T cells are largely dispensable for adaptive immune responses after laser-mediated epicutaneous immunization. Vaccine 2020; 38:1015-1024. [DOI: 10.1016/j.vaccine.2019.11.051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/06/2019] [Accepted: 11/19/2019] [Indexed: 12/21/2022]
|
40
|
Chuaychoo B, Kositanont U, Niyomthong P, Rittayamai N, Srisuma S, Rattanasaengloet K, Wongsrisakunkaew W, Thongam J, Songserm T. Comparison of immunogenicity between intradermal and intramuscular injections of repeated annual identical influenza virus strains post-pandemic (2011-2012) in COPD patients. Hum Vaccin Immunother 2019; 16:1371-1379. [PMID: 31770051 DOI: 10.1080/21645515.2019.1692559] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
We compared the antibody responses and persistence of the reduced-dose, 9 µg hemagglutinin (HA)/strain intradermal (ID) injection via the Mantoux technique and the 15 μg HA/strain intramuscular (IM) injection of the repeated annual identical trivalent, inactivated, split-virion vaccine 2011-2012 in chronic obstructive pulmonary disease (COPD) patients. Eighty patients were randomized to ID (n = 41) and IM (n = 39) groups. Four weeks post-vaccination, the antibody responses of the two groups were similar; those for influenza A(H1N1)pdm09 and influenza A(H3N2)-but not influenza B-met the criteria of the Committee for Proprietary Medicinal Products (CPMP). The antibody responses for influenza A(H1N1)pdm09 rapidly declined in both groups, especially with the ID injection, whereas those for influenza A(H3N2) maintained above the CPMP criteria throughout 12 months post-vaccination. The geometric mean titres for influenza A(H1N1)pdm09 persisted above the protective threshold (≥ 40) until 6 months post-vaccination in both the ID and IM groups. The seroprotection rates of the ID and IM groups were above 60% until 3 months and 6 months post-vaccination, respectively. In conclusion, the 9 μg HA/strain ID injection of vaccine 2011-2012 elicited antibody responses similar to the standard dose of 15 μg of the HA/strain IM injection at 4 weeks post-vaccination. However, the antibody responses for influenza A(H1N1)pdm09 rapidly declined, especially in the case of the ID injection, whereas they were comparable for influenza A(H3N2). Additional strategies for increasing vaccine durability should be considered, especially for new pandemic strains affecting elderly COPD patients.
Collapse
Affiliation(s)
- Benjamas Chuaychoo
- Division of Respiratory Disease and Tuberculosis, Department of Medicine, Faculty of Medicine Siriraj Hospital , Bangkok, Thailand
| | - Uraiwan Kositanont
- Department of Microbiology, Faculty of Medicine Siriraj Hospital , Bangkok, Thailand.,Faculty of Public Health, Thammasat University , Bangkok, Thailand
| | - Parichat Niyomthong
- Division of Respiratory Disease and Tuberculosis, Department of Medicine, Faculty of Medicine Siriraj Hospital , Bangkok, Thailand.,Medicine Unit, Phrae Medical Education Center, Naresuan University , Phitsanulok, Thailand
| | - Nuttapol Rittayamai
- Division of Respiratory Disease and Tuberculosis, Department of Medicine, Faculty of Medicine Siriraj Hospital , Bangkok, Thailand
| | - Sorachai Srisuma
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok, Thailand
| | - Kanokwan Rattanasaengloet
- Division of Respiratory Disease and Tuberculosis, Department of Medicine, Faculty of Medicine Siriraj Hospital , Bangkok, Thailand
| | - Walaiporn Wongsrisakunkaew
- Division of Respiratory Disease and Tuberculosis, Department of Medicine, Faculty of Medicine Siriraj Hospital , Bangkok, Thailand
| | - Julalux Thongam
- Department of Physiology, Faculty of Medicine Siriraj Hospital, Mahidol University , Bangkok, Thailand
| | - Thaweesak Songserm
- Department of Veterinary Pathology, Kamphaeng Saen, Kasetsart University , Nakhon Pathom, Thailand
| |
Collapse
|
41
|
Langedijk AC, De Pijper CA, Spijker R, Holman R, Grobusch MP, Stijnis C. Rabies Antibody Response After Booster Immunization: A Systematic Review and Meta-analysis. Clin Infect Dis 2019; 67:1932-1947. [PMID: 29788204 DOI: 10.1093/cid/ciy420] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/10/2018] [Indexed: 12/24/2022] Open
Abstract
Although fatal once symptomatic, rabies is preventable by administration of pre- and post-exposure vaccines. International guidelines suggest lifelong protection by a pre-exposure vaccination scheme followed by timely post-exposure vaccines. Rapidity and magnitude of the antibody recall response after booster inoculation are essential, as many people have been previously immunized a long time ago. The objective of this study was therefore to systematically review the evidence on the boostability of rabies immunization to date. We included 36 studies, of which 19 studies were suitable for meta-analysis. Reduced antibody levels were found after intradermal primary schedules as compared to intramuscular schedules. However, responses after booster immunization were adequate for both routes. Although studies showed that antibody levels decline over time, adequate booster responses were still retained over long time intervals indicating that post-exposure treatment is effective without extra measures after long periods of time.
Collapse
Affiliation(s)
- Annefleur C Langedijk
- Center for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, The Netherlands
| | - Cornelis A De Pijper
- Center for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, The Netherlands
| | - Rene Spijker
- Medical Library, Academic Medical Center, University of Amsterdam, The Netherlands.,Cochrane Netherlands, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, The Netherlands
| | - Rebecca Holman
- Clinical Research Unit, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Martin P Grobusch
- Center for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, The Netherlands
| | - Cornelis Stijnis
- Center for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, The Netherlands
| |
Collapse
|
42
|
Reliable and Standardized Animal Models to Study the Pathogenesis of Bluetongue and Schmallenberg Viruses in Ruminant Natural Host Species with Special Emphasis on Placental Crossing. Viruses 2019; 11:v11080753. [PMID: 31443153 PMCID: PMC6722754 DOI: 10.3390/v11080753] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/19/2019] [Accepted: 08/13/2019] [Indexed: 01/03/2023] Open
Abstract
Starting in 2006, bluetongue virus serotype 8 (BTV8) was responsible for a major epizootic in Western and Northern Europe. The magnitude and spread of the disease were surprisingly high and the control of BTV improved significantly with the marketing of BTV8 inactivated vaccines in 2008. During late summer of 2011, a first cluster of reduced milk yield, fever, and diarrhoea was reported in the Netherlands. Congenital malformations appeared in March 2012 and Schmallenberg virus (SBV) was identified, becoming one of the very few orthobunyaviruses distributed in Europe. At the start of both epizootics, little was known about the pathogenesis and epidemiology of these viruses in the European context and most assumptions were extrapolated based on other related viruses and/or other regions of the World. Standardized and repeatable models potentially mimicking clinical signs observed in the field are required to study the pathogenesis of these infections, and to clarify their ability to cross the placental barrier. This review presents some of the latest experimental designs for infectious disease challenges with BTV or SBV. Infectious doses, routes of infection, inoculum preparation, and origin are discussed. Particular emphasis is given to the placental crossing associated with these two viruses.
Collapse
|
43
|
Simmons JA, Davis J, Thomas J, Lopez J, Le Blanc A, Allison H, Slook H, Lewis P, Holtz J, Fisher P, Broderick KE, Marston JO. Characterization of skin blebs from intradermal jet injection: Ex-vivo studies. J Control Release 2019; 307:200-210. [PMID: 31252035 DOI: 10.1016/j.jconrel.2019.06.032] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 05/14/2019] [Accepted: 06/24/2019] [Indexed: 01/27/2023]
Abstract
This paper presents results from an ex-vivo study of intradermal jet injections, which is an attractive method to achieve both needle-free and fractional dose delivery of vaccines. Due to the fact that fluid properties of many novel therapeutics and vaccines can vary significantly, a key parameter for our study is the fluid viscosity, whilst the main focus is on determining the best correlation between the delivered volume and geometrical dimensions of the fluid deposit. For this we use a combination of top-view (skin wheal), underside (below the dermis), and cross-section (true skin bleb) perspectives and find that the top-view alone, as done in clinical practice, is insufficient to estimate the volume deposited in the dermis. Overall, the best correlation is found between the injection volume and cross-sectional diameter, however there is significant variation amongst the different fluids. For mean injection volumes of 60 μL the mean bleb diameter is ≈8 mm, with mean aspect ratio h¯/d=0.38, indicating the blebs are mostly oblate. However, the shape varies with viscosity and the higher viscosity does not spread laterally to the same degree as lower viscosity fluids. In addition, our high-speed video observations of the injection process, reveal some interesting dynamics of the jet injection method, and we modeled the bleb growth with an exponential saturation.
Collapse
Affiliation(s)
- Jonathan A Simmons
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States of America; Department of Mechanical Engineering, Texas Tech University, Lubbock, TX 79409, United States of America
| | - Justin Davis
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States of America
| | - James Thomas
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States of America
| | - Juan Lopez
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States of America
| | - Andrew Le Blanc
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States of America
| | - Haley Allison
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States of America
| | - Haley Slook
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States of America
| | - Paul Lewis
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States of America
| | - Joshua Holtz
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States of America
| | - Paul Fisher
- Inovio Pharmaceuticals, 10480 Wateridge Circle, San Diego, CA 92121, United States of America
| | - Kate E Broderick
- Inovio Pharmaceuticals, 10480 Wateridge Circle, San Diego, CA 92121, United States of America
| | - Jeremy O Marston
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States of America.
| |
Collapse
|
44
|
Cheeseman HM, Day S, McFarlane LR, Fleck S, Miller A, Cole T, Sousa-Santos N, Cope A, Cizmeci D, Tolazzi M, Hwekwete E, Hannaman D, Kratochvil S, McKay PF, Chung AW, Kent SJ, Cook A, Scarlatti G, Abraham S, Combadiere B, McCormack S, Lewis DJ, Shattock RJ. Combined Skin and Muscle DNA Priming Provides Enhanced Humoral Responses to a Human Immunodeficency Virus Type 1 Clade C Envelope Vaccine. Hum Gene Ther 2019; 29:1011-1028. [PMID: 30027768 PMCID: PMC6214652 DOI: 10.1089/hum.2018.075] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Intradermal (i.d.) and intramuscular (i.m.) injections when administered with or without electroporation (EP) have the potential to tailor the immune response to DNA vaccination. This Phase I randomized controlled clinical trial in human immunodeficiency virus type 1–negative volunteers investigated whether the site and mode of DNA vaccination influences the quality of induced cellular and humoral immune responses following the DNA priming phase and subsequent protein boost with recombinant clade C CN54 gp140. A strategy of concurrent i.d. and i.m. DNA immunizations administered with or without EP was adopted. Subtle differences were observed in the shaping of vaccine-induced virus-specific CD4+ and CD8+ T cell–mediated immune responses between groups receiving: i.d.EP + i.m., i.d. + i.m.EP, and i.d.EP + i.m.EP regimens. The DNA priming phase induced 100% seroconversion in all of the groups. A single, non-adjuvanted protein boost induced a rapid and profound increase in binding antibodies in all groups, with a trend for higher responses in i.d.EP + i.m.EP. The magnitude of antigen-specific binding immunoglobulin G correlated with neutralization of closely matched clade C 93MW965 virus and Fc-dimer receptor binding (FcγRIIa and FcγRIIIa). These results offer new perspectives on the use of combined skin and muscle DNA immunization in priming humoral and cellular responses to recombinant protein.
Collapse
Affiliation(s)
- Hannah Mary Cheeseman
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Suzanne Day
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Leon Robert McFarlane
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Sue Fleck
- 2 Medical Research Council Clinical Trials Unit at UCL, University College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Aleisha Miller
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Tom Cole
- 3 Imperial Clinical Research Facility, Hammersmith Hospital, Imperial College Healthcare NHS Trust, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Nelson Sousa-Santos
- 3 Imperial Clinical Research Facility, Hammersmith Hospital, Imperial College Healthcare NHS Trust, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Alethea Cope
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Deniz Cizmeci
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Monica Tolazzi
- 4 Viral Evolution and Transmission Unit, Division of Immunology, Transplant and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Edith Hwekwete
- 3 Imperial Clinical Research Facility, Hammersmith Hospital, Imperial College Healthcare NHS Trust, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Drew Hannaman
- 5 Ichor Medical Systems, Inc., San Diego, California; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Sven Kratochvil
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Paul Francis McKay
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Amy W Chung
- 6 Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, and UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Stephen J Kent
- 6 Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, and UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France .,7 ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, Australia; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France .,8 Melbourne Sexual Health Centre, Department of Infectious Diseases, Alfred Health, Central Clinical School, Monash University , Melbourne, Australia; and UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Adrian Cook
- 2 Medical Research Council Clinical Trials Unit at UCL, University College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Gabriella Scarlatti
- 4 Viral Evolution and Transmission Unit, Division of Immunology, Transplant and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Sonya Abraham
- 3 Imperial Clinical Research Facility, Hammersmith Hospital, Imperial College Healthcare NHS Trust, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Behazine Combadiere
- 9 Sorbonne Universités, UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Sheena McCormack
- 2 Medical Research Council Clinical Trials Unit at UCL, University College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - David John Lewis
- 3 Imperial Clinical Research Facility, Hammersmith Hospital, Imperial College Healthcare NHS Trust, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| | - Robin John Shattock
- 1 Department of Medicine, Section of Virology, Group of Mucosal Infection and Immunity, Imperial College London, London, United Kingdom; UPMC Univ Paris 06, INSERM, U1135, CNRS, ERL 8255, Centre d'Immunologie et des Maladies Infectieuses (CIMI-Paris), Paris, France
| |
Collapse
|
45
|
Marston JO, Lacerda CMR. Characterization of jet injection efficiency with mouse cadavers. J Control Release 2019; 305:101-109. [PMID: 31112720 DOI: 10.1016/j.jconrel.2019.05.023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 04/29/2019] [Accepted: 05/14/2019] [Indexed: 01/29/2023]
Abstract
Needle-free drug delivery is highly sought after for reduction in sharps waste, prevention of needle-stick injuries, and potential for improved drug dispersion and uptake. Whilst there is a wealth of literature on the array of different delivery methods, jet injection is proposed as the sole candidate for delivery of viscous fluids, which is especially relevant with the advent of DNA-based vaccines. The focus of this study was therefore to assess the role of viscosity and jet configuration (i.e. stand-off relative to the skin) upon injection efficiency for a fixed spring-loaded system (Bioject ID Pen). We performed this assessment in the context of mouse cadavers and found that the dominant factor in determining success rates was the time from euthanasia, which was taken as a proxy for the stiffness of the underlying tissue. For overall injection efficiency, ANOVA tests indicated that stiffness was highly significant (P < < 0.001), stand-off was moderately significant (P < 0.1), and viscosity was insignificant. In contrast, both viscosity and standoff were found to be significant (P < 0.01) when evaluating the percentage delivered intradermally. Using high-resolution micro-computed tomography (μ-CT), we also determined the depth and overall dispersion pattern immediately after injection.
Collapse
Affiliation(s)
- Jeremy O Marston
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States of America.
| | - Carla M R Lacerda
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX 79409, United States of America
| |
Collapse
|
46
|
Schaumburg F, De Pijper CA, Grobusch MP. Intradermal travel vaccinations-when less means more. Travel Med Infect Dis 2019; 28:3-5. [PMID: 30878310 DOI: 10.1016/j.tmaid.2019.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 01/04/2023]
Affiliation(s)
- Frieder Schaumburg
- Institute of Medical Microbiology, University Hospital Münster, Domagkstraße 10, 48149, Münster, Germany.
| | - Cornelis A De Pijper
- Center for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Infection & Immunity, Amsterdam Public Health, Meibergdreef 9, 1100, DD, Amsterdam, Netherlands
| | - Martin P Grobusch
- Center for Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Division of Internal Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam Infection & Immunity, Amsterdam Public Health, Meibergdreef 9, 1100, DD, Amsterdam, Netherlands
| |
Collapse
|
47
|
Criscuolo E, Caputo V, Diotti RA, Sautto GA, Kirchenbaum GA, Clementi N. Alternative Methods of Vaccine Delivery: An Overview of Edible and Intradermal Vaccines. J Immunol Res 2019; 2019:8303648. [PMID: 30949518 PMCID: PMC6425294 DOI: 10.1155/2019/8303648] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/08/2019] [Accepted: 02/14/2019] [Indexed: 01/26/2023] Open
Abstract
Vaccines are recognized worldwide as one of the most important tools for combating infectious diseases. Despite the tremendous value conferred by currently available vaccines toward public health, the implementation of additional vaccine platforms is also of key importance. In fact, currently available vaccines possess shortcomings, such as inefficient triggering of a cell-mediated immune response and the lack of protective mucosal immunity. In this regard, recent work has been focused on vaccine delivery systems, as an alternative to injectable vaccines, to increase antigen stability and improve overall immunogenicity. In particular, novel strategies based on edible or intradermal vaccine formulations have been demonstrated to trigger both a systemic and mucosal immune response. These novel vaccination delivery systems offer several advantages over the injectable preparations including self-administration, reduced cost, stability, and elimination of a cold chain. In this review, the latest findings and accomplishments regarding edible and intradermal vaccines are described in the context of the system used for immunogen expression, their molecular features and capacity to induce a protective systemic and mucosal response.
Collapse
Affiliation(s)
- E. Criscuolo
- Microbiology and Virology Unit, “Vita-Salute San Raffaele” University, Milan, Italy
| | - V. Caputo
- Microbiology and Virology Unit, “Vita-Salute San Raffaele” University, Milan, Italy
- Pomona Ricerca S.r.l., Turin, Italy
| | - R. A. Diotti
- Microbiology and Virology Unit, “Vita-Salute San Raffaele” University, Milan, Italy
- Pomona Ricerca S.r.l., Turin, Italy
| | - G. A. Sautto
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | | | - N. Clementi
- Microbiology and Virology Unit, “Vita-Salute San Raffaele” University, Milan, Italy
| |
Collapse
|
48
|
Gelfand JA, Nazarian RM, Kashiwagi S, Brauns T, Martin B, Kimizuka Y, Korek S, Botvinick E, Elkins K, Thomas L, Locascio J, Parry B, Kelly KM, Poznansky MC. A pilot clinical trial of a near-infrared laser vaccine adjuvant: safety, tolerability, and cutaneous immune cell trafficking. FASEB J 2019; 33:3074-3081. [PMID: 30192655 PMCID: PMC6338655 DOI: 10.1096/fj.201801095r] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/06/2018] [Indexed: 12/31/2022]
Abstract
Many vaccines require adjuvants to enhance immunogenicity, but there are few safe and effective intradermal (i.d.) adjuvants. Murine studies have validated the potency of laser illumination of skin as an adjuvant for i.d. vaccination with advantages over traditional adjuvants. We report a pilot clinical trial of low-power, continuous-wave, near-infrared laser adjuvant treatment, representing the first human trial of the safety, tolerability, and cutaneous immune cell trafficking changes produced by the laser adjuvant. In this trial we demonstrated a maximum tolerable energy dose of 300 J/cm2 to a spot on the lower back. The irradiated spot was biopsied 4 h later, as was a control spot. Paired biopsies were submitted for histomorphologic and immunohistochemical evaluation in a blinded fashion as well as quantitative PCR analysis for chemokines and cytokines. Similar to prior murine studies, highly significant reductions in CD1a+ Langerhans cells in the dermis and CD11c+ dermal dendritic cells were observed, corresponding to the increased migratory activity of these cells; changes in the epidermis were not significant. There was no evidence of skin damage. The laser adjuvant is a safe, well-tolerated adjuvant for i.d. vaccination in humans and results in significant cutaneous immune cell trafficking.-Gelfand, J. A., Nazarian, R. M., Kashiwagi, S., Brauns, T., Martin, B., Kimizuka, Y., Korek, S., Botvinick, E., Elkins, K., Thomas, L., Locascio, J., Parry, B., Kelly, K. M., Poznansky, M. C. A pilot clinical trial of a near-infrared laser vaccine adjuvant: safety, tolerability, and cutaneous immune cell trafficking.
Collapse
Affiliation(s)
- Jeffrey A. Gelfand
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Rosalynn M. Nazarian
- Dermatopathology Unit, Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Satoshi Kashiwagi
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Timothy Brauns
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Brent Martin
- Department of Dermatology, University of California, Irvine, School of Medicine, Irvine, California, USA
| | - Yoshifumi Kimizuka
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Skylar Korek
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Elliot Botvinick
- Beckman Laser Institute, University of California, Irvine, School of Medicine, Irvine, California, USA
| | - Kristen Elkins
- Department of Dermatology, University of California, Irvine, School of Medicine, Irvine, California, USA
| | - Logan Thomas
- Department of Dermatology, University of California, Irvine, School of Medicine, Irvine, California, USA
| | - Joseph Locascio
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Blair Parry
- Emergency Department, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kristen M. Kelly
- Department of Dermatology, University of California, Irvine, School of Medicine, Irvine, California, USA
- Beckman Laser Institute, University of California, Irvine, School of Medicine, Irvine, California, USA
| | - Mark C. Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
49
|
Evaluating new rabies post-exposure prophylaxis (PEP) regimens or vaccines. Vaccine 2018; 37 Suppl 1:A88-A93. [PMID: 30471958 DOI: 10.1016/j.vaccine.2018.10.103] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 10/12/2018] [Accepted: 10/31/2018] [Indexed: 12/25/2022]
Abstract
The development of human rabies vaccines has evolved dramatically from the first crude nerve tissue vaccine produced then administered in the presence of Louis Pasteur in 1885. New cell culture technology has enabled highly potent and well-tolerated rabies vaccines to be produced that have reduced the volume and number of doses required to save human lives after exposure. However, these highly potent vaccines are still unaffordable to many patients living at risk of exposure on a daily basis. The cost of post-exposure prophylaxis (PEP) is not only related to the direct cost of rabies biologicals and equipment but is also associated with indirect costs that patients incur as a result of travel, loss of work time (income loss), and accommodation over the period of time that a PEP regimen requires to be completed. This paper summarizes the particular criteria that the SAGE Working Group and WHO personnel reviewed as part of the evaluation process for recommending the new one-week intradermal vaccination regimen (2-2-2-0-0) for rabies post-exposure prophylaxis. These criteria included: Cost-effectiveness; evaluation of number of doses; seroconversion after vaccination; efficacy; safety; and patient follow-up.
Collapse
|
50
|
Rosenberg RN, Fu M, Lambracht-Washington D. Intradermal active full-length DNA Aβ42 immunization via electroporation leads to high anti-Aβ antibody levels in wild-type mice. J Neuroimmunol 2018; 322:15-25. [PMID: 29958693 PMCID: PMC6192700 DOI: 10.1016/j.jneuroim.2018.05.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 11/24/2022]
Abstract
Aβ immunotherapies with anti-Aβ antibody responses have high potential as possible prevention treatment for Alzheimer's disease. We have previously shown that active DNA Aβ1-42 immunization via gene gun delivery led to a non-inflammatory immune response resulting in decreased Aβ levels in brains of an immunized AD mouse model. To make DNA vaccination more applicable for clinical use, we used here intradermal electroporation. With fine tuning of the electropulse parameters, high antibody levels and low levels of inflammatory cytokines in the cellular immunoassays were observed. Full-length DNA Aβ1-42 immunization delivered via electroporation has potential to be used in the clinical setting.
Collapse
Affiliation(s)
- Roger N Rosenberg
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center Dallas, USA
| | - Min Fu
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center Dallas, USA
| | - Doris Lambracht-Washington
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center Dallas, USA.
| |
Collapse
|