scholarly journals Neutralizing and binding activities against SARS‐CoV ‐1/2, MERS‐CoV , and human coronaviruses 229E and OC43 by normal human intravenous immunoglobulin derived from healthy donors in Japan

Transfusion ◽  
2020 ◽  
Author(s):  
Ritsuko Kubota‐Koketsu ◽  
Yutaka Terada ◽  
Mikihiro Yunoki ◽  
Tadahiro Sasaki ◽  
Emi E. Nakayama ◽  
...  
2021 ◽  
Vol 100 (2) ◽  
pp. 174-181
Author(s):  
S.S. Vakhlayrskaya ◽  
◽  
M.N. Kostyleva ◽  
A.S. Botkina ◽  
E.S. Ilyina ◽  
...  

Human intravenous immunoglobulin (IVIG) – a blood product prepared from the serum of many healthy donors, is the mainstay of therapy for treatment of primary immunodeficiencies, is increasingly used in various fields of medicine. The article reflects practical aspects of use of this group of drugs in a multidisciplinary pediatric hospital not only taking the therapy of defects of the immune system as an example, but also oncohematological, neurological, dermatological diseases, including orphan diseases. The modern data on the mechanisms of action of immunoglobulins, schemes of use for various pathologies are presented. The issues of safety of application, interchangeability of immunoglobulins from different manufacturers are considered.


2021 ◽  
Author(s):  
Marinos C. Dalakas

AbstractIn the last 25 years, intravenous immunoglobulin (IVIg) has had a major impact in the successful treatment of previously untreatable or poorly controlled autoimmune neurological disorders. Derived from thousands of healthy donors, IVIg contains IgG1 isotypes of idiotypic antibodies that have the potential to bind pathogenic autoantibodies or cross-react with various antigenic peptides, including proteins conserved among the “common cold”-pre-pandemic coronaviruses; as a result, after IVIg infusions, some of the patients’ sera may transiently become positive for various neuronal antibodies, even for anti-SARS-CoV-2, necessitating caution in separating antibodies derived from the infused IVIg or acquired humoral immunity. IVIg exerts multiple effects on the immunoregulatory network by variably affecting autoantibodies, complement activation, FcRn saturation, FcγRIIb receptors, cytokines, and inflammatory mediators. Based on randomized controlled trials, IVIg is approved for the treatment of GBS, CIDP, MMN and dermatomyositis; has been effective in, myasthenia gravis exacerbations, and stiff-person syndrome; and exhibits convincing efficacy in autoimmune epilepsy, neuromyelitis, and autoimmune encephalitis. Recent evidence suggests that polymorphisms in the genes encoding FcRn and FcγRIIB may influence the catabolism of infused IgG or its anti-inflammatory effects, impacting on individualized dosing or efficacy. For chronic maintenance therapy, IVIg and subcutaneous IgG are effective in controlled studies only in CIDP and MMN preventing relapses and axonal loss up to 48 weeks; in practice, however, IVIg is continuously used for years in all the aforementioned neurological conditions, like is a “forever necessary therapy” for maintaining stability, generating challenges on when and how to stop it. Because about 35-40% of patients on chronic therapy do not exhibit objective neurological signs of worsening after stopping IVIg but express subjective symptoms of fatigue, pains, spasms, or a feeling of generalized weakness, a conditioning effect combined with fear that discontinuing chronic therapy may destabilize a multi-year stability status is likely. The dilemmas of continuing chronic therapy, the importance of adjusting dosing and scheduling or periodically stopping IVIg to objectively assess necessity, and concerns in accurately interpreting IVIg-dependency are discussed. Finally, the merit of subcutaneous IgG, the ineffectiveness of IVIg in IgG4-neurological autoimmunities, and genetic factors affecting IVIg dosing and efficacy are addressed.


Sign in / Sign up

Export Citation Format

Share Document