Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
1.
Blood Purif ; 53(5): 379-385, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38219716

RESUMO

INTRODUCTION: Novel hemoperfusion systems are emerging for the treatment of sepsis. These devices can directly remove pathogens, pathogen-associated molecular patterns, cytokines, and other inflammatory markers from circulation. However, significant safety concerns such as potential antibiotic clearance need to be addressed prior to these devices being used in large clinical studies. METHODS: Prospective, observational study of 34 participants undergoing treatment with the Seraph 100® Microbind Affinity Blood Filter (Seraph 100) device at 6 participating sites in the USA. Patients were included for analysis if they had a record of receiving an antibiotic concurrent with Seraph 100 treatment. Patients were excluded if there was missing information for blood flow rate. Blood samples were drawn pre- and post-filter at 1 h and 4 h after treatment initiation. These average pre- and post-filter time-concentration observations were then used to estimate antibiotic clearance in L/h (CLSeraph) due to the Seraph 100 device. RESULTS: Of the 34 participants in the study, 17 met inclusion and exclusion criteria for the antibiotic analysis. Data were obtained for 7 antibiotics (azithromycin, cefazolin, cefepime, ceftriaxone, linezolid, piperacillin, and vancomycin) and one beta-lactamase inhibitor. Mean CLSeraph for the antibiotics investigated ranged from -0.57 to 0.47 L/h. No antibiotic had a CLSeraph statistically significant from 0. DISCUSSION/CONCLUSION: The Seraph 100 did not significantly clear any measured antibiotic in clinical samples. These data give further evidence to suggest that these therapies may be safely administered to critically ill patients and will not impact concentrations of administered antibiotics.


Assuntos
Antibacterianos , Piperacilina , Humanos , Antibacterianos/uso terapêutico , Estudos Prospectivos , Piperacilina/uso terapêutico , Linezolida , Cefepima
2.
Haemophilia ; 27 Suppl 3: 28-36, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32608138

RESUMO

The development of inhibitory antibodies to therapeutic factor VIII (FVIII) in haemophilia A (HA) patients is the major complication in treatment/prevention of haemorrhages. The reasons some HA patients develop inhibitors while others do not remain unclear. This review briefly summarizes our understanding of anti-FVIII immune responses, the roles of T cells, both effector and regulatory, and generally discusses the interplay between FVIII and the immune system, both in factor replacement therapy and gene therapy, with some comparisons to factor IX and haemophilia B therapies. Notably, we propose that the prevailing observed active tolerance to FVIII in both HA and non-HA individuals rests to greater or lesser extents on peripherally induced immune tolerance. We also propose that the immune systems of inhibitor-negative HA patients do not merely ignore therapeutic FVIII, but rather have immunologically assessed and actively tolerized the patients to exogenous FVIII. Induction of such peripheral immune tolerance may further be triggered in HA patients who failed to tolerize upon initial FVIII exposure by 'appropriate' stimulation of their immune system, eg by immune tolerance induction therapy via intensive FVIII therapy, by oral administration of FVIII, by cellular therapies or by gene therapy directed to immuno-tolerogenic sites such as the liver.


Assuntos
Hemofilia A , Hemostáticos , Fator VIII/genética , Terapia Genética , Hemofilia A/tratamento farmacológico , Humanos , Tolerância Imunológica
3.
BMC Infect Dis ; 21(1): 544, 2021 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-34107889

RESUMO

BACKGROUND: SARS-CoV-2 is a recently emerged pandemic coronavirus (CoV) capable of causing severe respiratory illness. However, a significant number of infected people present as asymptomatic or pauci-symptomatic. In this prospective assessment of at-risk healthcare workers (HCWs) we seek to determine whether pre-existing antibody or T cell responses to previous seasonal human coronavirus (HCoV) infections affect immunological or clinical responses to SARS-CoV-2 infection or vaccination. METHODS: A cohort of 300 healthcare workers, confirmed negative for SARS-CoV-2 exposure upon study entry, will be followed for up to 1 year with monthly serology analysis of IgM and IgG antibodies against the spike proteins of SARS-CoV-2 and the four major seasonal human coronavirus - HCoV-OC43, HCoV-HKU1, HCoV-229E, and HCoV-NL63. Participants will complete monthly questionnaires that ask about Coronavirus Disease 2019 (COVID-19) exposure risks, and a standardized, validated symptom questionnaire (scoring viral respiratory disease symptoms, intensity and severity) at least twice monthly and any day when any symptoms manifest. SARS-CoV-2 PCR testing will be performed any time participants develop symptoms consistent with COVID-19. For those individuals that seroconvert and/or test positive by SARS-CoV-2 PCR, or receive the SARS-CoV-2 vaccine, additional studies of T cell activation and cytokine production in response to SARS-CoV-2 peptide pools and analysis of Natural Killer cell numbers and function will be conducted on that participant's cryopreserved baseline peripheral blood mononuclear cells (PBMCs). Following the first year of this study we will further analyze those participants having tested positive for COVID-19, and/or having received an authorized/licensed SARS-CoV-2 vaccine, quarterly (year 2) and semi-annually (years 3 and 4) to investigate immune response longevity. DISCUSSION: This study will determine the frequency of asymptomatic and pauci-symptomatic SARS-CoV-2 infection in a cohort of at-risk healthcare workers. Baseline and longitudinal assays will determine the frequency and magnitude of anti-spike glycoprotein antibodies to the seasonal HCoV-OC43, HCoV-HKU1, HCoV-229E, and HCoV-NL63, and may inform whether pre-existing antibodies to these human coronaviruses are associated with altered COVID-19 disease course. Finally, this study will evaluate whether pre-existing immune responses to seasonal HCoVs affect the magnitude and duration of antibody and T cell responses to SARS-CoV-2 vaccination, adjusting for demographic covariates.


Assuntos
COVID-19/imunologia , Pessoal de Saúde/estatística & dados numéricos , SARS-CoV-2/imunologia , Soroconversão , Vacinação/estatística & dados numéricos , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Infecções Assintomáticas , Vacinas contra COVID-19/imunologia , Coronavirus/imunologia , Reações Cruzadas , Humanos , Estudos Prospectivos , Glicoproteína da Espícula de Coronavírus/imunologia , Linfócitos T/imunologia
4.
Blood ; 142(3): 217-219, 2023 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-37471109
5.
Blood ; 128(16): 2007-2016, 2016 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-27587878

RESUMO

A normal hemostatic response to vascular injury requires both factor VIII (FVIII) and von Willebrand factor (VWF). In plasma, VWF and FVIII normally circulate as a noncovalent complex, and each has a critical function in the maintenance of hemostasis. Furthermore, the interaction between VWF and FVIII plays a crucial role in FVIII function, immunogenicity, and clearance, with VWF essentially serving as a chaperone for FVIII. Several novel recombinant FVIII (rFVIII) therapies for hemophilia A have been in clinical development, which aim to increase the half-life of FVIII (∼12 hours) and reduce dosing frequency by utilizing bioengineering techniques including PEGylation, Fc fusion, and single-chain design. However, these approaches have achieved only moderate increases in half-life of 1.5- to 2-fold compared with marketed FVIII products. Clearance of PEGylated rFVIII, rFVIIIFc, and rVIII-SingleChain is still regulated to a large extent by interaction with VWF. Therefore, the half-life of VWF (∼15 hours) appears to be the limiting factor that has confounded attempts to extend the half-life of rFVIII. A greater understanding of the interaction between FVIII and VWF is required to drive novel bioengineering strategies for products that either prolong the survival of VWF or limit VWF-mediated clearance of FVIII.


Assuntos
Fator VIII , Proteínas Recombinantes de Fusão , Fator de von Willebrand/metabolismo , Fator VIII/farmacocinética , Fator VIII/uso terapêutico , Meia-Vida , Hemofilia A/sangue , Hemofilia A/tratamento farmacológico , Humanos , Proteínas Recombinantes de Fusão/farmacocinética , Proteínas Recombinantes de Fusão/uso terapêutico
6.
Blood ; 128(16): 2043-2054, 2016 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-27471234

RESUMO

Factor VIII (FVIII)-neutralizing antibodies ("inhibitors") are a serious problem in hemophilia A (HA). The aim of this study was to characterize HLA-restricted T-cell responses from a severe HA subject with a persistent inhibitor and from 2 previously studied mild HA inhibitor subjects. Major histocompatibility complex II tetramers corresponding to both of the severe HA subject's HLA-DRA-DRB1 alleles were loaded with peptides spanning FVIII-A2, C1, and C2 domains. Interestingly, only 1 epitope was identified, in peptide FVIII2194-2213, and it was identical to the HLA-DRA*01-DRB1*01:01-restricted epitope recognized by the mild HA subjects. Multiple T-cell clones and polyclonal lines having different avidities for the peptide-loaded tetramer were isolated from all subjects. Only high- and medium-avidity T cells proliferated and secreted cytokines when stimulated with FVIII2194-2213 T-cell receptor ß (TCRB) gene sequencing of 15 T-cell clones from the severe HA subject revealed that all high-avidity clones expressed the same TCRB gene. High-throughput immunosequencing of high-, medium-, and low-avidity cells sorted from a severe HA polyclonal line revealed that 94% of the high-avidity cells expressed the same TCRB gene as the high-avidity clones. TCRB sequencing of clones and lines from the mild HA subjects also identified a limited TCRB gene repertoire. These results suggest a limited number of epitopes in FVIII drive inhibitor responses and that the T-cell repertoires of FVIII-responsive T cells can be quite narrow. The limited diversity of both epitopes and TCRB gene usage suggests that targeting of specific epitopes and/or T-cell clones may be a promising approach to achieve tolerance to FVIII.


Assuntos
Epitopos de Linfócito T , Fator VIII , Hemofilia A , Receptores de Antígenos de Linfócitos T alfa-beta , Linfócitos T/imunologia , Adolescente , Autoanticorpos/genética , Autoanticorpos/imunologia , Inibidores dos Fatores de Coagulação Sanguínea/genética , Inibidores dos Fatores de Coagulação Sanguínea/imunologia , Criança , Pré-Escolar , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Fator VIII/genética , Fator VIII/imunologia , Antígenos HLA/genética , Antígenos HLA/imunologia , Hemofilia A/genética , Hemofilia A/imunologia , Humanos , Masculino , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia
7.
Transfusion ; 58(5): 1171-1181, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29464723

RESUMO

BACKGROUND: Kell is a glycoprotein expressed on red blood cells (RBCs). Its K and k variants contain either Met (K antigen) or Thr (k antigen) at Position 193, respectively. Development of anti-K after K-mismatched antigen exposure via blood transfusions or pregnancy can destroy RBCs, leading to hemolytic transfusion reactions and hemolytic disease of the fetus and newborn. The immunogenicity of overlapping 15-mer Kell peptides with M193 or T193 at every possible position was investigated previously. Interestingly, Peptide W179 to M193, with the polymorphic M193T residue at the peptide's C-terminus, was the most effective at stimulating CD4 T cells from a series of K-immunized women. STUDY DESIGN AND METHODS: This study investigates the basis for HLA restriction of anti-K immune responses. Major histocompatibility complex Class II (MHCII)-binding prediction algorithms and quantitative peptide-MHCII-binding assays were employed to determine the binding registers; anchor residues; and affinities of wild-type, truncated, and sequence-modified K and k peptides. Predictions were generated using Immune Epitope Database and ProPred algorithms. Competitive peptide-MHCII-binding assays utilized 12 recombinant HLA-DR proteins, K and k peptides, and high-affinity MHCII-restricted reference peptides. RESULTS: The peptide-MHCII-binding assays identified a unique K peptide-binding register (W179-S187) restricted to HLA-DRB1*11:01, in addition to partially overlapping binding registers that included the K/k M193T polymorphic site and that bound promiscuously to multiple HLA-DR proteins. CONCLUSION: Three partially overlapping MHCII-binding motifs for HLA-DRB1*11:01 result in high-avidity K-peptide binding, which may contribute to HLA-DR11-restricted immunogenicity associated with the K allele.


Assuntos
Antígenos de Bactérias/imunologia , Antígenos de Superfície/imunologia , Subtipos Sorológicos de HLA-DR/imunologia , Antígenos de Bactérias/metabolismo , Antígenos de Superfície/metabolismo , Sítios de Ligação , Antígenos de Grupos Sanguíneos/imunologia , Cadeias HLA-DRB1 , Antígenos de Histocompatibilidade Classe II , Humanos , Ligação Proteica
8.
Blood ; 125(7): 1107-15, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25498909

RESUMO

Expansion of human regulatory T cells (Tregs) for clinical applications offers great promise for the treatment of undesirable immune responses in autoimmunity, transplantation, allergy, and antidrug antibody responses, including inhibitor responses in hemophilia A patients. However, polyclonal Tregs are nonspecific and therefore could potentially cause global immunosuppression. To avoid this undesirable outcome, the generation of antigen-specific Tregs would be advantageous. Herein, we report the production and properties of engineered antigen-specific Tregs, created by transduction of a recombinant T-cell receptor obtained from a hemophilia A subject's T-cell clone, into expanded human FoxP3(+) Tregs. Such engineered factor VIII (FVIII)-specific Tregs efficiently suppressed the proliferation and cytokine production of FVIII-specific T-effector cells. Moreover, studies with an HLA-transgenic, FVIII-deficient mouse model demonstrated that antibody production from FVIII-primed spleen cells in vitro were profoundly inhibited in the presence of these FVIII-specific Tregs, suggesting potential utility to treat anti-FVIII inhibitory antibody formation in hemophilia A patients.


Assuntos
Linfócitos B Reguladores/imunologia , Engenharia Celular , Fator VIII/imunologia , Tolerância Imunológica , Especificidade do Receptor de Antígeno de Linfócitos T , Linfócitos T Reguladores/imunologia , Adulto , Idoso , Animais , Linfócitos B Reguladores/metabolismo , Engenharia Celular/métodos , Células Cultivadas , Fator VIII/genética , Fator VIII/metabolismo , Engenharia Genética , Terapia Genética , Hemofilia A/genética , Hemofilia A/imunologia , Humanos , Tolerância Imunológica/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Especificidade do Receptor de Antígeno de Linfócitos T/genética , Linfócitos T Reguladores/metabolismo , Adulto Jovem
9.
Blood ; 126(7): 895-904, 2015 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-25617427

RESUMO

African American hemophilia A (HA) patients experience a higher incidence of neutralizing anti-factor VIII (FVIII) antibodies ("inhibitors") vis-à-vis white patients. Nonsynonymous single-nucleotide polymorphisms (ns-SNPs) in the F8 gene encoding FVIII-H484, FVIII-E1241, and FVIII-V2238 are more prevalent in African Americans. This study tested the hypothesis that immune responses to these sites provoke inhibitors. Blood samples were obtained from 174 African American and 198 white HA subjects and their F8 gene sequences determined. Major histocompatibility complex class II binding and T-cell recognition of polymorphic sequences were evaluated using quantitative binding assays and HLA-DRB1 tetramers. Peptides corresponding to 4 common ns-SNPs showed limited binding to 11 HLA-DRB1 proteins. CD4 T cells from 22 subjects treated with FVIII products having sequences at residues FVIII-484, 1241, and 2238 differing from those of putative proteins encoded by their F8 genes did not show high-avidity tetramer binding, whereas positive-control staining of tetanus-specific CD4 T cells was routinely successful. African Americans with an intron-22 inversion mutation showed a 2-3 times-higher inhibitor incidence than whites with the same mutation (odds ratio = 2.3 [1.1-5.0, P = .04]), but this did not correlate with any of the ns-SNPs. We conclude that immune responses to "sequence-mismatched" FVIII products are unlikely to contribute appreciably to the inhibitor incidence in African Americans.


Assuntos
Anticorpos Neutralizantes/sangue , Negro ou Afro-Americano/genética , Fator VIII/antagonistas & inibidores , Fator VIII/genética , Hemofilia A/genética , Hemofilia A/imunologia , Adolescente , Adulto , Sequência de Aminoácidos , Autoanticorpos/sangue , Linfócitos T CD4-Positivos/imunologia , Criança , Pré-Escolar , Mapeamento de Epitopos , Fator VIII/imunologia , Variação Genética , Cadeias HLA-DRB1/metabolismo , Haplótipos/genética , Haplótipos/imunologia , Hemofilia A/sangue , Humanos , Masculino , Dados de Sequência Molecular , Proteínas Mutantes/antagonistas & inibidores , Proteínas Mutantes/genética , Proteínas Mutantes/imunologia , Fragmentos de Peptídeos/sangue , Fragmentos de Peptídeos/genética , Polimorfismo de Nucleotídeo Único , Ligação Proteica , Fatores de Risco , População Branca/genética , Adulto Jovem
10.
Cell Immunol ; 301: 12-7, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26566286

RESUMO

The development of neutralizing antibodies against blood coagulation factor VIII (FVIII), referred to clinically as "inhibitors", is the most challenging and deleterious adverse event to occur following intravenous infusions of FVIII to treat hemophilia A. Inhibitors occlude FVIII surfaces that must bind to activated phospholipid membranes, the serine proteinase factor IXa, and other components of the 'intrinsic tenase complex' in order to carry out its important role in accelerating blood coagulation. Inhibitors develop in up to one of every three patients, yet remarkably, a substantial majority of severe hemophilia A patients, who circulate no detectable FVIII antigen or activity, acquire immune tolerance to FVIII during initial infusions or else after intensive FVIII therapy to overcome their inhibitor. The design of less immunogenic FVIII proteins through identification and modification ("de-immunization") of immunodominant T-cell epitopes is an important goal. For patients who develop persistent inhibitors, modification of B-cell epitopes through substitution of surface-exposed amino acid side chains and/or attachment of bulky moieties to interfere with FVIII attachment to antibodies and memory B cells is a promising approach. Both experimental and computational methods are being employed to achieve these goals. Future therapies for hemophilia A, as well as other monogenic deficiency diseases, are likely to involve administration of less immunogenic proteins in conjunction with other novel immunotherapies to promote a regulatory cellular environment promoting durable immune tolerance.


Assuntos
Fator VIII/imunologia , Hemofilia A/imunologia , Anticorpos Neutralizantes/imunologia , Epitopos de Linfócito B/imunologia , Epitopos de Linfócito T/imunologia , Humanos , Proteínas Recombinantes/síntese química , Proteínas Recombinantes/imunologia
11.
Cell Immunol ; 299: 30-41, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26653006

RESUMO

The role of the T cell receptor (TCR) in antigen recognition and activation of T lymphocytes is well established. However, how the TCR affects T-helper differentiation/skewing is less well understood, particularly for human CD4(+) (CD4) T cell subsets. Here we investigate the role of TCR specific antigen avidity in differentiation and maintenance of human Th1, Th2 and Th17 subsets. Two human TCRs, both specific for the same peptide antigen but with different avidities, were cloned and expressed in human CD4 T cells. These TCR engineered cells were then stimulated with specific antigen in unskewed and T-helper skewed conditions. We show that TCR avidity can control the percentage of IL-4 and IFN-γ co-expression in unskewed TCR engineered cells, that effector function can be maintained in a TCR avidity-dependent manner in skewed TCR engineered cells, and that increased TCR avidity can accelerate Th1 skewing of TCR engineered cells.


Assuntos
Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular , Receptores de Antígenos de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/imunologia , Linfócitos T Auxiliares-Indutores/citologia , Clonagem Molecular , Humanos , Engenharia de Proteínas
12.
Blood ; 123(17): 2732-9, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24591205

RESUMO

Neutralizing anti-factor VIII (FVIII) antibodies that develop in patients with hemophilia A and in murine hemophilia A models, clinically termed "inhibitors," bind to several distinct surfaces on the FVIII-C2 domain. To map these epitopes at high resolution, 60 recombinant FVIII-C2 proteins were generated, each having a single surface-exposed residue mutated to alanine or a conservative substitution. The binding kinetics of these muteins to 11 monoclonal, inhibitory anti-FVIII-C2 antibodies were evaluated by surface plasmon resonance and the results compared with those obtained for wild-type FVIII-C2. Clusters of residues with significantly altered binding kinetics identified "functional" B-cell epitopes, defined as those residues contributing appreciable antigen-antibody avidity. These antibodies were previously shown to neutralize FVIII activity by interfering with proteolytic activation of FVIII by thrombin or factor Xa, or with its binding to phospholipid surfaces, von Willebrand factor, or other components of the intrinsic tenase complex. Fine mapping of epitopes by surface plasmon resonance also indicated surfaces through which FVIII interacts with proteins and phospholipids as it participates in coagulation. Mutations that significantly altered the dissociation times/half-lives identified functionally important interactions within antigen-antibody interfaces and suggested specific sequence modifications to generate novel, less antigenic FVIII proteins with possible therapeutic potential for treatment of inhibitor patients.


Assuntos
Mapeamento de Epitopos , Fator VIII/química , Mutação Puntual , Ressonância de Plasmônio de Superfície , Alanina/química , Aminoácidos/química , Animais , Anticorpos Monoclonais/química , Antígenos/química , Coagulação Sanguínea , Cristalografia por Raios X , Ensaio de Imunoadsorção Enzimática , Epitopos/química , Epitopos de Linfócito B/química , Humanos , Camundongos , Mutagênese , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Suínos
14.
Blood ; 121(17): 3302-3, 2013 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-23620569

RESUMO

In this issue of Blood, Sorvillo et al investigate possible molecular triggers leading to idiopathic, autoimmune thrombotic thrombocytopenic purpura (TTP) by identifying naturally processed A Disintegrin And Metalloprotease with ThromboSpondin type 1 motif 13 (ADAMTS13)-derived peptides presented on human dendritic cells.


Assuntos
Proteínas ADAM/imunologia , Células Dendríticas/imunologia , Cadeias HLA-DRB1/imunologia , Fragmentos de Peptídeos/imunologia , Púrpura Trombocitopênica Trombótica/etiologia , Proteína ADAMTS13 , Humanos , Serina Endopeptidases/imunologia
15.
Blood ; 121(22): 4449-56, 2013 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-23502223

RESUMO

A major problem in treating hemophilia A patients with therapeutic factor VIII (FVIII) is that 20% to 30% of these patients produce neutralizing anti-FVIII antibodies. These antibodies block (inhibit) the procoagulant function of FVIII and thus are termed "inhibitors." The currently accepted clinical method to attempt to eliminate inhibitors is immune tolerance induction (ITI) via a protocol requiring intensive FVIII treatment until inhibitor titers drop. Although often successful, ITI is extremely costly and is less likely to succeed in patients with high-titer inhibitors. During the past decade, significant progress has been made in clarifying mechanisms of allo- and autoimmune responses to FVIII and in suppression of these responses. Animal model studies are suggesting novel, less costly methods to induce tolerance to FVIII. Complementary studies of anti-FVIII T-cell responses using blood samples from human donors are identifying immunodominant T-cell epitopes in FVIII and possible targets for tolerogenic efforts. Mechanistic experiments using human T-cell clones and lines are providing a clinically relevant counterpoint to the animal model studies. This review highlights recent progress toward the related goals of lowering the incidence of anti-FVIII immune responses and promoting durable, functional immune tolerance to FVIII in patients with an existing inhibitor.


Assuntos
Fator VIII/imunologia , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Hemofilia A/imunologia , Tolerância Imunológica/imunologia , Coagulantes/imunologia , Coagulantes/uso terapêutico , Humanos , Imunossupressores/uso terapêutico
16.
Blood ; 130(23): 2450-2451, 2017 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-29217699
17.
Antibodies (Basel) ; 13(3)2024 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-39189236

RESUMO

PURIFY-OBS-1 is an observational study evaluating the safety and efficacy of Seraph 100® Microbind Affinity Blood Filter (Seraph 100) use for COVID-19 patients with respiratory failure admitted to the intensive care unit (ICU). The Seraph 100 is a hemoperfusion device containing heparin-coated beads that can bind to, and reduce levels of, some circulating pathogens and inflammatory molecules. This study evaluated whether treatment with the Seraph 100 affected circulating and mucosal antibody levels in critically ill COVID-19 subjects. SARS-CoV-2 anti-spike and anti-nucleocapsid IgG and IgA levels in serum were evaluated at enrollment and on days 1, 4, 7, and 28 after Seraph 100 application, while anti-spike and nucleocapsid IgG, IgA, and secretory IgA levels in tracheal aspirates were evaluated at enrollment and on days 1, 2, 3, 7, and 28. Serum samples were also collected from the pre- and post-filter lines at 1 and 4 h following Seraph 100 application to evaluate the direct impact of the filter on circulating antibody levels. Treatment with the Seraph 100 did not alter the levels of circulating or mucosal antibodies in critically ill COVID-19 subjects admitted to the ICU.

18.
J Thromb Haemost ; 2024 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-39233012

RESUMO

BACKGROUND: Almost half of severe hemophilia A (HA) cases are caused by an intron-22 inversion mutation (Int22Inv), which truncates the 26-exon F8 mRNA after exon 22. Another F8 transcript, F8B, is initiated from within F8-intron-22. F8B mRNA consists of a short exon spliced to exons 23-26 and is expressed in multiple human cell types. It has been hypothesized that Int22Inv patients have self-tolerance to partial FVIII proteins expressed from these two transcripts. FVIII is expressed in endothelial cells, primarily in liver and lung. Several studies have reported FVIII expression in other cell types, although this has been controversial. OBJECTIVES: To determine if partial FVIII proteins are expressed from intron 22-inverted and/or F8B mRNA and if FVIII is expressed in non-endothelial cells. METHODS: A panel of FVIII-specific antibodies was validated and employed to label FVIII in cells and tissues, and for immunoprecipitation followed by western blots and mass spectrometry-proteomics analysis. RESULTS: Immunofluorescent (IF) staining localized FVIII to endothelial cells in liver sections from non-HA but not HA-Int22Inv dogs. Neither FVIII nor FVIIIB was detected in human PBMCs, B-cell or T-cell lines, or in cell lines expanded from PBMCs, whereas FVIII antigen and activity were readily detected in primary non-hemophilic liver sinusoidal endothelial cells. CONCLUSIONS: If FVIII is expressed in non-endothelial cells, or if partial FVIII proteins are expressed in HA-Int22Inv, the concentrations are below the detection limits of these sensitive assays. Our results argue against promotion of immune tolerance through expression of partial FVIII proteins in Int-22Inv patients.

19.
Curr Opin Hematol ; 19(5): 399-405, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22814650

RESUMO

PURPOSE OF REVIEW: The review describes recent advances in our understanding of mechanisms leading to development of neutralizing antibodies following factor VIII (FVIII) replacement therapy for hemophilia A. Novel interventions with translational potential to lessen the incidence of these deleterious immune responses are discussed. RECENT FINDINGS: Genetic and environmental risk factors for inhibitor development, and cellular mechanisms leading to antibody production versus immune tolerance to FVIII, are increasingly coming into focus. Human and animal model studies are identifying T-cell and B-cell epitopes in FVIII and characterizing the presentation of naturally processed FVIII peptides on antigen-presenting cells (APCs). Novel methods to promote immune tolerance include decreasing FVIII uptake by APCs, modifying co-stimulatory pathways, inducing regulatory T-cell production, and presenting FVIII antigen to immature dendritic cells in a tolerance-promoting manner. A complementary approach to reduce inhibitor incidence is the design of less immunogenic FVIII proteins through epitope modification. SUMMARY: Studies of FVIII immunogenicity are revealing mechanisms of anti-FVIII immune responses, suggesting new approaches to reduce the incidence of inhibitors. Rational design of FVIII variants is producing less immunogenic proteins targeted to specific patient sub-populations. Future therapies will likely involve administration of less immunogenic FVIII proteins under conditions that promote immune tolerance.


Assuntos
Inibidores dos Fatores de Coagulação Sanguínea/imunologia , Fator VIII/imunologia , Hemofilia A/imunologia , Animais , Linfócitos B/imunologia , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Humanos , Tolerância Imunológica/imunologia , Fatores de Risco , Linfócitos T/imunologia
20.
J Thromb Haemost ; 21(4): 800-813, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36696179

RESUMO

BACKGROUND: Several studies have suggested Black and Hispanic hemophilia A (HA) patients in the United States suffer higher incidences of neutralizing anti-FVIII antibodies (inhibitors) than their White counterparts. The possible influence of nonsynonymous single-nucleotide polymorphisms (ns-SNPs) in the F8 gene sequence has been proposed as a possible race-associated contributing factor. Some earlier studies indicated that intron-22 inversion mutations carry a lower inhibitor risk than other mutations resulting in large F8 gene disruptions. OBJECTIVES: The objectives of the study were to test the following hypotheses: (1) The risk of developing an inhibitor differs among racial/ethnic groups in the United States, (2) specific non-HA-causing ns-SNPs in the F8 gene are correlated with inhibitor risk, and (3) inhibitor risk associated with intron-22 inversions mutations is similar to that associated with other large structural changes in the F8 gene. METHODS: Adjusted logistic regression analysis of the "My Life Our Future" database containing demographic, clinical, and F8 sequence data from >6000 mild, moderate, and severe HA participants. RESULTS: Black and Hispanic severe HA subjects had a higher inhibitor risk than non-Hispanic Whites (adjusted odds ratio = 1.65, 95% CI: 1.22-2.21 and adjusted odds ratio = 1.88, 95% CI: 1.43-2.48), confirming this racial/ethnic/medical disparity; however, F8 ns-SNPs were not associated with inhibitor development. There was no difference in inhibitor risk among severe HA subjects with an intron-22 inversion vs other large structural changes in the F8 gene. CONCLUSIONS: Nonpathogenic ns-SNPs in the F8 gene are not correlated with inhibitor risk. Inhibitor risk associated with intron-22 inversion mutations is similar to that of other large structural changes in F8 that preclude intact FVIII expression.


Assuntos
Hemofilia A , Humanos , Hemofilia A/diagnóstico , Hemofilia A/genética , Etnicidade , Fator VIII/genética , Mutação , Íntrons
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA