1
|
Padilla-Bórquez DL, Matuz-Flores MG, Hernández-Bello J, Rosas-Rodríguez JA, Turrubiates-Hernández FJ, García-Arellano S, González-Estevez G, Ceja-Galvez HR, Oregon-Romero E, López-Reyes A, Muñoz-Valle JF. Influence of previous COVID-19 exposure and vaccine type (CoronaVac, ChAdOx1 nCov-19 or BNT162b2) on antibody and cytokine (Th1 or Th2) responses. Hum Vaccin Immunother 2024; 20:2394265. [PMID: 39246041 PMCID: PMC11385164 DOI: 10.1080/21645515.2024.2394265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/10/2024] Open
Abstract
To achieve global herd immunity, widespread vaccination is the most effective strategy. Vaccines stimulate the immune system, generating cytokines and chemokines, isotype antibodies, and neutralizing antibodies; all these molecules collectively provide a more comprehensive characterization of the immune response post-vaccination. We conducted a longitudinal study in northwestern Mexico, involving 120 individuals before vaccination and after the first dose of the SARS-CoV-2 vaccine, and 46 individuals after their second dose. Our findings reveal that antibody levels stabilize over time; cytokine levels generally increase following the first dose but decrease after the second dose and higher than normal levels in IgG1 and IgG3 concentrations are present. Most of the innate cytokines determined in this study were higher after the first dose of the vaccine. Regardless of previous infection history, this finding suggests that the first dose of the vaccine is crucial and may stimulate immunity by enhancing the innate immune response. Conversely, increased levels of IL-4, indicative of a Th2 response, were found in individuals without prior exposure to the virus and in those vaccinated with CoronaVac. These results suggest that the immune response to COVID-19 vaccines is multi-faceted, with preexisting immunity potentiating a more robust innate response. Vaccine type plays a critical role, with genetic vaccines favoring a Th1 response and inactivated vaccines like CoronaVac skewing toward a Th2 profile.
Collapse
Affiliation(s)
- Diana Lourdes Padilla-Bórquez
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Mónica Guadalupe Matuz-Flores
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Jorge Hernández-Bello
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Jesús Alfredo Rosas-Rodríguez
- Departamento de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora Unidad Regional Sur, Navojoa, México
| | - Francisco Javier Turrubiates-Hernández
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Samuel García-Arellano
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Guillermo González-Estevez
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Hazael Ramiro Ceja-Galvez
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Edith Oregon-Romero
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| | - Alberto López-Reyes
- Laboratorio de Gerociencias, Instituto Nacional de Rehabilitación "Luis Guillermo Ibarra Ibarra", Secretaria de Salud, Ciudad de México, México
| | - Jose Francisco Muñoz-Valle
- Instituto de Investigación en Ciencias Biomédicas (IICB), Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara (UdG), Guadalajara, México
| |
Collapse
|
2
|
Grewe I, Friedrich M, Dieck ML, Spohn M, Ly ML, Krähling V, Mayer L, Mellinghoff SC, Rottstegge M, Kraemer R, Volz A, Becker S, Fathi A, Dahlke C, Weskamm LM, Addo MM. MVA-based SARS-CoV-2 vaccine candidates encoding different spike protein conformations induce distinct early transcriptional responses which may impact subsequent adaptive immunity. Front Immunol 2024; 15:1500615. [PMID: 39749328 PMCID: PMC11693667 DOI: 10.3389/fimmu.2024.1500615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/27/2024] [Indexed: 01/04/2025] Open
Abstract
Introduction Vaccine platforms such as viral vectors and mRNA can accelerate vaccine development in response to newly emerging pathogens, as demonstrated during the COVID-19 pandemic. However, the differential effects of platform and antigen insert on vaccine immunogenicity remain incompletely understood. Innate immune responses induced by viral vector vaccines are suggested to have an adjuvant effect for subsequent adaptive immunity. Integrating data on both innate and adaptive immunity, systems vaccinology approaches can improve the understanding of vaccine-induced immune mechanisms. Methods Two vaccine candidates against SARS-CoV-2, both based on the viral vector Modified Vaccinia virus Ankara (MVA) and encoding the native (MVA-SARS-2-S) or prefusion-stabilized spike protein (MVA-SARS-2-ST), were evaluated in phase 1 clinical trials (ClinicalTrials.gov: NCT04569383, NCT04895449). Longitudinal dynamics of innate and early adaptive immune responses induced by vaccination in SARS-CoV-2-naïve individuals were analyzed based on transcriptome and flow cytometry data, in comparison to the licensed ChAd and mRNA vaccines. Results Compared to MVA-SARS-2-S, MVA-SARS-2-ST (encoding the prefusion-stabilized spike protein) induced a stronger transcriptional activation early after vaccination, as well as higher virus neutralizing antibodies. Positive correlations were observed between innate and adaptive immune responses induced by a second MVA-SARS-2-ST vaccination. MVA-, ChAd- and mRNA-based vaccines induced distinct immune signatures, with the overall strongest transcriptional activation as well as monocyte and circulating T follicular helper (cTFH) cell responses induced by ChAd. Discussion Our findings suggest a potential impact of the spike protein conformation not only on adaptive but also on innate immune responses. As indicated by positive correlations between several immune parameters induced by MVA-SARS-2-ST, the distinct transcriptional activation early after vaccination may be linked to the induction of classical monocytes and activation of cTFH1 cells, which may in turn result in the superior adaptive immunogenicity of MVA-SARS-2-ST, compared to MVA-SARS-2-S. Overall, our data demonstrate that both the vaccine platform and antigen insert can affect innate immune responses and subsequent vaccine immunogenicity in humans.
Collapse
Affiliation(s)
- Ilka Grewe
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- First Department of Medicine, Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Monika Friedrich
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Marie-Louise Dieck
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Michael Spohn
- Research Institute Children’s Cancer Center Hamburg, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - My Linh Ly
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Verena Krähling
- Institute of Virology, Philipps University Marburg, Marburg, Germany
- German Center for Infection Research, Partner Site Gießen-Marburg-Langen, Marburg, Germany
| | - Leonie Mayer
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Sibylle C. Mellinghoff
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Translational Research, Cluster of Excellence for Aging Research (CECAD), Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Department I of Internal Medicine, Faculty of Medicine and University Hospital of Cologne, University of Cologne, Cologne, Germany
| | - Monika Rottstegge
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Rebekka Kraemer
- Institute of Clinical Molecular Biology, Christian-Albrechts-University and University Medical Center Schleswig-Holstein, Kiel, Germany
| | - Asisa Volz
- Institute of Virology, University of Veterinary Medicine Hannover, Hanover, Germany
- German Center for Infection Research, Partner Site Hannover-Braunschweig, Hannover, Germany
| | - Stephan Becker
- Institute of Virology, Philipps University Marburg, Marburg, Germany
- German Center for Infection Research, Partner Site Gießen-Marburg-Langen, Marburg, Germany
| | - Anahita Fathi
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- First Department of Medicine, Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Christine Dahlke
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Leonie M. Weskamm
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Marylyn M. Addo
- Institute for Infection Research and Vaccine Development (IIRVD), Center for Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department for Clinical Immunology of Infectious Diseases, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| |
Collapse
|
3
|
Abdel-Haq H. Feasibility of Using a Type I IFN-Based Non-Animal Approach to Predict Vaccine Efficacy and Safety Profiles. Vaccines (Basel) 2024; 12:583. [PMID: 38932312 PMCID: PMC11209158 DOI: 10.3390/vaccines12060583] [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: 05/07/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Animal-based tests are used for the control of vaccine quality. However, because highly purified and safe vaccines are now available, alternative approaches that can replace or reduce animal use for the assessment of vaccine outcomes must be established. In vitro tests for vaccine quality control exist and have already been implemented. However, these tests are specifically designed for some next-generation vaccines, and this makes them not readily available for testing other vaccines. Therefore, universal non-animal tests are still needed. Specific signatures of the innate immune response could represent a promising approach to predict the outcome of vaccines by non-animal methods. Type I interferons (IFNs) have multiple immunomodulatory activities, which are exerted through effectors called interferon stimulated genes (ISGs), and are one of the most important immune signatures that might provide potential candidate molecular biomarkers for this purpose. This paper will mainly examine if this idea might be feasible by analyzing all relevant published studies that have provided type I IFN-related biomarkers for evaluating the safety and efficacy profiles of vaccines using an advanced transcriptomic approach as an alternative to the animal methods. Results revealed that such an approach could potentially provide biomarkers predictive of vaccine outcomes after addressing some limitations.
Collapse
Affiliation(s)
- Hanin Abdel-Haq
- Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy
| |
Collapse
|
4
|
Gillard J, Suffiotti M, Brazda P, Venkatasubramanian PB, Versteegen P, de Jonge MI, Kelly D, Bibi S, Pinto MV, Simonetti E, Babiceanu M, Kettring A, Teodosio C, de Groot R, Berbers G, Stunnenberg HG, Schanen B, Fenwick C, Huynen MA, Diavatopoulos DA. Antiviral responses induced by Tdap-IPV vaccination are associated with persistent humoral immunity to Bordetella pertussis. Nat Commun 2024; 15:2133. [PMID: 38459022 PMCID: PMC10923912 DOI: 10.1038/s41467-024-46560-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/01/2024] [Indexed: 03/10/2024] Open
Abstract
Many countries continue to experience pertussis epidemics despite widespread vaccination. Waning protection after booster vaccination has highlighted the need for a better understanding of the immunological factors that promote durable protection. Here we apply systems vaccinology to investigate antibody responses in adolescents in the Netherlands (N = 14; NL) and the United Kingdom (N = 12; UK) receiving a tetanus-diphtheria-acellular pertussis-inactivated poliovirus (Tdap-IPV) vaccine. We report that early antiviral and interferon gene expression signatures in blood correlate to persistence of pertussis-specific antibody responses. Single-cell analyses of the innate response identified monocytes and myeloid dendritic cells (MoDC) as principal responders that upregulate antiviral gene expression and type-I interferon cytokine production. With public data, we show that Tdap vaccination stimulates significantly lower antiviral/type-I interferon responses than Tdap-IPV, suggesting that IPV may promote antiviral gene expression. Subsequent in vitro stimulation experiments demonstrate TLR-dependent, IPV-specific activation of the pro-inflammatory p38 MAP kinase pathway in MoDCs. Together, our data provide insights into the molecular host response to pertussis booster vaccination and demonstrate that IPV enhances innate immune activity associated with persistent, pertussis-specific antibody responses.
Collapse
Affiliation(s)
- Joshua Gillard
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Madeleine Suffiotti
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Peter Brazda
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | | | - Pauline Versteegen
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Marien I de Jonge
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dominic Kelly
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Sagida Bibi
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Marta Valente Pinto
- Department of Paediatrics, Oxford Vaccine Group, University of Oxford, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Egas Moniz Center for Interdisciplinary Research (CiiEM), Egas Moniz School of Health & Science, Caparica, Almada, Portugal
| | - Elles Simonetti
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | | - Cristina Teodosio
- Leiden University Medical Center, Immunohematology & Blood Transfusion, Leiden, The Netherlands
| | - Ronald de Groot
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Guy Berbers
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | | | | | - Craig Fenwick
- Service of Immunology and Allergy, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Martijn A Huynen
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Dimitri A Diavatopoulos
- Laboratory of Medical Immunology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.
- Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands.
| |
Collapse
|
5
|
Rosati M, Terpos E, Homan P, Bergamaschi C, Karaliota S, Ntanasis-Stathopoulos I, Devasundaram S, Bear J, Burns R, Bagratuni T, Trougakos IP, Dimopoulos MA, Pavlakis GN, Felber BK. Rapid transient and longer-lasting innate cytokine changes associated with adaptive immunity after repeated SARS-CoV-2 BNT162b2 mRNA vaccinations. Front Immunol 2023; 14:1292568. [PMID: 38090597 PMCID: PMC10711274 DOI: 10.3389/fimmu.2023.1292568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/14/2023] [Indexed: 12/18/2023] Open
Abstract
Introduction Cytokines and chemokines play an important role in shaping innate and adaptive immunity in response to infection and vaccination. Systems serology identified immunological parameters predictive of beneficial response to the BNT162b2 mRNA vaccine in COVID-19 infection-naïve volunteers, COVID-19 convalescent patients and transplant patients with hematological malignancies. Here, we examined the dynamics of the serum cytokine/chemokine responses after the 3rd BNT162b2 mRNA vaccination in a cohort of COVID-19 infection-naïve volunteers. Methods We measured serum cytokine and chemokine responses after the 3rd dose of the BNT162b2 mRNA (Pfizer/BioNtech) vaccine in COVID-19 infection-naïve individuals by a chemiluminescent assay and ELISA. Anti-Spike binding antibodies were measured by ELISA. Anti-Spike neutralizing antibodies were measured by a pseudotype assay. Results Comparison to responses found after the 1st and 2nd vaccinations showed persistence of the coordinated responses of several cytokine/chemokines including the previously identified rapid and transient IL-15, IFN-γ, CXCL10/IP-10, TNF-α, IL-6 signature. In contrast to the transient (24hrs) effect of the IL-15 signature, an inflammatory/anti-inflammatory cytokine signature (CCL2/MCP-1, CCL3/MIP-1α, CCL4/MIP-1β, CXCL8/IL-8, IL-1Ra) remained at higher levels up to one month after the 2nd and 3rd booster vaccinations, indicative of a state of longer-lasting innate immune change. We also identified a systemic transient increase of CXCL13 only after the 3rd vaccination, supporting stronger germinal center activity and the higher anti-Spike antibody responses. Changes of the IL-15 signature, and the inflammatory/anti-inflammatory cytokine profile correlated with neutralizing antibody levels also after the 3rd vaccination supporting their role as immune biomarkers for effective development of vaccine-induced humoral responses. Conclusion These data revealed that repeated SARS-Cov-2 BNT162b2 mRNA vaccination induces both rapid transient as well as longer-lasting systemic serum cytokine changes associated with innate and adaptive immune responses. Clinical trial registration Clinicaltrials.gov, identifier NCT04743388.
Collapse
Affiliation(s)
- Margherita Rosati
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Philip Homan
- Center for Cancer Research Collaborative Bioinformatics Resource, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Cristina Bergamaschi
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Sevasti Karaliota
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
- Basic Science Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Santhi Devasundaram
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Jenifer Bear
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Robert Burns
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Tina Bagratuni
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis P. Trougakos
- Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Meletios A. Dimopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - George N. Pavlakis
- Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| | - Barbara K. Felber
- Human Retrovirus Pathogenesis Section, Center for Cancer Research, National Cancer Institute at Frederick, Frederick, MD, United States
| |
Collapse
|
6
|
Zhang J, Li H, Albakr L, Zhang Y, Lu A, Chen W, Shao T, Zhu L, Yuan H, Yang G, Wheate NJ, Kang L, Wu C. Microneedle-enabled therapeutics delivery and biosensing in clinical trials. J Control Release 2023; 360:687-704. [PMID: 37442203 DOI: 10.1016/j.jconrel.2023.07.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 06/16/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023]
Abstract
Microneedles (MNs) are micron-sized protrusions attached to a range of devices that are used in therapeutic delivery and diagnosis. Because MNs can be self-applied, are painless, and can carry multiple therapeutic agents, they have received extensive attention, and have been widely investigated, for local and systemic therapy. Many researchers are currently working to extend the use of MNs to clinical applications. In this review, we provide an update and analysis on MN-based clinical trials since their inception in 2007. The MNs in clinical trials are classified into five types based on their appearance and properties, including: hollow MNs, MN patches, radiofrequency MNs, MN rollers, and other MNs. The various aspects of MN trials are summarized, such as MN types, clinical trial time, and trial regions. This review aims to present an overview of MN development and provide insights for future research in this field. To our knowledge, this is the first review focused on MN clinical trials which showcases the latest applications of this advanced technology in medicine.
Collapse
Affiliation(s)
- Junying Zhang
- Department of TCMs Pharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
| | - Hailiang Li
- Department of TCMs Pharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
| | - Lamyaa Albakr
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Pharmacy and Bank Building A15, NSW 2006, Australia; Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11454, Saudi Arabia
| | - Yiwen Zhang
- Department of TCMs Pharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
| | - Aiyu Lu
- Department of TCMs Pharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
| | - Wenlin Chen
- Department of TCMs Pharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
| | - Tianyu Shao
- Department of TCMs Pharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
| | - Luying Zhu
- Department of TCMs Pharmaceuticals, China Pharmaceutical University, Nanjing 210009, China
| | - Hua Yuan
- KPC Pharmaceuticals Inc., Kunming 650106, China
| | - Gongjun Yang
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China
| | - Nial J Wheate
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Pharmacy and Bank Building A15, NSW 2006, Australia
| | - Lifeng Kang
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Pharmacy and Bank Building A15, NSW 2006, Australia.
| | - Chungyong Wu
- Department of Pharmaceutical Analysis, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
7
|
Muir R, Metcalf T, Fourati S, Bartsch Y, Kyosiimire-Lugemwa J, Canderan G, Alter G, Muyanja E, Okech B, Namatovu T, Namara I, Namuniina A, Ssetaala A, Mpendo J, Nanvubya A, Kitandwe PK, Bagaya BS, Kiwanuka N, Nassuna J, Biribawa VM, Elliott AM, de Dood CJ, Senyonga W, Balungi P, Kaleebu P, Mayanja Y, Odongo M, Connors J, Fast P, Price MA, Corstjens PLAM, van Dam GJ, Kamali A, Sekaly RP, Haddad EK. Schistosoma mansoni infection alters the host pre-vaccination environment resulting in blunted Hepatitis B vaccination immune responses. PLoS Negl Trop Dis 2023; 17:e0011089. [PMID: 37406029 PMCID: PMC10351710 DOI: 10.1371/journal.pntd.0011089] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 07/17/2023] [Accepted: 06/20/2023] [Indexed: 07/07/2023] Open
Abstract
Schistosomiasis is a disease caused by parasitic flatworms of the Schistosoma spp., and is increasingly recognized to alter the immune system, and the potential to respond to vaccines. The impact of endemic infections on protective immunity is critical to inform vaccination strategies globally. We assessed the influence of Schistosoma mansoni worm burden on multiple host vaccine-related immune parameters in a Ugandan fishing cohort (n = 75) given three doses of a Hepatitis B (HepB) vaccine at baseline and multiple timepoints post-vaccination. We observed distinct differences in immune responses in instances of higher worm burden, compared to low worm burden or non-infected. Concentrations of pre-vaccination serum schistosome-specific circulating anodic antigen (CAA), linked to worm burden, showed a significant bimodal distribution associated with HepB titers, which was lower in individuals with higher CAA values at month 7 post-vaccination (M7). Comparative chemokine/cytokine responses revealed significant upregulation of CCL19, CXCL9 and CCL17 known to be involved in T cell activation and recruitment, in higher CAA individuals, and CCL17 correlated negatively with HepB titers at month 12 post-vaccination. We show that HepB-specific CD4+ T cell memory responses correlated positively with HepB titers at M7. We further established that those participants with high CAA had significantly lower frequencies of circulating T follicular helper (cTfh) subpopulations pre- and post-vaccination, but higher regulatory T cells (Tregs) post-vaccination, suggesting changes in the immune microenvironment in high CAA could favor Treg recruitment and activation. Additionally, we found that changes in the levels of innate-related cytokines/chemokines CXCL10, IL-1β, and CCL26, involved in driving T helper responses, were associated with increasing CAA concentration. This study provides further insight on pre-vaccination host responses to Schistosoma worm burden which will support our understanding of vaccine responses altered by pathogenic host immune mechanisms and memory function and explain abrogated vaccine responses in communities with endemic infections.
Collapse
Affiliation(s)
- Roshell Muir
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Talibah Metcalf
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Slim Fourati
- PATRU, School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Yannic Bartsch
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
| | | | - Glenda Canderan
- Department of Medicine, Allergy and Immunology, University of Virginia, Charlottesville, Virginia, United States of America
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, United States of America
| | - Enoch Muyanja
- PATRU, School of Medicine, Emory University, Atlanta, Georgia, United States of America
- UVRI-IAVI HIV Vaccine Program, Entebbe, Uganda
| | | | | | | | | | | | | | | | | | - Bernard S. Bagaya
- Department of Immunology and Molecular Biology, School of Biomedical Sciences, Makerere University, College of Health Sciences, Kampala, Uganda
| | - Noah Kiwanuka
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University, College of Health Sciences, Kampala, Uganda
| | - Jacent Nassuna
- Department of Epidemiology and Biostatistics, School of Public Health, Makerere University, College of Health Sciences, Kampala, Uganda
| | | | - Alison M. Elliott
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- Department of Clinical Research, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Claudia J. de Dood
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | | | - Yunia Mayanja
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - Matthew Odongo
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
| | - Jennifer Connors
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Pat Fast
- International AIDS Vaccine Initiative, New York, New York, United States of America
- Pediatric Infectious Diseases, Stanford University School of Medicine, Palo Alto, California, United States of America
| | - Matt A. Price
- International AIDS Vaccine Initiative, New York, New York, United States of America
- Department of Epidemiology and Biostatistics, University of California at San Francisco, San Francisco, California, United States of America
| | - Paul L. A. M. Corstjens
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Govert J. van Dam
- Department of Parasitology, Leiden University Medical Center, Leiden, the Netherlands
| | - Anatoli Kamali
- UVRI-IAVI HIV Vaccine Program, Entebbe, Uganda
- International AIDS Vaccine Initiative, New York, New York, United States of America
- IAVI, New York, New York, United States of America, and Nairobi, Kenya
| | - Rafick Pierre Sekaly
- PATRU, School of Medicine, Emory University, Atlanta, Georgia, United States of America
| | - Elias K. Haddad
- Division of Infectious Diseases and HIV Medicine, Department of Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
8
|
Cadar AN, Martin DE, Bartley JM. Targeting the hallmarks of aging to improve influenza vaccine responses in older adults. Immun Ageing 2023; 20:23. [PMID: 37198683 PMCID: PMC10189223 DOI: 10.1186/s12979-023-00348-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 05/09/2023] [Indexed: 05/19/2023]
Abstract
Age-related declines in immune response pose a challenge in combating diseases later in life. Influenza (flu) infection remains a significant burden on older populations and often results in catastrophic disability in those who survive infection. Despite having vaccines designed specifically for older adults, the burden of flu remains high and overall flu vaccine efficacy remains inadequate in this population. Recent geroscience research has highlighted the utility in targeting biological aging to improve multiple age-related declines. Indeed, the response to vaccination is highly coordinated, and diminished responses in older adults are likely not due to a singular deficit, but rather a multitude of age-related declines. In this review we highlight deficits in the aged vaccine responses and potential geroscience guided approaches to overcome these deficits. More specifically, we propose that alternative vaccine platforms and interventions that target the hallmarks of aging, including inflammation, cellular senescence, microbiome disturbances, and mitochondrial dysfunction, may improve vaccine responses and overall immunological resilience in older adults. Elucidating novel interventions and approaches that enhance immunological protection from vaccination is crucial to minimize the disproportionate effect of flu and other infectious diseases on older adults.
Collapse
Affiliation(s)
- Andreia N Cadar
- UConn Center On Aging and Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - Dominique E Martin
- UConn Center On Aging and Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA
| | - Jenna M Bartley
- UConn Center On Aging and Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, 06030, USA.
| |
Collapse
|
9
|
Muir R, Metcalf T, Fourati S, Bartsch Y, Lugemwa JK, Canderan G, Alter G, Muyanja E, Okech B, Namatovu T, Namara I, Namuniina A, Ssetaala A, Mpendo J, Nanvubya A, Kitandwe PK, Bagaya BS, Kiwanuka N, Nassuna J, Biribawa VM, Elliott AM, de Dood CJ, Senyonga W, Balungi P, Kaleebu P, Mayanja Y, Odongo M, Fast P, Price MA, Corstjens PLAM, van Dam GJ, Kamali A, Sekaly RP, Haddad EK. Schistosoma mansoni infection alters the host pre-vaccination environment resulting in blunted Hepatitis B vaccination immune responses. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.02.24.23284435. [PMID: 36865336 PMCID: PMC9980246 DOI: 10.1101/2023.02.24.23284435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2023]
Abstract
The impact of endemic infections on protective immunity is critical to inform vaccination strategies. In this study, we assessed the influence of Schistosoma mansoni infection on host responses in a Ugandan fishing cohort given a Hepatitis B (HepB) vaccine. Concentrations of schistosome-specific circulating anodic antigen (CAA) pre-vaccination showed a significant bimodal distribution associated with HepB titers, which were lower in individuals with high CAA. We established that participants with high CAA had significantly lower frequencies of circulating T follicular helper (cTfh) subpopulations pre- and post-vaccination and higher regulatory T cells (Tregs) post-vaccination. Polarization towards higher frequencies of Tregs: cTfh cells can be mediated by changes in the cytokine environment favoring Treg differentiation. In fact, we observed higher levels of CCL17 and soluble IL-2R pre-vaccination (important for Treg recruitment and development), in individuals with high CAA that negatively associated with HepB titers. Additionally, alterations in pre-vaccination monocyte function correlated with HepB titers, and changes in innate-related cytokines/chemokine production were associated with increasing CAA concentration. We report, that by influencing the immune landscape, schistosomiasis has the potential to modulate immune responses to HepB vaccination. These findings highlight multiple Schistosoma -related immune associations that could explain abrogated vaccine responses in communities with endemic infections. Author Summary Schistosomiasis drives host immune responses for optimal pathogen survival, potentially altering host responses to vaccine-related antigen. Chronic schistosomiasis and co-infection with hepatotropic viruses are common in countries where schistosomiasis is endemic. We explored the impact of Schistosoma mansoni ( S. mansoni ) infection on Hepatitis B (HepB) vaccination of individuals from a fishing community in Uganda. We demonstrate that high schistosome-specific antigen (circulating anodic antigen, CAA) concentration pre-vaccination, is associated with lower HepB antibody titers post-vaccination. We show higher pre-vaccination levels of cellular and soluble factors in instances of high CAA that are negatively associated with HepB antibody titers post-vaccination, which coincided with lower frequencies of circulating T follicular helper cell populations (cTfh), proliferating antibody secreting cells (ASCs), and higher frequencies of regulatory T cells (Tregs). We also show that monocyte function is important in HepB vaccine responses, and that high CAA is associated with alterations in the early innate cytokine/chemokine microenvironment. Our findings suggest that in individuals with high CAA and likely high worm burden, schistosomiasis creates and sustains an environment that is polarized against optimal host immune responses to the vaccine, which puts many endemic communities at risk for infection against HepB and other diseases that are preventable by vaccines.
Collapse
|
10
|
Karimabad MN, Hassanshahi G, Kounis NG, Mplani V, Roditis P, Gogos C, Lagadinou M, Assimakopoulos SF, Dousdampanis P, Koniari I. The Chemokines CXC, CC and C in the Pathogenesis of COVID-19 Disease and as Surrogates of Vaccine-Induced Innate and Adaptive Protective Responses. Vaccines (Basel) 2022; 10:vaccines10081299. [PMID: 36016187 PMCID: PMC9416781 DOI: 10.3390/vaccines10081299] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/08/2022] [Accepted: 08/08/2022] [Indexed: 02/07/2023] Open
Abstract
COVID-19 is one of the progressive viral pandemics that originated from East Asia. COVID-19 or SARS-CoV-2 has been shown to be associated with a chain of physio-pathological mechanisms that are basically immunological in nature. In addition, chemokines have been proposed as a subgroup of chemotactic cytokines with different activities ranging from leukocyte recruitment to injury sites, irritation, and inflammation to angiostasis and angiogenesis. Therefore, researchers have categorized the chemotactic elements into four classes, including CX3C, CXC, CC, and C, based on the location of the cysteine motifs in their structures. Considering the severe cases of COVID-19, the hyperproduction of particular chemokines occurring in lung tissue as well as pro-inflammatory cytokines significantly worsen the disease prognosis. According to the studies conducted in the field documenting the changing expression of CXC and CC chemokines in COVID-19 cases, the CC and CXC chemokines contribute to this pandemic, and their impact could reflect the development of reasonable strategies for COVID-19 management. The CC and the CXC families of chemokines are important in host immunity to viral infections and along with other biomarkers can serve as the surrogates of vaccine-induced innate and adaptive protective responses, facilitating the improvement of vaccine efficacy. Furthermore, the immunogenicity elicited by the chemokine response to adenovirus vector vaccines may constitute the basis of vaccine-induced immune thrombotic thrombocytopaenia.
Collapse
Affiliation(s)
- Mojgan Noroozi Karimabad
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan 7717933777, Iran
| | - Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan 7717933777, Iran
| | - Nicholas G. Kounis
- Department of Internal Medicine, Division of Cardiology, University of Patras Medical School, 26500 Patras, Greece
- Correspondence:
| | - Virginia Mplani
- Intensive Care Unit, Patras University Hospital, 26500 Patras, Greece
| | - Pavlos Roditis
- Department of Cardiology, Mamatsio Kozanis General Hospital, 50100 Kozani, Greece
| | - Christos Gogos
- COVID-19 Unit, Papageorgiou General Hospital, 56403 Thessaloniki, Greece
| | - Maria Lagadinou
- Department of Internal Medicine, Division of Infectious Diseases, University of Patras Medical School, 26500 Patras, Greece
| | - Stelios F. Assimakopoulos
- Department of Internal Medicine, Division of Infectious Diseases, University of Patras Medical School, 26500 Patras, Greece
| | - Periklis Dousdampanis
- Department of Nephrology, Saint Andrews State General Hospital, 26221 Patras, Greece
| | - Ioanna Koniari
- Department of Cardiology, University Hospital of South Manchester, NHS Foundation Trust, Manchester M23 9LT, UK
| |
Collapse
|
11
|
Lee HK, Knabl L, Moliva JI, Knabl L, Werner AP, Boyoglu-Barnum S, Kapferer S, Pateter B, Walter M, Sullivan NJ, Furth PA, Hennighausen L. mRNA vaccination in octogenarians 15 and 20 months after recovery from COVID-19 elicits robust immune and antibody responses that include Omicron. Cell Rep 2022; 39:110680. [PMID: 35395191 PMCID: PMC8947943 DOI: 10.1016/j.celrep.2022.110680] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/16/2022] [Accepted: 03/23/2022] [Indexed: 01/20/2023] Open
Abstract
Knowledge about the impact of prior severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection of the elderly on mRNA vaccination response is needed to appropriately address the demand for additional vaccinations in this vulnerable population. Here, we show that octogenarians, a high-risk population, mount a sustained SARS-CoV-2 spike-specific immunoglobulin G (IgG) antibody response for 15 months following infection. This response boosts antibody levels 35-fold upon receiving a single dose of BNT162b2 mRNA vaccine 15 months after recovery from coronavirus disease 2019 (COVID-19). In contrast, antibody responses in naive individuals boost only 6-fold after a second vaccine. Spike-specific angiotensin-converting enzyme 2 (ACE2) antibody binding responses in the previously infected octogenarians following two vaccine doses exceed those found in a naive cohort after two doses. RNA sequencing (RNA-seq) demonstrates activation of interferon-induced genetic programs, which persist only in the previously infected. A preferential increase of specific immunoglobulin G heavy chain variable (IGHV) clonal transcripts that are the basis of neutralizing antibodies is observed only in the previously infected nuns.
Collapse
Affiliation(s)
- Hye Kyung Lee
- National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | - Juan I Moliva
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | - Anne P Werner
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Seyhan Boyoglu-Barnum
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | - Mary Walter
- Clinical Core, National Institute of Diabetes, Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Nancy J Sullivan
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Priscilla A Furth
- Departments of Oncology & Medicine, Georgetown University, Washington, DC, USA.
| | - Lothar Hennighausen
- National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
12
|
Tiwari N, Osorio‐Blanco ER, Sonzogni A, Esporrín‐Ubieto D, Wang H, Calderón M. Nanocarriers for Skin Applications: Where Do We Stand? Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202107960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Neha Tiwari
- POLYMAT Applied Chemistry Department Faculty of Chemistry University of the Basque Country UPV/EHU Paseo Manuel de Lardizabal 3 20018 Donostia-San Sebastián Spain
| | - Ernesto Rafael Osorio‐Blanco
- POLYMAT Applied Chemistry Department Faculty of Chemistry University of the Basque Country UPV/EHU Paseo Manuel de Lardizabal 3 20018 Donostia-San Sebastián Spain
| | - Ana Sonzogni
- Group of Polymers and Polymerization Reactors INTEC (Universidad Nacional del Litoral-CONICET) Güemes 3450 Santa Fe 3000 Argentina
| | - David Esporrín‐Ubieto
- POLYMAT Applied Chemistry Department Faculty of Chemistry University of the Basque Country UPV/EHU Paseo Manuel de Lardizabal 3 20018 Donostia-San Sebastián Spain
| | - Huiyi Wang
- POLYMAT Applied Chemistry Department Faculty of Chemistry University of the Basque Country UPV/EHU Paseo Manuel de Lardizabal 3 20018 Donostia-San Sebastián Spain
| | - Marcelo Calderón
- POLYMAT Applied Chemistry Department Faculty of Chemistry University of the Basque Country UPV/EHU Paseo Manuel de Lardizabal 3 20018 Donostia-San Sebastián Spain
- IKERBASQUE, Basque Foundation for Science 48009 Bilbao Spain
| |
Collapse
|
13
|
Tiwari N, Osorio‐Blanco ER, Sonzogni A, Esporrín‐Ubieto D, Wang H, Calderón M. Nanocarriers for Skin Applications: Where Do We Stand? Angew Chem Int Ed Engl 2022; 61:e202107960. [PMID: 34487599 PMCID: PMC9292798 DOI: 10.1002/anie.202107960] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Indexed: 12/15/2022]
Abstract
Skin penetration of active molecules for treatment of diverse diseases is a major field of research owing to the advantages associated with the skin like easy accessibility, reduced systemic-derived side effects, and increased therapeutic efficacy. Despite these advantages, dermal drug delivery is generally challenging due to the low skin permeability of therapeutics. Although various methods have been developed to improve skin penetration and permeation of therapeutics, they are usually aggressive and could lead to irreversible damage to the stratum corneum. Nanosized carrier systems represent an alternative approach for current technologies, with minimal damage to the natural barrier function of skin. In this Review, the use of nanoparticles to deliver drug molecules, genetic material, and vaccines into the skin is discussed. In addition, nanotoxicology studies and the recent clinical development of nanoparticles are highlighted to shed light on their potential to undergo market translation.
Collapse
Affiliation(s)
- Neha Tiwari
- POLYMATApplied Chemistry DepartmentFaculty of ChemistryUniversity of the Basque CountryUPV/EHUPaseo Manuel de Lardizabal 320018Donostia-San SebastiánSpain
| | - Ernesto Rafael Osorio‐Blanco
- POLYMATApplied Chemistry DepartmentFaculty of ChemistryUniversity of the Basque CountryUPV/EHUPaseo Manuel de Lardizabal 320018Donostia-San SebastiánSpain
| | - Ana Sonzogni
- Group of Polymers and Polymerization ReactorsINTEC (Universidad Nacional del Litoral-CONICET)Güemes 3450Santa Fe3000Argentina
| | - David Esporrín‐Ubieto
- POLYMATApplied Chemistry DepartmentFaculty of ChemistryUniversity of the Basque CountryUPV/EHUPaseo Manuel de Lardizabal 320018Donostia-San SebastiánSpain
| | - Huiyi Wang
- POLYMATApplied Chemistry DepartmentFaculty of ChemistryUniversity of the Basque CountryUPV/EHUPaseo Manuel de Lardizabal 320018Donostia-San SebastiánSpain
| | - Marcelo Calderón
- POLYMATApplied Chemistry DepartmentFaculty of ChemistryUniversity of the Basque CountryUPV/EHUPaseo Manuel de Lardizabal 320018Donostia-San SebastiánSpain
- IKERBASQUE, Basque Foundation for Science48009BilbaoSpain
| |
Collapse
|
14
|
The Involvement of CXC Motif Chemokine Ligand 10 (CXCL10) and Its Related Chemokines in the Pathogenesis of Coronary Artery Disease and in the COVID-19 Vaccination: A Narrative Review. Vaccines (Basel) 2021; 9:vaccines9111224. [PMID: 34835155 PMCID: PMC8623875 DOI: 10.3390/vaccines9111224] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022] Open
Abstract
Coronary artery disease (CAD) and coronary heart disease (CHD) constitute two of the leading causes of death in Europe, USA and the rest of the world. According to the latest reports of the Iranian National Health Ministry, CAD is the main cause of death in Iranian patients with an age over 35 years despite a significant reduction in mortality due to early interventional treatments in the context of an acute coronary syndrome (ACS). Inflammation plays a fundamental role in coronary atherogenesis, atherosclerotic plaque formation, acute coronary thrombosis and CAD establishment. Chemokines are well-recognized mediators of inflammation involved in several bio-functions such as leucocyte migration in response to inflammatory signals and oxidative vascular injury. Different chemokines serve as chemo-attractants for a wide variety of cell types including immune cells. CXC motif chemokine ligand 10 (CXCL10), also known as interferon gamma-induced protein 10 (IP-10/CXLC10), is a chemokine with inflammatory features whereas CXC chemokine receptor 3 (CXCR3) serves as a shared receptor for CXCL9, 10 and 11. These chemokines mediate immune responses through the activation and recruitment of leukocytes, eosinophils, monocytes and natural killer (NK) cells. CXCL10, interleukin (IL-15) and interferon (IFN-g) are increased after a COVID-19 vaccination with a BNT162b2 mRNA (Pfizer/BioNTech) vaccine and are enriched by tumor necrosis factor alpha (TNF-α) and IL-6 after the second vaccination. The aim of the present study is the presentation of the elucidation of the crucial role of CXCL10 in the patho-physiology and pathogenesis of CAD and in identifying markers associated with the vaccination resulting in antibody development.
Collapse
|
15
|
Lee HK, Knabl L, Knabl L, Kapferer S, Pateter B, Walter M, Furth PA, Hennighausen L. Robust immune response to the BNT162b mRNA vaccine in an elderly population vaccinated 15 months after recovery from COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.09.08.21263284. [PMID: 34545374 PMCID: PMC8452113 DOI: 10.1101/2021.09.08.21263284] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Knowledge about the impact of prior SARS-CoV-2 infection of the elderly on mRNA vaccination response is needed to appropriately address the need for booster vaccination in this vulnerable population. To address this, we investigated antibody and genomic immune responses in 16 elderly (avg. 81 yrs.) individuals that had received a single booster dose of BNT162b vaccine 15 months after recovering from COVID-19. Spike-specific IgG antibody levels increased in each of the study participants from an average of 710 U/ml prior to the vaccination to more than 40,000 U/ml within ten weeks after the vaccination. In contrast, anti-spike-specific IgG antibody levels averaged 2,190 U/ml in 14 healthy SARS-CoV-2-naïve individuals (avg. 58 yrs.) ten weeks after the second dose of BNT162b. RNA-seq conducted on PBMCs demonstrated the activation of interferon-activated genetic programs in both cohorts within one day. Unlike their transient induction in the younger naïve population, persistent activity and the initiation of additional cell cycle regulated programs were obtained in the older COVID-19 recovered population. Here we show that the elderly, a high-risk population, can mount a strong antibody and a persistent molecular immune response upon receiving a single dose of mRNA vaccine 15 months after recovery from COVID-19.
Collapse
Affiliation(s)
- Hye Kyung Lee
- National Institute of Diabetes, Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | | | | | | | | | - Mary Walter
- Clinical Core, National Institute of Diabetes, Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Priscilla A. Furth
- Departments of Oncology & Medicine, Georgetown University, Washington, DC, USA
| | - Lothar Hennighausen
- National Institute of Diabetes, Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| |
Collapse
|
16
|
Systemic IL-15, IFN-γ, and IP-10/CXCL10 signature associated with effective immune response to SARS-CoV-2 in BNT162b2 mRNA vaccine recipients. Cell Rep 2021; 36:109504. [PMID: 34352226 PMCID: PMC8299183 DOI: 10.1016/j.celrep.2021.109504] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/16/2021] [Accepted: 07/19/2021] [Indexed: 12/29/2022] Open
Abstract
Early responses to vaccination are important for shaping both humoral and cellular protective immunity. Dissecting innate vaccine signatures may predict immunogenicity to help optimize the efficacy of mRNA and other vaccine strategies. Here, we characterize the cytokine and chemokine responses to the 1st and 2nd dose of the BNT162b2 mRNA (Pfizer/BioNtech) vaccine in antigen-naive and in previously coronavirus disease 2019 (COVID-19)-infected individuals (NCT04743388). Transient increases in interleukin-15 (IL-15) and interferon gamma (IFN-γ) levels early after boost correlate with Spike antibody levels, supporting their use as biomarkers of effective humoral immunity development in response to vaccination. We identify a systemic signature including increases in IL-15, IFN-γ, and IP-10/CXCL10 after the 1st vaccination, which were enriched by tumor necrosis factor alpha (TNF-α) and IL-6 after the 2nd vaccination. In previously COVID-19-infected individuals, a single vaccination results in both strong cytokine induction and antibody titers similar to the ones observed upon booster vaccination in antigen-naive individuals, a result with potential implication for future public health recommendations.
Collapse
|
17
|
Lin X, Lin F, Liang T, Ducatez MF, Zanin M, Wong SS. Antibody Responsiveness to Influenza: What Drives It? Viruses 2021; 13:v13071400. [PMID: 34372607 PMCID: PMC8310379 DOI: 10.3390/v13071400] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/02/2021] [Accepted: 07/03/2021] [Indexed: 02/06/2023] Open
Abstract
The induction of a specific antibody response has long been accepted as a serological hallmark of recent infection or antigen exposure. Much of our understanding of the influenza antibody response has been derived from studying antibodies that target the hemagglutinin (HA) protein. However, growing evidence points to limitations associated with this approach. In this review, we aim to highlight the issue of antibody non-responsiveness after influenza virus infection and vaccination. We will then provide an overview of the major factors known to influence antibody responsiveness to influenza after infection and vaccination. We discuss the biological factors such as age, sex, influence of prior immunity, genetics, and some chronic infections that may affect the induction of influenza antibody responses. We also discuss the technical factors, such as assay choices, strain variations, and viral properties that may influence the sensitivity of the assays used to measure influenza antibodies. Understanding these factors will hopefully provide a more comprehensive picture of what influenza immunogenicity and protection means, which will be important in our effort to improve influenza vaccines.
Collapse
Affiliation(s)
- Xia Lin
- State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 195 Dongfengxi Rd, Guangzhou 510182, China; (X.L.); (F.L.); (T.L.); (M.Z.)
| | - Fangmei Lin
- State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 195 Dongfengxi Rd, Guangzhou 510182, China; (X.L.); (F.L.); (T.L.); (M.Z.)
| | - Tingting Liang
- State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 195 Dongfengxi Rd, Guangzhou 510182, China; (X.L.); (F.L.); (T.L.); (M.Z.)
| | | | - Mark Zanin
- State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 195 Dongfengxi Rd, Guangzhou 510182, China; (X.L.); (F.L.); (T.L.); (M.Z.)
- School of Public Health, The University of Hong Kong, Hong Kong, China
| | - Sook-San Wong
- State Key Laboratory of Respiratory Diseases, Guangzhou Medical University, 195 Dongfengxi Rd, Guangzhou 510182, China; (X.L.); (F.L.); (T.L.); (M.Z.)
- School of Public Health, The University of Hong Kong, Hong Kong, China
- Correspondence: ; Tel.: +86-178-2584-6078
| |
Collapse
|
18
|
Van Tilbeurgh M, Lemdani K, Beignon AS, Chapon C, Tchitchek N, Cheraitia L, Marcos Lopez E, Pascal Q, Le Grand R, Maisonnasse P, Manet C. Predictive Markers of Immunogenicity and Efficacy for Human Vaccines. Vaccines (Basel) 2021; 9:579. [PMID: 34205932 PMCID: PMC8226531 DOI: 10.3390/vaccines9060579] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 02/07/2023] Open
Abstract
Vaccines represent one of the major advances of modern medicine. Despite the many successes of vaccination, continuous efforts to design new vaccines are needed to fight "old" pandemics, such as tuberculosis and malaria, as well as emerging pathogens, such as Zika virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Vaccination aims at reaching sterilizing immunity, however assessing vaccine efficacy is still challenging and underscores the need for a better understanding of immune protective responses. Identifying reliable predictive markers of immunogenicity can help to select and develop promising vaccine candidates during early preclinical studies and can lead to improved, personalized, vaccination strategies. A systems biology approach is increasingly being adopted to address these major challenges using multiple high-dimensional technologies combined with in silico models. Although the goal is to develop predictive models of vaccine efficacy in humans, applying this approach to animal models empowers basic and translational vaccine research. In this review, we provide an overview of vaccine immune signatures in preclinical models, as well as in target human populations. We also discuss high-throughput technologies used to probe vaccine-induced responses, along with data analysis and computational methodologies applied to the predictive modeling of vaccine efficacy.
Collapse
Affiliation(s)
- Matthieu Van Tilbeurgh
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Katia Lemdani
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Anne-Sophie Beignon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Catherine Chapon
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Nicolas Tchitchek
- Unité de Recherche i3, Inserm UMR-S 959, Bâtiment CERVI, Hôpital de la Pitié-Salpêtrière, 75013 Paris, France;
| | - Lina Cheraitia
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Ernesto Marcos Lopez
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Quentin Pascal
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Roger Le Grand
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Pauline Maisonnasse
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| | - Caroline Manet
- Immunology of Viral Infections and Autoimmune Diseases (IMVA), IDMIT Department, Institut de Biologie François-Jacob (IBJF), University Paris-Sud—INSERM U1184, CEA, 92265 Fontenay-Aux-Roses, France; (M.V.T.); (K.L.); (A.-S.B.); (C.C.); (L.C.); (E.M.L.); (Q.P.); (R.L.G.); (P.M.)
| |
Collapse
|
19
|
Rosenbaum P, Tchitchek N, Joly C, Rodriguez Pozo A, Stimmer L, Langlois S, Hocini H, Gosse L, Pejoski D, Cosma A, Beignon AS, Dereuddre-Bosquet N, Levy Y, Le Grand R, Martinon F. Vaccine Inoculation Route Modulates Early Immunity and Consequently Antigen-Specific Immune Response. Front Immunol 2021; 12:645210. [PMID: 33959127 PMCID: PMC8093451 DOI: 10.3389/fimmu.2021.645210] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/06/2021] [Indexed: 01/12/2023] Open
Abstract
Vaccination is one of the most efficient public healthcare measures to fight infectious diseases. Nevertheless, the immune mechanisms induced in vivo by vaccination are still unclear. The route of administration, an important vaccination parameter, can substantially modify the quality of the response. How the route of administration affects the generation and profile of immune responses is of major interest. Here, we aimed to extensively characterize the profiles of the innate and adaptive response to vaccination induced after intradermal, subcutaneous, or intramuscular administration with a modified vaccinia virus Ankara model vaccine in non-human primates. The adaptive response following subcutaneous immunization was clearly different from that following intradermal or intramuscular immunization. The subcutaneous route induced a higher level of neutralizing antibodies than the intradermal and intramuscular vaccination routes. In contrast, polyfunctional CD8+ T-cell responses were preferentially induced after intradermal or intramuscular injection. We observed the same dichotomy when analyzing the early molecular and cellular immune events, highlighting the recruitment of cell populations, such as CD8+ T lymphocytes and myeloid-derived suppressive cells, and the activation of key immunomodulatory gene pathways. These results demonstrate that the quality of the vaccine response induced by an attenuated vaccine is shaped by early and subtle modifications of the innate immune response. In this immunization context, the route of administration must be tailored to the desired type of protective immune response. This will be achieved through systems vaccinology and mathematical modeling, which will be critical for predicting the efficacy of the vaccination route for personalized medicine.
Collapse
Affiliation(s)
- Pierre Rosenbaum
- UMR1184 IMVA-HB, IDMIT Department, Université Paris-Saclay – INSERM U1184 – CEA, Fontenay-aux-Roses, France
- Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| | - Nicolas Tchitchek
- UMR1184 IMVA-HB, IDMIT Department, Université Paris-Saclay – INSERM U1184 – CEA, Fontenay-aux-Roses, France
- Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| | - Candie Joly
- UMR1184 IMVA-HB, IDMIT Department, Université Paris-Saclay – INSERM U1184 – CEA, Fontenay-aux-Roses, France
- Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| | - André Rodriguez Pozo
- UMR1184 IMVA-HB, IDMIT Department, Université Paris-Saclay – INSERM U1184 – CEA, Fontenay-aux-Roses, France
- Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| | - Lev Stimmer
- INSERM, U1169, Kremlin-Bicêtre, France
- CEA – INSERM, MIRCen, UMS27, Fontenay-aux-Roses, France
| | - Sébastien Langlois
- UMR1184 IMVA-HB, IDMIT Department, Université Paris-Saclay – INSERM U1184 – CEA, Fontenay-aux-Roses, France
- Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| | - Hakim Hocini
- Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
- INSERM, U955, Team 16, Clinical and Infectious Diseases Department, Hospital Henri Mondor, University of Paris East, Créteil, France
| | - Leslie Gosse
- UMR1184 IMVA-HB, IDMIT Department, Université Paris-Saclay – INSERM U1184 – CEA, Fontenay-aux-Roses, France
- Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| | - David Pejoski
- UMR1184 IMVA-HB, IDMIT Department, Université Paris-Saclay – INSERM U1184 – CEA, Fontenay-aux-Roses, France
- Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| | - Antonio Cosma
- UMR1184 IMVA-HB, IDMIT Department, Université Paris-Saclay – INSERM U1184 – CEA, Fontenay-aux-Roses, France
- Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| | - Anne-Sophie Beignon
- UMR1184 IMVA-HB, IDMIT Department, Université Paris-Saclay – INSERM U1184 – CEA, Fontenay-aux-Roses, France
- Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| | - Nathalie Dereuddre-Bosquet
- UMR1184 IMVA-HB, IDMIT Department, Université Paris-Saclay – INSERM U1184 – CEA, Fontenay-aux-Roses, France
- Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| | - Yves Levy
- Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
- INSERM, U955, Team 16, Clinical and Infectious Diseases Department, Hospital Henri Mondor, University of Paris East, Créteil, France
| | - Roger Le Grand
- UMR1184 IMVA-HB, IDMIT Department, Université Paris-Saclay – INSERM U1184 – CEA, Fontenay-aux-Roses, France
- Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| | - Frédéric Martinon
- UMR1184 IMVA-HB, IDMIT Department, Université Paris-Saclay – INSERM U1184 – CEA, Fontenay-aux-Roses, France
- Vaccine Research Institute, Henri Mondor Hospital, Créteil, France
| |
Collapse
|
20
|
Adam L, Rosenbaum P, Bonduelle O, Combadière B. Strategies for Immunomonitoring after Vaccination and during Infection. Vaccines (Basel) 2021; 9:365. [PMID: 33918841 PMCID: PMC8070333 DOI: 10.3390/vaccines9040365] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/05/2021] [Accepted: 04/07/2021] [Indexed: 01/08/2023] Open
Abstract
Immunomonitoring is the study of an individual's immune responses over the course of vaccination or infection. In the infectious context, exploring the innate and adaptive immune responses will help to investigate their contribution to viral control or toxicity. After vaccination, immunomonitoring of the correlate(s) and surrogate(s) of protection is a major asset for measuring vaccine immune efficacy. Conventional immunomonitoring methods include antibody-based technologies that are easy to use. However, promising sensitive high-throughput technologies allowed the emergence of holistic approaches. This raises the question of data integration methods and tools. These approaches allow us to increase our knowledge on immune mechanisms as well as the identification of key effectors of the immune response. However, the depiction of relevant findings requires a well-rounded consideration beforehand about the hypotheses, conception, organization and objectives of the immunomonitoring. Therefore, well-standardized and comprehensive studies fuel insight to design more efficient, rationale-based vaccines and therapeutics to fight against infectious diseases. Hence, we will illustrate this review with examples of the immunomonitoring approaches used during vaccination and the COVID-19 pandemic.
Collapse
Affiliation(s)
| | | | | | - Behazine Combadière
- Inserm, Centre d’Immunologie et des Maladies Infectieuses, Sorbonne Université, 75013 Paris, France; (L.A.); (P.R.); (O.B.)
| |
Collapse
|
21
|
Blanc F, Maroilley T, Revilla M, Lemonnier G, Leplat JJ, Billon Y, Ravon L, Bouchez O, Bidanel JP, Bed'Hom B, Pinard-van der Laan MH, Estellé J, Rogel-Gaillard C. Influence of genetics and the pre-vaccination blood transcriptome on the variability of antibody levels after vaccination against Mycoplasma hyopneumoniae in pigs. Genet Sel Evol 2021; 53:24. [PMID: 33731010 PMCID: PMC7972226 DOI: 10.1186/s12711-021-00614-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 02/16/2021] [Indexed: 12/13/2022] Open
Abstract
Background The impact of individual genetic and genomic variations on immune responses is an emerging lever investigated in vaccination strategies. In our study, we used genetic and pre-vaccination blood transcriptomic data to study vaccine effectiveness in pigs. Results A cohort of 182 Large White pigs was vaccinated against Mycoplasma hyopneumoniae (M. hyo) at weaning (28 days of age), with a booster 21 days later. Vaccine response was assessed by measuring seric M. hyo antibodies (Ab) at 0 (vaccination day), 21 (booster day), 28, 35, and 118 days post-vaccination (dpv). Inter-individual variability of M. hyo Ab levels was observed at all time points and the corresponding heritabilities ranged from 0.46 to 0.57. Ab persistence was higher in females than in males. Genome-wide association studies with a 658 K SNP panel revealed two genomic regions associated with variations of M. hyo Ab levels at 21 dpv at positions where immunity-related genes have been mapped, DAB2IP on chromosome 1, and ASAP1, CYRIB and GSDMC on chromosome 4. We studied covariations of Ab responses with the pre-vaccination blood transcriptome obtained by RNA-Seq for a subset of 82 pigs. Weighted gene correlation network and differential expression analyses between pigs that differed in Ab responses highlighted biological functions that were enriched in heme biosynthesis and platelet activation for low response at 21 dpv, innate antiviral immunity and dendritic cells for high response at 28 and 35 dpv, and cell adhesion and extracellular matrix for high response at 118 dpv. Sparse partial least squares discriminant analysis identified 101 genes that efficiently predicted divergent responders at all time points. We found weak negative correlations of M. hyo Ab levels with body weight traits, which revealed a trade-off that needs to be further explored. Conclusions We confirmed the influence of the host genetics on vaccine effectiveness to M. hyo and provided evidence that the pre-vaccination blood transcriptome co-varies with the Ab response. Our results highlight that both genetic markers and blood biomarkers could be used as potential predictors of vaccine response levels and more studies are required to assess whether they can be exploited in breeding programs.
Collapse
Affiliation(s)
- Fany Blanc
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France.
| | - Tatiana Maroilley
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Manuel Revilla
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Gaëtan Lemonnier
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Jean-Jacques Leplat
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | | | | | | | - Jean-Pierre Bidanel
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Bertrand Bed'Hom
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | | | - Jordi Estellé
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | | |
Collapse
|
22
|
Th1/Th2 Cells and Associated Cytokines in Acute Hepatitis E and Related Acute Liver Failure. J Immunol Res 2020; 2020:6027361. [PMID: 33294465 PMCID: PMC7691005 DOI: 10.1155/2020/6027361] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/08/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023] Open
Abstract
Background and Aims The involvement of cellular immunity in the development of hepatitis E virus (HEV) infection is rare. We aimed to study the roles of viral load and Th cell responses in acute hepatitis E (AHE) and HEV-related acute liver failure (HEV-ALF). Methods We evaluated viral load and Th1/Th2 cytokine levels in 34 patients with HEV infection, including 17 each with AHE or HEV-ALF. Seventeen healthy controls (HCs) were also included who were negative for anti-HEV IgM and IgG. Results There was no significant difference in viral load and HEV RNA in the AHE and HEV-ALF groups (both P > 0.05). The Th lymphocyte levels (CD3+, CD4+) in the AHE and HEV-ALF groups were significantly higher than those in the HC group (both P < 0.05), but there was no significant difference between the AHE and HEV-ALF groups (P > 0.05). Both IFN-γ and IL-10 showed gradual upward trend from the HC group to the AHE (both P < 0.01), but IFN-γ showed a sharp downward trend from the AHE group to the HEV-ALF group (P < 0.01) and IL-4 showed gradual upward trend from the AHE group to the HEV-ALF group (P < 0.01).There was no significant difference in Th1 and Th2 cytokines between the HEV RNA(+) group and HEV RNA(-) group (all P > 0.05). Th2 bias was observed from the AHE (ratio = 58.65) to HEV-ALF (ratio = 1.20) groups. The level of IFN-γ was associated with the outcome of HEV-ALF patients. Conclusions HEV viral load was not associated with aggravation of AHE, and the HEV-ALF patients showed significant Th2 bias, which may be involved in the aggravation of AHE.
Collapse
|
23
|
Sanchez J, Gonçalves E, Llano A, Gonzáles P, Fernández-Maldonado M, Vogt A, Soria A, Perez S, Cedeño S, Fernández MA, Nourikyan J, de Bernard S, Ganoza C, Pedruzzi E, Bonduelle O, Mothe B, Gòmez CE, Esteban M, Garcia F, Lama JR, Brander C, Combadiere B. Immune Profiles Identification by Vaccinomics After MVA Immunization in Randomized Clinical Study. Front Immunol 2020; 11:586124. [PMID: 33244316 PMCID: PMC7683801 DOI: 10.3389/fimmu.2020.586124] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/12/2020] [Indexed: 12/04/2022] Open
Abstract
Background Our previous work has demonstrated the benefits of transcutaneous immunization in targeting Langerhans cells and preferentially inducing CD8 T-cell responses. Methods In this randomized phase Ib clinical trial including 20 HIV uninfected volunteers, we compared the safety and immunogenicity of the MVA recombinant vaccine expressing HIV-B antigen (MVA-B) by transcutaneous and intramuscular routes. We hypothesized that the quality of innate and adaptive immunity differs according to the route of immunization and explored the quality of the vector vaccine-induced immune responses. We also investigated the early blood transcriptome and serum cytokine levels to identify innate events correlated with the strength and quality of adaptive immunity. Results We demonstrate that MVA-B vaccine is safe by both routes, but that the quality and intensity of both innate and adaptive immunity differ significantly. Transcutaneous vaccination promoted CD8 responses in the absence of antibodies and slightly affected gene expression, involving mainly genes associated with metabolic pathways. Intramuscular vaccination, on the other hand, drove robust changes in the expression of genes involved in IL-6 and interferon signalling pathways, mainly those associated with humoral responses, and also some levels of CD8 response. Conclusion Thus, vaccine delivery route perturbs early innate responses that shape the quality of adaptive immunity. Clinical Trial Registration http://ClinicalTrials.gov, identifier PER-073-13.
Collapse
Affiliation(s)
- Jorge Sanchez
- Centro de Investigaciones Tecnológicas, Biomedicas y Medioambientales, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Elena Gonçalves
- Sorbonne Université, Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMIParis), Paris, France
| | - Anuska Llano
- IrsiCaixa AIDS Research Institute-HIVACAT, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | | | | | - Annika Vogt
- Clinical Research Center for Hair and Skin Science, Department of Dermatology, Venerology and Allergy, Charité-Universitatsmedizin Berlin, corporate member of Freie Universitaet Berlin, Humboldt-Universitaet zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | - Susana Perez
- Centro de Investigaciones Tecnológicas, Biomedicas y Medioambientales, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Samandhy Cedeño
- IrsiCaixa AIDS Research Institute-HIVACAT, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | - Marco Antonio Fernández
- Flow Cytometry Facility, Germans Trias i Pujol Research Institute (IGTP), Hospital Universitari Germans Trias i Pujol, Barcelona, Spain
| | | | | | | | - Eric Pedruzzi
- Sorbonne Université, Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMIParis), Paris, France
| | - Olivia Bonduelle
- Sorbonne Université, Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMIParis), Paris, France
| | - Beatriz Mothe
- IrsiCaixa AIDS Research Institute-HIVACAT, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain.,Fundació Lluita contra la Sida, Infectious Diseases Department, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Carmen E Gòmez
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Mariano Esteban
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Felipe Garcia
- Infectious Diseases Department, Hospital Clínic, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Javier R Lama
- Asociacion Civil Impacta Salud y Educacion, Lima, Peru
| | - Christian Brander
- IrsiCaixa AIDS Research Institute-HIVACAT, Hospital Universitari Germans Trias i Pujol, Barcelona, Spain.,Faculty of Medicine, Universitat de Vic-Central de Catalunya (UVic-UCC), Vic, Spain.,Institució Catalana de Recerca I Estudis Avançats (ICREA), Barcelona, Spain
| | - Behazine Combadiere
- Sorbonne Université, Inserm, Centre d'Immunologie et des Maladies Infectieuses (CIMIParis), Paris, France
| |
Collapse
|
24
|
Abstract
The impact of host adaptive immune response on COVID-19 has now become a critical issue in absence of specific therapy and immunotherapies. In SARS CoV-2 infection, the immune response is thought to contribute both to the pathogenesis of the disease and to protection during its resolution. While mild cases develop an immune response that contributes to host protection, immunity of severely infected patients is a balance between harmful and protective immune responses. The severity of the disease has raised many questions about the kinetic, amplitude and the quality of adaptive immunity to the virus and its generation during the early phases of infection in severe, mild and asymptomatic patients. The role of antibody and CD4+ and CD8+ T cell responses have been studied and the development of an adaptive immunity seems to correlate with convalescence. The bioinformatics study of the T and B epitopes of coronaviruses has raised the question of the existence of cross-immunity between SARS-CoV-2 and other coronaviruses such as MERS-CoV and SARS-CoV. In this review, we discuss the adaptive immune responses and their potential roles in protection during COVID-19.
Collapse
Affiliation(s)
- Béhazine Combadière
- Sorbonne Université, Inserm U1135, Centre d'Immunologie et des Maladies Infectieuses (Cimi-Paris), 91 boulevard de l'Hôpital, 75013 Paris, France
| |
Collapse
|
25
|
Boccard M, Albert-Vega C, Mouton W, Durieu I, Brengel-Pesce K, Venet F, Trouillet-Assant S, Ader F. [Functional immunoassays in the setting of infectious risk and immunosuppressive therapy of non-HIV immunocompromised patients]. Rev Med Interne 2020; 41:545-551. [PMID: 32624260 DOI: 10.1016/j.revmed.2020.04.008] [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: 11/11/2019] [Revised: 03/24/2020] [Accepted: 04/09/2020] [Indexed: 11/25/2022]
Abstract
The holistic approach of the human immune system is based on the study of its components collectively driving a functional response to an immunogenic stimulus. To appreciate a specific immune dysfunction, a condition is mimicked ex vivo and the immune response induced is assessed. The application field of such assays are broad and expanding, from the diagnosis of primary and secondary immunodeficiencies, immunotherapy for cancer to the management of patients at-risk for infections and vaccination. These assays are immune monitoring tools that may contribute to a personalised and precision medicine. The purpose of this review is to describe immune functional assays available in the setting of non-HIV acquired immune deficiency. First, we will address the use of theses assays in the diagnosis of opportunistic infections such as viral reactivation. Secondly, we will report the usefulness of these assays to assess vaccine efficacy and to manage immunosuppressive therapies.
Collapse
Affiliation(s)
- M Boccard
- Centre International de Recherche en Infectiologie (CIRI), Inserm 1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Univ Lyon, F-69007, Lyon, France; Département de médecine interne et vasculaire, centre hospitalier Lyon Sud, Hospices civils de Lyon, 69310 Pierre-Bénite, France; Unité mixte Hospices civils de Lyon-bioMérieux, centre hospitalier Lyon Sud, Hospices civils de Lyon, Pierre-Bénite, 69495 Lyon, France.
| | - C Albert-Vega
- Unité mixte Hospices civils de Lyon-bioMérieux, centre hospitalier Lyon Sud, Hospices civils de Lyon, Pierre-Bénite, 69495 Lyon, France
| | - W Mouton
- Unité mixte Hospices civils de Lyon-bioMérieux, centre hospitalier Lyon Sud, Hospices civils de Lyon, Pierre-Bénite, 69495 Lyon, France; Laboratoire virologie et pathologies humaines (VirPath), faculté de médecine Lyon Est, université Claude-Bernard Lyon 1, 69008 Lyon, France
| | - I Durieu
- Département de médecine interne et vasculaire, centre hospitalier Lyon Sud, Hospices civils de Lyon, 69310 Pierre-Bénite, France
| | - K Brengel-Pesce
- Unité mixte Hospices civils de Lyon-bioMérieux, centre hospitalier Lyon Sud, Hospices civils de Lyon, Pierre-Bénite, 69495 Lyon, France
| | - F Venet
- Unité mixte Hospices civils de Lyon-bioMérieux, centre hospitalier Lyon Sud, Hospices civils de Lyon, Pierre-Bénite, 69495 Lyon, France; Laboratoire d'immunologie, hôpital Édouard-Herriot, Hospices civils de Lyon, 69003 Lyon, France; EA7426 Pathophysiology of injury-induced immunosuppression, université Claude-Bernard Lyon 1, 69008 Lyon, France
| | - S Trouillet-Assant
- Unité mixte Hospices civils de Lyon-bioMérieux, centre hospitalier Lyon Sud, Hospices civils de Lyon, Pierre-Bénite, 69495 Lyon, France; Laboratoire virologie et pathologies humaines (VirPath), faculté de médecine Lyon Est, université Claude-Bernard Lyon 1, 69008 Lyon, France
| | - F Ader
- Centre International de Recherche en Infectiologie (CIRI), Inserm 1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, École Normale Supérieure de Lyon, Univ Lyon, F-69007, Lyon, France; Département des maladies infectieuses et tropicales, hôpital de la Croix-Rousse, Hospices civils de Lyon, 69004 Lyon, France
| |
Collapse
|
26
|
Adam L, Tchitchek N, Todorova B, Rosenbaum P, Joly C, Poux C, Chapon C, Spetz AL, Ustav M, Le Grand R, Martinon F. Innate Molecular and Cellular Signature in the Skin Preceding Long-Lasting T Cell Responses after Electroporated DNA Vaccination. THE JOURNAL OF IMMUNOLOGY 2020; 204:3375-3388. [PMID: 32385135 DOI: 10.4049/jimmunol.1900517] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 04/09/2020] [Indexed: 12/21/2022]
Abstract
DNA vaccines delivered with electroporation (EP) have shown promising results in preclinical models and are evaluated in clinical trials. In this study, we aim to characterize early mechanisms occurring in the skin after intradermal injection and EP of the auxoGTUmultiSIV DNA vaccine in nonhuman primates. First, we show that EP acts as an adjuvant by enhancing local inflammation, notably via granulocytes, monocytes/macrophages, and CD1aint-expressing cell recruitment. EP also induced Langerhans cell maturation, illustrated by CD86, CD83, and HLA-DR upregulation and their migration out of the epidermis. Second, we demonstrate the crucial role of the DNA vaccine in soluble factors release, such as MCP-1 or IL-15. Transcriptomic analysis showed that EP played a major role in gene expression changes postvaccination. However, the DNA vaccine is required to strongly upregulate several genes involved in inflammatory responses (e.g., Saa4), cell migration (e.g., Ccl3, Ccl5, or Cxcl10), APC activation (e.g., Cd86), and IFN-inducible genes (e.g., Ifit3, Ifit5, Irf7, Isg15, orMx1), illustrating an antiviral response signature. Also, AIM-2, a cytosolic DNA sensor, appeared to be strongly upregulated only in the presence of the DNA vaccine and trends to positively correlate with several IFN-inducible genes, suggesting the potential role of AIM-2 in vaccine sensing and the subsequent innate response activation leading to strong adaptive T cell responses. Overall, these results demonstrate that a combined stimulation of the immune response, in which EP and the auxoGTUmultiSIV vaccine triggered different components of the innate immunity, led to strong and persistent cellular recall responses.
Collapse
Affiliation(s)
- Lucille Adam
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Nicolas Tchitchek
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Biliana Todorova
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Pierre Rosenbaum
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Candie Joly
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Candice Poux
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Catherine Chapon
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Anna-Lena Spetz
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden; and
| | - Mart Ustav
- Institute of Technology, University of Tartu, 50411 Tartu, Estonia
| | - Roger Le Grand
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France
| | - Frédéric Martinon
- Immunology of Viral Infections and Autoimmune Diseases, Infectious Disease Models and Innovative Therapies Department, Commissariat à l'Energie Atomique et aux Energies Alternatives, Université Paris-Sud 11, INSERM U1184, 92265 Fontenay-aux-Roses, France;
| |
Collapse
|
27
|
Du Y, Thompson EG, Muller J, Valvo J, Braun J, Shankar S, van den Berg RA, Jongert E, Dover D, Sadoff J, Hendriks J, Gardner MJ, Ballou WR, Regules JA, van der Most R, Aderem A, Ockenhouse CF, Hill AV, Wille-Reece U, Zak DE. The Ratiometric Transcript Signature MX2/GPR183 Is Consistently Associated With RTS,S-Mediated Protection Against Controlled Human Malaria Infection. Front Immunol 2020; 11:669. [PMID: 32411130 PMCID: PMC7199517 DOI: 10.3389/fimmu.2020.00669] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 03/24/2020] [Indexed: 12/14/2022] Open
Abstract
The RTS,S/AS01 vaccine provides partial protection against Plasmodium falciparum infection but determinants of protection and/or disease are unclear. Previously, anti-circumsporozoite protein (CSP) antibody titers and blood RNA signatures were associated with RTS,S/AS01 efficacy against controlled human malaria infection (CHMI). By analyzing host blood transcriptomes from five RTS,S vaccination CHMI studies, we demonstrate that the transcript ratio MX2/GPR183, measured 1 day after third immunization, discriminates protected from non-protected individuals. This ratiometric signature provides information that is complementary to anti-CSP titer levels for identifying RTS,S/AS01 immunized people who developed protective immunity and suggests a role for interferon and oxysterol signaling in the RTS,S mode of action.
Collapse
Affiliation(s)
- Ying Du
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | | | | | - Joseph Valvo
- Center for Infectious Disease Research, Seattle, WA, United States
| | - Jackie Braun
- Center for Infectious Disease Research, Seattle, WA, United States
| | - Smitha Shankar
- Center for Infectious Disease Research, Seattle, WA, United States
| | | | | | - Drew Dover
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | - Jerald Sadoff
- Janssen Vaccines and Prevention BV, Leiden, Netherlands
| | | | - Malcolm J. Gardner
- Infectious Diseases, J. Craig Venter Institute, La Jolla, CA, United States
| | | | - Jason A. Regules
- Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | | | - Alan Aderem
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, WA, United States
| | | | | | | | - Daniel E. Zak
- Center for Infectious Disease Research, Seattle, WA, United States
| |
Collapse
|
28
|
Gonçalves E, Combadière B. Prédire la réponse à la vaccination contre la grippe. Med Sci (Paris) 2020; 36:31-37. [DOI: 10.1051/medsci/2019266] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
La vaccination est l’un des progrès majeurs de la médecine moderne. Mais afin d’améliorer l’efficacité des vaccins existants et d’en élaborer de nouveaux, nous devons mieux connaître les mécanismes d’action à l’origine de l’immunité protectrice et les stratégies vaccinales permettant d’induire une défense durable. La voie cutanée est une stratégie de vaccination importante, en raison de la richesse qu’elle présente en cellules de l’immunité innée qui ont un rôle clé dans la qualité, l’intensité et la persistance des réponses adaptatives qu’elles induisent. L’intégration des données biologiques obtenues au cours d’un essai clinique de vaccination antigrippale nous donne un aperçu de l’impact de la voie d’immunisation et de la signature innée sur la qualité des réponses immunitaires.
Collapse
|
29
|
Gonnet J, Poncelet L, Meriaux C, Gonçalves E, Weiss L, Tchitchek N, Pedruzzi E, Soria A, Boccara D, Vogt A, Bonduelle O, Hamm G, Ait-Belkacem R, Stauber J, Fournier I, Wisztorski M, Combadiere B. Mechanisms of innate events during skin reaction following intradermal injection of seasonal influenza vaccine. J Proteomics 2020; 216:103670. [PMID: 31991189 DOI: 10.1016/j.jprot.2020.103670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 12/03/2019] [Accepted: 01/25/2020] [Indexed: 12/15/2022]
Abstract
The skin plays a crucial role in host defences against microbial attack and the innate cells must provide the immune system with sufficient information to organize these defences. This unique feature makes the skin a promising site for vaccine administration. Although cellular innate immune events during vaccination have been widely studied, initial events remain poorly understood. Our aim is to determine molecular biomarkers of skin innate reaction after intradermal (i.d.) immunization. Using an ex vivo human explant model from healthy donors, we investigated by NanoLC-MS/MS analysis and MALDI-MSI imaging, to detect innate molecular events (lipids, metabolites, proteins) few hours after i.d. administration of seasonal trivalent influenza vaccine (TIV). This multimodel approach allowed to identify early molecules differentially expressed in dermal and epidermal layers at 4 and 18 h after TIV immunization compared with control PBS. In the dermis, the most relevant network of proteins upregulated were related to cell-to-cell signalling and cell trafficking. The molecular signatures detected were associated with chemokines such as CXCL8, a chemoattractant of neutrophils. In the epidermis, the most relevant networks were associated with activation of antigen-presenting cells and related to CXCL10. Our study proposes a novel step-forward approach to identify biomarkers of skin innate reaction. SIGNIFICANCE: To our knowledge, there is no study analyzing innate molecular reaction to vaccines at the site of skin immunization. What is known on skin reaction is based on macroscopic (erythema, redness…), microscopic (epidermal and dermal tissues) and cellular events (inflammatory cell infiltrate). Therefore, we propose a multimodal approach to analyze molecular events at the site of vaccine injection on skin tissue. We identified early molecular networks involved biological functions such cell migration, cell-to-cell interaction and antigen presentation, validated by chemokine expression, in the epidermis and dermis, then could be used as early indicator of success in immunization.
Collapse
Affiliation(s)
- Jessica Gonnet
- Sorbonne Université, Centre d'Immunologie et des Maladies Infectieuses - Paris (Cimi-Paris), INSERM U1135, Paris, France
| | - Lauranne Poncelet
- Univ. Lille, INSERM, CHU Lille, U1008 - Controlled Drug Delivery Systems and Biomaterials, F-59000 Lille, France; ImaBiotech, 152 rue du Docteur Yersin, 59120 Loos, France
| | - Celine Meriaux
- Univ. Lille, Inserm, U1192 - Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France
| | - Elena Gonçalves
- Sorbonne Université, Centre d'Immunologie et des Maladies Infectieuses - Paris (Cimi-Paris), INSERM U1135, Paris, France
| | - Lina Weiss
- Sorbonne Université, Centre d'Immunologie et des Maladies Infectieuses - Paris (Cimi-Paris), INSERM U1135, Paris, France; Clinical Research Center for Hair and Skin Science, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin (2), 10117 Berlin, Germany
| | - Nicolas Tchitchek
- CEA - Université Paris Sud 11 - INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, Institut de Biologie François Jacob, 92265 Fontenay-aux-Roses, France
| | - Eric Pedruzzi
- Sorbonne Université, Centre d'Immunologie et des Maladies Infectieuses - Paris (Cimi-Paris), INSERM U1135, Paris, France
| | - Angele Soria
- Sorbonne Université, Centre d'Immunologie et des Maladies Infectieuses - Paris (Cimi-Paris), INSERM U1135, Paris, France; Service de Dermatologie et d'Allergologie, Hôpital Tenon, 4 rue de la Chine, Hôpitaux Universitaire Est Parisien (HUEP), Assistance Publique Hôpitaux de Paris (APHP), 75020 Paris, France
| | - David Boccara
- Sorbonne Université, Centre d'Immunologie et des Maladies Infectieuses - Paris (Cimi-Paris), INSERM U1135, Paris, France; Service de chirurgie plastique reconstructrice, esthétique, centre des brûlés, Hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris (APHP), 1 avenue Claude Vellefaux, 75010 Paris, France
| | - Annika Vogt
- Sorbonne Université, Centre d'Immunologie et des Maladies Infectieuses - Paris (Cimi-Paris), INSERM U1135, Paris, France; Clinical Research Center for Hair and Skin Science, Department of Dermatology and Allergy, Charité - Universitätsmedizin Berlin (2), 10117 Berlin, Germany
| | - Olivia Bonduelle
- Sorbonne Université, Centre d'Immunologie et des Maladies Infectieuses - Paris (Cimi-Paris), INSERM U1135, Paris, France
| | - Gregory Hamm
- ImaBiotech, 152 rue du Docteur Yersin, 59120 Loos, France
| | | | | | - Isabelle Fournier
- Univ. Lille, Inserm, U1192 - Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France
| | - Maxence Wisztorski
- Univ. Lille, Inserm, U1192 - Protéomique, Réponse Inflammatoire et Spectrométrie de Masse-PRISM, F-59000 Lille, France
| | - Behazine Combadiere
- Sorbonne Université, Centre d'Immunologie et des Maladies Infectieuses - Paris (Cimi-Paris), INSERM U1135, Paris, France.
| |
Collapse
|
30
|
Maruyama SR, Carvalho B, González-Porta M, Rung J, Brazma A, Gustavo Gardinassi L, Ferreira BR, Banin TM, Veríssimo CJ, Katiki LM, de Miranda-Santos IKF. Blood transcriptome profile induced by an efficacious vaccine formulated with salivary antigens from cattle ticks. NPJ Vaccines 2019; 4:53. [PMID: 31871773 PMCID: PMC6920353 DOI: 10.1038/s41541-019-0145-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 11/13/2019] [Indexed: 12/11/2022] Open
Abstract
Ticks cause massive damage to livestock and vaccines are one sustainable alternative for the acaricide poisons currently heavily used to control infestations. An experimental vaccine adjuvanted with alum and composed by four recombinant salivary antigens mined with reverse vaccinology from a transcriptome of salivary glands from Rhipicephalus microplus ticks was previously shown to present an overall efficacy of 73.2% and cause a significant decrease of tick loads in artificially tick-infested, immunized heifers; this decrease was accompanied by increased levels of antigen-specific IgG1 and IgG2 antibodies, which were boosted during a challenge infestation. In order to gain insights into the systemic effects induced by the vaccine and by the tick challenge we now report the gene expression profile of these hosts' whole-blood leukocytes with RNA-seq followed by functional analyses. These analyses show that vaccination induced unique responses to infestations; genes upregulated in the comparisons were enriched for processes associated with chemotaxis, cell adhesion, T-cell responses and wound repair. Blood transcriptional modules were enriched for activation of dendritic cells, cell cycle, phosphatidylinositol signaling, and platelets. Together, the results indicate that by neutralizing the tick's salivary mediators of parasitism with vaccine-induced antibodies, the bovine host is able to mount normal homeostatic responses that hinder tick attachment and haematophagy and that the tick otherwise suppresses with its saliva.
Collapse
Affiliation(s)
- Sandra R. Maruyama
- Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, SP Brazil
- Present Address: Department of Genetics and Evolution, Center for Biological Sciences and Health, Federal University of São Carlos, São Carlos, SP Brazil
| | | | - Mar González-Porta
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Hinxton, UK
- Present Address: Illumina Centre, Cambridge, UK
| | - Johan Rung
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Hinxton, UK
- Present Address: Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Alvis Brazma
- European Molecular Biology Laboratory, European Bioinformatics Institute, EMBL-EBI, Hinxton, UK
| | - Luiz Gustavo Gardinassi
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP Brazil
| | - Beatriz R. Ferreira
- Ribeirão Preto School of Nursing, University of São Paulo, Ribeirão Preto, SP Brazil
| | - Tamy M. Banin
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP Brazil
| | | | | | | |
Collapse
|
31
|
Ruiz-Riol M, Brander C. Can we just kick-and-kill HIV: possible challenges posed by the epigenetically controlled interplay between HIV and host immunity. Immunotherapy 2019; 11:931-935. [PMID: 31218904 PMCID: PMC6609895 DOI: 10.2217/imt-2019-0092] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 06/04/2019] [Indexed: 02/06/2023] Open
Affiliation(s)
| | - Christian Brander
- IrsiCaixa AIDS Research Institute-HIVACAT, Badalona, Spain
- Universitat de Vic - Universitat Central de Catalunya (UVIC-UCC), Vic, Spain
- ICREA, Pg. Luis Companys 23, Barcelona, Spain
- AELIX Therapeutics, Barcelona, Spain
| |
Collapse
|