Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
PLoS Comput Biol ; 9(5): e1003066, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23696725

RESUMO

The development of neutralizing anti-drug-antibodies to the Factor VIII protein-therapeutic is currently the most significant impediment to the effective management of hemophilia A. Common non-synonymous single nucleotide polymorphisms (ns-SNPs) in the F8 gene occur as six haplotypes in the human population (denoted H1 to H6) of which H3 and H4 have been associated with an increased risk of developing anti-drug antibodies. There is evidence that CD4+ T-cell response is essential for the development of anti-drug antibodies and such a response requires the presentation of the peptides by the MHC-class-II (MHC-II) molecules of the patient. We measured the binding and half-life of peptide-MHC-II complexes using synthetic peptides from regions of the Factor VIII protein where ns-SNPs occur and showed that these wild type peptides form stable complexes with six common MHC-II alleles, representing 46.5% of the North American population. Next, we compared the affinities computed by NetMHCIIpan, a neural network-based algorithm for MHC-II peptide binding prediction, to the experimentally measured values and concluded that these are in good agreement (area under the ROC-curve of 0.778 to 0.972 for the six MHC-II variants). Using a computational binding predictor, we were able to expand our analysis to (a) include all wild type peptides spanning each polymorphic position; and (b) consider more MHC-II variants, thus allowing for a better estimation of the risk for clinical manifestation of anti-drug antibodies in the entire population (or a specific sub-population). Analysis of these computational data confirmed that peptides which have the wild type sequence at positions where the polymorphisms associated with haplotypes H3, H4 and H5 occur bind MHC-II proteins significantly more than a negative control. Taken together, the experimental and computational results suggest that wild type peptides from polymorphic regions of FVIII constitute potential T-cell epitopes and thus could explain the increased incidence of anti-drug antibodies in hemophilia A patients with haplotypes H3 and H4.


Assuntos
Biologia Computacional/métodos , Fator VIII/genética , Fator VIII/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/metabolismo , Sequência de Aminoácidos , Anticorpos/química , Anticorpos/imunologia , Anticorpos/metabolismo , População Negra , Fator VIII/química , Fator VIII/metabolismo , Haplótipos , Hemofilia A , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/metabolismo , Polimorfismo de Nucleotídeo Único , Ligação Proteica , Fatores de Risco
2.
Biologicals ; 40(3): 191-5, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22239993

RESUMO

The infusion of plasma-derived or recombinant factors to treat bleeding disorders such as hemophilia A and B is a success story in the management of a chronic disease. The effectiveness of this approach is however limited by challenges with adverse effects of treatment. The most notable of these are the development of inhibitory antibodies that target the protein therapeutic. The current standard of care for management of hemophiliacs is prophylactic treatment that includes frequent infusions of a Factor VIII product. Failure to comply with the prophylactic regimen is a major hurdle in the management of these patients. We discuss here more recent findings that argue for a pharmacogenetic approach to understanding (and eventually circumventing) immunogenicity. We also review strategies used to bioengineer coagulation factors to extend the half-lives of coagulation proteins. The rapid progress in the last few years to bioengineer coagulation factors in different ways to attain this goal is described. Finally, novel technologies and potential products are emerging that utilize synthetic molecules in lieu of replacement proteins obviating the limitations associated with replacement therapies.


Assuntos
Fator IX/uso terapêutico , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Hemofilia B/tratamento farmacológico , Plasma/química , Animais , Aptâmeros de Nucleotídeos/uso terapêutico , Fatores de Coagulação Sanguínea/uso terapêutico , Hemofilia A/sangue , Hemofilia B/sangue , Humanos , Lipoproteínas/antagonistas & inibidores
3.
Sci Transl Med ; 9(372)2017 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-28077675

RESUMO

Immunogenicity is an important consideration in the licensure of a therapeutic protein because the development of neutralizing anti-drug antibodies (ADAs) can affect both safety and efficacy. Neoantigens introduced by bioengineering of a protein drug are a particular cause for concern. The development of a bioengineered recombinant factor VIIa (rFVIIa) analog was discontinued after phase 3 trials because of the development of ADAs. The unmodified parent molecule (rFVIIa), on the other hand, has been successfully used as a drug for more than two decades with no reports of immunogenicity in congenital hemophilia patients with inhibitors. We used computational and experimental methods to demonstrate that the observed ADAs could have been elicited by neoepitopes in the engineered protein. The human leukocyte antigen type of the patients who developed ADAs is consistent with this hypothesis of a neoepitope-driven immune response, a finding that might have implications for the preclinical screening of therapeutic protein analogs.


Assuntos
Fator VIII/imunologia , Hemofilia A/sangue , Hemofilia A/terapia , Engenharia de Proteínas/métodos , Adolescente , Adulto , Anticorpos Neutralizantes/imunologia , Proliferação de Células , Criança , Estudos Cross-Over , Interpretação Estatística de Dados , Método Duplo-Cego , Epitopos/imunologia , Fator VIIa/imunologia , Antígenos HLA/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Masculino , Mutação , Proteínas Recombinantes/imunologia , Software , Resultado do Tratamento , Adulto Jovem
4.
PLoS One ; 10(7): e0132433, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26176629

RESUMO

Hemophilia A (HA) is a bleeding disorder caused by deficiency of functional plasma clotting factor VIII (FVIII). Genetic mutations in the gene encoding FVIII (F8) have been extensively studied. Over a thousand different mutations have been reported in the F8 gene. These span a diverse range of mutation types, namely, missense, splice-site, deletions of single and multiple exons, inversions, etc. There is nonetheless evidence that other molecular mechanisms, in addition to mutations in the gene encoding the FVIII protein, may be involved in the pathobiology of HA. In this study, global small ncRNA expression profiling analysis of whole blood from HA patients, and controls, was performed using high-throughput ncRNA microarrays. Patients were further sub-divided into those that developed neutralizing-anti-FVIII antibodies (inhibitors) and those that did not. Selected differentially expressed ncRNAs were validated by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) analysis. We identified several ncRNAs, and among them hsa-miR-1246 was significantly up-regulated in HA patients. In addition, miR-1246 showed a six-fold higher expression in HA patients without inhibitors. We have identified an miR-1246 target site in the noncoding region of F8 mRNA and were able to confirm the suppressory role of hsa-miR-1246 on F8 expression in a stable lymphoblastoid cell line expressing FVIII. These findings suggest several testable hypotheses vis-à-vis the role of nc-RNAs in the regulation of F8 expression. These hypotheses have not been exhaustively tested in this study as they require carefully curated clinical samples.


Assuntos
Fator VIII/metabolismo , Regulação da Expressão Gênica , Hemofilia A/genética , MicroRNAs/genética , MicroRNAs/metabolismo , RNA não Traduzido/sangue , Adolescente , Adulto , Sequência de Bases , Inibidores dos Fatores de Coagulação Sanguínea/sangue , Linhagem Celular Tumoral , Criança , Pré-Escolar , Fator VIII/química , Fator VIII/genética , Perfilação da Expressão Gênica , Hemofilia A/patologia , Humanos , Lactente , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Alinhamento de Sequência , Regulação para Cima , Adulto Jovem
5.
Biomed Res Int ; 2013: 793502, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555096

RESUMO

Flow cytometry is widely used in cancer research for diagnosis, detection of minimal residual disease, as well as immune monitoring and profiling following immunotherapy. Detection of specific host proteins for diagnosis predominantly uses quantitative PCR and western blotting assays. In this study, we optimized a flow cytometry-based detection assay for Factor VIII protein in peripheral blood mononuclear cells (PBMCs). An indirect intracellular staining (ICS) method was standardized using monoclonal antibodies to different domains of human Factor VIII protein. The FVIII protein expression level was estimated by calculating the mean and median fluorescence intensities (MFI) values for each monoclonal antibody. ICS staining of transiently transfected cell lines supported the method's specificity. Intracellular FVIII protein expression was also detected by the monoclonal antibodies used in the study in PBMCs of five blood donors. In summary, our data suggest that intracellular FVIII detection in PBMCs of hemophilia A patients can be a rapid and reliable method to detect intracellular FVIII levels.


Assuntos
Fator VIII/biossíntese , Citometria de Fluxo , Hemofilia A/diagnóstico , Leucócitos Mononucleares/citologia , Animais , Anticorpos Monoclonais/isolamento & purificação , Células CHO , Linhagem Celular , Cricetinae , Fator VIII/isolamento & purificação , Expressão Gênica , Células HEK293 , Hemofilia A/imunologia , Humanos , Leucócitos Mononucleares/metabolismo , Camundongos , Estrutura Terciária de Proteína
6.
Nat Med ; 19(10): 1318-24, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24037092

RESUMO

Neutralizing antibodies (inhibitors) to replacement factor VIII (FVIII, either plasma derived or recombinant) impair the effective management of hemophilia A. Individuals with hemophilia A due to major deletions of the FVIII gene (F8) lack antigenically cross-reactive material in their plasma ("CRM-negative"), and the prevalence of inhibitors in these individuals may be as high as 90%. Conversely, individuals with hemophilia A caused by F8 missense mutations are CRM-positive, and their overall prevalence of inhibitors is <10% (ref. 2). Individuals with the F8 intron 22 inversion (found in ∼50% of individuals with severe hemophilia A) have been grouped with the former on the basis of their genetic defect and CRM-negative status. However, only ∼20% of these individuals develop inhibitors. Here we demonstrate that the levels of F8 mRNA and intracellular FVIII protein in B lymphoblastoid cells and liver biopsies from individuals with the intron 22 inversion are comparable to those in healthy controls. These results support the hypothesis that most individuals with the intron 22 inversion are tolerized to FVIII and thus do not develop inhibitors. Furthermore, we developed a new pharmacogenetic algorithm that permits the stratification of inhibitor risk for individuals and subpopulations by predicting the immunogenicity of replacement FVIII using, as input, the number of putative T cell epitopes in the infused protein and the competence of major histocompatibility complex class II molecules to present such epitopes. This algorithm showed statistically significant accuracy in predicting the presence of inhibitors in 25 unrelated individuals with the intron 22 inversion.


Assuntos
Inversão Cromossômica , Fator VIII/biossíntese , Fator VIII/uso terapêutico , Hemofilia A/tratamento farmacológico , Íntrons , Anticorpos Neutralizantes/imunologia , Fator VIII/genética , Fator VIII/imunologia , Células HEK293 , Humanos , Farmacogenética
7.
PLoS One ; 7(2): e31948, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22384109

RESUMO

Therapeutic proteins are derived from complex expression/production systems, which can result in minor conformational changes due to preferential codon usage in different organisms, post-translational modifications, etc. Subtle conformational differences are often undetectable by bioanalytical methods but can sometimes profoundly impact the safety, efficacy and stability of products. Numerous bioanalytical methods exist to characterize the primary structure of proteins, post translational modifications; protein-substrate/protein/protein interactions and functional bioassays are available for most proteins that are developed as products. There are however few analytical techniques to detect changes in the tertiary structure of proteins suitable for use during drug development and quality control. For example, x-ray crystallography and NMR are impractical for routine use and do not capture the heterogeneity of the product. Conformation-sensitive antibodies can be used to map proteins. However the development of antibodies to represent sufficient epitopes can be challenging. Other limitations of antibodies include limited supply, high costs, heterogeneity and batch to batch variations in titer. Here we provide proof-of-principle that DNA aptamers to thrombin can be used as surrogate antibodies to characterize conformational changes. We show that aptamers can be used in assays using either an ELISA or a label-free platform to characterize different thrombin products. In addition we replicated a heat-treatment procedure that has previously been shown to not affect protein activity but can result in conformational changes that have serious adverse consequences. We demonstrate that a panel of aptamers (but not an antibody) can detect changes in the proteins even when specific activity is unaffected. Our results indicate a novel approach to monitor even small changes in the conformation of proteins which can be used in a routine drug-development and quality control setting. The technique can provide an early warning of structural changes during the manufacturing process that could have consequential outcomes downstream.


Assuntos
Aptâmeros de Peptídeos/química , Proteínas/química , Animais , Anticorpos Monoclonais/química , Técnicas Biossensoriais , Bovinos , Códon , Ensaio de Imunoadsorção Enzimática/métodos , Epitopos/química , Humanos , Interferometria/métodos , Cinética , Ligação Proteica , Conformação Proteica , Processamento de Proteína Pós-Traducional , Proteínas/uso terapêutico , Sensibilidade e Especificidade , Especificidade por Substrato , Temperatura , Trombina/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA