1
|
Lorenzen E, Contreras V, Olsen AW, Andersen P, Desjardins D, Rosenkrands I, Juel HB, Delache B, Langlois S, Delaugerre C, Joubert C, Dereuddre-Bosquet N, Bébéar C, De Barbeyrac B, Touati A, McKay PF, Shattock RJ, Le Grand R, Follmann F, Dietrich J. Multi-component prime-boost Chlamydia trachomatis vaccination regimes induce antibody and T cell responses and accelerate clearance of infection in a non-human primate model. Front Immunol 2022; 13:1057375. [PMID: 36505459 PMCID: PMC9726737 DOI: 10.3389/fimmu.2022.1057375] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/31/2022] [Indexed: 11/24/2022] Open
Abstract
It is of international priority to develop a vaccine against sexually transmitted Chlamydia trachomatis infections to combat the continued global spread of the infection. The optimal immunization strategy still remains to be fully elucidated. The aim of this study was to evaluate immunization strategies in a nonhuman primate (NHP) model. Cynomolgus macaques (Macaqua fascicularis) were immunized following different multi-component prime-boost immunization-schedules and subsequently challenged with C. trachomatis SvD in the lower genital tract. The immunization antigens included the recombinant protein antigen CTH522 adjuvanted with CAF01 or aluminium hydroxide, MOMP DNA antigen and MOMP vector antigens (HuAd5 MOMP and MVA MOMP). All antigen constructs were highly immunogenic raising significant systemic C. trachomatis-specific IgG responses. In particularly the CTH522 protein vaccinated groups raised a fast and strong pecificsIgG in serum. The mapping of specific B cell epitopes within the MOMP showed that all vaccinated groups, recognized epitopes near or within the variable domains (VD) of MOMP, with a consistent VD4 response in all animals. Furthermore, serum from all vaccinated groups were able to in vitro neutralize both SvD, SvE and SvF. Antibody responses were reflected on the vaginal and ocular mucosa, which showed detectable levels of IgG. Vaccines also induced C. trachomatis-specific cell mediated responses, as shown by in vitro stimulation and intracellular cytokine staining of peripheral blood mononuclear cells (PBMCs). In general, the protein (CTH522) vaccinated groups established a multifunctional CD4 T cell response, whereas the DNA and Vector vaccinated groups also established a CD8 T cells response. Following vaginal challenge with C. trachomatis SvD, several of the vaccinated groups showed accelerated clearance of the infection, but especially the DNA group, boosted with CAF01 adjuvanted CTH522 to achieve a balanced CD4/CD8 T cell response combined with an IgG response, showed accelerated clearance of the infection.
Collapse
Affiliation(s)
- Emma Lorenzen
- Chlamydia Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Vanessa Contreras
- Université Paris-Saclay, Inserm, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Anja W. Olsen
- Chlamydia Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Peter Andersen
- Novo Nordisk Foundation, Infectious Disease, Hellerup, Denmark
| | - Delphine Desjardins
- Université Paris-Saclay, Inserm, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Ida Rosenkrands
- Chlamydia Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Helene Bæk Juel
- Novo Nordisk Foundation, Center for Basic Metabolic Research, Copenhagen, Denmark
| | - Benoit Delache
- Université Paris-Saclay, Inserm, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Sebastien Langlois
- Université Paris-Saclay, Inserm, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Constance Delaugerre
- Laboratory of Virology, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris, Université de Paris, Paris Cité, Paris, France
| | - Christophe Joubert
- Université Paris-Saclay, Inserm, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Nathalie Dereuddre-Bosquet
- Université Paris-Saclay, Inserm, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Cécile Bébéar
- Bordeaux University Hopsital, Bacteriology Department, National Reference Centre for bacterial Sexually Transmitted Infections, Bordeaux, France
| | - Bertille De Barbeyrac
- Bordeaux University Hopsital, Bacteriology Department, National Reference Centre for bacterial Sexually Transmitted Infections, Bordeaux, France
| | - Arabella Touati
- Bordeaux University Hopsital, Bacteriology Department, National Reference Centre for bacterial Sexually Transmitted Infections, Bordeaux, France
| | - Paul F. McKay
- Department of Medicine, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Robin J. Shattock
- Department of Medicine, Imperial College London, St Mary’s Campus, London, United Kingdom
| | - Roger Le Grand
- Université Paris-Saclay, Inserm, Commissariat à l'Énergie Atomique et aux Énergies Alternatives (CEA), Center for Immunology of Viral, Auto-immune, Hematological and Bacterial diseases (IMVA-HB/IDMIT), Fontenay-aux-Roses & Le Kremlin-Bicêtre, France
| | - Frank Follmann
- Chlamydia Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | - Jes Dietrich
- Chlamydia Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark,*Correspondence: Jes Dietrich,
| |
Collapse
|
2
|
Chavda VP, Pandya A, Kypreos E, Patravale V, Apostolopoulos V. Chlamydia trachomatis: quest for an eye-opening vaccine breakthrough. Expert Rev Vaccines 2022; 21:771-781. [PMID: 35470769 DOI: 10.1080/14760584.2022.2061461] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Chlamydia trachomatis, commonly referred to as chlamydia (a bacterium), is a common sexually transmitted infection, and if attended to early, it can be treatable. However, if left untreated it can lead to serious consequences. C. trachomatis infects both females and males although its occurrence in females is more common, and it can spread to the eyes causing disease and in some case blindness. AREA COVERED With ongoing attempts in the most impoverished regions of the country, trachoma will be eradicated as a blinding disease by the year 2022. A prophylactic vaccine candidate with established safety and efficacy is a cogent tool to achieve this goal. This manuscript covers the vaccine development programs for chlamydial infection. EXPERT OPINION Currently, the Surgery Antibiotics Facial Environmental (SAFE) program is being implemented in endemic countries in order to reduce transmission and control of the disease. Vaccines have been shown over the years to protect against infectious diseases. Charge variant-based adjuvant can also be used for the successful delivery of chlamydial specific antigen for efficient vaccine delivery through nano delivery platform. Thus, a vaccine against C. trachomatis would be of great public health benefit.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad India
| | - Anjali Pandya
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai India
| | - Erica Kypreos
- Department of Immunology, Institute for Health and Sport, Victoria University, Melbourne VIC Australia
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai India
| | - Vasso Apostolopoulos
- Department of Immunology, Institute for Health and Sport, Victoria University, Melbourne VIC Australia
| |
Collapse
|
3
|
Blakney AK, McKay PF, Hu K, Samnuan K, Jain N, Brown A, Thomas A, Rogers P, Polra K, Sallah H, Yeow J, Zhu Y, Stevens MM, Geall A, Shattock RJ. Polymeric and lipid nanoparticles for delivery of self-amplifying RNA vaccines. J Control Release 2021; 338:201-210. [PMID: 34418521 PMCID: PMC8412240 DOI: 10.1016/j.jconrel.2021.08.029] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 01/09/2023]
Abstract
Self-amplifying RNA (saRNA) is a next-generation vaccine platform, but like all nucleic acids, requires a delivery vehicle to promote cellular uptake and protect the saRNA from degradation. To date, delivery platforms for saRNA have included lipid nanoparticles (LNP), polyplexes and cationic nanoemulsions; of these LNP are the most clinically advanced with the recent FDA approval of COVID-19 based-modified mRNA vaccines. While the effect of RNA on vaccine immunogenicity is well studied, the role of biomaterials in saRNA vaccine effectiveness is under investigated. Here, we tested saRNA formulated with either pABOL, a bioreducible polymer, or LNP, and characterized the protein expression and vaccine immunogenicity of both platforms. We observed that pABOL-formulated saRNA resulted in a higher magnitude of protein expression, but that the LNP formulations were overall more immunogenic. Furthermore, we observed that both the helper phospholipid and route of administration (intramuscular versus intranasal) of LNP impacted the vaccine immunogenicity of two model antigens (influenza hemagglutinin and SARS-CoV-2 spike protein). We observed that LNP administered intramuscularly, but not pABOL or LNP administered intranasally, resulted in increased acute interleukin-6 expression after vaccination. Overall, these results indicate that delivery systems and routes of administration may fulfill different delivery niches within the field of saRNA genetic medicines.
Collapse
Affiliation(s)
- Anna K. Blakney
- The University of British Columbia, Michael Smith Laboratories, School of Biomedical Engineering, Vancouver, BC V6T1Z4, Canada,Imperial College London, Department of Infectious Disease, London W21PG, United Kingdom,Corresponding authors at: Imperial College London, Department of Infectious Disease, London W21PG, United Kingdom
| | - Paul F. McKay
- Imperial College London, Department of Infectious Disease, London W21PG, United Kingdom
| | - Kai Hu
- Imperial College London, Department of Infectious Disease, London W21PG, United Kingdom
| | - Karnyart Samnuan
- Imperial College London, Department of Infectious Disease, London W21PG, United Kingdom
| | - Nikita Jain
- Precision NanoSystems Inc., Vancouver, BC V6P6T7, Canada
| | - Andrew Brown
- Precision NanoSystems Inc., Vancouver, BC V6P6T7, Canada
| | - Anitha Thomas
- Precision NanoSystems Inc., Vancouver, BC V6P6T7, Canada
| | - Paul Rogers
- Imperial College London, Department of Infectious Disease, London W21PG, United Kingdom
| | - Krunal Polra
- Imperial College London, Department of Infectious Disease, London W21PG, United Kingdom
| | - Hadijatou Sallah
- Imperial College London, Department of Infectious Disease, London W21PG, United Kingdom
| | - Jonathan Yeow
- Imperial College London, Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, London SW72BU, United Kingdom
| | - Yunqing Zhu
- Imperial College London, Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, London SW72BU, United Kingdom,School of Materials Science and Engineering, Tongji University, Shanghai 200092, China
| | - Molly M. Stevens
- Imperial College London, Department of Materials, Department of Bioengineering, Institute of Biomedical Engineering, London SW72BU, United Kingdom
| | - Andrew Geall
- Precision NanoSystems Inc., Vancouver, BC V6P6T7, Canada
| | - Robin J. Shattock
- Imperial College London, Department of Infectious Disease, London W21PG, United Kingdom,Corresponding authors at: Imperial College London, Department of Infectious Disease, London W21PG, United Kingdom
| |
Collapse
|
4
|
Murray SM, McKay PF. Chlamydia trachomatis: Cell biology, immunology and vaccination. Vaccine 2021; 39:2965-2975. [PMID: 33771390 DOI: 10.1016/j.vaccine.2021.03.043] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023]
Abstract
Chlamydia trachomatis is the causative agent of a highly prevalent sexually transmitted bacterial disease and is associated with a number of severe disease complications. Current therapy options are successful at treating disease, but patients are left without protective immunity and do not benefit the majority asymptomatic patients who do not seek treatment. As such, there is a clear need for a broad acting, protective vaccine that can prevent transmission and protect against symptomatic disease presentation. There are three key elements that underlie successful vaccine development: 1) Chlamydia biology and immune-evasion adaptations, 2) the correlates of protection that prevent disease in natural and experimental infection, 3) reflection upon the evidence provided by previous vaccine attempts. In this review, we give an overview of the unique intra-cellular biology of C. trachomatis and give insight into the dynamic combination of adaptations that allow Chlamydia to subvert host immunity and survive within the cell. We explore the current understanding of chlamydial immunity in animal models and in humans and characterise the key immune correlates of protection against infection. We discuss in detail the specific immune interactions involved in protection, with relevance placed on the CD4+ T lymphocyte and B lymphocyte responses that are key to pathogen clearance. Finally, we provide a timeline of C. trachomatis vaccine research to date and evaluate the successes and failures in development so far. With insight from these three key elements of research, we suggest potential solutions for chlamydial vaccine development and promising avenues for further exploration.
Collapse
Affiliation(s)
- Sam M Murray
- Department of Infectious Diseases, Imperial College London, Norfolk Place, London W2 1PG, UK.
| | - Paul F McKay
- Department of Infectious Diseases, Imperial College London, Norfolk Place, London W2 1PG, UK.
| |
Collapse
|
5
|
Blakney AK, McKay PF, Bouton CR, Hu K, Samnuan K, Shattock RJ. Innate Inhibiting Proteins Enhance Expression and Immunogenicity of Self-Amplifying RNA. Mol Ther 2021; 29:1174-1185. [PMID: 33352107 PMCID: PMC7935664 DOI: 10.1016/j.ymthe.2020.11.011] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 10/29/2020] [Accepted: 11/05/2020] [Indexed: 12/13/2022] Open
Abstract
Self-amplifying RNA (saRNA) is a cutting-edge platform for both nucleic acid vaccines and therapeutics. saRNA is self-adjuvanting, as it activates types I and III interferon (IFN), which enhances the immunogenicity of RNA vaccines but can also lead to inhibition of translation. In this study, we screened a library of saRNA constructs with cis-encoded innate inhibiting proteins (IIPs) and determined the effect on protein expression and immunogenicity. We observed that the PIV-5 V and Middle East respiratory syndrome coronavirus (MERS-CoV) ORF4a proteins enhance protein expression 100- to 500-fold in vitro in IFN-competent HeLa and MRC5 cells. We found that the MERS-CoV ORF4a protein partially abates dose nonlinearity in vivo, and that ruxolitinib, a potent Janus kinase (JAK)/signal transducer and activator of transcription (STAT) inhibitor, but not the IIPs, enhances protein expression of saRNA in vivo. Both the PIV-5 V and MERS-CoV ORF4a proteins were found to enhance the percentage of resident cells in human skin explants expressing saRNA and completely rescued dose nonlinearity of saRNA. Finally, we observed that the MERS-CoV ORF4a increased the rabies virus (RABV)-specific immunoglobulin G (IgG) titer and neutralization half-maximal inhibitory concentration (IC50) by ∼10-fold in rabbits, but not in mice or rats. These experiments provide a proof of concept that IIPs can be directly encoded into saRNA vectors and effectively abate the nonlinear dose dependency and enhance immunogenicity.
Collapse
Affiliation(s)
- Anna K Blakney
- Department of Infectious Disease, Imperial College London, London W21PG, UK.
| | - Paul F McKay
- Department of Infectious Disease, Imperial College London, London W21PG, UK
| | - Clément R Bouton
- Department of Infectious Disease, Imperial College London, London W21PG, UK
| | - Kai Hu
- Department of Infectious Disease, Imperial College London, London W21PG, UK
| | - Karnyart Samnuan
- Department of Infectious Disease, Imperial College London, London W21PG, UK
| | - Robin J Shattock
- Department of Infectious Disease, Imperial College London, London W21PG, UK.
| |
Collapse
|
6
|
Self-amplifying RNA SARS-CoV-2 lipid nanoparticle vaccine candidate induces high neutralizing antibody titers in mice. Nat Commun 2020; 11:3523. [PMID: 32647131 PMCID: PMC7347890 DOI: 10.1038/s41467-020-17409-9] [Citation(s) in RCA: 320] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023] Open
Abstract
The spread of the SARS-CoV-2 into a global pandemic within a few months of onset motivates the development of a rapidly scalable vaccine. Here, we present a self-amplifying RNA encoding the SARS-CoV-2 spike protein encapsulated within a lipid nanoparticle (LNP) as a vaccine. We observe remarkably high and dose-dependent SARS-CoV-2 specific antibody titers in mouse sera, as well as robust neutralization of both a pseudo-virus and wild-type virus. Upon further characterization we find that the neutralization is proportional to the quantity of specific IgG and of higher magnitude than recovered COVID-19 patients. saRNA LNP immunizations induce a Th1-biased response in mice, and there is no antibody-dependent enhancement (ADE) observed. Finally, we observe high cellular responses, as characterized by IFN-γ production, upon re-stimulation with SARS-CoV-2 peptides. These data provide insight into the vaccine design and evaluation of immunogenicity to enable rapid translation to the clinic.
Collapse
|
7
|
Blakney AK, Zhu Y, McKay PF, Bouton C, Yeow J, Tang J, Hu K, Samnuan K, Grigsby CL, Shattock RJ, Stevens MM. Big Is Beautiful: Enhanced saRNA Delivery and Immunogenicity by a Higher Molecular Weight, Bioreducible, Cationic Polymer. ACS NANO 2020; 14:5711-5727. [PMID: 32267667 PMCID: PMC7304921 DOI: 10.1021/acsnano.0c00326] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/08/2020] [Indexed: 05/18/2023]
Abstract
Self-amplifying RNA (saRNA) vaccines are highly advantageous, as they result in enhanced protein expression compared to mRNA (mRNA), thus minimizing the required dose. However, previous delivery strategies were optimized for siRNA or mRNA and do not necessarily deliver saRNA efficiently due to structural differences of these RNAs, thus motivating the development of saRNA delivery platforms. Here, we engineer a bioreducible, linear, cationic polymer called "pABOL" for saRNA delivery and show that increasing its molecular weight enhances delivery both in vitro and in vivo. We demonstrate that pABOL enhances protein expression and cellular uptake via both intramuscular and intradermal injection compared to commercially available polymers in vivo and that intramuscular injection confers complete protection against influenza challenge. Due to the scalability of polymer synthesis and ease of formulation preparation, we anticipate that this polymer is highly clinically translatable as a delivery vehicle for saRNA for both vaccines and therapeutics.
Collapse
Affiliation(s)
- Anna K. Blakney
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Yunqing Zhu
- Department
of Materials, Department of Bioengineering, Institute of Biomedical
Engineering, Imperial College London, London, SW7 2AZ, U.K.
- School
of Materials Science and Engineering, Tongji
University, Shanghai, 200092, China
| | - Paul F. McKay
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Clément
R. Bouton
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Jonathan Yeow
- Department
of Materials, Department of Bioengineering, Institute of Biomedical
Engineering, Imperial College London, London, SW7 2AZ, U.K.
| | - Jiaqing Tang
- Department
of Materials, Department of Bioengineering, Institute of Biomedical
Engineering, Imperial College London, London, SW7 2AZ, U.K.
| | - Kai Hu
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Karnyart Samnuan
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Christopher L. Grigsby
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 65, Sweden
| | - Robin J. Shattock
- Department
of Infectious Diseases, Imperial College
London, Norfolk Place, London, W2 1PG, U.K.
| | - Molly M. Stevens
- Department
of Materials, Department of Bioengineering, Institute of Biomedical
Engineering, Imperial College London, London, SW7 2AZ, U.K.
- Department
of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, 171 65, Sweden
| |
Collapse
|
8
|
Sathish S, Ishizu N, Shen AQ. Air Plasma-Enhanced Covalent Functionalization of Poly(methyl methacrylate): High-Throughput Protein Immobilization for Miniaturized Bioassays. ACS APPLIED MATERIALS & INTERFACES 2019; 11:46350-46360. [PMID: 31722179 DOI: 10.1021/acsami.9b14631] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Miniaturized systems, such as integrated microarray and microfluidic devices, are constantly being developed to satisfy the growing demand for sensitive and high-throughput biochemical screening platforms. Owing to its recyclability, and robust mechanical and optical properties, poly(methyl methacrylate) (PMMA) has become the most sought after material for the large-scale fabrication of these platforms. However, the chemical inertness of PMMA entails the use of complex chemical surface treatments for covalent immobilization of proteins. In addition to being hazardous and incompatible for large-scale operations, conventional biofunctionalization strategies pose high risks of compromising the biomolecular conformations, as well as the stability of PMMA. By exploiting radio frequency (RF) air plasma and standard 1-ethyl-3-(3-(dimethylamino)propyl) carbodiimide (EDC) and N-hydroxysuccinimide (NHS) chemistry in tandem, we demonstrate a simple yet scalable PMMA functionalization strategy for covalent immobilization (chemisorption) of proteins, such as green fluorescent protein (GFP), while preserving the structural integrities of the proteins and PMMA. The surface density of chemisorbed GFP is shown to be highly dependent on the air plasma energy, initial GFP concentration, and buffer pH, where a maximum GFP surface density of 4 × 10-7 mol/m2 is obtained, when chemisorbed on EDC-NHS-activated PMMA exposed to 27 kJ of air plasma, at pH 7.4. Furthermore, antibody-binding studies validate the preserved biofunctionality of the chemisorbed GFP molecules. Finally, the coupled air plasma and EDC-NHS PMMA biofunctionalization strategy is used to fabricate microfluidic antibody assay devices to detect clinically significant concentrations of Chlamydia trachomatis specific antibodies. By coupling our scalable and tailored air plasma-enhanced PMMA biofunctionalization strategy with microfluidics, we elucidate the potential of fabricating sensitive, reproducible, and sustainable high-throughput protein screening systems, without the need for harsh chemicals and complex instrumentation.
Collapse
Affiliation(s)
- Shivani Sathish
- Micro/Bio/Nanofluidics Unit , Okinawa Institute of Science and Technology Graduate University , 1919-1 Tancha , Onna-son, Kunigami-gun , Okinawa 904-0495 , Japan
| | - Noriko Ishizu
- Mechanical Engineering and Microfabrication Support Section , Okinawa Institute of Science and Technology Graduate University , 1919-1 Tancha , Onna-son, Kunigami-gun , Okinawa 904-0495 , Japan
| | - Amy Q Shen
- Micro/Bio/Nanofluidics Unit , Okinawa Institute of Science and Technology Graduate University , 1919-1 Tancha , Onna-son, Kunigami-gun , Okinawa 904-0495 , Japan
| |
Collapse
|
9
|
Li Y, Wang C, Sun Z, Xiao J, Yan X, Chen Y, Yu J, Wu Y. Simultaneous Intramuscular And Intranasal Administration Of Chitosan Nanoparticles-Adjuvanted Chlamydia Vaccine Elicits Elevated Protective Responses In The Lung. Int J Nanomedicine 2019; 14:8179-8193. [PMID: 31632026 PMCID: PMC6790120 DOI: 10.2147/ijn.s218456] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 09/11/2019] [Indexed: 12/13/2022] Open
Abstract
Background Chlamydia psittaci is a zoonotic bacteria closely associated with psittacosis/ornithosis. Vaccination has been recognized as the best way to inhibit the spread of C. psittaci due to the majority ignored of infections. The optimal Chlamydia vaccine was obstructed by the defect of single immunization route and the lack of availability of nontoxic and valid adjuvants. Methods In this study, we developed a novel immunization strategy, simultaneous (SIM) intramuscular (IM) and intranasal (IN) administration of a C. psittaci antigens (Ags) adjuvanted with chitosan nanoparticles (CNPs). And SIM-CNPs-Ags were used to determine the different types of immune response and the protective role in vivo. Results CNPs-Ags with zeta-potential values of 13.12 mV and of 276.1 nm showed excellent stability and optimal size for crossing the mucosal barrier with high 71.7% encapsulation efficiency. SIM-CPN-Ags mediated stronger humoral and mucosal responses by producing meaningfully high levels of IgG and secretory IgA (sIgA) antibodies. The SIM route also led to Ags-specific T-cell responses and increased IFN-γ, IL-2, TNF-α and IL-17A in the splenocyte supernatants. Following respiratory infection with C. psittaci, we found that SIM immunization remarkably reduced bacterial load and the degree of inflammation in the infected lungs and made for a lower level of IFN-γ, TNF-α and IL-6. Furthermore, SIM vaccination with CNPs-Ags had obviously inhibited C. psittaci disseminating to various organs in vivo. Conclusion SIM immunization with CNPs-adjuvanted C. psittaci Ags may present a novel strategy for the development of a vaccine against the C. psittaci infection.
Collapse
Affiliation(s)
- Yumeng Li
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institution of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang 421001, People's Republic of China
| | - Chuan Wang
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institution of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang 421001, People's Republic of China
| | - Zhenjie Sun
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institution of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang 421001, People's Republic of China
| | - Jian Xiao
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institution of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang 421001, People's Republic of China
| | - Xiaoliang Yan
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institution of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang 421001, People's Republic of China
| | - Yuqing Chen
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institution of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang 421001, People's Republic of China
| | - Jian Yu
- Department of Experimental Zoology, Hengyang Medical College, University of South China, Hengyang 421001, People's Republic of China
| | - Yimou Wu
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Institution of Pathogenic Biology, Hengyang Medical College, University of South China, Hengyang 421001, People's Republic of China
| |
Collapse
|
10
|
Blakney AK, McKay PF, Yus BI, Aldon Y, Shattock RJ. Inside out: optimization of lipid nanoparticle formulations for exterior complexation and in vivo delivery of saRNA. Gene Ther 2019; 26:363-372. [PMID: 31300730 PMCID: PMC6760535 DOI: 10.1038/s41434-019-0095-2] [Citation(s) in RCA: 146] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/18/2019] [Accepted: 06/28/2019] [Indexed: 12/17/2022]
Abstract
Self-amplifying RNA (saRNA) is a promising biotherapeutic tool that has been used as a vaccine against both infectious diseases and cancer. saRNA has been shown to induce protein expression for up to 60 days and elicit immune responses with lower dosing than messenger RNA (mRNA). Because saRNA is a large (~9500 nt), negatively charged molecule, it requires a delivery vehicle for efficient cellular uptake and degradation protection. Lipid nanoparticles (LNPs) have been widely used for RNA formulations, where the prevailing paradigm is to encapsulate RNA within the particle, including the first FDA-approved small-interfering siRNA therapy. Here, we compared LNP formulations with cationic and ionizable lipids with saRNA either on the interior or exterior of the particle. We show that LNPs formulated with cationic lipids protect saRNA from RNAse degradation, even when it is adsorbed to the surface. Furthermore, cationic LNPs deliver saRNA equivalently to particles formulated with saRNA encapsulated in an ionizable lipid particle, both in vitro and in vivo. Finally, we show that cationic and ionizable LNP formulations induce equivalent antibodies against HIV-1 Env gp140 as a model antigen. These studies establish formulating saRNA on the surface of cationic LNPs as an alternative to the paradigm of encapsulating RNA.
Collapse
Affiliation(s)
- Anna K Blakney
- Department of Medicine, Imperial College London, London, UK
| | - Paul F McKay
- Department of Medicine, Imperial College London, London, UK
| | | | - Yoann Aldon
- Department of Medicine, Imperial College London, London, UK
| | | |
Collapse
|
11
|
Blakney AK, McKay PF, Christensen D, Yus BI, Aldon Y, Follmann F, Shattock RJ. Effects of cationic adjuvant formulation particle type, fluidity and immunomodulators on delivery and immunogenicity of saRNA. J Control Release 2019; 304:65-74. [PMID: 31071377 DOI: 10.1016/j.jconrel.2019.04.043] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/07/2019] [Accepted: 04/29/2019] [Indexed: 01/07/2023]
Abstract
Self-amplifying RNA (saRNA) is well suited as a vaccine platform against chlamydia, as it is relatively affordable and scalable, has been shown to induce immunity against multivalent antigens, and can result in protein expression for up to 60 days. Cationic adjuvant formulations (CAFs) have been previously investigated as an adjuvant for protein subunit vaccines; here we optimize the CAFs for delivery of saRNA in vivo and observe the immunogenicity profile in the context of both cellular and humoral immunity against the major outer membrane protein (MOMP) of Chlamydia trachomatis. We tested both liposomal and emulsion based CAFs with solid and fluid phase lipids, with or without the TLR agonists R848 and 3M-052, for in vitro transfection efficiency and cytotoxicity. We then optimized the RNA/delivery system ratio for in vivo delivery using saRNA coding for firefly luciferase (fLuc) as a reporter protein in vivo. We observed that while the fluid phase liposome formulations showed the highest in vitro transfection efficiency, the fluid and solid phase liposomes had equivalent luciferase expression in vivo. Incorporation of R848 or 3M-052 into the formulation was not observed to affect the delivery efficiency of saRNA either in vitro or in vivo. MOMP-encoding saRNA complexed with CAFs resulted in both MOMP-specific cellular and humoral immunity, and while there was a slight enhancement of IFN-γ+ T-cell responses when R848 was incorporated into the formulation, the self-adjuvanting effects of RNA appeared to dominate the immune response. These studies establish that CAFs are efficient delivery vehicles for saRNA both for in vitro transfections and in vivo immunogenicity and generate cellular and humoral responses that are proportionate to protein expression.
Collapse
Affiliation(s)
- Anna K Blakney
- Department of Medicine, Imperial College London, London, UK
| | - Paul F McKay
- Department of Medicine, Imperial College London, London, UK
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | | | - Yoann Aldon
- Department of Medicine, Imperial College London, London, UK
| | - Frank Follmann
- Department of Infectious Disease Immunology, Statens Serum Institut, Copenhagen, Denmark
| | | |
Collapse
|
12
|
Badamchi-Zadeh A, Moynihan KD, Larocca RA, Aid M, Provine NM, Iampietro MJ, Kinnear E, Penaloza-MacMaster P, Abbink P, Blass E, Tregoning JS, Irvine DJ, Barouch DH. Combined HDAC and BET Inhibition Enhances Melanoma Vaccine Immunogenicity and Efficacy. THE JOURNAL OF IMMUNOLOGY 2018; 201:2744-2752. [PMID: 30249811 DOI: 10.4049/jimmunol.1800885] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 08/22/2018] [Indexed: 12/12/2022]
Abstract
The combined inhibition of histone deacetylases (HDAC) and the proteins of the bromodomain and extraterminal (BET) family have recently shown therapeutic efficacy against melanoma, pancreatic ductal adenocarcinoma, testicular, and lymphoma cancers in murine studies. However, in such studies, the role of the immune system in therapeutically controlling these cancers has not been explored. We sought to investigate the effect of the HDAC inhibitor romidepsin (RMD) and the BET inhibitor IBET151, both singly and in combination, on vaccine-elicited immune responses. C57BL/6 mice were immunized with differing vaccine systems (adenoviral, protein) in prime-boost regimens under treatment with RMD, IBET151, or RMD+IBET151. The combined administration of RMD+IBET151 during vaccination resulted in a significant increase in the frequency and number of Ag-specific CD8+ T cells. RMD+IBET151 treatment significantly increased the frequency of vaccine-elicited IFN-γ+ splenic CD8+ T cells and conferred superior therapeutic and prophylactic protection against B16-OVA melanoma. RNA sequencing analyses revealed strong transcriptional similarity between RMD+IBET151 and untreated Ag-specific CD8+ T cells except in apoptosis and IL-6 signaling-related genes that were differentially expressed. Serum IL-6 was significantly increased in vivo following RMD+IBET151 treatment, with recombinant IL-6 administration replicating the effect of RMD+IBET151 treatment on vaccine-elicited CD8+ T cell responses. IL-6 sufficiency for protection was not assessed. Combined HDAC and BET inhibition resulted in greater vaccine-elicited CD8+ T cell responses and enhanced therapeutic and prophylactic protection against B16-OVA melanoma. Increased IL-6 production and the differential expression of pro- and anti-apoptotic genes following RMD+IBET151 treatment are likely contributors to the enhanced cancer vaccine responses.
Collapse
Affiliation(s)
- Alexander Badamchi-Zadeh
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Kelly D Moynihan
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139
| | - Rafael A Larocca
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Malika Aid
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Nicholas M Provine
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - M Justin Iampietro
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Ekaterina Kinnear
- Department of Medicine, Imperial College London, London W2 1PG, United Kingdom
| | - Pablo Penaloza-MacMaster
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Peter Abbink
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - Eryn Blass
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215
| | - John S Tregoning
- Department of Medicine, Imperial College London, London W2 1PG, United Kingdom
| | - Darrell J Irvine
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139.,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139; and.,Howard Hughes Medical Institute, Chevy Chase, MD 20815
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215; .,Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139; and
| |
Collapse
|
13
|
The Chlamydia trachomatis PmpD adhesin forms higher order structures through disulphide-mediated covalent interactions. PLoS One 2018; 13:e0198662. [PMID: 29912892 PMCID: PMC6005502 DOI: 10.1371/journal.pone.0198662] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 05/23/2018] [Indexed: 12/27/2022] Open
Abstract
Chlamydia trachomatis (Ct) is the most common sexually transmitted bacterial pathogen, and the leading cause of infectious blindness worldwide. We have recently shown that immunization with the highly conserved antigenic passenger domain of recombinant Ct polymorphic membrane protein D (rPmpD) is protective in the mouse model of Ct genital tract infection, and previously, that ocular anti-rPmpD antibodies are elicited following vaccination. However, the mechanisms governing the assembly and structure-function relationship of PmpD are unknown. Here, we provide a biophysical analysis of this immunogenic 65 kDa passenger domain fragment of PmpD. Using differential cysteine labeling coupled with LC-MS/MS analysis, we show that widespread intra- and intermolecular disulphide interactions play important roles in the preservation of native monomeric secondary structure and the formation of higher-order oligomers. While it has been proposed that FxxN and GGA(I, L,V) repeat motifs in the Pmp21 ortholog in Chlamydia pneumoniae mediate self-interaction, no such role has previously been identified for cysteine residues in chlamydial Pmps. Further characterisation reveals that oligomeric proteoforms and rPmpD monomers adopt β-sheet folds, consistent with previously described Gram-negative bacterial type V secretion systems (T5SSs). We also highlight adhesin-like properties of rPmpD, showing that both soluble rPmpD and anti-rPmpD serum from immunized mice abrogate binding of rPmpD-coated beads to mammalian cells in a dose-dependent fashion. Hence, our study provides further evidence that chlamydial Pmps may function as adhesins, while elucidating yet another important mechanism of self-association of bacterial T5SS virulence factors that may be unique to the Chlamydiaceae.
Collapse
|
14
|
Therapeutic Efficacy of Vectored PGT121 Gene Delivery in HIV-1-Infected Humanized Mice. J Virol 2018; 92:JVI.01925-17. [PMID: 29321310 PMCID: PMC5972893 DOI: 10.1128/jvi.01925-17] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/04/2018] [Indexed: 01/15/2023] Open
Abstract
Broadly neutralizing antibodies (bNAbs) are being explored for HIV-1 prevention and cure strategies. However, administration of purified bNAbs poses challenges in resource-poor settings, where the HIV-1 disease burden is greatest. In vivo vector-based production of bNAbs represents an alternative strategy. We investigated adenovirus serotype 5 (Ad5) and adeno-associated virus serotype 1 (AAV1) vectors to deliver the HIV-1-specific bNAb PGT121 in wild-type and immunocompromised C57BL/6 mice as well as in HIV-1-infected bone marrow-liver-thymus (BLT) humanized mice. Ad5.PGT121 and AAV1.PGT121 produced functional antibody in vivo. Ad5.PGT121 produced PGT121 rapidly within 6 h, whereas AAV1.PGT121 produced detectable PGT121 in serum by 72 h. Serum PGT121 levels were rapidly reduced by the generation of anti-PGT121 antibodies in immunocompetent mice but were durably maintained in immunocompromised mice. In HIV-1-infected BLT humanized mice, Ad5.PGT121 resulted in a greater reduction of viral loads than did AAV1.PGT121. Ad5.PGT121 also led to more-sustained virologic control than purified PGT121 IgG. Ad5.PGT121 afforded more rapid, robust, and durable antiviral efficacy than AAV1.PGT121 and purified PGT121 IgG in HIV-1-infected humanized mice. Further evaluation of vector delivery of HIV-1 bNAbs is warranted, although approaches to prevent the generation of antiantibody responses may also be required. IMPORTANCE Broadly neutralizing antibodies (bNAbs) are being explored for HIV-1 prevention and cure strategies, but delivery of purified antibodies may prove challenging. We investigated adenovirus serotype 5 (Ad5) and adeno-associated virus serotype 1 (AAV1) vectors to deliver the HIV-1-specific bNAb PGT121. Ad5.PGT121 afforded more rapid, robust, and durable antiviral efficacy than AAV1.PGT121 and purified PGT121 IgG in HIV-1-infected humanized mice.
Collapse
|
15
|
Lactobacillus plantarum producing a Chlamydia trachomatis antigen induces a specific IgA response after mucosal booster immunization. PLoS One 2017; 12:e0176401. [PMID: 28467432 PMCID: PMC5415134 DOI: 10.1371/journal.pone.0176401] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 04/09/2017] [Indexed: 01/23/2023] Open
Abstract
Mucosal immunity is important for the protection against a wide variety of pathogens. Traditional vaccines administered via parenteral routes induce strong systemic immunity, but they often fail to generate mucosal IgA. In contrast, bacteria-based vaccines comprise an appealing strategy for antigen delivery to mucosal sites. Vaginal infection with Chlamydia trachomatis can develop into upper genital tract infections that can lead to infertility. Therefore, the development of an effective vaccine against Chlamydia is a high priority. In the present study, we have explored the use of a common lactic acid bacterium, Lactobacillus plantarum, as a vector for delivery of a C. trachomatis antigen to mucosal sites. The antigen, referred as Hirep2 (H2), was anchored to the surface of L. plantarum cells using an N-terminal lipoprotein anchor. After characterization, the constructed strain was used as an immunogenic agent in mice. We explored a heterologous prime-boost strategy, consisting of subcutaneous priming with soluble H2 antigen co-administered with CAF01 adjuvant, followed by an intranasal boost with H2-displaying L. plantarum. The results show that, when used as a booster, the recombinant L. plantarum strain was able to evoke cellular responses. Most importantly, booster immunization with the Lactobacillus-based vaccine induced generation of antigen-specific IgA in the vaginal cavity.
Collapse
|
16
|
Badamchi-Zadeh A, McKay PF, Korber BT, Barinaga G, Walters AA, Nunes A, Gomes JP, Follmann F, Tregoning JS, Shattock RJ. A Multi-Component Prime-Boost Vaccination Regimen with a Consensus MOMP Antigen Enhances Chlamydia trachomatis Clearance. Front Immunol 2016; 7:162. [PMID: 27199987 PMCID: PMC4848310 DOI: 10.3389/fimmu.2016.00162] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 04/15/2016] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND A vaccine for Chlamydia trachomatis is of urgent medical need. We explored bioinformatic approaches to generate an immunogen against C. trachomatis that would induce cross-serovar T-cell responses as (i) CD4(+) T cells have been shown in animal models and human studies to be important in chlamydial protection and (ii) antibody responses may be restrictive and serovar specific. METHODS A consensus antigen based on over 1,500 major outer membrane protein (MOMP) sequences provided high epitope coverage against the most prevalent C. trachomatis strains in silico. Having designed the T-cell immunogen, we assessed it for immunogenicity in prime-boost regimens. This consensus MOMP transgene was delivered using plasmid DNA, Human Adenovirus 5 (HuAd5) or modified vaccinia Ankara (MVA) vectors with or without MF59(®) adjuvanted recombinant MOMP protein. RESULTS Different regimens induced distinct immune profiles. The DNA-HuAd5-MVA-Protein vaccine regimen induced a cellular response with a Th1-biased serum antibody response, alongside high serum and vaginal MOMP-specific antibodies. This regimen significantly enhanced clearance against intravaginal C. trachomatis serovar D infection in both BALB/c and B6C3F1 mouse strains. This enhanced clearance was shown to be CD4(+) T-cell dependent. Future studies will need to confirm the specificity and precise mechanisms of protection. CONCLUSION A C. trachomatis vaccine needs to induce a robust cellular response with broad cross-serovar coverage and a heterologous prime-boost regimen may be an approach to achieve this.
Collapse
Affiliation(s)
| | - Paul F McKay
- Mucosal Infection and Immunity Group, Imperial College London , London , UK
| | - Bette T Korber
- Los Alamos National Laboratory, Theoretical Division , Los Alamos, NM , USA
| | - Guillermo Barinaga
- Mucosal Infection and Immunity Group, Imperial College London , London , UK
| | - Adam A Walters
- Mucosal Infection and Immunity Group, Imperial College London , London , UK
| | - Alexandra Nunes
- Department of Infectious Diseases, National Institute of Health , Lisbon , Portugal
| | - João Paulo Gomes
- Department of Infectious Diseases, National Institute of Health , Lisbon , Portugal
| | - Frank Follmann
- Chlamydia Vaccine Research, Department of Infectious Disease Immunology, Statens Serum Institute , Copenhagen , Denmark
| | - John S Tregoning
- Mucosal Infection and Immunity Group, Imperial College London , London , UK
| | - Robin J Shattock
- Mucosal Infection and Immunity Group, Imperial College London , London , UK
| |
Collapse
|
17
|
Transient CD4+ T Cell Depletion Results in Delayed Development of Functional Vaccine-Elicited Antibody Responses. J Virol 2016; 90:4278-4288. [PMID: 26865713 PMCID: PMC4836333 DOI: 10.1128/jvi.00039-16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 02/05/2016] [Indexed: 01/09/2023] Open
Abstract
UNLABELLED We have recently demonstrated that CD4(+)T cell help is required at the time of adenovirus (Ad) vector immunization for the development of functional CD8(+)T cell responses, but the temporal requirement for CD4(+)T cell help for the induction of antibody responses remains unclear. Here we demonstrate that induction of antibody responses in C57BL/6 mice can occur at a time displaced from the time of Ad vector immunization by depletion of CD4(+)T cells. Transient depletion of CD4(+)T cells at the time of immunization delays the development of antigen-specific antibody responses but does not permanently impair their development or induce tolerance against the transgene. Upon CD4(+)T cell recovery, transgene-specific serum IgG antibody titers develop and reach a concentration equivalent to that in undepleted control animals. These delayed antibody responses exhibit no functional defects with regard to isotype, functional avidity, expansion after boosting immunization, or the capacity to neutralize a simian immunodeficiency virus (SIV) Env-expressing pseudovirus. The development of this delayed transgene-specific antibody response is temporally linked to the expansion of de novo antigen-specific CD4(+)T cell responses, which develop after transient depletion of CD4(+)T cells. These data demonstrate that functional vaccine-elicited antibody responses can be induced even if CD4(+)T cell help is provided at a time markedly separated from the time of vaccination. IMPORTANCE CD4(+)T cells have a critical role in providing positive help signals to B cells, which promote robust antibody responses. The paradigm is that helper signals must be provided immediately upon antigen exposure, and their absence results in tolerance against the antigen. Here we demonstrate that, in contrast to the current model that the absence of CD4(+)T cell help at priming results in long-term antibody nonresponsiveness, antibody responses can be induced by adenovirus vector immunization or alum-adjuvanted protein immunization even if CD4(+)T cell help is not provided until >1 month after immunization. These data demonstrate that the time when CD4(+)T cell help signals must be provided is more dynamic and flexible than previously appreciated. These data suggest that augmentation of CD4(+)T cell helper function even after the time of vaccination can enhance vaccine-elicited antibody responses and thereby potentially enhance the immunogenicity of vaccines in immunocompromised individuals.
Collapse
|