1
|
Thomson-Luque R, Stabler TC, Fürle K, Silva JC, Daubenberger C. Plasmodium falciparum merozoite surface protein 1 as asexual blood stage malaria vaccine candidate. Expert Rev Vaccines 2024; 23:160-173. [PMID: 38100310 DOI: 10.1080/14760584.2023.2295430] [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: 09/25/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023]
Abstract
INTRODUCTION Malaria represents a public health challenge in tropical and subtropical regions, and currently deployed control strategies are likely insufficient to drive elimination of malaria. Development and improvement of malaria vaccines might be key to reduce disease burden. Vaccines targeting asexual blood stages of the parasite have shown limited efficacy when studied in human trials conducted over the past decades. AREAS COVERED Vaccine candidates based on the merozoite surface protein 1 (MSP1) were initially envisioned as one of the most promising approaches to provide immune protection against asexual blood-stage malaria. Successful immunization studies in monkey involved the use of the full-length MSP1 (MSP1FL) as vaccine construct. Vaccines using MSP1FL for immunization have the potential benefit of including numerous conserved B-cell and T-cell epitopes. This could result in improved parasite strain-transcending, protective immunity in the field. We review outcomes of clinical trials that utilized a variety of MSP1 constructs and formulations, including MSP1FL, either alone or in combination with other antigens, in both animal models and humans. EXPERT OPINION Novel approaches to analyze breadth and magnitude of effector functions of MSP1-targeting antibodies in volunteers undergoing experimental vaccination and controlled human malaria infection will help to define correlates of protective immunity.
Collapse
Affiliation(s)
- Richard Thomson-Luque
- Centre for Infectious Diseases-Parasitology, Heidelberg University Hospital, Heidelberg, Germany
- Sumaya-Biotech GmbH & Co. KG Heidelberg, Germany
| | - Thomas C Stabler
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
- University of Basel Basel, Switzerland
- Swiss Tropical and Public Health Institute Allschwil, Switzerland
| | - Kristin Fürle
- Centre for Infectious Diseases-Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Joana C Silva
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
- Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade NOVA de Lisboa (GHTM IHMT, UNL), Lisbon, Portugal
| | - Claudia Daubenberger
- University of Basel Basel, Switzerland
- Swiss Tropical and Public Health Institute Allschwil, Switzerland
| |
Collapse
|
2
|
Methodological advances in the design of peptide-based vaccines. Drug Discov Today 2022; 27:1367-1380. [DOI: 10.1016/j.drudis.2022.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/02/2021] [Accepted: 03/07/2022] [Indexed: 12/11/2022]
|
3
|
Nabi SU, Ali SI, Rather MA, Sheikh WM, Altaf M, Singh H, Mumtaz PT, Mishra NC, Nazir SU, Bashir SM. Organoids: A new approach in toxicity testing of nanotherapeutics. J Appl Toxicol 2021; 42:52-72. [PMID: 34060108 DOI: 10.1002/jat.4206] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 05/04/2021] [Accepted: 05/20/2021] [Indexed: 12/18/2022]
Abstract
Nanotechnology has revolutionized diverse fields, which include agriculture, the consumer market, medicine, and other fields. Widespread use of nanotechnology-based products has led to increased prevalence of these novel formulations in the environment, which has raised concerns regarding their deleterious effects. The application of nanotechnology-based formulations into clinical use is hampered by the lack of the availability of effective in vitro systems, which could accurately assess their in vivo toxic effects. A plethora of studies has shown the hazardous effects of nanoparticle-based formulations in two-dimensional in vitro cell cultures and animal models. These have some associated disadvantages when used for the evaluation of nano-toxicity. Organoid technology fills the space between existing two-dimensional cell line culture and in vivo models. The uniqueness of organoids over other systems for evaluating toxicity caused by nano-drug formulation includes them being a co-culture of diverse cell types, dynamic flow within them that simulates the actual flow of nanoparticles within biological systems, extensive cell-cell, cell-matrix interactions, and a tissue-like morphology. Thus, it mimics the actual tissue microenvironment and, subsequently, provides an opportunity to study drug metabolism and toxico-dynamics of nanotechnology-based novel formulations. The use of organoids in the evaluation of nano-drug toxicity is in its infancy. A limited number of studies conducted so far have shown good predictive value and efficiently significant data correlation with the clinical trials. In this review, we attempt to introduce organoids of the liver, lungs, brain, kidney intestine, and potential applications to evaluate toxicity caused by nanoparticles.
Collapse
Affiliation(s)
- Showkat Ul Nabi
- Large Animal Diagnostic Laboratory, Department of Clinical Veterinary Medicine, Ethics & Jurisprudence, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, Jammu and Kashmir, India
| | - Sofi Imtiyaz Ali
- Biochemistry & Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, Jammu and Kashmir, India
| | - Muzafar Ahmad Rather
- Biochemistry & Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, Jammu and Kashmir, India
| | - Wajid Mohammad Sheikh
- Biochemistry & Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, Jammu and Kashmir, India
| | - Mehvish Altaf
- Department of Food Technology, Islamic University of Science & Technology, Awantipora, Pulwama, Jammu and Kashmir, India
| | - Hemant Singh
- Department of Polymer and Process Engineering, Indian Institute of Technology Roorkee, Roorkee, India
| | - Peerzada Tajamul Mumtaz
- Biochemistry & Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, Jammu and Kashmir, India
| | - Narayan Chandra Mishra
- Department of Polymer and Process Engineering, Indian Institute of Technology Roorkee, Roorkee, India
| | - Sheikh Uzma Nazir
- Biochemistry & Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, Jammu and Kashmir, India
| | - Showkeen Muzamil Bashir
- Biochemistry & Molecular Biology Lab, Division of Veterinary Biochemistry, Faculty of Veterinary Sciences and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology, Srinagar, Jammu and Kashmir, India
| |
Collapse
|
4
|
Virosome-based nanovaccines; a promising bioinspiration and biomimetic approach for preventing viral diseases: A review. Int J Biol Macromol 2021; 182:648-658. [PMID: 33862071 PMCID: PMC8049750 DOI: 10.1016/j.ijbiomac.2021.04.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 04/02/2021] [Accepted: 04/03/2021] [Indexed: 01/08/2023]
Abstract
Vaccination is the most effective means of controlling infectious disease-related morbidity and mortality. However, due to low immunogenicity of viral antigens, nanomedicine as a new opportunity in new generation of vaccine advancement attracted researcher encouragement. Virosome is a lipidic nanomaterial emerging as FDA approved nanocarriers with promising bioinspiration and biomimetic potency against viral infections. Virosome surface modification with critical viral fusion proteins is the cornerstone of vaccine development. Surface antigens at virosomes innovatively interact with targeted receptors on host cells that evoke humoral or cellular immune responses through antibody-producing B cell and internalization by endocytosis-mediated pathways. To date, several nanovaccine based on virosome formulations have been commercialized against widespread and life-threatening infections. Recently, Great efforts were made to fabricate a virosome-based vaccine platform against a new severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus. Thus, this review provides a novel overview of the virosome based nanovaccine production, properties, and application on the viral disease, especially its importance in SARS-CoV-2 vaccine discovery.
Collapse
|
5
|
González-Romo F, Picazo JJ. [Development of new vaccines]. Enferm Infecc Microbiol Clin 2015; 33:557-68. [PMID: 26341041 DOI: 10.1016/j.eimc.2015.06.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 06/10/2015] [Indexed: 01/01/2023]
Abstract
Recent and important advances in the fields of immunology, genomics, functional genomics, immunogenetics, immunogenomics, bioinformatics, microbiology, genetic engineering, systems biology, synthetic biochemistry, proteomics, metabolomics and nanotechnology, among others, have led to new approaches in the development of vaccines. The better identification of ideal epitopes, the strengthening of the immune response due to new adjuvants, and the search of new routes of vaccine administration, are good examples of advances that are already a reality and that will favour the development of more vaccines, their use in indicated population groups, or its production at a lower cost. There are currently more than 130 vaccines are under development against the more wished (malaria or HIV), difficult to get (CMV or RSV), severe re-emerging (Dengue or Ebola), increasing importance (Chagas disease or Leishmania), and nosocomial emerging (Clostridium difficile or Staphylococcus aureus) infectious diseases.
Collapse
Affiliation(s)
- Fernando González-Romo
- Servicio de Microbiología Clínica, Hospital Clínico San Carlos, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, España.
| | - Juan J Picazo
- Servicio de Microbiología Clínica, Hospital Clínico San Carlos, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, España
| |
Collapse
|
6
|
Soema PC, Rosendahl Huber SK, Willems GJ, Jiskoot W, Kersten GFA, Amorij JP. Influenza T-cell epitope-loaded virosomes adjuvanted with CpG as a potential influenza vaccine. Pharm Res 2014; 32:1505-15. [PMID: 25344321 PMCID: PMC4356889 DOI: 10.1007/s11095-014-1556-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 10/20/2014] [Indexed: 11/30/2022]
Abstract
Purpose Influenza CD8+ T-cell epitopes are conserved amongst influenza strains and can be recognized by influenza-specific cytotoxic T-cells (CTLs), which can rapidly clear infected cells. An influenza peptide vaccine that elicits these CTLs would therefore be an alternative to current influenza vaccines, which are not cross-reactive. However, peptide antigens are poorly immunogenic due to lack of delivery to antigen presenting cells, and therefore need additional formulation with a suitable delivery system. In this study, the potential of virosomes as a delivery system for an influenza T-cell peptide was investigated. Methods The conserved human HLA-A2.1 influenza T-cell epitope M158–66 was formulated with virosomes. The immunogenicity and protective effect of the peptide-loaded virosomes was assessed in HLA-A2 transgenic mice. Delivery properties of the virosomes were studied in mice and in in vitro dendritic cell cultures. Results Immunization of HLA-A2.1 transgenic C57BL/6 mice with peptide-loaded virosomes in the presence of the adjuvant CpG-ODN 1826 increased the number of peptide-specific CTLs. Vaccination with adjuvanted peptide-loaded virosomes reduced weight loss in mice after heterologous influenza infection. Association with fusion-active virosomes was found to be crucial for antigen uptake by dendritic cells, and subsequent induction of CTLs in mice. Conclusions These results show that influenza virosomes loaded with conserved influenza epitopes could be the basis of a novel cross-protective influenza vaccine. Electronic supplementary material The online version of this article (doi:10.1007/s11095-014-1556-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Peter C Soema
- Intravacc (Institute for Translational Vaccinology), Antonie van Leeuwenhoeklaan 9, 3721 MA, Bilthoven, The Netherlands,
| | | | | | | | | | | |
Collapse
|
7
|
Dey AK, Srivastava IK. Novel adjuvants and delivery systems for enhancing immune responses induced by immunogens. Expert Rev Vaccines 2014; 10:227-51. [DOI: 10.1586/erv.10.142] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
8
|
Moser C, Amacker M, Kammer AR, Rasi S, Westerfeld N, Zurbriggen R. Influenza virosomes as a combined vaccine carrier and adjuvant system for prophylactic and therapeutic immunizations. Expert Rev Vaccines 2014; 6:711-21. [DOI: 10.1586/14760584.6.5.711] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
9
|
Virosome presents multimodel cancer therapy without viral replication. BIOMED RESEARCH INTERNATIONAL 2013; 2013:764706. [PMID: 24369016 PMCID: PMC3866828 DOI: 10.1155/2013/764706] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 10/31/2013] [Indexed: 12/11/2022]
Abstract
A virosome is an artificial envelope that includes viral surface proteins and lacks the ability to produce progeny virus. Virosomes are able to introduce an encapsulated macromolecule into the cytoplasm of cells using their viral envelope fusion ability. Moreover, virus-derived factors have an adjuvant effect for immune stimulation. Therefore, many virosomes have been utilized as drug delivery vectors and adjuvants for cancer therapy. This paper introduces the application of virosomes for cancer treatment. In Particular, we focus on virosomes derived from the influenza and Sendai viruses which have been widely used for cancer therapy. Influenza virosomes have been mainly applied as drug delivery vectors and adjuvants. By contrast, the Sendai virosomes have been mainly applied as anticancer immune activators and apoptosis inducers.
Collapse
|
10
|
Vaccination for the prevention and treatment of breast cancer with special focus on Her-2/neu peptide vaccines. Breast Cancer Res Treat 2013; 138:1-12. [PMID: 23340862 DOI: 10.1007/s10549-013-2410-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2012] [Accepted: 01/07/2013] [Indexed: 01/18/2023]
Abstract
Immunologic interventions in a subset of breast cancer patients represent a well-established therapeutic approach reflecting individualized treatment modalities. Thus, the therapeutic administration of monoclonal antibodies targeting tumor-associated antigens (TAA), such as Her-2/neu, represents a milestone in cancer treatment. However, passive antibody administration suffers from several drawbacks, including frequency and long duration of treatment. These undesirables may be avoidable in an approach based on generating active immune responses against these same targets. Only recently has the significance of tumors in relation to their microenvironments been understood as essential for creating an effective cancer vaccine. In particular, the immune system plays an important role in suppressing or promoting tumor formation and growth. Therefore, activation of appropriate triggers (such as induction of Th1 cells, CD8+ T cells, and suppression of regulatory cells in combination with generation of antibodies with anti-tumor activity) is a desirable goal. Current vaccination approaches have concentrated on therapeutic vaccines using certain TAA. Many cancer antigens, including breast cancer antigens, have been described and also given priority ranking for use as vaccine antigens by the US National Cancer Institute. One of the TAA antigens which has been thoroughly examined in numerous trials is Her-2/neu. This review will discuss delivery systems for this antigen with special focus on T and B cell peptide vaccines. Attention will be given to their advantages and limitations, as well as the use of certain adjuvants to improve anti-cancer responses.
Collapse
|
11
|
Joris F, Manshian BB, Peynshaert K, De Smedt SC, Braeckmans K, Soenen SJ. Assessing nanoparticle toxicity in cell-based assays: influence of cell culture parameters and optimized models for bridging the in vitro–in vivo gap. Chem Soc Rev 2013; 42:8339-59. [DOI: 10.1039/c3cs60145e] [Citation(s) in RCA: 159] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
12
|
Cech PG, Aebi T, Abdallah MS, Mpina M, Machunda EB, Westerfeld N, Stoffel SA, Zurbriggen R, Pluschke G, Tanner M, Daubenberger C, Genton B, Abdulla S. Virosome-formulated Plasmodium falciparum AMA-1 & CSP derived peptides as malaria vaccine: randomized phase 1b trial in semi-immune adults & children. PLoS One 2011; 6:e22273. [PMID: 21799810 PMCID: PMC3142124 DOI: 10.1371/journal.pone.0022273] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2011] [Accepted: 06/22/2011] [Indexed: 11/29/2022] Open
Abstract
Background This trial was conducted to evaluate the safety and immunogenicity of two virosome formulated malaria peptidomimetics derived from Plasmodium falciparum AMA-1 and CSP in malaria semi-immune adults and children. Methods The design was a prospective randomized, double-blind, controlled, age-deescalating study with two immunizations. 10 adults and 40 children (aged 5–9 years) living in a malaria endemic area were immunized with PEV3B or virosomal influenza vaccine Inflexal®V on day 0 and 90. Results No serious or severe adverse events (AEs) related to the vaccines were observed. The only local solicited AE reported was pain at injection site, which affected more children in the Inflexal®V group compared to the PEV3B group (p = 0.014). In the PEV3B group, IgG ELISA endpoint titers specific for the AMA-1 and CSP peptide antigens were significantly higher for most time points compared to the Inflexal®V control group. Across all time points after first immunization the average ratio of endpoint titers to baseline values in PEV3B subjects ranged from 4 to 15 in adults and from 4 to 66 in children. As an exploratory outcome, we found that the incidence rate of clinical malaria episodes in children vaccinees was half the rate of the control children between study days 30 and 365 (0.0035 episodes per day at risk for PEV3B vs. 0.0069 for Inflexal®V; RR = 0.50 [95%-CI: 0.29–0.88], p = 0.02). Conclusion These findings provide a strong basis for the further development of multivalent virosomal malaria peptide vaccines. Trial Registration ClinicalTrials.gov NCT00513669
Collapse
Affiliation(s)
- Patrick Georges Cech
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Thomas Aebi
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
- Bagamoyo Research and Training Unit, Ifakara Health Institute, Dar es Salaam, Tanzania
| | | | - Maxmillian Mpina
- Bagamoyo Research and Training Unit, Ifakara Health Institute, Dar es Salaam, Tanzania
| | - Ester Barnabas Machunda
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | | | | | | | - Gerd Pluschke
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Marcel Tanner
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Claudia Daubenberger
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
| | - Blaise Genton
- Swiss Tropical and Public Health Institute, Basel, Switzerland
- University of Basel, Basel, Switzerland
- Infectious Disease Service and Travel Clinic, Lausanne University Hospital, Lausanne, Switzerland
- * E-mail:
| | - Salim Abdulla
- Bagamoyo Research and Training Unit, Ifakara Health Institute, Dar es Salaam, Tanzania
| |
Collapse
|
13
|
Chauhan A, Swaleha Z, Ahmad N, Farazuddin M, Vasco A, Abida M, Mohammad O. Escheriosome mediated cytosolic delivery of Candida albicans cytosolic proteins induces enhanced cytotoxic T lymphocyte response and protective immunity. Vaccine 2011; 29:5424-33. [DOI: 10.1016/j.vaccine.2011.05.066] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 05/16/2011] [Accepted: 05/20/2011] [Indexed: 01/04/2023]
|
14
|
Affiliation(s)
| | - Marco Tamborrini
- Swiss Tropical & Public Health Institute, Socinstr. 57, 4002 Basel, Switzerland
- University of Basel, Petersplatz 1, 4003 Basel, Switzerland
| |
Collapse
|
15
|
Tamborrini M, Mueller MS, Stoffel SA, Westerfeld N, Vogel D, Boato F, Zurbriggen R, Robinson JA, Pluschke G. Design and pre-clinical profiling of a Plasmodium falciparum MSP-3 derived component for a multi-valent virosomal malaria vaccine. Malar J 2009; 8:314. [PMID: 20042100 PMCID: PMC2805693 DOI: 10.1186/1475-2875-8-314] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Accepted: 12/30/2009] [Indexed: 01/22/2023] Open
Abstract
Background Clinical profiling of two components for a synthetic peptide-based virosomal malaria vaccine has yielded promising results, encouraging the search for additional components for inclusion in a final multi-valent vaccine formulation. This report describes the immunological characterization of linear and cyclized synthetic peptides comprising amino acids 211-237 of Plasmodium falciparum merozoite surface protein (MSP-3). Methods These peptides were coupled to phosphatidylethanolamine (PE); the conjugates were intercalated into immunopotentiating reconstituted influenza virosomes (IRIVs) and then used for immunizations in mice to evaluate their capacity to elicit P. falciparum cross-reactive antibodies. Results While all MSP-3-derived peptides were able to elicit parasite-binding antibodies, stabilization of turn structures by cyclization had no immune-enhancing effect. Therefore, further pre-clinical profiling was focused on FB-12, a PE conjugate of the linear peptide. Consistent with the immunological results obtained in mice, all FB-12 immunized rabbits tested seroconverted and consistently elicited antibodies that interacted with blood stage parasites. It was observed that a dose of 50 μg was superior to a dose of 10 μg and that influenza pre-existing immunity improved the immunogenicity of FB-12 in rabbits. FB-12 production was successfully up-scaled and the immunogenicity of a vaccine formulation, produced according to the rules of Good Manufacturing Practice (GMP), was tested in mice and rabbits. All animals tested developed parasite-binding antibodies. Comparison of ELISA and IFA titers as well as the characterization of a panel of anti-FB-12 monoclonal antibodies indicated that at least the majority of antibodies specific for the virosomally formulated synthetic peptide were parasite cross-reactive. Conclusion These results reconfirm the suitability of IRIVs as a carrier/adjuvant system for the induction of strong humoral immune responses against a wide range of synthetic peptide antigens. The virosomal formulation of the FB-12 peptidomimetic is suitable for use in humans and represents a candidate component for a virosomal multi-valent malaria subunit vaccine.
Collapse
Affiliation(s)
- Marco Tamborrini
- Swiss Tropical Institute, Molecular Immunology, CH-4002 Basel, Switzerland.
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Faisal SM, Yan W, McDonough SP, Chang CF, Pan MJ, Chang YF. Leptosome-entrapped leptospiral antigens conferred significant higher levels of protection than those entrapped with PC-liposomes in a hamster model. Vaccine 2009; 27:6537-45. [PMID: 19729088 DOI: 10.1016/j.vaccine.2009.08.051] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2009] [Revised: 07/08/2009] [Accepted: 08/16/2009] [Indexed: 12/18/2022]
Abstract
We prepared novel liposomes from total polar lipids of non-pathogenic Leptospira biflexa serovar Potac (designated leptosomes) and evaluated their vaccine delivery/adjuvant potential with novel protective antigens (Lp0607, Lp1118 and Lp1454) of L. interrogans serovar Pomona in a hamster model. The immune response induced by three individual antigens and protective efficacy were evaluated and compared to those induced by same antigens entrapped with PC-liposomes and E. coli lipid liposomes (escheriosomes). Four-week-old hamsters were immunized subcutaneously twice at a 3-week interval, bled at various time points to evaluate antibody response and sacrificed to isolate splenocytes for lymphocyte proliferation and cytokine profiles in response to recall antigen. For the challenge test, 10x MLD(50) (modified lethal dose 50%) of virulent L. interrogans serovar Pomona were administered intraperitoneally. Our results demonstrate that leptosome are better adjuvant than PC-liposomes as revealed by enhanced long term antibody response, lymphocyte proliferation and significant enhancement of both Th1 (IFN-gamma) and Th2 (IL-4 and IL-10) cytokines. Additionally, leptosomes and escheriosomes induced significantly higher level of memory responses than PC-liposome did. Moreover, the novel leptosomal vaccine induced significantly higher levels of protection than those prepared with PC-liposomes as revealed by enhanced survival, reduced histopathological lesions in vital organs and reduced leptospiral load in kidneys. Taken together, the results of the present study clearly reveal that both leptosomes and escheriosomes have emerged as promising delivery vehicles/adjuvants that can be widely exploited with newly discovered antigens in future leptospira vaccines.
Collapse
Affiliation(s)
- Syed M Faisal
- College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | | | |
Collapse
|
17
|
Dwivedi V, Vasco A, Vedi S, Dangi A, Arif K, Bhattacharya SM, Owais M. Adjuvanticity and protective immunity of Plasmodium yoelii nigeriensis blood-stage soluble antigens encapsulated in fusogenic liposome. Vaccine 2008; 27:473-82. [PMID: 18996429 DOI: 10.1016/j.vaccine.2008.10.054] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Revised: 10/13/2008] [Accepted: 10/20/2008] [Indexed: 10/21/2022]
Abstract
In our previous studies we established fusogenic properties of lipids isolated from edible yeast Saccharomyces cerevisiae (S. cerevisiae). We demonstrated that liposomes prepared from S. cerevisiae membrane lipid (saccharosome) can deliver encapsulated antigen into cytosol of the antigen presenting cells and elicit antigen specific cell mediated as well as humoral immune responses. In this study, we evaluated immunological behavior of saccharosome encapsulated cytosolic proteins (sAg) of Plasmodium yoelii nigeriensis in BALB/c mice. Immunization with antigen (sAg) encapsulated in saccharosome resulted in enhancement of CD4+ and CD8+ T cell populations and also up-regulated the expression of CD80 and CD86 molecules on the surface of antigen presenting cells. Further, immunization with saccharosome-encapsulated sAg-induced elevated levels of both IFN-gamma and IL-4 cytokines in the immunized mice when compared to egg PC liposome encapsulated sAg or its IFA emulsified form. Saccharosome-mediated immunization resulted in induction of high level of total antibody response with preponderance of IgG2a isotype as well. The data of this study suggest that saccharosome-based vehicle can emerge as an effective vaccine in imparting protection against various intracellular pathogens including Plasmodium yoelii nigeriensis.
Collapse
Affiliation(s)
- Varun Dwivedi
- Department of Biochemistry, J.N. Medical College, Aligarh Muslim University, Aligarh, India
| | | | | | | | | | | | | |
Collapse
|
18
|
Daubenberger CA, Pluschke G, Zurbriggen R, Westerfeld N. Development of influenza virosome-based synthetic malaria vaccines. Expert Opin Drug Discov 2008; 3:415-23. [DOI: 10.1517/17460441.3.4.415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
19
|
Contribution of influenza immunity and virosomal-formulated synthetic peptide to cellular immune responses in a phase I subunit malaria vaccine trial. Clin Immunol 2008; 127:188-97. [PMID: 18337175 DOI: 10.1016/j.clim.2008.01.012] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2007] [Revised: 01/15/2008] [Accepted: 01/15/2008] [Indexed: 12/14/2022]
Abstract
We have demonstrated recently in a phase Ia clinical trial that synthetic malaria peptides delivered by immuno-potentiating reconstituted influenza virosomes (IRIV) induced long-lived peptide-specific antibody responses in all volunteers. In the current ancillary study to this clinical trial we have investigated the cellular immune responses specific for IRIV and the surface bound synthetic malaria peptides tested. After vaccination, in 50% (8/16) of the volunteers at least one positive lymphoproliferative response specific for the 49mer peptide derived from the Plasmodium falciparum apical membrane antigen-1 (AMA-1) was observed with stimulation indices ranging from 2 to 4.5. All volunteers showed pre-existing IRIV specific cellular immunity assessed by ex vivo IFN-gamma ELISpot analysis and lymphoproliferation. The pre-existing influenza specific T cell responses did not interfere negatively with the induction of malaria peptide-specific humoral and cellular immune responses. Our results support the view that IRIV constitute a safe antigen delivery system for induction of peptide-specific immune responses in human populations.
Collapse
|
20
|
Okitsu SL, Silvie O, Westerfeld N, Curcic M, Kammer AR, Mueller MS, Sauerwein RW, Robinson JA, Genton B, Mazier D, Zurbriggen R, Pluschke G. A virosomal malaria peptide vaccine elicits a long-lasting sporozoite-inhibitory antibody response in a phase 1a clinical trial. PLoS One 2007; 2:e1278. [PMID: 18060072 PMCID: PMC2093993 DOI: 10.1371/journal.pone.0001278] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2007] [Accepted: 11/06/2007] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Peptides delivered on the surface of influenza virosomes have been shown to induce solid humoral immune responses in experimental animals. High titers of peptide-specific antibodies were also induced in a phase 1a clinical trial in volunteers immunized with virosomal formulations of two peptides derived from the circumsporozoite protein (CSP) and the apical membrane antigen 1 (AMA-1) of Plasmodium falciparum. The main objective of this study was to perform a detailed immunological and functional analysis of the CSP-specific antibodies elicited in this phase 1a trial. METHODOLOGY/PRINCIPAL FINDINGS 46 healthy malaria-naïve adults were immunized with virosomal formulations of two peptide-phosphatidylethanolamine conjugates, one derived from the NANP repeat region of P. falciparum CSP (designated UK-39) the other from P. falciparum AMA-1 (designated AMA49-C1). The two antigens were delivered in two different concentrations, alone and in combination. One group was immunized with empty virosomes as control. In this report we show a detailed analysis of the antibody response against UK-39. Three vaccinations with a 10 microg dose of UK-39 induced high titers of sporozoite-binding antibodies in all volunteers. This IgG response was affinity maturated and long-lived. Co-administration of UK-39 and AMA49-C1 loaded virosomes did not interfere with the immunogenicity of UK-39. Purified total IgG from UK-39 immunized volunteers inhibited sporozoite migration and invasion of hepatocytes in vitro. Sporozoite inhibition closely correlated with titers measured in immunogenicity assays. CONCLUSIONS Virosomal delivery of a short, conformationally constrained peptide derived from P. falciparum CSP induced a long-lived parasite-inhibitory antibody response in humans. Combination with a second virosomally-formulated peptide derived from P. falciparum AMA-1 did not interfere with the immunogenicity of either peptide, demonstrating the potential of influenza virosomes as a versatile, human-compatible antigen delivery platform for the development of multivalent subunit vaccines. TRIAL REGISTRATION ClinicalTrials.gov NCT00400101.
Collapse
Affiliation(s)
- Shinji L. Okitsu
- Molecular Immunology, Swiss Tropical Institute, Basel, Switzerland
| | - Olivier Silvie
- INSERM/UPMC UMR S U511, Immunobiologie Cellulaire et Moléculaire des Infections Parasitaires, Faculté de Médecine Pierre et Marie Curie, Centre Hospitalier Universitaire Pitié-Salpêtrière, Paris, France
| | | | - Marija Curcic
- Molecular Immunology, Swiss Tropical Institute, Basel, Switzerland
| | | | | | - Robert W. Sauerwein
- Department of Medical Microbiology, University Medical Centre St Radboud, Nijmegen, The Netherlands
| | - John A. Robinson
- Institute of Organic Chemistry, University of Zurich, Zurich, Switzerland
| | - Blaise Genton
- Molecular Immunology, Swiss Tropical Institute, Basel, Switzerland
| | - Dominique Mazier
- INSERM/UPMC UMR S U511, Immunobiologie Cellulaire et Moléculaire des Infections Parasitaires, Faculté de Médecine Pierre et Marie Curie, Centre Hospitalier Universitaire Pitié-Salpêtrière, Paris, France
| | | | - Gerd Pluschke
- Molecular Immunology, Swiss Tropical Institute, Basel, Switzerland
- * To whom correspondence should be addressed. E-mail:
| |
Collapse
|
21
|
Mallick A, Singha H, Khan S, Anwar T, Ansari M, Khalid R, Chaudhuri P, Owais M. Escheriosome-mediated delivery of recombinant ribosomal L7/L12 protein confers protection against murine brucellosis. Vaccine 2007; 25:7873-84. [DOI: 10.1016/j.vaccine.2007.09.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2007] [Revised: 08/31/2007] [Accepted: 09/03/2007] [Indexed: 11/29/2022]
|
22
|
A randomized placebo-controlled phase Ia malaria vaccine trial of two virosome-formulated synthetic peptides in healthy adult volunteers. PLoS One 2007; 2:e1018. [PMID: 17925866 PMCID: PMC2001290 DOI: 10.1371/journal.pone.0001018] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Accepted: 09/25/2007] [Indexed: 11/19/2022] Open
Abstract
Background and Objectives Influenza virosomes represent an innovative human-compatible antigen delivery system that has already proven its suitability for subunit vaccine design. The aim of the study was to proof the concept that virosomes can also be used to elicit high titers of antibodies against synthetic peptides. The specific objective was to demonstrate the safety and immunogenicity of two virosome-formulated P. falciparum protein derived synthetic peptide antigens given in two different doses alone or in combination. Methodology/Principal Findings The design was a single blind, randomized, placebo controlled, dose-escalating study involving 46 healthy Caucasian volunteers aged 18–45 years. Five groups of 8 subjects received virosomal formulations containing 10 µg or 50 µg of AMA 49-CPE, an apical membrane antigen-1 (AMA-1) derived synthetic phospatidylethanolamine (PE)-peptide conjugate or 10 ug or 50 ug of UK39, a circumsporozoite protein (CSP) derived synthetic PE-peptide conjugate or 50 ug of both antigens each. A control group of 6 subjects received unmodified virosomes. Virosomal formulations of the antigens (designated PEV301 and PEV302 for the AMA-1 and the CSP virosomal vaccine, respectively) or unmodified virosomes were injected i. m. on days 0, 60 and 180. In terms of safety, no serious or severe adverse events (AEs) related to the vaccine were observed. 11/46 study participants reported 16 vaccine related local AEs. Of these 16 events, all being pain, 4 occurred after the 1st, 7 after the 2nd and 5 after the 3rd vaccination. 6 systemic AEs probably related to the study vaccine were reported after the 1st injection, 10 after the 2nd and 6 after the 3rd. Generally, no difference in the distribution of the systemic AEs between either the doses applied (10 respectively 50 µg) or the synthetic antigen vaccines (PEV301 and PEV302) used for immunization was found. In terms of immunogenicity, both PEV301 and PEV302 elicited already after two injections a synthetic peptide-specific antibody response in all volunteers immunized with the appropriate dose. In the case of PEV301 the 50 µg antigen dose was associated with a higher mean antibody titer and seroconversion rate than the 10 µg dose. In contrast, for PEV302 mean titer and seroconversion rate were higher with the lower dose. Combined delivery of PEV301 and PEV302 did not interfere with the development of an antibody response to either of the two antigens. No relevant antibody responses against the two malaria antigens were observed in the control group receiving unmodified virosomes. Conclusions The present study demonstrates that three immunizations with the virosomal malaria vaccine components PEV301 or/and PEV302 (containing 10 µg or 50 µg of antigen) are safe and well tolerated. At appropriate antigen doses seroconversion rates of 100% were achieved. Two injections may be sufficient for eliciting an appropriate immune response, at least in individuals with pre-existing anti-malarial immunity. These results justify further development of a final multi-stage virosomal vaccine formulation incorporating additional malaria antigens. Trial Registration ClinicalTrials.gov NCT00400101
Collapse
|
23
|
Okitsu SL, Boato F, Mueller MS, Li DB, Vogel D, Westerfeld N, Zurbriggen R, Robinson JA, Pluschke G. Antibodies elicited by a virosomally formulated Plasmodium falciparum serine repeat antigen-5 derived peptide detect the processed 47 kDa fragment both in sporozoites and merozoites. Peptides 2007; 28:2051-60. [PMID: 17875342 DOI: 10.1016/j.peptides.2007.08.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2007] [Revised: 08/04/2007] [Accepted: 08/07/2007] [Indexed: 11/22/2022]
Abstract
Serine repeat antigen-5 (SERA5) is a candidate antigen for inclusion into a malaria subunit vaccine. During merozoite release and reinvasion the 120 kDa SERA5 precursor protein (P120) is processed, and a complex consisting of an N-terminal 47 kDa (P47) and a C-terminal 18kDa (P18) processing product associates with the surface of merozoites. This complex is thought to be involved in merozoite invasion of and/or egress from host erythrocytes. Here we describe the synthesis and immunogenic properties of virosomally formulated synthetic phosphatidylethanolamine (PE)-peptide conjugates, incorporating amino acid sequence stretches from the N-terminus of Plasmodium falciparum SERA5. Choosing an appropriate sequence was crucial for the development of a peptide that elicited high titers of parasite cross-reactive antibodies in mice. Monoclonal antibodies (mAbs) raised against the optimized peptide FB-23 incorporating amino acids 57-94 of SERA5 bound to both P120 and to P47. Western blotting analysis proved for the first time the presence of SERA5 P47 in sporozoites. In immunofluorescence assays, the mAbs stained SERA5 in all its predicted localizations. The virosomal formulation of peptide FB-23 is suitable for use in humans and represents a candidate component for a multi-valent malaria subunit vaccine targeting both sporozoites and blood stage parasites.
Collapse
Affiliation(s)
- Shinji L Okitsu
- Molecular Immunology, Swiss Tropical Institute, CH-4002 Basel, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Westerfeld N, Pluschke G, Zurbriggen R. Optimized Malaria-antigens delivered by immunostimulating reconstituted influenza virosomes. Wien Klin Wochenschr 2007; 118:50-7. [PMID: 17131241 DOI: 10.1007/s00508-006-0684-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Malaria remains a serious cause of morbidity and mortality in millions of individuals each year. The development of widespread resistance of the parasite to drugs as well as resistance of the transmitting mosquito-vector to insecticides in combination with the poor economic situation of many malaria-endemic countries make the development of an effective and inexpensive treatment and prevention a main focus of research. Vaccines remain to be one of the most cost effective and feasible means of disease control and have remarkable success in the control of many infectious disease: eradication of small pox, virtual eradication of polio and the reduction of measles and diphtheria. Next generation vaccines should focus on specific antigens rather than whole inactivated or attenuated pathogens, since the requirements by regulatory authorities concerning safety are becoming more stringent over time. But sub-unit and in particular peptide-based vaccines are poorly immunogenic themselves, and alum represents only a sub-optimal adjuvant for recombinant proteins and synthetic peptides. This emphasizes the need for suitable carrier- and adjuvant systems promoting protective immune responses by delivering protein and peptide antigens in an appropriate conformation. Here, we review the development of a new approach combining peptide-based malaria vaccine candidate antigens with an immune stimulatory carrier-system based on influenza virosomes focusing on the induction of protective antibodies.
Collapse
|
25
|
Okitsu SL, Kienzl U, Moehle K, Silvie O, Peduzzi E, Mueller MS, Sauerwein RW, Matile H, Zurbriggen R, Mazier D, Robinson JA, Pluschke G. Structure-activity-based design of a synthetic malaria peptide eliciting sporozoite inhibitory antibodies in a virosomal formulation. ACTA ACUST UNITED AC 2007; 14:577-87. [PMID: 17524988 DOI: 10.1016/j.chembiol.2007.04.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2006] [Revised: 03/13/2007] [Accepted: 04/02/2007] [Indexed: 10/23/2022]
Abstract
The circumsporozoite protein (CSP) of Plasmodium falciparum is a leading candidate antigen for inclusion in a malaria subunit vaccine. We describe here the design of a conformationally constrained synthetic peptide, designated UK-39, which has structural and antigenic similarity to the NPNA-repeat region of native CSP. NMR studies on the antigen support the presence of helical turn-like structures within consecutive NPNA motifs in aqueous solution. Intramuscular delivery of UK-39 to mice and rabbits on the surface of reconstituted influenza virosomes elicited high titers of sporozoite crossreactive antibodies. Influenza virus proteins were crucially important for the immunostimulatory activity of the virosome-based antigen delivery system, as a liposomal formulation of UK-39 was not immunogenic. IgG antibodies elicited by UK-39 inhibited invasion of hepatocytes by P. falciparum sporozoites, but not by antigenically distinct P. yoelii sporozoites. Our approach to optimized virosome-formulated synthetic peptide vaccines should be generally applicable for other infectious and noninfectious diseases.
Collapse
Affiliation(s)
- Shinji L Okitsu
- Molecular Immunology, Swiss Tropical Institute, CH-4002 Basel, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Plebanski M, Lopez E, Proudfoot O, Cooke BM, Itzstein MV, Coppel RL. Economic and practical challenges to the formulation of vaccines against endemic infectious diseases such as malaria. Methods 2007; 40:77-85. [PMID: 16997716 DOI: 10.1016/j.ymeth.2006.05.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2006] [Accepted: 05/02/2006] [Indexed: 11/30/2022] Open
Abstract
Herein, we analyze in general the current vaccine market and identify potential factors driving and modulating supply and demand for vaccines. An emphasis is placed on changes in regulation in the last 20 years which have led to increased indirect costs of production, and which can create a barrier against the timely use of technological advances to reduce direct costs. Other defining industry characteristics, such as firm numbers and sizes, cost and pricing strategies, nature extent and impact of Government involvement and international regulation are noted. These considerations, far from being removed from basic vaccine research, influence its ability to achieve aims that can be then progressed into effective vaccine products. We discuss specifically the development of particulate vaccines against malaria, a major lethal disease and health problem prevalent in Africa, including some key economic and methodological challenges and opportunities. We note some practical issues blocking the development of effective particulate vaccines for the Third World, mainly driven by the regulatory spiral noted above.
Collapse
Affiliation(s)
- Magdalena Plebanski
- Vaccine and Infectious Diseases Laboratory, Burnet Institute at Austin, Studley Road, Heidelberg, Vic. 3084, Australia.
| | | | | | | | | | | |
Collapse
|
27
|
James S, Moehle K, Renard A, Mueller MS, Vogel D, Zurbriggen R, Pluschke G, Robinson JA. Synthesis, Solution Structure and Immune Recognition of an Epidermal Growth Factor-Like Domain from Plasmodium falciparum Merozoite Surface Protein-1. Chembiochem 2006; 7:1943-50. [PMID: 17068840 DOI: 10.1002/cbic.200600357] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The Plasmodium falciparum merozoite surface protein-1 19 kDa fragment (MSP-1(19)) comprises two closely packed EGF-like domains (EGF=epidermal growth factor), each stabilized by three disulfide bonds. The native conformation of this protein is important for eliciting P. falciparum growth inhibitory antibodies. Here we show that the N-terminal EGF domain alone can be chemically synthesized and efficiently refolded to a native-like state, as shown by its solution structure as determined by NMR spectroscopy. In order to study its immunogenicity, the domain was coupled through its N terminus to a phospholipid and incorporated into reconstituted influenza virus-like particles (virosomes). When used to immunize mice, the peptide-loaded virosomes elicited potent humoral immune responses that were shown by Western blots and immunofluorescence assays to cross-react with native MSP-1 on the surfaces of P. falciparum blood stage parasites. This opens the way for a medicinal chemistry-oriented approach to the study and optimization of the antigenicity of the protein as a potential malaria vaccine candidate, whilst exploiting the immunopotentiating properties of influenza virosomes as a delivery vehicle.
Collapse
Affiliation(s)
- Sonya James
- Department of Chemistry, University of Zürich, Winterthurerstrasse 190, 8057 Zürich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Ahmad N, Deeba F, Faisal SM, Khan A, Agrewala JN, Dwivedi V, Owais M. Role of fusogenic non-PC liposomes in elicitation of protective immune response against experimental murine salmonellosis. Biochimie 2006; 88:1391-400. [PMID: 16765503 DOI: 10.1016/j.biochi.2006.04.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2005] [Revised: 04/07/2006] [Accepted: 04/28/2006] [Indexed: 11/28/2022]
Abstract
Earlier we have demonstrated that novel fusogenic liposomes made up of lipid from Escherichia coli (escheriosomes) have strong tendency to fuse with the plasma membrane of target cells and thereby delivering the entrapped contents into their cytosol. The delivery of entrapped antigen in cytosol of the target cells ensues its processing and presentation along with MHC class I pathway that eventually elicit antigen specific cytotoxic T cells. The result of the present study revealed that immunization of BALB/c mice with escheriosome-encapsulated Salmonella typhimurium (S. typhimurium) cytosolic antigens resulted in the augmentation of antigen specific cytotoxic T cell lymphocyte as well as IgG responses. In contrast, free or conventional liposome (PC liposome) encapsulated antigen failed to induce CD8+ CTLs in the immunized animals. Further, immunization with escheriosome-encapsulated antigen resulted in significant enhancement in the release of IFN-gamma and IgG2a in the experimental animals. Interestingly, the immunization with escheriosome-encapsulated antigen resulted in upregulation of CD80 and CD86 on the surface of antigen presenting cells (APCs) as well. Finally, the results of the present study reveal that immunization of animals with escheriosomes encapsulated antigen protected them against virulent S. typhimurium infection. This was evident by increased survival, and reduced bacterial burden in vital organs of the immunized animals. The data of the present study suggest that escheriosomes can emerge as an effective vehicle for intracellular delivery of antigen and thus hold promise in development of liposome based vaccine against Salmonella and other intracellular pathogens.
Collapse
Affiliation(s)
- N Ahmad
- Faculty of Pharmacy, Jamia Hamdard, New Delhi 62, India
| | | | | | | | | | | | | |
Collapse
|
29
|
Liu X, Siegrist S, Amacker M, Zurbriggen R, Pluschke G, Seeberger PH. Enhancement of the immunogenicity of synthetic carbohydrates by conjugation to virosomes: a leishmaniasis vaccine candidate. ACS Chem Biol 2006; 1:161-4. [PMID: 17163663 DOI: 10.1021/cb600086b] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Novel virosomal formulations of a synthetic oligosaccharide were prepared and evaluated as vaccine candidates against leishmaniasis. A lipophosphoglycan-related synthetic tetrasaccharide antigen was conjugated to a phospholipid and to the influenza virus coat protein hemagglutinin. These glycan conjugates were embedded into the lipid membrane of reconstituted influenza virus virosomes. The virosomal formulations elicited both IgM and IgG anti-glycan antibodies in mice, indicating an antibody isotype class switch to IgG. The antisera cross-reacted in vitro with the corresponding natural carbohydrate antigens expressed by leishmania cells. These findings support the concept of using virosomes as universal antigen delivery platform for synthetic carbohydrate vaccines.
Collapse
Affiliation(s)
- Xinyu Liu
- Laboratorium für Organische Chemie, ETH Zürich, Wolfgang-Pauli Strasse 10, 8093 Zürich, Switzerland
| | | | | | | | | | | |
Collapse
|
30
|
Huckriede A, Bungener L, Stegmann T, Daemen T, Medema J, Palache AM, Wilschut J. The virosome concept for influenza vaccines. Vaccine 2005; 23 Suppl 1:S26-38. [PMID: 16026906 DOI: 10.1016/j.vaccine.2005.04.026] [Citation(s) in RCA: 151] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
There is a need for more efficacious inactivated influenza vaccines, since current formulations show suboptimal immunogenicity in at-risk populations, like the elderly. More effective vaccines are also urgently needed for an improved influenza pandemic preparedness. In this context, there is considerable interest in virosomes. Virosomes are virus-like particles, consisting of reconstituted influenza virus envelopes, lacking the genetic material of the native virus. Virosomes are produced from influenza virus through a detergent solubilization and removal procedure. Properly reconstituted virosomes retain the cell binding and membrane fusion properties of the native virus, mediated by the viral envelope glycoprotein haemagglutinin. These functional characteristics of virosomes form the basis for their enhanced immunogenicity. First, the repetitive arrangement of haemagglutinin molecules on the virosomal surface mediates a cooperative interaction of the antigen with Ig receptors on B lymphocytes, stimulating strong antibody responses. In addition, virosomes interact efficiently with antigen-presenting cells, such as dendritic cells, resulting in activation of T lymphocytes. In a murine model system, virosomes, as compared to conventional subunit vaccine, which consists of isolated influenza envelope glycoproteins, induce a more balanced T helper 1 versus T helper 2 response, virosomes in particular eliciting stronger T helper 1 responses than subunit vaccine. Also, as a result of fusion of the virosomes with the endosomal membrane, part of the virosomal antigen gains access to the major histocompatibility class I presentation pathway, thus priming cytotoxic T lymphocyte activity. Finally, virosomes represent an excellent platform for inclusion of lipophilic adjuvants for further stimulation of vaccine immunogenicity. By virtue of these characteristics, virosomes represent a promising novel class of inactivated influenza vaccines, which not only induce high virus-neutralizing antibody titres, but also prime the cellular arm of the immune system.
Collapse
Affiliation(s)
- Anke Huckriede
- University Medical Center Groningen, Department of Medical Microbiology, Molecular Virology Section, University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
31
|
Musoke A, Rowlands J, Nene V, Nyanjui J, Katende J, Spooner P, Mwaura S, Odongo D, Nkonge C, Mbogo S, Bishop R, Morzaria S. Subunit vaccine based on the p67 major surface protein of Theileria parva sporozoites reduces severity of infection derived from field tick challenge. Vaccine 2005; 23:3084-95. [PMID: 15811656 DOI: 10.1016/j.vaccine.2004.09.039] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2004] [Revised: 08/30/2004] [Accepted: 09/08/2004] [Indexed: 11/15/2022]
Abstract
Two recombinant vaccines against Theileriaparva, based on a near full-length version of the sporozoite surface antigen p67 (p67(635)), or an 80 amino acid C-terminal section (p67C), were evaluated by exposure of immunized cattle to natural tick challenge in two sites at the Kenya Coast and one in Central Kenya. Vaccination reduced severe ECF by 47% at the coast and by 52% in central Kenya from an average incidence of 0.53+/-0.07 (S.E.) in 50 non-immunised controls to an average of 0.27+/-0.05 in 83 immunised animals. The reduction in severe East Coast fever was similar to that observed in laboratory experiments with p67(635) and p67C. The p67 coding sequence from thirteen T. parva field isolates including seven from vaccinated cattle that were not protected, was 100% identical to the gene on which the recombinant vaccine is based, suggesting a predominantly homologous p67 antigenic challenge. The same parasite isolates were however genetically heterogeneous at several loci other than p67.
Collapse
Affiliation(s)
- Antony Musoke
- International Livestock Research Institute, P.O. Box 30709 Nairobi. Kenya
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Glück R, Moser C, Metcalfe IC. Influenza virosomes as an efficient system for adjuvanted vaccine delivery. Expert Opin Biol Ther 2005; 4:1139-45. [PMID: 15268680 DOI: 10.1517/14712598.4.7.1139] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Immunopotentiating reconstituted influenza virosomes possess several characteristics defining them as vaccine adjuvants. Virosomes have been shown to provide vaccine components with protection from extracellular degradation; a regular, repetitive antigen structure aiding presentation to B lymphocytes and fully functional, fusion-active, influenza haemagglutinin envelope proteins that enables receptor-mediated uptake and intracellular processing of the antigen. In addition, virosomes, as vaccine delivery systems, have been shown to be safe and not to engender any antibodies against the phospholipid components. Through the use of virosomes as a delivery vehicle, a number of vaccines have been developed. In humans, virosome-based vaccines containing inactivated hepatitis A and influenza antigens have been found to be efficacious and well-tolerated and have been on the market for several years. Hepatitis B, nucleic acids, cytotoxic drugs, and tetanus and diphtheria toxoids have also been incorporated into virosomes. Further investigations are ongoing in order to define the full potential of virosomes in both prophylactic and immunotherapeutic applications.
Collapse
Affiliation(s)
- Reinhard Glück
- Berna Biotech Ltd, Rehhagstrasse 79, Berne, Switzerland.
| | | | | |
Collapse
|
33
|
Westerfeld N, Zurbriggen R. Peptides delivered by immunostimulating reconstituted influenza virosomes. J Pept Sci 2005; 11:707-12. [PMID: 16059967 DOI: 10.1002/psc.700] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Vaccines have been well accepted and used effectively for more than 100 years. Traditional vaccines are generally composed of whole inactivated or attenuated microorganisms that have lost their disease-causing properties. These classical prophylactic live vaccines evoke protective immune responses, but have often been associated with an unfavorable safety profile, as observed, for example, for smallpox and polio myelitis vaccines [1,2]. First improvements were subunit vaccines that do not focus on attenuation of whole organisms but concentrate on particular proteins. These vaccines are able to generate protective immune responses (e.g. diphtheria, tetanus, pertussis)3. However, next generation vaccines should focus on specific antigens (e.g. proteins, peptides), since the requirements by regulatory authorities to crude biological material are becoming more stringent over time. An increasing number of such antigens capable of inducing protective humoral or cellular immune responses have been identified in the last few years. But most of these are weak immunogens. This reemphasizes the need for adjuvants to promote a potent immune response and also for delivery antigens to the immune system in an appropriate way (carrier capability). Here we review a new approach for prophylactic and therapeutic vaccines, which focuses on the induction of highly specific immune responses directed against antigen-derived peptides using a suitable carrier system.
Collapse
|
34
|
Schwaninger R, Waelti E, Zajac P, Wetterwald A, Mueller D, Gimmi CD. Virosomes as new carrier system for cancer vaccines. Cancer Immunol Immunother 2004; 53:1005-17. [PMID: 15185010 PMCID: PMC11032935 DOI: 10.1007/s00262-004-0545-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2003] [Accepted: 03/22/2004] [Indexed: 11/24/2022]
Abstract
HER-2/neu, a tumor-associated antigen (TAAg), plays a critical role in oncogenesis of various tumor types, and its selective overexpression by malignant tumor cells makes it an ideal target for immunotherapy. A prerequisite for clinical vaccines is the construction of safe and highly immunogenic reagents able to generate efficient immune responses against TAAg. Previous protein vaccines, consisting of the extracellular domain of HER-2/neu (pNeuECD), were shown to elicit an immune response that did not provide protection from transplantable tumors expressing HER-2/neu. Here we showed that virosomes, which consist of reconstituted viral envelopes without viral genetic material, can act as a carrier and an adjuvant for a truncated protein pNeuECD. Mice vaccinated with pNeuECD either encapsulated in virosomes or bound to the virosomal membrane (Vir-pNeuECD), generated rNeu-specific humoral and cytotoxic immune responses. In addition, Vir-p(NeuECD) induced significant tumor rejection and additionally did not lead to delayed tumor formation when compared with free pNeuECD in complete Freund's adjuvant. There was no difference between the virosomal constructs. Taken together these results suggest that virosomes, as clinically approved safe vaccines, can be used to elicit both humoral and cell-mediated responses against TAAg and induce tumor rejection. Our model is providing important preclinical data to design human vaccination trials for patients with tumors overexpressing HER-2/neu, either as a primary vaccination or as a boost in combination with other vaccines in a context of an adjuvant treatment plan.
Collapse
Affiliation(s)
- Ruth Schwaninger
- Department of Clinical Research, University of Bern, Bern, Switzerland
| | - Ernst Waelti
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Paul Zajac
- Research Department, Kantonsspital Basel, Basel, Switzerland
| | | | | | - Claude D. Gimmi
- Department of Clinical Research, University of Bern, Bern, Switzerland
- Global Drug Development, Oncology, F. Hoffmann-LaRoche, PDM2, Grenzacherstrasse, 4070 Basel, Switzerland
| |
Collapse
|
35
|
Mueller MS, Renard A, Boato F, Vogel D, Naegeli M, Zurbriggen R, Robinson JA, Pluschke G. Induction of parasite growth-inhibitory antibodies by a virosomal formulation of a peptidomimetic of loop I from domain III of Plasmodium falciparum apical membrane antigen 1. Infect Immun 2003; 71:4749-58. [PMID: 12874357 PMCID: PMC166038 DOI: 10.1128/iai.71.8.4749-4758.2003] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Apical membrane antigen 1 (AMA-1) of Plasmodium falciparum is a leading candidate antigen for inclusion in a malaria subunit vaccine. Its ectodomain can be divided into three subdomains, each with disulfide bond-stabilized structures. Since the majority of antibodies raised against the ectodomain appear to recognize strain-specific epitopes in domain I, we attempted to develop a vaccine formulation which directs the immune response to a region that contains more conserved epitopes. Here we demonstrate that a virosomal formulation of a peptide that mimics the semiconserved loop I of domain III elicits parasite growth-inhibitory antibodies. A synthetic peptide comprising residues 446 to 490 of AMA-1 (AMA-1(446-490)) was conjugated through the N terminus to a derivative of phosphatidylethanolamine and the phosphatidylethanolamine-peptide conjugate was incorporated into immunopotentiating reconstituted influenza virosomes as a human-compatible antigen delivery system. Both cyclized and linear versions of the peptide antigen elicited antibodies which specifically bound to parasite-expressed AMA-1 in Western blotting with parasite lysates as well as in immunofluorescence assays with blood stage parasites. All 11 peptidomimetic-specific monoclonal antibodies generated were cross-reactive with parasite-expressed AMA-1. Antigen binding assays with a library of overlapping cyclic peptides covering the target sequence revealed differences in the fine specificity of these monoclonal antibodies and provided evidence that at least some of them recognized discontinuous epitopes. The two immunodominant epitopes comprised the conserved linear sequences K(459)RIKLN(464) and D(467)DEGNKKII(475). A key feature of the synthetic vaccine formulation proposed here is the display of the peptide antigen in a native-like state on the surface of the virosome.
Collapse
Affiliation(s)
- Markus S Mueller
- Molecular Immunology, Swiss Tropical Institute, CH-4002 Basel, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Syed FM, Khan MA, Nasti TH, Ahmad N, Mohammad O. Antigen entrapped in the escheriosomes leads to the generation of CD4(+) helper and CD8(+) cytotoxic T cell response. Vaccine 2003; 21:2383-93. [PMID: 12744869 DOI: 10.1016/s0264-410x(03)00106-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
In previous study, we demonstrated the potential of Escherichia coli (E. coli) lipid liposomes (escheriosomes) to undergo membrane-membrane fusion with cytoplasmic membrane of the target cells including professional antigen presenting cells. Our present study demonstrates that antigen encapsulated in escheriosomes could be successfully delivered simultaneously to the cytosolic as well as endosomal processing pathways of antigen presenting cells, leading to the generation of both CD4(+) T-helper and CD8(+) cytotoxic T cell response. In contrast, encapsulation of same antigen in egg phosphatidyl-choline (egg PC) liposomes, just like antigen-incomplete Freund's adjuvant (IFA) complex, has inefficient access to the cytosolic pathway of MHC I-dependent antigen presentation and failed to generate antigen-specific CD8(+) cytotoxic T cell response. However, both egg PC liposomes as well as escheriosomes-encapsulated antigen elicited strong humoral immune response in immunized animals but antibody titre was significantly higher in the group of animals immunized with escheriosomes-encapsulated antigen. These results imply usage of liposome-based adjuvant as potential candidate vaccine capable of eliciting both cell-mediated as well as humoral immune responses. Furthermore, antigen entrapped in escheriosomes stimulates antigen-specific CD4(+) T cell proliferation and also enhances the level of IL-2, IFN-gamma and IL-4 in the immunized animals.
Collapse
Affiliation(s)
- Faisal M Syed
- Interdisciplinary Biotechnology Unit, Aligarh Muslim University, 202002, Aligarh, India
| | | | | | | | | |
Collapse
|
37
|
Daubenberger CA, Diaz D, Curcic M, Mueller MS, Spielmann T, Certa U, Lipp J, Pluschke G. Identification and characterization of a conserved, stage-specific gene product of Plasmodium falciparum recognized by parasite growth inhibitory antibodies. Infect Immun 2003; 71:2173-81. [PMID: 12654839 PMCID: PMC152076 DOI: 10.1128/iai.71.4.2173-2181.2003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We have identified a novel conserved protein of Plasmodium falciparum, designated D13, that is stage-specifically expressed in asexual blood stages of the parasite. The predicted open reading frame (ORF) D13 contains 863 amino acids with a calculated molecular mass of 99.7 kDa and displays a repeat region composed of pentapeptide motives. Northern blot analysis with lysates of synchronized blood stage parasites showed that D13 is highly expressed at the mRNA level during schizogony. The first N'-terminal 138 amino acids of D13 were expressed in Escherichia coli and the purified protein was used to generate anti-D13 monoclonal antibodies (MAbs). Using total lysates of blood stage parasites and Western blot analysis, these MAbs stained one single band of approximately 100 kDa, corresponding to the predicted molecular mass of ORF D13. Western blot analysis demonstrated further that D13 is expressed during schizogony, declines rapidly in early ring stages and is undetectable in trophozoites. D13 protein is localized in individual merozoites in a distinct area, as demonstrated by indirect immunofluorescence analysis. After subcellular fractionation, D13 was confined to the pelleted fraction of the parasite lysate and its extraction by alkaline carbonate buffer treatment indicated that D13 is not a membrane-integral protein. Inclusion of certain anti-D13 MAbs into in vitro cultures of blood stage parasites resulted in considerable reduction in parasite growth. The N'-terminal domain encompassing 158 amino acids is 94 and 95%, respectively, identical at the amino acid level between Plasmodium knowlesi, Plasmodium yoelii, and P. falciparum. In summary, we describe a novel stage-specifically expressed, highly conserved gene product of P. falciparum that is recognized by parasite growth inhibitory antibodies.
Collapse
Affiliation(s)
- Claudia A Daubenberger
- Molecular Immunology, Swiss Tropical Institute, Socinstrasse 57, 4002 Basel, Switzerland.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Affiliation(s)
- Lorne A Babiuk
- Veterinary Infectious Disease Organization Saskatoon, Saskatchewan S7N 5E3, Canada
| | | | | |
Collapse
|
39
|
Abstract
Influenza virosomes can be regarded as unilamellar liposomes carrying the spike proteins of influenza virus on their surface. Vaccination with influenza virosomes elicits high titers of influenza-specific antibodies, indicating that HA (and NA) reconstituted into a membranous environment exhibit strong immunogenicity. Moreover, virosomes can be used as presentation systems for unrelated antigens bound to the virosome surface. Because of their intrinsic adjuvant activity, virosomes support antibody formation and induction of T-helper cell responses against such surface-associated antigens. Provided that the fusogenic properties of the reconstituted HA are retained, virosomes can also be used to elicit cytotoxic T-cell responses against encapsulated antigens. Vaccines capable of activating the cellular branch of the immune response can be very important for protection against acute virus infections, especially for viruses with rapidly changing envelope glycoproteins like HIV and influenza virus. Moreover, virosomes can suit as powerful carriers in the development of prophylactic and immunotherapeutic strategies against cancer and premalignant disease. The use of virosomes as commercial influenza vaccine and as commercial adjuvant for a hepatitis A vaccine demonstrates that production of virosomes on an industrial scale is feasible, both technically and economically. The industrial production procedure currently followed has not been designed to retain the functional properties of HA. In fact, several steps in the procedure are probably incompatible with retention of fusion activity. As mentioned previously the fusogenic properties of virosomes are important for CTL activation and might also play a role in the induction of T-helper cell and antibody responses. Therefore, a number of key adaptations in the virosome production protocol will be necessary. Thus improved, virosomes are very attractive devices for the development of highly efficacious vaccines against a range of antigens.
Collapse
Affiliation(s)
- Anke Huckriede
- Department of Medical Microbiology, Molecular Virology Section, University of Groningen, 9713 AV Groningen, The Netherlands
| | | | | | | |
Collapse
|
40
|
Abstract
The desire for improved quality of life in both industrialised and under-developed nations has led to the quest for greater understanding and subsequent prevention and treatment of diseases. Here we discuss some of the latest of modern medicine's approaches to vaccination and disease treatment. Our main subject of discussion being the novel antigen delivery systems termed immunopotentiating reconstituted influenza virosomes (IRIVs) and their use as vaccines. Particular attention is paid to the currently licensed Epaxal and Inflexal V, good examples of the improvements being made in vaccinology. Alternative uses of virosomes such as peptide delivery, cytosolic drug delivery and gene delivery are also considered, highlighting the flexibility of the IRIV formulation and method of action. The paper concludes with consideration of alternative novel approaches to vaccinology including bacterial carriers for DNA vaccines, recombinant MV vaccines and polysaccharide-protein conjugates.
Collapse
Affiliation(s)
- R Glück
- Berna Biotech Ltd., Rehhagstr. 79, CH-3018 Berne, Switzerland.
| | | |
Collapse
|
41
|
Ahmad N, Masood AK, Owais M. Fusogenic potential of prokaryotic membrane lipids. Implication in vaccine development. EUROPEAN JOURNAL OF BIOCHEMISTRY 2001; 268:5667-75. [PMID: 11722550 DOI: 10.1046/j.0014-2956.2001.02507.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Development of protective immunity against many pathogens, particularly viruses, requires fine orchestration of both humoral- and cell mediated-immunity. The immunization of animals with soluble antigens usually leads to the induction of humoral immune responses. In contrast, the activation of a cell-mediated immune response against exogenous antigens has always been a challenge, requiring special strategies to expose them to the proteasome, a multifunctional protease complex in the cytosol of the target cells. The degradation of the protein by the cytosolic proteolytic system forms a cardinal step for the induction of cytotoxic T lymphocytes (CTLs). In the present study, we report that a potent primary CTL response against a soluble protein, ovalbumin, can be induced in mice by encapsulating it in the liposomes comprised of Escherichia coli membrane lipids. These lipids were shown to induce strong membrane-membrane fusion as evident from resonance energy transfer and content mixing assays. Furthermore, the fusion of these liposomes with living cells (J774 A1) was demonstrated to result in effective transfer of a fluorescent lipid probe to the plasma membrane of the cells. Moreover, ricin A, a protein synthesis inhibitor that does not cross plasma membrane, was demonstrated to gain access to the cytosol when it was encapsulated in these liposomes. Finally, the liposomes were demonstrated to behave like efficient vehicles for the in vivo delivery of the antigens to the target cells resulting in the elicitation of antigen reactive CD8+ T cell responses.
Collapse
Affiliation(s)
- N Ahmad
- Pharmaceutics Department, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | | | | |
Collapse
|
42
|
Lioy E, Suarez J, Guzmàn F, Siegrist S, Pluschke G, Patarroyo ME. Synthesis, Biological, and Immunological Properties of Cyclic Peptides fromPlasmodium Falciparum Merozoite Surface Protein-1. Angew Chem Int Ed Engl 2001; 40:2631-2635. [DOI: 10.1002/1521-3773(20010716)40:14<2631::aid-anie2631>3.0.co;2-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2001] [Indexed: 11/08/2022]
|
43
|
Lioy E, Suarez J, Guzmàn F, Siegrist S, Pluschke G, Patarroyo ME. Synthesis, Biological, and Immunological Properties of Cyclic Peptides fromPlasmodium Falciparum Merozoite Surface Protein-1. Angew Chem Int Ed Engl 2001. [DOI: 10.1002/1521-3757(20010716)113:14<2701::aid-ange2701>3.0.co;2-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
44
|
Owais M, Masood AK, Agrewala JN, Bisht D, Gupta CM. Use of liposomes as an immunopotentiating delivery system: in perspective of vaccine development. Scand J Immunol 2001; 54:125-32. [PMID: 11439158 DOI: 10.1046/j.1365-3083.2001.00944.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Liposomes have been widely used to deliver antigens to the antigen-presenting cells (APCs) and also to modify their immunological behaviour in model animals. We recently demonstrated the potential of yeast lipid liposomes to undergo membrane-membrane fusion with cytoplasmic membrane of the target cells. Interestingly, studies in the present report revealed that antigen encapsulated in yeast lipid liposomes could be successfully delivered simultaneously into the cytosolic as well as endosomal processing pathways of APCs, leading to the generation of both CD4+ T helper and CD8+ cytotoxic T cells. In contrast, encapsulation of same antigen in egg phosphatidyl-choline (PC) liposomes, just like its free form, has inefficient access to the cytosolic pathway of major histocompatibility complex (MHC) I dependent antigen presentation and failed to generate antigen specific CD8+ cytotoxic T-cell response. However, both egg PC as well as yeast lipid liposomes have elicited strong antigen specific antibody responses in immunized animals. These results imply usage of liposome encapsulated antigen as potential candidate vaccine capable of eliciting both cell mediated as well as humoral immune responses.
Collapse
|
45
|
Moreno R, Pöltl-Frank F, Stüber D, Matile H, Mutz M, Weiss NA, Pluschke G. Rhoptry-associated protein 1-binding monoclonal antibody raised against a heterologous peptide sequence inhibits Plasmodium falciparum growth in vitro. Infect Immun 2001; 69:2558-68. [PMID: 11254620 PMCID: PMC98192 DOI: 10.1128/iai.69.4.2558-2568.2001] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Monoclonal antibodies (MAbs) specific for Plasmodium falciparum rhoptry-associated protein 1 (RAP-1) were generated and tested for inhibition of parasite growth in vitro. The majority of indirect immunofluorescence assay (IFA)-positive MAbs raised against recombinant RAP-1 positions 23 to 711 (rRAP-1(23-711)) recognized epitopes located in the immunodominant N-terminal third of RAP-1. MAbs specific for the building block 35.1 of the synthetic peptide malaria vaccine SPf66 also yielded an IFA staining pattern characteristic for rhoptry-associated proteins and reacted specifically with rRAP-1 and parasite-derived RAP-1 molecules p67 and p82. Cross-reactivity with RAP-1 was blocked by the 35.1 peptide. Epitope mapping with truncated rRAP-1 molecules and overlapping peptides identified the linear RAP-1 sequence Y218KYSL222 as a target of the anti-35.1 MAbs. This sequence lacks primary sequence similarity with the 35.1 peptide (YGGPANKKNAG). Cross-reactivity of the anti-35.1 MAbs thus appears to be associated with conformational rather than sequence homology. While the anti-35.1 MAb SP8.18 exhibited parasite growth-inhibitory activity, none of the tested anti-rRAP-1(23-711) MAbs inhibited parasite growth, independently of their fine specificity for the RAP-1 sequences at positions 33 to 42, 213 to 222, 243 to 247, 280 to 287, or 405 to 446. The growth-inhibitory activity of MAb SP8.18 was, however, accelerated by noninhibitory anti-RAP-1 MAbs. Results demonstrate that in addition to fine specificity, other binding parameters are also crucial for the inhibitory potential of an antibody.
Collapse
Affiliation(s)
- R Moreno
- Swiss Tropical Institute, CH-4002 Basel, Switzerland
| | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Needle-free delivery of vaccines should not only increase compliance, but should also prove to be a safer and less traumatic method of vaccine delivery. One of the potential ways to achieve needle-free delivery is with the use of lipid-based delivery systems. To demonstrate the utility of these systems, we have shown them to be effective with proteins produced by recombinant DNA technology, plasmid-based vaccines, as well as conventional vaccines. Furthermore, these lipid-based delivery systems were shown to be effective in inducing mucosal immunity if delivered to mucosal surfaces or systemic immunity if different transdermally. These approaches have the potential to revolutionize vaccine delivery in humans and animals.
Collapse
Affiliation(s)
- M E Baca-Estrada
- Veterinary Infectious Disease Organization, University of Saskatchewan, 120 Veterinary Road, S7N 5E3, Saskatoon, Canada.
| | | | | | | |
Collapse
|
47
|
Affiliation(s)
- D C Drummond
- Research Institute, California Pacific Medical Center, 94115, San Francisco, CA, USA
| | | | | |
Collapse
|
48
|
Kurella S, Manocha M, Sabhnani L, Thomas B, Rao DN. New age adjuvants and delivery systems for subunit vaccines. Indian J Clin Biochem 2000; 15:83-100. [PMID: 23105272 PMCID: PMC3454077 DOI: 10.1007/bf02867548] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The dramatic advancements in the field of vaccinology has led to the formulation of chemically well defined vaccines composed of synthetic peptides and recombinant proteins derived from the immunologically dominant regions of the pathogens. Though these subunit vaccines are safer compared to the traditional vaccines they are known to be poorly immunogenic. This necessitates the use of adjuvants to enhance the immunogenicity of these vaccine formulations. The most common adjuvant for human use is alum. Research in the past has focused on the development of systemic immunity using conventional immunization protocols. In the present are, the emphasis is on the development and formulation of alternative adjuvants and delivery systems in generating systemic as well as mucosal immunity. This review mainly focuses on a variety of adjuvants (particulate as well as non-particulate) used with protective antigens of HIV, malaria, plague, leprosy using modified delivery vehicles. The experience of our laboratory and other researchers in this field clearly proves that these new age adjuvants and delivery systems undoubtedly generate enhanced immune response-both humoral and cell mediated. The choice of antigens, the nature of adjuvant used and the mode of delivery employed have a profound effect on the type of immune response generated. Besides the quantity, the quality of the antibodies generated also play a vital role in protection against these diseases. Some of the adjuvants and delivery systems used promoted high titre and affinity antibodies, which were shown to be cytophilic in nature, an important criteria in providing protection to the host. Thus the studies on these adjuvants/delivery systems with respect to various infectious diseases indicate their active role in efficient modulation of immune response along with safety and permissibility.
Collapse
Affiliation(s)
- S Kurella
- Department of Biochemistry, All India Institute Of Medical Sciences, 110029 New Delhi, India
| | | | | | | | | |
Collapse
|
49
|
Owais M, Gupta CM. Liposome-mediated cytosolic delivery of macromolecules and its possible use in vaccine development. EUROPEAN JOURNAL OF BIOCHEMISTRY 2000; 267:3946-56. [PMID: 10866793 DOI: 10.1046/j.1432-1327.2000.01447.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the majority of bacterial and viral infections the generation of cytotoxic T cells is of particular interest because such pathogens are able to escape the host defence mechanisms by surviving intracellularly within the phagocytic cells. To generate a CD8+ T lymphocyte response against exogenous antigens, the prerequisite is their delivery into the cytosol followed by processing and presentation along with class I major histocompatibility complex (MHC-I) molecules. In the present study we describe the method of liposome-based delivery of antigens and other macromolecules into the cytosol of target cells. To develop safe and effective methods for generating CD8+ T lymphocytes, we exploited the fusogenic character of lipids derived from lower organisms, that is baker's yeast (Saccharomyces cerevisiae). The degree of fusion with model membrane systems using yeast lipid liposomes varied from 40-70%, as opposed to 1-8% observed with egg PtdCho liposomes, depending on the assay system used. The fusion of yeast lipid liposomes with macrophages resulted in effective delivery of the entrapped solutes into the cytoplasmic compartment. This was further supported by the inhibition of cellular protein synthesis in J774 A1 cells by ricin A, encapsulated in the yeast lipid liposomes. Interestingly, the model antigen ovalbumin, when entrapped in the yeast lipid liposomes, successfully elicited antigen reactive CD8+ T cell responses. It may be concluded that the liposomes made of lipids derived from S. cerevisiae can spontaneously fuse with macrophages, delivering a significant portion of their contents into the cytoplasmic compartment of the cells.
Collapse
Affiliation(s)
- M Owais
- Inter-disciplinary Biotechnology Unit, Aligarh Muslim University, India
| | | |
Collapse
|
50
|
Daubenberger CA, Pöltl-Frank F, Jiang G, Lipp J, Certa U, Pluschke G. Identification and recombinant expression of glyceraldehyde-3-phosphate dehydrogenase of Plasmodium falciparum. Gene 2000; 246:255-64. [PMID: 10767547 DOI: 10.1016/s0378-1119(00)00069-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The gene coding for the cytosolic glyceraldehyde-3-phosphate dehydrogenase (GAPDH; EC 1.2.1.12) was isolated from Plasmodium falciparum. The gene contains 1 intron and the A+T content is characteristic for the codon usage of P. falciparum. The predicted open reading frame codes for 337 amino acids (36651Da) and is 63.5% identical to the human erythrocytic GAPDH. GAPDH sequences from several field isolates of P. falciparum displayed 100% conservation. Phylogenetic analysis supports the hypothesis that dinoflagellates and Plasmodium are closely related. The protein encoded by the pfGAPDH was expressed recombinantly in Escherichia coli and exhibited enzymatic activity with NAD(+) but not with NADP(+) as cofactor. Antiserum raised against the recombinantly expressed enzyme detected specifically all developmental stages of cultured P. falciparum blood-stage parasites.
Collapse
Affiliation(s)
- C A Daubenberger
- Swiss Tropical Institute, Molecular Immunology, Socinstrasse 57, 4002, Basel, Switzerland.
| | | | | | | | | | | |
Collapse
|