1
|
Long AH, Aftandilian C, Barmettler S, Alexander S. Hypogammaglobulinemia in Children Receiving Targeted Immunotherapies for B Lineage Malignancies: Practical Guidance for Assessment and Management. Pediatr Blood Cancer 2025:e31779. [PMID: 40372257 DOI: 10.1002/pbc.31779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/21/2025] [Accepted: 04/24/2025] [Indexed: 05/16/2025]
Abstract
Hypogammaglobulinemia is a well-defined risk factor for infection. B-cell-directed immunotherapies given in addition to conventional chemotherapy are now core elements of effective therapy for children with B lymphoid malignancies. These therapies are associated with depletion of normal B cells and consequent hypogammaglobulinemia. This review summarizes the current state of knowledge regarding the mechanism, incidence, and clinical outcomes related to hypogammaglobulinemia in children with mature B-cell non-Hodgkin lymphoma and B-cell acute lymphoblastic leukemia, as well as provides practical guidance for laboratory monitoring and considerations for immunoglobulin replacement therapy.
Collapse
Affiliation(s)
- Adrienne H Long
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Catherine Aftandilian
- Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, California, USA
| | - Sara Barmettler
- Division of Rheumatology, Allergy and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sarah Alexander
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Lane A, Quach HQ, Ovsyannikova IG, Kennedy RB, Ross TM, Einav T. Characterizing the Short- and Long-Term Temporal Dynamics of Antibody Responses to Influenza Vaccination. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.26.25322965. [PMID: 40061340 PMCID: PMC11888507 DOI: 10.1101/2025.02.26.25322965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Most influenza vaccine studies evaluate acute antibody responses 1 month post-vaccination, leaving long-term immunity poorly understood. Here, we performed a combined analysis of 14 large-scale vaccine studies and conducted two new studies mapping antibody responses in high resolution from their inception out to 1 year post-vaccination. Vaccine antibody responses were classified as weak (<4x fold-change at 1 month and 1 year), transient (≥4x at 1 month, <4x at 1 year), or durable (≥4x at 1 month and 1 year). Surprisingly, >50% of vaccine recipients were weak across seasons, age groups, sexes, pre-vaccination titers, and high or standard vaccine doses. Peak fold-change at 1 month post-vaccination was strongly associated with the long-term response, with most transient responders achieving a maximum fold-change of 4x, while most durable responders reached ≥16x, with both groups maintaining these titers for 2 months (10-75 days post-vaccination). Using the weak, transient, and durable trajectories, a single time point early in the response (days 7-8 or 21) predicted an individual's response out to 1 year post-vaccination. These results demonstrate that influenza vaccine responses range from little-to-no response to eliciting strong-and-durable immunity, highlighting the stark heterogeneity that is consistently seen across influenza seasons.
Collapse
Affiliation(s)
- Aaron Lane
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Huy Q Quach
- Vaccine Research Group, Mayo Clinic, Rochester, MN 55905, USA
| | | | | | - Ted M Ross
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
- Department of Infectious Diseases, University of Georgia, Athens, GA 30602, USA
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
- Department of Infection Biology, Lehner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Tal Einav
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
- Department of Medicine, University of California San Diego, La Jolla, CA 92037, USA
| |
Collapse
|
3
|
Li M, Feng M, Liu T, Duan S, Man X, Yuan X, Wang L, Sun Y, Wei X, Fu Q, Sun B, Lin W. Increased ocular plasma cells induce damaging α-synuclein + microglia in autoimmune uveitis. Mucosal Immunol 2025:S1933-0219(25)00025-X. [PMID: 40015479 DOI: 10.1016/j.mucimm.2025.02.007] [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: 05/05/2024] [Revised: 02/23/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
Autoimmune uveitis (AIU) is an immune-inflammatory disease that can lead to blindness. However, incomplete understanding of the involved immune cell subsets and their contributions to retinal injury has hindered the development of effective AIU therapies. Using single-cell RNA sequencing and immunofluorescence, we identified α-synuclein+ microglia as the primary subset of damaged ocular cells in the eyes of the experimental autoimmune uveitis (EAU) mouse model. Ocular-infiltrating plasma cells (PCs) were shown to express multiple inflammatory factors, particularly TNF-α, which promoted the production of α-synuclein+ microglia. Studies of heterogeneous PC subtypes revealed that MUC1- PCs represent the primary pathogenic subset, secreting multiple cytokines. Although MUC1+ PCs expressed TGF-β, they exhibited long-lived characteristics and secreted IgG and IgM, thereby prolonging disease progression. Finally, the small G protein Rab1A, also expressed in the PCs of Vogt-Koyanagi-Harada (VKH) patients, was found to mediate autophagy and NF-κB expression, influencing PCs survival and inflammatory responses. Silencing or knocking down Rab1A in PCs inhibited their survival. This study elucidates potential mechanisms underlying the neuroimmune inflammatory response and highlights the previously unrecognized role of infiltrating PCs in AIU, offering novel therapeutic targets for this disease.
Collapse
Affiliation(s)
- Minghao Li
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China; School of Clinical and Basic Medicine, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, China; Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Medicine and Health Key Laboratory of Rheumatism, The First Affiliated Hospital of Shandong First Medical University, Jinan, China; Department of Critical-care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Meng Feng
- Department of Laboratory Medicine, The Affiliated Hospital of Shaanxi University of Chinese Medicine, Deputy 2, West Road Weiyang, Qindu District, Xianyang, Shaanxi 712000, China; The First Affiliated Hospital of Shaanxi University of Traditional Chinese Medicine, Weiyang West Road, Xianyang City 710077 Shaanxi Province, China
| | - Tingting Liu
- Shandong Eye Hospital, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China; Department of Ophthalmology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, China
| | - Songqi Duan
- College of Food Science, Sichuan Agricultural University, Ya an, China
| | - Xuejing Man
- Department of Ophthalmology, Yuhuangding Hospital, Yantai, China
| | - Xiaomeng Yuan
- Shandong Eye Hospital, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, Shandong Eye Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Lijie Wang
- School of Clinical and Basic Medicine, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, China
| | - Yu Sun
- Shanghai Jiao Tong University, Subei Research Institute, Anti-aging Innovation Center, Shanghai, China; School of Pharmacology, Institute of Aging Medicine, Binzhou Medical University,Yantai, China
| | - Xunbin Wei
- Biomedical Engineering Department, Peking University, Beijing, China; School of Biomedical Engineering, Anhui Medical University, Hefei, China; Institute of Medical Technology, Peking University Health Science Center, Beijing, China; Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China; International Cancer Institute, Peking University, Beijing, China
| | - Qiang Fu
- Shanghai Jiao Tong University, Subei Research Institute, Anti-aging Innovation Center, Shanghai, China; School of Pharmacology, Institute of Aging Medicine, Binzhou Medical University,Yantai, China.
| | - Baofa Sun
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Center for Cell Responses, College of Life Sciences, Nankai University, Tianjin, China.
| | - Wei Lin
- Department of Rheumatology and Autoimmunology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China; School of Clinical and Basic Medicine, Shandong First Medical University &Shandong Academy of Medical Sciences, Jinan, China; Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Shandong Medicine and Health Key Laboratory of Rheumatism, The First Affiliated Hospital of Shandong First Medical University, Jinan, China; Department of Critical-care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| |
Collapse
|
4
|
Stocks D, Thomas A, Finn A, Danon L, Brooks-Pollock E. Mechanistic models of humoral kinetics following COVID-19 vaccination. J R Soc Interface 2025; 22:20240445. [PMID: 39876790 PMCID: PMC11775660 DOI: 10.1098/rsif.2024.0445] [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: 07/01/2024] [Revised: 10/07/2024] [Accepted: 11/01/2024] [Indexed: 01/31/2025] Open
Abstract
COVID-19 vaccine programmes must account for variable immune responses and waning protection. Existing descriptions of antibody responses to COVID-19 vaccination convey limited information about the mechanisms of antibody production and maintenance. We describe antibody dynamics after COVID-19 vaccination with two biologically motivated mathematical models. We fit the models using Markov chain Monte Carlo to seroprevalence data from 14 602 uninfected individuals in England between May 2020 and September 2022. We analyse the effect of age, vaccine type, number of doses and the interval between doses on antibody production and longevity. We find evidence that individuals over 35 years old twice vaccinated with ChAdOx1-S generate a persistent antibody response suggestive of long-lived plasma cell induction. We also find that plasmablast productive capacity is greater in: younger people than older people (≤4.5-fold change in point estimates); people vaccinated with two doses than one dose (≤12-fold change); and people vaccinated with BNT162b2 than ChAdOx1-S (≤440-fold change). We find the half-life of an antibody to be 23-106 days. Routinely collected seroprevalence data are invaluable for characterizing within-host mechanisms of antibody production and persistence. Extended sampling and linking seroprevalence data to outcomes would enable conclusions about how humoral kinetics protect against disease.
Collapse
Affiliation(s)
- Daniel Stocks
- School of Engineering Mathematics and Technology, University of Bristol, Tankard’s Close, Bristol, BS8 1TW, UK
| | - Amy Thomas
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Adam Finn
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Leon Danon
- School of Engineering Mathematics and Technology, University of Bristol, Tankard’s Close, Bristol, BS8 1TW, UK
| | - Ellen Brooks-Pollock
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield Grove, Bristol, BS8 2BN, UK
| |
Collapse
|
5
|
Arana C, Garcia-Busquets A, Nicoli M, Betriu S, Gille I, Heemskerk MHM, Heidt S, Palou E, Rovira J, Diekmann F. Chimeric HLA antibody receptor T cell therapy for humoral transplant rejection. Nephrol Dial Transplant 2024; 40:19-26. [PMID: 39025810 DOI: 10.1093/ndt/gfae160] [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: 04/03/2024] [Indexed: 07/20/2024] Open
Abstract
Antibody-mediated rejection (ABMR) is a significant obstacle to achieving optimal long-term outcomes after solid organ transplantation. The presence of donor-specific antibodies (DSAs), particularly against human leucocyte antigen (HLA), increases the risk of allograft rejection and subsequent graft loss. No effective treatment for ABMR currently exists, warranting novel approaches to target the HLA-specific humoral alloimmune response. Cellular therapies may hold promise to this end. According to publicly available sources as of now, three independent laboratories have genetically engineered a chimeric HLA antibody receptor (CHAR) and transduced it into human T cells, based on the demonstrated efficacy of chimeric antigen receptor T cell therapies in malignancies. These CHAR-T cells are designed to exclusively eliminate B cells that produce donor-specific HLA antibodies, which form the cornerstone of ABMR. CHAR technology generates potent and functional human cytotoxic T cells to target alloreactive HLA-specific B cells, sparing B cells with other specificities. Thus CHAR technology may be used as a selective desensitization protocol and to treat ABMR after solid organ transplantation.
Collapse
Affiliation(s)
- Carolt Arana
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Nephrology and Kidney Transplantation. Institut Clínic de Nefrologia i Urologia (ICNU), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Ainhoa Garcia-Busquets
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Michael Nicoli
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sergi Betriu
- Department of Immunology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Ilse Gille
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Mirjam H M Heemskerk
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Internal Medicine, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Eduard Palou
- Department of Immunology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Jordi Rovira
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Fritz Diekmann
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Department of Nephrology and Kidney Transplantation. Institut Clínic de Nefrologia i Urologia (ICNU), Hospital Clínic de Barcelona, Barcelona, Spain
| |
Collapse
|
6
|
Hirano H, Asada H. Exponential decline, ceiling effect, downregulation, and T-cell response in immunoglobulin G antibody levels after messenger RNA vaccine boosters: a case report. J Med Case Rep 2024; 18:631. [PMID: 39707550 DOI: 10.1186/s13256-024-04889-2] [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: 01/25/2024] [Accepted: 10/09/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND Vaccine protection against severe acute respiratory syndrome coronavirus 2 infection reduces gradually over time, requiring administration of updated boosters. However, long-term immune response following up to the sixth dose of the messenger RNA vaccine has not been well studied. CASE PRESENTATION We longitudinally determined anti-spike protein immunoglobulin G antibody levels in a 69-year-old Japanese man 76 times (first to sixth dose) to investigate their dynamics. Regarding the messenger RNA BNT162b2 vaccine, first to fourth doses were identical monovalent vaccines, and fifth and sixth doses were identical bivalent vaccines. T-cell responses after fourth and fifth doses were studied using T-SPOT. Immunoglobulin G levels peaked at 1-2 weeks after second to sixth dose, declining exponentially after each dose. The decline was approximated using the formula f (t) = Ae-t/τ + C. Time constant τ increased with each booster vaccination, indicating a decreasing rate of antibody titer decay with increasing number of doses. Baseline and peak immunoglobulin G levels were similar in the second and third dose. Conversely, baseline immunoglobulin G levels after the fourth dose increased over fivefold over the second and third dose; however, peak immunoglobulin G levels after fourth dose decreased to 60% of those after the third dose. Baseline immunoglobulin G levels after the sixth dose increased 1.4-fold over the fifth dose; however, peak immunoglobulin G levels after the sixth dose decreased to 56% of those after the fifth dose. Dynamics of T-cell responses differed from those of immunoglobulin G antibodies. T cell responses increased gradually; however, their peak level was difficult to determine. CONCLUSIONS Ceiling effect or downregulation of peak immunoglobulin G levels was clearly observed after messenger RNA booster vaccination. After peaking, the IgG level declined exponentially, and the rate of decay decreased with each subsequent booster. Although this was a single-case study, this data may provide a generalized mathematical decay model for humoral immunity in healthy older adults. Moreover, our study provides insights into the immunogenicity after booster vaccination with messenger RNA vaccines.
Collapse
Affiliation(s)
- Harukazu Hirano
- Koyo Seikyo Clinic, Fukui Health Cooperative Association, 3-9-23 Koyo, Fukui, 910-0026, Japan.
| | - Hiroshi Asada
- Department of Applied Physics, Faculty of Engineering, University of Fukui, 3-9-1 Bunkyo, Fukui, 910-8507, Japan
| |
Collapse
|
7
|
Kolachala VL, Wei C, Venkateswaran S, Hill AL, Warren V, Espinoza H, Sanz I, Gupta NA. Increased IgD and CD27 Double Negative (DN) B cell population in pediatric onset autoimmune hepatitis. Autoimmunity 2024; 57:2356089. [PMID: 38770919 DOI: 10.1080/08916934.2024.2356089] [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: 06/04/2023] [Accepted: 05/12/2024] [Indexed: 05/22/2024]
Abstract
Autoimmune hepatitis (AIH) is a chronic, inflammatory liver disease of unknown aetiology which requires lifelong immunosuppression. Most therapeutic and outcome studies of AIH have been conducted predominantly in Caucasian (European Ancestry, EA) cohorts, with the exclusion of African American (AA) patients due to inadequate sample size. It is known that AA patients have a severe phenotype of autoimmune diseases and demonstrate a poor response to conventional medical therapy. Understanding cellular and molecular pathways which determine AIH severity and progression in AA patients is likely to lead to the discovery of novel, personalised and better tolerated therapies. The aim of the study is to determine the distinct effector B cell phenotypes which contribute to disease severity and progression of AIH in AA children as compared to their EA cohorts. PBMCs were isolated from blood samples collected from patients visiting Children's Healthcare of Atlanta (CHOA) and were grouped into AA, (n = 12), EA, (n = 11) and controls (n = 12) and were processed for flow cytometry. Markers of B cell development, maturation and activation were assessed namely CD19, CD21, IgD, CD27, CD38, CD11c, CD24, CD138. AA children with AIH demonstrated an expansion of CD19 + ve, Activated Naïve (aN), (CD19+ IgD-/CD27- Double Negative (DN2) ([CD19+/IgD-/CD27++CD38++) cells. Plasmablasts were significantly higher along with Signalling Lymphocytic activation molecule F7 (SLAMF7). Unswitched memory [CD19+] IgD+CD27+ (USM) B cells were significantly contracted in AA patients with AIH. B cell phenotyping reveals a distinct profile in AA AIH patients with a major skewing towards the expansion of effector pathways which have been previously characterised in severe SLE in AA patients. These results suggest that the quantification and therapeutic target of B cell pathway could contribute substantially to the clinical approach to AIH especially in the AA population.
Collapse
Affiliation(s)
| | - Chungwen Wei
- Division of Rheumatology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Medicine, Lowance Center for Human Immunology, Atlanta, GA, USA
- Department of Medicine, Emory Autoimmunity Center of Excellence, Atlanta, GA, USA
| | | | - Aisha Latrece Hill
- Division of Rheumatology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Medicine, Lowance Center for Human Immunology, Atlanta, GA, USA
- Department of Medicine, Emory Autoimmunity Center of Excellence, Atlanta, GA, USA
| | - Vivian Warren
- Division of Rheumatology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Medicine, Lowance Center for Human Immunology, Atlanta, GA, USA
- Department of Medicine, Emory Autoimmunity Center of Excellence, Atlanta, GA, USA
| | - Hillary Espinoza
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Pediatric Transplant, Children's Healthcare of Atlanta, Transplant Services, Atlanta, GA, USA
| | - Iñaki Sanz
- Division of Rheumatology, Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
- Department of Medicine, Lowance Center for Human Immunology, Atlanta, GA, USA
- Department of Medicine, Emory Autoimmunity Center of Excellence, Atlanta, GA, USA
| | - Nitika A Gupta
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
- Pediatric Transplant, Children's Healthcare of Atlanta, Transplant Services, Atlanta, GA, USA
| |
Collapse
|
8
|
Rogers GL, Huang C, Mathur A, Huang X, Chen HY, Stanten K, Morales H, Chang CH, Kezirian EJ, Cannon PM. Reprogramming human B cells with custom heavy-chain antibodies. Nat Biomed Eng 2024; 8:1700-1714. [PMID: 39039240 DOI: 10.1038/s41551-024-01240-4] [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: 06/07/2024] [Accepted: 06/22/2024] [Indexed: 07/24/2024]
Abstract
The immunoglobulin locus of B cells can be reprogrammed by genome editing to produce custom or non-natural antibodies that are not induced by immunization. However, current strategies for antibody reprogramming require complex expression cassettes and do not allow for customization of the constant region of the antibody. Here we show that human B cells can be edited at the immunoglobulin heavy-chain locus to express heavy-chain-only antibodies that support alterations to both the fragment crystallizable domain and the antigen-binding domain, which can be based on both antibody and non-antibody components. Using the envelope protein (Env) from the human immunodeficiency virus as a model antigen, we show that B cells edited to express heavy-chain antibodies to Env support the regulated expression of B cell receptors and antibodies through alternative splicing and that the cells respond to the Env antigen in a tonsil organoid model of immunization. This strategy allows for the reprogramming of human B cells to retain the potential for in vivo amplification while producing molecules with flexibility of composition beyond that of standard antibodies.
Collapse
Affiliation(s)
- Geoffrey L Rogers
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Chun Huang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Atishay Mathur
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Xiaoli Huang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Hsu-Yu Chen
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Kalya Stanten
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Heidy Morales
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Chan-Hua Chang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Eric J Kezirian
- Department of Otolaryngology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Paula M Cannon
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
9
|
Lam N, Lee Y, Farber DL. A guide to adaptive immune memory. Nat Rev Immunol 2024; 24:810-829. [PMID: 38831162 DOI: 10.1038/s41577-024-01040-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/26/2024] [Indexed: 06/05/2024]
Abstract
Immune memory - comprising T cells, B cells and plasma cells and their secreted antibodies - is crucial for human survival. It enables the rapid and effective clearance of a pathogen after re-exposure, to minimize damage to the host. When antigen-experienced, memory T cells become activated, they proliferate and produce effector molecules at faster rates and in greater magnitudes than antigen-inexperienced, naive cells. Similarly, memory B cells become activated and differentiate into antibody-secreting cells more rapidly than naive B cells, and they undergo processes that increase their affinity for antigen. The ability of T cells and B cells to form memory cells after antigen exposure is the rationale behind vaccination. Understanding immune memory not only is crucial for the design of more-efficacious vaccines but also has important implications for immunotherapies in infectious disease and cancer. This 'guide to' article provides an overview of the current understanding of the phenotype, function, location, and pathways for the generation, maintenance and protective capacity of memory T cells and memory B cells.
Collapse
Affiliation(s)
- Nora Lam
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - YoonSeung Lee
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
| | - Donna L Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
10
|
Yu-Hong Cheng R, Helmers AE, Kreuser S, Dahl N, Honaker Y, Lopez C, Rawlings DJ, James RG. Generation, expansion, gene delivery, and single-cell profiling in rhesus macaque plasma B cells. CELL REPORTS METHODS 2024; 4:100878. [PMID: 39406231 PMCID: PMC11573788 DOI: 10.1016/j.crmeth.2024.100878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 07/18/2024] [Accepted: 09/20/2024] [Indexed: 10/25/2024]
Abstract
A key step in developing engineered B cells for therapeutic purposes is evaluation in immunocompetent, large-animal models. Therefore, we developed methods to purify, expand, and differentiate non-human primate (NHP; rhesus macaque) B cells. After 7 days in culture, B cells expanded 10-fold, differentiated into a plasma cell phenotype (CD38, CD138), and secreted immunoglobulin G. Using single-cell sequencing and flow cytometry, we verified the presence of plasma cell genes in differentiated NHP B cells and unearthed less-recognized markers, such as CD59 and CD79A. In contrast with human cells, we found that the immune checkpoint molecule CD274 (PD-L1) and major histocompatibility complex (MHC) class I molecules were upregulated in NHP plasma cells in the transcriptional data. Lastly, we established the conditions for efficient transduction of NHP B cells with adeno-associated virus (AAV) vectors, achieving a delivery rate of approximately 60%. We envision that this work will accelerate proof-of-concept studies using engineered B cells in NHPs.
Collapse
Affiliation(s)
- Rene Yu-Hong Cheng
- Center of Immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA 98101, USA; Molecular Engineering and Science Institute, University of Washington, Seattle, WA 98195, USA
| | - Anna E Helmers
- Center of Immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA 98101, USA
| | - Shannon Kreuser
- Center of Immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA 98101, USA
| | - Noelle Dahl
- Center of Immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA 98101, USA
| | - Yuchi Honaker
- Center of Immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA 98101, USA
| | - Christina Lopez
- Center of Immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA 98101, USA
| | - David J Rawlings
- Center of Immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98195, USA; Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | - Richard G James
- Center of Immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA 98101, USA; Molecular Engineering and Science Institute, University of Washington, Seattle, WA 98195, USA; Department of Pharmacology, University of Washington, Seattle, WA 98195, USA; Brotman-Baty Institute for Precision Medicine, Seattle, WA 98195, USA.
| |
Collapse
|
11
|
Hill TF, Narvekar P, Asher GD, Edelstein JN, Camp ND, Grimm A, Thomas KR, Leiken MD, Molloy KM, Cook PJ, Arlauckas SP, Morgan RA, Tasian SK, Rawlings DJ, James RG. Human plasma cells engineered to secrete bispecifics drive effective in vivo leukemia killing. Mol Ther 2024; 32:2676-2691. [PMID: 38959896 PMCID: PMC11405176 DOI: 10.1016/j.ymthe.2024.06.004] [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: 01/23/2024] [Revised: 05/09/2024] [Accepted: 06/04/2024] [Indexed: 07/05/2024] Open
Abstract
Bispecific antibodies are an important tool for the management and treatment of acute leukemias. As a next step toward clinical translation of engineered plasma cells, we describe approaches for secretion of bispecific antibodies by human plasma cells. We show that human plasma cells expressing either fragment crystallizable domain-deficient anti-CD19 × anti-CD3 (blinatumomab) or anti-CD33 × anti-CD3 bispecific antibodies mediate T cell activation and direct T cell killing of B acute lymphoblastic leukemia or acute myeloid leukemia cell lines in vitro. We demonstrate that knockout of the self-expressed antigen, CD19, boosts anti-CD19-bispecific secretion by plasma cells and prevents self-targeting. Plasma cells secreting anti-CD19-bispecific antibodies elicited in vivo control of acute lymphoblastic leukemia patient-derived xenografts in immunodeficient mice co-engrafted with autologous T cells. In these studies, we found that leukemic control elicited by engineered plasma cells was similar to CD19-targeted chimeric antigen receptor-expressing T cells. Finally, the steady-state concentration of anti-CD19 bispecifics in serum 1 month after cell delivery and tumor eradication was comparable with that observed in patients treated with a steady-state infusion of blinatumomab. These findings support further development of ePCs for use as a durable delivery system for the treatment of acute leukemias, and potentially other cancers.
Collapse
Affiliation(s)
- Tyler F Hill
- University of Washington, Medical Scientist Training Program, Seattle, WA, USA; Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | - Parnal Narvekar
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | - Gregory D Asher
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | | | - Nathan D Camp
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | - Annaiz Grimm
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | - Kerri R Thomas
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | | | | | - Peter J Cook
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA
| | | | | | - Sarah K Tasian
- Children's Hospital of Philadelphia, Division of Oncology and Center for Childhood Cancer Research, Philadelphia, PA, USA; Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David J Rawlings
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA; University of Washington, Departments of Pediatrics and Immunology, Seattle, WA, USA
| | - Richard G James
- Seattle Children's Research Institute, Center for Immunity and Immunotherapy, Seattle, WA, USA; University of Washington, Departments of Pediatrics and Pharmacology, Seattle, WA, USA.
| |
Collapse
|
12
|
Kim W. Germinal Center Response to mRNA Vaccination and Impact of Immunological Imprinting on Subsequent Vaccination. Immune Netw 2024; 24:e28. [PMID: 39246619 PMCID: PMC11377948 DOI: 10.4110/in.2024.24.e28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 04/04/2024] [Accepted: 04/29/2024] [Indexed: 09/10/2024] Open
Abstract
Vaccines are the most effective intervention currently available, offering protective immunity against targeted pathogens. The emergence of the coronavirus disease 2019 pandemic has prompted rapid development and deployment of lipid nanoparticle encapsulated, mRNA-based vaccines. While these vaccines have demonstrated remarkable immunogenicity, concerns persist regarding their ability to confer durable protective immunity to continuously evolving severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants. This review focuses on human B cell responses induced by SARS-CoV-2 mRNA vaccination, with particular emphasis on the crucial role of germinal center reactions in shaping enduring protective immunity. Additionally, we explored observations of immunological imprinting and dynamics of recalled pre-existing immunity following variants of concern-based booster vaccination. Insights from this review contribute to comprehensive understanding B cell responses to mRNA vaccination in humans, thereby refining vaccination strategies for optimal and sustained protection against evolving coronavirus variants.
Collapse
Affiliation(s)
- Wooseob Kim
- Department of Microbiology, Korea University College of Medicine, Seoul 02841, Korea
- Vaccine Innovation Center, Korea University College of Medicine, Seoul 02841, Korea
| |
Collapse
|
13
|
Fooksman DR, Jing Z, Park R. New insights into the ontogeny, diversity, maturation and survival of long-lived plasma cells. Nat Rev Immunol 2024; 24:461-470. [PMID: 38332373 DOI: 10.1038/s41577-024-00991-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 02/10/2024]
Abstract
Plasma cells are unique immune effectors, capable of producing large amounts of high-affinity antibodies that protect against pathogenic infections. Although most plasma cells have short lifespans, certain conditions or vaccinations can give rise to long-lived plasma cells (LLPCs) that provide individuals with lifelong protection against pathogen exposure. The nature of these LLPCs is poorly understood; however, recent studies have shed new light on the ontogeny, diversity, maturation and survival of these unique cells. Whereas LLPCs had been thought to arise preferentially from germinal centres, novel genetic tools have revealed that they can originate from various stages throughout the humoral response. Furthermore, new single-cell analyses have shown that mouse and human plasma cells are heterogeneous and may undergo further maturation in situ in the bone marrow niche. Finally, plasma cells were previously considered to be sessile cells maintained in fixed survival niches, but new data show that plasma cell subsets can differentially migrate and organize into clusters that may be associated with survival niches. These descriptive findings provide new insights into how cell-intrinsic programmes and extrinsic factors may regulate the longevity of plasma cells in various contexts, which suggest new research avenues for their functional validation.
Collapse
Affiliation(s)
- David R Fooksman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA.
| | - Zhixin Jing
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, MD, USA
| | - Rosa Park
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
14
|
Ganusov VV. Appropriate Sampling and Longer Follow-Up Are Required to Rigorously Evaluate Longevity of Humoral Memory After Vaccination. Immunohorizons 2024; 8:397-403. [PMID: 38864816 PMCID: PMC11220738 DOI: 10.4049/immunohorizons.2300057] [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: 07/18/2023] [Accepted: 04/25/2024] [Indexed: 06/13/2024] Open
Abstract
One of the goals of vaccination is to induce long-lived immunity against the infection and/or disease. Many studies have followed the generation of humoral immunity to SARS-CoV-2 after vaccination; however, such studies typically varied by the duration of the follow-up and the number of time points at which immune response measurements were done. How these parameters (the number of time points and the overall duration of the follow-up) impact estimates of immunity longevity remain largely unknown. Several studies, including one by Arunachalam et al. (2023. J. Clin. Invest. 133: e167955), evaluated the humoral immune response in individuals receiving either a third or fourth dose of mRNA COVID-19 vaccine; by measuring Ab levels at three time points (prior to vaccination and at 1 and 6 mo), Arunachalam et al. found similar half-life times for serum Abs in the two groups and thus suggested that additional boosting is unnecessary to prolong immunity to SARS-CoV-2. I demonstrate that measuring Ab levels at these three time points and only for 6 mo does not allow one to accurately evaluate the long-term half-life of vaccine-induced Abs. By using the data from a cohort of blood donors followed for several years, I show that after revaccination with vaccinia virus, vaccinia virus-specific Abs decay biphasically, and even the late decay rate exceeds the true slow loss rate of humoral memory observed years prior to the boosting. Mathematical models of Ab response kinetics, parameterized using preliminary data, should be used for power analysis to determine the most appropriate timing and duration of sampling to rigorously determine the duration of humoral immunity after vaccination.
Collapse
Affiliation(s)
- Vitaly V. Ganusov
- Address correspondence and reprint request to: Vitaly V. Ganusov, Texas Biomedical Research Institute, 8715 W. Military Dr., San Antonio, TX 78227-5302. E-mail address:
| |
Collapse
|
15
|
Lee Z, Wan J, Shen A, Barnard G. Gene copy number, gene configuration and LC/HC mRNA ratio impact on antibody productivity and product quality in targeted integration CHO cell lines. Biotechnol Prog 2024; 40:e3433. [PMID: 38321634 DOI: 10.1002/btpr.3433] [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: 10/13/2023] [Revised: 12/01/2023] [Accepted: 01/03/2024] [Indexed: 02/08/2024]
Abstract
The augmentation of transgene copy numbers is a prevalent approach presumed to enhance transcriptional activity and product yield. CHO cell lines engineered via targeted integration (TI) offer an advantageous platform for investigating the interplay between gene copy number, mRNA abundance, product yield, and product quality. Our investigation revealed that incrementally elevating the gene copy numbers of both IgG heavy chain (HC) and light chain (LC) concurrently resulted in the attainment of plateaus in mRNA levels and product titers, notably occurring beyond four to five gene copies integrated at the same TI site. Furthermore, maintaining a fixed gene copy number while varying the position of genes within the vector influenced the LC/HC mRNA ratio, which subsequently exerted a substantial impact on product titer. Moreover, manipulation of the LC/HC gene ratio through the introduction of surplus LC gene copies led to heightened LC mRNA expression and a reduction in the levels of high molecular weight species. It is noteworthy that the effects of excess LC on product titer were dependent on the specific molecule under consideration. The strategic utilization of PCR tags enabled precise quantification of transcription from each expression slot within the vector, facilitating the identification of highly expressive and less expressive slots. Collectively, these findings significantly enhance our understanding of stable antibody production in TI CHO cell lines.
Collapse
Affiliation(s)
- Zion Lee
- Department of Cell Culture and Bioprocess Operations, Genentech, Inc., San Francisco, California, USA
| | - Jun Wan
- Department of Cell Culture and Bioprocess Operations, Genentech, Inc., San Francisco, California, USA
| | - Amy Shen
- Department of Cell Culture and Bioprocess Operations, Genentech, Inc., San Francisco, California, USA
| | - Gavin Barnard
- Department of Cell Culture and Bioprocess Operations, Genentech, Inc., San Francisco, California, USA
| |
Collapse
|
16
|
Taubmann J, Müller F, Yalcin Mutlu M, Völkl S, Aigner M, Bozec A, Mackensen A, Grieshaber-Bouyer R, Schett G. CD19 Chimeric Antigen Receptor T Cell Treatment: Unraveling the Role of B Cells in Systemic Lupus Erythematosus. Arthritis Rheumatol 2024; 76:497-504. [PMID: 38114423 DOI: 10.1002/art.42784] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/09/2023] [Accepted: 12/14/2023] [Indexed: 12/21/2023]
Abstract
B cell generation of autoantibodies is a crucial step in the pathogenesis of systemic lupus erythematosus (SLE). After their differentiation in the bone marrow, B cells populate the secondary lymphatic organs, where they undergo further maturation leading to the development of memory B cells as well as antibody-producing plasmablasts and plasma cells. Targeting B cells is an important strategy to treat autoimmune diseases such as SLE, in which B cell tolerance is disturbed and autoimmune B cells and autoantibodies emerge. This review discusses the functional aspects of antibody- and cell-based B cell-depleting therapy in SLE. It thereby particularly focuses on lessons learned from chimeric antigen receptor (CAR) T cell treatment on the role of B cells in SLE for understanding B cell pathology in SLE. CAR T cells model a deep B cell depletion and thereby allow understanding the role of aberrant B cell activation in the pathogenesis of SLE. Furthermore, the effects of B cell depletion on autoantibody production can be better described, ie, explaining the concept of different cellular sources of (auto-) antibodies in the form of short-lived plasmablasts and long-lived plasma cells, which differ in their susceptibility to B cell depletion and require different targeted therapeutic approaches. Finally, the safety of deep B cell depletion in autoimmune disease is discussed.
Collapse
Affiliation(s)
- Jule Taubmann
- Friedrich-Alexander-Universität Erlangen Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Fabian Müller
- Friedrich-Alexander-Universität Erlangen Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Melek Yalcin Mutlu
- Friedrich-Alexander-Universität Erlangen Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Simon Völkl
- Friedrich-Alexander-Universität Erlangen Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Michael Aigner
- Friedrich-Alexander-Universität Erlangen Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Aline Bozec
- Friedrich-Alexander-Universität Erlangen Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Andreas Mackensen
- Friedrich-Alexander-Universität Erlangen Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Ricardo Grieshaber-Bouyer
- Friedrich-Alexander-Universität Erlangen Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Friedrich-Alexander-Universität Erlangen Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
17
|
Simons BD, Karin O. Tuning of plasma cell lifespan by competition explains the longevity and heterogeneity of antibody persistence. Immunity 2024; 57:600-611.e6. [PMID: 38447570 DOI: 10.1016/j.immuni.2024.02.005] [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: 07/07/2023] [Revised: 01/12/2024] [Accepted: 02/07/2024] [Indexed: 03/08/2024]
Abstract
Plasma cells that emerge after infection or vaccination exhibit heterogeneous lifespans; most survive for days to months, whereas others persist for decades, providing antigen-specific long-term protection. We developed a mathematical framework to explore the dynamics of plasma cell removal and its regulation by survival factors. Analyses of antibody persistence following hepatitis A and B and HPV vaccination revealed specific patterns of longevity and heterogeneity within and between responses, implying that this process is fine-tuned near a critical "flat" state between two dynamic regimes. This critical state reflects the tuning of rates of the underlying regulatory network and is highly sensitive to variation in parameters, which amplifies lifespan differences between cells. We propose that fine-tuning is the generic outcome of competition over shared survival signals, with a competition-based mechanism providing a unifying explanation for a wide range of experimental observations, including the dynamics of plasma cell accumulation and the effects of survival factor deletion. Our theory is testable, and we provide specific predictions.
Collapse
Affiliation(s)
- Benjamin D Simons
- Department of Applied Mathematics and Theoretical Physics, Centre for Mathematical Sciences, University of Cambridge, Cambridge CB3 0WA, UK; Wellcome Trust, Cancer Research UK Gurdon Institute, University of Cambridge, Cambridge CB2 1QN, UK; Wellcome Trust-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
| | - Omer Karin
- Department of Mathematics, Imperial College London, London SW7 2AZ, UK.
| |
Collapse
|
18
|
Nguyen DC, Saney C, Hentenaar IT, Cabrera-Mora M, Capric V, Woodruff MC, Andrews J, Lonial S, Sanz I, Lee FEH. Majority of human circulating IgG plasmablasts stop blasting in a cell-free pro-survival culture. Sci Rep 2024; 14:3616. [PMID: 38350990 PMCID: PMC10864258 DOI: 10.1038/s41598-024-53977-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 02/07/2024] [Indexed: 02/16/2024] Open
Abstract
Following infection or vaccination, early-minted antibody secreting cells (ASC) or plasmablasts appear in circulation transiently, and a small fraction migrates to the spleen or bone marrow (BM) to mature into long-lived plasma cells (LLPC). While LLPC, by definition, are quiescent or non-dividing, the majority of blood ASC are thought to be "blasting" or proliferative. In this study, we find > 95% nascent blood ASC in culture express Ki-67 but only 6-12% incorporate BrdU after 4 h or 24 h labeling. In contrast, < 5% BM LLPC in culture are Ki-67+ with no BrdU uptake. Due to limitations of traditional flow cytometry, we utilized a novel optofluidic technology to evaluate cell division with simultaneous functional IgG secretion. We find 11% early-minted blood ASC undergo division, and none of the terminally differentiated BM LLPC (CD19-CD38hiCD138+) divide during the 7-21 days in culture. While BM LLPC undergo complete cell cycle arrest, the process of differentiation into an ASC or plasmablasts also discourages entry into S phase. Since the majority of Ki-67+ nascent blood ASC have exited cell cycle and are no longer actively "blasting", the term "plasmablast", which traditionally refers to an ASC that still has the capacity to divide, may probably be a misnomer.
Collapse
Affiliation(s)
- Doan C Nguyen
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Celia Saney
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
- Center for Vaccines and Immunology, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Ian T Hentenaar
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Monica Cabrera-Mora
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Violeta Capric
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Matthew C Woodruff
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | - Joel Andrews
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Sagar Lonial
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Ignacio Sanz
- Division of Rheumatology, Department of Medicine, Emory University, Atlanta, GA, USA
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA
| | - F Eun-Hyung Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Emory University, Atlanta, GA, USA.
- Lowance Center for Human Immunology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
19
|
Matsuoka T, Araki M, Lin Y, Okamoto T, Gold R, Chihara N, Sato W, Kimura A, Tachimori H, Miyamoto K, Kusunoki S, Yamamura T. Long-term Effects of IL-6 Receptor Blockade Therapy on Regulatory Lymphocytes and Neutrophils in Neuromyelitis Optica Spectrum Disorder. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200173. [PMID: 37863660 PMCID: PMC10691226 DOI: 10.1212/nxi.0000000000200173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 08/29/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND AND OBJECTIVES Neuromyelitis optica spectrum disorder (NMOSD) is a disabling autoimmune neurologic disease. Anti-IL-6 receptor (IL-6R) therapy prevents relapses in patients with anti-aquaporin 4 (AQP4)-IgG-positive NMOSD; however, it remains unclear how cellular immune components are altered by anti-IL-6R therapy. In this study, we examined the long-term effects of the anti-IL-6R monoclonal antibody tocilizumab (TCZ) on immune cell profiles in patients with NMOSD. METHODS Monthly IV injections of TCZ (8 mg/kg) were administered as an add-on therapy to 19 anti-AQP4-IgG-positive patients, who had been refractory to corticosteroids and immunosuppressive drugs. Peripheral blood was collected before infusion of TCZ for flow cytometry analysis of lymphocyte subsets. Seven patients provided whole blood samples for gene expression profiles. RESULTS Patients with NMOSD had reduced numbers of lymphocyte subsets with regulatory functions, including transitional B cells, CD56high NK cells, and CD45RA-FoxP3high regulatory T cells. However, after initiating TCZ therapy, the numbers increased to normal levels within 1 year. Gene expression analysis revealed that neutrophil granule-related genes, predominated by those related to azurophil granules, were significantly upregulated in patients with NMOSD. Such alterations suggestive of accelerated myeloid turnover were not observed 1 year after TCZ therapy, and the effects of TCZ on some neutrophil genes were observed as early as 5 days after starting TCZ. In vitro analysis demonstrated that naïve T-cell division was impaired in the enrolled patients, which was fully recovered after 18 months of therapy. DISCUSSION In patients with active NMOSD not treated with molecular targeting drugs, we observed reduction or deficiency in lymphocytes with regulatory potentials and activation of neutrophils. However, introduction of anti-IL-6R therapy accompanied by tapering concomitant drugs corrected such abnormalities, which might contribute to persistent relapse prevention. The recovery in the naïve T-cell division after starting TCZ may underlie the relatively low risk of infection in patients under anti-IL-6R therapy. TRIAL REGISTRATION INFORMATION University Hospital Medical Information Network Clinical Trials Registry: UMIN000005889 (July 8, 2011) and UMIN000007866 (May 1, 2012) (umin.ac.jp/ctr/index.htm). The first participant was enrolled on November 2, 2011.
Collapse
Affiliation(s)
- Takako Matsuoka
- From the Department of Immunology (T.M., W.S., A.K., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Department of Pediatrics (T.M.), Graduate School of Medicine, The University of Tokyo, Bunkyo; Multiple Sclerosis Center (M.A., Y.L., T.O., W.S., T.Y.), National Center of Neurology and Psychiatry, Kodaira; Department of Neurology (M.A.), Kawakita General Hospital, Suginami; Department of Neurology (Y.L., T.O.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (R.G.), Ruhr University, Bochum, Germany; Division of Neurology (N.C.), Kobe University Graduate School of Medicine; Department of Clinical Epidemiology (H.T.), Translational Medical Center, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira; Bureau of International Health Cooperation (H.T.), National Center for Global Health and Medicine, Shinjuku, Tokyo; Department of Neurology (K.M., S.K.), Kindai University Faculty of Medicine, Osakasayama, Osaka; and Department of Neurology (K.M.), Wakayama Medical University, Japan
| | - Manabu Araki
- From the Department of Immunology (T.M., W.S., A.K., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Department of Pediatrics (T.M.), Graduate School of Medicine, The University of Tokyo, Bunkyo; Multiple Sclerosis Center (M.A., Y.L., T.O., W.S., T.Y.), National Center of Neurology and Psychiatry, Kodaira; Department of Neurology (M.A.), Kawakita General Hospital, Suginami; Department of Neurology (Y.L., T.O.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (R.G.), Ruhr University, Bochum, Germany; Division of Neurology (N.C.), Kobe University Graduate School of Medicine; Department of Clinical Epidemiology (H.T.), Translational Medical Center, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira; Bureau of International Health Cooperation (H.T.), National Center for Global Health and Medicine, Shinjuku, Tokyo; Department of Neurology (K.M., S.K.), Kindai University Faculty of Medicine, Osakasayama, Osaka; and Department of Neurology (K.M.), Wakayama Medical University, Japan
| | - Youwei Lin
- From the Department of Immunology (T.M., W.S., A.K., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Department of Pediatrics (T.M.), Graduate School of Medicine, The University of Tokyo, Bunkyo; Multiple Sclerosis Center (M.A., Y.L., T.O., W.S., T.Y.), National Center of Neurology and Psychiatry, Kodaira; Department of Neurology (M.A.), Kawakita General Hospital, Suginami; Department of Neurology (Y.L., T.O.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (R.G.), Ruhr University, Bochum, Germany; Division of Neurology (N.C.), Kobe University Graduate School of Medicine; Department of Clinical Epidemiology (H.T.), Translational Medical Center, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira; Bureau of International Health Cooperation (H.T.), National Center for Global Health and Medicine, Shinjuku, Tokyo; Department of Neurology (K.M., S.K.), Kindai University Faculty of Medicine, Osakasayama, Osaka; and Department of Neurology (K.M.), Wakayama Medical University, Japan
| | - Tomoko Okamoto
- From the Department of Immunology (T.M., W.S., A.K., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Department of Pediatrics (T.M.), Graduate School of Medicine, The University of Tokyo, Bunkyo; Multiple Sclerosis Center (M.A., Y.L., T.O., W.S., T.Y.), National Center of Neurology and Psychiatry, Kodaira; Department of Neurology (M.A.), Kawakita General Hospital, Suginami; Department of Neurology (Y.L., T.O.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (R.G.), Ruhr University, Bochum, Germany; Division of Neurology (N.C.), Kobe University Graduate School of Medicine; Department of Clinical Epidemiology (H.T.), Translational Medical Center, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira; Bureau of International Health Cooperation (H.T.), National Center for Global Health and Medicine, Shinjuku, Tokyo; Department of Neurology (K.M., S.K.), Kindai University Faculty of Medicine, Osakasayama, Osaka; and Department of Neurology (K.M.), Wakayama Medical University, Japan
| | - Ralf Gold
- From the Department of Immunology (T.M., W.S., A.K., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Department of Pediatrics (T.M.), Graduate School of Medicine, The University of Tokyo, Bunkyo; Multiple Sclerosis Center (M.A., Y.L., T.O., W.S., T.Y.), National Center of Neurology and Psychiatry, Kodaira; Department of Neurology (M.A.), Kawakita General Hospital, Suginami; Department of Neurology (Y.L., T.O.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (R.G.), Ruhr University, Bochum, Germany; Division of Neurology (N.C.), Kobe University Graduate School of Medicine; Department of Clinical Epidemiology (H.T.), Translational Medical Center, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira; Bureau of International Health Cooperation (H.T.), National Center for Global Health and Medicine, Shinjuku, Tokyo; Department of Neurology (K.M., S.K.), Kindai University Faculty of Medicine, Osakasayama, Osaka; and Department of Neurology (K.M.), Wakayama Medical University, Japan
| | - Norio Chihara
- From the Department of Immunology (T.M., W.S., A.K., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Department of Pediatrics (T.M.), Graduate School of Medicine, The University of Tokyo, Bunkyo; Multiple Sclerosis Center (M.A., Y.L., T.O., W.S., T.Y.), National Center of Neurology and Psychiatry, Kodaira; Department of Neurology (M.A.), Kawakita General Hospital, Suginami; Department of Neurology (Y.L., T.O.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (R.G.), Ruhr University, Bochum, Germany; Division of Neurology (N.C.), Kobe University Graduate School of Medicine; Department of Clinical Epidemiology (H.T.), Translational Medical Center, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira; Bureau of International Health Cooperation (H.T.), National Center for Global Health and Medicine, Shinjuku, Tokyo; Department of Neurology (K.M., S.K.), Kindai University Faculty of Medicine, Osakasayama, Osaka; and Department of Neurology (K.M.), Wakayama Medical University, Japan
| | - Wakiro Sato
- From the Department of Immunology (T.M., W.S., A.K., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Department of Pediatrics (T.M.), Graduate School of Medicine, The University of Tokyo, Bunkyo; Multiple Sclerosis Center (M.A., Y.L., T.O., W.S., T.Y.), National Center of Neurology and Psychiatry, Kodaira; Department of Neurology (M.A.), Kawakita General Hospital, Suginami; Department of Neurology (Y.L., T.O.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (R.G.), Ruhr University, Bochum, Germany; Division of Neurology (N.C.), Kobe University Graduate School of Medicine; Department of Clinical Epidemiology (H.T.), Translational Medical Center, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira; Bureau of International Health Cooperation (H.T.), National Center for Global Health and Medicine, Shinjuku, Tokyo; Department of Neurology (K.M., S.K.), Kindai University Faculty of Medicine, Osakasayama, Osaka; and Department of Neurology (K.M.), Wakayama Medical University, Japan
| | - Atsuko Kimura
- From the Department of Immunology (T.M., W.S., A.K., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Department of Pediatrics (T.M.), Graduate School of Medicine, The University of Tokyo, Bunkyo; Multiple Sclerosis Center (M.A., Y.L., T.O., W.S., T.Y.), National Center of Neurology and Psychiatry, Kodaira; Department of Neurology (M.A.), Kawakita General Hospital, Suginami; Department of Neurology (Y.L., T.O.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (R.G.), Ruhr University, Bochum, Germany; Division of Neurology (N.C.), Kobe University Graduate School of Medicine; Department of Clinical Epidemiology (H.T.), Translational Medical Center, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira; Bureau of International Health Cooperation (H.T.), National Center for Global Health and Medicine, Shinjuku, Tokyo; Department of Neurology (K.M., S.K.), Kindai University Faculty of Medicine, Osakasayama, Osaka; and Department of Neurology (K.M.), Wakayama Medical University, Japan
| | - Hisateru Tachimori
- From the Department of Immunology (T.M., W.S., A.K., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Department of Pediatrics (T.M.), Graduate School of Medicine, The University of Tokyo, Bunkyo; Multiple Sclerosis Center (M.A., Y.L., T.O., W.S., T.Y.), National Center of Neurology and Psychiatry, Kodaira; Department of Neurology (M.A.), Kawakita General Hospital, Suginami; Department of Neurology (Y.L., T.O.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (R.G.), Ruhr University, Bochum, Germany; Division of Neurology (N.C.), Kobe University Graduate School of Medicine; Department of Clinical Epidemiology (H.T.), Translational Medical Center, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira; Bureau of International Health Cooperation (H.T.), National Center for Global Health and Medicine, Shinjuku, Tokyo; Department of Neurology (K.M., S.K.), Kindai University Faculty of Medicine, Osakasayama, Osaka; and Department of Neurology (K.M.), Wakayama Medical University, Japan
| | - Katsuichi Miyamoto
- From the Department of Immunology (T.M., W.S., A.K., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Department of Pediatrics (T.M.), Graduate School of Medicine, The University of Tokyo, Bunkyo; Multiple Sclerosis Center (M.A., Y.L., T.O., W.S., T.Y.), National Center of Neurology and Psychiatry, Kodaira; Department of Neurology (M.A.), Kawakita General Hospital, Suginami; Department of Neurology (Y.L., T.O.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (R.G.), Ruhr University, Bochum, Germany; Division of Neurology (N.C.), Kobe University Graduate School of Medicine; Department of Clinical Epidemiology (H.T.), Translational Medical Center, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira; Bureau of International Health Cooperation (H.T.), National Center for Global Health and Medicine, Shinjuku, Tokyo; Department of Neurology (K.M., S.K.), Kindai University Faculty of Medicine, Osakasayama, Osaka; and Department of Neurology (K.M.), Wakayama Medical University, Japan
| | - Susumu Kusunoki
- From the Department of Immunology (T.M., W.S., A.K., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Department of Pediatrics (T.M.), Graduate School of Medicine, The University of Tokyo, Bunkyo; Multiple Sclerosis Center (M.A., Y.L., T.O., W.S., T.Y.), National Center of Neurology and Psychiatry, Kodaira; Department of Neurology (M.A.), Kawakita General Hospital, Suginami; Department of Neurology (Y.L., T.O.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (R.G.), Ruhr University, Bochum, Germany; Division of Neurology (N.C.), Kobe University Graduate School of Medicine; Department of Clinical Epidemiology (H.T.), Translational Medical Center, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira; Bureau of International Health Cooperation (H.T.), National Center for Global Health and Medicine, Shinjuku, Tokyo; Department of Neurology (K.M., S.K.), Kindai University Faculty of Medicine, Osakasayama, Osaka; and Department of Neurology (K.M.), Wakayama Medical University, Japan
| | - Takashi Yamamura
- From the Department of Immunology (T.M., W.S., A.K., T.Y.), National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira; Department of Pediatrics (T.M.), Graduate School of Medicine, The University of Tokyo, Bunkyo; Multiple Sclerosis Center (M.A., Y.L., T.O., W.S., T.Y.), National Center of Neurology and Psychiatry, Kodaira; Department of Neurology (M.A.), Kawakita General Hospital, Suginami; Department of Neurology (Y.L., T.O.), National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Neurology (R.G.), Ruhr University, Bochum, Germany; Division of Neurology (N.C.), Kobe University Graduate School of Medicine; Department of Clinical Epidemiology (H.T.), Translational Medical Center, National Institute of Mental Health, National Center of Neurology and Psychiatry, Kodaira; Bureau of International Health Cooperation (H.T.), National Center for Global Health and Medicine, Shinjuku, Tokyo; Department of Neurology (K.M., S.K.), Kindai University Faculty of Medicine, Osakasayama, Osaka; and Department of Neurology (K.M.), Wakayama Medical University, Japan.
| |
Collapse
|
20
|
Gao X, Wang X, Li S, Saif Ur Rahman M, Xu S, Liu Y. Nanovaccines for Advancing Long-Lasting Immunity against Infectious Diseases. ACS NANO 2023; 17:24514-24538. [PMID: 38055649 DOI: 10.1021/acsnano.3c07741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/08/2023]
Abstract
Infectious diseases, particularly life-threatening pathogens such as small pox and influenza, have substantial implications on public health and global economies. Vaccination is a key approach to combat existing and emerging pathogens. Immunological memory is an essential characteristic used to evaluate vaccine efficacy and durability and the basis for the long-term effects of vaccines in protecting against future infections; however, optimizing the potency, improving the quality, and enhancing the durability of immune responses remains challenging and a focus for research involving investigation of nanovaccine technologies. In this review, we describe how nanovaccines can address the challenges for conventional vaccines in stimulating adaptive immune memory responses to protect against reinfection. We discuss protein and nonprotein nanoparticles as useful antigen platforms, including those with highly ordered and repetitive antigen array presentation to enhance immunogenicity through cross-linking with multiple B cell receptors, and with a focus on antigen properties. In addition, we describe how nanoadjuvants can improve immune responses by providing enhanced access to lymph nodes, lymphnode targeting, germinal center retention, and long-lasting immune response generation. Nanotechnology has the advantage to facilitate vaccine induction of long-lasting immunity against infectious diseases, now and in the future.
Collapse
Affiliation(s)
- Xinglong Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Xinlian Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Shilin Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
- University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | | | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen 518060, P.R. China
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, P.R. China
| |
Collapse
|
21
|
Alexandre M, Prague M, McLean C, Bockstal V, Douoguih M, Thiébaut R. Prediction of long-term humoral response induced by the two-dose heterologous Ad26.ZEBOV, MVA-BN-Filo vaccine against Ebola. NPJ Vaccines 2023; 8:174. [PMID: 37940656 PMCID: PMC10632397 DOI: 10.1038/s41541-023-00767-y] [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: 03/23/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023] Open
Abstract
The persistence of the long-term immune response induced by the heterologous Ad26.ZEBOV, MVA-BN-Filo two-dose vaccination regimen against Ebola has been investigated in several clinical trials. Longitudinal data on IgG-binding antibody concentrations were analyzed from 487 participants enrolled in six Phase I and Phase II clinical trials conducted by the EBOVAC1 and EBOVAC2 consortia. A model based on ordinary differential equations describing the dynamics of antibodies and short- and long-lived antibody-secreting cells (ASCs) was used to model the humoral response from 7 days after the second vaccination to a follow-up period of 2 years. Using a population-based approach, we first assessed the robustness of the model, which was originally estimated based on Phase I data, against all data. Then we assessed the longevity of the humoral response and identified factors that influence these dynamics. We estimated a half-life of the long-lived ASC of at least 15 years and found an influence of geographic region, sex, and age on the humoral response dynamics, with longer antibody persistence in Europeans and women and higher production of antibodies in younger participants.
Collapse
Affiliation(s)
- Marie Alexandre
- Department of Public Health, Bordeaux University, Inserm UMR 1219 Bordeaux Population Health Research Center, Inria SISTM, Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | - Mélanie Prague
- Department of Public Health, Bordeaux University, Inserm UMR 1219 Bordeaux Population Health Research Center, Inria SISTM, Bordeaux, France
- Vaccine Research Institute, Créteil, France
| | - Chelsea McLean
- Janssen Vaccines and Prevention, Leiden, the Netherlands
| | - Viki Bockstal
- Janssen Vaccines and Prevention, Leiden, the Netherlands
- ExeVir, Ghent, Belgium
| | | | - Rodolphe Thiébaut
- Department of Public Health, Bordeaux University, Inserm UMR 1219 Bordeaux Population Health Research Center, Inria SISTM, Bordeaux, France.
- Vaccine Research Institute, Créteil, France.
| |
Collapse
|
22
|
He R, Zheng X, Zhang J, Liu B, Wang Q, Wu Q, Liu Z, Chang F, Hu Y, Xie T, Liu Y, Chen J, Yang J, Teng S, Lu R, Pan D, Wang Y, Peng L, Huang W, Terzieva V, Liu W, Wang Y, Li YP, Qu X. SARS-CoV-2 spike-specific T FH cells exhibit unique responses in infected and vaccinated individuals. Signal Transduct Target Ther 2023; 8:393. [PMID: 37802996 PMCID: PMC10558553 DOI: 10.1038/s41392-023-01650-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 10/08/2023] Open
Abstract
Long-term humoral immunity to SARS-CoV-2 is essential for preventing reinfection. The production of neutralizing antibody (nAb) and B cell differentiation are tightly regulated by T follicular help (TFH) cells. However, the longevity and functional role of TFH cell subsets in COVID-19 convalescents and vaccine recipients remain poorly defined. Here, we show that SARS-CoV-2 infection and inactivated vaccine elicited both spike-specific CXCR3+ TFH cell and CXCR3- TFH cell responses, which showed distinct response patterns. Spike-specific CXCR3+ TFH cells exhibit a dominant and more durable response than CXCR3- TFH cells that positively correlated with antibody responses. A third booster dose preferentially expands the spike-specific CXCR3+ TFH cell subset induced by two doses of inactivated vaccine, contributing to antibody maturation and potency. Functionally, spike-specific CXCR3+ TFH cells have a greater ability to induce spike-specific antibody secreting cells (ASCs) differentiation compared to spike-specific CXCR3- TFH cells. In conclusion, the persistent and functional role of spike-specific CXCR3+ TFH cells following SARS-CoV-2 infection and vaccination may play an important role in antibody maintenance and recall response, thereby conferring long-term protection. The findings from this study will inform the development of SARS-CoV-2 vaccines aiming to induce long-term protective immune memory.
Collapse
Affiliation(s)
- Rongzhang He
- College of Basic Medical Sciences, Hengyang Medical School, University of South China & MOE Key Lab of Rare Pediatric Diseases, 421001, Hengyang, China
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, 423000, Chenzhou, China
| | - Xingyu Zheng
- College of Basic Medical Sciences, Hengyang Medical School, University of South China & MOE Key Lab of Rare Pediatric Diseases, 421001, Hengyang, China
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, 423000, Chenzhou, China
| | - Jian Zhang
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, 423000, Chenzhou, China
| | - Bo Liu
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, 423000, Chenzhou, China
| | - Qijie Wang
- The Central Hospital of Shaoyang, 422000, Shaoyang, China
| | - Qian Wu
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, 423000, Chenzhou, China
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, 501180, Guangzhou, China
| | - Ziyan Liu
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, 423000, Chenzhou, China
| | - Fangfang Chang
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, 501180, Guangzhou, China
| | - Yabin Hu
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, 423000, Chenzhou, China
| | - Ting Xie
- The Central Hospital of Shaoyang, 422000, Shaoyang, China
| | - Yongchen Liu
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, 501180, Guangzhou, China
| | - Jun Chen
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, 423000, Chenzhou, China
| | - Jing Yang
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, 423000, Chenzhou, China
| | - Shishan Teng
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, 423000, Chenzhou, China
| | - Rui Lu
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, 423000, Chenzhou, China
| | - Dong Pan
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, 423000, Chenzhou, China
| | - You Wang
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, 423000, Chenzhou, China
- School of Public Health, University of South China, 421001, Hengyang, China
| | - Liting Peng
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, 423000, Chenzhou, China
| | - Weijin Huang
- National Institutes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Key Laboratory of Biological Product Quality Research and Evaluation of National Medical Products Administration, 102629, Beijing, China
| | - Velislava Terzieva
- Laboratory of OMICs Technologies, Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, 1113, Bulgaria
| | - Wenpei Liu
- College of Basic Medical Sciences, Hengyang Medical School, University of South China & MOE Key Lab of Rare Pediatric Diseases, 421001, Hengyang, China
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, 423000, Chenzhou, China
| | - Youchun Wang
- National Institutes for Food and Drug Control, Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, Key Laboratory of Biological Product Quality Research and Evaluation of National Medical Products Administration, 102629, Beijing, China.
| | - Yi-Ping Li
- Institute of Human Virology, Zhongshan School of Medicine, and Key Laboratory of Tropical Disease Control of Ministry of Education, Sun Yat-sen University, 501180, Guangzhou, China.
| | - Xiaowang Qu
- College of Basic Medical Sciences, Hengyang Medical School, University of South China & MOE Key Lab of Rare Pediatric Diseases, 421001, Hengyang, China.
- Translational Medicine Institute, The First People's Hospital of Chenzhou, Hengyang Medical School, University of South China, 423000, Chenzhou, China.
| |
Collapse
|
23
|
Dari A, Jacqmin P, Iwaki Y, Neyens M, Le Gars M, Sadoff J, Hardt K, Ruiz‐Guiñazú J, Pérez‐Ruixo JJ. Mechanistic modeling projections of antibody persistence after homologous booster regimens of COVID-19 vaccine Ad26.COV2.S in humans. CPT Pharmacometrics Syst Pharmacol 2023; 12:1485-1498. [PMID: 37715342 PMCID: PMC10583247 DOI: 10.1002/psp4.13025] [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: 06/21/2023] [Accepted: 07/25/2023] [Indexed: 09/17/2023] Open
Abstract
Mechanistic model-based simulations can be deployed to project the persistence of humoral immune response following vaccination. We used this approach to project the antibody persistence through 24 months from the data pooled across five clinical trials in severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-seronegative participants following vaccination with Ad26.COV2.S (5 × 1010 viral particles), given either as a single-dose or a homologous booster regimen at an interval of 2, 3, or 6 months. Antibody persistence was quantified as the percentage of participants with detectable anti-spike binding and wild-type virus neutralizing antibodies. The projected overall 24-month persistence after single-dose Ad26.COV2.S was 70.5% for binding antibodies and 55.2% for neutralizing antibodies, and increased after any homologous booster regimen to greater than or equal to 89.9% for binding and greater than or equal to 80.0% for neutralizing antibodies. The estimated model parameters quantifying the rates of antibody production attributed to short-lived and long-lived plasma cells decreased with increasing age, whereas the rate of antibody production mediated by long-lived plasma cells was higher in women relative to men. Accordingly, a more pronounced waning of antibody responses was predicted in men aged greater than or equal to 60 years and was markedly attenuated following any homologous boosting regimen. The findings suggest that homologous boosting might be a viable strategy for maintaining protective effects of Ad26.COV2.S for up to 24 months following prime vaccination. The estimation of mechanistic modeling parameters identified the long-lived plasma cell pathway as a key contributor mediating antibody persistence following single-dose and homologous booster vaccination with Ad26.COV2.S in different subgroups of recipients stratified by age and sex.
Collapse
Affiliation(s)
- Anna Dari
- Janssen Research & DevelopmentBeerseBelgium
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Dari A, Solforosi L, Roozendaal R, Hoetelmans RMW, Pérez-Ruixo JJ, Boulton M. Mechanistic Model Describing the Time Course of Humoral Immunity Following Ad26.COV2.S Vaccination in Non-Human Primates. J Pharmacol Exp Ther 2023; 387:121-130. [PMID: 37536955 DOI: 10.1124/jpet.123.001591] [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: 01/23/2023] [Revised: 07/14/2023] [Accepted: 07/19/2023] [Indexed: 08/05/2023] Open
Abstract
Mechanistic modeling can be used to describe the time course of vaccine-induced humoral immunity and to identify key biologic drivers in antibody production. We used a six-compartment mechanistic model to describe a 20-week time course of humoral immune responses in 56 non-human primates (NHPs) elicited by vaccination with Ad26.COV2.S according to either a single-dose regimen (1 × 1011 or 5 × 1010 viral particles [vp]) or a two-dose homologous regimen (5 × 1010 vp) given in an interval of 4 or 8 weeks. Humoral immune responses were quantified by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike-specific binding antibody concentrations as determined by spike protein-enzyme-linked immunosorbent assay. The mechanistic model adequately described the central tendency and variability of binding antibody concentrations through 20 weeks in all vaccination arms. The estimation of mechanistic modeling parameters revealed greater contribution of the antibody production mediated by short-lived cells as compared with long-lived cells in driving the peak response, especially post second dose when a more rapid peak response was observed. The antibody production mediated by long-lived cells was identified as relevant for generating the first peak and for contributing to the long-term time course of sustained antibody concentrations in all vaccination arms. The findings contribute evidence on the key biologic components responsible for the observed time course of vaccine-induced humoral immunity in NHPs and constitute a step toward defining immune biomarkers of protection against SARS-CoV-2 that might translate across species. SIGNIFICANCE STATEMENT: We demonstrate the adequacy of a mechanistic modeling approach describing the time course of binding antibody concentrations in non-human primates (NHPs) elicited by different dose levels and regimens of Ad26.COV2.S. The findings are relevant for informing the mechanism-based accounts of vaccine-induced humoral immunity in NHPs and translational research efforts aimed at identifying immune biomarkers of protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Anna Dari
- Janssen Research and Development, Beerse, Belgium (A.D., R.M.W.H., J.-J.P.-R., M.B.); and Janssen Vaccines and Prevention B.V., Leiden, The Netherlands (L.S., R.R.)
| | - Laura Solforosi
- Janssen Research and Development, Beerse, Belgium (A.D., R.M.W.H., J.-J.P.-R., M.B.); and Janssen Vaccines and Prevention B.V., Leiden, The Netherlands (L.S., R.R.)
| | - Ramon Roozendaal
- Janssen Research and Development, Beerse, Belgium (A.D., R.M.W.H., J.-J.P.-R., M.B.); and Janssen Vaccines and Prevention B.V., Leiden, The Netherlands (L.S., R.R.)
| | - Richard M W Hoetelmans
- Janssen Research and Development, Beerse, Belgium (A.D., R.M.W.H., J.-J.P.-R., M.B.); and Janssen Vaccines and Prevention B.V., Leiden, The Netherlands (L.S., R.R.)
| | - Juan-José Pérez-Ruixo
- Janssen Research and Development, Beerse, Belgium (A.D., R.M.W.H., J.-J.P.-R., M.B.); and Janssen Vaccines and Prevention B.V., Leiden, The Netherlands (L.S., R.R.)
| | - Muriel Boulton
- Janssen Research and Development, Beerse, Belgium (A.D., R.M.W.H., J.-J.P.-R., M.B.); and Janssen Vaccines and Prevention B.V., Leiden, The Netherlands (L.S., R.R.)
| |
Collapse
|
25
|
Nguyen DC, Saney C, Hentenaar IT, Cabrera-Mora M, Woodruff MC, Andrews J, Lonial S, Sanz I, Lee FEH. Majority of human circulating plasmablasts stop blasting: A probable misnomer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.10.557057. [PMID: 37745615 PMCID: PMC10515790 DOI: 10.1101/2023.09.10.557057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Following infection or vaccination, early-minted antibody secreting cells (ASC) or plasmablasts appear in circulation transiently, and a small fraction migrates to the spleen or bone marrow (BM) to mature into long-lived plasma cells (LLPC). While LLPC, by definition, are quiescent or non-dividing, the majority of blood ASC are thought to be "blasting" or proliferative. In this study, we find >95% nascent blood ASC in culture express Ki-67 but only 6-12% incorporate BrdU after 4h or 24h labeling. In contrast, <5% BM LLPC in culture are Ki-67 + with no BrdU uptake. Due to limitations of traditional flow cytometry, we utilized a novel optofluidic technology to evaluate cell division with simultaneous functional Ig secretion. We find 11% early-minted blood ASC undergo division, and none of the terminally differentiated BM LLPC (CD19 - CD38 hi CD138 + ) divide during the 7-21 days in culture. While BM LLPC undergo complete cell cycle arrest, the process of differentiation into an ASC of plasmablasts discourages entry into S phase. Since the majority of Ki-67 + nascent blood ASC have exited cell cycle and are no longer actively "blasting", the term "plasmablast", which traditionally refers to an ASC that still has the capacity to divide, may probably be a misnomer.
Collapse
|
26
|
Zhen Z, Zhang L, Li Q, Zhu Y, Wang X, Fu X, Ai J, Wang R, Xie Z, Ma S. Cross-reactive antibodies against monkeypox virus exist in the population immunized with vaccinia Tian Tan strain in China. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023; 113:105477. [PMID: 37392823 DOI: 10.1016/j.meegid.2023.105477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 06/24/2023] [Accepted: 06/28/2023] [Indexed: 07/03/2023]
Abstract
Monkeypox virus (MPXV) belongs to the Orthopoxvirus genus. The worldwide outbreak of MPXV in 2022 has caused widespread concerns. Cross-reactive antibodies induced by vaccinia-inoculation can provide protection against reinfection by MPXV. The vaccinia Tian Tan (VTT) strain, which was widely inoculated in the Chinese population before the 1980s, has genomic differences from other vaccinia strains, although they all belong to the orthopoxviruses family. The current seroprevalence of VTT-vaccinated populations remains unclear more than four decades after the termination of vaccination campaigns in China. Our results showed that cross-reactive IgG antibodies against MPXV were present in 31.8% (75/236) of vaccinees four decades after VTT-vaccination, suggesting that vaccination with VTT may provide long-term protection against MPXV infection in some individuals.
Collapse
Affiliation(s)
- Zida Zhen
- Department of Transfusion Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Linlin Zhang
- Beijing Key Laboratory of Pediatric Respiratory Infectious Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Research Unit of Critical Infection in Children, 2019RU016, Chinese Academy of Medical Sciences, Beijing 100045, China
| | - Qi Li
- Beijing Key Laboratory of Pediatric Respiratory Infectious Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Research Unit of Critical Infection in Children, 2019RU016, Chinese Academy of Medical Sciences, Beijing 100045, China
| | - Yun Zhu
- Beijing Key Laboratory of Pediatric Respiratory Infectious Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Research Unit of Critical Infection in Children, 2019RU016, Chinese Academy of Medical Sciences, Beijing 100045, China
| | - Xiaohuan Wang
- Department of Transfusion Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Xiaoyan Fu
- Department of Transfusion Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China
| | - Junhong Ai
- Beijing Key Laboratory of Pediatric Respiratory Infectious Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Research Unit of Critical Infection in Children, 2019RU016, Chinese Academy of Medical Sciences, Beijing 100045, China
| | - Ran Wang
- Beijing Key Laboratory of Pediatric Respiratory Infectious Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Research Unit of Critical Infection in Children, 2019RU016, Chinese Academy of Medical Sciences, Beijing 100045, China.
| | - Zhengde Xie
- Beijing Key Laboratory of Pediatric Respiratory Infectious Diseases, Key Laboratory of Major Diseases in Children, Ministry of Education, National Clinical Research Center for Respiratory Diseases, Laboratory of Infection and Virology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China; Research Unit of Critical Infection in Children, 2019RU016, Chinese Academy of Medical Sciences, Beijing 100045, China
| | - Shuxuan Ma
- Department of Transfusion Medicine, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing 100045, China.
| |
Collapse
|
27
|
Hill TF, Narvekar P, Asher G, Camp N, Thomas KR, Tasian SK, Rawlings DJ, James RG. Human plasma cells engineered to secrete bispecifics drive effective in vivo leukemia killing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554523. [PMID: 37662410 PMCID: PMC10473709 DOI: 10.1101/2023.08.24.554523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Bispecific antibodies are an important tool for the management and treatment of acute leukemias. Advances in genome-engineering have enabled the generation of human plasma cells that secrete therapeutic proteins and are capable of long-term in vivo engraftment in humanized mouse models. As a next step towards clinical translation of engineered plasma cells (ePCs) towards cancer therapy, here we describe approaches for the expression and secretion of bispecific antibodies by human plasma cells. We show that human ePCs expressing either fragment crystallizable domain deficient anti-CD19 × anti-CD3 (blinatumomab) or anti-CD33 × anti-CD3 bispecific antibodies mediate T cell activation and direct T cell killing of specific primary human cell subsets and B-acute lymphoblastic leukemia or acute myeloid leukemia cell lines in vitro. We demonstrate that knockout of the self-expressed antigen, CD19, boosts anti-CD19 bispecific secretion by ePCs and prevents self-targeting. Further, anti-CD19 bispecific-ePCs elicited tumor eradication in vivo following local delivery in flank-implanted Raji lymphoma cells. Finally, immunodeficient mice engrafted with anti-CD19 bispecific-ePCs and autologous T cells potently prevented in vivo growth of CD19+ acute lymphoblastic leukemia in patient-derived xenografts. Collectively, these findings support further development of ePCs for use as a durable, local delivery system for the treatment of acute leukemias, and potentially other cancers.
Collapse
Affiliation(s)
- Tyler F. Hill
- University of Washington, Medical Scientist Training Program, Seattle WA
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
| | - Parnal Narvekar
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
| | - Gregory Asher
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
| | - Nathan Camp
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
| | - Kerri R. Thomas
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
| | - Sarah K. Tasian
- Children’s Hospital of Philadelphia, Division of Oncology and Center for Childhood Cancer Research, Philadelphia PA
- Department of Pediatrics and Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia PA
| | - David J. Rawlings
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
- University of Washington, Departments of Pediatrics and Immunology, Seattle WA
| | - Richard G. James
- Seattle Children’s Research Institute, Center for Immunity and Immunotherapy, Seattle WA
- University of Washington, Departments of Pediatrics and Pharmacology, Seattle WA
| |
Collapse
|
28
|
Steinmetz TD, Verstappen GM, Suurmond J, Kroese FGM. Targeting plasma cells in systemic autoimmune rheumatic diseases - Promises and pitfalls. Immunol Lett 2023; 260:44-57. [PMID: 37315847 DOI: 10.1016/j.imlet.2023.06.005] [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: 02/03/2023] [Revised: 05/12/2023] [Accepted: 06/10/2023] [Indexed: 06/16/2023]
Abstract
Plasma cells are the antibody secretors of the immune system. Continuous antibody secretion over years can provide long-term immune protection but could also be held responsible for long-lasting autoimmunity in case of self-reactive plasma cells. Systemic autoimmune rheumatic diseases (ARD) affect multiple organ systems and are associated with a plethora of different autoantibodies. Two prototypic systemic ARDs are systemic lupus erythematosus (SLE) and Sjögren's disease (SjD). Both diseases are characterized by B-cell hyperactivity and the production of autoantibodies against nuclear antigens. Analogues to other immune cells, different subsets of plasma cells have been described. Plasma cell subsets are often defined dependent on their current state of maturation, that also depend on the precursor B-cell subset from which they derived. But, a universal definition of plasma cell subsets is not available so far. Furthermore, the ability for long-term survival and effector functions may differ, potentially in a disease-specific manner. Characterization of plasma cell subsets and their specificity in individual patients can help to choose a suitable targeting approach for either a broad or more selective plasma cell depletion. Targeting plasma cells in systemic ARDs is currently challenging because of side effects or varying depletion efficacies in the tissue. Recent developments, however, like antigen-specific targeting and CAR-T-cell therapy might open up major benefits for patients beyond current treatment options.
Collapse
Affiliation(s)
- Tobit D Steinmetz
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
| | - Gwenny M Verstappen
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jolien Suurmond
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Frans G M Kroese
- University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
29
|
Sgrò D, Rossi P, Piaggi P, Brancatella A, Lorusso L, Bottici V, Molinaro E, Latrofa F, Elisei R, Agate L. Significance of Thyroglobulin Autoantibodies in Patients With Thyroid Cancer Treated With Lenvatinib. J Endocr Soc 2023; 7:bvad084. [PMID: 37440964 PMCID: PMC10334479 DOI: 10.1210/jendso/bvad084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Indexed: 07/15/2023] Open
Abstract
Context Serum thyroglobulin (Tg) is a highly sensitive and specific tumor marker, employed in post-operative management of patients with differentiated thyroid carcinomas. Tumor shrinkage of radioiodine-refractory thyroid cancer (RAIR-DTC) treated with multitarget kinase inhibitors as lenvatinib, expressed according to the Response Evaluation Criteria in Solid Tumors (RECIST), is also associated with a drastic reduction of Tg levels. However, interference caused by circulating thyroglobulin autoantibodies (TgAb) represents the main limitation in the clinical use of Tg. Objective To evaluate if in RAIR-DTC TgAb could be considered a surrogate marker of Tg in monitoring response to treatment with lenvatinib. Design We retrospectively evaluated patients who had started lenvatinib and correlated serum Tg and TgAb with the radiological response across visits. Setting University of Pisa, Italy. Patients We selected 9/97 RAIR-DTC patients with detectable TgAb. Intervention None. Main Outcome Measures None. Results Tg values correlated neither with TgAb title nor with radiological response across visits. Greater decreases in TgAb titer correlated with favorable radiological response to lenvatinib after 1 month (Spearman's correlation = 0.74, P = .021) and 6 months (correlation = 0.61, P = .079). According to RECIST, patients with partial response showed a ∼10-fold greater decrease in TgAb compared to those with stable disease at 1 month (median TgAb decrease: -142 vs -14 IU/mL, P = .01) and those with progressive disease at 6 months (median TgAb decrease: -264 vs-24 IU/mL, P = .04). Conclusion TgAb evaluation may represent a reliable surrogate marker for Tg trend in evaluating response of RAIR-DTC to treatment with lenvatinib. A multicentric study would be useful to confirm our results.
Collapse
Affiliation(s)
- Daniele Sgrò
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Piercarlo Rossi
- Radiology, Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, Pisa 56127, Italy
| | - Paolo Piaggi
- Department of Information Engineering, University of Pisa, Pisa 56127, Italy
| | - Alessandro Brancatella
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Loredana Lorusso
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Valeria Bottici
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Eleonora Molinaro
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Francesco Latrofa
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| | - Rossella Elisei
- Correspondence: Rossella Elisei, MD, Endocrinology Unit I, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Via Paradisa 2, Pisa 56127, Italy.
| | - Laura Agate
- Endocrinology Unit, Department of Clinical and Experimental Medicine, University of Pisa, Pisa 56127, Italy
| |
Collapse
|
30
|
Rogers GL, Huang C, Mathur A, Huang X, Chen HY, Stanten K, Morales H, Chang CH, Kezirian EJ, Cannon PM. Reprogramming human B cells with custom heavy chain antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.28.546944. [PMID: 37425794 PMCID: PMC10327003 DOI: 10.1101/2023.06.28.546944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
We describe a genome editing strategy to reprogram the immunoglobulin heavy chain (IgH) locus of human B cells to express custom molecules that respond to immunization. These heavy chain antibodies (HCAbs) comprise a custom antigen-recognition domain linked to an Fc domain derived from the IgH locus and can be differentially spliced to express either B cell receptor (BCR) or secreted antibody isoforms. The HCAb editing platform is highly flexible, supporting antigen-binding domains based on both antibody and non-antibody components, and also allowing alterations in the Fc domain. Using HIV Env protein as a model antigen, we show that B cells edited to express anti-Env HCAbs support the regulated expression of both BCRs and antibodies, and respond to Env antigen in a tonsil organoid model of immunization. In this way, human B cells can be reprogrammed to produce customized therapeutic molecules with the potential for in vivo amplification.
Collapse
Affiliation(s)
- Geoffrey L. Rogers
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Chun Huang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Atishay Mathur
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Xiaoli Huang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Hsu-Yu Chen
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Kalya Stanten
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Heidy Morales
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Chan-Hua Chang
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Eric J. Kezirian
- Department of Otolaryngology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| | - Paula M. Cannon
- Department of Molecular Microbiology and Immunology, Keck School of Medicine of the University of Southern California, Los Angeles, California, USA
| |
Collapse
|
31
|
Haas M, Fest T. Final step of B-cell differentiation into plasmablasts; the right time to activate plasma cell PIM2 kinase. Immunol Lett 2023; 258:45-50. [PMID: 37207916 DOI: 10.1016/j.imlet.2023.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 05/13/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023]
Abstract
The differentiation of B cells into antibody-secreting plasma cells is a complex process that involves extensive changes in morphology, lifespan, and cellular metabolism to support the high rates of antibody production. During the final stage of differentiation, B cells undergo significant expansion of their endoplasmic reticulum and mitochondria, which induces cellular stress and may lead to cell death in absence of effective inhibition of the apoptotic pathway. These changes are tightly regulated at transcriptional and epigenetic levels, as well as at post-translational level, with protein modifications playing a critical role in the process of cellular modification and adaptation. Our recent research has highlighted the pivotal role of the serine/threonine kinase PIM2 in B cell differentiation, from commitment stage to plasmablast and maintenance of expression in mature plasma cells. PIM2 has been shown to promote cell cycle progression during the final stage of differentiation and to inhibit Caspase 3 activation, raising the threshold for apoptosis. In this review, we examine the key molecular mechanisms controlled by PIM2 that contribute to plasma cell development and maintenance.
Collapse
Affiliation(s)
- Marion Haas
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, Team B_DEVIL, UMR_S1236, Rennes, France; Laboratoire d'Hématologie, Centre Hospitalier Universitaire, Rennes, France
| | - Thierry Fest
- Université de Rennes 1, INSERM, Établissement Français du Sang de Bretagne, Team B_DEVIL, UMR_S1236, Rennes, France; Laboratoire d'Hématologie, Centre Hospitalier Universitaire, Rennes, France.
| |
Collapse
|
32
|
Azar JH, Evans JP, Sikorski MH, Chakravarthy KB, McKenney S, Carmody I, Zeng C, Teodorescu R, Song NJ, Hamon JL, Bucci D, Velegraki M, Bolyard C, Weller KP, Reisinger SA, Bhat SA, Maddocks KJ, Denlinger N, Epperla N, Gumina RJ, Vlasova AN, Oltz EM, Saif LJ, Chung D, Woyach JA, Shields PG, Liu SL, Li Z, Rubinstein MP. Selective suppression of de novo SARS-CoV-2 vaccine antibody responses in patients with cancer on B cell-targeted therapy. JCI Insight 2023; 8:e163434. [PMID: 36749632 PMCID: PMC10070099 DOI: 10.1172/jci.insight.163434] [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: 07/08/2022] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
We assessed vaccine-induced antibody responses to the SARS-CoV-2 ancestral virus and Omicron variant before and after booster immunization in 57 patients with B cell malignancies. Over one-third of vaccinated patients at the pre-booster time point were seronegative, and these patients were predominantly on active cancer therapies such as anti-CD20 monoclonal antibody. While booster immunization was able to induce detectable antibodies in a small fraction of seronegative patients, the overall booster benefit was disproportionately evident in patients already seropositive and not receiving active therapy. While ancestral virus- and Omicron variant-reactive antibody levels among individual patients were largely concordant, neutralizing antibodies against Omicron tended to be reduced. Interestingly, in all patients, including those unable to generate detectable antibodies against SARS-CoV-2 spike, we observed comparable levels of EBV- and influenza-reactive antibodies, demonstrating that B cell-targeting therapies primarily impair de novo but not preexisting antibody levels. These findings support rationale for vaccination before cancer treatment.
Collapse
Affiliation(s)
- Joseph H. Azar
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - John P. Evans
- Center for Retrovirus Research
- Department of Veterinary Biosciences
- Molecular, Cellular and Developmental Biology Program
| | - Madison H. Sikorski
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Karthik B. Chakravarthy
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Selah McKenney
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Ian Carmody
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Cong Zeng
- Center for Retrovirus Research
- Department of Veterinary Biosciences
| | - Rachael Teodorescu
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - No-Joon Song
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Jamie L. Hamon
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Donna Bucci
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Maria Velegraki
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Chelsea Bolyard
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Kevin P. Weller
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Sarah A. Reisinger
- The Ohio State University Comprehensive Cancer Center – James, The James Cancer Hospital
| | - Seema A. Bhat
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center – James
| | - Kami J. Maddocks
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center – James
| | - Nathan Denlinger
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center – James
| | - Narendranath Epperla
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center – James
| | - Richard J. Gumina
- Department of Internal Medicine, Division of Cardiovascular Medicine; and
| | - Anastasia N. Vlasova
- Center for Food Animal Health, Animal Sciences Department, Ohio Agricultural Research and Development Center, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Columbus, Ohio, USA
- Veterinary Preventive Medicine Department, College of Veterinary Medicine, The Ohio State University, Wooster, Ohio, USA
- Viruses and Emerging Pathogens Program, Infectious Diseases Institute
| | - Eugene M. Oltz
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
- Department of Microbial Infection and Immunity; and
| | - Linda J. Saif
- Center for Food Animal Health, Animal Sciences Department, Ohio Agricultural Research and Development Center, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Columbus, Ohio, USA
- Veterinary Preventive Medicine Department, College of Veterinary Medicine, The Ohio State University, Wooster, Ohio, USA
- Viruses and Emerging Pathogens Program, Infectious Diseases Institute
| | - Dongjun Chung
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
- Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio, USA
| | - Jennifer A. Woyach
- Division of Hematology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center – James
| | - Peter G. Shields
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Shan-Lu Liu
- Center for Retrovirus Research
- Department of Veterinary Biosciences
- Viruses and Emerging Pathogens Program, Infectious Diseases Institute
- Department of Microbial Infection and Immunity; and
| | - Zihai Li
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| | - Mark P. Rubinstein
- Division of Medical Oncology, Department of Internal Medicine
- The Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center – James
| |
Collapse
|
33
|
Swift M, Horns F, Quake SR. Lineage tracing reveals fate bias and transcriptional memory in human B cells. Life Sci Alliance 2023; 6:e202201792. [PMID: 36639222 PMCID: PMC9840405 DOI: 10.26508/lsa.202201792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 01/15/2023] Open
Abstract
We combined single-cell transcriptomics and lineage tracing to understand fate choice in human B cells. Using the antibody sequences of B cells, we tracked clones during in vitro differentiation. Clonal analysis revealed a subset of IgM+ B cells which were more proliferative than other B-cell types. Whereas the population of B cells adopted diverse states during differentiation, clones had a restricted set of fates available to them; there were two times more single-fate clones than expected given population-level cell-type diversity. This implicated a molecular memory of initial cell states that was propagated through differentiation. We then identified the genes which had strongest coherence within clones. These genes significantly overlapped known B-cell fate determination programs, suggesting the genes which determine cell identity are most robustly controlled on a clonal level. Persistent clonal identities were also observed in human plasma cells from bone marrow, indicating that these transcriptional programs maintain long-term cell identities in vivo. Thus, we show how cell-intrinsic fate bias influences differentiation outcomes in vitro and in vivo.
Collapse
Affiliation(s)
- Michael Swift
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA, USA
| | - Felix Horns
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA, USA
| | - Stephen R Quake
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Applied Physics, Stanford University, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| |
Collapse
|
34
|
Hill JA, Kiem ES, Bhatti A, Liu W, Keane-Candib J, Fitzpatrick KS, Boonyaratanakornkit J, Gardner RA, Green DJ, Maloney DG, Turtle CJ, Smith JM, Gimferrer I, Blosser CD, Jackson SW. Anti-HLA antibodies in recipients of CD19 versus BCMA-targeted CAR T-cell therapy. Am J Transplant 2023; 23:416-422. [PMID: 36748802 PMCID: PMC10266802 DOI: 10.1016/j.ajt.2022.11.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/05/2022] [Accepted: 11/06/2022] [Indexed: 01/15/2023]
Abstract
Antibodies against foreign human leukocyte antigen (HLA) molecules are barriers to successful organ transplantation. B cell-depleting treatments are used to reduce anti-HLA antibodies but have limited efficacy. We hypothesized that the primary source for anti-HLA antibodies is long-lived plasma cells, which are ineffectively targeted by B cell depletion. To study this, we screened for anti-HLA antibodies in a prospectively enrolled cohort of 49 patients who received chimeric antigen receptor T-cell therapy (CARTx), targeting naïve and memory B cells (CD19-targeted, n = 21) or plasma cells (BCMA-targeted, n = 28) for hematologic malignancies. Longitudinal samples were collected before and up to 1 year after CARTx. All individuals were in sustained remission. We identified 4 participants with anti-HLA antibodies before CD19-CARTx. Despite B cell depletion, anti-HLA antibodies and calculated panel reactive antibody scores were stable for 1 year after CD19-CARTx. Only 1 BCMA-CARTx recipient had pre-CARTx low-level anti-HLA antibodies, with no follow-up samples available. These data implicate CD19neg long-lived plasma cells as an important source for anti-HLA antibodies, a model supported by infrequent HLA sensitization in BCMA-CARTx subjects receiving previous plasma cell-targeted therapies. Thus, plasma cell-targeted therapies may be more effective against HLA antibodies, thereby enabling improved access to organ transplantation and rejection management.
Collapse
Affiliation(s)
- Joshua A Hill
- Departments of Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA.
| | - Erika S Kiem
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Atif Bhatti
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Winnie Liu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Jacob Keane-Candib
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Kristin S Fitzpatrick
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA; Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Jim Boonyaratanakornkit
- Departments of Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Rebecca A Gardner
- Seattle Children's Research Institute, Seattle, Washington, USA; Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| | - Damian J Green
- Departments of Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - David G Maloney
- Departments of Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Cameron J Turtle
- Departments of Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Jodi M Smith
- Seattle Children's Research Institute, Seattle, Washington, USA; Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| | - Idoia Gimferrer
- Immunogenetics/HLA laboratory Bloodworks Northwest, Seattle, Washington, USA
| | - Christopher D Blosser
- Departments of Medicine, University of Washington School of Medicine, Seattle, Washington, USA; Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
| | - Shaun W Jackson
- Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, Washington, USA; Seattle Children's Research Institute, Seattle, Washington, USA; Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA.
| |
Collapse
|
35
|
Desai N, Pradhan V, Chougule D, Tiwari S, Mandke C, Yadav RM, Athvale A, Kawle J, Pai V, Pawaskar S, Kharkar H, Bhosale S, Parab A, Ansari S, Kumar KH, Mhashal S, Redkar N, Madkaikar M. Perturbations of immune landscape in COVID-19 associated mucormycosis. Mycoses 2023; 66:226-236. [PMID: 36380699 DOI: 10.1111/myc.13546] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 11/11/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND A rise in secondary fungal infections during the COVID-19 pandemic necessitates a deeper understanding of the associated immunological perturbations. OBJECTIVES To evaluate the clinical and immunological characteristics observed in patients with COVID-19 associated mucormycosis (CAM) infection. PATIENTS/ METHODS Cases of mucormycosis with or post-COVID-19 infection were compared with cases of acute COVID-19 and convalescent COVID-19. Lymphocyte subsets, cytokines and other laboratory markers were compared between the groups. RESULTS The frequency of proposed risk factors for CAM was diabetes mellitus (77%), recent history of steroid use (69%) and hypoxia during COVID-19 infection (52%). Iron metabolism was dysregulated in CAM patients with low TIBC and total iron. Further, CAM was accompanied with lymphopenia with drastic reduction in B cell counts; however, plasmablasts were not altered. Further, CAM patients had low immunoglobulin levels and antibodies specific to mucor peptide did not increase in CAM suggesting dysfunction in B-cell response. There was increase in activated effector cytotoxic CD8 T cells and NK cells in CAM compared with COVID-19 infection and healthy controls. Among T helper cells, Tregs were reduced and Th-1 frequency was increased in CAM compared with COVID-19 infection. A distinct cytokine signature was evident in CAM with increase in IL-1β, IFN-γ, IL-6, IL-22, IL-17A, IL-10, IL-2, IL-8, IL-7, IL-21 and GM-CSF. CONCLUSION This is the first study on immunophenotyping in CAM suggesting the need for long-term monitoring of B-cell function after SARS-CoV-2 in patients with dysregulated glycaemic control and the possible benefit of therapeutic supplementation with intravenous immunoglobulins in CAM.
Collapse
Affiliation(s)
- Nidhi Desai
- Department of Pediatric Immunology and Leukocyte Biology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Vandana Pradhan
- Department of Clinical & Experimental Immunology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Durga Chougule
- Department of Clinical & Experimental Immunology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Smrati Tiwari
- Department of Medicine, G.S. Medical College, King Edward Memorial Hospital, Mumbai, India
| | - Charuta Mandke
- Department of Ophthalmology, HBT Medical College and Dr R N Cooper Hospital, Mumbai, India
| | - Reetika Malik Yadav
- Department of Pediatric Immunology and Leukocyte Biology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Amita Athvale
- Department of Pulmonary Medicine, G.S. Medical College, King Edward Memorial Hospital, Mumbai, India
| | - Juhi Kawle
- Department of Medicine, G.S. Medical College, King Edward Memorial Hospital, Mumbai, India
| | - Vinayak Pai
- Department of Medicine, G.S. Medical College, King Edward Memorial Hospital, Mumbai, India
| | - Swapnal Pawaskar
- Department of Clinical & Experimental Immunology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Harshada Kharkar
- Department of Clinical & Experimental Immunology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Snehal Bhosale
- Department of Pediatric Immunology and Leukocyte Biology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Ankita Parab
- Department of Pediatric Immunology and Leukocyte Biology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| | - Shazia Ansari
- Department of Ophthalmology, HBT Medical College and Dr R N Cooper Hospital, Mumbai, India
| | - Kinnera Harish Kumar
- Department of Otorhinolaryngology, HBT Medical College and Dr R N Cooper Hospital, Mumbai, India
| | - Shashikant Mhashal
- Department of Otolaryngology, HBT Medical College and Dr R N Cooper Hospital, Mumbai, India
| | - Neelam Redkar
- Department of Medicine, HBT Medical College and Dr R N Cooper Hospital, Mumbai, India
| | - Manisha Madkaikar
- Department of Pediatric Immunology and Leukocyte Biology, Indian Council of Medical Research- National Institute of Immunohaematology, Mumbai, India
| |
Collapse
|
36
|
Notch Signaling in Acute Inflammation and Sepsis. Int J Mol Sci 2023; 24:ijms24043458. [PMID: 36834869 PMCID: PMC9967996 DOI: 10.3390/ijms24043458] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Notch signaling, a highly conserved pathway in mammals, is crucial for differentiation and homeostasis of immune cells. Besides, this pathway is also directly involved in the transmission of immune signals. Notch signaling per se does not have a clear pro- or anti-inflammatory effect, but rather its impact is highly dependent on the immune cell type and the cellular environment, modulating several inflammatory conditions including sepsis, and therefore significantly impacts the course of disease. In this review, we will discuss the contribution of Notch signaling on the clinical picture of systemic inflammatory diseases, especially sepsis. Specifically, we will review its role during immune cell development and its contribution to the modulation of organ-specific immune responses. Finally, we will evaluate to what extent manipulation of the Notch signaling pathway could be a future therapeutic strategy.
Collapse
|
37
|
Pampusch MS, Sevcik EN, Quinn ZE, Davey BC, Berg JM, Gorrell-Brown I, Abdelaal HM, Rakasz EG, Rendahl A, Skinner PJ. Assessment of anti-CD20 antibody pre-treatment for augmentation of CAR-T cell therapy in SIV-infected rhesus macaques. Front Immunol 2023; 14:1101446. [PMID: 36825014 PMCID: PMC9941136 DOI: 10.3389/fimmu.2023.1101446] [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: 11/17/2022] [Accepted: 01/25/2023] [Indexed: 02/10/2023] Open
Abstract
During chronic HIV and SIV infections, the majority of viral replication occurs within lymphoid follicles. In a pilot study, infusion of SIV-specific CD4-MBL-CAR-T cells expressing the follicular homing receptor, CXCR5, led to follicular localization of the cells and a reduction in SIV viral loads in rhesus macaques. However, the CAR-T cells failed to persist. We hypothesized that temporary disruption of follicles would create space for CAR-T cell engraftment and lead to increased abundance and persistence of CAR-T cells. In this study we treated SIV-infected rhesus macaques with CAR-T cells and preconditioned one set with anti-CD20 antibody to disrupt the follicles. We evaluated CAR-T cell abundance and persistence in four groups of SIVmac239-infected and ART-suppressed animals: untreated, CAR-T cell treated, CD20 depleted, and CD20 depleted/CAR-T cell treated. In the depletion study, anti-CD20 was infused one week prior to CAR-T infusion and cessation of ART. Anti-CD20 antibody treatment led to temporary depletion of CD20+ cells in blood and partial depletion in lymph nodes. In this dose escalation study, there was no impact of CAR-T cell infusion on SIV viral load. However, in both the depleted and non-depleted animals, CAR-T cells accumulated in and around lymphoid follicles and were Ki67+. CAR-T cells increased in number in follicles from 2 to 6 days post-treatment, with a median 15.2-fold increase in follicular CAR-T cell numbers in depleted/CAR-T treated animals compared to an 8.1-fold increase in non-depleted CAR-T treated animals. The increase in CAR T cells in depleted animals was associated with a prolonged elevation of serum IL-6 levels and a rapid loss of detectable CAR-T cells. Taken together, these data suggest that CAR-T cells likely expanded to a greater extent in depleted/CAR-T cell treated animals. Further studies are needed to elucidate mechanisms mediating the rapid loss of CAR-T cells and to evaluate strategies to improve engraftment and persistence of HIV-specific CAR-T cells. The potential for an inflammatory cytokine response appears to be enhanced with anti-CD20 antibody treatment and future studies may require CRS control strategies. These studies provide important insights into cellular immunotherapy and suggest future studies for improved outcomes.
Collapse
Affiliation(s)
- Mary S. Pampusch
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Emily N. Sevcik
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Zoe E. Quinn
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Brianna C. Davey
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - James M. Berg
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Ian Gorrell-Brown
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Hadia M. Abdelaal
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin, Madison WI, United States
| | - Aaron Rendahl
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| | - Pamela J. Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, United States
| |
Collapse
|
38
|
Abstract
Access to kidney transplantation is limited by HLA-specific sensitization. Desensitization strategies enable crossmatch-positive kidney transplantation. In this review, we describe clinical experience gained over the last 20 y using desensitization strategies before kidney transplantation and describe the different tools used (both drugs and apheresis options), including IVIg, rituximab, apheresis techniques, interleukin-6 interference, proteasome inhibition, enzymatic degradation of HLA antibodies, complement inhibition, and B cytokine interference. Although access to transplantation for highly sensitized kidney transplantation candidates has been vastly improved by desensitization strategies, it remains, however, limited by the recurrence of HLA antibodies after transplantation and the occurrence of antibody-mediated rejection.
Collapse
|
39
|
Dari A, Boulton M, Neyens M, Le Gars M, Valenzuela B, Shukarev G, Cárdenas V, Ruiz-Guiñazú J, Sadoff J, Hoetelmans RMW, Ruixo JJP. Quantifying Antibody Persistence After a Single Dose of COVID-19 Vaccine Ad26.COV2.S in Humans Using a Mechanistic Modeling and Simulation Approach. Clin Pharmacol Ther 2023; 113:380-389. [PMID: 36377532 PMCID: PMC10107600 DOI: 10.1002/cpt.2796] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022]
Abstract
Understanding persistence of humoral immune responses elicited by vaccination against coronavirus disease 2019 (COVID-19) is critical for informing the duration of protection and appropriate booster timing. We developed a mechanistic model to characterize the time course of humoral immune responses in severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2)-seronegative adults after primary vaccination with the Janssen COVID-19 vaccine, Ad26.COV2.S. The persistence of antibody responses was quantified through mechanistic modeling-based simulations. Two biomarkers of humoral immune responses were examined: SARS-CoV-2 neutralizing antibodies determined by wild-type virus neutralization assay (wtVNA) and spike protein-binding antibodies determined by indirect spike protein enzyme-linked immunosorbent assay (S-ELISA). The persistence of antibody responses was defined as the period of time during which wtVNA and S-ELISA titers remained above the lower limit of quantification. A total of 442 wtVNA and 1,185 S-ELISA titers from 82 and 220 participants, respectively, were analyzed following administration of a single dose of Ad26.COV2.S (5 × 1010 viral particles). The mechanistic model adequately described the time course of observed wtVNA and S-ELISA serum titers and its associated variability up to 8 months following vaccination. Mechanistic model-based simulations show that single-dose Ad26.COV2.S elicits durable but waning antibody responses up to 24 months following immunization. Of the estimated model parameters, the production rate of memory B cells was decreased in older adults relative to younger adults, and the antibody production rate mediated by long-lived plasma cells was increased in women relative to men. A steeper waning of antibody responses was predicted in men and in older adults.
Collapse
Affiliation(s)
- Anna Dari
- Janssen Research and Development, Beerse, Belgium
| | | | | | | | - Belén Valenzuela
- Janssen-Cilag Spain, Part of Janssen Pharmaceutical Companies, Madrid, Spain
| | | | - Vicky Cárdenas
- Janssen Research and Development, Spring House, Pennsylvania, USA
| | | | - Jerald Sadoff
- Janssen Vaccines and Prevention, Leiden, The Netherlands
| | | | | |
Collapse
|
40
|
The cellular biology of plasma cells: Unmet challenges and opportunities. Immunol Lett 2023; 254:6-12. [PMID: 36646289 DOI: 10.1016/j.imlet.2023.01.005] [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: 11/06/2022] [Revised: 12/27/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
Plasma cells and the antibodies they secrete are paramount for protection against infection but can also be implicated in diseases including autoantibody-mediated disease and multiple myeloma. Plasma cell terminal differentiation relies on a transcriptional switch and on important morphological changes. The cellular and molecular mechanisms underlying these processes are partly understood and how plasma cells manage to survive for long periods of time while secreting large quantities of antibodies remains unclear. In this review we aim to put in perspective what is known about plasma cell cellular biology to highlight the challenges faced by this field of research but also to illustrate how new opportunities may arise from the study of the fundamental mechanisms sustaining plasma cell survival and function.
Collapse
|
41
|
Yang N, Maskey AR, Srivastava K, Kim M, Wang Z, Musa I, Shi Y, Gong Y, Fidan O, Wang J, Dunkin D, Chung D, Zhan J, Miao M, Sampson HA, Li XM. Inhibition of pathologic immunoglobulin E in food allergy by EBF-2 and active compound berberine associated with immunometabolism regulation. Front Immunol 2023; 14:1081121. [PMID: 36825019 PMCID: PMC9941740 DOI: 10.3389/fimmu.2023.1081121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/02/2023] [Indexed: 02/10/2023] Open
Abstract
Introduction Food allergy is a significant public health problem with limited treatment options. As Food Allergy Herbal Formula 2 (FAHF-2) showed potential as a food allergy treatment, we further developed a purified version named EBF-2 and identified active compounds. We investigated the mechanisms of EBF-2 on IgE-mediated peanut (PN) allergy and its active compound, berberine, on IgE production. Methods IgE plasma cell line U266 cells were cultured with EBF-2 and FAHF-2, and their effects on IgE production were compared. EBF-2 was evaluated in a murine PN allergy model for its effect on PN-specific IgE production, number of IgE+ plasma cells, and PN anaphylaxis. Effects of berberine on IgE production, the expression of transcription factors, and mitochondrial glucose metabolism in U266 cells were evaluated. Results EBF-2 dose-dependently suppressed IgE production and was over 16 times more potent than FAHF-2 in IgE suppression in U266 cells. EBF-2 significantly suppressed PN-specific IgE production (70%, p<0.001) and the number of IgE-producing plasma cells in PN allergic mice, accompanied by 100% inhibition of PN-induced anaphylaxis and plasma histamine release (p<0.001) without affecting IgG1 or IgG2a production. Berberine markedly suppressed IgE production, which was associated with suppression of XBP1, BLIMP1, and STAT6 transcription factors and a reduced rate of mitochondrial oxidation in an IgE-producing plasma cell line. Conclusions EBF-2 and its active compound berberine are potent IgE suppressors, associated with cellular regulation of immunometabolism on IgE plasma cells, and may be a potential therapy for IgE-mediated food allergy and other allergic disorders.
Collapse
Affiliation(s)
- Nan Yang
- General Nutraceutical Technology, Elmsford, NY, United States.,Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | - Anish R Maskey
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | - Kamal Srivastava
- General Nutraceutical Technology, Elmsford, NY, United States.,Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | - Monica Kim
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Zixi Wang
- Department of Allergy, Peking Union Medical College Hospital, Beijing, China
| | - Ibrahim Musa
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States
| | - Yanmei Shi
- Academy of Chinese Medicine Sciences, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Yixuan Gong
- Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ozkan Fidan
- Department of Biological Engineering, Utah State University, Logan, UT, United States.,Department of Bioengineering, Abdullah Gul University, Kayseri, Türkiye
| | - Julie Wang
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - David Dunkin
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Danna Chung
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Jixun Zhan
- Department of Biological Engineering, Utah State University, Logan, UT, United States
| | - Mingsan Miao
- Academy of Chinese Medicine Sciences, Henan University of Traditional Chinese Medicine, Zhengzhou, Henan, China
| | - Hugh A Sampson
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Xiu-Min Li
- Department of Pathology, Microbiology and Immunology, New York Medical College, Valhalla, NY, United States.,Department of Otolaryngology, New York Medical College, Valhalla, NY, United States
| |
Collapse
|
42
|
Single cell multi-omic reference atlases of non-human primate immune tissues reveals CD102 as a biomarker for long-lived plasma cells. Commun Biol 2022; 5:1399. [PMID: 36543997 PMCID: PMC9770566 DOI: 10.1038/s42003-022-04216-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 11/02/2022] [Indexed: 12/24/2022] Open
Abstract
In response to infection or immunization, antibodies are produced that provide protection against re-exposure with the same pathogen. These antibodies can persist at high titers for decades and are maintained by bone marrow-resident long-lived plasma cells (LLPC). However, the durability of antibody responses to immunization varies amongst vaccines. It is unknown what factors contribute to the differential longevity of serum antibody responses and whether heterogeneity in LLPC contributes to this phenomenon. While LLPC differentiation has been studied extensively in mice, little is known about this population in humans or non-human primates (NHP). Here, we use multi-omic single-cell profiling to identify and characterize the LLPC compartment in NHP. We identify LLPC biomarkers including the marker CD102 and show that CD102 in combination with CD31 identifies LLPC in NHP bone marrow. Additionally, we find that CD102 is expressed by LLPC in mouse and humans. These results further our understanding of the LLPC compartment in NHP, identify biomarkers of LLPC, and provide tissue-specific single cell references for future studies.
Collapse
|
43
|
A Retrospective Study of the Safety and Immunogenicity of MVC-COV1901 Vaccine for People Living with HIV. Vaccines (Basel) 2022; 11:vaccines11010018. [PMID: 36679862 PMCID: PMC9863561 DOI: 10.3390/vaccines11010018] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND This study aimed to assess the safety and immunogenicity of MVC-COV1901, a recombinant COVID-19 protein vaccine, containing S-2P protein adjuvanted with CpG 1018 and aluminum hydroxide, for people living with HIV (PWH). METHODS A total of 57 PWH of ≥20 years of age who are on stable antiretroviral therapy were compared with 882 HIV-negative participants. Participants received two doses of MVC-COV1901 28 days apart. RESULTS No vaccine-related serious adverse events (SAEs) were recorded. Seroconversion rates (SCRs) of 100% and 99.8% were achieved in PWH and comparators, respectively, 28 days after the second dose. After adjusting for sex, age, BMI category, and comorbidity, the adjusted GMT ratio of comparator/PWH was 3.2 (95% CI 2.5-4). A higher CD4/CD8 ratio was associated with a higher GMT (R = 0.27, p = 0.039). MVC-COV1901 has shown robust safety but elicited weaker immune responses in PWH. CONCLUSIONS Further investigations may be needed to determine whether PWH require distinct immunization strategies with improved immunogenicity. The main study is registered at ClinicalTrials.gov (NCT04695652).
Collapse
|
44
|
Notario GR, Kwak K. Increased B Cell Understanding Puts Improved Vaccine Platforms Just Over the Horizon. Immune Netw 2022; 22:e47. [PMID: 36627934 PMCID: PMC9807965 DOI: 10.4110/in.2022.22.e47] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 10/27/2022] [Accepted: 11/08/2022] [Indexed: 12/30/2022] Open
Abstract
In the face of an endlessly expanding repertoire of Ags, vaccines are constantly being tested, each more effective than the last. As viruses and other pathogens evolve to become more infectious, the need for efficient and effective vaccines grows daily, which is especially obvious in an era that is still attempting to remove itself from the clutches of the severe acute respiratory syndrome coronavirus 2, the cause of coronavirus pandemic. To continue evolving alongside these pathogens, it is proving increasingly essential to consider one of the main effector cells of the immune system. As one of the chief orchestrators of the humoral immune response, the B cell and other lymphocytes are essential to not only achieving immunity, but also maintaining it, which is the vital objective of every vaccine.
Collapse
Affiliation(s)
- Geneva Rose Notario
- Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea.,Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Kihyuck Kwak
- Institute for Immunology and Immunological Diseases, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea.,Department of Microbiology and Immunology, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
45
|
Selective Silencing of Disease-Associated B Lymphocytes from Hashimoto's Thyroiditis Patients by Chimeric Protein Molecules. Int J Mol Sci 2022; 23:ijms232315083. [PMID: 36499407 PMCID: PMC9738561 DOI: 10.3390/ijms232315083] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/03/2022] Open
Abstract
Hashimoto's thyroiditis is one of the most common endocrine disorders, affecting up to 20% of the adult population. No treatment or prevention exists except hormonal substitution for hypothyroidism. We hypothesize that it may be possible to selectively suppress anti-thyroglobulin (Tg) IgG antibody-producing B lymphocytes from HT patients by a chimeric protein molecule containing a monoclonal antibody specific for the human inhibitory receptor CR1, coupled to peptide epitopes derived from Tg protein. We expect that this treatment will down-regulate B-cell autoreactivity by delivering a strong inhibitory signal. Three peptides-two epitope-predicted ones derived from Tg and another irrelevant peptide-were synthesized and then coupled with monoclonal anti-human CR1 antibody to construct three chimeric molecules. The binding to CD35 on human B cells and the effects of the chimeric constructs on PBMC and TMC from patients with HT were tested using flow cytometry, ELISpot assay, and immunoenzyme methods. We found that after the chemical conjugation, all chimeras retained their receptor-binding capacity, and the Tg epitopes could be recognized by anti-Tg autoantibodies in the patients' sera. This treatment downregulated B-cell autoreactivity and cell proliferation, inhibited Tg-specific B-cell differentiation to plasmablasts and promoted apoptosis to the targeted cells. The treatment of PBMCs from HT patients with Tg-epitope-carrying chimeric molecules affects the activity of Tg-specific autoreactive B lymphocytes, delivering to them a strong suppressive signal.
Collapse
|
46
|
Kwiatkowska KM, Mkindi CG, Nielsen CM. Human lymphoid tissue sampling for vaccinology. Front Immunol 2022; 13:1045529. [PMID: 36466924 PMCID: PMC9714609 DOI: 10.3389/fimmu.2022.1045529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 10/31/2022] [Indexed: 02/15/2024] Open
Abstract
Long-lived plasma cells (LLPCs) - largely resident in the bone marrow - secrete antibody over months and years, thus maintaining serum antibody concentrations relevant for vaccine-mediated immunity. Little is known regarding factors that can modulate the induction of human LLPC responses in draining lymph node germinal centres, or those that maintain LLPCs in bone marrow niches following vaccination. Here, we review human and non-human primate vaccination studies which incorporate draining lymph node and/or bone marrow aspirate sampling. We emphasise the key contributions these samples can make to improve our understanding of LLPC immunology and guide rational vaccine development. Specifically, we highlight findings related to the impact of vaccine dosing regimens, adjuvant/vaccine platform selection, duration of germinal centre reactions in draining lymph nodes and relevance for timing of tissue sampling, and heterogeneity in bone marrow plasma cell populations. Much of this work has come from recent studies with SARS-CoV-2 vaccine candidates or, with respect to the non-human primate work, HIV vaccine development.
Collapse
Affiliation(s)
| | - Catherine G. Mkindi
- Department of Intervention and Clinical Trials, Ifakara Health Institute, Bagamoyo, Tanzania
| | - Carolyn M. Nielsen
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
47
|
Cheng RYH, Hung KL, Zhang T, Stoffers CM, Ott AR, Suchland ER, Camp ND, Khan IF, Singh S, Yang YJ, Rawlings DJ, James RG. Ex vivo engineered human plasma cells exhibit robust protein secretion and long-term engraftment in vivo. Nat Commun 2022; 13:6110. [PMID: 36245034 PMCID: PMC9573882 DOI: 10.1038/s41467-022-33787-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 10/04/2022] [Indexed: 12/24/2022] Open
Abstract
Due to their unique longevity and capacity to secrete high levels of protein, plasma B cells have the potential to be used as a cell therapy for protein replacement. Here, we show that ex vivo engineered human plasma cells exhibit single-cell RNA profiles, scanning electron micrograph ultrastructural features, and in vivo homing capacity of long-lived plasma cells. After transferring human plasma cells to immunodeficient mice in the presence of the human cytokines BAFF and IL-6, we observe increases in retention of plasma cells in the bone marrow, with engraftment exceeding a year. The most profound in vivo effects of human IL-6 are observed within 20 days of transfer and could be explained by decreased apoptosis in newly differentiated plasma cells. Collectively, these results show that ex vivo engineered and differentiated human plasma cells have the potential for long-lived in vivo protein secretion, which can be modeled in small animals.
Collapse
Affiliation(s)
- Rene Yu-Hong Cheng
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
- Molecular Engineering and Science Institute, University of Washington, Seattle, WA, 98195, USA
| | - King L Hung
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Tingting Zhang
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
| | - Claire M Stoffers
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
| | - Andee R Ott
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
| | - Emmaline R Suchland
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
| | - Nathan D Camp
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
| | - Iram F Khan
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
| | - Swati Singh
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA
| | - Ying-Jen Yang
- Department of Applied Mathematics, University of Washington, Seattle, WA, 98195, USA
| | - David J Rawlings
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA.
- Departments of Pediatrics, University of Washington, Seattle, WA, 98195, USA.
- Department of Immunology, University of Washington, Seattle, WA, 98195, USA.
| | - Richard G James
- Center of immunotherapy and Immunity, Seattle Children Research Institute, Seattle, WA, 98101, USA.
- Molecular Engineering and Science Institute, University of Washington, Seattle, WA, 98195, USA.
- Departments of Pediatrics, University of Washington, Seattle, WA, 98195, USA.
- Department of pharmacology, University of Washington, Seattle, WA, 98195, USA.
- Brotman-Baty Institute for Precision Medicine, Seattle, WA, 98195, USA.
| |
Collapse
|
48
|
Nishio A, Hasan S, Park H, Park N, Salas JH, Salinas E, Kardava L, Juneau P, Frumento N, Massaccesi G, Moir S, Bailey JR, Grakoui A, Ghany MG, Rehermann B. Serum neutralization activity declines but memory B cells persist after cure of chronic hepatitis C. Nat Commun 2022; 13:5446. [PMID: 36114169 PMCID: PMC9481596 DOI: 10.1038/s41467-022-33035-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/30/2022] [Indexed: 11/09/2022] Open
Abstract
The increasing incidence of hepatitis C virus (HCV) infections underscores the need for an effective vaccine. Successful vaccines to other viruses generally depend on a long-lasting humoral response. However, data on the half-life of HCV-specific responses are lacking. Here we study archived sera and mononuclear cells that were prospectively collected up to 18 years after cure of chronic HCV infection to determine the role of HCV antigen in maintaining neutralizing antibody and B cell responses. We show that HCV-neutralizing activity decreases rapidly in potency and breadth after curative treatment. In contrast, HCV-specific memory B cells persist, and display a restored resting phenotype, normalized chemokine receptor expression and preserved ability to differentiate into antibody-secreting cells. The short half-life of HCV-neutralizing activity is consistent with a lack of long-lived plasma cells. The persistence of HCV-specific memory B cells and the reduced inflammation after cure provide an opportunity for vaccination to induce protective immunity against re-infection.
Collapse
Affiliation(s)
- Akira Nishio
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892, USA
| | - Sharika Hasan
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892, USA
| | - Heiyoung Park
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892, USA
| | - Nana Park
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892, USA
| | - Jordan H Salas
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Eduardo Salinas
- Division of Infectious Diseases, Emory Vaccine Center, Division of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Emory National Primate Research Center, Emory Vaccine Center, Atlanta, GA, 30329, USA
| | - Lela Kardava
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892, USA
| | - Paul Juneau
- Division of Data Services, NIH Library, Office of Research Services, National Institutes of Health, Bethesda, MD, USA
- Contractor- Zimmerman Associates, Inc, Fairfax, VA, USA
| | - Nicole Frumento
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Guido Massaccesi
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Susan Moir
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892, USA
| | - Justin R Bailey
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Arash Grakoui
- Division of Infectious Diseases, Emory Vaccine Center, Division of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA, 30322, USA
- Emory National Primate Research Center, Emory Vaccine Center, Atlanta, GA, 30329, USA
| | - Marc G Ghany
- Clinical Research Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892, USA
| | - Barbara Rehermann
- Immunology Section, Liver Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, DHHS, Bethesda, MD, 20892, USA.
| |
Collapse
|
49
|
Lelievre JD, Bauer J. What are the prospects for durable immune control? Infect Dis Now 2022; 52:S4-S6. [PMID: 36113766 PMCID: PMC9472462 DOI: 10.1016/j.idnow.2022.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
During the SARS CoV-2 primary infection, the neutralizing antibodies focused against the spike (S) glycoproteins are responsible for blockage of virus-host cell interaction. The cellular response mediated by CD4+ and CD8+ T-cells is responsible for control of viremia. Immune memory against SARS-CoV-2 depends on virus type, replication kinetics and route of penetration. The formation and persistence of germinal centers are critical for the generation of affinity-matured plasma cells and memory B cells capable of mediating durable immunity. They can persist up to 30 weeks after vaccination and several months after infection. Heterogeneity in the longevity of the vaccination-induced GC response is significant.
Collapse
Affiliation(s)
- J D Lelievre
- Immunologie clinique et maladies infectieuses, Hôpital Henri-Mondor, 94000 Créteil, France
| | - J Bauer
- Service Universitaire des Maladies Infectieuses et du Voyageurs, CH Dron, 59200 Tourcoing, France.
| |
Collapse
|
50
|
Nguyen DC, Lamothe PA, Woodruff MC, Saini AS, Faliti CE, Sanz I, Lee FE. COVID-19 and plasma cells: Is there long-lived protection? Immunol Rev 2022; 309:40-63. [PMID: 35801537 PMCID: PMC9350162 DOI: 10.1111/imr.13115] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Infection with SARS-CoV-2, the etiology of the ongoing COVID-19 pandemic, has resulted in over 450 million cases with more than 6 million deaths worldwide, causing global disruptions since early 2020. Memory B cells and durable antibody protection from long-lived plasma cells (LLPC) are the mainstay of most effective vaccines. However, ending the pandemic has been hampered by the lack of long-lived immunity after infection or vaccination. Although immunizations offer protection from severe disease and hospitalization, breakthrough infections still occur, most likely due to new mutant viruses and the overall decline of neutralizing antibodies after 6 months. Here, we review the current knowledge of B cells, from extrafollicular to memory populations, with a focus on distinct plasma cell subsets, such as early-minted blood antibody-secreting cells and the bone marrow LLPC, and how these humoral compartments contribute to protection after SARS-CoV-2 infection and immunization.
Collapse
Affiliation(s)
- Doan C. Nguyen
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
| | - Pedro A. Lamothe
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
| | - Matthew C. Woodruff
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Emory Autoimmunity Center of ExcellenceEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Ankur S. Saini
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Emory Autoimmunity Center of ExcellenceEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Caterina E. Faliti
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Ignacio Sanz
- Division of Rheumatology, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Emory Autoimmunity Center of ExcellenceEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| | - Frances Eun‐Hyung Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of MedicineEmory UniversityAtlantaGeorgiaUSA
- Lowance Center for Human ImmunologyEmory UniversityAtlantaGeorgiaUSA
| |
Collapse
|