Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Blood Purif ; 53(5): 379-385, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38219716

RESUMEN

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.


Asunto(s)
Antibacterianos , Piperacilina , Humanos , Antibacterianos/uso terapéutico , Estudios Prospectivos , Piperacilina/uso terapéutico , Linezolid , Cefepima
2.
Haemophilia ; 27 Suppl 3: 28-36, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32608138

RESUMEN

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.


Asunto(s)
Hemofilia A , Hemostáticos , Factor VIII/genética , Terapia Genética , Hemofilia A/tratamiento farmacológico , Humanos , Tolerancia Inmunológica
3.
BMC Infect Dis ; 21(1): 544, 2021 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-34107889

RESUMEN

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.


Asunto(s)
COVID-19/inmunología , Personal de Salud/estadística & datos numéricos , SARS-CoV-2/inmunología , Seroconversión , Vacunación/estadística & datos numéricos , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Infecciones Asintomáticas , Vacunas contra la COVID-19/inmunología , Coronavirus/inmunología , Reacciones Cruzadas , Humanos , Estudios Prospectivos , Glicoproteína de la Espiga del Coronavirus/inmunología , Linfocitos T/inmunología
4.
Blood ; 142(3): 217-219, 2023 07 20.
Artículo en Inglés | MEDLINE | ID: mdl-37471109
5.
Blood ; 128(16): 2007-2016, 2016 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-27587878

RESUMEN

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.


Asunto(s)
Factor VIII , Proteínas Recombinantes de Fusión , Factor de von Willebrand/metabolismo , Factor VIII/farmacocinética , Factor VIII/uso terapéutico , Semivida , Hemofilia A/sangre , Hemofilia A/tratamiento farmacológico , Humanos , Proteínas Recombinantes de Fusión/farmacocinética , Proteínas Recombinantes de Fusión/uso terapéutico
6.
Blood ; 128(16): 2043-2054, 2016 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-27471234

RESUMEN

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.


Asunto(s)
Epítopos de Linfocito T , Factor VIII , Hemofilia A , Receptores de Antígenos de Linfocitos T alfa-beta , Linfocitos T/inmunología , Adolescente , Autoanticuerpos/genética , Autoanticuerpos/inmunología , Inhibidores de Factor de Coagulación Sanguínea/genética , Inhibidores de Factor de Coagulación Sanguínea/inmunología , Niño , Preescolar , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Factor VIII/genética , Factor VIII/inmunología , Antígenos HLA/genética , Antígenos HLA/inmunología , Hemofilia A/genética , Hemofilia A/inmunología , Humanos , Masculino , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología
7.
Transfusion ; 58(5): 1171-1181, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29464723

RESUMEN

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.


Asunto(s)
Antígenos Bacterianos/inmunología , Antígenos de Superficie/inmunología , Subtipos Serológicos HLA-DR/inmunología , Antígenos Bacterianos/metabolismo , Antígenos de Superficie/metabolismo , Sitios de Unión , Antígenos de Grupos Sanguíneos/inmunología , Cadenas HLA-DRB1 , Antígenos de Histocompatibilidad Clase II , Humanos , Unión Proteica
8.
Blood ; 125(7): 1107-15, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-25498909

RESUMEN

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.


Asunto(s)
Linfocitos B Reguladores/inmunología , Ingeniería Celular , Factor VIII/inmunología , Tolerancia Inmunológica , Especificidad del Receptor de Antígeno de Linfocitos T , Linfocitos T Reguladores/inmunología , Adulto , Anciano , Animales , Linfocitos B Reguladores/metabolismo , Ingeniería Celular/métodos , Células Cultivadas , Factor VIII/genética , Factor VIII/metabolismo , Ingeniería Genética , Terapia Genética , Hemofilia A/genética , Hemofilia A/inmunología , Humanos , Tolerancia Inmunológica/genética , Masculino , Ratones , Persona de Mediana Edad , Especificidad del Receptor de Antígeno de Linfocitos T/genética , Linfocitos T Reguladores/metabolismo , Adulto Joven
9.
Blood ; 126(7): 895-904, 2015 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-25617427

RESUMEN

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.


Asunto(s)
Anticuerpos Neutralizantes/sangre , Negro o Afroamericano/genética , Factor VIII/antagonistas & inhibidores , Factor VIII/genética , Hemofilia A/genética , Hemofilia A/inmunología , Adolescente , Adulto , Secuencia de Aminoácidos , Autoanticuerpos/sangre , Linfocitos T CD4-Positivos/inmunología , Niño , Preescolar , Mapeo Epitopo , Factor VIII/inmunología , Variación Genética , Cadenas HLA-DRB1/metabolismo , Haplotipos/genética , Haplotipos/inmunología , Hemofilia A/sangre , Humanos , Masculino , Datos de Secuencia Molecular , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/genética , Proteínas Mutantes/inmunología , Fragmentos de Péptidos/sangre , Fragmentos de Péptidos/genética , Polimorfismo de Nucleótido Simple , Unión Proteica , Factores de Riesgo , Población Blanca/genética , Adulto Joven
10.
Cell Immunol ; 301: 12-7, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26566286

RESUMEN

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.


Asunto(s)
Factor VIII/inmunología , Hemofilia A/inmunología , Anticuerpos Neutralizantes/inmunología , Epítopos de Linfocito B/inmunología , Epítopos de Linfocito T/inmunología , Humanos , Proteínas Recombinantes/síntesis química , Proteínas Recombinantes/inmunología
11.
Cell Immunol ; 299: 30-41, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26653006

RESUMEN

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.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Diferenciación Celular , Receptores de Antígenos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/inmunología , Linfocitos T Colaboradores-Inductores/citología , Clonación Molecular , Humanos , Ingeniería de Proteínas
12.
Blood ; 123(17): 2732-9, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24591205

RESUMEN

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.


Asunto(s)
Mapeo Epitopo , Factor VIII/química , Mutación Puntual , Resonancia por Plasmón de Superficie , Alanina/química , Aminoácidos/química , Animales , Anticuerpos Monoclonales/química , Antígenos/química , Coagulación Sanguínea , Cristalografía por Rayos X , Ensayo de Inmunoadsorción Enzimática , Epítopos/química , Epítopos de Linfocito B/química , Humanos , Ratones , Mutagénesis , Mutación , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Porcinos
14.
Blood ; 121(17): 3302-3, 2013 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-23620569

RESUMEN

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.


Asunto(s)
Proteínas ADAM/inmunología , Células Dendríticas/inmunología , Cadenas HLA-DRB1/inmunología , Fragmentos de Péptidos/inmunología , Púrpura Trombocitopénica Trombótica/etiología , Proteína ADAMTS13 , Humanos , Serina Endopeptidasas/inmunología
15.
Blood ; 121(22): 4449-56, 2013 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-23502223

RESUMEN

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.


Asunto(s)
Factor VIII/inmunología , Factor VIII/uso terapéutico , Hemofilia A/tratamiento farmacológico , Hemofilia A/inmunología , Tolerancia Inmunológica/inmunología , Coagulantes/inmunología , Coagulantes/uso terapéutico , Humanos , Inmunosupresores/uso terapéutico
16.
Blood ; 130(23): 2450-2451, 2017 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-29217699
17.
Curr Opin Hematol ; 19(5): 399-405, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22814650

RESUMEN

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.


Asunto(s)
Inhibidores de Factor de Coagulación Sanguínea/inmunología , Factor VIII/inmunología , Hemofilia A/inmunología , Animales , Linfocitos B/inmunología , Factor VIII/uso terapéutico , Hemofilia A/tratamiento farmacológico , Humanos , Tolerancia Inmunológica/inmunología , Factores de Riesgo , Linfocitos T/inmunología
18.
J Thromb Haemost ; 21(4): 800-813, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36696179

RESUMEN

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.


Asunto(s)
Hemofilia A , Humanos , Hemofilia A/diagnóstico , Hemofilia A/genética , Etnicidad , Factor VIII/genética , Mutación , Intrones
19.
Front Immunol ; 14: 1128641, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36936969

RESUMEN

Background: Almost half of severe hemophilia A (HA) is caused by an intron 22 inversion mutation (Int22Inv), which disrupts the 26-exon F8 gene. Inverted F8 mRNA exons 1-22 are transcribed, while F8B mRNA, containing F8 exons 23-26, is transcribed from a promoter within intron 22. Neither FVIII activity nor FVIII antigen (cross-reacting material, CRM) are detectable in plasma of patients with an intron-22 inversion. Objectives: To test the hypothesis that (putative) intracellular synthesis of FVIII proteins encoded by inverted F8 and F8B mRNAs confers T-cell tolerance to almost the entire FVIII sequence, and to evaluate the immunogenicity of the region encoded by the F8 exon 22-23 junction sequence. Patients/Methods: Peripheral blood mononuclear cells (PBMCs) from 30 severe or moderate HA subjects (17 with an Int22Inv mutation) were tested by ELISPOT assays to detect cytokine secretion in response to FVIII proteins and peptides and to map immunodominant T-cell epitopes. Potential immunogenicity of FVIII sequences encoded by the F8 exon 22-23 junction region was also tested using peptide-MHCII binding assays. Results: Eight of the Int22Inv subjects showed robust cytokine secretion from PBMCs stimulated with FVIII proteins and/or peptides, consistent with earlier publications from the Conti-Fine group. Peptide ELISPOT assays identified immunogenic regions of FVIII. Specificity for sequences encoded within F8 mRNA exons 1-22 and F8B mRNA was confirmed by staining Int22Inv CD4+ T cells with peptide-loaded HLA-Class II tetramers. FVIII peptides spanning the F8 exon 22-23 junction (encoding M2124-V2125) showed limited binding to MHCII proteins and low immunogenicity, with cytokine secretion from only one Int22Inv subject. Conclusions: PBMCs from multiple subjects with an Int22Inv mutation, with and without a current FVIII inhibitor, responded to FVIII epitopes. Furthermore, the FVIII region encoded by the exon 22-23 junction sequence was not remarkably immunoreactive and is therefore unlikely to contain an immunodominant, promiscuous CD4+ T-cell epitope. Our results indicate that putative intracellular expression of partial FVIII proteins does not confer T-cell tolerance to FVIII regions encoded by inverted F8 mRNA or F8B mRNA.


Asunto(s)
Hemofilia A , Humanos , Factor VIII , Intrones/genética , Leucocitos Mononucleares , Mutación , Péptidos/genética , Epítopos de Linfocito T/genética , Inversión Cromosómica , Linfocitos T CD4-Positivos , ARN Mensajero/genética , Citocinas/genética
20.
Blood Adv ; 7(17): 4983-4998, 2023 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-36459498

RESUMEN

The most common complication in hemophilia A (HA) treatment, affecting 25% to 30% of patients with severe HA, is the development of alloimmune inhibitors that foreclose the ability of infused factor VIII (FVIII) to participate in coagulation. Inhibitors confer significant pathology on affected individuals and present major complexities in their management. Inhibitors are more common in African American patients, and it has been hypothesized that this is a consequence of haplotype (H)-treatment product mismatch. F8 haplotypes H1 to H5 are defined by nonsynonymous single-nucleotide polymorphisms encoding sequence variations at FVIII residues 1241, 2238, and 484. Haplotypes H2 to H5 are more prevalent in individuals with Black African ancestry, whereas 80% to 90% of the White population has the H1 haplotype. This study used an established multiplex fluorescence immunoassay to determine anti-FVIII antibody titers in plasma from 394 individuals with HA (188 Black, 206 White), measuring their binding to recombinant full-length H1 and H2 and B-domain-deleted (BDD) H1/H2, H3/H5, and H4 FVIII proteins. Inhibitor titers were determined using a chromogenic assay and linear B-cell epitopes characterized using peptide microarrays. FVIII-reactive antibodies were readily detected in most individuals with HA, with higher titers in those with a current inhibitor, as expected. Neither total nor inhibitory antibody titers correlated with F8 haplotype mismatches, and peptides with D1241E and M2238V polymorphisms did not comprise linear B-cell epitopes. Interestingly, compared with the full-length FVIII products, the BDD-FVIII proteins were markedly more reactive with plasma antibodies. The stronger immunoreactivity of BDD-FVIII suggests that B-domain removal might expose novel B-cell epitopes, perhaps through conformational rearrangements of FVIII domains.


Asunto(s)
Hemofilia A , Hemostáticos , Humanos , Factor VIII/metabolismo , Haplotipos , Epítopos de Linfocito B , Blanco , Anticuerpos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA