Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-32514273

RESUMO

In this letter to the editor, we present results of claims data analysis. This claims data analysis supports a hypothesis that in patients with hereditary angioedema due to C1-esterase inhibitor (C1-INH) deficiency, the occurrence and/or symptomatology of coexisting autoimmune disease may be positively influenced by a replacement therapy with plasma derived C1-INH.

2.
Transfusion ; 58 Suppl 3: 3096-3105, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30536430

RESUMO

Immune-deficient patients depend on the antibodies in pooled human immunoglobulin G (IgG) preparations to remain free from serious infections. The potency of IgG preparations is therefore an ongoing concern. The use of pooled IgG to prevent infection is based on the concept that healthy adults have recovered from infections earlier in life and maintain relatively high antibody titers. In general, vaccine-induced immunity is less robust or long-lasting than immunity after natural infection, and many infectious diseases which were formerly widely prevalent have become much less common due to improved hygiene and vaccines. This raises questions as to the adequacy of protective antibodies in current IgG preparations. This paper reviews available data on antibodies against selected bacterial and virus vaccine antigens in current IgG products. Most products contain sufficient antibody to yield levels above minimal protective concentrations to a broad range of pathogens and toxins. Illustrative examples of effects of vaccines on antibody content of IgG products are also discussed: antibody titers to hepatitis A virus in donor plasma pools in both the US and EU are dropping due to decreased natural infection, but they are still sufficient to provide robust protection. Increasing seroprevalence of hepatitis B virus as a result of immunization suggests that antibody titers against this virus may actually be increasing. Finally, serial studies suggest that pooled IgG provides protection against seasonal influenza viruses despite year-to-year antigenic drift, and is also likely to provide at least some protective antibody against potentially pandemic strains.


Assuntos
Anticorpos Antivirais/análise , Imunoglobulina G/administração & dosagem , Imunoglobulina G/imunologia , Vacinas Virais/imunologia , Adulto , Anticorpos Neutralizantes/imunologia , Anticorpos Neutralizantes/uso terapêutico , Anticorpos Antivirais/sangue , Anticorpos Antivirais/uso terapêutico , Bactérias/imunologia , Toxinas Bacterianas/imunologia , Combinação de Medicamentos , Humanos , Imunoglobulina G/uso terapêutico , Ligação Proteica , Vacinação/métodos
3.
Immunotherapy ; 10(11): 919-933, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29764262

RESUMO

Subcutaneous IgG (SCIG) administration may be preferred over the intravenous route (IVIG) in chronic inflammatory demyelinating polyneuropathy (CIDP) because it minimizes 'end of cycle' treatment-related fluctuations, reduces systemic adverse effects, improves convenience/quality of life and potentially lowers overall costs. Early reports of the use of highly concentrated SCIG preparations suggested they were effective and well-tolerated in chronic inflammatory demyelinating polyneuropathy. This was confirmed in the Polyneuropathy and Treatment with Hizentra® study of 172 subjects randomized to receive maintenance therapy with placebo or one of two doses of IgPro20 (20% IgG stabilized with L-Proline) for 6 months. Risk of relapse was reduced by SCIG in a dose-related manner as compared with placebo. A total of 88% of polyneuropathy and treatment with hizentra subjects felt the subcutaneous method was 'easy to learn'. Local adverse events were mostly mild or moderate, and systemic adverse events were infrequent. Some patients may prefer maintenance therapy with SCIG over IVIG.


Assuntos
Imunoglobulina G/uso terapêutico , Imunoglobulinas Intravenosas/uso terapêutico , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/tratamento farmacológico , Autoimunidade , Humanos , Injeções Subcutâneas , Estimativa de Kaplan-Meier , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/mortalidade , Qualidade de Vida , Ensaios Clínicos Controlados Aleatórios como Assunto , Resultado do Tratamento
4.
J Peripher Nerv Syst ; 23(2): 78-87, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29573033

RESUMO

Despite the well-recognized importance of immunoglobulin therapy individualization during the treatment of chronic inflammatory demyelinating polyneuropathy (CIDP), the pathway to best achieve optimization is unknown. There are many pharmacokinetic and immunobiologic variables that can potentially influence the appropriateness of any individual therapy. Although identification of specific autoantibodies and their targets has only been accomplished in a minority of patients with CIDP, already the diagnostic and treatment implications of specific autoantibody detection are being realized. Individual variability in IgG pharmacokinetic properties including IgG catabolic rates and distribution, as well as the IgG level necessary for disease control also require consideration during the optimization process. For optimization to be successful there must be a measure of treatment response that has a clinically meaningful interpretation. There are currently available well-defined and validated clinical assessment tools and outcome measures that are well suited for this purpose. While there remains much to learn on how best to manipulate immunopathology and immunoglobulin pharmacokinetics in the most favorable way, there currently exists an understanding of these principles to a degree sufficient to begin to develop rational and evidence-based treatment optimization strategies.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Imunoglobulinas Intravenosas/uso terapêutico , Doenças do Sistema Nervoso Periférico/tratamento farmacológico , Humanos , Imunoglobulinas Intravenosas/administração & dosagem , Doenças do Sistema Nervoso Periférico/imunologia , Medicina de Precisão , Resultado do Tratamento
5.
Blood Rev ; 32(2): 106-115, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-28958644

RESUMO

Immunoglobulins are used to prevent or reduce infection risk in primary immune deficiencies and in settings which exploit its anti-inflammatory and immune-modulatory effects. Rigorous proof of immunoglobulin efficacy in persons with lympho-proliferative neoplasms, plasma cell myeloma, and persons receiving hematopoietic cell transplants is lacking despite many clinical trials. Further, there are few consensus guidelines or algorithms for use in these conditions. Rapid development of new therapies targeting B-cell signaling and survival pathways and increased use of chimeric antigen receptor T-cell (CAR-T) therapy will likely result in more acquired deficiencies of humoral immunity and infections in persons with cancer. We review immunoglobulin formulations and discuss efficacy and potential adverse effects in the context of preventing infections and in graft-versus-host disease. We suggest an algorithm for evaluating acquired deficiencies of humoral immunity in persons with hematologic neoplasms and recommend appropriate use of immunoglobulin therapy.


Assuntos
Neoplasias Hematológicas/terapia , Imunização Passiva , Antineoplásicos Imunológicos/administração & dosagem , Antineoplásicos Imunológicos/efeitos adversos , Antineoplásicos Imunológicos/uso terapêutico , Doenças Autoimunes/complicações , Neoplasias Hematológicas/complicações , Neoplasias Hematológicas/diagnóstico , Neoplasias Hematológicas/etiologia , Transplante de Células-Tronco Hematopoéticas/efeitos adversos , Transplante de Células-Tronco Hematopoéticas/métodos , Humanos , Imunização Passiva/efeitos adversos , Imunização Passiva/métodos , Imunoterapia/efeitos adversos , Imunoterapia/métodos , Resultado do Tratamento
6.
Respir Med ; 132: 181-188, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29229095

RESUMO

BACKGROUND: More than half of all primary immune deficiency diseases (PIDD) affect antibody production and are well known as causes of recurrent sinusitis and lung infections. Chronic and recurrent infections of the upper and/or lower airways can contribute to inflammatory and obstructive processes in the lower airways which are initially reversible and considered "asthma", but can eventually cause irreversible remodeling and chronic obstructive pulmonary disease (COPD). Conversely, several lines of evidence suggest that many patients who present with a diagnosis of asthma have an increased incidence of infection, suggesting underlying host-defense defects. Asthma and respiratory infections in the first decades of life are recognized as risk factors for development of COPD, but when patients present with COPD as adults, underlying primary immune deficiency disease may be unrecognized. MAIN FINDINGS AND CONCLUSIONS: Detection of PIDD as a potentially treatable underlying contributor to recurrent/acute exacerbations and morbidity of COPD, and provision of immunoglobulin (Ig) G replacement therapy, when appropriate, may decrease the progression of COPD. Decreasing the severity and rate of exacerbations and admissions should improve the quality of life and longevity of an important subset of patients with COPD, while decreasing costs. Major steps toward achieving these goals include developing a high index of suspicion, more frequent use and appropriate interpretation of screening tests such as quantitative immunoglobulins and vaccine responses, and prompt institution of IgG replacement therapy when antibody deficiency has been diagnosed.


Assuntos
Asma/imunologia , Bronquiectasia/imunologia , Imunoglobulina G/imunologia , Síndromes de Imunodeficiência/imunologia , Doença Pulmonar Obstrutiva Crônica/imunologia , Infecções Respiratórias/imunologia , Rinite/imunologia , Sinusite/imunologia , Anticorpos/imunologia , Asma/etiologia , Bronquiectasia/etiologia , Doença Crônica , Progressão da Doença , Humanos , Imunoglobulinas Intravenosas/uso terapêutico , Síndromes de Imunodeficiência/complicações , Síndromes de Imunodeficiência/diagnóstico , Síndromes de Imunodeficiência/tratamento farmacológico , Fatores Imunológicos/uso terapêutico , Doença Pulmonar Obstrutiva Crônica/etiologia , Recidiva , Infecções Respiratórias/etiologia , Rinite/etiologia , Sinusite/etiologia , Vacinas/imunologia
7.
Autoimmun Rev ; 16(10): 1029-1035, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28778706

RESUMO

The pathophysiology of SSc-mediated organ damage is complex and not well understood. Hallmarks of the disease include skin thickening, vasculopathy and gastrointestinal dysmotility. Diverse anti-nuclear antibodies can be used as biomarkers for classification and prognosis, but their role in producing tissue pathology/organ dysfunction is not established. In contrast, antibodies against cell surface receptors for platelet derived growth factor, angiotensin II, endothelin A, ICAM-1, and type 3 muscarinic acetyl choline receptors may play a major role in skin thickening, vasoconstriction/pulmonary and renal hypertension, ischemia and gastrointestinal dysmotility, respectively. In addition, antibodies to an inhibitory B-lymphocyte surface molecule, CD 22, may allow increased production of other autoantibodies. Each of these types of antibodies have been reported in some SSc patients, and laboratory studies suggest signaling pathways and mechanisms by which they may contribute to disease activity. However, we are far from a consensus on their importance. Additional epidemiologic, mechanistic and physiologic studies are needed. Confirmation of the roles of anti-receptor antibodies and identification of the signaling pathways by which they alter cellular functions would have major implications for treatment of SSc, both in terms of targeting autoantibodies and the cells that produce them, and in the use of small molecules which inhibit their pernicious effects.


Assuntos
Autoanticorpos/imunologia , Escleroderma Sistêmico/genética , Humanos , Escleroderma Sistêmico/fisiopatologia
9.
Muscle Nerve ; 51(3): 315-26, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25418426

RESUMO

Prolonged intravenous immunoglobulin (IVIG) therapy is used for the chronic autoimmune neuropathies chronic idiopathic demyelinating polyneuropathy and multifocal motor neuropathy, but the doses and treatment intervals are usually chosen empirically due to a paucity of data from dose-response studies. Recent studies of the electrophysiology and immunology of these diseases suggest that antibody-induced reversible dysfunction of nodes of Ranvier may play a role in conduction block and disability which responds to immunotherapy more rapidly than would be expected for demyelination or axonal damage per se. Clinical reports suggest that in some cases, the effects of each dose of IVIG may be transient, wearing-off before the next dose is due. These observations lead us to hypothesize that that therapeutic IgG acts by competing with pathologic autoantibodies and that individual patients may require different IgG levels for optimal therapeutic effects. Frequent IVIG dosing and weekly subcutaneous IgG have been tried as ways of continuously maintaining high serum IgG levels, resulting in stabilization of neuromuscular function in small case series. Frequent grip strength and disability measurements, performed by the patient at home and reported electronically, can be used to assess the extent and duration of responses to IgG doses. Individualization of IgG treatment regimens may optimize efficacy, minimize disability, and identify nonresponders.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Imunoglobulina G/administração & dosagem , Imunoglobulinas Intravenosas/administração & dosagem , Polineuropatias/tratamento farmacológico , Animais , Autoanticorpos/sangue , Autoanticorpos/imunologia , Doenças Autoimunes/sangue , Doenças Autoimunes/imunologia , Doença Crônica , Humanos , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Imunoglobulinas Intravenosas/sangue , Imunoglobulinas Intravenosas/imunologia , Polineuropatias/sangue , Polineuropatias/imunologia
10.
J Clin Immunol ; 34(7): 804-12, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24981039

RESUMO

PURPOSE: This retrospective study evaluated the effectiveness and tolerability in clinical practice of an L-proline-stabilized 10 % intravenous immunoglobulin (IVIG; Privigen®) in patients with primary (PID) or secondary immunodeficiency (SID). METHODS: Patients from 6 centers in Europe and the US were treated with individually determined regimens of Privigen® for ≥3 months. Serum immunoglobulin G (IgG) trough levels, annualized rates of infection, hospitalization and antibiotics use, and the incidence of adverse events (AEs) were analyzed. RESULTS: Of 72 patients, three infants with severe combined immunodeficiency (SCID) were analyzed separately. The remaining 69 patients (52.2 % male; median age 38 years [range: 0.1-90.0]) with PID (82.6 %) or SID (17.4 %) received a mean (±standard deviation) Privigen® dose of 532 ± 250 mg/kg/month resulting in trough serum IgG levels of 407-1,581 mg/dL (median: 954 mg/dL). Ten patients (14.5 %) experienced 11 serious bacterial infections over 22.0 ± 15.0 months of treatment (0.087 events/patient/year, upper one-sided 99 % confidence interval: 0.170), the most common being pneumonia (11.6 %). The rates for any infection and hospitalization were 1.082 events/patient/year and 3.63 days/patient/year, respectively. Two patients with severe disease accounted for 303 of 460 hospital days. Across all 72 patients, 13 (18.1 %) patients experienced AEs, including 10 (13.9 %) patients with AEs at least possibly related to Privigen®, including headache (8.3 %), fever, and chills (2.8 % each). No related serious AEs were reported. One infant with SCID died due to severe viral infection. CONCLUSIONS: Despite the heterogeneous population, effectiveness and tolerability of Privigen® in clinical practice closely matched those reported in clinical studies.


Assuntos
Antibacterianos/uso terapêutico , Imunoglobulinas Intravenosas/administração & dosagem , Síndromes de Imunodeficiência/terapia , Infecções/terapia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Europa (Continente) , Feminino , Cefaleia/etiologia , Hospitalização , Humanos , Imunoglobulinas Intravenosas/efeitos adversos , Imunoglobulinas Intravenosas/química , Síndromes de Imunodeficiência/complicações , Síndromes de Imunodeficiência/imunologia , Lactente , Infecções/etiologia , Infecções/imunologia , Masculino , Pessoa de Meia-Idade , Padrões de Prática Médica , Prolina/química , Estabilidade Proteica , Estudos Retrospectivos , Estados Unidos , Adulto Jovem
11.
Immunotherapy ; 6(1): 71-83, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24341886

RESUMO

Subcutaneous administration of IgG (SCIG) has become widely used in primary immune deficiency diseases but it has only recently been studied for maintenance therapy in autoimmune peripheral neuropathies, such as chronic idiopathic demyelinating polyneuropathy and multifocal motor neuropathy. Weekly self-administration of SCIG is safe and well-tolerated, and results in steady-state serum IgG levels, as contrasted with the peaks and troughs of monthly immune globulin (human) for intravenous use. Freedom from the need for venous access or medical personnel for infusions, flexibility in scheduling, convenience of home therapy, and improved clinical stability due to the steady-state IgG levels, lead many patients to prefer SCIG to immune globulin (human) for intravenous use. Long-term studies are needed to determine if the constant IgG levels and clinical stability translate into better long-term outcomes.


Assuntos
Doenças Autoimunes do Sistema Nervoso/terapia , Imunoglobulina G/uso terapêutico , Doenças Autoimunes do Sistema Nervoso/imunologia , Ensaios Clínicos como Assunto , Humanos , Imunoglobulinas Intravenosas/uso terapêutico , Infusões Subcutâneas , Autoadministração , Resultado do Tratamento
12.
J Peripher Nerv Syst ; 18(4): 275-96, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24200120

RESUMO

Intravenous immunoglobulin (IVIG) is widely used in autoimmune neuromuscular diseases whose pathogenesis is undefined. Many different effects of IVIG have been demonstrated in vitro, but few studies actually identify the mechanism(s) most important in vivo. Doses and treatment intervals are generally chosen empirically. Recent studies in Guillain-Barré syndrome and chronic inflammatory demyelinating polyneuropathy show that some effects of IVIG are readily reversible and highly dependent on the serum IgG level. This suggests that in some autoantibody-mediated neuromuscular diseases, IVIG directly competes with autoantibodies that reversibly interfere with nerve conduction. Mechanisms of action of IVIG which most likely involve direct competition with autoantibodies include: neutralization of autoantibodies by anti-idiotypes, inhibition of complement deposition, and increasing catabolism of pathologic antibodies by saturating FcRn. Indirect immunomodulatory effects are not as likely to involve competition and may not have the same reversibility and dose-dependency. Pharmacodynamic analyses should be informative regarding most relevant mechanism(s) of action of IVIG as well as the role of autoantibodies in the immunopathogenesis of each disease. Better understanding of the role of autoantibodies and of the target(s) of IVIG could lead to more efficient use of this therapy and better patient outcomes.


Assuntos
Autoanticorpos/metabolismo , Doenças Autoimunes/terapia , Imunoglobulinas Intravenosas/uso terapêutico , Fatores Imunológicos/uso terapêutico , Doenças Neuromusculares/terapia , Animais , Doenças Autoimunes/complicações , Humanos , Imunoglobulinas Intravenosas/farmacologia , Fatores Imunológicos/farmacologia , Doenças Neuromusculares/complicações , Ligação Proteica/efeitos dos fármacos
13.
Vaccine ; 31(51): 6107-12, 2013 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-24120547

RESUMO

BACKGROUND: Hypersensitivity disorders following vaccinations are a cause for concern. OBJECTIVE: To determine the type and rate by age, gender, and vaccine received for reported hypersensitivity reactions following monovalent 2009 pandemic influenza A (H1N1) vaccines. DESIGN: A systematic review of reports to the Vaccine Adverse Event Reporting System (VAERS) following monovalent 2009 pandemic influenza A (H1N1) vaccines. SETTING/PATIENTS: US Civilian reports following vaccine received from October 1, 2009 through May 31, 2010. MEASUREMENTS: Age, gender, vaccines received, diagnoses, clinical signs, and treatment were reviewed by nurses and physicians with expertise in vaccine adverse events. A panel of experts, including seven allergists reviewed complex illnesses and those with conflicting evidence for classification of the event. RESULTS: Of 1984 reports, 1286 were consistent with immediate hypersensitivity disorders and 698 were attributed to anxiety reactions, syncope, or other illnesses. The female-to-male ratio was ≥4:1 for persons 20-to-59 years of age, but approximately equal for children under 10. One hundred eleven reports met Brighton Collaboration criteria for anaphylaxis; only one-half received epinephrine for initial therapy. The overall rate of reported hypersensitivity reactions was 10.7 per million vaccine doses distributed, with a 2-fold higher rate for live vaccine. LIMITATIONS: Underreporting, especially of mild events, would result in an underestimate of the true rate of immediate hypersensitivity reactions. Selective reporting of events in adult females could have resulted in higher rates than reported for males. CONCLUSIONS: Adult females may be at higher risk of hypersensitivity reactions after influenza vaccination than men. Although the risk of hypersensitivity reactions following 2009 pandemic influenza A (H1N1) vaccines was low, all clinics administering vaccines should be familiar with treatment guidelines for these adverse events, including the use of intramuscular epinephrine early in the course of serious hypersensitivity reactions.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Hipersensibilidade Imediata/induzido quimicamente , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/efeitos adversos , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/patologia , Feminino , Humanos , Lactente , Recém-Nascido , Vírus da Influenza A Subtipo H1N1/imunologia , Influenza Humana/prevenção & controle , Masculino , Pessoa de Meia-Idade , Fatores Sexuais , Estados Unidos/epidemiologia , Adulto Jovem
14.
Postgrad Med ; 125(2): 65-72, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23816772

RESUMO

AIMS: We reviewed the intravenous immunoglobulin G (IVIG) dispensing records of a specialty pharmacy to characterize the IVIG treatment regimens used for chronic inflammatory demyelinating polyneuropathy (CIDP) and myasthenia gravis (MG) in community practice. METHODS: Anonymized records were selected based on International Classification of Diseases, Ninth Revision (ICD-9) codes and IVIG treatment for > 1 month. Each patient's immunoglobulin G (IgG) dose per infusion (mg/kg/dose) was multiplied by the number of doses per month (30.5 days divided by the dosing interval in days) to yield the total monthly dose (mg/kg/month). Data were analyzed and summarized using descriptive statistics. RESULTS: Forty-six patients (median age, 56.5 years; range, 8-86 years) fulfilled the inclusion criteria. Thirty-one patients with CIDP received IgG at 7- to 92-day intervals (mean [standard deviation (SD)], 28 [16] days). The mean (SD) IgG dose was 75 (60) g/dose, equivalent to 866 (623) mg/kg/dose and 1145 (778) mg/kg/month. Six patients with stable MG received IVIG or subcutaneous IgG at 3.5- to 61-day intervals (28 [20] days) at a mean (SD) IgG dose of 39 (15) g/dose, equivalent to 405 (108) mg/kg/dose and 783 (680) mg/kg/month. Nine patients with MG with acute exacerbations received IgG at 7- to 42-day intervals (22 [12] days) at a mean (SD) dose of 40 (21) g/dose, equivalent to 403 (172) mg/kg/dose and 641 (288) mg/kg/month. One patient with CIDP and 4 patients with MG were treated with weekly subcutaneous IgG injections. CONCLUSION: Although patients with CIDP and MG are treated with mean total monthly IgG doses similar to those approved by the US Food and Drug Administration, the individual doses and intervals vary considerably, suggesting that physicians may be adjusting IgG dosing according to each patient's clinical condition and treatment response. Further study is necessary to determine the criteria used to adjust IgG treatment regimens and whether these adjustments optimize clinical outcomes while limiting overall costs.


Assuntos
Imunoglobulinas Intravenosas/administração & dosagem , Fatores Imunológicos/administração & dosagem , Miastenia Gravis/tratamento farmacológico , Polirradiculoneuropatia Desmielinizante Inflamatória Crônica/tratamento farmacológico , Padrões de Prática Médica/estatística & dados numéricos , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Estudos Transversais , Esquema de Medicação , Feminino , Humanos , Imunoglobulinas Intravenosas/uso terapêutico , Fatores Imunológicos/uso terapêutico , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Adulto Jovem
16.
J Clin Immunol ; 33(5): 984-90, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23456255

RESUMO

PURPOSE: US licensing studies of subcutaneous IgG (SCIG) calculate dose adjustments necessary to achieve area under the curve (AUC) of serum IgG vs. time on SCIG that is non-inferior to that on intravenous IgG (IVIG), within the FDA-set limit of ±20%. The results are interpreted as showing that different SCIGs differ in bioavailability. We used three approaches to determine if the bioavailabilities were actually different. METHODS: Dose adjustments and AUCs from published licensing studies were used to calculate bioavailabilities using the formula: Bioavailability (% of IVIG) = AUC(SCIG) ÷ AUC(IVIG) x 1/Dose Adjustment. We also compared the increment in serum IgG concentration achieved with varying doses of SCIG in recent meta-analyses with the increment with different doses of IVIG, and determined the serum IgG concentrations when patients switched SCIG products at the same dose. RESULTS: The actual bioavailabilities were: Gamunex® 65.0%, Hizentra® 65.5%, Gammagard® 67.2%, Vivaglobin® 69.0%. Regression analyses of serum IgG vs. dose showed that the mean increase in serum IgG resulting from a 100 mg/kg/month increment in SCIG dosing was 69.4% of the increase with the same increment in IVIG dosing (84 mg/dL vs. 121 mg/dL). Patients switching SCIG preparations at the same dose had no change in serum IgG levels, confirming that bioavailabilities of the SCIG preparations did not differ. CONCLUSIONS: Decreased bioavailability appears to be a basic property of SCIG and not a result of any manufacturing process or concentration. Because serum IgG levels do not vary with different SCIG products at the same dose, adjustments are not necessary when switching products.


Assuntos
Imunoglobulina G/administração & dosagem , Imunoglobulina G/metabolismo , Adolescente , Adulto , Idoso , Área Sob a Curva , Disponibilidade Biológica , Criança , Humanos , Injeções Subcutâneas , Pessoa de Meia-Idade , Adulto Jovem
17.
Pediatr Infect Dis J ; 32(2): 163-8, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23334340

RESUMO

BACKGROUND: Monovalent 2009 H1N1 influenza vaccines were licensed and administered in the United States during the H1N1 influenza pandemic between 2009 and 2013. METHODS: Vaccine Adverse Event Reporting System received reports of adverse events following immunization (AEFI) after H1N1 vaccination. Selected reports were referred to the Centers for Disease Control and Prevention's Clinical Immunization Safety Assessment network for additional review. We assessed causality using modified World Health Organization criteria. RESULTS: There were 3,928 reports of AEFI in children younger than age 18 years after 2009 H1N1 vaccination received by January 31, 2010. Of these, 214 (5.4%) were classified as serious nonfatal and 109 were referred to Clinical Immunization Safety Assessment for further evaluation. Ninety-nine (91%) had sufficient initial information to begin investigation and are described here. The mean age was 8 years (range, 6 months-17 years) and 38% were female. Median number of days between vaccination and symptom onset was 2 (range, -11 days to +41 days). Receipt of inactivated, live attenuated, or unknown type of 2009 H1N1 vaccines was reported by 68, 26 and 5 cases, respectively. Serious AEFI were categorized as neurologic events in 47 cases, as hypersensitivity in 15 cases and as respiratory events in 10 cases. At the time of evaluation, recovery was described as complete (61), partial (16), no improvement (1), or unknown (21). Causality assessment yielded the following likelihood of association with 2009 H1N1 vaccination: 8 definitely; 8 probably; 21 possibly; 43 unlikely; 17 unrelated; and 2 unclassifiable. CONCLUSIONS: Most AEFI in children evaluated were not causally related to vaccine and resolved without sequelae. Detailed clinical assessment of individual serious AEFI can provide reassurance of vaccine safety.


Assuntos
Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/efeitos adversos , Adolescente , Sistemas de Notificação de Reações Adversas a Medicamentos , Centers for Disease Control and Prevention, U.S. , Criança , Pré-Escolar , Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos/epidemiologia , Feminino , Humanos , Lactente , Vacinas contra Influenza/administração & dosagem , Masculino , Vacinação em Massa/estatística & dados numéricos , Estados Unidos/epidemiologia
18.
J Allergy Clin Immunol Pract ; 1(6): 558-66, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24565701

RESUMO

IgG is widely used for patients with immune deficiencies and in a broad range of autoimmune and inflammatory disorders. Up to 40% of intravenous infusions of IgG may be associated with adverse effects (AEs), which are mostly uncomfortable or unpleasant but often are not serious. The most common infusion-related AE is headache. More serious reactions, including true anaphylaxis and anaphylactoid reactions, occur less frequently. Most reactions are related to the rate of infusion and can be prevented or treated just by slowing the infusion rate. Medications such as nonsteroidal anti-inflammatory drugs, antihistamines, or corticosteroids also may be helpful in preventing or treating these common AEs. IgA deficiency with the potential of IgG or IgE antibodies against IgA increases the risk of some AEs but should not be viewed as a contraindication if IgG therapy is needed. Potentially serious AEs include renal dysfunction and/or failure, thromboembolic events, and acute hemolysis. These events usually are multifactorial, related to combinations of constituents in the IgG product as well as risk factors for the recipient. Awareness of these factors should allow minimization of the risks and consequences of these AEs. Subcutaneous IgG is absorbed more slowly into the circulation and has a lower incidence of AEs, but awareness and diligence are necessary whenever IgG is administered.


Assuntos
Imunoglobulina G/efeitos adversos , Imunoglobulinas Intravenosas/efeitos adversos , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/prevenção & controle , Anafilaxia/etiologia , Anafilaxia/prevenção & controle , Anemia Hemolítica/etiologia , Anemia Hemolítica/prevenção & controle , Transtornos da Cefaleia/etiologia , Transtornos da Cefaleia/prevenção & controle , Humanos , Hiponatremia/etiologia , Hiponatremia/prevenção & controle , Imunoglobulina A/efeitos adversos , Meningite Asséptica/etiologia , Meningite Asséptica/prevenção & controle , Neutropenia/etiologia , Neutropenia/prevenção & controle , Fatores de Risco , Dermatopatias/etiologia , Dermatopatias/prevenção & controle , Tromboembolia/etiologia , Tromboembolia/prevenção & controle
20.
Expert Rev Clin Immunol ; 8(2): 169-78, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22288455

RESUMO

Privigen(®) (immune globulin intravenous [human], 10% liquid) and Hizentra(®) (immune globulin subcutaneous [human], 20% liquid) are stabilized by proline. The clinical implications of administering proline-containing immunoglobulin products to patients with defects of proline metabolism have not been addressed; Privigen and Hizentra are contraindicated in these patients. Some patients with chromosome 22q11.2 deletion syndrome have elevated proline levels; however, only 3-4% of patients also have an immunodeficiency that requires IgG therapy. This review summarizes the evidence related to the safety and pharmacokinetics of proline assessed in Privigen and Hizentra preclinical and clinical studies, and subsequent implications for patients with defects in proline metabolism. Clinical data indicate that proline does not accumulate after Privigen or Hizentra treatment and is not associated with adverse events. There is no evidence to suggest that patients with defects of proline metabolism would be affected by transient elevations in plasma proline following Privigen and/or Hizentra treatment.


Assuntos
Efeitos Colaterais e Reações Adversas Relacionados a Medicamentos , Excipientes , Imunoglobulina G , Imunoglobulinas Intravenosas , Prolina , Animais , Deleção Cromossômica , Cromossomos Humanos Par 22/genética , Estabilidade de Medicamentos , Excipientes/efeitos adversos , Excipientes/farmacocinética , Excipientes/uso terapêutico , Doenças Genéticas Inatas/tratamento farmacológico , Doenças Genéticas Inatas/genética , Humanos , Imunoglobulina G/efeitos adversos , Imunoglobulina G/uso terapêutico , Imunoglobulinas Intravenosas/efeitos adversos , Imunoglobulinas Intravenosas/farmacocinética , Imunoglobulinas Intravenosas/uso terapêutico , Prolina/efeitos adversos , Prolina/farmacocinética , Prolina/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...