1
|
Fleischmann E, Middelkamp V, van den Broek T. Deciphering the Human Germinal Center: A Review of Models to Study T-B Cell Interactions. Eur J Immunol 2025; 55:e202451460. [PMID: 39931794 PMCID: PMC11811811 DOI: 10.1002/eji.202451460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/09/2025] [Accepted: 01/13/2025] [Indexed: 02/13/2025]
Abstract
Interactions between T- and B cells in the germinal center reaction are instrumental for the initiation, maintenance, and downregulation of the human adaptive immune response, leading to the production of antigen-specific antibodies and long-lasting immunological memory. Replicating the human immune system remains challenging, with an over-reliance on animal models with limited translational accuracy. There is an increasing need for new tools that accurately model human immune function. This review evaluates existing 2D and 3D in vitro and ex vivo human models for their ability to reproduce the germinal center reaction, with a particular focus on T- and B-cell interaction. We conclude that although current models are able to replicate certain features of the germinal center reaction, no current model is able to completely replicate the complex human GC process. We outline the challenges in recreating a fully functional germinal center and suggest future directions of research to improve existing models, ultimately bringing us closer to completely reproducing the human lymph node.
Collapse
Affiliation(s)
- Elisa Fleischmann
- Center for Translational ImmunologyUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Vera Middelkamp
- Center for Translational ImmunologyUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| | - Theo van den Broek
- Center for Translational ImmunologyUniversity Medical Center UtrechtUtrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
2
|
Zatorski JM, Raskovic D, Arneja A, Kiridena S, Ozulumba T, Hammel JH, Anbaei P, Ortiz-Cárdenas JE, Braciale TJ, Munson JM, Luckey CJ, Pompano RR. Initiation of primary T cell-B cell interactions and extrafollicular antibody responses in an organized microphysiological model of the human lymph node. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.12.632545. [PMID: 39868310 PMCID: PMC11761657 DOI: 10.1101/2025.01.12.632545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Antibody production is central to protection against new pathogens and cancers, as well as to certain forms of autoimmunity. Antibodies often originate in the lymph node (LN), specifically at the extrafollicular border of B cell follicles, where T and B lymphocytes physically interact to drive B cell maturation into antibody-secreting plasmablasts. In vitro models of this process are sorely needed to predict aspects of the human immune response. Microphysiological systems (MPSs) offer the opportunity to approximate the lymphoid environment, but so far have focused primarily on memory recall responses to antigens previously encountered by donor cells. To date, no 3D culture system has replicated the engagement between T cells and B cells (T-B interaction) that leads to antibody production when starting with naïve cells. Here, we developed a LN-MPS to model early T-B interactions at the extrafollicular border built from primary, naïve human lymphocytes encapsulated within a collagen-based 3D matrix. Within the MPS, naïve T cells exhibited CCL21-dependent chemotaxis and chemokinesis as predicted. Naïve T and B cells were successfully skewed on chip to an early T follicular helper (pre-Tfh) and activated state, respectively, and co-culture of the latter cells led to CD38+ plasmablast cells and T cell dependent production of IgM. These responses required differentiation of the T cells into pre-Tfhs, physical cell-cell contact, and were sensitive to the ratio at which pre-Tfh and activated B cells were seeded on-chip. Dependence on T cell engagement was greatest at a 1:5 T:B ratio, while cell proliferation and CD38+ signal was greatest at a 1:1 T:B ratio. Furthermore, plasmablast formation was established starting from naïve T and B cells on-chip. We envision that this MPS model of primary lymphocyte physiology will enable new mechanistic analyses of human humoral immunity in vitro.
Collapse
Affiliation(s)
- Jonathan M Zatorski
- Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
| | - Djuro Raskovic
- Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
| | - Abhinav Arneja
- Department of Pathology, University of Virginia, Charlottesville, VA 22908
| | - Saweetha Kiridena
- Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
| | - Tochukwu Ozulumba
- Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
| | - Jennifer H Hammel
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Roanoke, VA
| | - Parastoo Anbaei
- Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
| | - Jennifer E Ortiz-Cárdenas
- Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
- Stanford University, Department of Bioengineering, 443 Via Ortega, Rm 119, Stanford, CA 94305, United States
| | - Thomas J Braciale
- Department of Pathology, University of Virginia, Charlottesville, VA 22908
| | - Jennifer M Munson
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Polytechnic Institute and State University, Roanoke, VA
| | - Chance John Luckey
- Department of Pathology, University of Virginia, Charlottesville, VA 22908
| | - Rebecca R Pompano
- Department of Chemistry, 409 McCormick Road, University of Virginia, Charlottesville, VA 22904
- Department of Pathology, University of Virginia, Charlottesville, VA 22908
- Department of Biomedical Engineering, University of Virginia School of Engineering and Applied Sciences, Thornton Hall, 351 McCormick Rd, Charlottesville, VA 22904
| |
Collapse
|
3
|
Gong S, Beukema M, De Vries-Idema J, Huckriede A. Assessing human B cell responses to influenza virus vaccines and adjuvants in a PBMC-derived in vitro culture system. Vaccine 2025; 44:126563. [PMID: 39616951 DOI: 10.1016/j.vaccine.2024.126563] [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: 07/08/2024] [Revised: 11/19/2024] [Accepted: 11/23/2024] [Indexed: 12/20/2024]
Abstract
In vitro systems based on human peripheral blood mononuclear cells (PBMCs) can bridge the gap between preclinical and clinical vaccine evaluation but have so far mainly been exploited to assess vaccine effects on antigen-presenting cells and T cells. Our study aimed to assess whether B cells present in PBMCs also respond to vaccines and reflect the effects of different vaccine formulations and adjuvants. We stimulated PBMCs with whole inactivated virus (WIV) or split virus (SIV) H5N1 influenza vaccine, with or without the addition of the adjuvant cytosine phosphoguanine (CpG) ODN 2395, and collected the cells and supernatants at different timepoints. B cell subsets were measured by flow cytometry, immunoglobulin (IgG) levels by ELISA, B cell-related genes by qPCR, and cytokine levels by intracellular staining. B cells differentiated more readily to plasmablasts and plasma cells and produced more IgG when PBMC cultures were stimulated with WIV than when stimulated with SIV. In line, PRDM1, XBP1, and AICDA, genes associated with the differentiation of B cells to antibody-secreting cells, were expressed at higher levels in WIV- than in SIV-stimulated PBMCs. The combination of WIV and CpG consistently induced the highest levels of antibody-secreting cell differentiation, IgG production, and B-cells secreting IL-6 and IL-10. Taken together, B cells in human PBMC cultures show distinct responses to different types of vaccines and vaccine/CpG combinations. This underlines the suitability of unfractionated PBMCs for evaluating vaccine effects on different types of human immune cells before running costly clinical trials.
Collapse
MESH Headings
- Humans
- Influenza Vaccines/immunology
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/pharmacology
- Leukocytes, Mononuclear/immunology
- B-Lymphocytes/immunology
- Immunoglobulin G/immunology
- Immunoglobulin G/blood
- Oligodeoxyribonucleotides/immunology
- Oligodeoxyribonucleotides/pharmacology
- Antibodies, Viral/immunology
- Antibodies, Viral/blood
- Influenza A Virus, H5N1 Subtype/immunology
- Cytokines/metabolism
- Cells, Cultured
- Influenza, Human/prevention & control
- Influenza, Human/immunology
- X-Box Binding Protein 1/immunology
- X-Box Binding Protein 1/genetics
- Adult
- Vaccines, Inactivated/immunology
- Cell Differentiation/immunology
- Interleukin-10/metabolism
- Interleukin-10/immunology
- Positive Regulatory Domain I-Binding Factor 1
Collapse
Affiliation(s)
- Shuran Gong
- Department of Medical Microbiology & Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Martin Beukema
- Department of Medical Microbiology & Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jacqueline De Vries-Idema
- Department of Medical Microbiology & Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anke Huckriede
- Department of Medical Microbiology & Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| |
Collapse
|
4
|
Jeger-Madiot R, Planas D, Staropoli I, Debarnot H, Kervevan J, Mary H, Collina C, Fonseca BF, Robinot R, Gellenoncourt S, Schwartz O, Ewart L, Bscheider M, Gobaa S, Chakrabarti LA. Modeling memory B cell responses in a lymphoid organ-chip to evaluate mRNA vaccine boosting. J Exp Med 2024; 221:e20240289. [PMID: 39240335 PMCID: PMC11383861 DOI: 10.1084/jem.20240289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 06/26/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024] Open
Abstract
Predicting the immunogenicity of candidate vaccines in humans remains a challenge. To address this issue, we developed a lymphoid organ-chip (LO chip) model based on a microfluidic chip seeded with human PBMC at high density within a 3D collagen matrix. Perfusion of the SARS-CoV-2 spike protein mimicked a vaccine boost by inducing a massive amplification of spike-specific memory B cells, plasmablast differentiation, and spike-specific antibody secretion. Features of lymphoid tissue, including the formation of activated CD4+ T cell/B cell clusters and the emigration of matured plasmablasts, were recapitulated in the LO chip. Importantly, myeloid cells were competent at capturing and expressing mRNA vectored by lipid nanoparticles, enabling the assessment of responses to mRNA vaccines. Comparison of on-chip responses to Wuhan monovalent and Wuhan/Omicron bivalent mRNA vaccine boosts showed equivalent induction of Omicron neutralizing antibodies, pointing at immune imprinting as reported in vivo. The LO chip thus represents a versatile platform suited to the preclinical evaluation of vaccine-boosting strategies.
Collapse
Affiliation(s)
- Raphaël Jeger-Madiot
- Control of Chronic Viral Infections Group, Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | - Delphine Planas
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | - Isabelle Staropoli
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | - Hippolyte Debarnot
- Control of Chronic Viral Infections Group, Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | - Jérôme Kervevan
- Control of Chronic Viral Infections Group, Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | - Héloïse Mary
- Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Camilla Collina
- Control of Chronic Viral Infections Group, Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | - Barbara F. Fonseca
- Control of Chronic Viral Infections Group, Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
- Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Rémy Robinot
- Control of Chronic Viral Infections Group, Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | - Stacy Gellenoncourt
- Control of Chronic Viral Infections Group, Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | - Olivier Schwartz
- Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| | | | - Michael Bscheider
- Roche Pharma Research & Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Samy Gobaa
- Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Lisa A. Chakrabarti
- Control of Chronic Viral Infections Group, Virus and Immunity Unit, Institut Pasteur, Université de Paris Cité, Centre National de la Recherche Scientifique UMR3569, Paris, France
| |
Collapse
|
5
|
Pasupuleti D, Bagwe P, Ferguson A, Uddin MN, D'Souza MJ, Zughaier SM. Evaluating Nanoparticulate Vaccine Formulations for Effective Antigen Presentation and T-Cell Proliferation Using an In Vitro Overlay Assay. Vaccines (Basel) 2024; 12:1049. [PMID: 39340079 PMCID: PMC11435973 DOI: 10.3390/vaccines12091049] [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: 08/01/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Inducing T lymphocyte (T-cell) activation and proliferation with specificity against a pathogen is crucial in vaccine formulation. Assessing vaccine candidates' ability to induce T-cell proliferation helps optimize formulation for its safety, immunogenicity, and efficacy. Our in-house vaccine candidates use microparticles (MPs) and nanoparticles (NPs) to enhance antigen stability and target delivery to antigen-presenting cells (APCs), providing improved immunogenicity. Typically, vaccine formulations are screened for safety and immunostimulatory effects using in vitro methods, but extensive animal testing is often required to assess immunogenic responses. We identified the need for a rapid, intermediate screening process to select promising candidates before advancing to expensive and time-consuming in vivo evaluations. In this study, an in vitro overlay assay system was demonstrated as an effective high-throughput preclinical testing method to evaluate the immunogenic properties of early-stage vaccine formulations. The overlay assay's effectiveness in testing particulate vaccine candidates for immunogenic responses has been evaluated by optimizing the carboxyfluorescein succinimidyl ester (CFSE) T-cell proliferation assay. DCs were overlaid with T-cells, allowing vaccine-stimulated DCs to present antigens to CFSE-stained T-cells. T-cell proliferation was quantified using flow cytometry on days 0, 1, 2, 4, and 6 upon successful antigen presentation. The assay was tested with nanoparticulate vaccine formulations targeting Neisseria gonorrhoeae (CDC F62, FA19, FA1090), measles, H1N1 flu prototype, canine coronavirus, and Zika, with adjuvants including Alhydrogel® (Alum) and AddaVax™. The assay revealed robust T-cell proliferation in the vaccine treatment groups, with variations between bacterial and viral vaccine candidates. A dose-dependent study indicated immune stimulation varied with antigen dose. These findings highlight the assay's potential to differentiate and quantify effective antigen presentation, providing valuable insights for developing and optimizing vaccine formulations.
Collapse
Affiliation(s)
- Dedeepya Pasupuleti
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Priyal Bagwe
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Amarae Ferguson
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Mohammad N Uddin
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Martin J D'Souza
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery Research, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Susu M Zughaier
- College of Medicine, QU Health, Qatar University, Doha P.O. Box 2731, Qatar
| |
Collapse
|
6
|
Zareein A, Mahmoudi M, Jadhav SS, Wilmore J, Wu Y. Biomaterial engineering strategies for B cell immunity modulations. Biomater Sci 2024; 12:1981-2006. [PMID: 38456305 PMCID: PMC11019864 DOI: 10.1039/d3bm01841e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
B cell immunity has a penetrating effect on human health and diseases. Therapeutics aiming to modulate B cell immunity have achieved remarkable success in combating infections, autoimmunity, and malignancies. However, current treatments still face significant limitations in generating effective long-lasting therapeutic B cell responses for many conditions. As the understanding of B cell biology has deepened in recent years, clearer regulation networks for B cell differentiation and antibody production have emerged, presenting opportunities to overcome current difficulties and realize the full therapeutic potential of B cell immunity. Biomaterial platforms have been developed to leverage these emerging concepts to augment therapeutic humoral immunity by facilitating immunogenic reagent trafficking, regulating T cell responses, and modulating the immune microenvironment. Moreover, biomaterial engineering tools have also advanced our understanding of B cell biology, further expediting the development of novel therapeutics. In this review, we will introduce the general concept of B cell immunobiology and highlight key biomaterial engineering strategies in the areas including B cell targeted antigen delivery, sustained B cell antigen delivery, antigen engineering, T cell help optimization, and B cell suppression. We will also discuss our perspective on future biomaterial engineering opportunities to leverage humoral immunity for therapeutics.
Collapse
Affiliation(s)
- Ali Zareein
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Mina Mahmoudi
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Shruti Sunil Jadhav
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
| | - Joel Wilmore
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Yaoying Wu
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
7
|
Witten J, Hu Y, Langer R, Anderson DG. Recent advances in nanoparticulate RNA delivery systems. Proc Natl Acad Sci U S A 2024; 121:e2307798120. [PMID: 38437569 PMCID: PMC10945842 DOI: 10.1073/pnas.2307798120] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024] Open
Abstract
Nanoparticle-based RNA delivery has shown great progress in recent years with the approval of two mRNA vaccines for Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) and a liver-targeted siRNA therapy. Here, we discuss the preclinical and clinical advancement of new generations of RNA delivery therapies along multiple axes. Improvements in cargo design such as RNA circularization and data-driven untranslated region optimization can drive better mRNA expression. New materials discovery research has driven improved delivery to extrahepatic targets such as the lung and splenic immune cells, which could lead to pulmonary gene therapy and better cancer vaccines, respectively. Other organs and even specific cell types can be targeted for delivery via conjugation of small molecule ligands, antibodies, or peptides to RNA delivery nanoparticles. Moreover, the immune response to any RNA delivery nanoparticle plays a crucial role in determining efficacy. Targeting increased immunogenicity without induction of reactogenic side effects is crucial for vaccines, while minimization of immune response is important for gene therapies. New developments have addressed each of these priorities. Last, we discuss the range of RNA delivery clinical trials targeting diverse organs, cell types, and diseases and suggest some key advances that may play a role in the next wave of therapies.
Collapse
Affiliation(s)
- Jacob Witten
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Yizong Hu
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- Harvard and Massachusetts Institute of Technology Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Anesthesiology, Boston Children’s Hospital, Boston, MA02115
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Daniel G. Anderson
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
- Harvard and Massachusetts Institute of Technology Division of Health Science and Technology, Massachusetts Institute of Technology, Cambridge, MA02139
- Department of Anesthesiology, Boston Children’s Hospital, Boston, MA02115
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA02139
| |
Collapse
|
8
|
Pullen RH, Sassano E, Agrawal P, Escobar J, Chehtane M, Schanen B, Drake DR, Luna E, Brennan RJ. A Predictive Model of Vaccine Reactogenicity Using Data from an In Vitro Human Innate Immunity Assay System. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:904-916. [PMID: 38276072 DOI: 10.4049/jimmunol.2300185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 01/02/2024] [Indexed: 01/27/2024]
Abstract
A primary concern in vaccine development is safety, particularly avoiding an excessive immune reaction in an otherwise healthy individual. An accurate prediction of vaccine reactogenicity using in vitro assays and computational models would facilitate screening and prioritization of novel candidates early in the vaccine development process. Using the modular in vitro immune construct model of human innate immunity, PBMCs from 40 healthy donors were treated with 10 different vaccines of varying reactogenicity profiles and then cell culture supernatants were analyzed via flow cytometry and a multichemokine/cytokine assay. Differential response profiles of innate activity and cell viability were observed in the system. In parallel, an extensive adverse event (AE) dataset for the vaccines was assembled from clinical trial data. A novel reactogenicity scoring framework accounting for the frequency and severity of local and systemic AEs was applied to the clinical data, and a machine learning approach was employed to predict the incidence of clinical AEs from the in vitro assay data. Biomarker analysis suggested that the relative levels of IL-1B, IL-6, IL-10, and CCL4 have higher predictive importance for AE risk. Predictive models were developed for local reactogenicity, systemic reactogenicity, and specific individual AEs. A forward-validation study was performed with a vaccine not used in model development, Trumenba (meningococcal group B vaccine). The clinically observed Trumenba local and systemic reactogenicity fell on the 26th and 93rd percentiles of the ranges predicted by the respective models. Models predicting specific AEs were less accurate. Our study presents a useful framework for the further development of vaccine reactogenicity predictive models.
Collapse
|
9
|
Morrocchi E, van Haren S, Palma P, Levy O. Modeling human immune responses to vaccination in vitro. Trends Immunol 2024; 45:32-47. [PMID: 38135599 PMCID: PMC11688643 DOI: 10.1016/j.it.2023.11.002] [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: 10/31/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/24/2023]
Abstract
The human immune system is a complex network of coordinated components that are crucial for health and disease. Animal models, commonly used to study immunomodulatory agents, are limited by species-specific differences, low throughput, and ethical concerns. In contrast, in vitro modeling of human immune responses can enable species- and population-specific mechanistic studies and translational development within the same study participant. Translational accuracy of in vitro models is enhanced by accounting for genetic, epigenetic, and demographic features such as age, sex, and comorbidity. This review explores various human in vitro immune models, considers evidence that they may resemble human in vivo responses, and assesses their potential to accelerate and de-risk vaccine discovery and development.
Collapse
Affiliation(s)
- Elena Morrocchi
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, Rome, Italy; Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA
| | - Simon van Haren
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Paolo Palma
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, Rome, Italy; Chair of Pediatrics, Department of Systems Medicine, University of Rome 'Tor Vergata', Rome, Italy.
| | - Ofer Levy
- Precision Vaccines Program, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
10
|
Ozulumba T, Montalbine AN, Ortiz-Cárdenas JE, Pompano RR. New tools for immunologists: models of lymph node function from cells to tissues. Front Immunol 2023; 14:1183286. [PMID: 37234163 PMCID: PMC10206051 DOI: 10.3389/fimmu.2023.1183286] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/20/2023] [Indexed: 05/27/2023] Open
Abstract
The lymph node is a highly structured organ that mediates the body's adaptive immune response to antigens and other foreign particles. Central to its function is the distinct spatial assortment of lymphocytes and stromal cells, as well as chemokines that drive the signaling cascades which underpin immune responses. Investigations of lymph node biology were historically explored in vivo in animal models, using technologies that were breakthroughs in their time such as immunofluorescence with monoclonal antibodies, genetic reporters, in vivo two-photon imaging, and, more recently spatial biology techniques. However, new approaches are needed to enable tests of cell behavior and spatiotemporal dynamics under well controlled experimental perturbation, particularly for human immunity. This review presents a suite of technologies, comprising in vitro, ex vivo and in silico models, developed to study the lymph node or its components. We discuss the use of these tools to model cell behaviors in increasing order of complexity, from cell motility, to cell-cell interactions, to organ-level functions such as vaccination. Next, we identify current challenges regarding cell sourcing and culture, real time measurements of lymph node behavior in vivo and tool development for analysis and control of engineered cultures. Finally, we propose new research directions and offer our perspective on the future of this rapidly growing field. We anticipate that this review will be especially beneficial to immunologists looking to expand their toolkit for probing lymph node structure and function.
Collapse
Affiliation(s)
- Tochukwu Ozulumba
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Alyssa N. Montalbine
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, United States
| | - Jennifer E. Ortiz-Cárdenas
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Bioengineering, Stanford University, Stanford, CA, United States
| | - Rebecca R. Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
- Carter Immunology Center and University of Virginia (UVA) Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
11
|
A roadmap for translational cancer glycoimmunology at single cell resolution. J Exp Clin Cancer Res 2022; 41:143. [PMID: 35428302 PMCID: PMC9013178 DOI: 10.1186/s13046-022-02335-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/17/2022] [Indexed: 11/11/2022] Open
Abstract
Cancer cells can evade immune responses by exploiting inhibitory immune checkpoints. Immune checkpoint inhibitor (ICI) therapies based on anti-CTLA-4 and anti-PD-1/PD-L1 antibodies have been extensively explored over the recent years to unleash otherwise compromised anti-cancer immune responses. However, it is also well established that immune suppression is a multifactorial process involving an intricate crosstalk between cancer cells and the immune systems. The cancer glycome is emerging as a relevant source of immune checkpoints governing immunosuppressive behaviour in immune cells, paving an avenue for novel immunotherapeutic options. This review addresses the current state-of-the-art concerning the role played by glycans controlling innate and adaptive immune responses, while shedding light on available experimental models for glycoimmunology. We also emphasize the tremendous progress observed in the development of humanized models for immunology, the paramount contribution of advances in high-throughput single-cell analysis in this context, and the importance of including predictive machine learning algorithms in translational research. This may constitute an important roadmap for glycoimmunology, supporting careful adoption of models foreseeing clinical translation of fundamental glycobiology knowledge towards next generation immunotherapies.
Collapse
|
12
|
Immunogenicity and protective efficacy of RSV G central conserved domain vaccine with a prefusion nanoparticle. NPJ Vaccines 2022; 7:74. [PMID: 35773301 PMCID: PMC9244890 DOI: 10.1038/s41541-022-00487-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 05/19/2022] [Indexed: 11/10/2022] Open
Abstract
Respiratory syncytial virus (RSV) G glycoprotein has recently reemerged as a vaccine antigen due to its ability to elicit potent neutralizing antibodies and ameliorate disease in animal models. Here we designed three constructs to display the G central conserved domain (Gcc) focused on inducing broad and potent neutralizing antibodies. One construct displaying Gcc from both RSV subgroups trimerized via a C-terminal foldon (Gcc-Foldon) was highly immunogenic in mice and in MIMIC, a pre-immune human in vitro model. To explore an optimal RSV vaccine, we combined the Gcc-Foldon antigen with a stabilized pre-fusion-F nanoparticle (pre-F-NP) as a bivalent vaccine and detected no antigenic interference between the two antigens in the MIMIC model. In RSV-primed macaques, the bivalent vaccine elicited potent humoral responses. Furthermore, both Gcc-Foldon and the bivalent vaccine conferred effective protection against RSV challenge in mice. This two-component vaccine could potentially provide effective protection against RSV infection in humans and warrants further clinical evaluation.
Collapse
|
13
|
Hammel JH, Zatorski JM, Cook SR, Pompano RR, Munson JM. Engineering in vitro immune-competent tissue models for testing and evaluation of therapeutics. Adv Drug Deliv Rev 2022; 182:114111. [PMID: 35031388 PMCID: PMC8908413 DOI: 10.1016/j.addr.2022.114111] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 11/16/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022]
Abstract
Advances in 3D cell culture, microscale fluidic control, and cellular analysis have enabled the development of more physiologically-relevant engineered models of human organs with precise control of the cellular microenvironment. Engineered models have been used successfully to answer fundamental biological questions and to screen therapeutics, but these often neglect key elements of the immune system. There are immune elements in every tissue that contribute to healthy and diseased states. Including immune function will be essential for effective preclinical testing of therapeutics for inflammatory and immune-modulated diseases. In this review, we first discuss the key components to consider in designing engineered immune-competent models in terms of physical, chemical, and biological cues. Next, we review recent applications of models of immunity for screening therapeutics for cancer, preclinical evaluation of engineered T cells, modeling autoimmunity, and screening vaccine efficacy. Future work is needed to further recapitulate immune responses in engineered models for the most informative therapeutic screening and evaluation.
Collapse
Affiliation(s)
- Jennifer H. Hammel
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Tech, Roanoke, Virginia 24016, USA
| | - Jonathan M. Zatorski
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Sophie R. Cook
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA
| | - Rebecca R. Pompano
- Department of Chemistry, University of Virginia, Charlottesville, Virginia 22904, USA,Department of Biomedical Engineering, University of Virginia; Charlottesville, Virginia 22904, USA,Carter Immunology Center and UVA Cancer Center, University of Virginia School of Medicine, Charlottesville, Virginia 22903
| | - Jennifer M. Munson
- Fralin Biomedical Research Institute and Department of Biomedical Engineering and Mechanics, Virginia Tech, Roanoke, Virginia 24016, USA
| |
Collapse
|
14
|
Shanti A, Hallfors N, Petroianu GA, Planelles L, Stefanini C. Lymph Nodes-On-Chip: Promising Immune Platforms for Pharmacological and Toxicological Applications. Front Pharmacol 2021; 12:711307. [PMID: 34483920 PMCID: PMC8415712 DOI: 10.3389/fphar.2021.711307] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/04/2021] [Indexed: 12/14/2022] Open
Abstract
Organs-on-chip are gaining increasing attention as promising platforms for drug screening and testing applications. However, lymph nodes-on-chip options remain limited although the lymph node is one of the main determinants of the immunotoxicity of newly developed pharmacological drugs. In this review, we describe existing biomimetic lymph nodes-on-chip, their design, and their physiological relevance to pharmacology and shed the light on future directions associated with lymph node-on-chip design and implementation in drug discovery and development.
Collapse
Affiliation(s)
- Aya Shanti
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Nicholas Hallfors
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Georg A Petroianu
- College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Lourdes Planelles
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Cesare Stefanini
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
15
|
Noe AR, Terry FE, Schanen BC, Sassano E, Hindocha P, Phares TW, Moise L, Christen JM, Tucker KD, Kotraiah V, Drake DR, Martin WD, De Groot AS, Gutierrez GM. Bridging Computational Vaccinology and Vaccine Development Through Systematic Identification, Characterization, and Downselection of Conserved and Variable Circumsporozoite Protein CD4 T Cell Epitopes From Diverse Plasmodium falciparum Strains. Front Immunol 2021; 12:689920. [PMID: 34168657 PMCID: PMC8217813 DOI: 10.3389/fimmu.2021.689920] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 05/06/2021] [Indexed: 11/13/2022] Open
Abstract
An effective malaria vaccine must prevent disease in a range of populations living in regions with vastly different transmission rates and protect against genetically-diverse Plasmodium falciparum (Pf) strains. The protective efficacy afforded by the currently licensed malaria vaccine, Mosquirix™, promotes strong humoral responses to Pf circumsporozoite protein (CSP) 3D7 but protection is limited in duration and by strain variation. Helper CD4 T cells are central to development of protective immune responses, playing roles in B cell activation and maturation processes, cytokine production, and stimulation of effector T cells. Therefore, we took advantage of recent in silico modeling advances to predict and analyze human leukocyte antigen (HLA)-restricted class II epitopes from PfCSP – across the entire PfCSP 3D7 sequence as well as in 539 PfCSP sequence variants – with the goal of improving PfCSP-based malaria vaccines. Specifically, we developed a systematic workflow to identify peptide sequences capable of binding HLA-DR in a context relevant to achieving broad human population coverage utilizing cognate T cell help and with limited T regulatory cell activation triggers. Through this workflow, we identified seven predicted class II epitope clusters in the N- and C-terminal regions of PfCSP 3D7 and an additional eight clusters through comparative analysis of 539 PfCSP sequence variants. A subset of these predicted class II epitope clusters was synthesized as peptides and assessed for HLA-DR binding in vitro. Further, we characterized the functional capacity of these peptides to prime and activate human peripheral blood mononuclear cells (PBMCs), by monitoring cytokine response profiles using MIMIC® technology (Modular IMmune In vitro Construct). Utilizing this decision framework, we found sufficient differential cellular activation and cytokine profiles among HLA-DR-matched PBMC donors to downselect class II epitope clusters for inclusion in a vaccine targeting PfCSP. Importantly, the downselected clusters are not highly conserved across PfCSP variants but rather, they overlap a hypervariable region (TH2R) in the C-terminus of the protein. We recommend assessing these class II epitope clusters within the context of a PfCSP vaccine, employing a test system capable of measuring immunogenicity across a broad set of HLA-DR alleles.
Collapse
Affiliation(s)
- Amy R Noe
- Leidos Life Sciences, Leidos Inc., Frederick, MD, United States
| | | | - Brian C Schanen
- Sanofi Pasteur, VaxDesign Campus, Orlando, FL, United States
| | - Emily Sassano
- Sanofi Pasteur, VaxDesign Campus, Orlando, FL, United States
| | | | | | | | | | | | | | - Donald R Drake
- Sanofi Pasteur, VaxDesign Campus, Orlando, FL, United States
| | | | - Anne S De Groot
- EpiVax Inc., Providence, RI, United States.,University of Georgia Center for Vaccines and Immunology, Athens, GA, United States
| | | |
Collapse
|
16
|
Tucker KD, Schanen BC, Phares TW, Sassano E, Terry FE, Hindocha P, Moise L, Kotraiah V, Martin WD, De Groot AS, Drake DR, Gutierrez GM, Noe AR. Identification, Selection and Immune Assessment of Liver Stage CD8 T Cell Epitopes From Plasmodium falciparum. Front Immunol 2021; 12:684116. [PMID: 34025684 PMCID: PMC8138313 DOI: 10.3389/fimmu.2021.684116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/13/2021] [Indexed: 11/13/2022] Open
Abstract
Immunization with radiation-attenuated sporozoites (RAS) has been shown to protect against malaria infection, primarily through CD8 T cell responses, but protection is limited based on parasite strain. Therefore, while CD8 T cells are an ideal effector population target for liver stage malaria vaccine development strategies, such strategies must incorporate conserved epitopes that cover a large range of class I human leukocyte antigen (HLA) supertypes to elicit cross-strain immunity across the target population. This approach requires identifying and characterizing a wide range of CD8 T cell epitopes for incorporation into a vaccine such that coverage across a large range of class I HLA alleles is attained. Accordingly, we devised an experimental framework to identify CD8 T cell epitopes from novel and minimally characterized antigens found at the pre-erythrocytic stage of parasite development. Through in silico analysis we selected conserved P. falciparum proteins, using P. vivax orthologues to establish stringent conservation parameters, predicted to have a high number of T cell epitopes across a set of six class I HLA alleles representative of major supertypes. Using the decision framework, five proteins were selected based on the density and number of predicted epitopes. Selected epitopes were synthesized as peptides and evaluated for binding to the class I HLA alleles in vitro to verify in silico binding predictions, and subsequently for stimulation of human T cells using the Modular IMmune In-vitro Construct (MIMIC®) technology to verify immunogenicity. By combining the in silico tools with the ex vivo high throughput MIMIC platform, we identified 15 novel CD8 T cell epitopes capable of stimulating an immune response in alleles across the class I HLA panel. We recommend these epitopes should be evaluated in appropriate in vivo humanized immune system models to determine their protective efficacy for potential inclusion in future vaccines.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Anne S. De Groot
- EpiVax Inc., Providence, RI, United States
- University of Georgia Center for Vaccines and Immunology, Athens, GA, United States
| | | | | | - Amy R. Noe
- Leidos Life Sciences, Leidos Inc., Frederick, MD, United States
| |
Collapse
|
17
|
Swanson KA, Rainho-Tomko JN, Williams ZP, Lanza L, Peredelchuk M, Kishko M, Pavot V, Alamares-Sapuay J, Adhikarla H, Gupta S, Chivukula S, Gallichan S, Zhang L, Jackson N, Yoon H, Edwards D, Wei CJ, Nabel GJ. A respiratory syncytial virus (RSV) F protein nanoparticle vaccine focuses antibody responses to a conserved neutralization domain. Sci Immunol 2020; 5:5/47/eaba6466. [PMID: 32358170 DOI: 10.1126/sciimmunol.aba6466] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/09/2020] [Indexed: 12/18/2022]
Abstract
A stabilized form of the respiratory syncytial virus (RSV) fusion (F) protein has been explored as a vaccine to prevent viral infection because it presents several potent neutralizing epitopes. Here, we used a structure-based rational design to optimize antigen presentation and focus antibody (Ab) responses to key epitopes on the pre-fusion (pre-F) protein. This protein was fused to ferritin nanoparticles (pre-F-NP) and modified with glycans to mask nonneutralizing or poorly neutralizing epitopes to further focus the Ab response. The multimeric pre-F-NP elicited durable pre-F-specific Abs in nonhuman primates (NHPs) after >150 days and elicited potent neutralizing Ab (NAb) responses in mice and NHPs in vivo, as well as in human cells evaluated in the in vitro MIMIC system. This optimized pre-F-NP stimulated a more potent Ab response than a representative pre-F trimer, DS-Cav1. Collectively, this pre-F vaccine increased the generation of NAbs targeting the desired pre-F conformation, an attribute that facilitates the development of an effective RSV vaccine.
Collapse
Affiliation(s)
| | | | - Zachary P Williams
- Sanofi Pasteur VaxDesign, 2501 Discovery Drive, Suite 300, Orlando, FL 32826, USA
| | - Lilibeth Lanza
- Sanofi Pasteur VaxDesign, 2501 Discovery Drive, Suite 300, Orlando, FL 32826, USA
| | - Michael Peredelchuk
- Sanofi Pasteur VaxDesign, 2501 Discovery Drive, Suite 300, Orlando, FL 32826, USA
| | - Michael Kishko
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, USA
| | - Vincent Pavot
- Sanofi Pasteur, 1541 Avenue Marcel Mérieux, Marcy l'Etoile, France
| | | | | | - Sankalp Gupta
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, USA
| | | | - Scott Gallichan
- Sanofi Pasteur, 95 Willowdale Blvd, Toronto, Ontario, Canada
| | - Linong Zhang
- Sanofi Pasteur, 38 Sidney Street, Cambridge, MA 02139, USA
| | | | - Heesik Yoon
- Sanofi Pasteur VaxDesign, 2501 Discovery Drive, Suite 300, Orlando, FL 32826, USA
| | - Darin Edwards
- Sanofi Pasteur VaxDesign, 2501 Discovery Drive, Suite 300, Orlando, FL 32826, USA
| | | | - Gary J Nabel
- Sanofi, 640 Memorial Drive, Cambridge, MA 02139, USA.
| |
Collapse
|
18
|
Safety biomarkers for development of vaccines and biologics: Report from the safety biomarkers symposium held on November 28-29, 2017, Marcy l'Etoile, France. Vaccine 2020; 38:8055-8063. [PMID: 33187767 DOI: 10.1016/j.vaccine.2020.10.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 09/14/2020] [Accepted: 10/06/2020] [Indexed: 12/27/2022]
Abstract
Vaccines prevent infectious diseases, but vaccination is not without risk and adverse events are reported although they are more commonly reported for biologicals than for vaccines. Vaccines and biologicals must undergo vigorous assessment before and after licensure to minimise safety concerns. Potential safety concerns should be identified as early as possible during the development for vaccines and biologicals to minimize investment risk. State-of-the art tools and methods to identify safety concerns and biomarkers that are predictive of clinical outcomes are indispensable. For vaccines and adjuvant formulations, systems biology approaches, supported by single-cell microfluidics applied to translational studies between preclinical and clinical studies, could improve reactogenicity and safety predictions. Next-generation animal models for clinical assessment of injection-site reactions with greater relevance for target human population and criteria to define the level of acceptability of local reactogenicity at vaccine injection sites in pre-clinical animal species should be assessed. Advanced in silico machine-learning-based analytics, species-specific cell or tissue expression, receptor occupancy and kinetics and cell-based assays for functional activity are needed to improve pre-clinical safety assessment of biologicals. The in vitro MIMIC® system could be used to compliment preclinical and clinical studies for assessing immune-toxicity, immunogenicity, immuno-inflammatory and mode of action of biologicals and vaccines. Sanofi Pasteur brought together leading experts in this field to review the state-of-the-art at a unique 'Safety Biomarkers Symposium' on 28-29 November 2017. Here we summarise the proceedings of this symposium. This unique scientific meeting confirmed the importance for institutions and industrial organizations to collaborate to develop tools and methods needed for predicting reactogenicity and immune-inflammatory reactions to vaccines and biologicals, and to develop more accuracy, reliability safety biomarkers, to inform decisions on the attrition or advancement of vaccines and biologicals.
Collapse
|
19
|
Najibi AJ, Mooney DJ. Cell and tissue engineering in lymph nodes for cancer immunotherapy. Adv Drug Deliv Rev 2020; 161-162:42-62. [PMID: 32750376 PMCID: PMC7736208 DOI: 10.1016/j.addr.2020.07.023] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/03/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023]
Abstract
In cancer, lymph nodes (LNs) coordinate tumor antigen presentation necessary for effective antitumor immunity, both at the levels of local cellular interactions and tissue-level organization. In this review, we examine how LNs may be engineered to improve the therapeutic outcomes of cancer immunotherapy. At the cellular scale, targeting the LNs impacts the potency of cancer vaccines, immune checkpoint blockade, and adoptive cell transfer. On a tissue level, macro-scale biomaterials mimicking LN features can function as immune niches for cell reprogramming or delivery in vivo, or be utilized in vitro to enable preclinical testing of drugs and vaccines. We additionally review strategies to induce ectopic lymphoid sites reminiscent of LNs that may improve antitumor T cell priming.
Collapse
Affiliation(s)
- Alexander J Najibi
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138
| | - David J Mooney
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138; Wyss Institute for Biologically Inspired Engineering, Harvard University, Cambridge, MA 02138.
| |
Collapse
|
20
|
Jawa V, Terry F, Gokemeijer J, Mitra-Kaushik S, Roberts BJ, Tourdot S, De Groot AS. T-Cell Dependent Immunogenicity of Protein Therapeutics Pre-clinical Assessment and Mitigation-Updated Consensus and Review 2020. Front Immunol 2020; 11:1301. [PMID: 32695107 PMCID: PMC7338774 DOI: 10.3389/fimmu.2020.01301] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 05/22/2020] [Indexed: 01/01/2023] Open
Abstract
Immune responses to protein and peptide drugs can alter or reduce their efficacy and may be associated with adverse effects. While anti-drug antibodies (ADA) are a standard clinical measure of protein therapeutic immunogenicity, T cell epitopes in the primary sequences of these drugs are the key drivers or modulators of ADA response, depending on the type of T cell response that is stimulated (e.g., T helper or Regulatory T cells, respectively). In a previous publication on T cell-dependent immunogenicity of biotherapeutics, we addressed mitigation efforts such as identifying and reducing the presence of T cell epitopes or T cell response to protein therapeutics prior to further development of the protein therapeutic for clinical use. Over the past 5 years, greater insight into the role of regulatory T cell epitopes and the conservation of T cell epitopes with self (beyond germline) has improved the preclinical assessment of immunogenic potential. In addition, impurities contained in therapeutic drug formulations such as host cell proteins have also attracted attention and become the focus of novel risk assessment methods. Target effects have come into focus, given the emergence of protein and peptide drugs that target immune receptors in immuno-oncology applications. Lastly, new modalities are entering the clinic, leading to the need to revise certain aspects of the preclinical immunogenicity assessment pathway. In addition to drugs that have multiple antibody-derived domains or non-antibody scaffolds, therapeutic drugs may now be introduced via viral vectors, cell-based constructs, or nucleic acid based therapeutics that may, in addition to delivering drug, also prime the immune system, driving immune response to the delivery vehicle as well as the encoded therapeutic, adding to the complexity of assessing immunogenicity risk. While it is challenging to keep pace with emerging methods for the preclinical assessment of protein therapeutics and new biologic therapeutic modalities, this collective compendium provides a guide to current best practices and new concepts in the field.
Collapse
Affiliation(s)
- Vibha Jawa
- Predictive and Clinical Immunogenicity, PPDM, Merck & Co., Kenilworth, NJ, United States
| | | | - Jochem Gokemeijer
- Discovery Biotherapeutics, Bristol-Myers Squibb, Cambridge, MA, United States
| | | | | | - Sophie Tourdot
- BioMedicine Design, Pfizer Inc., Andover, MA, United States
| | - Anne S De Groot
- EpiVax, Inc., Providence, RI, United States.,Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
| |
Collapse
|
21
|
Current In Vitro Assays for Prediction of T Cell Mediated Immunogenicity of Biotherapeutics and Manufacturing Impurities. J Pharm Innov 2019. [DOI: 10.1007/s12247-019-09412-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
22
|
Dull P, Friede M, Hwang A, Hall BF. Meeting report: Global vaccine and immunization research forum, 2018. Vaccine 2019; 37:7519-7526. [PMID: 31623915 PMCID: PMC6899432 DOI: 10.1016/j.vaccine.2019.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 09/27/2019] [Accepted: 10/01/2019] [Indexed: 11/26/2022]
Abstract
Every two years, the Global Vaccine and Immunization Research Forum takes stock of global research in vaccines and immunization. As in prior years, the 2018 meeting addressed vaccine discovery, development, decision-making, and deployment. This time, however, it also featured two overarching themes: "Innovating for Equity" and "End-to-End Integration." Significant advances have been made in the last two years, but participants noted that some important goals of the Global Vaccine Action Plan are not being met and called urgently for innovation in improving access to vaccines. Two factors were highlighted as crucial to improving coverage: a focus on equity and sustainability throughout the immunization ecosystem, and an enabling political environment that prioritizes health and immunization.
Collapse
Affiliation(s)
- Peter Dull
- Bill & Melinda Gates Foundation, PO Box 23350, Seattle, WA 98102. USA.
| | - Martin Friede
- Department of Immunization, Vaccines, and Biologicals, World Health Organization, Avenue Appia 20, 1211 Geneva 27, Switzerland.
| | - Angela Hwang
- Angela Hwang Consulting, PO Box 6601, Albany, CA 94706. USA.
| | - B Fenton Hall
- Parasitology & International Programs Branch, Division of Microbiology and Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, MSC 9825, Bethesda, MD 20892-9825, USA.
| |
Collapse
|
23
|
Influenza A and B virus-like particles produced in mammalian cells are highly immunogenic and induce functional antibodies. Vaccine 2019; 37:6857-6867. [DOI: 10.1016/j.vaccine.2019.09.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 09/13/2019] [Accepted: 09/18/2019] [Indexed: 12/16/2022]
|
24
|
Azizi A. Protective Efficacy of Candidate Vaccines Prior to Human Clinical Trials. J Pharm Sci 2018; 107:2992-2994. [PMID: 30121314 DOI: 10.1016/j.xphs.2018.08.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 07/31/2018] [Accepted: 08/07/2018] [Indexed: 12/23/2022]
Affiliation(s)
- Ali Azizi
- Sanofi Pasteur, Analytical Research and Development North America, Toronto, Ontario M2R 3T4, Canada.
| |
Collapse
|
25
|
Wagar LE, DiFazio RM, Davis MM. Advanced model systems and tools for basic and translational human immunology. Genome Med 2018; 10:73. [PMID: 30266097 PMCID: PMC6162943 DOI: 10.1186/s13073-018-0584-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/17/2018] [Indexed: 12/31/2022] Open
Abstract
There are fundamental differences between humans and the animals we typically use to study the immune system. We have learned much from genetically manipulated and inbred animal models, but instances in which these findings have been successfully translated to human immunity have been rare. Embracing the genetic and environmental diversity of humans can tell us about the fundamental biology of immune cell types and the elasticity of the immune system. Although people are much more immunologically diverse than conventionally housed animal models, tools and technologies are now available that permit high-throughput analysis of human samples, including both blood and tissues, which will give us deep insights into human immunity in health and disease. As we gain a more detailed picture of the human immune system, we can build more sophisticated models to better reflect this complexity, both enabling the discovery of new immunological mechanisms and facilitating translation into the clinic.
Collapse
Affiliation(s)
- Lisa E Wagar
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Robert M DiFazio
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Mark M Davis
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
26
|
Streatfield SJ, Karczewski J, Yusibov V. Introduction. Vaccine 2017; 35:5435-5436. [PMID: 28826749 PMCID: PMC7130944 DOI: 10.1016/j.vaccine.2017.08.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | - Jerzy Karczewski
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| | - Vidadi Yusibov
- Fraunhofer USA Center for Molecular Biotechnology, Newark, DE, USA
| |
Collapse
|