1
|
Alam R, Samad A, Ahammad F, Nur SM, Alsaiari AA, Imon RR, Talukder MEK, Nain Z, Rahman MM, Mohammad F, Karpiński TM. In silico formulation of a next-generation multiepitope vaccine for use as a prophylactic candidate against Crimean-Congo hemorrhagic fever. BMC Med 2023; 21:36. [PMID: 36726141 PMCID: PMC9891764 DOI: 10.1186/s12916-023-02750-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Crimean-Congo hemorrhagic fever (CCHF) is a widespread disease transmitted to humans and livestock animals through the bite of infected ticks or close contact with infected persons' blood, organs, or other bodily fluids. The virus is responsible for severe viral hemorrhagic fever outbreaks, with a case fatality rate of up to 40%. Despite having the highest fatality rate of the virus, a suitable treatment option or vaccination has not been developed yet. Therefore, this study aimed to formulate a multiepitope vaccine against CCHF through computational vaccine design approaches. METHODS The glycoprotein, nucleoprotein, and RNA-dependent RNA polymerase of CCHF were utilized to determine immunodominant T- and B-cell epitopes. Subsequently, an integrative computational vaccinology approach was used to formulate a multi-epitopes vaccine candidate against the virus. RESULTS After rigorous assessment, a multiepitope vaccine was constructed, which was antigenic, immunogenic, and non-allergenic with desired physicochemical properties. Molecular dynamics (MD) simulations of the vaccine-receptor complex show strong stability of the vaccine candidates to the targeted immune receptor. Additionally, the immune simulation of the vaccine candidates found that the vaccine could trigger real-life-like immune responses upon administration to humans. CONCLUSIONS Finally, we concluded that the formulated multiepitope vaccine candidates would provide excellent prophylactic properties against CCHF.
Collapse
Affiliation(s)
- Rahat Alam
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
- Laboratory of Computational Biology, Biological Solution Centre (BioSol Centre), Jashore, 7408, Bangladesh
| | - Abdus Samad
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
- Laboratory of Computational Biology, Biological Solution Centre (BioSol Centre), Jashore, 7408, Bangladesh
| | - Foysal Ahammad
- Laboratory of Computational Biology, Biological Solution Centre (BioSol Centre), Jashore, 7408, Bangladesh
- Division of Biological and Biomedical Sciences (BBS), College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), 34110, Doha, Qatar
| | - Suza Mohammad Nur
- Department of Biochemistry, School of Medicine Case, Western Reserve University, Cleveland, OH, 44106, USA
| | - Ahad Amer Alsaiari
- College of Applied Medical Science, Clinical Laboratories Science Department, Taif University, Taif, 21944, Saudi Arabia
| | - Raihan Rahman Imon
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
- Laboratory of Computational Biology, Biological Solution Centre (BioSol Centre), Jashore, 7408, Bangladesh
| | - Md Enamul Kabir Talukder
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
- Laboratory of Computational Biology, Biological Solution Centre (BioSol Centre), Jashore, 7408, Bangladesh
| | - Zulkar Nain
- School of Biomedical Sciences, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Md Mashiar Rahman
- Department of Genetic Engineering and Biotechnology, Jashore University of Science and Technology, Jashore, 7408, Bangladesh
| | - Farhan Mohammad
- Division of Biological and Biomedical Sciences (BBS), College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), 34110, Doha, Qatar.
| | - Tomasz M Karpiński
- Chair and Department of Medical Microbiology, Poznań University of Medical Sciences, Rokietnicka 10, 60-806, Poznań, Poland.
| |
Collapse
|
2
|
Abstract
Influenza viruses cause respiratory tract infections, which lead to human disease outbreaks and pandemics. Influenza A virus (IAV) circulates in diverse animal species, predominantly aquatic birds. This often results in the emergence of novel viral strains causing severe human disease upon zoonotic transmission. Innate immune sensing of the IAV infection promotes host cell death and inflammatory responses to confer antiviral host defense. Dysregulated respiratory epithelial cell death and excessive proinflammatory responses drive immunopathology in highly pathogenic influenza infections. Here, we discuss the critical mechanisms regulating IAV-induced cell death and proinflammatory responses. We further describe the essential role of the Z-form nucleic acid sensor ZBP1/DAI and RIPK3 in triggering apoptosis, necroptosis, and pyroptosis during IAV infection and their impact on host defense and pathogenicity in vivo. We also discuss the functional importance of ZBP1-RIPK3 signaling in recent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other viral infections. Understanding these mechanisms of RNA virus-induced cytopathic and pathogenic inflammatory responses is crucial for targeting pathogenic lung infections and human respiratory illness.
Collapse
|
3
|
Ferrantelli F, Chiozzini C, Manfredi F, Giovannelli A, Leone P, Federico M. Simultaneous CD8 + T-Cell Immune Response against SARS-Cov-2 S, M, and N Induced by Endogenously Engineered Extracellular Vesicles in Both Spleen and Lungs. Vaccines (Basel) 2021; 9:240. [PMID: 33801926 PMCID: PMC7999804 DOI: 10.3390/vaccines9030240] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 12/29/2022] Open
Abstract
Most advanced vaccines against severe acute respiratory syndrome coronavirus (SARS-CoV)-2 are designed to induce antibodies against spike (S) protein. Differently, we developed an original strategy to induce CD8+ T cytotoxic lymphocyte (CTL) immunity based on in vivo engineering of extracellular vesicles (EVs). This is a new vaccination approach based on intramuscular injection of DNA expression vectors coding for a biologically inactive HIV-1 Nef protein (Nefmut) with an unusually high efficiency of incorporation into EVs, even when foreign polypeptides are fused to its C-terminus. Nanovesicles containing Nefmut-fused antigens released by muscle cells can freely circulate into the body and are internalized by antigen-presenting cells. Therefore, EV-associated antigens can be cross-presented to prime antigen-specific CD8+ T-cells. To apply this technology to a strategy of anti-SARS-CoV-2 vaccine, we designed DNA vectors expressing the products of fusion between Nefmut and different viral antigens, namely N- and C-terminal moieties of S (referred to as S1 and S2), M, and N. We provided evidence that all fusion products are efficiently uploaded in EVs. When the respective DNA vectors were injected in mice, a strong antigen-specific CD8+ T cell immunity became detectable in spleens and, most important, in lung airways. Co-injection of DNA vectors expressing the diverse SARS-CoV-2 antigens resulted in additive immune responses in both spleen and lungs. Hence, DNA vectors expressing Nefmut-based fusion proteins can be proposed for new anti-SARS-CoV-2 vaccine strategies.
Collapse
Affiliation(s)
- Flavia Ferrantelli
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (F.F.); (C.C.); (F.M.); (P.L.)
| | - Chiara Chiozzini
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (F.F.); (C.C.); (F.M.); (P.L.)
| | - Francesco Manfredi
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (F.F.); (C.C.); (F.M.); (P.L.)
| | - Andrea Giovannelli
- National Center for Animal Experimentation and Welfare, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy;
| | - Patrizia Leone
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (F.F.); (C.C.); (F.M.); (P.L.)
| | - Maurizio Federico
- National Center for Global Health, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy; (F.F.); (C.C.); (F.M.); (P.L.)
| |
Collapse
|
4
|
Kotomina T, Isakova-Sivak I, Kim KH, Park BR, Jung YJ, Lee Y, Mezhenskaya D, Matyushenko V, Kang SM, Rudenko L. Generation and Characterization of Universal Live-Attenuated Influenza Vaccine Candidates Containing Multiple M2e Epitopes. Vaccines (Basel) 2020; 8:vaccines8040648. [PMID: 33153089 PMCID: PMC7711583 DOI: 10.3390/vaccines8040648] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 10/20/2020] [Accepted: 10/29/2020] [Indexed: 12/24/2022] Open
Abstract
Influenza viruses constantly evolve, reducing the overall protective effect of routine vaccination campaigns. Many different strategies are being explored to design universal influenza vaccines capable of protecting against evolutionary diverged viruses. The ectodomain of influenza A M2e protein (M2e) is among the most promising targets for universal vaccine design. Here, we generated two recombinant live attenuated influenza vaccines (LAIVs) expressing additional four M2e tandem repeats (4M2e) from the N-terminus of the viral hemagglutinin (HA) protein, in an attempt to enhance the M2e-mediated cross-protection. The recombinant H1N1+4M2e and H3N2+4M2e viruses retained growth characteristics attributable to traditional LAIV viruses and induced robust influenza-specific antibody responses in BALB/c mice, although M2e-specific antibodies were raised only after two-dose vaccination with LAIV+4M2e viruses. Mice immunized with either LAIV or LAIV+4M2e viruses were fully protected against a panel of heterologous influenza challenge viruses suggesting that antibody and cell-mediated immunity contributed to the protection. The protective role of the M2e-specific antibody was seen in passive serum transfer experiments, where enhancement in the survival rates between classical LAIV and chimeric H3N2+4M2e LAIV was demonstrated for H3N2 and H5N1 heterologous challenge viruses. Overall, the results of our study suggest that M2e-specific antibodies induced by recombinant LAIV+4M2e in addition to cellular immunity by LAIV play an important role in conferring protection against heterologous viruses.
Collapse
Affiliation(s)
- Tatiana Kotomina
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg 197376, Russia; (T.K.); (D.M.); (V.M.); (L.R.)
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg 197376, Russia; (T.K.); (D.M.); (V.M.); (L.R.)
- Correspondence:
| | - Ki-Hye Kim
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (K.-H.K.); (B.R.P.); (Y.-J.J.); (Y.L.); (S.-M.K.)
| | - Bo Ryoung Park
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (K.-H.K.); (B.R.P.); (Y.-J.J.); (Y.L.); (S.-M.K.)
| | - Yu-Jin Jung
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (K.-H.K.); (B.R.P.); (Y.-J.J.); (Y.L.); (S.-M.K.)
| | - Youri Lee
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (K.-H.K.); (B.R.P.); (Y.-J.J.); (Y.L.); (S.-M.K.)
| | - Daria Mezhenskaya
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg 197376, Russia; (T.K.); (D.M.); (V.M.); (L.R.)
| | - Victoria Matyushenko
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg 197376, Russia; (T.K.); (D.M.); (V.M.); (L.R.)
| | - Sang-Moo Kang
- Center for Inflammation, Immunity and Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA; (K.-H.K.); (B.R.P.); (Y.-J.J.); (Y.L.); (S.-M.K.)
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg 197376, Russia; (T.K.); (D.M.); (V.M.); (L.R.)
| |
Collapse
|
5
|
Skarlupka AL, Handel A, Ross TM. Influenza hemagglutinin antigenic distance measures capture trends in HAI differences and infection outcomes, but are not suitable predictive tools. Vaccine 2020; 38:5822-5830. [PMID: 32682618 DOI: 10.1016/j.vaccine.2020.06.042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 05/28/2020] [Accepted: 06/16/2020] [Indexed: 01/24/2023]
Abstract
Vaccination is the most effective method to combat influenza. Vaccine effectiveness is influenced by the antigenic distance between the vaccine strain and the actual circulating virus. Amino acid sequence based methods of quantifying the antigenic distance were designed to predict influenza vaccine effectiveness in humans. The use of these antigenic distance measures has been proposed as an additive method for seasonal vaccine selection. In this report, several antigenic distance measures were evaluated as predictors of hemagglutination inhibition titer differences and clinical outcomes following influenza vaccination or infection in mice or ferrets. The antigenic distance measures described the increasing trend in the change of HAI titer, lung viral titer and percent weight loss in mice and ferrets. However, the variability of outcome variables produced wide prediction intervals for any given antigenic distance value. The amino acid substitution based antigenic distance measures were no better predictors of viral load and weight loss than HAI titer differences, the current predictive measure of immunological correlate of protection for clinical signs after challenge.
Collapse
Affiliation(s)
- Amanda L Skarlupka
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA
| | - Andreas Handel
- Department of Epidemiology and Biostatistics, University of Georgia, Athens, GA, USA
| | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, USA; Department of Infectious Diseases, University of Georgia, Athens, GA, USA.
| |
Collapse
|
6
|
Kirsteina A, Akopjana I, Bogans J, Lieknina I, Jansons J, Skrastina D, Kazaka T, Tars K, Isakova-Sivak I, Mezhenskaya D, Kotomina T, Matyushenko V, Rudenko L, Kazaks A. Construction and Immunogenicity of a Novel Multivalent Vaccine Prototype Based on Conserved Influenza Virus Antigens. Vaccines (Basel) 2020; 8:vaccines8020197. [PMID: 32344753 PMCID: PMC7349063 DOI: 10.3390/vaccines8020197] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/17/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023] Open
Abstract
Influenza, an acute, highly contagious respiratory disease, remains a significant threat to public health. More effective vaccination strategies aimed at inducing broad cross-protection not only against seasonal influenza variants, but also zoonotic and emerging pandemic influenza strains are urgently needed. A number of conserved protein targets to elicit such cross-protective immunity have been under investigation, with long alpha-helix (LAH) from hemagglutinin stalk and ectodomain of matrix protein 2 ion channel (M2e) being the most studied ones. Recently, we have reported the three-dimensional structure and some practical applications of LAH expressed in Escherichia coli system (referred to as tri-stalk protein). In the present study, we investigated the immunogenicity and efficacy of a panel of broadly protective influenza vaccine prototypes based on both influenza tri-stalk and triple M2e (3M2e) antigens integrated into phage AP205 virus-like particles (VLPs). While VLPs containing the 3M2e alone induced protection against standard homologous and heterologous virus challenge in mice, only the combination of both conserved influenza antigens into a single VLP fully protected mice from a high-dose homologous H1N1 influenza infection. We propose that a combination of genetic fusion and chemical coupling techniques to expose two different foreign influenza antigens on a single particle is a perspective approach for generation of a broadly-effective vaccine candidate that could protect against the constantly emerging influenza virus strains.
Collapse
Affiliation(s)
- Anna Kirsteina
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (A.K.); (I.A.); (J.B.); (I.L.); (J.J.); (D.S.); (T.K.); (K.T.)
| | - Inara Akopjana
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (A.K.); (I.A.); (J.B.); (I.L.); (J.J.); (D.S.); (T.K.); (K.T.)
| | - Janis Bogans
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (A.K.); (I.A.); (J.B.); (I.L.); (J.J.); (D.S.); (T.K.); (K.T.)
| | - Ilva Lieknina
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (A.K.); (I.A.); (J.B.); (I.L.); (J.J.); (D.S.); (T.K.); (K.T.)
| | - Juris Jansons
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (A.K.); (I.A.); (J.B.); (I.L.); (J.J.); (D.S.); (T.K.); (K.T.)
| | - Dace Skrastina
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (A.K.); (I.A.); (J.B.); (I.L.); (J.J.); (D.S.); (T.K.); (K.T.)
| | - Tatjana Kazaka
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (A.K.); (I.A.); (J.B.); (I.L.); (J.J.); (D.S.); (T.K.); (K.T.)
| | - Kaspars Tars
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (A.K.); (I.A.); (J.B.); (I.L.); (J.J.); (D.S.); (T.K.); (K.T.)
| | - Irina Isakova-Sivak
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg 197376, Russia; (I.I.-S.); (D.M.); (T.K.); (V.M.); (L.R.)
| | - Daria Mezhenskaya
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg 197376, Russia; (I.I.-S.); (D.M.); (T.K.); (V.M.); (L.R.)
| | - Tatiana Kotomina
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg 197376, Russia; (I.I.-S.); (D.M.); (T.K.); (V.M.); (L.R.)
| | - Victoria Matyushenko
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg 197376, Russia; (I.I.-S.); (D.M.); (T.K.); (V.M.); (L.R.)
| | - Larisa Rudenko
- Department of Virology, Institute of Experimental Medicine, Saint Petersburg 197376, Russia; (I.I.-S.); (D.M.); (T.K.); (V.M.); (L.R.)
| | - Andris Kazaks
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (A.K.); (I.A.); (J.B.); (I.L.); (J.J.); (D.S.); (T.K.); (K.T.)
- Correspondence:
| |
Collapse
|
7
|
Kim J, Hajam IA, Lee JH. Human antigen presenting cells stimulated with Salmonella delivered influenza antigens induce cytokine production and proliferation of human CD4 + T cells in vitro. J Immunol Methods 2019; 470:20-26. [PMID: 31028753 DOI: 10.1016/j.jim.2019.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/19/2019] [Accepted: 04/23/2019] [Indexed: 11/15/2022]
Abstract
This study aimed to investigate whether the human antigen presenting cells (APCs) can process and present Salmonella expressing H7N9 hemagglutinin (Sal-HA), neuraminidase (Sal-NA) or M2 ectodomain (Sal-M2e) to T cells and subsequently activate CD4+ T cell responses in vitro. In this study, APCs generated from human peripheral blood mononuclear cells (PBMCs) were first treated with mitomycin-C, followed by stimulation with Sal-HA, Sal-M2e, Sal-NA or Salmonella alone for 24 h. Subsequently, stimulated APCs were coincubated with untreated PBMCs (1:10) of the same individual for 24 or 72 h and then analysed for cytokine induction and T cell proliferations by qRT-PCR assay and flow cytometry, respectively. Our results demonstrated that APCs stimulated with Sal-HA, Sal-M2e or Sal-NA induced significantly (p < .05) higher CD3+CD4+ T cell proliferations compared to the APCs treated with Salmonella alone. Our data further revealved that APCs treated with Sal-HA induced significantly (p < .05) higher CD3+CD4+ T cell responses compared to the APCs treated with either Sal-M2e or Sal-NA, which both induced almost comparable levels. The T cell proliferation responses were further measured by lymphocyte proliferation assay and the results showed that Sal-HA and Sal-M2e stimulated APCs induced significantly (p < .05) higher proliferations in T cells compared to the APCs stimulated with either Sal-NA or Salmonella alone. With respect to cytokine inductions, APCs treated with either Sal-HA or Sal-M2e induced significantly (p < .05) higher mRNA transcription levels of proinflammatory (IL-1β, IL-6, IL-12 and IL-23), Th1 (IFN-γ), Th17 (IL-17 and IL-21) and Th2 (IL-10 and TGF-β) cytokines in T cells compared to Sal-NA or Salmonella alone treated APCs. In conclusion, we show that Salmonella system can efficiently deliver vaccine antigens to APCs and is, thus, capable to elicit heterologous antigen-specific adaptive immunity.
Collapse
Affiliation(s)
- Jehyoung Kim
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - Irshad Ahmed Hajam
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea
| | - John Hwa Lee
- College of Veterinary Medicine, Chonbuk National University, Iksan 54596, Republic of Korea.
| |
Collapse
|
8
|
Mancini N, Marrone L, Clementi N, Sautto GA, Clementi M, Burioni R. Adoptive T-cell therapy in the treatment of viral and opportunistic fungal infections. Future Microbiol 2016; 10:665-82. [PMID: 25865200 DOI: 10.2217/fmb.14.122] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Viral infections and opportunistic fungal pathogens represent a major menace for immunocompromised patients. Despite the availability of antifungal and antiviral drugs, mortality in these patients remains high, underlining the need of novel therapeutic options based on completely different strategies. This review describes the potential of several T-cell-based therapeutic approaches in the prophylaxis and treatment of infectious diseases with a particular focus on persistent viral infections and opportunistic fungal infections, as these mostly affect immunocompromised patients.
Collapse
Affiliation(s)
- Nicasio Mancini
- Laboratorio di Microbiologia e Virologia, Università 'Vita-Salute' San Raffaele, DIBIT2, via Olgettina 58, 20132, Milan, Italy
| | | | | | | | | | | |
Collapse
|
9
|
Bouguyon E, Goncalves E, Shevtsov A, Maisonnasse P, Remyga S, Goryushev O, Deville S, Bertho N, Ben Arous J. A New Adjuvant Combined with Inactivated Influenza Enhances Specific CD8 T Cell Response in Mice and Decreases Symptoms in Swine Upon Challenge. Viral Immunol 2015; 28:524-31. [PMID: 26447972 DOI: 10.1089/vim.2014.0149] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Vaccination is the most effective way to control swine influenza virus (SIV) in the field. Classical vaccines are based on inactivated antigens formulated with an oil emulsion or a polymeric adjuvant. Standard adjuvants enhance the humoral response and orient the immune response toward a Th2 response. An important issue is that current vaccines do not protect against new strains. One approach to improve cross-protection is to enhance Th1 and cytotoxic responses. The development of adjuvants orienting the immune response of inactivated vaccines toward Th1/Cytotoxic responses would be highly beneficial. This study shows that the water in oil in water emulsion adjuvant Montanide™ ISA 201 VG allows the induction of anti-influenza CD8 T cell in mice and induces homologous protection against an H1N1 challenge in swine. Such adjuvants that induce both humoral and cell-mediated immunity could improve the protection conferred by SIV vaccines in the field.
Collapse
Affiliation(s)
- Edwige Bouguyon
- 1 Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique , Domaine de Vilvert, Jouy-en-Josas, France
| | | | - Alexander Shevtsov
- 3 FGBI "Federal Centre for Animal Health" (FGBI "ARRIAH") , Yur'evets, Vladimir, Russia
| | - Pauline Maisonnasse
- 1 Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique , Domaine de Vilvert, Jouy-en-Josas, France
| | - Stepan Remyga
- 3 FGBI "Federal Centre for Animal Health" (FGBI "ARRIAH") , Yur'evets, Vladimir, Russia
| | - Oleg Goryushev
- 3 FGBI "Federal Centre for Animal Health" (FGBI "ARRIAH") , Yur'evets, Vladimir, Russia
| | | | - Nicolas Bertho
- 1 Virologie et Immunologie Moléculaires UR892, Institut National de la Recherche Agronomique , Domaine de Vilvert, Jouy-en-Josas, France
| | | |
Collapse
|
10
|
Stab V, Nitsche S, Niezold T, Storcksdieck genannt Bonsmann M, Wiechers A, Tippler B, Hannaman D, Ehrhardt C, Überla K, Grunwald T, Tenbusch M. Protective efficacy and immunogenicity of a combinatory DNA vaccine against Influenza A Virus and the Respiratory Syncytial Virus. PLoS One 2013; 8:e72217. [PMID: 23967287 PMCID: PMC3743785 DOI: 10.1371/journal.pone.0072217] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 07/05/2013] [Indexed: 12/18/2022] Open
Abstract
The Respiratory Syncytial Virus (RSV) and Influenza A Virus (IAV) are both two major causative agents of severe respiratory tract infections in humans leading to hospitalization and thousands of deaths each year. In this study, we evaluated the immunogenicity and efficacy of a combinatory DNA vaccine in comparison to the single component vaccines against both diseases in a mouse model. Intramuscular electroporation with plasmids expressing the hemagglutinin (HA) of IAV and the F protein of RSV induced strong humoral immune responses regardless if they were delivered in combination or alone. In consequence, high neutralizing antibody titers were detected, which conferred protection against a lethal challenge with IAV. Furthermore, the viral load in the lungs after a RSV infection could be dramatically reduced in vaccinated mice. Concurrently, substantial amounts of antigen-specific, polyfunctional CD8⁺ T-cells were measured after vaccination. Interestingly, the cellular response to the hemagglutinin was significantly reduced in the presence of the RSV-F encoding plasmid, but not vice versa. Although these results indicate a suppressive effect of the RSV-F protein, the protective efficacy of the combinatory vaccine was comparable to the efficacy of both single-component vaccines. In conclusion, the novel combinatory vaccine against RSV and IAV may have great potential to reduce the rate of severe respiratory tract infections in humans without increasing the number of necessary vaccinations.
Collapse
Affiliation(s)
- Viktoria Stab
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Sandra Nitsche
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Thomas Niezold
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | | | - Andrea Wiechers
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Bettina Tippler
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Drew Hannaman
- Ichor Medical Systems, San Diego, California, United States of America
| | - Christina Ehrhardt
- Institute of Molecular Virology, Centre of Molecular Biology of Inflammation, Westfaelische Wilhelms University, Muenster, Germany
| | - Klaus Überla
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Thomas Grunwald
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
| | - Matthias Tenbusch
- Department of Molecular and Medical Virology, Ruhr-University Bochum, Bochum, Germany
- * E-mail:
| |
Collapse
|
11
|
Budimir N, Huckriede A, Meijerhof T, Boon L, Gostick E, Price DA, Wilschut J, de Haan A. Induction of heterosubtypic cross-protection against influenza by a whole inactivated virus vaccine: the role of viral membrane fusion activity. PLoS One 2012; 7:e30898. [PMID: 22303469 PMCID: PMC3267744 DOI: 10.1371/journal.pone.0030898] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 12/23/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The inability of seasonal influenza vaccines to effectively protect against infection with antigenically drifted viruses or newly emerging pandemic viruses underlines the need for development of cross-reactive influenza vaccines that induce immunity against a variety of virus subtypes. Therefore, potential cross-protective vaccines, e.g., whole inactivated virus (WIV) vaccine, that can target conserved internal antigens such as the nucleoprotein (NP) and/or matrix protein (M1) need to be explored. METHODOLOGY/PRINCIPAL FINDINGS In the current study we show that a WIV vaccine, through induction of cross-protective cytotoxic T lymphocytes (CTLs), protects mice from heterosubtypic infection. This protection was abrogated after depletion of CD8+ cells in vaccinated mice, indicating that CTLs were the primary mediators of protection. Previously, we have shown that different procedures used for virus inactivation influence optimal activation of CTLs by WIV, most likely by affecting the membrane fusion properties of the virus. Specifically, inactivation with formalin (FA) severely compromises fusion activity of the virus, while inactivation with β-propiolactone (BPL) preserves fusion activity. Here, we demonstrate that vaccination of mice with BPL-inactivated H5N1 WIV vaccine induces solid protection from lethal heterosubtypic H1N1 challenge. By contrast, vaccination with FA-inactivated WIV, while preventing death after lethal challenge, failed to protect against development of disease and severe body weight loss. Vaccination with BPL-inactivated WIV, compared to FA-inactivated WIV, induced higher levels of specific CD8+ T cells in blood, spleen and lungs, and a higher production of granzyme B in the lungs upon H1N1 virus challenge. CONCLUSION/SIGNIFICANCE The results underline the potential use of WIV as a cross-protective influenza vaccine candidate. However, careful choice of the virus inactivation procedure is important to retain membrane fusion activity and full immunogenicity of the vaccine.
Collapse
MESH Headings
- Animals
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/immunology
- Body Weight/drug effects
- Cross Protection/drug effects
- Cross Protection/immunology
- Formaldehyde/pharmacology
- Hemagglutination Inhibition Tests
- Humans
- Immune Sera/drug effects
- Immune Sera/immunology
- Influenza A Virus, H1N1 Subtype/drug effects
- Influenza A Virus, H1N1 Subtype/immunology
- Influenza A Virus, H5N1 Subtype/drug effects
- Influenza A Virus, H5N1 Subtype/immunology
- Influenza Vaccines/immunology
- Influenza, Human/immunology
- Influenza, Human/prevention & control
- Influenza, Human/virology
- Lung/drug effects
- Lung/immunology
- Lung/pathology
- Lung/virology
- Mice
- Nucleoproteins/immunology
- Orthomyxoviridae Infections/immunology
- Orthomyxoviridae Infections/prevention & control
- Orthomyxoviridae Infections/virology
- Propiolactone/pharmacology
- Species Specificity
- Survival Analysis
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- Vaccination
- Vaccines, Inactivated/immunology
- Viral Load/drug effects
- Viral Load/immunology
- Virus Inactivation/drug effects
- Virus Internalization/drug effects
Collapse
Affiliation(s)
- Natalija Budimir
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center of Groningen, Groningen, The Netherlands
| | - Anke Huckriede
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center of Groningen, Groningen, The Netherlands
| | - Tjarko Meijerhof
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center of Groningen, Groningen, The Netherlands
| | | | - Emma Gostick
- Department of Infection, Immunity & Biochemistry, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - David A. Price
- Department of Infection, Immunity & Biochemistry, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Jan Wilschut
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center of Groningen, Groningen, The Netherlands
| | - Aalzen de Haan
- Department of Medical Microbiology, Molecular Virology Section, University Medical Center of Groningen, Groningen, The Netherlands
- * E-mail:
| |
Collapse
|
12
|
Iorio AM, Bistoni O, Galdiero M, Lepri E, Camilloni B, Russano AM, Neri M, Basileo M, Spinozzi F. Influenza viruses and cross-reactivity in healthy adults: humoral and cellular immunity induced by seasonal 2007/2008 influenza vaccination against vaccine antigens and 2009 A(H1N1) pandemic influenza virus. Vaccine 2012; 30:1617-23. [PMID: 22245606 DOI: 10.1016/j.vaccine.2011.12.107] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 12/16/2011] [Accepted: 12/22/2011] [Indexed: 01/10/2023]
Abstract
We analyzed humoral and cellular immune responses against vaccine antigens and the new A(H1N1) virus in healthy adults before and after immunization with the 2007/2008 commercially available trivalent subunit MF59-adjuvanted influenza vaccine during the Fall 2007, prior to the emergence of the new virus. Antibody titers were significantly boosted only against the three vaccine antigens. Seasonal vaccination boosted pre-existing cellular responses upon stimulation of peripheral blood mononuclear cells not only with the homologous three vaccine antigens, but also with the heterologous new 2009 A(H1N1) and with a highly conserved peptide present in the stalk region of hemagglutinin (HA). These results show that cross-reactive cell responses against the new virus were present before the circulation of the virus and were boosted by seasonal vaccination. The cross-reactivity of cellular responses might, at least in part, explain the low pathogenicity of the new pandemic virus. The finding of cellular immunity, that can be increased by seasonal vaccination, against the conserved HA peptide, underline the potential use, in human vaccines, of conserved peptides of the stalk region of HA characterized by broad immunogenicity in experimental systems.
Collapse
Affiliation(s)
- Anna M Iorio
- Department of Medical and Surgical Specialties and Public Health, University of Perugia, Via del Giochetto, 06123 Perugia, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Quinn K, Quirion MR, Lo CY, Misplon JA, Epstein SL, Chiorini JA. Intranasal administration of adeno-associated virus type 12 (AAV12) leads to transduction of the nasal epithelia and can initiate transgene-specific immune response. Mol Ther 2011; 19:1990-8. [PMID: 21829176 DOI: 10.1038/mt.2011.146] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A critical aspect in defining the utility of a vector for gene therapy applications is the cell tropism and biodistribution of the vector. Adeno-associated virus type 12 (AAV12) has several unique biological and immunological properties that could be exploited for gene therapy purposes, including a unique cell surface receptor, transduction of epithelial cells, and limited neutralization by pooled human antibodies. However, little is known about its cell tropism and biodistribution in vivo. In vivo biodistribution studies with AAV12 vectors encoding a cytomegalovirus promoted luciferase transgene indicated preferential transduction of the nasal epithelia which was not observed with AAV2-based vectors. Expression peaked 2 weeks postadministration, before decreasing to a persistent level. The level of neutralizing antibodies (Nab) induced was sevenfold lower for AAV12 than for AAV2, an advantage for use in repeat administration. Furthermore, vectors encoding influenza A nucleoprotein (NP), an antigen which has previously been shown to induce immune protection against challenge, resulted in generation of both anti-A/NP antibodies and lung anti-A/NP T cells. Our findings suggest further evaluation of AAV12 as a vector for gene therapy and as a potential nasal vaccine.
Collapse
Affiliation(s)
- Kathrina Quinn
- Molecular Physiology and Therapeutics Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | |
Collapse
|
14
|
Abstract
The mechanisms responsible for heterosubtypic immunity to influenza virus are not well understood but might hold the key for new vaccine strategies capable of providing lasting protection against both seasonal and pandemic strains. Memory CD4 T cells are capable of providing substantial protection against influenza both through direct effector mechanisms and indirectly through regulatory and helper functions. Here, we discuss the broad impact of memory CD4 T cells on heterosubtypic immunity against influenza and the prospects of translating findings from animal models into improved human influenza vaccines.
Collapse
Affiliation(s)
- K K McKinstry
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA.
| | | | | |
Collapse
|
15
|
Scheible K, Zhang G, Baer J, Azadniv M, Lambert K, Pryhuber G, Treanor JJ, Topham DJ. CD8+ T cell immunity to 2009 pandemic and seasonal H1N1 influenza viruses. Vaccine 2011; 29:2159-68. [PMID: 21211588 PMCID: PMC3061835 DOI: 10.1016/j.vaccine.2010.12.073] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 12/08/2010] [Accepted: 12/16/2010] [Indexed: 12/17/2022]
Abstract
A novel strain of H1N1 influenza A virus (pH1N1) emerged in 2009, causing a worldwide pandemic. Several studies suggest that this virus is antigenically more closely related to human influenza viruses that circulated prior to 1957 than viruses of more recent seasonal influenza varieties. The extent to which individuals who are naïve to the 2009 pH1N1 virus carry cross-reactive CD8+ T cells is not known, but a certain degree of reactivity would be expected since there is substantial conservation among the internal proteins of the virus. In the present study, we examined the production of multiple cytokines in response to virus from CD8+ T cells in healthy adult subjects, between 18 and 50 years of age (born post 1957), who had no evidence of exposure to the 2009 pH1N1 virus, and had blood collected prior to the emergence of the pandemic in April of 2009. Human peripheral blood mononuclear cells (PBMCs) were stimulated in vitro with a panel of live viruses, and assayed by intracellular cytokine staining and flow cytometry. Although results were variable, most subjects exhibited cytokine positive CD8+ T cells in response to pH1N1. Cytokine producing cells were predominantly single positive (IL2, IFNγ, or TNFα); triple-cytokine producing cells were relatively rare. This result suggests that although many adults carry cross-reactive T cells against the emergent pandemic virus, these cells are in a functionally limited state, possibly because these subjects have not had recent exposure to either seasonal or pandemic influenza strains.
Collapse
Affiliation(s)
- Kristin Scheible
- New York Influenza Center of Excellence, David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology; University of Rochester Medical Center, Rochester, NY 14642, USA
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Gang Zhang
- Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jane Baer
- New York Influenza Center of Excellence, David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology; University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Mitra Azadniv
- New York Influenza Center of Excellence, David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology; University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Kris Lambert
- New York Influenza Center of Excellence, David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology; University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Gloria Pryhuber
- Division of Neonatology, Department of Pediatrics, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - John J. Treanor
- New York Influenza Center of Excellence, David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology; University of Rochester Medical Center, Rochester, NY 14642, USA
- Division of Infectious Diseases, Department of Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - David J. Topham
- New York Influenza Center of Excellence, David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology; University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
16
|
Gupta SK, Srivastava M, Akhoon BA, Smita S, Schmitz U, Wolkenhauer O, Vera J, Gupta SK. Identification of immunogenic consensus T-cell epitopes in globally distributed influenza-A H1N1 neuraminidase. INFECTION GENETICS AND EVOLUTION 2010; 11:308-19. [PMID: 21094280 DOI: 10.1016/j.meegid.2010.10.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Revised: 10/15/2010] [Accepted: 10/18/2010] [Indexed: 02/01/2023]
Abstract
Antigenic drift is the ability of the swine influenza virus to undergo continuous and progressive changes in response to the host immune system. These changes dictate influenza vaccine updates annually to ensure inclusion of antigens of the most current strains. The identification of those peptides that stimulate T-cell responses, termed T-cell epitopes, is essential for the development of successful vaccines. In this study, the highly conserved and specific epitopes from neuraminidase of globally distributed H1N1 strains were predicted so that these potential vaccine candidates may escape with antigenic drift. A total of nine novel CD8(+) T-cell epitopes for MHC class-I and eight novel CD4(+) T-cell epitopes for MHC class-II alleles were proposed as novel epitope based vaccine candidates. Additionally, the epitope FSYKYGNGV was identified as a highly conserved, immunogenic and potential vaccine candidate, capable for generating both CD8(+) and CD4(+) responses.
Collapse
Affiliation(s)
- Shishir K Gupta
- Society for Biological Research & Rural Development, Lucknow, UP, India.
| | | | | | | | | | | | | | | |
Collapse
|