1
|
Leung WY, Wu HHL, Nair B, Woywodt A, Ponnusamy A. Unexpected seronegative response in relapsed PLA2R-associated membranous nephropathy. Clin Kidney J 2024; 17:sfae189. [PMID: 39056067 PMCID: PMC11272037 DOI: 10.1093/ckj/sfae189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Indexed: 07/28/2024] Open
Affiliation(s)
- Wing Yin Leung
- Department of Renal Medicine, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, UK
| | - Henry H L Wu
- Renal Research, Kolling Institute of Medical Research, Royal North Shore Hospital & The University of Sydney, Sydney, NSW, Australia
| | - Beena Nair
- Department of Pathology, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, UK
| | - Alexander Woywodt
- Department of Renal Medicine, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, UK
- Faculty of Biology, Medicine & Health, The University of Manchester, Manchester, UK
| | - Arvind Ponnusamy
- Department of Renal Medicine, Lancashire Teaching Hospitals NHS Foundation Trust, Preston, UK
- Faculty of Biology, Medicine & Health, The University of Manchester, Manchester, UK
| |
Collapse
|
2
|
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: 0] [Impact Index Per Article: 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
|
3
|
Bass LE, Bonami RH. Factors Governing B Cell Recognition of Autoantigen and Function in Type 1 Diabetes. Antibodies (Basel) 2024; 13:27. [PMID: 38651407 PMCID: PMC11036271 DOI: 10.3390/antib13020027] [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: 02/21/2024] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Islet autoantibodies predict type 1 diabetes (T1D) but can be transient in murine and human T1D and are not thought to be directly pathogenic. Rather, these autoantibodies signal B cell activity as antigen-presenting cells (APCs) that present islet autoantigen to diabetogenic T cells to promote T1D pathogenesis. Disrupting B cell APC function prevents T1D in mouse models and has shown promise in clinical trials. Autoantigen-specific B cells thus hold potential as sophisticated T1D biomarkers and therapeutic targets. B cell receptor (BCR) somatic hypermutation is a mechanism by which B cells increase affinity for islet autoantigen. High-affinity B and T cell responses are selected in protective immune responses, but immune tolerance mechanisms are known to censor highly autoreactive clones in autoimmunity, including T1D. Thus, different selection rules often apply to autoimmune disease settings (as opposed to protective host immunity), where different autoantigen affinity ceilings are tolerated based on variations in host genetics and environment. This review will explore what is currently known regarding B cell signaling, selection, and interaction with T cells to promote T1D pathogenesis.
Collapse
Affiliation(s)
- Lindsay E. Bass
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
| | - Rachel H. Bonami
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA;
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
4
|
Wang L, Li Q, Deng C, Liu Z, Wang F, Li S, Dong L, Jiang J. Preclinical Pharmacokinetics and Pharmacology Study of RC98: A Programmed Cell Death Ligand 1 Monoclonal Antibody in Cynomolgus Monkeys. Curr Pharm Des 2024; 30:1240-1246. [PMID: 38623974 DOI: 10.2174/0113816128248929230920071937] [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: 02/14/2023] [Revised: 06/27/2023] [Accepted: 08/02/2023] [Indexed: 04/17/2024]
Abstract
INTRODUCTION RC98 is the monoclonal antibody against Programmed Cell Death Ligand 1 (PD-L1). Relevant reports have confirmed that the influence of PD-L1 expressed by tumor cells on antitumor CD8+ T cell responses is well characterized, but the impact of PD-L1 expressed by immune cells has not been well defined. OBJECTIVE This study aimed to design a Pharmacokinetics/Pharmacology (PK/PD) study of RC98 in normal cynomolgus monkeys to research the effect on the immune system. METHODS RC98 and vehicle were administered to cynomolgus monkeys at 15 mg/kg via intravenous infusion once a week for 4 weeks to evaluate the relationship between PK and PD. The pharmacodynamic activity was measured by the PD-L1 receptor occupancy (RO) in CD3+ T cells, A T-cell-dependent antibody response (TDAR), and the concentration of soluble PD-L1. RESULTS The pharmacokinetic result showed that the exposure from the last administration was lower than that of the first administration, probably due to immunogenicity production. There was a strong correlation between systemic exposure and RO in CD3+ T cells but decreased RO levels after the last dose, which indirectly reflected the activation of T cells. The keyhole limpet hemocyanin (KLH)-induced TDAR in the RC98 group was higher than in the vehicle group. The concentration of soluble PD-L1 had increased feedback with RC98, and the concentration of soluble PD-L1 was maintained at a higher level after multiple doses than before dosing. CONCLUSION These data indicate that the immune system was clearly activated. In addition, the non-clinical data could provide a basis for its efficacy evaluation in clinical trials.
Collapse
Affiliation(s)
- Ling Wang
- Non-clinical Research Department, RemeGen, Ltd., Yantai 264000, Shandong, China
- Rongchang Industry College, Yantai 264003, Shandong, China
| | - Qiaoning Li
- Non-clinical Research Department, RemeGen, Ltd., Yantai 264000, Shandong, China
- Rongchang Industry College, Yantai 264003, Shandong, China
| | - Chenglian Deng
- Immunogenicity Department, United-Power Pharma Tech Co. Ltd, Beijing 102206, China
| | - Zhihao Liu
- Non-clinical Research Department, RemeGen, Ltd., Yantai 264000, Shandong, China
- Rongchang Industry College, Yantai 264003, Shandong, China
| | - Fang Wang
- Immunogenicity Department, United-Power Pharma Tech Co. Ltd, Beijing 102206, China
| | - Shenjun Li
- Non-clinical Research Department, RemeGen, Ltd., Yantai 264000, Shandong, China
- Rongchang Industry College, Yantai 264003, Shandong, China
| | - Lihou Dong
- Immunogenicity Department, United-Power Pharma Tech Co. Ltd, Beijing 102206, China
| | - Jing Jiang
- Rongchang Industry College, Yantai 264003, Shandong, China
- Department of Pharmacology, Binzhou Medical University, Yantai 264003, Shandong, China
| |
Collapse
|
5
|
Levin D, Osman MS, Durand C, Kim H, Hemmati I, Jamani K, Howlett JG, Johannson KA, Weatherald J, Woo M, Lee J, Storek J. Hematopoietic Cell Transplantation for Systemic Sclerosis-A Review. Cells 2022; 11:3912. [PMID: 36497169 PMCID: PMC9739132 DOI: 10.3390/cells11233912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/18/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune, multi-organ, connective tissue disease associated with significant morbidity and mortality. Conventional immunosuppressive therapies demonstrate limited efficacy. Autologous hematopoietic stem cell transplantation (HCT) is more efficacious but carries associated risks, including treatment-related mortality. Here, we review HCT as a treatment for SSc, its efficacy and toxicity in comparison to conventional therapies, and the proposed mechanisms of action. Furthermore, we discuss the importance of and recent developments in patient selection. Finally, we highlight the knowledge gaps and future work required to further improve patient outcomes.
Collapse
Affiliation(s)
- Daniel Levin
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Mohammed S. Osman
- Faculty of Medicine, University of Alberta, Edmonton, AB T6G 2R7, Canada
| | - Caylib Durand
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Hyein Kim
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Iman Hemmati
- Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Kareem Jamani
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jonathan G. Howlett
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Kerri A. Johannson
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jason Weatherald
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Faculty of Medicine, University of Alberta, Edmonton, AB T6G 2R7, Canada
| | - Matthew Woo
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jason Lee
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jan Storek
- Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|
6
|
DeWolf S, Laracy JC, Perales MA, Kamboj M, van den Brink MRM, Vardhana S. SARS-CoV-2 in immunocompromised individuals. Immunity 2022; 55:1779-1798. [PMID: 36182669 PMCID: PMC9468314 DOI: 10.1016/j.immuni.2022.09.006] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/24/2022] [Accepted: 09/08/2022] [Indexed: 12/15/2022]
Abstract
Immunocompromised individuals and particularly those with hematologic malignancies are at increased risk for SARS-CoV-2-associated morbidity and mortality due to immunologic deficits that limit prevention, treatment, and clearance of the virus. Understanding the natural history of viral infections in people with impaired immunity due to underlying conditions, immunosuppressive therapy, or a combination thereof has emerged as a critical area of investigation during the COVID-19 pandemic. Studies focused on these individuals have provided key insights into aspects of innate and adaptive immunity underlying both the antiviral immune response and excess inflammation in the setting of COVID-19. This review presents what is known about distinct states of immunologic vulnerability to SARS-CoV-2 and how this information can be harnessed to improve prevention and treatment strategies for immunologically high-risk populations.
Collapse
Affiliation(s)
- Susan DeWolf
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Justin C Laracy
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | - Mini Kamboj
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marcel R M van den Brink
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA; Department of Immunology, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Santosha Vardhana
- Weill Cornell Medical College, New York, NY, USA; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
7
|
Venturini C, Petrovic Fabijan A, Fajardo Lubian A, Barbirz S, Iredell J. Biological foundations of successful bacteriophage therapy. EMBO Mol Med 2022; 14:e12435. [PMID: 35620963 PMCID: PMC9260219 DOI: 10.15252/emmm.202012435] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/13/2022] [Accepted: 04/19/2022] [Indexed: 12/20/2022] Open
Abstract
Bacteriophages (phages) are selective viral predators of bacteria. Abundant and ubiquitous in nature, phages can be used to treat bacterial infections (phage therapy), including refractory infections and those resistant to antibiotics. However, despite an abundance of anecdotal evidence of efficacy, significant hurdles remain before routine implementation of phage therapy into medical practice, including a dearth of robust clinical trial data. Phage-bacterium interactions are complex and diverse, characterized by co-evolution trajectories that are significantly influenced by the environments in which they occur (mammalian body sites, water, soil, etc.). An understanding of the molecular mechanisms underpinning these dynamics is essential for successful clinical translation. This review aims to cover key aspects of bacterium-phage interactions that affect bacterial killing by describing the most relevant published literature and detailing the current knowledge gaps most likely to influence therapeutic success.
Collapse
Affiliation(s)
- Carola Venturini
- Centre for Infectious Diseases and MicrobiologyWestmead Institute for Medical ResearchWestmeadNSWAustralia
- Faculty of ScienceSydney School of Veterinary ScienceThe University of SydneySydneyNSWAustralia
| | - Aleksandra Petrovic Fabijan
- Centre for Infectious Diseases and MicrobiologyWestmead Institute for Medical ResearchWestmeadNSWAustralia
- Faculty of Health and MedicineSchool of MedicineSydney Medical SchoolThe University of SydneySydneyNSWAustralia
| | - Alicia Fajardo Lubian
- Centre for Infectious Diseases and MicrobiologyWestmead Institute for Medical ResearchWestmeadNSWAustralia
- Faculty of Health and MedicineSchool of MedicineSydney Medical SchoolThe University of SydneySydneyNSWAustralia
| | - Stefanie Barbirz
- Department of MedicineScience FacultyMSB Medical School BerlinBerlinGermany
| | - Jonathan Iredell
- Centre for Infectious Diseases and MicrobiologyWestmead Institute for Medical ResearchWestmeadNSWAustralia
- Faculty of Health and MedicineSchool of MedicineSydney Medical SchoolThe University of SydneySydneyNSWAustralia
- Westmead HospitalWestern Sydney Local Health DistrictWestmeadNSWAustralia
| |
Collapse
|
8
|
Martire B, Ottaviano G, Sangerardi M, Sgrulletti M, Chini L, Dellepiane RM, Montin D, Rizzo C, Pignata C, Marseglia GL, Moschese V. Vaccinations in Children and Adolescents Treated With Immune-Modifying Biologics: Update and Current Developments. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:1485-1496. [PMID: 35085809 DOI: 10.1016/j.jaip.2022.01.007] [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: 07/02/2021] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 06/14/2023]
Abstract
Treatment with immune-modifying biologics has positively impacted disease control and quality of life in many patients with immune-mediated disorders. However, the higher susceptibility to common and opportunistic pathogens is of concern. Thus, immunization strategies to control vaccine-preventable diseases represent a critical issue in this population. However, limited data exist on the safety, immunogenicity, and efficacy of available vaccines in patients on biologics, particularly in children. Here, according to published literature and real-life experience and practice, we report the interim indications of the Italian Society of Pediatric Allergology and Immunology (SIAIP) Vaccine Committee and of the Italian Primary Immunodeficiency Network (IPINet) Centers on immunization of children and adolescents receiving biologics. Our aim is to provide a practical guidance for the clinician to ensure optimal protection for patients and the community.
Collapse
Affiliation(s)
- Baldassarre Martire
- Pediatrics and Neonatology Unit, Maternal-Infant Department, Monsignor A. R. Dimiccoli Hospital, Barletta, Italy.
| | - Giorgio Ottaviano
- Molecular and Cellular Immunology Unit, Great Ormond Street Institute of Child Health, University College of London, London, UK
| | - Maria Sangerardi
- Department of Pediatrics and Emergency, Pediatric Hospital, Policlinico - University of Bari, Bari, Italy
| | - Mayla Sgrulletti
- Pediatric Immunopathology and Allergology Unit, University of Rome, Tor Vergata, Policlinico Tor Vergata, Rome, Italy
| | - Loredana Chini
- Pediatric Immunopathology and Allergology Unit, University of Rome, Tor Vergata, Policlinico Tor Vergata, Rome, Italy
| | - Rosa Maria Dellepiane
- Pediatric Intermediate Care Unit, Scientific Institute for Research, Hospitalization and Healthcare Foundation (IRCSS); Ca Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Davide Montin
- Department of Public Health and Pediatrics, Regina Margherita Children Hospital, University of Turin, Turin, Italy
| | - Caterina Rizzo
- Innovation and Clinical Pathways Unit, Bambino Gesù Children Hospital, IRCCS, Rome, Italy
| | - Claudio Pignata
- Department of Translational Medical Sciences-Section of Pediatrics, Federico II University of Naples, Naples, Italy
| | - Gian Luigi Marseglia
- Department of Pediatrics, IRCCS Foundation, Policlinico San Matteo, University of Pavia, Pavia, Italy
| | - Viviana Moschese
- Pediatric Immunopathology and Allergology Unit, University of Rome, Tor Vergata, Policlinico Tor Vergata, Rome, Italy
| |
Collapse
|
9
|
Otani IM, Lehman HK, Jongco AM, Tsao LR, Azar AE, Tarrant TK, Engel E, Walter JE, Truong TQ, Khan DA, Ballow M, Cunningham-Rundles C, Lu H, Kwan M, Barmettler S. Practical guidance for the diagnosis and management of secondary hypogammaglobulinemia: A Work Group Report of the AAAAI Primary Immunodeficiency and Altered Immune Response Committees. J Allergy Clin Immunol 2022; 149:1525-1560. [PMID: 35176351 DOI: 10.1016/j.jaci.2022.01.025] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 12/31/2021] [Accepted: 01/21/2022] [Indexed: 11/17/2022]
Abstract
Secondary hypogammaglobulinemia (SHG) is characterized by reduced immunoglobulin levels due to acquired causes of decreased antibody production or increased antibody loss. Clarification regarding whether the hypogammaglobulinemia is secondary or primary is important because this has implications for evaluation and management. Prior receipt of immunosuppressive medications and/or presence of conditions associated with SHG development, including protein loss syndromes, are histories that raise suspicion for SHG. In patients with these histories, a thorough investigation of potential etiologies of SHG reviewed in this report is needed to devise an effective treatment plan focused on removal of iatrogenic causes (eg, discontinuation of an offending drug) or treatment of the underlying condition (eg, management of nephrotic syndrome). When iatrogenic causes cannot be removed or underlying conditions cannot be reversed, therapeutic options are not clearly delineated but include heightened monitoring for clinical infections, supportive antimicrobials, and in some cases, immunoglobulin replacement therapy. This report serves to summarize the existing literature regarding immunosuppressive medications and populations (autoimmune, neurologic, hematologic/oncologic, pulmonary, posttransplant, protein-losing) associated with SHG and highlights key areas for future investigation.
Collapse
Affiliation(s)
- Iris M Otani
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, UCSF Medical Center, San Francisco, Calif.
| | - Heather K Lehman
- Division of Allergy, Immunology, and Rheumatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| | - Artemio M Jongco
- Division of Allergy and Immunology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY
| | - Lulu R Tsao
- Division of Pulmonary, Critical Care, Allergy and Sleep Medicine, UCSF Medical Center, San Francisco, Calif
| | - Antoine E Azar
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore
| | - Teresa K Tarrant
- Division of Rheumatology and Immunology, Duke University, Durham, NC
| | - Elissa Engel
- Division of Hematology and Oncology, Cincinnati Children's Hospital, Cincinnati, Ohio
| | - Jolan E Walter
- Division of Allergy and Immunology, Johns Hopkins All Children's Hospital, St Petersburg, Fla; Division of Allergy and Immunology, Morsani College of Medicine, University of South Florida, Tampa; Division of Allergy and Immunology, Massachusetts General Hospital for Children, Boston
| | - Tho Q Truong
- Divisions of Rheumatology, Allergy and Clinical Immunology, National Jewish Health, Denver
| | - David A Khan
- Division of Allergy and Immunology, University of Texas Southwestern Medical Center, Dallas
| | - Mark Ballow
- Division of Allergy and Immunology, Morsani College of Medicine, Johns Hopkins All Children's Hospital, St Petersburg
| | | | - Huifang Lu
- Department of General Internal Medicine, Section of Rheumatology and Clinical Immunology, The University of Texas MD Anderson Cancer Center, Houston
| | - Mildred Kwan
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, University of North Carolina School of Medicine, Chapel Hill
| | - Sara Barmettler
- Allergy and Immunology, Massachusetts General Hospital, Boston.
| |
Collapse
|
10
|
Kampouri E, Walti CS, Gauthier J, Hill JA. Managing Hypogammaglobulinemia in Patients Treated with CAR-T-cell Therapy: Key Points for Clinicians. Expert Rev Hematol 2022; 15:305-320. [PMID: 35385358 DOI: 10.1080/17474086.2022.2063833] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION The unprecedented success of chimeric antigen receptor (CAR)-T-cell therapy in the management of B-cell malignancies comes with a price of specific side effects. Healthy B-cell depletion is an anticipated 'on-target' 'off-tumor' side effect and can contribute to severe and prolonged hypogammaglobulinemia. Evidence-based guidelines for the use of immunoglobulin replacement therapy (IGRT) for infection prevention are lacking in this population. AREAS COVERED This article reviews the mechanisms and epidemiology of hypogammaglobulinemia and antibody deficiency, association with infections, and strategies to address these issues in CD19- and BCMA-CAR-T-cell recipients. EXPERT OPINION CD19 and BCMA CAR-T-cell therapy result in unique immune deficits due to depletion of specific B-lineage cells and may require different infection prevention strategies. Hypogammaglobulinemia before and after CAR-T-cell therapy is frequent, but data on the efficacy and cost-effectiveness of IGRT are lacking. Monthly IGRT should be prioritized for patients with severe or recurrent bacterial infections. IGRT may be more broadly necessary to prevent infections in BCMA-CAR-T-cell recipients and children with severe hypogammaglobulinemia irrespective of infection history. Vaccinations are indicated to augment humoral immunity and can be immunogenic despite cytopenias; re-vaccination(s) may be required. Controlled trials are needed to better understand the role of IGRT and vaccines in this population.
Collapse
Affiliation(s)
- Eleftheria Kampouri
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Infectious Diseases Service, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Carla S Walti
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Infectious Disease and Hospital Epidemiology Division, University Hospital Basel, Basel, Switzerland
| | - Jordan Gauthier
- Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Joshua A Hill
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
11
|
Galati A, McElrath T, Bove R. Use of B-Cell–Depleting Therapy in Women of Childbearing Potential With Multiple Sclerosis and Neuromyelitis Optica Spectrum Disorder. Neurol Clin Pract 2022; 12:154-163. [PMID: 35733945 PMCID: PMC9208398 DOI: 10.1212/cpj.0000000000001147] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Accepted: 12/16/2021] [Indexed: 11/17/2022]
Abstract
Purpose of Review There is considerable heterogeneity in the use of B-cell depletion in women of childbearing age, likely driven at least in part by the discrepancy between the product labels and what is known about the physiology of IgG1, including breastmilk and placental transfer. Recent Findings We provide practical considerations on the use of this medication class in women of childbearing potential. We discuss prepregnancy planning including vaccinations, safety of B-cell depletion during pregnancy, and postpartum considerations including breastfeeding. Summary B-cell–depleting monoclonal antibodies have shown to be effective for prepregnancy and postpartum prevention of inflammatory activity in MS and neuromyelitis optica spectrum disorder. B-cell–depleting therapies are large IgG1 monoclonal antibodies, which have minimal transfer across the placenta and into breastmilk. Consideration of risks and benefits of these therapies should be considered in counseling women planning pregnancy and postpartum.
Collapse
Affiliation(s)
- Alexandra Galati
- Division of Neuroimmunology and Glial Biology (A.G., R.B.), Department of Neurology, University of California, San Francisco, UCSF Weill Institute for the Neurosciences, San Francisco, CA; and Brigham & Women's Hospital, Harvard Medical School, Boston, MA
| | - Thomas McElrath
- Division of Neuroimmunology and Glial Biology (A.G., R.B.), Department of Neurology, University of California, San Francisco, UCSF Weill Institute for the Neurosciences, San Francisco, CA; and Brigham & Women's Hospital, Harvard Medical School, Boston, MA
| | - Riley Bove
- Division of Neuroimmunology and Glial Biology (A.G., R.B.), Department of Neurology, University of California, San Francisco, UCSF Weill Institute for the Neurosciences, San Francisco, CA; and Brigham & Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
12
|
Peeters M, Verbruggen L, Teuwen L, Vanhoutte G, Vande Kerckhove S, Peeters B, Raats S, Van der Massen I, De Keersmaecker S, Debie Y, Huizing M, Pannus P, Neven K, Ariën KK, Martens GA, Van Den Bulcke M, Roelant E, Desombere I, Anguille S, Goossens M, Vandamme T, van Dam P. Reduced humoral immune response after BNT162b2 coronavirus disease 2019 messenger RNA vaccination in cancer patients under antineoplastic treatment. ESMO Open 2021; 6:100274. [PMID: 34597941 PMCID: PMC8423808 DOI: 10.1016/j.esmoop.2021.100274] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/16/2021] [Accepted: 09/01/2021] [Indexed: 12/26/2022] Open
Abstract
Background Cancer patients are at a higher risk of developing severe coronavirus disease 2019 (COVID-19). However, the safety and efficacy of COVID-19 vaccination in cancer patients undergoing treatment remain unclear. Patients and methods In this interventional prospective multicohort study, priming and booster doses of the BNT162b2 COVID-19 vaccine were administered 21 days apart to solid tumor patients receiving chemotherapy, immunotherapy, targeted or hormonal therapy, and patients with a hematologic malignancy receiving rituximab or after allogeneic hematopoietic stem cell transplantation. Vaccine safety and efficacy (until 3 months post-booster) were assessed. Anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor-binding domain (RBD) antibody levels were followed over time (until 28 days after the booster) and in vitro SARS-CoV-2 50% neutralization titers (NT50) toward the wild-type Wuhan strain were analyzed 28 days after the booster. Results Local and systemic adverse events (AEs) were mostly mild to moderate (only 1%-3% of patients experienced severe AEs). Local, but not systemic, AEs occurred more frequently after the booster dose. Twenty-eight days after the booster vaccination of 197 cancer patients, RBD-binding antibody titers and NT50 were lower in the chemotherapy group {234.05 IU/ml [95% confidence interval (CI) 122.10-448.66] and 24.54 (95% CI 14.50-41.52), respectively} compared with healthy individuals [1844.93 IU/ml (95% CI 1383.57-2460.14) and 122.63 (95% CI 76.85-195.67), respectively], irrespective of timing of vaccination during chemotherapy cycles. Extremely low antibody responses were seen in hematology patients receiving rituximab; only two patients had RBD-binding antibody titers necessary for 50% protection against symptomatic SARS-CoV-2 infection (<200 IU/ml) and only one had NT50 above the limit of detection. During the study period, five cancer patients tested positive for SARS-CoV-2 infection, including a case of severe COVID-19 in a patient receiving rituximab, resulting in a 2-week hospital admission. Conclusion The BNT162b2 vaccine is well-tolerated in cancer patients under active treatment. However, the antibody response of immunized cancer patients was delayed and diminished, mainly in patients receiving chemotherapy or rituximab, resulting in breakthrough infections. The BNT162b2 vaccine is well-tolerated in cancer patients, including patients under immunotherapy. Full BNT162b2 vaccination results in a blunted humoral immune response in cancer patients under active treatment. The humoral immune response after BNT162b2 vaccination varies between different antineoplastic treatments. Two doses of BNT162b2 vaccination may insufficiently protect patients receiving chemotherapy or rituximab against SARS-CoV-2.
Collapse
Affiliation(s)
- M Peeters
- Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp and Antwerp University Hospital, Edegem, Belgium.
| | - L Verbruggen
- Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
| | - L Teuwen
- Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
| | - G Vanhoutte
- Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
| | - S Vande Kerckhove
- SD Infectious Diseases in Humans, Service Immune response, Sciensano, Brussels, Belgium
| | - B Peeters
- Department of Laboratory Medicine, Antwerp University Hospital, Edegem, Belgium
| | - S Raats
- Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
| | - I Van der Massen
- Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
| | - S De Keersmaecker
- Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
| | - Y Debie
- Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
| | | | - P Pannus
- SD Epidemiology and Public Health, Sciensano, Brussels, Belgium
| | - K Neven
- SD Epidemiology and Public Health, Sciensano, Brussels, Belgium
| | - K K Ariën
- Virology Unit, Institute of Tropical Medicine Antwerp, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Edegem, Belgium
| | - G A Martens
- Department of Laboratory Medicine, AZ Delta General Hospital, Roeselare, Belgium
| | | | - E Roelant
- Clinical Trial Center (CTC), CRC Antwerp, Antwerp University Hospital, University of Antwerp, Edegem, Belgium; StatUa, Center for Statistics, University of Antwerp, Antwerp, Belgium
| | - I Desombere
- SD Infectious Diseases in Humans, Service Immune response, Sciensano, Brussels, Belgium
| | - S Anguille
- Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium
| | - M Goossens
- SD Epidemiology and Public Health, Sciensano, Brussels, Belgium
| | - T Vandamme
- Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp and Antwerp University Hospital, Edegem, Belgium
| | - P van Dam
- Multidisciplinary Oncologic Centre Antwerp (MOCA), Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp and Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
13
|
Diks AM, Overduin LA, van Leenen LD, Slobbe L, Jolink H, Visser LG, van Dongen JJM, Berkowska MA. B-Cell Immunophenotyping to Predict Vaccination Outcome in the Immunocompromised - A Systematic Review. Front Immunol 2021; 12:690328. [PMID: 34557188 PMCID: PMC8452967 DOI: 10.3389/fimmu.2021.690328] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 08/13/2021] [Indexed: 11/13/2022] Open
Abstract
Vaccination is the most effective measure to prevent infections in the general population. Its efficiency strongly depends on the function and composition of the immune system. If the immune system lacks critical components, patients will not be fully protected despite a completed vaccination schedule. Antigen-specific serum immunoglobulin levels are broadly used correlates of protection. These are the products of terminally differentiated B cells - plasma cells. Here we reviewed the literature on how aberrancies in B-cell composition and function influence immune responses to vaccinations. In a search through five major literature databases, 6,537 unique articles published from 2000 and onwards were identified. 75 articles were included along three major research lines: extremities of life, immunodeficiency and immunosuppression. Details of the protocol can be found in the International Prospective Register of Systematic Reviews [PROSPERO (registration number CRD42021226683)]. The majority of articles investigated immune responses in adults, in which vaccinations against pneumococci and influenza were strongly represented. Lack of baseline information was the most common reason of exclusion. Irrespective of study group, three parameters measured at baseline seemed to have a predictive value in assessing vaccine efficacy: (1) distribution of B-cell subsets (mostly a reduction in memory B cells), (2) presence of exhausted/activated B cells, or B cells with an aberrant phenotype, and (3) pre-existing immunological memory. In this review we showed how pre-immunization (baseline) knowledge of circulating B cells can be used to predict vaccination efficacy. We hope that this overview will contribute to optimizing vaccination strategies, especially in immunocompromised patients.
Collapse
Affiliation(s)
- Annieck M Diks
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Lisanne A Overduin
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Department of Infectious Diseases, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Laurens D van Leenen
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Lennert Slobbe
- Department of Internal Medicine, Section of Infectious Diseases, Institute for Tropical Diseases, Erasmus Medical Center (MC), Rotterdam, Netherlands
| | - Hetty Jolink
- Department of Infectious Diseases, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Leonardus G Visser
- Department of Infectious Diseases, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | | | - Magdalena A Berkowska
- Department of Immunology, Leiden University Medical Center (LUMC), Leiden, Netherlands
| |
Collapse
|
14
|
Wolff ASB, Braun S, Husebye ES, Oftedal BE. B Cells and Autoantibodies in AIRE Deficiency. Biomedicines 2021; 9:1274. [PMID: 34572460 PMCID: PMC8466229 DOI: 10.3390/biomedicines9091274] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/11/2021] [Accepted: 09/17/2021] [Indexed: 12/15/2022] Open
Abstract
Autoimmune polyendocrine syndrome type 1 (APS-1) is a rare but severe monogenetic autoimmune endocrine disease caused by failure of the Autoimmune Regulator (AIRE). AIRE regulates the negative selection of T cells in the thymus, and the main pathogenic mechanisms are believed to be T cell-mediated, but little is known about the role of B cells. Here, we give an overview of the role of B cells in thymic and peripheral tolerance in APS-1 patients and different AIRE-deficient mouse models. We also look closely into which autoantibodies have been described for this disorder, and their implications. Based on what is known about B cell therapy in other autoimmune disorders, we outline the potential of B cell therapies in APS-1 and highlight the unresolved research questions to be answered.
Collapse
Affiliation(s)
- Anette S. B. Wolff
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (A.S.B.W.); (S.B.); (E.S.H.)
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- KG Jebsen Center for Autoimmune Disorders, University of Bergen, 5021 Bergen, Norway
| | - Sarah Braun
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (A.S.B.W.); (S.B.); (E.S.H.)
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- Institute of Pharmacy and Molecular Biotechnology, Ruprecht-Karls University, 69120 Heidelberg, Germany
| | - Eystein S. Husebye
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (A.S.B.W.); (S.B.); (E.S.H.)
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- KG Jebsen Center for Autoimmune Disorders, University of Bergen, 5021 Bergen, Norway
| | - Bergithe E. Oftedal
- Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (A.S.B.W.); (S.B.); (E.S.H.)
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway
- KG Jebsen Center for Autoimmune Disorders, University of Bergen, 5021 Bergen, Norway
| |
Collapse
|
15
|
Vaccine response following anti-CD20 therapy: a systematic review and meta-analysis of 905 patients. Blood Adv 2021; 5:2624-2643. [PMID: 34152403 PMCID: PMC8216656 DOI: 10.1182/bloodadvances.2021004629] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/23/2021] [Indexed: 11/20/2022] Open
Abstract
The objective of this study was to perform a systematic review of the literature on vaccine responsiveness in patients who have received anti-CD20 therapy. PubMed and EMBASE were searched up to 4 January 2021 to identify studies of vaccine immunogenicity in patients treated with anti-CD20 therapy, including patients with hematologic malignancy or autoimmune disease. The primary outcomes were seroprotection (SP), seroconversion (SC), and/or seroresponse rates for each type of vaccine reported. As the pandemic influenza vaccine (2009 H1N1) has standardized definitions for SP and SC, and represented a novel primary antigen similar to the COVID-19 vaccine, meta-analysis was conducted for SC of studies of this vaccine. Pooled estimates, relative benefit ratios (RBs), and 95% confidence intervals (CIs) were calculated using a random-effects model. Thirty-eight studies (905 patients treated with anti-CD20 therapy) were included (19 studies of patients with hematologic malignancies). Patients on active (<3 months since last dose) anti-CD20 therapy had poor responses to all types of vaccines. The pooled estimate for SC after 1 pandemic influenza vaccine dose in these patients was 3% (95% CI, 0% to 9%), with an RB of 0.05 (95% CI, 0-0.73) compared with healthy controls and 0.22 (95% CI, 0.09-0.56) compared with disease controls. SC compared with controls seems abrogated for at least 6 months following treatment (3-6 months post anti-CD20 therapy with an RB of 0.50 [95% CI, 0.24-1.06] compared with healthy and of 0.44 [95% CI, 0.23-0.84] compared with disease controls). For all vaccine types, response to vaccination improves incrementally over time, but may not reach the level of healthy controls even 12 months after therapy.
Collapse
|
16
|
Joosse BA, Jackson JH, Cisneros A, Santhin AB, Smith SA, Moore DJ, Crofford LJ, Wilfong EM, Bonami RH. High-Throughput Detection of Autoantigen-Specific B Cells Among Distinct Functional Subsets in Autoimmune Donors. Front Immunol 2021; 12:685718. [PMID: 34234784 PMCID: PMC8256427 DOI: 10.3389/fimmu.2021.685718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 05/06/2021] [Indexed: 11/17/2022] Open
Abstract
Antigen-specific B cells (ASBCs) can drive autoimmune disease by presenting autoantigen to cognate T cells to drive their activation, proliferation, and effector cell differentiation and/or by differentiating into autoantibody-secreting cells. Autoantibodies are frequently used to predict risk and diagnose several autoimmune diseases. ASBCs can drive type 1 diabetes even when immune tolerance mechanisms block their differentiation into antibody-secreting cells. Furthermore, anti-histidyl tRNA synthetase syndrome patients have expanded IgM+ Jo-1-binding B cells, which clinically diagnostic IgG Jo-1 autoantibodies may not fully reflect. Given the potential disconnect between the pathologic function of ASBCs and autoantibody secretion, direct study of ASBCs is a necessary step towards developing better therapies for autoimmune diseases, which often have no available cure. We therefore developed a high-throughput screening pipeline to 1) phenotypically identify specific B cell subsets, 2) expand them in vitro, 3) drive them to secrete BCRs as antibody, and 4) identify wells enriched for ASBCs through ELISA detection of antibody. We tested the capacity of several B cell subset(s) to differentiate into antibody-secreting cells following this robust stimulation. IgM+ and/or IgD+, CD27- memory, memory, switched memory, and BND B cells secreted B cell receptor (BCR) as antibody following in vitro stimulation, whereas few plasmablasts responded. Bimodal responses were observed across autoimmune donors for IgM+ CD21lo and IgM- CD21lo B cells, consistent with documented heterogeneity within the CD21lo subset. Using this approach, we detected insulin-binding B cell bias towards CD27- memory and CD27+ memory subsets in pre-symptomatic type 1 diabetes donors. We took advantage of routine detection of Jo-1-binding B cells in Jo-1+ anti-histidyl tRNA synthetase syndrome patients to show that Jo-1-binding B cells and total B cells expanded 20-30-fold using this culture system. Overall, these studies highlight technology that is amenable to small numbers of cryopreserved peripheral blood mononuclear cells that enables interrogation of phenotypic and repertoire attributes of ASBCs derived from autoimmune patients.
Collapse
Affiliation(s)
- Bryan A Joosse
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - James H Jackson
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Biomedical Sciences, School of Medicine Greenville, University of South Carolina, Greenville, SC, United States
| | - Alberto Cisneros
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Austin B Santhin
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Scott A Smith
- Department of Medicine, Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Nashville, TN, United States
| | - Daniel J Moore
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Nashville, TN, United States.,Department of Pediatrics, Division of Endocrinology & Diabetes, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Leslie J Crofford
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Nashville, TN, United States
| | - Erin M Wilfong
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Medicine, Allergy, Pulmonary, and Critical Care, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Rachel H Bonami
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, United States.,Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Nashville, TN, United States
| |
Collapse
|
17
|
Stable HLA antibodies following sustained CD19+ cell depletion implicate a long-lived plasma cell source. Blood Adv 2021; 4:4292-4295. [PMID: 32915973 DOI: 10.1182/bloodadvances.2020002435] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/12/2020] [Indexed: 11/20/2022] Open
Abstract
Key Points
HLA-specific alloantibodies can be maintained despite profound CD19+ cell aplasia, likely due to production by CD19− plasma cells.
Collapse
|
18
|
Reder AT, Centonze D, Naylor ML, Nagpal A, Rajbhandari R, Altincatal A, Kim M, Berdofe A, Radhakrishnan M, Jung E, Sandrock AW, Smirnakis K, Popescu C, de Moor C. COVID-19 in Patients with Multiple Sclerosis: Associations with Disease-Modifying Therapies. CNS Drugs 2021; 35:317-330. [PMID: 33743151 PMCID: PMC7980129 DOI: 10.1007/s40263-021-00804-1] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/10/2021] [Indexed: 12/18/2022]
Abstract
BACKGROUND Disease-modifying therapies (DMTs) for multiple sclerosis (MS) target immunity and have the potential to increase the risk of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection and alter its clinical course. We assessed these risks in patients with MS (PwMS). OBJECTIVE The objective of this study was to describe the overall risk of coronavirus disease 2019 (COVID-19) infection, severe disease course, and potential population-level predictors of COVID-19 infection in PwMS, and to provide a context using a cohort of patients with systemic lupus erythematosus (SLE). In addition, the association of different MS DMTs with the incidence and clinical course of COVID-19 was evaluated. Safety data from the Biogen Global Safety Database are also presented on reported cases of COVID-19 in patients treated with Biogen MS therapies. METHODS The IBM® Explorys electronic health record database of > 72,000,000 patients from US healthcare networks identified patients with MS or SLE, with and without polymerase chain reaction-confirmed COVID-19. COVID-19 cumulative incidence, hospitalization, and deaths among DMT classes were compared using logistic regression (adjusted for age, sex, body mass index, comorbidities, and race/ethnicity). As a secondary data source to assess safety data, COVID-19 reports for Biogen MS therapies were extracted and described from Biogen's Global Safety Database. RESULTS 30,478 PwMS with an open DMT prescription were identified within Explorys; 344 were COVID-19 positive. The most significant risk factors for acquiring COVID-19 were comorbidity score ≥ 1, body mass index ≥ 30, and Black/African ancestry. Similar risk factors were also identified for patients with SLE. Patients with MS were less likely to develop COVID-19 when treated with interferons (0.61%) and glatiramer acetate (0.51%), vs all other MS DMTs (both p < 0.001); anti-CD20 therapy was associated with the highest risk (3.45%; p < 0.0001). In the Biogen Global Safety Database, we identified 1217 patients who were COVID-19 positive treated with intramuscular interferon beta-1a, peginterferon beta-1a, natalizumab, dimethyl fumarate, diroximel fumarate, or fampridine. CONCLUSIONS Comorbidities, obesity, and Black/African ancestry, but not age, were associated with a higher risk of SARS-CoV-2 infection in PwMS. Interferons and glatiramer acetate were associated with a reduced COVID-19 risk, whereas anti-CD20 therapies were associated with an increased risk, within the treated MS cohort. COVID-19 safety reports for patients receiving Biogen MS therapies were consistent with the Explorys database and MS literature, illustrating the replicability and power of this approach.
Collapse
Affiliation(s)
- Anthony T Reder
- Department of Neurology and Brain Research Institute, University of Chicago, Chicago, IL, USA.
| | - Diego Centonze
- Laboratory of Synaptic Immunopathology, Department of Systems Medicine, Tor Vergata University, Rome, Italy
- Unit of Neurology, IRCCS Neuromed, Pozzilli, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Kuderer NM, Hill JA, Carpenter PA, Lyman GH. Challenges and Opportunities for COVID-19 Vaccines in Patients with Cancer. Cancer Invest 2021; 39:205-213. [PMID: 33534645 DOI: 10.1080/07357907.2021.1885596] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Given the rapidly expanding global spread of the SARS-Co-V-2 virus and the expanding number of individuals with the serious and potentially fatal illness, COVID-19, there is an urgent need for safe and effective vaccines. Based on compelling evidence that patients with cancer are at increased risk for greater morbidity and mortality with COVID-19, several professional organizations have provided early guidance on the role of COVID-19 vaccines in patients with malignant disease. In this commentary we review the available data on the efficacy and safety of the approved and forthcoming vaccines in patients with cancer. Based on a review of the totality of available evidence, we recommend that most patients with cancer should receive the recommended dose and schedule of one of the COVID-19 vaccines when available. We encourage industry, regulators and professional research organizations to carefully track the efficacy and safety of COVID-19 vaccination in patients with cancer in the real world setting and routinely report unanticipated adverse events and signs of loss of efficacy. Particular attention is needed for patients on active cancer therapy to carefully evaluate efficacy and safety in relationship to the timing of vaccination relative to that of active cancer treatment and immunosuppression.
Collapse
Affiliation(s)
| | - Joshua A Hill
- Department of Medicine, University of Washington, Seattle, Washington, USA.,Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Paul A Carpenter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Gary H Lyman
- Department of Medicine, University of Washington, Seattle, Washington, USA.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| |
Collapse
|
20
|
Felton JL, Conway H, Bonami RH. B Quiet: Autoantigen-Specific Strategies to Silence Raucous B Lymphocytes and Halt Cross-Talk with T Cells in Type 1 Diabetes. Biomedicines 2021; 9:biomedicines9010042. [PMID: 33418839 PMCID: PMC7824835 DOI: 10.3390/biomedicines9010042] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 01/10/2023] Open
Abstract
Islet autoantibodies are the primary biomarkers used to predict type 1 diabetes (T1D) disease risk. They signal immune tolerance breach by islet autoantigen-specific B lymphocytes. T-B lymphocyte interactions that lead to expansion of pathogenic T cells underlie T1D development. Promising strategies to broadly prevent this T-B crosstalk include T cell elimination (anti-CD3, teplizumab), B cell elimination (anti-CD20, rituximab), and disruption of T cell costimulation/activation (CTLA-4/Fc fusion, abatacept). However, global disruption or depletion of immune cell subsets is associated with significant risk, particularly in children. Therefore, antigen-specific therapy is an area of active investigation for T1D prevention. We provide an overview of strategies to eliminate antigen-specific B lymphocytes as a means to limit pathogenic T cell expansion to prevent beta cell attack in T1D. Such approaches could be used to prevent T1D in at-risk individuals. Patients with established T1D would also benefit from such targeted therapies if endogenous beta cell function can be recovered or islet transplant becomes clinically feasible for T1D treatment.
Collapse
Affiliation(s)
- Jamie L. Felton
- Department of Pediatrics, Division of Pediatric Endocrinology and the Herman B. Wells Center for Pediatric Research, Indianapolis, IN 46202, USA; (J.L.F.); (H.C.)
| | - Holly Conway
- Department of Pediatrics, Division of Pediatric Endocrinology and the Herman B. Wells Center for Pediatric Research, Indianapolis, IN 46202, USA; (J.L.F.); (H.C.)
| | - Rachel H. Bonami
- Department of Medicine, Division of Rheumatology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Correspondence:
| |
Collapse
|
21
|
Gembara K, Dąbrowska K. Phage-specific antibodies. Curr Opin Biotechnol 2020; 68:186-192. [PMID: 33388538 DOI: 10.1016/j.copbio.2020.11.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 11/12/2020] [Accepted: 11/22/2020] [Indexed: 12/29/2022]
Abstract
Antibodies specific to phage virions have been observed many times, both in animals and in humans. Phages induce the T-dependent type of immune response, which is fundamental for immunological memory and long retention of abilities to recognize and respond to foreign epitopes. Experimental models have shown that phage-specific antibodies can be devastating for a phage in vivo. Observations from phage treatment in humans demonstrate however that antibodies do not necessarily hinder phage therapy. Controlling effective doses may help in controlling the immune response to phage and to create 'therapeutic windows' for the phage. Phage therapy design may also benefit from understanding phage antigenicity and predicting possible cross-reactions between antibodies specific to different phages.
Collapse
Affiliation(s)
- Katarzyna Gembara
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wrocław, Poland
| | - Krystyna Dąbrowska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Weigla 12, 53-114, Wrocław, Poland.
| |
Collapse
|
22
|
Esmaeil Amini M, Shomali N, Bakhshi A, Rezaei S, Hemmatzadeh M, Hosseinzadeh R, Eslami S, Babaie F, Aslani S, Torkamandi S, Mohammadi H. Gut microbiome and multiple sclerosis: New insights and perspective. Int Immunopharmacol 2020; 88:107024. [PMID: 33182024 DOI: 10.1016/j.intimp.2020.107024] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 02/07/2023]
Abstract
The human gastrointestinal microbiota, also known as the gut microbiota living in the human gastrointestinal tract, has been shown to have a significant impact on several human disorders including rheumatoid arthritis, diabetes, obesity, and multiple sclerosis (MS). MS is an inflammatory disease characterized by the destruction of the spinal cord and nerve cells in the brain due to an attack of immune cells, causing a wide range of harmful symptoms related to inflammation in the central nervous system (CNS). Despite extensive studies on MS that have shown that many external and genetic factors are involved in its pathogenesis, the exact role of external factors in the pathophysiology of MS is still unclear. Recent studies on MS and experimental autoimmune encephalomyelitis (EAE), an animal model of encephalitis, have shown that intestinal microbiota may play a key role in the pathogenesis of MS. Therefore, modification of the intestinal microbiome could be a promising strategy for the future treatment of MS. In this study, the characteristics of intestinal microbiota, the relationship between intestine and brain despite the blood-brain barrier, various factors involved in intestinal microbiota modification, changes in intestinal microbial composition in MS, intestinal microbiome modification strategies, and possible use of intestinal microbiome and factors affecting it have been discussed.
Collapse
Affiliation(s)
- Mohammad Esmaeil Amini
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran; Student Research Committee, Guilan University of Medical Sciences, Rasht, Iran
| | - Navid Shomali
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Arash Bakhshi
- Student Research Committee, Guilan University of Medical Sciences, Rasht, Iran
| | - Somaye Rezaei
- Department of Neurology, Imam Khomeini Hospital, Urmia University of Medical Sciences, Urmia, Iran
| | - Maryam Hemmatzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Hosseinzadeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Solat Eslami
- Dietary Supplements & Probiotic Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Farhad Babaie
- Department of Medical Genetics and Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Saeed Aslani
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahram Torkamandi
- Department of Medical Genetics and Immunology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran; Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.
| |
Collapse
|
23
|
Robinson MJ, Webster RH, Tarlinton DM. How intrinsic and extrinsic regulators of plasma cell survival might intersect for durable humoral immunity. Immunol Rev 2020; 296:87-103. [DOI: 10.1111/imr.12895] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/01/2020] [Accepted: 06/03/2020] [Indexed: 12/11/2022]
Affiliation(s)
- Marcus J. Robinson
- Department of Immunology & Pathology Alfred Medical Research and Education Precinct Monash University Melbourne Vic. Australia
| | - Rosela H. Webster
- Department of Immunology & Pathology Alfred Medical Research and Education Precinct Monash University Melbourne Vic. Australia
| | - David M. Tarlinton
- Department of Immunology & Pathology Alfred Medical Research and Education Precinct Monash University Melbourne Vic. Australia
| |
Collapse
|
24
|
Mustafa SS, Jamshed S, Vadamalai K, Ramsey A. The Use of 20% Subcutaneous Immunoglobulin Replacement Therapy in Patients With B Cell Non-Hodgkin Lymphoma With Humoral Immune Dysfunction After Treatment With Rituximab. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:e590-e596. [PMID: 32646834 DOI: 10.1016/j.clml.2020.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/09/2020] [Accepted: 04/11/2020] [Indexed: 01/14/2023]
Abstract
BACKGROUND Rituximab is an anti-CD20 chimeric antibody used to treat autoimmune conditions and B cell neoplasms. We characterized immunoglobulin (Ig) levels and vaccine responses in rituximab-treated B cell non-Hodgkin lymphoma (NHL) patients. Patients with impaired vaccine responses were offered therapy with 20% subcutaneous (subq) Ig. PATIENTS AND METHODS Patients with a biopsy-proven diagnosis of B cell NHL who had received rituximab within the past 24 months were eligible for the study and underwent the following immune evaluation: serum IgG, IgM, IgA, IgE, T/B cell lymphocyte panel, and pre/post vaccine IgG titers to diphtheria, tetanus, and streptococcus pneumoniae. Patients were vaccinated with tetanus, diphtheria and pneumococcal polysaccharide vaccine. Patients with abnormal vaccine responses were offered prophylactic subq Ig for 52 weeks. RESULTS Fifteen patients with NHL were enrolled in the study. The median IgG was 628 mg/dL [interquartile range, 489-718 mg/dL]. Three (20%) of 15 patients responded to diphtheria vaccination, 1 (6.7%) of 15 responded to tetanus vaccination, and 3 (20%) of 15 responded to vaccination to streptococcus pneumoniae. Thirteen (86.7%) of 15 met criteria for humoral immunodeficiency. Ten patients received subq Ig, and experienced a significant increase in serum IgG (P = .008). There were no serious adverse events, and there was a decrease in nonneutropenic infections while on subq Ig therapy. CONCLUSIONS Patients with NHL treated with rituximab may have significant humoral immunodeficiency as defined by abnormal vaccine responses even in the setting of relatively normal IgG levels. For these patients, subq Ig replacement therapy is well-tolerated and efficacious in improving serum IgG, and may decrease reliance on antibiotics for the treatment of nonneutropenic infections.
Collapse
Affiliation(s)
- S Shahzad Mustafa
- Division of Allergy, Immunology, and Rheumatology, Rochester Regional Health, Rochester, NY; Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY.
| | - Saad Jamshed
- Division of Hematology and Oncology, Rochester Regional Health, Rochester, NY
| | | | - Allison Ramsey
- Division of Allergy, Immunology, and Rheumatology, Rochester Regional Health, Rochester, NY; Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY
| |
Collapse
|
25
|
Anti-CD20–mediated B-cell depletion in autoimmune diseases: successes, failures and future perspectives. Kidney Int 2020; 97:885-893. [DOI: 10.1016/j.kint.2019.12.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/09/2019] [Accepted: 12/12/2019] [Indexed: 12/11/2022]
|
26
|
Mina MJ, Kula T, Leng Y, Li M, de Vries RD, Knip M, Siljander H, Rewers M, Choy DF, Wilson MS, Larman HB, Nelson AN, Griffin DE, de Swart RL, Elledge SJ. Measles virus infection diminishes preexisting antibodies that offer protection from other pathogens. Science 2019; 366:599-606. [PMID: 31672891 PMCID: PMC8590458 DOI: 10.1126/science.aay6485] [Citation(s) in RCA: 244] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/30/2019] [Indexed: 12/25/2022]
Abstract
Measles virus is directly responsible for more than 100,000 deaths yearly. Epidemiological studies have associated measles with increased morbidity and mortality for years after infection, but the reasons why are poorly understood. Measles virus infects immune cells, causing acute immune suppression. To identify and quantify long-term effects of measles on the immune system, we used VirScan, an assay that tracks antibodies to thousands of pathogen epitopes in blood. We studied 77 unvaccinated children before and 2 months after natural measles virus infection. Measles caused elimination of 11 to 73% of the antibody repertoire across individuals. Recovery of antibodies was detected after natural reexposure to pathogens. Notably, these immune system effects were not observed in infants vaccinated against MMR (measles, mumps, and rubella), but were confirmed in measles-infected macaques. The reduction in humoral immune memory after measles infection generates potential vulnerability to future infections, underscoring the need for widespread vaccination.
Collapse
Affiliation(s)
- Michael J Mina
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Boston, MA 02115, USA.
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Tomasz Kula
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Boston, MA 02115, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Yumei Leng
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Boston, MA 02115, USA
| | - Mamie Li
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Rory D de Vries
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, 3015 CN, Rotterdam, Netherlands
| | - Mikael Knip
- Children's Hospital, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, 00014 Helsinki, Finland
| | - Heli Siljander
- Children's Hospital, University of Helsinki and Helsinki University Hospital, 00290 Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, 00014 Helsinki, Finland
| | - Marian Rewers
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Denver, CO 80045, USA
| | - David F Choy
- Genentech Inc., South San Francisco, CA 94080, USA
| | | | - H Benjamin Larman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ashley N Nelson
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Rik L de Swart
- Department of Viroscience, Postgraduate School of Molecular Medicine, Erasmus MC, University Medical Centre Rotterdam, 3015 CN, Rotterdam, Netherlands
| | - Stephen J Elledge
- Division of Genetics, Brigham and Women's Hospital, Howard Hughes Medical Institute, Boston, MA 02115, USA.
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
27
|
Garcia Garrido HM, Veurink AM, Leeflang M, Spijker R, Goorhuis A, Grobusch MP. Hepatitis A vaccine immunogenicity in patients using immunosuppressive drugs: A systematic review and meta-analysis. Travel Med Infect Dis 2019; 32:101479. [PMID: 31521804 DOI: 10.1016/j.tmaid.2019.101479] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 09/02/2019] [Accepted: 09/09/2019] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Inactivated hepatitis A (HepA) vaccines are very immunogenic in healthy individuals; however, it remains unclear how different immunosuppressive regimens affect HepA vaccine immunogenicity. Our objective was to summarise the current evidence on immunogenicity of HepA vaccination in patients using immunosuppressive drugs. METHODS We systematically searched the literature for studies on immunogenicity of inactivated HepA vaccines in adults using immunosuppressive drugs. Studies reporting seroconversion rates (SCR) 4-8 weeks after 1 and 2 doses of HepA vaccine in organ transplant recipients and patients with chronic inflammatory conditions were included in a meta-analysis. RESULTS We included 17 studies, comprising 1,332 individuals. In healthy controls (2 studies), SCRs were 90-94% after the first dose and 100% after the second dose. In organ transplant recipients, SCRs ranged from 0 to 67% after the first dose of vaccine and 0-97% after the second dose. In patients with chronic inflammatory conditions, SCRs ranged from 6% to 100% after the first dose and from 48 to 100% after the second dose of vaccine. Patients using a TNF-alpha inhibitor versus conventional immune-modulators (e.g. methotrexate, azathioprine, corticosteroids) were more likely to seroconvert after the first dose of vaccine (OR12.1 [2.14-68.2]) but not after the second dose of vaccine (OR 0.78 [0.21-2.92]) in a meta-analysis. CONCLUSION Studies evaluating HepA vaccine immunogenicity in immunosuppressive agents are heterogeneous. Overall, there is an impaired immune response following HepA vaccination in patients using immunosuppressive drugs, especially after only one dose of vaccine and in organ transplant recipients. HepA vaccination should therefore be considered before immunosuppressive therapy. Future research should focus on alternative vaccination regimens and long-term immunogenicity. PROSPERO ID CRD42018102607.
Collapse
Affiliation(s)
- Hannah M Garcia Garrido
- Amsterdam UMC, University of Amsterdam, Centre of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Meibergdreef 9, Amsterdam, the Netherlands.
| | - Ati M Veurink
- Amsterdam UMC, University of Amsterdam, Centre of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Meibergdreef 9, Amsterdam, the Netherlands
| | - Mariska Leeflang
- Amsterdam UMC, University of Amsterdam, Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam Public Health, Meibergdreef 9, Amsterdam, the Netherlands
| | - René Spijker
- Amsterdam UMC, University of Amsterdam, Medical Library, Amsterdam Public Health, Meibergdreef 9, Amsterdam, the Netherlands; Cochrane Netherlands, Julius Center for Health Sciences and Primary Care, UMC Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Abraham Goorhuis
- Amsterdam UMC, University of Amsterdam, Centre of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Meibergdreef 9, Amsterdam, the Netherlands
| | - Martin P Grobusch
- Amsterdam UMC, University of Amsterdam, Centre of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Meibergdreef 9, Amsterdam, the Netherlands
| |
Collapse
|
28
|
Hill JA, Giralt S, Torgerson TR, Lazarus HM. CAR-T - and a side order of IgG, to go? - Immunoglobulin replacement in patients receiving CAR-T cell therapy. Blood Rev 2019; 38:100596. [PMID: 31416717 DOI: 10.1016/j.blre.2019.100596] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/22/2019] [Accepted: 08/06/2019] [Indexed: 12/15/2022]
Abstract
The development and regulatory approval of chimeric antigen receptor T cell (CAR-T) therapies targeting the B-lineage surface antigen CD19 represents a major milestone in cancer immunotherapy. This treatment also results in depletion of normal CD19+ B cells and is associated with hypogammaglobulinemia. These on-target, off-tumor toxicities may result in an increased risk for infection, particularly for encapsulated bacteria. Data regarding the efficacy and cost-effectiveness of prophylactic IgG replacement in CD19-targeted CAR-T cell therapy recipients is lacking, and current expert recommendations are extrapolated from the data for individuals with primary immune deficiencies. This article reviews CAR-T cell therapies targeting B-lineage lymphocytes, associated side effects, and considerations for the approach to management of hypogamaglobulinemia in this patient population. Studies are needed to establish evidence-based approaches to prophylactic immunoglobulin administration in this context, and strategies may differ by patient and CAR-T cell product characteristics.
Collapse
Affiliation(s)
- Joshua A Hill
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA; Department of Medicine, University of Washington, Seattle, WA, USA.
| | - Sergio Giralt
- Adult Bone Marrow Transplant Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Troy R Torgerson
- Department of Pediatrics, University of Washington, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Hillard M Lazarus
- Department of Medicine, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
29
|
Baker D, Pryce G, Amor S, Giovannoni G, Schmierer K. Learning from other autoimmunities to understand targeting of B cells to control multiple sclerosis. Brain 2019; 141:2834-2847. [PMID: 30212896 DOI: 10.1093/brain/awy239] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/01/2018] [Indexed: 12/15/2022] Open
Abstract
Although many suspected autoimmune diseases are thought to be T cell-mediated, the response to therapy indicates that depletion of B cells consistently inhibits disease activity. In multiple sclerosis, it appears that disease suppression is associated with the long-term reduction of memory B cells, which serves as a biomarker for disease activity in many other CD20+ B cell depletion-sensitive, autoimmune diseases. Following B cell depletion, the rapid repopulation by transitional (immature) and naïve (mature) B cells from the bone marrow masks the marked depletion and slow repopulation of lymphoid tissue-derived, memory B cells. This can provide long-term protection from a short treatment cycle. It seems that memory B cells, possibly via T cell stimulation, drive relapsing disease. However, their sequestration in ectopic follicles and the chronic activity of B cells and plasma cells in the central nervous system may drive progressive neurodegeneration directly via antigen-specific mechanisms or indirectly via glial-dependent mechanisms. While unproven, Epstein-Barr virus may be an aetiological trigger of multiple sclerosis. This infects mature B cells, drives the production of memory B cells and possibly provides co-stimulatory signals promoting T cell-independent activation that breaks immune tolerance to generate autoreactivity. Thus, a memory B cell centric mechanism can integrate: potential aetiology, genetics, pathology and response to therapy in multiple sclerosis and other autoimmune conditions with ectopic B cell activation that are responsive to memory B cell-depleting strategies.
Collapse
Affiliation(s)
- David Baker
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Gareth Pryce
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sandra Amor
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Pathology Department, Free University Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Gavin Giovannoni
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, UK
| | - Klaus Schmierer
- BartsMS, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, UK
| |
Collapse
|
30
|
Slifka MK, Amanna IJ. Role of Multivalency and Antigenic Threshold in Generating Protective Antibody Responses. Front Immunol 2019; 10:956. [PMID: 31118935 PMCID: PMC6504826 DOI: 10.3389/fimmu.2019.00956] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 04/15/2019] [Indexed: 12/03/2022] Open
Abstract
Vaccines play a vital role in protecting our communities against infectious disease. Unfortunately, some vaccines provide only partial protection or in some cases vaccine-mediated immunity may wane rapidly, resulting in either increased susceptibility to that disease or a requirement for more booster vaccinations in order to maintain immunity above a protective level. The durability of antibody responses after infection or vaccination appears to be intrinsically determined by the structural biology of the antigen, with multivalent protein antigens often providing more long-lived immunity than monovalent antigens. This forms the basis for the Imprinted Lifespan model describing the differential survival of long-lived plasma cell populations. There are, however, exceptions to this rule with examples of highly attenuated live virus vaccines that are rapidly cleared and elicit only short-lived immunity despite the expression of multivalent surface epitopes. These exceptions have led to the concept that multivalency alone may not reliably determine the duration of protective humoral immune responses unless a minimum number of long-lived plasma cells are generated by reaching an appropriate antigenic threshold of B cell stimulation. Examples of long-term and in some cases, potentially lifelong antibody responses following immunization against human papilloma virus (HPV), Japanese encephalitis virus (JEV), Hepatitis B virus (HBV), and Hepatitis A virus (HAV) provide several lessons in understanding durable serological memory in human subjects. Moreover, studies involving influenza vaccination provide the unique opportunity to compare the durability of hemagglutinin (HA)-specific antibody titers mounted in response to antigenically repetitive whole virus (i.e., multivalent HA), or detergent-disrupted “split” virus, in comparison to the long-term immune responses induced by natural influenza infection. Here, we discuss the underlying mechanisms that may be associated with the induction of protective immunity by long-lived plasma cells and their importance in future vaccine design.
Collapse
Affiliation(s)
- Mark K Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, United States
| | - Ian J Amanna
- Najít Technologies, Inc., Beaverton, OR, United States
| |
Collapse
|
31
|
Linsley PS, Greenbaum CJ, Rosasco M, Presnell S, Herold KC, Dufort MJ. Elevated T cell levels in peripheral blood predict poor clinical response following rituximab treatment in new-onset type 1 diabetes. Genes Immun 2019; 20:293-307. [PMID: 29925930 PMCID: PMC6477779 DOI: 10.1038/s41435-018-0032-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 12/11/2022]
Abstract
Biologic treatment of type 1 diabetes (T1D) with agents including anti-CD3 (otelixizumab and teplizumab), anti-CD20 (rituximab), LFA3Ig (alafacept), and CTLA4Ig (abatacept) results in transient stabilization of insulin C-peptide, a surrogate for endogenous insulin secretion. With the goal of inducing more robust immune tolerance, we used systems biology approaches to elucidate mechanisms associated with C-peptide stabilization in clinical trial blood samples from new-onset T1D subjects treated with the B cell-depleting drug, rituximab. RNA sequencing (RNA-seq) analysis of whole-blood samples from this trial revealed a transient increase in heterogeneous T cell populations, which were associated with decreased pharmacodynamic activity of rituximab, increased proliferative responses to islet antigens, and more rapid C-peptide loss. Our findings illustrate complexity in hematopoietic remodeling that accompanies B cell depletion by rituximab, which impacts and predicts therapeutic efficacy in T1D. Our data also suggest that a combination of rituximab with therapy targeting CD4 + T cells may be beneficial for T1D subjects.
Collapse
Affiliation(s)
- Peter S Linsley
- Systems Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
| | - Carla J Greenbaum
- Diabetes Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Mario Rosasco
- Systems Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Scott Presnell
- Systems Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Kevan C Herold
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT, 06520, USA
| | - Matthew J Dufort
- Systems Immunology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| |
Collapse
|
32
|
Sánchez-Ramón S, Bermúdez A, González-Granado LI, Rodríguez-Gallego C, Sastre A, Soler-Palacín P. Primary and Secondary Immunodeficiency Diseases in Oncohaematology: Warning Signs, Diagnosis, and Management. Front Immunol 2019; 10:586. [PMID: 30984175 PMCID: PMC6448689 DOI: 10.3389/fimmu.2019.00586] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Accepted: 03/05/2019] [Indexed: 11/13/2022] Open
Abstract
Background: Immunodeficiencies (ID), in particular primary immunodeficiencies (PID), are often associated with haematological manifestations, such as peripheral cytopenias or lymphoproliferative syndromes. Early diagnosis and management have significant prognostic implications. Secondary immunodeficiencies (SID) may also be induced by oncohaematological diseases and their treatments. Haematologists and oncologists must therefore be aware of the association between blood disorders and cancer and ID, and be prepared to offer their patients appropriate treatment without delay. Our aim was to define the warning signs of primary and secondary IDs in paediatric and adult patients with oncohaematological manifestations. Methods: A multidisciplinary group of six experts (2 haematologists, 2 immunologists, and 2 paediatricians specializing in ID) conducted a literature review and prepared a document based on agreements reached an in-person meeting. An external group of 44 IDs specialists from all over Spain assessed the document and were consulted regarding their level of agreement. Results: This document identifies the haematological and extra-haematological diseases that should prompt a suspicion of PIDs in adults and children, in both primary care and haematology and oncology departments. Cytopenia and certain lymphoproliferative disorders are key diagnostic pointers. The diagnosis must be based on a detailed clinical history, physical exploration, complete blood count and standard laboratory tests. The immunological and haematological tests included in the diagnostic process will depend on the care level. Patients who are candidates for immunoglobulin replacement therapy must be carefully selected, and treatment should be offered as soon as possible to avoid the development of complications. Finally, this document recommends procedures for monitoring these patients. Conclusions: This document combines scientific evidence with the opinion of a broad panel of experts, and emphasizes the importance of an early diagnosis and treatment to avoid complications. The resulting document is a useful tool for primary care physicians and specialists who see both adult and paediatric patients with oncohaematological diseases.
Collapse
Affiliation(s)
| | | | | | - Carlos Rodríguez-Gallego
- Hospital Universitario de Gran Canaria Dr. Negrín, Las Palmas de Gran Canaria, Canary Islands, Spain
| | | | | | | |
Collapse
|
33
|
Linsley PS, Greenbaum CJ, Speake C, Long SA, Dufort MJ. B lymphocyte alterations accompany abatacept resistance in new-onset type 1 diabetes. JCI Insight 2019; 4:126136. [PMID: 30830871 PMCID: PMC6478438 DOI: 10.1172/jci.insight.126136] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/17/2019] [Indexed: 02/06/2023] Open
Abstract
Costimulatory interactions control T cell activation at sites of activated antigen-presenting cells, including B cells. Blockade of the CD28/CD80/CD86 costimulatory axis with CTLA4Ig (abatacept) is widely used to treat certain autoimmune diseases. While transiently effective in subjects with new-onset type 1 diabetes (T1D), abatacept did not induce long-lasting immune tolerance. To elucidate mechanisms limiting immune tolerance in T1D, we performed unbiased analysis of whole blood transcriptomes and targeted measurements of cell subset levels in subjects from a clinical trial of abatacept in new-onset T1D. We showed that individual subjects displayed age-related immune phenotypes ("immunotypes") at baseline, characterized by elevated levels of B cells or neutrophils, that accompanied rapid or slow progression, respectively, in both abatacept- and placebo-treated groups. A more pronounced immunotype was exhibited by a subset of subjects showing poor response (resistance) to abatacept. This resistance immunotype was characterized by a transient increase in activated B cells (one of the cell types that binds abatacept), reprogrammed costimulatory ligand gene expression, and reduced inhibition of anti-insulin antibodies. Our findings identify immunotypes in T1D subjects that are linked to the rate of disease progression, both in placebo- and abatacept-treated subjects. Furthermore, our results suggest therapeutic approaches to restore immune tolerance in T1D.
Collapse
Affiliation(s)
| | | | | | - S. Alice Long
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington, USA
| | | |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW The immunosuppressive agent cyclosporine was first reported to lower daily insulin dose and improve glycemic control in patients with new-onset type 1 diabetes (T1D) in 1984. While renal toxicity limited cyclosporine's extended use, this observation ignited collaborative efforts to identify immunotherapeutic agents capable of safely preserving β cells in patients with or at risk for T1D. RECENT FINDINGS Advances in T1D prediction and early diagnosis, together with expanded knowledge of the disease mechanisms, have facilitated trials targeting specific immune cell subsets, autoantigens, and pathways. In addition, clinical responder and non-responder subsets have been defined through the use of metabolic and immunological readouts. Herein, we review emerging T1D biomarkers within the context of recent and ongoing T1D immunotherapy trials. We also discuss responder/non-responder analyses in an effort to identify therapeutic mechanisms, define actionable pathways, and guide subject selection, drug dosing, and tailored combination drug therapy for future T1D trials.
Collapse
Affiliation(s)
- Laura M Jacobsen
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Brittney N Newby
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Daniel J Perry
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA
| | - Michael J Haller
- Department of Pediatrics, College of Medicine, University of Florida Diabetes Institute, Gainesville, FL, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida Diabetes Institute, 1275 Center Drive, Biomedical Sciences Building J-589, Box 100275, Gainesville, FL, 32610, USA.
| |
Collapse
|
35
|
Greenbaum CJ, Speake C, Krischer J, Buckner J, Gottlieb PA, Schatz DA, Herold KC, Atkinson MA. Strength in Numbers: Opportunities for Enhancing the Development of Effective Treatments for Type 1 Diabetes-The TrialNet Experience. Diabetes 2018; 67:1216-1225. [PMID: 29769238 PMCID: PMC6014559 DOI: 10.2337/db18-0065] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/20/2018] [Indexed: 12/12/2022]
Abstract
The early to mid-1980s were an inflection point in the history of type 1 diabetes research. Two landmark events occurred: the initiation of immune-based interventions seeking to prevent type 1 diabetes and the presentation of an innovative model describing the disorder's natural history. Both formed the basis for hundreds of subsequent studies designed to achieve a dramatic therapeutic goal-a means to prevent and/or reverse type 1 diabetes. However, the need to screen large numbers of individuals and prospectively monitor them using immunologic and metabolic tests for extended periods of time suggested such efforts would require a large collaborative network. Hence, the National Institutes of Health formed the landmark Diabetes Prevention Trial-Type 1 (DPT-1) in the mid-1990s, an effort that led to Type 1 Diabetes TrialNet. TrialNet studies have helped identify novel biomarkers; delineate type 1 diabetes progression, resulting in identification of highly predictable stages defined by the accumulation of autoantibodies (stage 1), dysglycemia (stage 2), and disease meeting clinical criteria for diagnosis (stage 3); and oversee numerous clinical trials aimed at preventing disease progression. Such efforts pave the way for stage-specific intervention trials with improved hope that a means to effectively disrupt the disorder's development will be identified.
Collapse
Affiliation(s)
- Carla J Greenbaum
- Clinical Research Center, Diabetes Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Cate Speake
- Clinical Research Center, Diabetes Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Jeffrey Krischer
- Diabetes Center and Pediatric Epidemiology Center, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Jane Buckner
- Clinical Research Center, Diabetes Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA
| | - Peter A Gottlieb
- Barbara Davis Center for Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Desmond A Schatz
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
| | - Kevan C Herold
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT
| | - Mark A Atkinson
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL
- Department of Pathology, College of Medicine, University of Florida, Gainesville, FL
| |
Collapse
|
36
|
Da Rosa LC, Boldison J, De Leenheer E, Davies J, Wen L, Wong FS. B cell depletion reduces T cell activation in pancreatic islets in a murine autoimmune diabetes model. Diabetologia 2018; 61:1397-1410. [PMID: 29594371 PMCID: PMC6449006 DOI: 10.1007/s00125-018-4597-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/21/2018] [Indexed: 01/01/2023]
Abstract
AIMS/HYPOTHESIS Type 1 diabetes is a T cell-mediated autoimmune disease characterised by the destruction of beta cells in the islets of Langerhans, resulting in deficient insulin production. B cell depletion therapy has proved successful in preventing diabetes and restoring euglycaemia in animal models of diabetes, as well as in preserving beta cell function in clinical trials in the short term. We aimed to report a full characterisation of B cell kinetics post B cell depletion, with a focus on pancreatic islets. METHODS Transgenic NOD mice with a human CD20 transgene expressed on B cells were injected with an anti-CD20 depleting antibody. B cells were analysed using multivariable flow cytometry. RESULTS There was a 10 week delay in the onset of diabetes when comparing control and experimental groups, although the final difference in the diabetes incidence, following prolonged observation, was not statistically significant (p = 0.07). The co-stimulatory molecules CD80 and CD86 were reduced on stimulation of B cells during B cell depletion and repopulation. IL-10-producing regulatory B cells were not induced in repopulated B cells in the periphery, post anti-CD20 depletion. However, the early depletion of B cells had a marked effect on T cells in the local islet infiltrate. We demonstrated a lack of T cell activation, specifically with reduced CD44 expression and effector function, including IFN-γ production from both CD4+ and CD8+ T cells. These CD8+ T cells remained altered in the pancreatic islets long after B cell depletion and repopulation. CONCLUSIONS/INTERPRETATION Our findings suggest that B cell depletion can have an impact on T cell regulation, inducing a durable effect that is present long after repopulation. We suggest that this local effect of reducing autoimmune T cell activity contributes to delay in the onset of autoimmune diabetes.
Collapse
Affiliation(s)
- Larissa C Da Rosa
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas, Campinas, SP, Brazil
| | - Joanne Boldison
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - Evy De Leenheer
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
- University of Sheffield, New Spring House, Sheffield, UK
| | - Joanne Davies
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK
| | - Li Wen
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, USA
| | - F Susan Wong
- Division of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, CF14 4XN, UK.
| |
Collapse
|
37
|
Wong KY, Baron R, Seldon TA, Jones ML, Rice AM, Munster DJ. CD83 Antibody Inhibits Human B Cell Responses to Antigen as well as Dendritic Cell-Mediated CD4 T Cell Responses. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 200:3383-3396. [PMID: 29643191 DOI: 10.4049/jimmunol.1700064] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Accepted: 03/20/2018] [Indexed: 01/11/2023]
Abstract
Anti-CD83 Ab capable of Ab-dependent cellular cytotoxicity can deplete activated CD83+ human dendritic cells, thereby inhibiting CD4 T cell-mediated acute graft-versus-host disease. As CD83 is also expressed on the surface of activated B lymphocytes, we hypothesized that anti-CD83 would also inhibit B cell responses to stimulation. We found that anti-CD83 inhibited total IgM and IgG production in vitro by allostimulated human PBMC. Also, Ag-specific Ab responses to immunization of SCID mice xenografted with human PBMC were inhibited by anti-CD83 treatment. This inhibition occurred without depletion of all human B cells because anti-CD83 lysed activated CD83+ B cells by Ab-dependent cellular cytotoxicity and spared resting (CD83-) B cells. In cultured human PBMC, anti-CD83 inhibited tetanus toxoid-stimulated B cell proliferation and concomitant dendritic cell-mediated CD4 T cell proliferation and expression of IFN-γ and IL-17A, with minimal losses of B cells (<20%). In contrast, the anti-CD20 mAb rituximab depleted >80% of B cells but had no effect on CD4 T cell proliferation and cytokine expression. By virtue of the ability of anti-CD83 to selectively deplete activated, but not resting, B cells and dendritic cells, with the latter reducing CD4 T cell responses, anti-CD83 may be clinically useful in autoimmunity and transplantation. Advantages might include inhibited expansion of autoantigen- or alloantigen-specific B cells and CD4 T cells, thus preventing further production of pathogenic Abs and inflammatory cytokines while preserving protective memory and regulatory cells.
Collapse
Affiliation(s)
- Kuan Y Wong
- Mater Research Institute, University of Queensland, Brisbane, Queensland 4102, Australia; and
| | - Rebecca Baron
- Mater Research Institute, University of Queensland, Brisbane, Queensland 4102, Australia; and
| | - Therese A Seldon
- Mater Research Institute, University of Queensland, Brisbane, Queensland 4102, Australia; and
| | - Martina L Jones
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Alison M Rice
- Mater Research Institute, University of Queensland, Brisbane, Queensland 4102, Australia; and
| | - David J Munster
- Mater Research Institute, University of Queensland, Brisbane, Queensland 4102, Australia; and
| |
Collapse
|
38
|
Bingley PJ, Wherrett DK, Shultz A, Rafkin LE, Atkinson MA, Greenbaum CJ. Type 1 Diabetes TrialNet: A Multifaceted Approach to Bringing Disease-Modifying Therapy to Clinical Use in Type 1 Diabetes. Diabetes Care 2018; 41:653-661. [PMID: 29559451 PMCID: PMC5860837 DOI: 10.2337/dc17-0806] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 12/13/2017] [Indexed: 02/03/2023]
Abstract
What will it take to bring disease-modifying therapy to clinical use in type 1 diabetes? Coordinated efforts of investigators involved in discovery, translational, and clinical research operating in partnership with funders and industry and in sync with regulatory agencies are needed. This Perspective describes one such effort, Type 1 Diabetes TrialNet, a National Institutes of Health-funded and JDRF-supported international clinical trials network that emerged from the Diabetes Prevention Trial-Type 1 (DPT-1). Through longitudinal natural history studies, as well as trials before and after clinical onset of disease combined with mechanistic and ancillary investigations to enhance scientific understanding and translation to clinical use, TrialNet is working to bring disease-modifying therapies to individuals with type 1 diabetes. Moreover, TrialNet uses its expertise and experience in clinical studies to increase efficiencies in the conduct of trials and to reduce the burden of participation on individuals and families. Herein, we highlight key contributions made by TrialNet toward a revised understanding of the natural history of disease and approaches to alter disease course and outline the consortium's plans for the future.
Collapse
Affiliation(s)
- Polly J Bingley
- Diabetes and Metabolism, Bristol Medical School, University of Bristol, Bristol, U.K
| | - Diane K Wherrett
- Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | - Ann Shultz
- Diabetes Research Program, Benaroya Research Institute, Seattle, WA
| | - Lisa E Rafkin
- University of Miami Diabetes Research Institute, Miami, FL
| | - Mark A Atkinson
- Departments of Pathology and Pediatrics, University of Florida College of Medicine,Gainesville, FL
| | | |
Collapse
|
39
|
Plasma cell survival in the absence of B cell memory. Nat Commun 2017; 8:1781. [PMID: 29176567 PMCID: PMC5701209 DOI: 10.1038/s41467-017-01901-w] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/24/2017] [Indexed: 01/28/2023] Open
Abstract
Pre-existing serum antibodies play an important role in vaccine-mediated protection against infection but the underlying mechanisms of immune memory are unclear. Clinical studies indicate that antigen-specific antibody responses can be maintained for many years, leading to theories that reactivation/differentiation of memory B cells into plasma cells is required to sustain long-term antibody production. Here, we present a decade-long study in which we demonstrate site-specific survival of bone marrow-derived plasma cells and durable antibody responses to multiple virus and vaccine antigens in rhesus macaques for years after sustained memory B cell depletion. Moreover, BrdU+ cells with plasma cell morphology can be detected for 10 years after vaccination/BrdU administration, indicating that plasma cells may persist for a prolonged period of time in the absence of cell division. On the basis of these results, long-lived plasma cells represent a key cell population responsible for long-term antibody production and serological memory. The long-term maintenance of antibody-secreting plasma cells and the requirement for memory B cells are unclear. Here, the authors show that plasma cells and the antibodies secreted are long-lived and maintained over a decade in the absence of memory B cells in non-human primates.
Collapse
|
40
|
Sun J, Yuan Z, Abajas YL, Szollosi DE, Hu G, Hua B, Xiao X, Li C. A Retrospective Study of the Cytokine Profile Changes in Mice with FVIII Inhibitor Development After Adeno-Associated Virus-Mediated Gene Therapy in a Hemophilia A Mouse Model. Hum Gene Ther 2017; 29:381-389. [PMID: 28922951 DOI: 10.1089/hum.2017.094] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The development of inhibitory autoantibodies to the infused clotting factor VIII (FVIII) is a major complication for severe hemophilia A management. Novel therapy options for hemophilia have significantly progressed in the last decade, and a gene therapy cure for hemophilia is becoming a reality. However, mechanistic studies of FVIII autoantibodies (FVIII inhibitors) have lagged behind and remain a challenge for both protein replacement and gene therapy. FVIII inhibitor formation is assumed to be a classical T cell-dependent immune response in which cytokines/chemokines play an important role. The study of cytokine profile changes during FVIII inhibitor development may be helpful to understand the mechanism of inhibitor development and to explore potential novel approaches that will minimize the risk. After FVIII-/- mice were treated with intravenous administration of an adeno-associated virus 8 vector encoding human FVIII, FVIII expression peaked at week 2 (W2), and FVIII inhibitor was thoroughly developed at week 8 (W8). W8 plasma that showed positive FVIII inhibitor, and W2 samples with negative FVIII inhibitor (anti-FVIII[+]), were subjected to multiplex cytokines measurement. W8 and W2 samples were both negative for FVIII inhibitor (anti-FVIII[-]) as the control. In comparison to mice in the anti-FVIII(-) group, mice in the anti-FVIII(+) group exhibited significantly elevated pro-inflammatory cytokines of interleukin (IL)-1, IL-6, IL-12p40, monocyte chemoattractant protein-1, macrophage inflammatory protein (MIP)-1, MIP-2, and tumor necrosis factor alpha (TNF-α), especially at higher titers. The anti-inflammatory cytokine of transforming growth factor beta (TGF-β) was decreased at W2 in both groups. Multivariate analysis of the risk factors for FVIII inhibitor development showed peak FVIII activity at W2. IL-6 and TNF-α at W8 were positively correlated with inhibitor formation, and negatively correlated with the age starting gene therapy. Collectively, the elevated monocyte derived pro-inflammatory cytokines/chemokines, together with the decreased anti-inflammatory cytokine of TGF-β at an early time point, may contribute to the persistent inflammatory environment in favor of an immune response toward FVIII inhibitor development.
Collapse
Affiliation(s)
- Junjiang Sun
- 1 Gene Therapy Center, University of North Carolina , Chapel Hill, North Carolina.,2 Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina
| | - Zhenhua Yuan
- 2 Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina
| | - Yasmina L Abajas
- 3 Pediatric Hematology Oncology, University of North Carolina , Chapel Hill, North Carolina
| | - Doreen E Szollosi
- 4 Department of Pharmaceutical Sciences, University of Saint Joseph School of Pharmacy , Hartford, Connecticut
| | - Genlin Hu
- 1 Gene Therapy Center, University of North Carolina , Chapel Hill, North Carolina
| | - Baolai Hua
- 5 Department of Hematology, Northern Jiangsu People's Hospital , Yangzhou, China .,6 Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing, China
| | - Xiao Xiao
- 2 Division of Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina , Chapel Hill, North Carolina
| | - Chengwen Li
- 1 Gene Therapy Center, University of North Carolina , Chapel Hill, North Carolina.,7 Department of Pediatrics, University of North Carolina , Chapel Hill, North Carolina
| |
Collapse
|
41
|
van den Hoogen WJ, Laman JD, 't Hart BA. Modulation of Multiple Sclerosis and Its Animal Model Experimental Autoimmune Encephalomyelitis by Food and Gut Microbiota. Front Immunol 2017; 8:1081. [PMID: 28928747 PMCID: PMC5591889 DOI: 10.3389/fimmu.2017.01081] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 08/21/2017] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is an autoimmune neurological disease characterized by chronic inflammation of the central nervous system (CNS), leading to demyelination, axonal damage, and symptoms such as fatigue and disability. Although the cause of MS is not known, the infiltration of peripherally activated immune cells into the CNS has a key pathogenic role. Accumulating evidence supports an important role of diet and gut microbiota in immune-mediated diseases. Preclinical as well as clinical studies suggest a role for gut microbiota and dietary components in MS. Here, we review these recent studies on gut microbiota and dietary interventions in MS and its animal model experimental autoimmune encephalomyelitis. We also propose directions for future research.
Collapse
Affiliation(s)
| | - Jon D Laman
- Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Bert A 't Hart
- Department of Neuroscience, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.,Department of Immunobiology, Biomedical Primate Research Center, Rijswijk, Netherlands
| |
Collapse
|
42
|
An unexpected tetanus case. THE LANCET. INFECTIOUS DISEASES 2017; 16:746-752. [PMID: 27301930 DOI: 10.1016/s1473-3099(16)00075-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2015] [Revised: 12/29/2015] [Accepted: 02/01/2016] [Indexed: 10/21/2022]
Abstract
1 million cases of tetanus are estimated to occur worldwide each year, with more than 200 000 deaths. Tetanus is a life-threatening but preventable disease caused by a toxin produced by Clostridium tetani-a Gram-positive bacillus found in high concentrations in soil and animal excrement. Tetanus is almost completely preventable by active immunisation, but very rarely unexpected cases can occur in individuals who have been previously vaccinated. We report a case of generalised tetanus in a 22-year-old woman that arose despite the protective antitoxin antibody in her serum. The patient received all her vaccinations in the USA; her last vaccination was 6 years ago. The case was unusual because the patient had received all standard vaccinations, had no defined port of entry at disease onset, and had symptoms lasting for 6 months. Tetanus can present with unusual clinical forms; therefore, the diagnosis and management of this rare but difficult disease should be updated. In this Grand Round, we review the clinical features, epidemiology, treatment, and prognosis of C tetani infections.
Collapse
|
43
|
Brousseau N, Murphy DG, Gilca V, Larouche J, Mandal S, Tedder RS. Acute hepatitis B virus infection with delayed appearance of hepatitis B core antibody in an immunocompromised patient: a case report. J Med Case Rep 2017; 11:111. [PMID: 28412974 PMCID: PMC5393022 DOI: 10.1186/s13256-017-1264-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 03/09/2017] [Indexed: 11/29/2022] Open
Abstract
Background Despite the introduction of universal hepatitis B immunization programs worldwide, outbreaks of acute infection still occur in unimmunized individuals. A timely diagnosis of hepatitis B is necessary to ensure adequate clinical care and public health interventions that will reduce transmission. Yet, interpretation of hepatitis B serological markers can be complex. We present a case of hepatitis B with atypical markers, including delayed appearance of hepatitis B core antibody. Case presentation A 62-year-old white woman was identified as a sexual contact of a male individual with acute hepatitis B virus infection. She had a history of recurrent low-grade non-Hodgkin lymphoma and had recently received immunosuppressive therapy. At baseline she had a negative serology and received three double doses (40 μg) of Engerix-B vaccine (hepatitis B vaccine) with a 0-month, 1-month, and 6-month schedule. One month following the last dose, hepatitis B surface antigen was positive in the absence of hepatitis B core antibody. The only sign of infection was a slight elevation of alanine aminotransferase enzymes a few months after first sexual contacts with the male individual. Hepatitis B virus infection was later confirmed despite the absence of hepatitis B core antibody. The development of hepatitis B core antibody was finally noted more than 6 months after the first positive hepatitis B surface antigen and more than 12 months after elevation of alanine aminotransferase enzymes. Immunosuppression including rituximab treatment was the most likely explanation for this serological profile. On her last medical assessment, she had not developed HBeAg seroconversion despite lower hepatitis B virus deoxyribonucleic acid levels with tenofovir treatment. Conclusions When confronted with positive hepatitis B surface antigen in the absence of hepatitis B core antibody, consideration should be given to the possibility of both acute and persistent infection particularly in the setting of immunosuppression so that appropriate clinical management and public health interventions can take place. Given the increasing use of biologicals such as anti-tumor necrosis factor therapies either alone or with other immunosuppressive agents, this phenomenon may be encountered more frequently.
Collapse
Affiliation(s)
- Nicholas Brousseau
- CIUSSS de la Mauricie-et-Centre-du-Québec, 858 terrasse Turcotte, Trois-Rivières, Québec, G9A 5C5, Canada. .,Institut national de santé publique du Québec, 945 av Wolfe, Québec, G1V 5B3, Canada.
| | - Donald G Murphy
- Institut national de santé publique du Québec, 945 av Wolfe, Québec, G1V 5B3, Canada
| | - Vladimir Gilca
- Institut national de santé publique du Québec, 945 av Wolfe, Québec, G1V 5B3, Canada
| | - Jacynthe Larouche
- CIUSSS de la Mauricie-et-Centre-du-Québec, 858 terrasse Turcotte, Trois-Rivières, Québec, G9A 5C5, Canada
| | - Sema Mandal
- Public Health England, 61 Colindale Ave, London, NW9 5EQ, UK
| | | |
Collapse
|
44
|
Łusiak-Szelachowska M, Żaczek M, Weber-Dąbrowska B, Międzybrodzki R, Letkiewicz S, Fortuna W, Rogóż P, Szufnarowski K, Jończyk-Matysiak E, Olchawa E, Walaszek KM, Górski A. Antiphage activity of sera during phage therapy in relation to its outcome. Future Microbiol 2017; 12:109-117. [DOI: 10.2217/fmb-2016-0156] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Aim: The aim was to study the association between the phage neutralization of patients’ sera and the clinical outcome of phage therapy (PT). Patients: About 62 patients with various bacterial infections receiving PT as well as 30 healthy volunteers were studied. Materials & methods: Antiphage activity of sera (AAS) was examined using the phage neutralization test of different types of phages before and during PT in relation to the route of phage administration and correlated with the results of PT. Results & conclusion: The analysis of the association between AAS level and clinical results indicated that the level of AAS is not correlated with the outcome of PT.
Collapse
Affiliation(s)
- Marzanna Łusiak-Szelachowska
- Laboratory of Bacteriophages, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), Wrocław, Poland
| | - Maciej Żaczek
- Laboratory of Bacteriophages, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), Wrocław, Poland
| | - Beata Weber-Dąbrowska
- Laboratory of Bacteriophages, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), Wrocław, Poland
- Phage Therapy Unit, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Ryszard Międzybrodzki
- Laboratory of Bacteriophages, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), Wrocław, Poland
- Phage Therapy Unit, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
- Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, Warsaw, Poland
| | - Sławomir Letkiewicz
- Phage Therapy Unit, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
- Medical Sciences Institute, Katowice School of Economics, Katowice, Poland
| | - Wojciech Fortuna
- Laboratory of Bacteriophages, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), Wrocław, Poland
- Phage Therapy Unit, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Paweł Rogóż
- Phage Therapy Unit, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Krzysztof Szufnarowski
- Phage Therapy Unit, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Ewa Jończyk-Matysiak
- Laboratory of Bacteriophages, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), Wrocław, Poland
| | | | - Kinga M Walaszek
- Faculty of Chemistry, Wrocław University of Technology, Wrocław, Poland
| | - Andrzej Górski
- Laboratory of Bacteriophages, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences (HIIET PAS), Wrocław, Poland
- Phage Therapy Unit, Hirszfeld Institute of Immunology & Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
- Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
45
|
Żaczek M, Łusiak-Szelachowska M, Jończyk-Matysiak E, Weber-Dąbrowska B, Międzybrodzki R, Owczarek B, Kopciuch A, Fortuna W, Rogóż P, Górski A. Antibody Production in Response to Staphylococcal MS-1 Phage Cocktail in Patients Undergoing Phage Therapy. Front Microbiol 2016; 7:1681. [PMID: 27822205 PMCID: PMC5075762 DOI: 10.3389/fmicb.2016.01681] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 10/07/2016] [Indexed: 01/12/2023] Open
Abstract
In this study, we investigated the humoral immune response (through the release of IgG, IgA, and IgM antiphage antibodies) to a staphylococcal phage cocktail in patients undergoing experimental phage therapy at the Phage Therapy Unit, Medical Center of the Ludwik Hirszfeld Institute of Immunology and Experimental Therapy in Wrocław, Poland. We also evaluated whether occurring antiphage antibodies had neutralizing properties toward applied phages (K rate). Among 20 examined patients receiving the MS-1 phage cocktail orally and/or locally, the majority did not show a noticeably higher level of antiphage antibodies in their sera during phage administration. Even in those individual cases with an increased immune response, mostly by induction of IgG and IgM, the presence of antiphage antibodies did not translate into unsatisfactory clinical results of phage therapy. On the other hand, a negative outcome of the treatment occurred in some patients who showed relatively weak production of antiphage antibodies before and during treatment. This may imply that possible induction of antiphage antibodies is not an obstacle to the implementation of phage therapy and support our assumption that the outcome of the phage treatment does not primarily depend on the appearance of antiphage antibodies in sera of patients during therapy. These conclusions are in line with our previous findings. The confirmation of this thesis is of great interest as regards the efficacy of phage therapy in humans.
Collapse
Affiliation(s)
- Maciej Żaczek
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWrocław, Poland
| | - Marzanna Łusiak-Szelachowska
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWrocław, Poland
| | - Ewa Jończyk-Matysiak
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWrocław, Poland
| | - Beata Weber-Dąbrowska
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWrocław, Poland
- Phage Therapy Unit, Medical Center of the Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWrocław, Poland
| | - Ryszard Międzybrodzki
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWrocław, Poland
- Phage Therapy Unit, Medical Center of the Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWrocław, Poland
- Department of Clinical Immunology, Transplantation Institute, Medical University of WarsawWarsaw, Poland
| | - Barbara Owczarek
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWrocław, Poland
| | - Agnieszka Kopciuch
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWrocław, Poland
| | - Wojciech Fortuna
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWrocław, Poland
- Phage Therapy Unit, Medical Center of the Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWrocław, Poland
| | - Paweł Rogóż
- Phage Therapy Unit, Medical Center of the Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWrocław, Poland
| | - Andrzej Górski
- Bacteriophage Laboratory, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWrocław, Poland
- Phage Therapy Unit, Medical Center of the Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of SciencesWrocław, Poland
- Department of Clinical Immunology, Transplantation Institute, Medical University of WarsawWarsaw, Poland
| |
Collapse
|
46
|
Sánchez-Ramón S, Dhalla F, Chapel H. Challenges in the Role of Gammaglobulin Replacement Therapy and Vaccination Strategies for Hematological Malignancy. Front Immunol 2016; 7:317. [PMID: 27597852 PMCID: PMC4993076 DOI: 10.3389/fimmu.2016.00317] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/05/2016] [Indexed: 12/13/2022] Open
Abstract
Patients with chronic lymphocytic leukemia (CLL) and multiple myeloma (MM) are prone to present with antibody production deficits associated with recurrent or severe bacterial infections that might benefit from human immunoglobulin (Ig) (IVIg/SCIg) replacement therapy. However, the original IVIg trial data were done before modern therapies were available, and the current indications do not take into account the shift in the immune situation of current treatment combinations and changes in the spectrum of infections. Besides, patients affected by other B cell malignancies present with similar immunodeficiency and manifestations while they are not covered by the current IVIg indications. A potential beneficial strategy could be to vaccinate patients at monoclonal B lymphocytosis and monoclonal gammopathy of undetermined significance stages (for CLL and MM, respectively) or at B-cell malignancy diagnosis, when better antibody responses are attained. We have to re-emphasize the need for assessing and monitoring specific antibody responses; these are warranted to select adequately those patients for whom early intervention with prophylactic anti-infective therapy and/or IVIg is preferred. This review provides an overview of the current scenario, with a focus on prevention of infection in patients with hematological malignancies and the role of Ig replacement therapy.
Collapse
Affiliation(s)
- Silvia Sánchez-Ramón
- Department of Clinical Immunology and IdISSC, Hospital Clínico San Carlos, Madrid, Spain; Department of Microbiology I, Complutense University School of Medicine, Madrid, Spain
| | - Fatima Dhalla
- Nuffield Department of Medicine, University of Oxford, Oxford, UK; Department of Clinical Immunology, John Radcliffe Hospital, Headington, Oxford, UK
| | - Helen Chapel
- Nuffield Department of Medicine, University of Oxford, Oxford, UK; Department of Clinical Immunology, John Radcliffe Hospital, Headington, Oxford, UK
| |
Collapse
|
47
|
Persistence of long-lived plasma cells and humoral immunity in individuals responding to CD19-directed CAR T-cell therapy. Blood 2016; 128:360-70. [PMID: 27166358 DOI: 10.1182/blood-2016-01-694356] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 04/29/2016] [Indexed: 12/28/2022] Open
Abstract
The mechanisms underlying the maintenance of long-lasting humoral immunity are not well understood. Studies in mice indicate that plasma cells (PCs) can survive up to a lifetime, even in the absence of regeneration by B cells, implying the presence of long-lived PCs as a mechanism for long-lasting immunity. Evidence from humans treated with anti-CD20, which depletes circulating B cells, also suggests B-cell-independent long-term survival of some PCs. On the other hand, antibody responses may be sustained solely by short-lived PCs with repopulation from clonally related memory B cells. To explore PC longevity and humoral immunity in humans, we investigated the fate of PCs and their antibodies in adult and pediatric patients who received chimeric antigen receptor-based adoptive T-cell immunotherapy targeting CD19 to treat B-cell lineage malignancies (CTL019). Treatment with CTL019 is frequently associated with B-cell aplasia that can persist for years. Serum antibody titers to vaccine-related antigens were measured, and quantitative assessment of B cells and PCs in blood and bone marrow was performed at various time points before and after CTL019 therapy. While total serum immunoglobulin concentrations decline following CTL019-induced B-cell aplasia, several vaccine/pathogen-specific serum immunoglobulin G and A (IgG and IgA) titers remain relatively stable for at least 6 and 12 months posttreatment, respectively. Analysis of bone marrow biopsies after CTL019 revealed 8 patients with persistence of antibody-secreting PCs at least 25 months post-CTL019 infusion despite absence of CD19(+)CD20(+) B cells. These results provide strong evidence for the existence of memory B-cell-independent, long-lived PCs in humans that contribute to long-lasting humoral immunity.
Collapse
|
48
|
Hamad ARA, Ahmed R, Donner T, Fousteri G. B cell-targeted immunotherapy for type 1 diabetes: What can make it work? DISCOVERY MEDICINE 2016; 21:213-219. [PMID: 27115172 PMCID: PMC5266520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Immunotherapy has revolutionized treatment of cancers and autoimmune diseases. Bucking the trend, however, is type 1 diabetes (T1D), although it is one of best understood autoimmune diseases and individuals at genetic risk are identifiable with high certainty. Here we review the major obstacles associated with pan-B-cell-depletion using rituximab (RTX) and discuss the notion that B cell-directed therapy may be most effective as a preventive measure. We suggest that it will be more productive to aim at identifying and targeting autoreactive B cells rather than making adjustments to pan-B cell depletion and that non-conventional alternative therapies such as antibody blockade of FasL to bolster IL-10-producing Breg cells, which work successfully in mice, should be considered.
Collapse
Affiliation(s)
- Abdel Rahim A Hamad
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rizwan Ahmed
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Thomas Donner
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Georgia Fousteri
- Diabetes Research Institute (DRI), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
49
|
Abstract
Pediatric pemphigus vulgaris (PV) is a rare autoimmune bullous dermatosis that poses a diagnostic and therapeutic challenge. Rituximab, an anti-CD20 monoclonal antibody, is an important medication in adult PV but has rarely been used to treat pediatric disease. We describe successful rituximab therapy in a 4-year-old, the youngest patient with PV ever reported to receive rituximab. A live attenuated vaccination was later given without incident. We also review rituximab clinical outcomes, toxicity, dosing protocols, and relapse in children with PV.
Collapse
Affiliation(s)
- Leah Kincaid
- Division of Dermatology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,Section of Dermatology, Division of Pediatric Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Miriam Weinstein
- Division of Dermatology, Department of Medicine, University of Toronto, Toronto, Ontario, Canada.,Section of Dermatology, Division of Pediatric Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|
50
|
Bonami RH, Thomas JW. Targeting Anti-Insulin B Cell Receptors Improves Receptor Editing in Type 1 Diabetes-Prone Mice. THE JOURNAL OF IMMUNOLOGY 2015; 195:4730-41. [PMID: 26432895 DOI: 10.4049/jimmunol.1500438] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 09/08/2015] [Indexed: 12/11/2022]
Abstract
Autoreactive B lymphocytes that commonly arise in the developing repertoire can be salvaged by receptor editing, a central tolerance mechanism that alters BCR specificity through continued L chain rearrangement. It is unknown whether autoantigens with weak cross-linking potential, such as insulin, elicit receptor editing, or whether this process is dysregulated in related autoimmunity. To resolve these issues, we developed an editing-competent model in which anti-insulin Vκ125 was targeted to the Igκ locus and paired with anti-insulin VH125Tg. Physiologic, circulating insulin increased RAG-2 expression and was associated with BCR replacement that eliminated autoantigen recognition in a proportion of developing anti-insulin B lymphocytes. The proportion of anti-insulin B cells that underwent receptor editing was reduced in the type 1 diabetes-prone NOD strain relative to a nonautoimmune strain. Resistance to editing was associated with increased surface IgM expression on immature (but not transitional or mature) anti-insulin B cells in the NOD strain. The actions of mAb123 on central tolerance were also investigated, because selective targeting of insulin-occupied BCR by mAb123 eliminates anti-insulin B lymphocytes and prevents type 1 diabetes. Autoantigen targeting by mAb123 increased RAG-2 expression and dramatically enhanced BCR replacement in newly developed B lymphocytes. Administering F(ab')2123 induced IgM downregulation and reduced the frequency of anti-insulin B lymphocytes within the polyclonal repertoire of VH125Tg/NOD mice, suggesting enhanced central tolerance by direct BCR interaction. These findings indicate that weak or faulty checkpoints for central tolerance can be overcome by autoantigen-specific immunomodulatory therapy.
Collapse
Affiliation(s)
- Rachel H Bonami
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232; and
| | - James W Thomas
- Division of Rheumatology and Immunology, Department of Medicine, Vanderbilt University, Nashville, TN 37232; and Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville TN 37232
| |
Collapse
|