Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
2.
Mol Ther ; 4(3): 201-10, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11545610

RESUMO

The safety of several gene therapy approaches for treatment of the severe, X-linked bleeding disorder hemophilia is currently being evaluated in early phase clinical trials. One strategy seeks to correct deficiency of functional coagulation factor IX (hemophilia B) by intramuscular (IM) administration of an adeno-associated viral (AAV) vector. A potentially serious complication of any treatment for hemophilia is formation of inhibitory antibodies against the coagulation factor protein, a risk that increases in the setting of null mutations in the factor IX gene (F9). Here, we describe hemophilia B mice with a large F9 deletion that form inhibitors within 1 to 2 months after IM administration of an AAV vector expressing mouse F9 or after repeated intravenous infusion of mouse F9 concentrate. In both cases, inhibitors are primarily IgG1 immunoglobulins representing a Th2-driven humoral immune response. We further demonstrate that anti-mouse F9 antibody formation in the gene-based approach can be reduced by transient immune modulation at the time of vector administration. Moreover, this maneuver resulted in complete absence of anti-mouse F9 and sustained expression of functional mouse F9 in some hemophilia B mice, particularly in those animals treated with the immunosuppressive drug cyclophosphamide. These data have direct relevance for design of clinical trials and strategies aimed at avoiding immune responses against a secreted transgene product.


Assuntos
Dependovirus/genética , Fator IX/genética , Fator IX/imunologia , Deleção de Genes , Terapia Genética/métodos , Hemofilia B/genética , Hemofilia B/imunologia , Animais , Anticorpos/imunologia , Células CHO , Cricetinae , Ciclofosfamida/farmacologia , Fator IX/administração & dosagem , Fator IX/uso terapêutico , Expressão Gênica/efeitos dos fármacos , Vetores Genéticos/genética , Hemofilia B/tratamento farmacológico , Hemofilia B/terapia , Imunossupressores/farmacologia , Injeções Intramusculares , Injeções Intravenosas , Camundongos , Camundongos Endogâmicos C57BL , Tempo de Tromboplastina Parcial , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Risco , Fatores de Tempo , Transgenes/genética
3.
Gene Ther ; 8(5): 354-61, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11313811

RESUMO

Inbred immunocompetent C57BL/6 mice have been a favored strain to study transgene expression of human blood coagulation factor IX (hF.IX) from viral vectors because systemic expression of the secreted protein is not limited by antibody responses following intravenous (i.v.) injection of vector. For example, i.v. injection of an adenoviral (Ad) vector results in sustained expression of hF.IX in normal or hemophilic C57BL/6 mice, while anti-hF.IX antibodies rapidly emerge in other strains (Gene Therapy 4: 473; Blood 91: 784). To investigate these observations further, we injected naive C57BL/6 mice and C57BL/6 mice with pre-existing anti-hF.IX with Ad-hF.IX vector via peripheral vein. All mice expressed hF.IX antigen without detectable anti-hF.IX, even when challenged with hF.IX in different immunogenic settings at later time points. Moreover, in mice with pre-existing immunity, anti-hF.IX titers diminished to undetectable levels after i.v. administration of Ad-hF.IX. Lymphocytes from mice that had received Ad-hF.IX i.v. failed to proliferate when stimulated with hF.IX in vitro after the animals had been repeatedly challenged with hF.IX protein formulated in complete Freund's adjuvant. Thus, absence of anti-hF.IX in C57BL/6 mice after i.v. injection of Ad vector is not due to ignorance to the foreign transgene product. Similar experiments in other strains showed that immune tolerance to hF.IX does not correlate with the strain haplotype or expression of IL-10 cytokine. Given the well-documented immunogenicity of the first-generation adenoviral vector, data from C57BL/6 mice may therefore grossly underestimate immunological consequences in certain gene therapy protocols.


Assuntos
Adenoviridae/genética , Fator IX/imunologia , Vetores Genéticos , Tolerância Imunológica , Proteínas E1 de Adenovirus/genética , Proteínas E3 de Adenovirus/genética , Animais , Divisão Celular/imunologia , Haplótipos , Injeções Intravenosas , Interleucina-10/metabolismo , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Linfócitos T/imunologia
4.
Blood ; 97(1): 130-8, 2001 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-11133752

RESUMO

Recent data demonstrate that the introduction into skeletal muscle of an adeno-associated viral (AAV) vector expressing blood coagulation factor IX (F.IX) can result in long-term expression of the transgene product and amelioration of the bleeding diathesis in animals with hemophilia B. These data suggest that biologically active F.IX can be synthesized in skeletal muscle. Factor IX undergoes extensive posttranslational modifications in the liver, the normal site of synthesis. In addition to affecting specific activity, these posttranslational modifications can also affect recovery, half-life in the circulation, and the immunogenicity of the protein. Before initiating a human trial of an AAV-mediated, muscle-directed approach for treating hemophilia B, a detailed biochemical analysis of F.IX synthesized in skeletal muscle was carried out. As a model system, human myotubes transduced with an AAV vector expressing F.IX was used. F.IX was purified from conditioned medium using a novel strategy designed to purify material representative of all species of rF.IX in the medium. Purified F.IX was analyzed by sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE), N-terminal sequence analysis, chemical gamma-carboxyglutamyl analysis, carbohydrate analysis, assays for tyrosine sulfation, and serine phosphorylation, and for specific activity. Results show that myotube-synthesized F.IX has specific activity similar to that of liver-synthesized F.IX. Posttranslational modifications critical for specific activity, including removal of the signal sequence and propeptide, and gamma-carboxylation of the N-terminal glutamic acid residues, are also similar, but carbohydrate analysis and assessment of tyrosine sulfation and serine phosphorylation disclose differences. In vivo experiments in mice showed that these differences affect recovery but not half-life of muscle-synthesized F.IX.


Assuntos
Fator IX/biossíntese , Fibras Musculares Esqueléticas/citologia , Processamento de Proteína Pós-Traducional , Ácido 1-Carboxiglutâmico/análise , Adenoviridae/genética , Carboidratos/análise , Técnicas de Cultura de Células , Meios de Cultivo Condicionados/química , Fator IX/química , Fator IX/genética , Vetores Genéticos , Humanos , Dados de Sequência Molecular , Fibras Musculares Esqueléticas/metabolismo , Tempo de Tromboplastina Parcial , Fosforilação , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Análise de Sequência de Proteína , Serina/metabolismo , Sulfatos , Transdução Genética , Tirosina/metabolismo
5.
Am J Pharmacogenomics ; 1(2): 137-44, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-12174674

RESUMO

The year 2000 saw the first successful treatment of a genetic disorder by gene therapy. Pediatric patients with X-linked severe combined immunodeficiency disorder (SCID-X1) received autologous CD34+ hematopoietic cells following ex vivo gene transfer using a retroviral vector, with subsequent demonstration of improved immune responses. A number of preclinical and clinical studies have been conducted with the aim of developing gene therapy for hemophilia, Fanconi anemia, sickle cell disease, beta-thalassemia, chronic granulomatous disease, and other inherited hematological disorders. The greatest advances in novel approaches toward treatment of hematological disorders have been made in hemophilia, with 3 current phase I clinical trials ongoing. Two trials are investigating the safety and feasibility of utilizing either an ex vivo, non-viral gene transfer technique or an intravenous infusion of a retroviral vector to treat adults with severe hemophilia A (factor VIII deficiency). The third study involves intramuscular administration of an adeno-associated viral (AAV) vector for expression of factor IX in adult patients with hemophilia B. Results from this study and from preclinical studies preceding the trial demonstrate that it is possible to safely administer high doses of a viral vector in vivo.


Assuntos
Terapia Genética/métodos , Doenças Hematológicas/genética , Doenças Hematológicas/terapia , Animais , Anemia de Fanconi/genética , Anemia de Fanconi/terapia , Terapia Genética/estatística & dados numéricos , Terapia Genética/tendências , Doença Granulomatosa Crônica/genética , Doença Granulomatosa Crônica/terapia , Hemoglobinopatias/genética , Hemoglobinopatias/terapia , Hemofilia A/genética , Hemofilia A/terapia , Humanos , Imunodeficiência Combinada Severa/genética , Imunodeficiência Combinada Severa/terapia
6.
Mol Ther ; 1(3): 225-35, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10933938

RESUMO

Defining immune responses against the secreted transgene product in a gene therapy setting is critical for treatment of genetic diseases such as hemophilia B (coagulation factor IX deficiency). We have previously shown that intramuscular administration of an adeno-associated viral (AAV) vector results in stable expression of therapeutic levels of factor IX (F.IX) and may be associated with humoral immune responses against F.IX. This study demonstrates that intramuscular injection of an AAV vector expressing F.IX fails to activate F.IX-specific cytotoxic T lymphocytes (CTLs) in hemostatically normal or in hemophilia B mice, so that there is an absence of cellular immune responses against F.IX. However, transgene-derived F.IX can cause B cell responses characterized by production of T helper cell-dependent antibodies (predominantly IgG1, but also IgG2 subclasses) resulting from activation of CD4+ T helper cells primarily of the Th2 subset. In contrast, administration of an adenoviral vector efficiently activated F.IX-specific CTLs and T helper cells of both Th1 and Th2 subsets, leading to inflammation and destruction of transduced muscle tissue and activation of B cells as well. Therefore, vector sequences fundamentally influence T cell responses against transgene-encoded F.IX. In conclusion, activation of the immune system in AAV-mediated gene transfer is restricted to pathways mediated by F.IX antigen presentation through MHC class II determinants resulting in T and B cell responses that are more comparable to responses in the setting of protein infusion rather than of viral infection/gene transfer.


Assuntos
Fator IX/metabolismo , Técnicas de Transferência de Genes/efeitos adversos , Vetores Genéticos/imunologia , Subpopulações de Linfócitos T/imunologia , Transgenes , Adenoviridae/genética , Transferência Adotiva , Animais , Citocinas/metabolismo , Fator IX/genética , Fator IX/imunologia , Imunofluorescência , Hemofilia B/imunologia , Hemofilia B/metabolismo , Imunoglobulina G/biossíntese , Imunoglobulina G/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo
7.
Blood ; 95(8): 2536-42, 2000 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-10753832

RESUMO

Hemophilia B is caused by the absence of functional coagulation factor IX (F.IX) and represents an important model for treatment of genetic diseases by gene therapy. Recent studies have shown that intramuscular injection of an adeno-associated viral (AAV) vector into mice and hemophilia B dogs results in vector dose-dependent, long-term expression of biologically active F.IX at therapeutic levels. In this study, we demonstrate that levels of expression of approximately 300 ng/mL (6% of normal human F.IX levels) can be reached by intramuscular injection of mice using a 2- to 4-fold lower vector dose (1 x 10(11) vector genomes/mouse, injected into 4 intramuscular sites) than previously described. This was accomplished through the use of an improved expression cassette that uses the cytomegalovirus (CMV) immediate early enhancer/promoter in combination with a 1.2-kilobase portion of human skeletal actin promoter. These results correlated with enhanced levels of F.IX transcript and secreted F.IX protein in transduced murine C2C12 myotubes. Systemic F.IX expression from constructs containing the CMV enhancer/promoter alone was 120 to 200 ng/mL in mice injected with 1 x 10(11) vector genomes. Muscle-specific promoters performed poorly for F.IX transgene expression in vitro and in vivo. However, the incorporation of a sequence from the alpha-skeletal actin promoter containing at least 1 muscle-specific enhancer and 1 enhancer-like element further improved muscle-derived expression of F.IX from a CMV enhancer/promoter-driven expression cassette over previously published results. These findings will allow the design of a clinical protocol for therapeutic levels of F.IX expression with lower vector doses, thus enhancing efficacy and safety of the protocol. (Blood. 2000;95:2536-2542)


Assuntos
Fator IX/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Actinas/genética , Animais , Citomegalovirus/genética , Cães , Elementos Facilitadores Genéticos/genética , Fator IX/biossíntese , Expressão Gênica , Humanos , Camundongos , Músculo Esquelético
8.
Nat Med ; 5(1): 56-63, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9883840

RESUMO

Hemophilia B is a severe X-linked bleeding diathesis caused by the absence of functional blood coagulation factor IX, and is an excellent candidate for treatment of a genetic disease by gene therapy. Using an adeno-associated viral vector, we demonstrate sustained expression (>17 months) of factor IX in a large-animal model at levels that would have a therapeutic effect in humans (up to 70 ng/ml, adequate to achieve phenotypic correction, in an animal injected with 8.5x10(12) vector particles/kg). The five hemophilia B dogs treated showed stable, vector dose-dependent partial correction of the whole blood clotting time and, at higher doses, of the activated partial thromboplastin time. In contrast to other viral gene delivery systems, this minimally invasive procedure, consisting of a series of percutaneous intramuscular injections at a single timepoint, was not associated with local or systemic toxicity. Efficient gene transfer to muscle was shown by immunofluorescence staining and DNA analysis of biopsied tissue. Immune responses against factor IX were either absent or transient. These data provide strong support for the feasibility of the approach for therapy of human subjects.


Assuntos
Dependovirus , Fator IX/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Hemofilia B/terapia , Animais , DNA Viral/análise , Dependovirus/genética , Modelos Animais de Doenças , Cães , Fator IX/imunologia , Expressão Gênica , Hemofilia B/imunologia , Humanos , Injeções Intramusculares , Masculino , Fatores de Tempo , Células Tumorais Cultivadas
9.
J Pediatr ; 133(6): 777-81, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9842043

RESUMO

OBJECTIVE: Resistance to activated protein C (APC) has been identified as a risk factor for thrombotic disease in adults. In over 90% of cases, the basis for the APC resistance is a mutation in the coagulation factor V gene (factor V Leiden) that renders the protein more resistant to inactivation by APC. We sought to determine the prevalence of the factor V Leiden (FVL) mutation in neonates and children who had experienced an arterial or venous thromboembolic event. STUDY DESIGN: We retrospectively analyzed the clinical records of 33 neonates and 52 children with thromboembolic disease. Screening for the FVL mutation was performed by DNA analysis, allowing for identification of patients as normal, heterozygous, or homozygous. RESULTS: Of the 85 patients studied, 12 (14.1%) were heterozygous for FVL; none were homozygous. Of the 47 patients who had arterial central nervous system events, 8 (17%) were positive for the FVL mutation, including 6 of 22 (27%) neonates. Of those patients who had a venous thrombosis, 4 of 32 (12.5%) were FVL positive. None of the 85 patients had protein C deficiency, 3.5% had protein S deficiency, 1.2% had antithrombin III deficiency, and 16.5% had anti-phospholipid antibodies. CONCLUSION: These data suggest that the FVL mutation plays a role in the development of arterial and venous thrombotic events in neonates and children.


Assuntos
Fator V/genética , Mutação , Tromboembolia/genética , Adolescente , Adulto , Criança , Pré-Escolar , Análise Mutacional de DNA , Humanos , Lactente , Recém-Nascido , Estudos Retrospectivos , Trombose Venosa/genética
10.
Blood ; 91(12): 4600-7, 1998 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-9616156

RESUMO

Recombinant adeno-associated virus vectors (AAV) were prepared in high titer (10(12) to 10(13) particles/mL) for the expression of human factor IX after in vivo transduction of murine hepatocytes. Injection of AAV-CMV-F.IX (expression from the human cytomegalovirus IE enhancer/promoter) into the portal vein of adult mice resulted in no detectable human factor IX in plasma, but in mice injected intravenously as newborns with the same vector, expression was initially 55 to 110 ng/mL. The expression in the liver was mostly transient, and plasma levels decreased to undetectable levels within 5 weeks. However, long-term expression of human F.IX was detected by immunofluorescence staining in 0.25% of hepatocytes 8 to 10 months postinjection. The loss of expression was likely caused by suppression of the CMV promoter, because polymerase chain reaction data showed no substantial loss of vector DNA in mouse liver. A second vector in which F.IX expression was controlled by the human EF1alpha promoter was constructed and injected into the portal vein of adult C57BL/6 mice at a dose of 6.3 x 10(10) particles. This resulted in therapeutic plasma levels (200 to 320 ng/mL) for a period of at least 6 months, whereas no human F.IX was detected in plasma of mice injected with AAV-CMV-F.IX. Doses of AAV-EF1alpha-F. IX of 2.7 x 10(11) particles resulted in plasma levels of 700 to 3, 200 ng/mL. Liver-derived expression of human F.IX from the AAV-EF1alpha-F.IX vector was confirmed by immunofluorescence staining. We conclude that recombinant AAV can efficiently transduce hepatocytes and direct stable expression of an F.IX transgene in mouse liver, but sustained expression is critically dependent on the choice of promoter.


Assuntos
Dependovirus , Fator IX/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Fígado/fisiologia , Adulto , Animais , Imunofluorescência , Regulação da Expressão Gênica , Humanos , Camundongos
11.
Blood ; 91(3): 784-90, 1998 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9446637

RESUMO

Mice with hemophilia B have been engineered using gene targeting techniques. These animals exhibit severe factor IX deficiency and a clinical phenotype that mirrors the human disease. We have bred the founder animals onto two different strains of mice, C57B1/6 and CD-1, and have sought to determine whether adenoviral vectors expressing human factor IX could correct the bleeding diathesis of mice with hemophilia B. Initial experiments showed that purified plasma-derived human factor IX added to murine factor IX-deficient plasma resulted in complete correction of the activated partial thromboplastin time (aPTT), and that injection of 10(11) particles of an adenoviral vector expressing human factor IX resulted in normalization of a modified aPTT in mouse plasma. As an additional method of assessing the function of human factor IX in the murine coagulation system, bleeding times were performed in normal, hemophilic, and adenoviral-treated hemophilic mice. By two different bleeding-time techniques, the treated hemophilic mice gave values identical to normal littermate controls, whereas the untreated hemophilic mice exhibited heavy blood loss and prolonged bleeding. There was a marked difference in antibody formation in the two strains of mice; 100% of the hemophilic CD-1 mice formed antibodies to human factor IX, but none of the C57B1/6 mice did. These data suggest that the C57B1/6 hemophilic mice will be more useful for gene transfer studies, while the CD-1 hemophilic mice may be of greater utility in studying the development of inhibitors.


Assuntos
Fator IX/uso terapêutico , Hemofilia B/tratamento farmacológico , Hemorragia/prevenção & controle , Adenoviridae/genética , Animais , Anticorpos/sangue , Tempo de Sangramento , Coagulação Sanguínea , Fator IX/genética , Fator IX/imunologia , Feminino , Expressão Gênica , Técnicas de Transferência de Genes , Vetores Genéticos , Hemofilia B/sangue , Hemofilia B/complicações , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Tempo de Tromboplastina Parcial
12.
Proc Natl Acad Sci U S A ; 94(11): 5804-9, 1997 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-9159155

RESUMO

We sought to determine whether intramuscular injection of a recombinant adeno-associated virus (rAAV) vector expressing human factor IX (hF.IX) could direct expression of therapeutic levels of the transgene in experimental animals. High titer (10(12)-10(13) vector genomes/ml) rAAV expressing hF.IX was prepared, purified, and injected into hindlimb muscles of C57BL/6 mice and Rag 1 mice. In the immunocompetent C57BL/6 mice, immunofluorescence staining of muscle harvested 3 months after injection demonstrated the presence of hF.IX protein, and PCR analysis of muscle DNA was positive for AAV DNA, but no hF.IX was detected in mouse plasma. Further studies showed that these mice had developed circulating antibodies to hF.IX. In follow-up experiments in Rag 1 mice, which carry a mutation in the recombinase activating gene-1 and thus lack functional B and T cells, similar results were seen on DNA analysis of muscle, but these mice also demonstrated therapeutic levels (200-350 ng/ml) of F. IX in the plasma. The time course of F.IX expression demonstrates that levels gradually increase over a period of several weeks before reaching a plateau that is stable 6 months after injection. In other experiments we demonstrate colocalization of hF.IX and collagen IV in intersitial spaces between muscle fibers. Collagen IV has recently been identified as a F.IX-binding protein; this finding explains the unusual pattern of immunofluorescent staining for F.IX shown in these experiments. Thus rAAV can be used to direct stable expression of therapeutic levels of F.IX after intramuscular injection and is a feasible strategy for treatment of patients with hemophilia B.


Assuntos
Dependovirus , Fator IX/biossíntese , Fator IX/genética , Técnicas de Transferência de Genes , Proteínas de Homeodomínio , Músculo Esquelético/metabolismo , Animais , Anticorpos Heterófilos/sangue , Primers do DNA , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Terapia Genética/métodos , Humanos , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Síndromes de Imunodeficiência/terapia , Injeções Intramusculares , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Músculo Esquelético/citologia , Reação em Cadeia da Polimerase , Biossíntese de Proteínas , Proteínas/genética
13.
Proc Natl Acad Sci U S A ; 93(7): 3056-61, 1996 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-8610167

RESUMO

Adenoviral vectors can direct high-level expression of a transgene, but, due to a host immune response to adenoviral antigens, expression is of limited duration, and repetitive administration has generally been unsuccessful. Exposure to foreign proteins beginning in the neonatal period may alter or ablate the immune response. We injected adult and neonatal (immunocompetent) CD-1 mice intravenously with an adenoviral vector expressing human blood coagulation factor IX. In both groups of mice, expression of human factor IX persisted for 12-16 weeks. However, in mice initially injected as adults, repeat administration of the vector resulted in no detectable expression of the transgene, whereas in mice initially injected in the neonatal period, repeat administration resulted in high-level expression of human factor IX. We show that animals that fail to express the transgene on repeat administration have developed high-titer neutralizing antibodies to adenovirus, whereas those that do express factor IX have not. This experimental model suggests that newborn mice can be tolerized to adenoviral vectors and demonstrates that at least one repeat injection of the adenoviral vector is possible; the model will be useful in elucidating the immunologic mechanisms underlying successful repeat administration of adenoviral vectors.


Assuntos
Fator IX/biossíntese , Expressão Gênica , Vetores Genéticos , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Animais Recém-Nascidos , Anticorpos Antivirais/biossíntese , Anticorpos Antivirais/sangue , Fator IX/análise , Fator IX/genética , Feminino , Humanos , Cinética , Óperon Lac , Masculino , Camundongos , Camundongos Endogâmicos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/sangue , Fatores de Tempo , beta-Galactosidase/análise , beta-Galactosidase/biossíntese
14.
Thromb Haemost ; 74(6): 1486-90, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8772225

RESUMO

Prenatal maternal vitamin K1 supplementation to improve the hemostatic status of the fetus may depend upon the route of administration and subsequent presentation at the placental barrier. We investigated intramuscular (IM) vs oral (PO) vitamin K1 supplementation in eight healthy, nonpregnant women of childbearing age. Pharmacokinetics were studied in each subject after a 5 mg IM dose and after a 5 mg oral dose of vitamin K1 approximately one month later. Plasma collected at the peak vitamin K level for each treatment was separated into very low density lipoproteins (VLDL), low density lipoprotein (LDL), high density lipoprotein (HDL) and lipoprotein-free fractions by density gradient ultracentrifugation. Vitamin K1 was measured in the plasma and lipoprotein fractions using HPLC. The concentration of vitamin K1 in plasma reached a peak 2 h after an IM dose and remained high throughout the 30 h course of the study. In contrast, the oral dose of vitamin K1 peaked at 4 h and rapidly decreased to near baseline by 18 to 30 h. The distribution of vitamin K1 in the lipid fractions was different for IM compared to PO. The percentage of vitamin K1 in the VLDL fraction at the peak for an oral dose was significantly higher than for an IM dose (80.8% +/- 3.5 vs 10.8% +/- 6.5, p < 0.0001). After the oral absorption stage, the subjects took 5 mg of vitamin K1 orally, once a day, for 12 days. No significant differences were observed for the following coagulation proteins and hemostatic markers measured immediately before and after long-term oral vitamin K supplementation: factor II, factor VII, protein C, and thrombin-antithrombin III complex. In conclusion, physiological processing of supplemented vitamin K1 differs in the IM vs PO routes of administration and 12 days of oral vitamin K1 does not alter the concentration of selected vitamin K-dependent coagulation proteins or thrombin-antithrombin complex generation.


Assuntos
Hemostasia/efeitos dos fármacos , Lipoproteínas/sangue , Vitamina K 1/farmacocinética , Administração Oral , Adulto , Fracionamento Químico , Feminino , Humanos , Injeções Intramusculares , Vitamina K 1/administração & dosagem
16.
J Am Coll Cardiol ; 21(5): 1039-47, 1993 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8459055

RESUMO

OBJECTIVES: In a Phase I clinical trial, we studied the antithrombotic and clinical effects of the synthetic competitive thrombin inhibitor, argatroban, in 43 patients with unstable angina pectoris. BACKGROUND: Thrombin has a pivotal role in platelet-mediated thrombosis associated with atheromatous plaque rupture in patients with an acute ischemic coronary syndrome. However, the efficacy of conventional heparin therapy to prevent ischemic events is limited and has been surpassed by that of specific thrombin inhibitors in experimental models of arterial thrombosis. METHODS: Intravenous infusion of the drug (0.5 to 5.0 micrograms/kg per min) for 4 h was monitored by sequential measurements of coagulation times and of indexes of thrombin activity in vivo followed by a 24-h clinical observation period. RESULTS: Significant dose-related increases in plasma drug concentrations and activated partial thromboplastin times (aPTT), but no bleeding time prolongation or spontaneous bleeding, was observed. Myocardial ischemia did not occur during therapy but, surprisingly, 9 of the 43 patients experienced an episode of unstable angina 5.8 +/- 2.6 h (mean +/- SD) after infusion. This early recurrent angina was correlated significantly with a higher argatroban dose and with greater prolongation of aPTT but not with other demographic, clinical, laboratory and angiographic characteristics. Pretreatment plasma concentrations of thrombin-antithrombin III complex and fibrinopeptide A were elevated two to three times above normal values. During infusion, thrombin-antithrombin III complex levels remained unchanged, whereas a significant 2.3-fold decrease in fibrinopeptide A concentrations was observed. By contrast, 2 h after infusion, thrombin-antithrombin III complex concentrations increased 3.9-fold over baseline measurements together with return of fibrinopeptide A levels to values before treatment with argatroban. CONCLUSIONS: In patients with unstable angina, argatroban inhibits clotting (aPTT prolongation) and thrombin activity toward fibrinogen (fibrinopeptide A decrease), but in vivo thrombin (thrombin-antithrombin III complex) formation is not suppressed. However, cessation of infusion is associated with rebound thrombin (thrombin-antithrombin III complex) generation and with an early dose-related recurrence of unstable angina. Although the mechanism of this clinical and biochemical rebound phenomenon remains to be determined, its implication for the clinical use of specific thrombin inhibitors in the management of ischemic coronary syndromes may be significant.


Assuntos
Angina Instável/tratamento farmacológico , Antitrombinas/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Ácidos Pipecólicos/uso terapêutico , Idoso , Angina Instável/sangue , Antitrombina III/análise , Antitrombinas/administração & dosagem , Antitrombinas/farmacologia , Arginina/análogos & derivados , Esquema de Medicação , Feminino , Fibrinopeptídeo A/análise , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Ácidos Pipecólicos/administração & dosagem , Ácidos Pipecólicos/farmacologia , Recidiva , Sulfonamidas , Trombina/análise , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA