Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Allergy ; 73(9): 1871-1880, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29688579

RESUMO

BACKGROUND: Effective inhibition of plasma kallikrein may have significant benefits for patients with hereditary angioedema due to deficiency of C1 inhibitor (C1-INH-HAE) by reducing the frequency of angioedema attacks. Avoralstat is a small molecule inhibitor of plasma kallikrein. This study (OPuS-2) evaluated the efficacy and safety of prophylactic avoralstat 300 or 500 mg compared with placebo. METHODS: OPuS-2 was a Phase 3, multicenter, randomized, double-blind, placebo-controlled, parallel-group study. Subjects were administered avoralstat 300 mg, avoralstat 500 mg, or placebo orally 3 times per day for 12 weeks. The primary efficacy endpoint was the angioedema attack rate based on adjudicator-confirmed attacks. RESULTS: A total of 110 subjects were randomized and dosed. The least squares (LS) mean attack rates per week were 0.589, 0.675, and 0.593 for subjects receiving avoralstat 500 mg, avoralstat 300 mg, and placebo, respectively. Overall, 1 subject in each of the avoralstat groups and no subjects in the placebo group were attack-free during the 84-day treatment period. The LS mean duration of all confirmed attacks was 25.4, 29.4, and 31.4 hours for the avoralstat 500 mg, avoralstat 300 mg, and placebo groups, respectively. Using the Angioedema Quality of Life Questionnaire (AE-QoL), improved QoL was observed for the avoralstat 500 mg group compared with placebo. Avoralstat was generally safe and well tolerated. CONCLUSIONS: Although this study did not demonstrate efficacy of avoralstat in preventing angioedema attacks in C1-INH-HAE, it provided evidence of shortened angioedema episodes and improved QoL in the avoralstat 500 mg treatment group compared with placebo.


Assuntos
Angioedemas Hereditários/prevenção & controle , Inibidores Enzimáticos/uso terapêutico , Calicreína Plasmática/antagonistas & inibidores , Administração Oral , Adulto , Angioedemas Hereditários/diagnóstico , Angioedemas Hereditários/tratamento farmacológico , Progressão da Doença , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/efeitos adversos , Inibidores Enzimáticos/farmacocinética , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Qualidade de Vida , Recidiva , Resultado do Tratamento
2.
Allergy ; 71(12): 1676-1683, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27154593

RESUMO

BACKGROUND: Avoralstat is a potent small-molecule oral plasma kallikrein inhibitor under development for treatment of hereditary angioedema (HAE). This first-in-human study evaluated the safety, tolerability, pharmacokinetics, and pharmacodynamics of avoralstat. METHODS: This double-blind, placebo-controlled, ascending-dose cohort trial evaluated avoralstat single doses of 50, 125, 250, 500, and 1000 mg and multiple doses up to 2400 mg daily (100, 200, 400, and 800 mg every 8 h [q8 h] up to 7 days). RESULTS: Avoralstat (n = 71) was generally well tolerated with no signals for a safety concern; there were no serious adverse events (AEs) or discontinuations due to AEs, and compared to placebo (n = 18), no notable difference in AEs. Four moderate severity AEs were reported in two subjects; syncope after a single 250 mg dose (one subject) and abdominal pain, back pain, and eczema after multiple doses of 800 mg avoralstat (one subject). For multiple-dose cohorts, the incidence of gastrointestinal AEs was highest at the 2400 mg/day dose. Elimination of avoralstat was bi-exponential with a terminal half-life of 12-31 h. Inhibition of plasma kallikrein was observed at all doses, and the degree of inhibition was highly correlated with avoralstat concentrations (R = 0.93). Mean avoralstat concentrations at doses ≥400 mg q8 h met or exceeded plasma kallikrein EC50 values throughout the dosing interval. CONCLUSION: Avoralstat was well tolerated, and drug exposure was sufficient to meet target levels for inhibition of plasma kallikrein. Based on these results, the 400 mg q8 h dose was selected for further evaluation in patients with HAE.


Assuntos
Angioedemas Hereditários/tratamento farmacológico , Calicreína Plasmática/antagonistas & inibidores , Inibidores de Proteases/administração & dosagem , Inibidores de Proteases/farmacocinética , Administração Oral , Adolescente , Adulto , Esquema de Medicação , Monitoramento de Medicamentos , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Inibidores de Proteases/efeitos adversos , Fatores de Tempo , Resultado do Tratamento , Adulto Jovem
3.
Clin Pharmacol Ther ; 89(2): 172-4, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21252934

RESUMO

The 2009 H1N1 influenza pandemic prompted the US Food and Drug Administration (FDA) to issue an emergency use authorization (EUA) for the intravenous antiviral peramivir, an unapproved neuraminidase inhibitor (NAI) currently under development. Peramivir use was limited to patients for whom other NAI therapy had failed or in whom oral or inhalational drug absorption was believed to be unreliable. This introduced a patient selection bias that precluded safety and efficacy assessment. Despite the challenges and risks, there was a compelling public health need for an intravenous agent during the 2009 H1N1 pandemic.


Assuntos
Antivirais/uso terapêutico , Ciclopentanos/uso terapêutico , Aprovação de Drogas , Indústria Farmacêutica , Serviços Médicos de Emergência , Guanidinas/uso terapêutico , Vírus da Influenza A Subtipo H1N1/efeitos dos fármacos , Influenza Humana/tratamento farmacológico , Ácidos Carbocíclicos , Ciclopentanos/efeitos adversos , Guanidinas/efeitos adversos , Humanos , Estados Unidos , United States Food and Drug Administration
4.
Oncologist ; 5(2): 162-8, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10794807

RESUMO

Prostate cancer has become the most common cancer among American men and is second only to lung cancer as a cause of male cancer-related death. Several treatment options exist for different stages of prostate cancer including observation, prostatectomy, radiation therapy, chemotherapy, and hormone therapy. Hormone therapy has evolved from the use of estrogens to gonadotropin-releasing hormone (GnRH) agonists and recently, investigational GnRH antagonists. GnRH receptor agonists such as leuprolide, bruserelin and goserelin have been used for the treatment of prostate cancer. These agonists eventually cause the inhibition of lutenizing hormone production, which in turn causes a suppression of testosterone and dihydrotestosterone, on which continued growth of prostate cancer cells depend. Several comparative studies of leuprorelin administered as daily injections or monthly depot injections have been reported. Disease progression was prevented in more than 72% of men administered daily leuprorelin, and in 82% to 89% of those receiving monthly depots. Another synthetic GnRH analog, goserelin, has been studied in a similar population of men with daily injections producing partial responses in 60% to 80% of men with previously untreated prostate cancer. Abarelix, a peptide antagonist of GnRH receptor, is also being studied for the treatment of prostate cancer. The discovery and development of GnRH antagonists may provide an important advance for patients with prostate cancer. Clearly the studies described herein, as well as many others, outline an exciting era of research to define the optimal use of hormonal therapy in prostate cancer.


Assuntos
Antineoplásicos Hormonais/uso terapêutico , Hormônio Liberador de Gonadotropina/antagonistas & inibidores , Neoplasias da Próstata/terapia , Testosterona/farmacologia , Antineoplásicos Hormonais/farmacologia , Busserrelina/farmacologia , Busserrelina/uso terapêutico , Gosserrelina/farmacologia , Gosserrelina/uso terapêutico , Humanos , Leuprolida/farmacologia , Leuprolida/uso terapêutico , Masculino , Neoplasias da Próstata/fisiopatologia
5.
J Clin Oncol ; 17(1): 82-92, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10458221

RESUMO

PURPOSE: To determine the safety and efficacy of multiple cycles of dose-intensive, nonablative chemotherapy in women with poor-prognosis breast cancer. PATIENTS AND METHODS: Women with stage II breast cancer and 10 or more involved nodes or four or more involved nodes and estrogen receptor-negative tumors and women with stage III disease received three cycles of epirubicin 200 mg/m2 and cyclophosphamide 4 g/m2, with progenitor cell and filgrastim support every 28 days (n = 79) or 21 days (n = 20). Patients were reviewed at least twice yearly thereafter. Twenty-six patients had bone marrow and apheresis collections assessed for the presence of micrometastatic tumor cells. RESULTS: Ninety-nine women (median age, 43 years; range, 24 to 60 years) were treated. Ninety-two completed all three cycles of chemotherapy. The major toxicity was severe, reversible myelosuppression that was more prolonged with successive cycles, and this did not differ between patients given treatment every 28 days and those treated every 21 days. Febrile neutropenia occurred in 176 (61%) of 287 cycles. Severe mucositis (grade 3 or 4) occurred in 23% of cycles but tended to be short-lived and was reversible. The cardiac ejection fraction fell by a median of 4% during treatment, and three patients developed evidence of cardiac failure after chemotherapy. Two patients (2%) died of acute toxicity. Three of 26 patients had evidence of circulating micrometastatic tumor cells. The actuarial distant disease-free and overall survival rates at 60-month follow-up were 64% (95% confidence interval [CI], 53% to 75%) and 67% (95% CI, 56% to 78%), respectively. CONCLUSION: Multiple cycles of dose-intensive, nonablative chemotherapy is a feasible and safe approach. Disease control and survival are similar to those in other studies of myeloablative chemotherapy in poor-prognosis breast cancer. The regimen is being evaluated in a randomized trial of the International Breast Cancer Study Group.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Mobilização de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Hematopoéticas , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Neoplasias da Mama/terapia , Terapia Combinada , Ciclofosfamida/administração & dosagem , Ciclofosfamida/efeitos adversos , Epirubicina/administração & dosagem , Epirubicina/efeitos adversos , Feminino , Filgrastim , Seguimentos , Humanos , Pessoa de Meia-Idade , Células Neoplásicas Circulantes , Prognóstico , Proteínas Recombinantes , Taxa de Sobrevida
6.
Exp Hematol ; 27(1): 117-30, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9923450

RESUMO

A single injection of > or =10 microg/kg PEG-rHuMGDF in mice causes a dose-dependent increase in circulating platelets beginning on day 3 and peaking on days 5-6. The mean platelet volume and platelet distribution width at doses > or =100 microg/kg initially increase in a dose-dependent fashion and later decrease. However, the mean platelet volume does not change when platelets are incubated with PEG-rHuMGDF in vitro. The number of marrow megakaryocytes increases in a dose-dependent fashion as early as day 1 and peaks on day 3. Marrow megakaryocyte colony-forming units (CFU-Meg) do not increase on days 1-3 at a dose of 100 microg/kg (a dose that increases platelet numbers two- to threefold and may be clinically relevant), but the relative frequency of high ploidy megakaryocytes and the proportion of large marrow megakaryocytes (29-50 microm in diameter) increases. After a dose of 1,000 microg/kg the percentage of megakaryocytes in mitosis peaks at 24-48 hours and the percentage of megakaryocytes incorporating BrdU is maximal at 48 hours, the relatively delayed peak of BrdU incorporation most likely representing endomitosis. The relative frequency of type II and III megakaryocytes peaks on days 3 and 4, respectively. Pharmacokinetic analysis of PEG-rHuMGDF shows peak serum concentrations at 2-4 hours and a terminal half-life of 11.4+/-2.5 hours. A single injection of PEG-rHuMGDF ameliorates carboplatin-induced megakaryocytopenia and thrombocytopenia in a dose-response dependent fashion. In conclusion, a single injection of PEG-rHuMGDF increases megakaryocyte and platelet production in normal and myelo-suppressed mice.


Assuntos
Polietilenoglicóis/farmacologia , Polietilenoglicóis/uso terapêutico , Trombocitopenia/fisiopatologia , Trombopoetina/farmacologia , Trombopoetina/uso terapêutico , Acetilcolinesterase/metabolismo , Animais , Plaquetas/citologia , Plaquetas/efeitos dos fármacos , Medula Óssea/química , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/fisiologia , Carboplatina/farmacologia , Contagem de Células/efeitos dos fármacos , Membrana Celular/ultraestrutura , Tamanho Celular/efeitos dos fármacos , Corantes , DNA/análise , DNA/metabolismo , Relação Dose-Resposta a Droga , Fêmur/citologia , Humanos , Injeções , Fígado/citologia , Megacariócitos/citologia , Megacariócitos/efeitos dos fármacos , Megacariócitos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Microscopia Eletrônica , Mitose/efeitos dos fármacos , Contagem de Plaquetas/efeitos dos fármacos , Ploidias , Polietilenoglicóis/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Reticulina/análise , Baço/citologia , Trombocitopenia/tratamento farmacológico , Trombopoetina/metabolismo , Fatores de Tempo
7.
Am J Clin Oncol ; 21(3): 263-9, 1998 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9626795

RESUMO

This clinical trial was designed to explore dose escalation of carboplatin and cyclophosphamide when supported with filgrastim. Twenty-seven patients who had advanced solid tumors received up to six cycles of treatment; a total of 92 cycles of chemotherapy were delivered. Two control groups received standard-dose carboplatin (300 mg/m2) and cyclophosphamide (600 mg/m2), with and without filgrastim. Subsequently, the doses of both carboplatin and cyclophosphamide were increased simultaneously by 50% of the standard dose in sequential cohorts. Doses of up to 2.5 times the standard dose were explored. A final dose of carboplatin, 600 mg/m2, and cyclophosphamide, 1,500 mg/m2, was tested in 4 patients. The duration of neutropenia was brief, even at the highest dose levels. The mean duration of grade 3 or 4 neutropenia was 5.8 days at standard dose without filgrastim and 5.4 days at 2.5 times standard dose with filgrastim. More severe neutropenia was more prolonged at higher doses but remained brief in duration. The mean duration of neutropenia of less than 100 x 10(6)/l was 0.4 days at standard dose without filgrastim and 1.3 days at 2.5 times standard dose. There was no evidence of cumulative neutropenia over repeated cycles of treatment. In contrast, thrombocytopenia was both dose limiting and cumulative. The mean duration of grade 3 or 4 thrombocytopenia was 1.6 days at standard dose and 9.6 days at 2.5 times standard dose. An average of 2.3 platelet transfusions per cycle of treatment was required at the highest dose. Thrombocytopenia was worse with repetitive cycles of therapy. The mean duration of grade 3 or 4 thrombocytopenia was 2.2 days after the first cycle of chemotherapy and 7.8 days after cycle four. The maximum tolerated dose, as defined prospectively, was not reached but further dose escalation was not thought to be warranted because of the severity of thrombocytopenia. When supported with filgrastim, carboplatin and cyclophosphamide can be administered safely with substantially increased dose and acceptable toxicity.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Neoplasias/tratamento farmacológico , Adolescente , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Carboplatina/administração & dosagem , Carboplatina/efeitos adversos , Ciclofosfamida/administração & dosagem , Ciclofosfamida/efeitos adversos , Relação Dose-Resposta a Droga , Feminino , Filgrastim , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos/efeitos adversos , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Recombinantes , Trombocitopenia/induzido quimicamente
8.
J Clin Oncol ; 16(5): 1899-908, 1998 May.
Artigo em Inglês | MEDLINE | ID: mdl-9586908

RESUMO

PURPOSE: To assess the mobilization potential and safety of recombinant human stem-cell factor (SCF) when coadministered with filgrastim to untreated women with poor-prognosis breast cancer. PATIENTS AND METHODS: Eligible women had breast cancer with 10 or more positive axillary nodes, or estrogen receptor-negative tumor with 4 positive nodes, or stage III disease. Patients were randomized to receive SCF plus filgrastim or filgrastim alone. Filgrastim 12 microg/kg daily was administered for 6 days by continuous subcutaneous infusion. SCF was administered by daily subcutaneous injection at 5, 10, or 15 microg/kg concurrent with filgrastim for 7 days, or 10 microg/kg daily starting 3 days before filgrastim for a total of 10 days (SCF pretreatment). Apheresis was performed on days 5, 6, and 7 of filgrastim administration. Patients then had three cycles of epirubicin 200 mg/m2 and cyclophosphamide 4 g/m2 every 28 days, each supported by one third of the apheresis product. RESULTS: Sixty-two women were treated. Greater yields occurred in patients who received SCF 10 microg/kg daily plus filgastim than those who received filgrastim alone (P=.013 for CD34+ cells; P=.07 for granulocyte-macrophage colony-forming cells [GM-CFCs]). The difference was more marked with SCF-pretreatment than concurrent SCF. Fewer aphereses were required to reach the predetermined target of peripheral-blood progenitor/stem cells (PBPCs) in women who received SCF. SCF was generally well tolerated. Hematologic recovery was rapid after each of the three cycles of chemotherapy. There was no difference in recovery between the different treatment groups. CONCLUSION: Mobilization of PBPCs by filgrastim is significantly enhanced by coadministration of SCF, and commencing SCF before filgrastim can optimize this effect. SCF has the potential to reduce the number of aphereses required to collect a target number of PBPCs.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Neoplasias da Mama/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Hematopoese , Mobilização de Células-Tronco Hematopoéticas , Fator de Células-Tronco/administração & dosagem , Adulto , Antígenos CD34/análise , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Contagem de Células Sanguíneas , Remoção de Componentes Sanguíneos , Neoplasias da Mama/sangue , Ensaio de Unidades Formadoras de Colônias , Ciclofosfamida/administração & dosagem , Ciclofosfamida/efeitos adversos , Epirubicina/administração & dosagem , Epirubicina/efeitos adversos , Células Precursoras Eritroides , Feminino , Filgrastim , Hematopoese/efeitos dos fármacos , Mobilização de Células-Tronco Hematopoéticas/métodos , Hemoglobinas/análise , Humanos , Subpopulações de Linfócitos , Pessoa de Meia-Idade , Proteínas Recombinantes/administração & dosagem , Fator de Células-Tronco/efeitos adversos
9.
Blood ; 90(9): 3378-89, 1997 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9345020

RESUMO

Administration of hematopoietic growth factors is being used increasingly to obtain populations of blood progenitor/stem cells (PBPC) for clinical transplantation. Here we examined the effect of combining stem cell factor (SCF ) and granulocyte colony-stimulating factor (G-CSF ) versus G-CSF alone in a randomized clinical study involving 62 women with early-stage breast cancer. In the first patient cohorts, escalating doses of SCF were administered for 7 days with concurrent G-CSF administration. At baseline, levels of progenitor cells in the bone marrow or blood were comparable in the different patient groups. As with administration of G-CSF alone, the combination of SCF plus G-CSF did not alter the wide variation in levels of PBPC observed between individuals and did not alter the selective nature of PBPC release, with preferential release of day-14 granulocyte-macrophage colony-stimulating factor (GM-CFC) versus day-7 GM-CFC. However, SCF acted to sustain the levels of PBPC after cessation of growth factor treatment; levels of PBPC were elevated 100-fold at later timepoints compared with G-CSF alone. In addition, the maximum levels of PBPC observed were increased approximately fivefold at day 5 of growth-factor administration. The increased levels of PBPC resulted in significantly increased levels of PBPC obtained by leukapheresis. In a subsequent patient cohort, 3-days pretreatment with SCF was introduced and followed by 7 days concurrent SCF plus G-CSF. The 3-days pretreatment with SCF resulted in an earlier wave of PBPC release in response to commencement of G-CSF. In addition, maximum PBPC levels in blood and PBPC yield in leukapheresis products were further increased. Unexpectedly however, SCF pretreatment resulted in progenitor cells with enhanced self-generation potential. Recloning assays documented the ability of approximately 30% of primary granulocyte-macrophage (GM) colonies from control cell populations to generate secondary GM colonies (n = 1,106 primary colonies examined). In contrast approximately 90% of GM colonies from PBPC after SCF pretreatment generated secondary clones and 65% generated secondary colonies. The action of SCF was not explicable in terms of altered SCF, GM-CSF, or G-CSF responsiveness, but SCF pretreatment was associated with maximum serum SCF levels at the time G-CSF was commenced. These results show that PBPC populations mobilized by different growth factor regimens can differ in their functional properties and caution against solely considering number of harvested progenitor cells without regard to their function.


Assuntos
Neoplasias da Mama/terapia , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Mobilização de Células-Tronco Hematopoéticas , Transplante de Células-Tronco Hematopoéticas , Fator de Células-Tronco/administração & dosagem , Administração Cutânea , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Feminino , Humanos , Transplante Autólogo
10.
Blood ; 90(8): 2939-51, 1997 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-9376574

RESUMO

The safety and optimal dose and schedule of stem cell factor (SCF) administered in combination with filgrastim for the mobilization of peripheral blood progenitor cells (PBPCs) was determined in 215 patients with high-risk breast cancer. Patients received either filgrastim alone (10 microg/kg/d for 7 days) or the combination of 10 microg/kg/d filgrastim and 5 to 30 microg/kg/d SCF for either 7, 10, or 13 days. SCF patients were premedicated with antiallergy prophylaxis. Leukapheresis was performed on the final 3 days of cytokine therapy and, after high-dose chemotherapy and infusion of PBPCs, patients received 10 microg/kg/d filgrastim until absolute neutrophil count recovery. The median number of CD34+ cells collected was greater for patients receiving the combination of filgrastim and SCF, at doses greater than 10 microg/kg/d, than for those receiving filgrastim alone (7.7 v 3.2 x 10(6)/kg, P < .05). There were significantly (P < .05) more CD34+ cells harvested for the 20 microg/kg/d SCF (median, 7.9 x 10(6)/kg) and 25 microg/kg/d SCF (median, 13.6 x 10(6)/kg) 7-day combination groups than for the filgrastim alone patients (median, 3.2 x 10(6)/kg). The duration of administration of SCF and filgrastim (7, 10, or 13 days) did not significantly affect CD34+ cell yield. Treatment groups mobilized with filgrastim alone or with the cytokine combination had similar hematopoietic engraftment and overall survival after PBPC infusion. In conclusion, the results of this study indicate that SCF therapy enhances CD34+ cell yield and is associated with manageable levels of toxicity when combined with filgrastim for PBPC mobilization. The combination of 20 microg/kg/d SCF and 10 microg/kg/d filgrastim with daily apheresis beginning on day 5 was selected as the optimal dose and schedule for the mobilization of PBPCs.


Assuntos
Neoplasias da Mama Masculina/tratamento farmacológico , Neoplasias da Mama/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Fator de Células-Tronco/uso terapêutico , Adolescente , Adulto , Idoso , Antígenos CD34/análise , Coleta de Amostras Sanguíneas , Neoplasias da Mama/sangue , Neoplasias da Mama Masculina/sangue , Esquema de Medicação , Quimioterapia Combinada , Feminino , Filgrastim , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Humanos , Leucaférese , Masculino , Pessoa de Meia-Idade , Transfusão de Plaquetas , Proteínas Recombinantes , Fator de Células-Tronco/administração & dosagem
11.
Curr Opin Hematol ; 4(5): 312-6, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9288463

RESUMO

Mpl ligand is the hematopoietic growth factor responsible for regulating the production of platelets. Since its discovery just 3 years ago, it has provided both unique insights into megakaryocytopoiesis and the means to stimulate platelet production in numerous clinical situations. In animals deficient in Mpl ligand, the number of megakaryocytes and platelets decreases by more than 80%. In addition to having an effect on the megakaryocyte lineage, Mpl ligand supports the growth of stem cells, multipotential cells, and erythroid precursors. The endogenous Mpl ligand, thrombopoietin, is produced in the liver and kidneys, and circulating levels appear to be regulated primarily by the mass of platelets and megakaryocytes, which bind and catabolize Mpl ligand. The clinical utility of recombinant Mpl ligands is currently under intense investigation. Early results have demonstrated a marked stimulation of megakaryocyte and platelet production with no apparent adverse effects. In patients with cancer who are treated with chemotherapy, the duration of thrombocytopenia was shorter in patients treated with Mpl ligand than in those receiving placebo. The eventual role of Mpl ligand in clinical practice will be determined by the results of many ongoing clinical studies.


Assuntos
Antineoplásicos/uso terapêutico , Hematopoese , Megacariócitos/citologia , Neoplasias/tratamento farmacológico , Trombopoetina/uso terapêutico , Ensaios Clínicos Fase I como Assunto , Expressão Gênica , Humanos , Modelos Biológicos , Polietilenoglicóis , Trombopoetina/administração & dosagem , Trombopoetina/genética
12.
Br J Haematol ; 97(4): 871-80, 1997 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-9217191

RESUMO

The most important physiological regulator of megakaryocytopoiesis is the ligand for the c-mpl receptor (thrombopoietin/megakaryocyte growth and development factor, MGDF). We examined the effect of pegylated-recombinant human MGDF (PEG-rHuMGDF): patients received PEG-rHuMGDF at doses of 0.03, 0.1, 0.3 or 1.0 microg/kg/d or placebo for 10d maximum in a double-blinded randomized study. There was a dose-dependent elevation in circulating platelet counts but no alteration in erythrocyte or total leucocyte counts. The number of bone marrow megakaryocytes was increased approximately 2-fold. The frequency of bone marrow progenitor cells was not altered. In contrast, both to the bone marrow results and to published pre-clinical data, there was a dose-dependent mobilization into the blood of progenitor cells of multiple cell lineages. Increased levels of Meg-CFC (maximum increase 30-fold), day 7 and day 14 GM-CFC and BFU-E were demonstrated at doses of 0.3 and 1.0 microg/kg/d PEG-rHuMGDF. At 0.1 microg/kg/d, mobilization of Meg-CFC alone occurred in two-thirds of patients. Maximum blood levels of progenitor cells occurred at day 12. Thus, administration of PEG-rHuMGDF to humans resulted in mobilization of progenitor cells of multiple lineages despite its 'lineage-specific' activity on mature cell development.


Assuntos
Células-Tronco Hematopoéticas/efeitos dos fármacos , Megacariócitos/efeitos dos fármacos , Proteínas de Neoplasias , Proteínas Proto-Oncogênicas/uso terapêutico , Receptores de Citocinas , Adulto , Idoso , Plaquetas/patologia , Linhagem da Célula , Tamanho Celular , Humanos , Megacariócitos/patologia , Pessoa de Meia-Idade , Neoplasias/terapia , Contagem de Plaquetas , Receptores de Trombopoetina
14.
Blood ; 87(12): 5006-15, 1996 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-8652813

RESUMO

PEG-rHuMGDF injected daily in normal mice causes a rapid dose-dependent increase in megakaryocytes and platelets. At the same time that platelet numbers are increased, the mean platelet volume (MPV) and platelet distribution width (PDW) can be either decreased, normal, or increased depending on the dose and time after administration. Thus, PEG-rHuMGDF at a low dose causes decreases in MPV and PDW, MGDF at an intermediate dose causes an initial increase followed by a decrease in MPV and PDW, and PEG-rHuMGDF at higher doses causes an increase in MPV and PDW followed by a gradual normalization of these platelet indices. In addition to the expected thrombocytosis after 7 to 10 days of daily injection of high doses of PEG-rHuMGDF, a transient decrease in peripheral red blood cell numbers and hemoglobin is noted accompanied in the bone marrow by megakaryocytic hyperplasia, myeloid hyperplasia, erythroid and lymphoid hypoplasia, and deposition of a fine network of reticulin fibers. Splenomegaly, an increase in splenic megakaryocytes, and extramedullary hematopoiesis accompany the hematologic changes in the peripheral blood and marrow to complete a spectrum of pathologic features similar to those reported in patients with myelofibrosis and megakaryocyte hyperplasia. However, all the PEG-rHuMGDF-initiated hematopathology including the increase in marrow reticulin is completely and rapidly reversible upon the cessation of administration of PEG-rHuMGDF. Thus, transient hyperplastic proliferation of megakaryocytes does not cause irreversible tissue injury. Furthermore, PEG-rHuMGDF completely ameliorates carboplatin-induced thrombocytopenia at a low-dose that does not cause the hematopathology associated with myelofibrosis.


Assuntos
Anemia/induzido quimicamente , Megacariócitos/efeitos dos fármacos , Mielofibrose Primária/induzido quimicamente , Esplenomegalia/induzido quimicamente , Trombocitose/induzido quimicamente , Trombopoetina/toxicidade , Anemia/patologia , Animais , Medula Óssea/química , Medula Óssea/patologia , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Humanos , Hiperplasia , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos BALB C , Polietilenoglicóis/administração & dosagem , Mielofibrose Primária/patologia , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/toxicidade , Proteínas Recombinantes , Reticulina/análise , Esplenomegalia/patologia , Trombocitose/patologia , Trombopoetina/administração & dosagem , Trombopoetina/genética
15.
J Clin Invest ; 95(6): 2973-8, 1995 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-7539462

RESUMO

The present study shows that recombinant human megakaryocyte growth and development factor (r-HuMGDF) behaves both as a megakaryocyte colony stimulating factor and as a differentiation factor in human progenitor cell cultures. Megakaryocyte colony formation induced with r-HuMGDF is synergistically affected by stem cell factor but not by interleukin 3. Megakaryocytes stimulated with r-HuMGDF demonstrate progressive cytoplasmic and nuclear maturation. Measurable levels of megakaryocyte growth and development factor in serum from patients undergoing myeloablative therapy and transplantation are shown to be elaborated in response to thrombocytopenic stress. These data support the concept that megakaryocyte growth and development factor is a physiologically regulated cytokine that is capable of supporting several aspects of megakaryopoiesis.


Assuntos
Hematopoese/efeitos dos fármacos , Megacariócitos/citologia , Trombocitopenia/sangue , Trombopoetina/farmacologia , Adulto , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Humanos , Técnicas In Vitro , Interleucina-3/farmacologia , Proteínas Recombinantes , Fator de Células-Tronco
16.
Ann Intern Med ; 121(7): 492-501, 1994 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-7520676

RESUMO

OBJECTIVE: To determine if filgrastim (recombinant human methionyl granulocyte colony-stimulating factor) used in addition to standard inpatient antibiotic therapy accelerated recovery from infection associated with chemotherapy-induced neutropenia. DESIGN: Randomized, double-blind, placebo-controlled trial. SETTING: Hematology and oncology wards of four teaching hospitals. PATIENTS: 218 patients with cancer who had fever (temperature > 38.2 degrees C) and neutropenia (neutrophil count < 1.0 x 10(9)/L) after chemotherapy. INTERVENTION: Patients were randomly assigned to receive filgrastim (12 micrograms/kg of body weight per day) (n = 109) or placebo (n = 107) beginning within 12 hours of empiric therapy with tobramycin and piperacillin. Patients received treatment and remained in the study until the neutrophil count was greater than 0.5 x 10(9)/L and until 4 days without fever (temperature < 37.5 degrees C) had elapsed. MEASUREMENTS: Days of neutropenia and fever and days in the study (hospitalization); time to resolution of fever and febrile neutropenia; and frequency of the use of alternative antibiotics. RESULTS: Compared with placebo, filgrastim reduced the median number of days of neutropenia (3.0 compared with 4.0 days of a neutrophil count of < 0.5 x 10(9)/L; P = 0.005) and the time to resolution of febrile neutropenia (5.0 compared with 6.0 days; P = 0.01) but not days of fever (3.0 days for both groups). The frequency of the use of alternative antibiotics was similar in the two groups (46% compared with 41%; P = 0.48). The median number of days patients were hospitalized while on study was the same (8.0 days; P = 0.09); however, filgrastim decreased the risk for prolonged hospitalization (> 11 days, 4th quartile) by half (relative risk, 2.1 [95% CI, 1.1 to 4.1]; P = 0.02). In exploratory subset analyses, filgrastim appeared to provide the greatest benefit in patients with documented infection and in patients presenting with neutrophil counts of less than 0.1 x 10(9)/L. CONCLUSIONS: Filgrastim treatment used with antibiotics at the onset of febrile neutropenia in patients with cancer who have received chemotherapy accelerated neutrophil recovery and shortened the duration of febrile neutropenia.


Assuntos
Antineoplásicos/efeitos adversos , Quimioterapia Combinada/uso terapêutico , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Infecções/tratamento farmacológico , Neutropenia/tratamento farmacológico , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Método Duplo-Cego , Feminino , Filgrastim , Humanos , Infecções/etiologia , Tempo de Internação , Contagem de Leucócitos , Masculino , Pessoa de Meia-Idade , Neutropenia/induzido quimicamente , Neutrófilos , Piperacilina/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Tobramicina/uso terapêutico
17.
Leuk Lymphoma ; 14(3-4): 279-83, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7524888

RESUMO

We retrospectively reviewed the regimen-related toxicity associated with busulphan (1 mg/kg orally QID days -7 to -4) and cyclophosphamide (60 mg/kg IV days -3 and -2) (Bu/Cy) chemotherapy in 69 consecutive patients who underwent autologous bone marrow transplantation (ABMT). Twenty-four patients received bone marrow (BM) alone, 22 received BM plus post-transplant granulocyte-colony stimulating factor (G-CSF) and 23 received peripheral blood progenitor cells (PBPC) +/- BM plus post-transplant G-CSF. Toxicity was scored using the criteria of Bearman. Grade II and III toxicities included mucosa (38%), liver (8%), central nervous system (5%), kidney (5%), heart (3%), pericardium (2%), bladder (2%) and lung (2%). There were five treatment related deaths (7%) from pneumonitis (2) and veno-occlusive disease, pulmonary hemorrhage and sepsis (1 each). Post-transplant G-CSF (+/- PBPC) resulted in a trend (p = 0.07) towards a reduction in post-transplant stomatitis, but did not impact on the already low incidence of other organ toxicities. As Bu/Cy for ABMT is associated with minimal non-hemopoietic toxicity, the addition of other cytotoxic agents is justified in an attempt to augment the anti-tumour effect of this conditioning regimen.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Transplante de Medula Óssea , Doença de Hodgkin/tratamento farmacológico , Doença de Hodgkin/terapia , Leucemia Mieloide/tratamento farmacológico , Leucemia Mieloide/terapia , Linfoma não Hodgkin/tratamento farmacológico , Linfoma não Hodgkin/terapia , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Doença Aguda , Administração Oral , Adolescente , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Bussulfano/administração & dosagem , Bussulfano/efeitos adversos , Terapia Combinada , Ciclofosfamida/administração & dosagem , Ciclofosfamida/efeitos adversos , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Fator Estimulador de Colônias de Granulócitos/efeitos adversos , Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Transplante de Células-Tronco Hematopoéticas , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos , Estomatite/induzido quimicamente , Análise de Sobrevida
18.
Bone Marrow Transplant ; 14(1): 105-11, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7524904

RESUMO

The hemopoietic growth factor filgrastim (r-metHu G-CSF) stimulates granulopoiesis after autologous BMT and can also be used as a peripheral blood progenitor cell (PBPC)-mobilizing agent. Rapid platelet recovery follows the addition of filgrastim-mobilized PBPC to autologous BMT. We have now studied 29 adults with malignant lymphoma, Hodgkin's disease or ALL to assess the ability of filgrastim-mobilized PBPC to rapidly and durably restore hemopoiesis without bone marrow (BM) infusion. Patients with a high yield of PBPC from three leukaphereses, defined as > 30 x 10(4)/kg GM-CFC, were eligible for PBPC transplant without BM. Patients with a low yield of GM-CFC received both PBPC and BM infusion. After filgrastim therapy 12 or 24 micrograms/kg/day by continuous sc infusion for 6 or 7 days, a high yield was obtained in 11 of 29 patients. Kinetics of recovery of both the platelet and neutrophil counts were more rapid in the high yield group than in the low yield group. The platelet count recovered to > 20 x 10(9)/l at a median of 9 days, to > 50 x 10(9)/l at 11 days and the neutrophil count to > 0.5 x 10(9)/l at 9 days in the high yield group compared with 12 days, 37 days and 10 days, respectively, in the low yield group (p = 0.028, p < 0.001 and p = 0.027). Fewer platelet transfusions were required in the high yield group (median 11 vs 29.5 units, p = 0.021).(ABSTRACT TRUNCATED AT 250 WORDS)


Assuntos
Fator Estimulador de Colônias de Granulócitos/uso terapêutico , Transplante de Células-Tronco Hematopoéticas/métodos , Adolescente , Adulto , Antineoplásicos/administração & dosagem , Feminino , Filgrastim , Hematopoese , Células-Tronco Hematopoéticas/efeitos dos fármacos , Doença de Hodgkin/sangue , Doença de Hodgkin/terapia , Humanos , Leucemia Mieloide Aguda/sangue , Leucemia Mieloide Aguda/terapia , Contagem de Leucócitos , Linfoma não Hodgkin/sangue , Linfoma não Hodgkin/terapia , Masculino , Pessoa de Meia-Idade , Contagem de Plaquetas , Proteínas Recombinantes/uso terapêutico , Transplante Autólogo
20.
J Hematother ; 3(4): 349-52, 1994.
Artigo em Inglês | MEDLINE | ID: mdl-7537610

RESUMO

Autologous bone marrow transplantation, although successful, is limited in the rate of hematopoietic recovery achieved. Myeloablative chemotherapy results in prolonged pancytopenia with standard autologous bone marrow support. High-dose myelosuppressive chemotherapy results in severe but short-term pancytopenia, and hematopoietic recovery is routine without cellular support. Cellular support becomes necessary when multiple-cycle, severely myelosuppressive regimens are used because of the cumulative stem cell damage and unacceptably long duration of severe pancytopenia. Peripheral blood progenitor cells (PBPCs) may be used as cellular support, and colony-stimulating factors with or without chemotherapy are currently the most potent available PBPC-mobilizing tools. Filgrastim (rmethG-CSF) has been shown to enhance the number of PBPCs for harvest in both cancer patients and normal donors. When Filgrastim is used in conjunction with chemotherapy, there appears to be greater PBPC mobilization, which seems to be dependent on dose and schedule of chemotherapy as well as the type of chemotherapeutic agent used. Platelet recovery has been shown to be more rapid when PBPCs are used compared with historical controls given autologous bone marrow infusions. It may be concluded that PBPC transplantation is useful supportive care following myeloablative chemotherapy.


Assuntos
Fator Estimulador de Colônias de Granulócitos/farmacologia , Transplante de Células-Tronco Hematopoéticas , Transplante de Medula Óssea , Filgrastim , Humanos , Contagem de Plaquetas/efeitos dos fármacos , Proteínas Recombinantes/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...