1
|
Crispin P, Henderson R, Yun J, Crosbie C, Tognarini D, Youssef S, Barrese G, Fleischmann S. Outcomes of a patient support programme for subcutaneous immunoglobulin therapy in patients with primary or secondary immunodeficiencies or chronic inflammatory demyelinating polyneuropathy. Intern Med J 2024; 54:1827-1837. [PMID: 39291857 DOI: 10.1111/imj.16520] [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: 02/09/2024] [Accepted: 08/04/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Subcutaneous immunoglobulin (SCIg) therapy is important in the treatment of primary (PID) and secondary immunodeficiencies (SID) and chronic inflammatory demyelinating polyneuropathy (CIDP). Patient support programmes (PSPs) help patients self-administer medication regimens and play a more active role in the self-management of their medical conditions. AIM To describe the effectiveness of the CSL Behring CARES PSP in optimising the quality use of SCIg in a hospital-free environment. DESIGN This retrospective, observational study analysed records of patients enroled in the CSL Behring CARES PSP. Key outcomes were accessibility and effectiveness. Data were extracted from the patient database and analysed using descriptive methods. RESULTS Seven hundred eighty-nine patients with PID (30.8%), SID (53.4%) and CIDP (15.8%) were enroled in the CARES PSP, 92.8% of whom were referred from public hospitals and the remaining from private hospitals. Of the total patient population, 697 (88.3%) received the nurse-led SCIg self-administration training and education (COACH), out of which 656 (94.1%) completed training and achieved competency after an average of 2.3 training sessions. The proportions of patients who achieved competency were similar across age groups and prior SCIg hospital education status. CONCLUSION This is the largest real-world evidence study that describes the effectiveness of SCIg PSPs across three therapeutic disease states. These PSPs can optimise hospital resources such as infusion nurse time and allocation of infusion chairs that were once used for intravenous immunoglobulin infusions, improve patient access to SCIg therapy and enable patients self-administer SCIg outside a hospital environment.
Collapse
Affiliation(s)
- Philip Crispin
- School of Medicine and Psychology, Australian National University, Canberra, Australian Capital Territory, Australia
- Haematology Department, The Canberra Hospital, Canberra, Australian Capital Territory, Australia
| | - Robert Henderson
- Department of Neurology, Royal Brisbane & Women's Hospital, Brisbane, Queensland, Australia
| | - James Yun
- Department of Immunology, Prince of Wales Hospital, Sydney, New South Wales, Australia
| | - Christina Crosbie
- Haematology Department, Sir Charles Gardiner Hospital, Perth, Western Australia, Australia
| | | | | | | | | |
Collapse
|
2
|
Farini A, Tripodi L, Villa C, Strati F, Facoetti A, Baselli G, Troisi J, Landolfi A, Lonati C, Molinaro D, Wintzinger M, Gatti S, Cassani B, Caprioli F, Facciotti F, Quattrocelli M, Torrente Y. Microbiota dysbiosis influences immune system and muscle pathophysiology of dystrophin-deficient mice. EMBO Mol Med 2023; 15:e16244. [PMID: 36533294 PMCID: PMC9994487 DOI: 10.15252/emmm.202216244] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 11/24/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive severe muscle-wasting disease caused by mutations in DMD, encoding dystrophin, that leads to loss of muscle function with cardiac/respiratory failure and premature death. Since dystrophic muscles are sensed by infiltrating inflammatory cells and gut microbial communities can cause immune dysregulation and metabolic syndrome, we sought to investigate whether intestinal bacteria support the muscle immune response in mdx dystrophic murine model. We highlighted a strong correlation between DMD disease features and the relative abundance of Prevotella. Furthermore, the absence of gut microbes through the generation of mdx germ-free animal model, as well as modulation of the microbial community structure by antibiotic treatment, influenced muscle immunity and fibrosis. Intestinal colonization of mdx mice with eubiotic microbiota was sufficient to reduce inflammation and improve muscle pathology and function. This work identifies a potential role for the gut microbiota in the pathogenesis of DMD.
Collapse
Affiliation(s)
- Andrea Farini
- Neurology UnitFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
| | - Luana Tripodi
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Dino Ferrari CenterUniversity of MilanMilanItaly
| | - Chiara Villa
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Dino Ferrari CenterUniversity of MilanMilanItaly
| | - Francesco Strati
- Mucosal Immunology Lab, Department of Experimental OncologyIEO‐European Institute of OncologyMilanItaly
| | - Amanda Facoetti
- Humanitas UniversityMilanItaly
- Humanitas Clinical and Research Center IRCCSMilanItaly
| | - Guido Baselli
- Translational Medicine – Department of Transfusion Medicine and HematologyFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
- Present address:
SciLifeLab, Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetSolnaSweden
| | - Jacopo Troisi
- Department of Medicine, Surgery and Dentistry, Scuola Medica SalernitanaUniversity of SalernoBaronissiItaly
- Theoreo Srl, Spinoff Company of the University of SalernoMontecorvino PuglianoItaly
| | - Annamaria Landolfi
- Department of Medicine, Surgery and Dentistry, Scuola Medica SalernitanaUniversity of SalernoBaronissiItaly
- Theoreo Srl, Spinoff Company of the University of SalernoMontecorvino PuglianoItaly
| | - Caterina Lonati
- Center for Surgical ResearchFondazione IRCCS Ca' Granda, Ospedale Maggiore PoliclinicoMilanItaly
| | - Davide Molinaro
- Neurology UnitFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Dino Ferrari CenterUniversity of MilanMilanItaly
| | - Michelle Wintzinger
- Molecular Cardiovascular Biology Division, Heart InstituteCincinnati Children's Hospital Medical CenterCincinnatiOHUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOHUSA
| | - Stefano Gatti
- Center for Surgical ResearchFondazione IRCCS Ca' Granda, Ospedale Maggiore PoliclinicoMilanItaly
| | - Barbara Cassani
- Humanitas Clinical and Research Center IRCCSMilanItaly
- Department of Medical Biotechnologies and Translational MedicineUniversità Degli Studi di MilanoMilanItaly
| | - Flavio Caprioli
- Unit of Gastroenterology and Endoscopy, Department of Pathophysiology and TransplantationUniversità degli Studi di Milano, Fondazione IRCCS Ca' Granda, Ospedale Policlinico di MilanoMilanItaly
| | - Federica Facciotti
- Unit of Gastroenterology and Endoscopy, Department of Pathophysiology and TransplantationUniversità degli Studi di Milano, Fondazione IRCCS Ca' Granda, Ospedale Policlinico di MilanoMilanItaly
| | - Mattia Quattrocelli
- Molecular Cardiovascular Biology Division, Heart InstituteCincinnati Children's Hospital Medical CenterCincinnatiOHUSA
- Department of PediatricsUniversity of Cincinnati College of MedicineCincinnatiOHUSA
| | - Yvan Torrente
- Neurology UnitFondazione IRCCS Ca' Granda Ospedale Maggiore PoliclinicoMilanItaly
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Dino Ferrari CenterUniversity of MilanMilanItaly
| |
Collapse
|
3
|
Farini A, Villa C, Tripodi L, Legato M, Torrente Y. Role of Immunoglobulins in Muscular Dystrophies and Inflammatory Myopathies. Front Immunol 2021; 12:666879. [PMID: 34335568 PMCID: PMC8316973 DOI: 10.3389/fimmu.2021.666879] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/25/2021] [Indexed: 01/15/2023] Open
Abstract
Muscular dystrophies and inflammatory myopathies are heterogeneous muscular disorders characterized by progressive muscle weakness and mass loss. Despite the high variability of etiology, inflammation and involvement of both innate and adaptive immune response are shared features. The best understood immune mechanisms involved in these pathologies include complement cascade activation, auto-antibodies directed against muscular proteins or de-novo expressed antigens in myofibers, MHC-I overexpression in myofibers, and lymphocytes-mediated cytotoxicity. Intravenous immunoglobulins (IVIGs) administration could represent a suitable immunomodulator with this respect. Here we focus on mechanisms of action of immunoglobulins in muscular dystrophies and inflammatory myopathies highlighting results of IVIGs from pre-clinical and case reports evidences.
Collapse
Affiliation(s)
- Andrea Farini
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, University of Milan, Dino Ferrari Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | | | | | - Yvan Torrente
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, University of Milan, Dino Ferrari Center, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
4
|
Wehrli M, Schneider C, Cortinas-Elizondo F, Verschoor D, Frias Boligan K, Adams OJ, Hlushchuk R, Engelmann C, Daudel F, Villiger PM, Seibold F, Yawalkar N, Vonarburg C, Miescher S, Lötscher M, Kaufmann T, Münz C, Mueller C, Djonov V, Simon HU, von Gunten S. IgA Triggers Cell Death of Neutrophils When Primed by Inflammatory Mediators. THE JOURNAL OF IMMUNOLOGY 2020; 205:2640-2648. [PMID: 33008951 DOI: 10.4049/jimmunol.1900883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 09/10/2020] [Indexed: 12/14/2022]
Abstract
IVIG preparations consisting of pooled IgG are increasingly used for the treatment of autoimmune diseases. IVIG is known to regulate the viability of immune cells, including neutrophils. We report that plasma-derived IgA efficiently triggers death of neutrophils primed by cytokines or TLR agonists. IgA-mediated programmed neutrophil death was PI3K-, p38 MAPK-, and JNK-dependent and evoked anti-inflammatory cytokines in macrophage cocultures. Neutrophils from patients with acute Crohn's disease, rheumatoid arthritis, or sepsis were susceptible to both IgA- and IVIG-mediated death. In contrast to IVIG, IgA did not promote cell death of quiescent neutrophils. Our findings suggest that plasma-derived IgA might provide a therapeutic option for the treatment of neutrophil-associated inflammatory disorders.
Collapse
Affiliation(s)
- Marc Wehrli
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | | | | | | | | | - Olivia Joan Adams
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Ruslan Hlushchuk
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland
| | - Christine Engelmann
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Fritz Daudel
- Intensive Care Unit, Spital Thun, 3600 Thun, Switzerland
| | - Peter M Villiger
- Department of Rheumatology/Clinical Immunology/Allergology, University Hospital Bern, 3008 Bern, Switzerland
| | - Frank Seibold
- Gastroenterologie, Spitalnetz Bern, 3004 Bern, Switzerland.,Gastroenterologie, Praxis Balsiger, Seibold und Partner am Lindenhofspital, 3012 Bern, Switzerland
| | - Nikhil Yawalkar
- Department of Dermatology, University Hospital Bern, University of Bern, 3010 Bern, Switzerland
| | | | | | | | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Christoph Mueller
- Institute of Pathology, University of Bern, 3008 Bern, Switzerland; and
| | - Valentin Djonov
- Institute of Anatomy, University of Bern, 3012 Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, 3010 Bern, Switzerland.,Department of Clinical Immunology and Allergology, Sechenov University, Moscow 119991, Russia
| | | |
Collapse
|
5
|
Gumanova NG. Analytical complex of biochemical markers for preclinical diagnosis and prevention of cardiovascular diseases. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2019. [DOI: 10.15829/1728-8800-2019-5-117-127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Affiliation(s)
- N. G. Gumanova
- National Medical Research Center for Preventive Medicine
| |
Collapse
|
6
|
Tortorici MA, Lawo JP, Weide R, Jochems J, Puli S, Hofmann J, Pfruender D, Rojavin MA. Privigen® has similar pharmacokinetic properties in primary and secondary immune deficiency. Int Immunopharmacol 2019; 66:119-126. [DOI: 10.1016/j.intimp.2018.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/05/2018] [Accepted: 11/06/2018] [Indexed: 10/27/2022]
|
7
|
Mallick R, Jolles S, Kanegane H, Agbor-Tarh D, Rojavin M. Treatment Satisfaction with Subcutaneous Immunoglobulin Replacement Therapy in Patients with Primary Immunodeficiency: a Pooled Analysis of Six Hizentra® Studies. J Clin Immunol 2018; 38:886-897. [PMID: 30465179 PMCID: PMC6292975 DOI: 10.1007/s10875-018-0562-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 10/15/2018] [Indexed: 12/21/2022]
Abstract
Purpose Primary immunodeficiency diseases (PIDDs) are a heterogenous group of disorders characterized by intrinsic impairment in the immune system. Most patients with PIDD require life-long immunoglobulin G replacement therapy, which has been shown to reduce the rate of infections and, related hospitalizations and reduce health-related quality of life (HRQOL). Here, treatment satisfaction and HRQOL in patients with PIDD was evaluated upon switching from intravenous (IVIG) or subcutaneous immunoglobulins (SCIGs) to 20% SCIG (Hizentra®), and during long-term steady-state Hizentra® treatment. Methods Analyses were based on two pivotal (switch) and four extension/follow-up (maintenance) Phase III studies of Hizentra® conducted in Europe (EU), Japan (JP), and the United States (US). Two validated questionnaires were used: Life Quality Index (LQI) for assessment of IgG-specific perceptions of HRQOL and Short Form 36 version 2 (SF-36v2). Results In the EU and JP switch studies, there was significant and meaningful improvement from Screening in LQI domain scores at all time points, largely driven by patients switching from IVIG to SCIG. In the EU switch study, there were also significant increases in mean SF-36v2 domain scores for Physical Function and General Health from Screening to Week 12. These improvements were observed also at Week 24. Overall, LQI and SF-36v2 domain scores were generally sustained in the maintenance studies. Conclusions These results showed that switching patients from IVIG to SCIG improves patient self-reported health status and IgG-specific HRQOL perception. The maintenance studies generally showed no deterioration of this improved health status over a long follow-up period. Electronic supplementary material The online version of this article (10.1007/s10875-018-0562-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | - Hirokazu Kanegane
- Department of Child Health and Development, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | |
Collapse
|
8
|
Jolles S, Rojavin MA, Lawo JP, Nelson R, Wasserman RL, Borte M, Tortorici MA, Imai K, Kanegane H. Long-Term Efficacy and Safety of Hizentra® in Patients with Primary Immunodeficiency in Japan, Europe, and the United States: a Review of 7 Phase 3 Trials. J Clin Immunol 2018; 38:864-875. [PMID: 30415311 PMCID: PMC6292970 DOI: 10.1007/s10875-018-0560-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 10/04/2018] [Indexed: 12/28/2022]
Abstract
Many patients with primary immunodeficiency (PID) require immunoglobulin G (IgG) replacement therapy, delivered as intravenous IgG (IVIG) or subcutaneous IgG (SCIG). We aim to identify trends in efficacy and safety that would not be evident in individual studies of small patient numbers. Seven open-label, Phase 3, prospective, multicenter studies of the efficacy and safety of Hizentra® (a SCIG), conducted in Japan, Europe, and the US were summarized. Overall, 125 unique patients received 15,013 weekly infusions during a total observation period of 250.9 patient-years. Mean weekly doses of Hizentra® were 83.22–221.3 mg/kg body weight; infusion rates per patient (total body rate) were 25.2–49.3 mL/h across studies. The rates of infections and serious bacterial infections were 3.10 and 0.03 events per patient/year, respectively. Annualized rates of days hospitalized due to infection, out of work/school, and prophylactic antibiotic use were 0.95, 5.14, and 36.78 per patient, respectively. For the equivalent monthly dose, weekly Hizentra® SCIG administration resulted in expectedly-increased serum IgG trough levels in patients switching from IVIG, and maintained levels in patients switching from previous SCIG. Adverse events (AEs) totaled 5039 (events/infusion 0.094–0.773), almost all of which were mild/moderate. Three thousand one hundred ninety-seven were considered treatment-related, the most common of which were injection site reactions (2919 events; 0.001–0.592 AEs per infusion). Systemic AEs were very uncommon. The results from these seven studies indicate that Hizentra® therapy was both efficacious and well tolerated during long-term treatment. This is particularly important in patients with PID, who may require lifelong IgG replacement therapy.
Collapse
Affiliation(s)
- Stephen Jolles
- Immunodeficiency Centre for Wales, University Hospital of Wales, Cardiff, UK.
| | | | | | - Robert Nelson
- School of Medicine and the Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, USA
| | | | - Michael Borte
- Hospital St. Georg GmbH Leipzig, Academic Teaching Hospital of the University of Leipzig, Leipzig, Germany
| | | | - Kohsuke Imai
- Department of Community Pediatrics, Perinatal and Maternal Medicine, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hirokazu Kanegane
- Department of Child Health and Development, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
9
|
Pecoraro A, Crescenzi L, Granata F, Genovese A, Spadaro G. Immunoglobulin replacement therapy in primary and secondary antibody deficiency: The correct clinical approach. Int Immunopharmacol 2017; 52:136-142. [DOI: 10.1016/j.intimp.2017.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/04/2017] [Accepted: 09/07/2017] [Indexed: 12/14/2022]
|
10
|
Nunes BG, Loures FV, Bueno HMS, Cangussu EB, Goulart E, Coatti GC, Caldini EG, Condino-Neto A, Zatz M. Immunoglobulin therapy ameliorates the phenotype and increases lifespan in the severely affected dystrophin-utrophin double knockout mice. Eur J Hum Genet 2017; 25:1388-1396. [PMID: 29255177 DOI: 10.1038/s41431-017-0017-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 09/13/2017] [Accepted: 09/14/2017] [Indexed: 12/15/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive disorder, caused by mutations in the dystrophin gene, affecting 1:3500-5000 boys worldwide. The lack of dystrophin induces degeneration of muscle cells and elicits an immune response characterized by an intensive secretion of pro-inflammatory cytokines. Immunoglobulins modulate the inflammatory response through several mechanisms and have been widely used as an adjuvant therapy for autoimmune diseases. Here we evaluated the effect of immunoglobulin G (IG) injected intraperitoneally in a severely affected double knockout (dko) mouse model for Duchenne muscular dystrophy. The IG dko treated mice were compared regarding activity rates, survival and histopathology with a control untreated group. Additionally, dendritic cells and naïve lymphocytes from these two groups and WT mice were obtained to study in vitro the role of the immune system associated to DMD pathophysiology. We show that IG therapy significantly enhances activity rate and lifespan of dko mice. It diminishes muscle tissue inflammation by decreasing the expression of costimulatory molecules MHC, CD86 and CD40 and reducing Th1-related cytokines IFN-γ, IL-1β and TNF-α release. IG therapy dampens the effector immune responses supporting the hypothesis according to which the immune response accelerates DMD progression. As IG therapy is already approved by FDA for treating autoimmune disorders, with less side-effects than currently used glucocorticoids, our results may open a new therapeutic option aiming to improve life quality and lifespan of DMD patients.
Collapse
Affiliation(s)
- Bruno Ghirotto Nunes
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, Sao Paulo, SP, Brazil
| | - Flávio Vieira Loures
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, SP, Brazil
| | - Heloisa Maria Siqueira Bueno
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, Sao Paulo, SP, Brazil
| | - Erica Baroni Cangussu
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, Sao Paulo, SP, Brazil
| | - Ernesto Goulart
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, Sao Paulo, SP, Brazil
| | - Giuliana Castello Coatti
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, Sao Paulo, SP, Brazil
| | - Elia Garcia Caldini
- Department of Pathology, School of Medicine, University of São Paulo, Sao Paulo, SP, Brazil
| | - Antonio Condino-Neto
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, Sao Paulo, SP, Brazil.
| | - Mayana Zatz
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, Sao Paulo, SP, Brazil.
| |
Collapse
|
11
|
Schneider C, Wicki S, Graeter S, Timcheva TM, Keller CW, Quast I, Leontyev D, Djoumerska-Alexieva IK, Käsermann F, Jakob SM, Dimitrova PA, Branch DR, Cummings RD, Lünemann JD, Kaufmann T, Simon HU, von Gunten S. IVIG regulates the survival of human but not mouse neutrophils. Sci Rep 2017; 7:1296. [PMID: 28465620 PMCID: PMC5430961 DOI: 10.1038/s41598-017-01404-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 03/30/2017] [Indexed: 12/12/2022] Open
Abstract
Intravenous immunoglobulin (IVIG) are purified IgG preparations made from the pooled plasma from thousands of healthy donors and are being tested in preclinical mouse models. Inherent challenges, however, are the pluripotency of IVIG and its xenogeneicity in animals. IVIG can alter the viability of human neutrophils via agonistic antibodies to Fas and Siglec-9. In this study, we compared the effects of IVIG on human and mouse neutrophils using different death assays. Different commercial IVIG preparations similarly induced cytokine-dependent death in human neutrophils, whereas they had no effects on the survival of either peripheral blood or bone marrow neutrophils from C57BL/6 or BALB/c mice. F(ab’)2 but not Fc fragments of IVIG induced death of human neutrophils, whereas neither of these IVIG fragments, nor agonistic monoclonal antibodies to human Fas or Siglec-9 affected the viability of mouse neutrophils. Pooled mouse IgG, which exhibited a different immunoprofile compared to IVIG, also had no effect on mouse cells. Together, these observations demonstrate that effects of IVIG on neutrophil survival are not adequately reflected in current mouse models, despite the key role of these cells in human inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
| | - Simone Wicki
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Stefanie Graeter
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | | | - Christian W Keller
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland
| | - Isaak Quast
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland.,Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - Danila Leontyev
- Department of Medicine, University of Toronto and Centre for Innovation, Canadian Blood Services, Toronto, Ontario, Canada
| | - Iglika K Djoumerska-Alexieva
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | | | - Stephan M Jakob
- Department of Intensive Care Medicine, University Hospital Bern (Inselspital), University of Bern, Bern, Switzerland
| | - Petya A Dimitrova
- Department of Immunology, Stefan Angelov Institute of Microbiology, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Donald R Branch
- Department of Medicine, University of Toronto and Centre for Innovation, Canadian Blood Services, Toronto, Ontario, Canada
| | - Richard D Cummings
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Jan D Lünemann
- Institute of Experimental Immunology, Laboratory of Neuroinflammation, University of Zurich, Zurich, Switzerland
| | - Thomas Kaufmann
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Hans-Uwe Simon
- Institute of Pharmacology, University of Bern, Bern, Switzerland
| | | |
Collapse
|
12
|
Späth PJ, Schneider C, von Gunten S. Clinical Use and Therapeutic Potential of IVIG/SCIG, Plasma-Derived IgA or IgM, and Other Alternative Immunoglobulin Preparations. Arch Immunol Ther Exp (Warsz) 2016; 65:215-231. [DOI: 10.1007/s00005-016-0422-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 08/31/2016] [Indexed: 12/22/2022]
|
13
|
Lee CY, Lin WC, Wu MS, Yang CY, Yeh CC, Tsai MK. Repeated cycles of high-dose intravenous immunoglobulin and plasmapheresis for treatment of late antibody-mediated rejection of renal transplants. J Formos Med Assoc 2016; 115:845-852. [PMID: 27542515 DOI: 10.1016/j.jfma.2016.07.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 07/05/2016] [Accepted: 07/05/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND/PURPOSE Intravenous immunoglobulin (IVIG) plays a central role in the treatment of antibody-mediated rejection (AMR) of renal allografts, but the treatment outcomes for late AMR (>6 months after transplantation) are poor. METHODS We performed a retrospective study to assess the response patterns of IVIG-based (2 g/kg) desensitization for late AMR. Patients who received desensitization after the pathological diagnosis of late AMR positive for complement component C4d were grouped as the Desensitized Group and compared to a historical Control Group with complement component C4d positivity in retrospective stainings. RESULTS The 10-year graft survival of the Desensitized Group (73.9%, n = 35) was significantly better than that of the historical Control Group (35.0%, n = 40) without desensitization. In the Desensitized Group, a subgroup of patients (D2 Subgroup, n = 11), who responded to desensitization initially but deteriorated later, was identified to benefit from repeated cycles of desensitization at 31.1 ± 20.9 months. Patients receiving only one cycle of desensitization were further grouped into D1-good (n = 10) and D1-poor (n = 14) based on their long-term renal function. The D2 Subgroup patients did not exhibit significant improvements in renal function compared to the D1-poor patients, until 30 months after IVIG-based desensitization, suggesting desensitization therapy has a working window of approximately 24 months. CONCLUSION Repeated cycles of IVIG-based desensitization help stabilize long-term renal function in patients with persistent AMR.
Collapse
Affiliation(s)
- Chih-Yuan Lee
- Department of Surgery, National Taiwan University Hospital and National Taiwan University, College of Medicine, Taipei, Taiwan
| | - Wei-Chou Lin
- Department of Pathology, National Taiwan University Hospital and National Taiwan University, College of Medicine, Taipei, Taiwan
| | - Ming-Shiou Wu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University, College of Medicine, Taipei, Taiwan
| | - Ching-Yao Yang
- Department of Surgery, National Taiwan University Hospital and National Taiwan University, College of Medicine, Taipei, Taiwan
| | - Chi-Chuan Yeh
- Department of Surgery, National Taiwan University Hospital and National Taiwan University, College of Medicine, Taipei, Taiwan
| | - Meng-Kun Tsai
- Department of Surgery, National Taiwan University Hospital and National Taiwan University, College of Medicine, Taipei, Taiwan.
| |
Collapse
|