Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.466
Filtrar
1.
Science ; 232(4746): 58-60, 1986 Apr 04.
Artigo em Inglês | MEDLINE | ID: mdl-3082008

RESUMO

The pathophysiology of anaphylaxis is very complex, and the sequelae of events are not fully explained in terms of the effects of histamine and peptide leukotrienes alone. Platelet-activating factor (1-O-alkyl-2-acetyl-sn-glyceryl-3-phosphorylcholine, PAF-acether) has been detected in animals undergoing anaphylaxis. Injection of synthetic PAF-acether induces similar effects, including bronchoconstriction, respiratory arrest, systemic hypotension, neutropenia, and thrombocytopenia. The results reported here demonstrate that the histamine- and leukotriene-independent component of guinea pig anaphylaxis in vivo and in isolated lung parenchymal strips in vitro is mediated by PAF-acether. However, PAF-acether is not responsible for the anaphylaxis-induced thrombocytopenia.


Assuntos
Anafilaxia , Pulmão/imunologia , Fator de Ativação de Plaquetas/imunologia , 4,5-Di-Hidro-1-(3-(Trifluormetil)Fenil)-1H-Pirazol-3-Amina , Alprazolam , Animais , Anti-Inflamatórios/farmacologia , Benzodiazepinas/farmacologia , Pressão Sanguínea , Difenidramina/farmacologia , Cobaias , Técnicas In Vitro , Cinética , Pulmão/efeitos dos fármacos , Masculino , Ovalbumina , Contagem de Plaquetas/efeitos dos fármacos , Pirazóis/farmacologia
2.
J Clin Invest ; 96(2): 1125-30, 1995 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-7635948

RESUMO

HST-1 (FGF-4) gene product is a member of the fibroblast growth factor family with a signal peptide and plays a crucial role in limb development. We showed previously that an intraperitoneal injection of replication-deficient adenovirus containing the HST-1 gene (Adex1HST-1) into normal mice caused a twofold increase in peripheral platelet count. To investigate whether Adex1HST-1 could effectively prevent experimentally induced thrombocytopenia in mice, we injected Adex1HST-1 intraperitoneally into thrombocytopenic mice induced by administration of a chemotherapeutic agent and/or by irradiation. A single Adex1HST-1 injection caused continuously increased levels of serum HST-1 protein for at least 30 d and increased the count of large megakaryocytes in bone marrow, which specifically recovered platelet counts and more efficiently diminished the extent and duration of thrombocytopenia than any other reported cytokine or any combination of cytokines so far. In the other peripheral hematological parameters, no discernible differences were detected. No other apparent side effects were observed. Therefore, this method could be useful for treatment and/or prevention of thrombocytopenia induced by chemotherapy and/or irradiation for cancer treatment.


Assuntos
Adenovírus Humanos/genética , Cisplatino/toxicidade , Fatores de Crescimento de Fibroblastos/genética , Terapia Genética , Vetores Genéticos , Proteínas Proto-Oncogênicas/genética , Lesões Experimentais por Radiação/terapia , Trombocitopenia/prevenção & controle , Animais , Feminino , Fator 4 de Crescimento de Fibroblastos , Humanos , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos ICR , Agregação Plaquetária , Contagem de Plaquetas/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , Trombocitopenia/induzido quimicamente , Trombocitopenia/etiologia
3.
J Clin Invest ; 89(5): 1669-73, 1992 May.
Artigo em Inglês | MEDLINE | ID: mdl-1314853

RESUMO

Interleukin-2 was recently shown to cause acute lung injury characterized by microvascular permeability defect, interstitial edema, and leukosequestration. Similar responses can also be produced by platelet activating factor (PAF). Thus, the present study aimed to examine whether PAF plays a key role in the development of IL-2-induced lung injury in the anesthetized rat. Intravenous infusion (60 min) of recombinant human IL-2 at 10(5)-10(6) U/rat (n = 7-9) dose-dependently elevated lung water content (27 +/- 1%, P less than 0.01), myeloperoxidase activity (+84 +/- 23%, P less than 0.05), and serum thromboxane B2 (990 +/- 70%, P less than 0.01), but failed to alter blood pressure, hematocrit, serum tumor necrosis factor-alpha, and circulating leukocytes and platelets. Pretreatment (-30 min) with a potent and specific PAF antagonist, BN 50739 (10 mg/kg, intraperitoneally, n = 6) prevented the pulmonary edema (P less than 0.05) and thromboxane B2 production (P less than 0.01), and attenuated the elevation of lung myeloperoxidase activity (+18 +/- 16%, P less than 0.05) induced by IL-2. These data suggest that PAF is involved in the pathophysiological processes leading to IL-2-induced lung injury, and point to the potential therapeutic capacity of PAF antagonists in preventing pulmonary edema during IL-2 therapy.


Assuntos
Azepinas/farmacologia , Interleucina-2/toxicidade , Pneumopatias/induzido quimicamente , Fator de Ativação de Plaquetas/fisiologia , Triazóis/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Contagem de Leucócitos , Masculino , Peroxidase/metabolismo , Contagem de Plaquetas/efeitos dos fármacos , Ratos , Ratos Endogâmicos , Tromboxano B2/sangue , Fator de Necrose Tumoral alfa/metabolismo
4.
J Clin Invest ; 81(4): 1162-72, 1988 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-3258319

RESUMO

In addition to activating T and B lymphocytes, interleukin 1 (IL-1) induces several hematologic and metabolic changes typical of host responses to infection and injury. We now report a new biological property, namely, the induction of hypotension. Rabbits given a single intravenous injection of recombinant human IL-1-beta (5 micrograms/kg) rapidly developed decreased systemic arterial pressure, which reached the lowest levels after 50-60 min and slowly returned to pre-IL-1 values after 3 h. Associated with the hypotension, systemic vascular resistance and central venous pressure fell, while cardiac output and heart rate increased. These responses were prevented by ibuprofen given 15 min before the IL-1. A bolus injection of IL-1 followed by a 2-h infusion sustained the hypotension and was associated with leukopenia and thrombocytopenia. Ibuprofen given at the mid-point of the infusion reversed the changes in all hemodynamic parameters, but had no effect on the leukopenia or thrombocytopenia. Tumor necrosis factor (TNF) also induced a shock-like state in rabbits. When the dose of IL-1 or TNF was reduced to 1 microgram/kg, no hemodynamic changes were observed; however, the combination of these low doses of both cytokines resulted in a profound shock-like state including histological evidence of severe pulmonary edema and hemorrhage. Pretreatment with ibuprofen prevented the hemodynamic, leukocyte, and platelet changes induced by the low-dose cytokine combination, and ameliorated the pulmonary tissue damage. These results demonstrate that IL-1, like TNF, possesses the ability to induce hemodynamic and hematological changes typical of septic shock, and that the combination of IL-1 and TNF is more potent than either agent alone. These effects seem to require cyclooxygenase products, and suggest that intravenous cyclooxygenase inhibitors may be of therapeutic value in patients with IL-1/TNF-mediated shock.


Assuntos
Ibuprofeno/farmacologia , Interleucina-1/farmacologia , Choque/induzido quimicamente , Fator de Necrose Tumoral alfa/farmacologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Sinergismo Farmacológico , Hemodinâmica/efeitos dos fármacos , Interleucina-1/antagonistas & inibidores , Contagem de Leucócitos/efeitos dos fármacos , Pulmão/patologia , Contagem de Plaquetas/efeitos dos fármacos , Coelhos , Choque/patologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores
5.
J Clin Invest ; 105(2): 173-81, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10642595

RESUMO

Bone marrow transplantation (BMT) has considerable potential for the treatment of malignancies, hemoglobinopathies, and autoimmune diseases, as well as the induction of transplantation allograft tolerance. Toxicities associated with standard preparative regimens for bone marrow transplantation, however, make this approach unacceptable for all but the most severe of these clinical situations. Here, we demonstrate that stable mixed hematopoietic cell chimerism and donor-specific tolerance can be established in miniature swine, using a relatively mild, non-myeloablative preparative regimen. We conditioned recipient swine with whole-body and thymic irradiation, and we depleted their T-cells by CD3 immunotoxin-treatment. Infusion of either bone marrow cells or cytokine-mobilized peripheral blood stem cells from leukocyte antigen-matched animals resulted in stable mixed chimerism, as detected by flow cytometry in the peripheral blood, thymus, and bone marrow, without any clinical evidence of graft-versus-host disease (GvHD). Long-term acceptance of donor skin and consistent rejection of third-party skin indicated that the recipients had developed donor-specific tolerance.


Assuntos
Transplante de Medula Óssea/imunologia , Quimera/imunologia , Transplante de Células-Tronco Hematopoéticas , Tolerância Imunológica/imunologia , Condicionamento Pré-Transplante/métodos , Animais , Complexo CD3/imunologia , Linhagem da Célula/imunologia , Esquema de Medicação , Citometria de Fluxo , Sobrevivência de Enxerto/efeitos dos fármacos , Sobrevivência de Enxerto/efeitos da radiação , Imunotoxinas/administração & dosagem , Imunotoxinas/efeitos adversos , Contagem de Leucócitos/efeitos dos fármacos , Contagem de Leucócitos/efeitos da radiação , Depleção Linfocítica , Contagem de Plaquetas/efeitos dos fármacos , Contagem de Plaquetas/efeitos da radiação , Transplante de Pele/imunologia , Suínos , Porco Miniatura , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Timo/citologia , Timo/imunologia , Timo/efeitos da radiação , Irradiação Corporal Total
6.
J Natl Cancer Inst ; 81(15): 1172-4, 1989 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-2746669

RESUMO

Twenty-two patients with hairy cell leukemia were treated with low-dose interferon alfa-2b (0.2 X 10(6) U/m2 given three times weekly) for 6-12 months. The overall response rate was 54%, with only 18% complete plus partial responses. The therapy had to be terminated early in five of these patients because their progressive disease led to severe cytopenia. Although the toxic effects with this regimen were minimal, the significantly lower response rate and the poorer quality of the responses prohibit its use as initial therapy in hairy cell leukemia.


Assuntos
Interferon Tipo I/administração & dosagem , Interferon-alfa/administração & dosagem , Leucemia de Células Pilosas/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Medula Óssea/patologia , Esquema de Medicação , Avaliação de Medicamentos , Feminino , Seguimentos , Hemoglobinas/análise , Humanos , Interferon alfa-2 , Interferon-alfa/efeitos adversos , Leucemia de Células Pilosas/sangue , Contagem de Leucócitos/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Contagem de Plaquetas/efeitos dos fármacos , Proteínas Recombinantes , Indução de Remissão
7.
J Natl Cancer Inst ; 86(1): 39-45, 1994 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-8271281

RESUMO

BACKGROUND: Macrophage colony-stimulating factor is a bone marrow-derived glycoprotein that can stimulate monocytes and macrophages, resulting in production of factors involved in immune response. In vitro and in vivo preclinical studies in animals have demonstrated that recombinant human macrophage colony-stimulating factor (rHuM-CSF) can have antitumor activity. PURPOSE: A phase I clinical trial was undertaken to evaluate the toxicity, pharmacokinetics, and immunologic effects of rHuM-CSF given by continuous intravenous infusion in patients with cancer. METHODS: Eighteen patients with metastatic solid tumors refractory to conventional therapy were treated with rHuM-CSF. Twelve patients received two 14-day cycles of rHuM-CSF by continuous infusion, with a 2-week interval. Dose escalation levels were 50, 100, and 150 micrograms/kg over 24 hours. Consecutive cohorts of three to six patients were planned at each dose level. Six patients received a modified regimen of four 7-day periods of infusion at 100 micrograms/kg over 24 hours, with 1-week intervals. RESULTS: Dose-limiting toxicity was grade 4 thrombocytopenia at a dose of 150 micrograms/kg over 24 hours in two patients receiving the 2-week regimen. Platelet count nadirs and concomitant monocytosis were seen on days 7-9, but recovery occurred during the treatment period. Macrophage colony-stimulating factor serum levels were maximal on day 1 and returned to near baseline on day 7 of infusion. Patients treated with four 7-day infusions had no treatment-limiting thrombocytopenia. There were no cumulative effects on platelet or monocyte counts or significant constitutional symptoms. Subclinical conjunctival injection was noted in five of 10 patients receiving screening ophthalmologic evaluation. Grade 2 episcleritis was diagnosed in one patient, and asymptomatic perilimbal and retinal hemorrhages were seen in two. Two patients developed sepsis caused by the intravenous line, which required cessation of therapy. No objective responses were documented. CONCLUSION: The maximum tolerated dose of rHuM-CSF given by continuous intravenous infusion for 14 days was 100 micrograms/kg over 24 hours, with rapidly reversible, dose-limiting thrombocytopenia at 150 micrograms/kg over 24 hours. A regimen alternating weekly cycles of infusion avoids dose-limiting toxicity and allows long-term treatment. IMPLICATIONS: The regimen of repeated 7-day infusions may be useful for future studies evaluating rHuM-CSF-activated monocytes in therapy for long-term infectious diseases or in investigation of new modes of cancer therapy using rHuM-CSF in conjunction with a tumor-specific antibody.


Assuntos
Fator Estimulador de Colônias de Macrófagos/uso terapêutico , Neoplasias/tratamento farmacológico , Adulto , Idoso , Feminino , Humanos , Infusões Intravenosas , Contagem de Leucócitos/efeitos dos fármacos , Fator Estimulador de Colônias de Macrófagos/administração & dosagem , Fator Estimulador de Colônias de Macrófagos/efeitos adversos , Masculino , Pessoa de Meia-Idade , Monócitos/efeitos dos fármacos , Metástase Neoplásica , Contagem de Plaquetas/efeitos dos fármacos , Proteínas Recombinantes/uso terapêutico , Resultado do Tratamento
8.
J Natl Cancer Inst ; 83(23): 1748-53, 1991 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-1770554

RESUMO

Recombinant human granulocyte-macrophage colony-stimulating factor (rHuGM-CSF) may reduce myelosuppression and, thus, allow dose escalation of certain chemotherapeutic agents. We conducted two sequential phase I trials of escalating doses of carboplatin and a fixed dose and schedule of rHuGM-CSF in ovarian cancer patients who had not previously had chemotherapy, i.e., chemotherapy-naive patients. In the first trial, patients were assigned to regimens of increasing dose levels of carboplatin (starting at 400 mg/m2) and fixed doses and schedules of cyclophosphamide (600 mg/m2) and rHuGM-CSF (10 micrograms/kg given subcutaneously once daily on days 2-11). Chemotherapy was given every 3 weeks (regimen A). In the subsequent trial, the design was the same except that cyclophosphamide was omitted (regimen B). Fifteen patients received regimen A, and seven patients received regimen B. In regimen A, all three patients treated at the first dose level tolerated five cycles at full doses. Hematologic toxicity was dose limiting at the 600-mg/m2 dose level. When 500 mg/m2 carboplatin was given, six of eight patients tolerated three or four cycles at full doses before requiring dose reductions or treatment delays. In regimen B, doses could not be escalated above the first dose level (600 mg/m2) because of severe hematological toxicity. Nonhematological toxicity was tolerable and managed with acetaminophen, antihistamines, and/or nonsteroidal, anti-inflammatory medication. Compliance was excellent. We conclude that (a) rHuGM-CSF can be given safely and reliably to chemotherapy-naive ovarian cancer patients receiving these treatment regimens, (b) early and severe thrombocytopenia was a major problem with or without cyclophosphamide with doses of carboplatin at or above 600 mg/m2, and (c) 500 mg/m2 carboplatin administered every 3 weeks is the highest dose in regimen A that can be given safely in the outpatient setting.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/toxicidade , Carboplatina/toxicidade , Fator Estimulador de Colônias de Granulócitos e Macrófagos/uso terapêutico , Neoplasias Ovarianas/terapia , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Carboplatina/administração & dosagem , Carboplatina/uso terapêutico , Ciclofosfamida/administração & dosagem , Relação Dose-Resposta a Droga , Avaliação de Medicamentos , Feminino , Humanos , Contagem de Leucócitos/efeitos dos fármacos , Estadiamento de Neoplasias , Neutrófilos/efeitos dos fármacos , Neoplasias Ovarianas/patologia , Contagem de Plaquetas/efeitos dos fármacos , Proteínas Recombinantes/uso terapêutico
9.
J Natl Cancer Inst ; 88(19): 1393-8, 1996 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-8827017

RESUMO

BACKGROUND: Increased proliferation of endogenous bone marrow progenitor cells in response to the administration of hematopoietic growth factors, followed by reduced cell cycling or entrance of the cells into a quiescent state upon withdrawal of the growth factors, may have clinically relevant effects on the tolerance of the hematopoietic system to subsequent myelotoxic treatments. PURPOSE: We investigated the ability of granulocyte colony-stimulating factor (G-CSF) to protect progenitor cells in the bone marrow of cancer patients from the toxic effects of subsequent treatments with chemotherapeutic agents. METHODS: Thirty-six patients with histologically documented, locally advanced or metastatic breast cancer were randomly assigned to receive doxorubicin once every 3 weeks at a dose of 75 mg/m2 and cyclophosphamide at a dose of 1000 mg/m2, with G-CSF administered either before and after chemotherapy (18 patients) or after chemotherapy only (18 patients). For prechemotherapy administration of G-CSF, recombinant human methionyl (r-met Hu) G-CSF was administered subcutaneously to patients twice per day for 5 days at a dose of 5 micrograms/kg, with the last dose given 48 hours before the start of chemotherapy. For postchemotherapy administration of G-CSF, r-met Hu G-CSF was administered subcutaneously to patients once per day for 7 days at a dose of 5 micrograms/kg, with the first dose given 24 hours after chemotherapy. RESULTS: The incidence or the duration of grade 4 neutropenia was not reduced in all patients by the use of prechemotherapy G-CSF; the incidence over all cycles of chemotherapy was 74% for patients treated with prechemotherapy and postchemotherapy G-CSF and 66% for patients treated with postchemotherapy G-CSF only (two-sided P, adjusted for dose = .21) and the median duration in both treatment arms was 3 days (two-sided P = .19). Unexpectedly, the incidence of grades 3 and 4 thrombocytopenia was much greater in patients who received prechemotherapy G-CSF compared with those who did not (54% versus 6%, respectively, over all chemotherapy cycles; two-sided P, adjusted for dose < .001). No difference in the decrease in hemoglobin level (adjusted for red blood cell transfusions) between patients in the two treatment arms was observed. CONCLUSIONS AND IMPLICATIONS: No beneficial effects were associated with the administration of G-CSF to cancer patients prior to chemotherapy. The observation of more severe thrombocytopenia in patients treated with prechemotherapy G-CSF led us to conclude that the proliferation of progenitor cells was still increased 48 hours after the last dose of G-CSF and that the administration of chemotherapy at or within this time period actually worsens the toxic effects on bone marrow. This result has important ramifications for the design of clinical cancer treatment protocols, especially those that involve shortened intervals between cycles of chemotherapeutic agent administration.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Medula Óssea/efeitos dos fármacos , Neoplasias da Mama/tratamento farmacológico , Fator Estimulador de Colônias de Granulócitos/efeitos adversos , Leucopenia/prevenção & controle , Adulto , Idoso , Antibióticos Antineoplásicos/efeitos adversos , Antineoplásicos Alquilantes/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Neoplasias da Mama/patologia , Ciclofosfamida/efeitos adversos , Doxorrubicina/efeitos adversos , Esquema de Medicação , Feminino , Fator Estimulador de Colônias de Granulócitos/administração & dosagem , Humanos , Incidência , Leucopenia/induzido quimicamente , Pessoa de Meia-Idade , Neutropenia/prevenção & controle , Contagem de Plaquetas/efeitos dos fármacos , Proteínas Recombinantes/efeitos adversos , Índice de Gravidade de Doença , Fatores de Tempo
10.
Cancer Res ; 47(10): 2727-32, 1987 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-2952261

RESUMO

We administered anti-Leu 2a, a murine monoclonal antibody to the human suppressor-cytotoxic T-cell subset, to 20 patients with advanced cancer to determine the toxicity and its ability to deplete circulating Leu 2(+) lymphocytes. Four doses were tested, 1, 5, 25, and 100 mg, and the infusion rate varied from 2 to 24 h. Administration of anti-Leu 2a produced rapid (within 4 h) depletion of circulating Leu 2(+) lymphocytes, the magnitude and duration of which were dose dependent: the 1-mg dose caused a less than 50% fall in circulating Leu 2(+) lymphocytes, while the higher doses caused 75-98% depletion of those cells in 10 of 17 patients so treated. The duration of depletion of Leu 2(+) cells was 1-2 days after 5 mg of anti-Leu 2a, 3-4 days after 25 mg, and 4-30+ days after 100 mg. In seven patients (3-5, 3-25, and 1-100 mg), there was persistence of variable numbers of circulating mouse Ig-coated Leu 2(+) cells, possibly indicating impaired capacity to clear these cells. Modulation of the Leu 2a antigen after antibody treatment was not observed. Peak serum levels of anti-Leu 2a were (medians): 5-mg dose = 350 ng/ml, 25-mg dose = 5500 ng/ml, and 100-mg dose = 48,000 ng/ml; murine antibody was detectable in the serum for 7-14 days after the 100-mg dose. All patients had increased titers of antimouse antibody following treatment with peak titers on day 14. The most common toxicity was shaking chills. This occurred within 1.5 h of beginning the infusion, lasted for no more than 30 min, and did not require cessation of the treatment. Anti-Leu 2a administration caused a clinically unimportant, but statistically significant fall in hematocrit (mean = -7%) and platelet (mean = -23%) count and a transient (less than 1 day duration) fall in the total lymphocyte count. Thus, infusion of murine monoclonal anti-Leu 2a can cause substantial depletion of the suppressor-cytotoxic T-cell subset with modest toxicity.


Assuntos
Anticorpos Monoclonais , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/imunologia , Adulto , Idoso , Animais , Antígenos de Diferenciação de Linfócitos T , Antígenos de Superfície/imunologia , Avaliação de Medicamentos , Contagem de Eritrócitos/efeitos dos fármacos , Feminino , Humanos , Imunoglobulinas/imunologia , Masculino , Camundongos , Pessoa de Meia-Idade , Contagem de Plaquetas/efeitos dos fármacos
11.
Cancer Res ; 50(16): 5095-101, 1990 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-2379173

RESUMO

Epirubicin, a stereoisomer of doxorubicin, is reported to have equal antitumor activity with lower cardiac and systemic toxicity. Recently the maximum tolerated dose of this drug has been revised upwards with reported increased response rates. However, the pharmacokinetics of epirubicin at high doses have never been reported. Accordingly, this study was designed to evaluate the pharmacokinetics of epirubicin when administered as either a 15-min i.v. bolus or a 6-h i.v. infusion in a phase I study at high doses. Nineteen patients with a variety of malignancies were given a total of 52 cycles of epirubicin at doses of 90 to 150 mg/m2 given once every 3 weeks. The maximum tolerated dose was 150 mg/m2 epirubicin given either as a bolus or as an infusion. The major dose-limiting toxicity was neutropenia. Interpatient variation occurred in the pharmacokinetics at each dose level but overall there were dose-dependent pharmacokinetics. This was manifested as a disproportionate increase in plasma levels and areas under the curve as the epirubicin dose was increased from 90 to 150 mg/m2. The pharmacokinetics of epirubicin could best be described by an open two-compartment model. Peak plasma concentrations were attained at a median of 12 min following the bolus injection and concentrations approached the steady state within a median of 55 min following the start of the 6-h infusion. Administration of the 150 mg/m2 dose over the 6 h compared to the bolus administration was associated with a 92% decrease in peak concentration from 3088 +/- 1503 to 234 +/- 126 ng/ml. This was not associated with an appreciable change in hematological or nonhematological toxicities. The median distribution half-life was 10 min and the median elimination half-life was 42.0 h. The cumulative renal excretion of the parent compound accounted for less than 2% of the administered dose. The major metabolites in both plasma and urine samples were 4'-O-beta-D-glucuronyl-4'-epidoxorubicin, 13-S-dihydro-4'-epidoxorubicin, and 4'-O-beta-D-glucuronyl-13-S-dihydro-4'-epidoxorubicin. This study demonstrates that a 135 mg/m2 bolus infusion given on a 3-weekly schedule is an appropriate initial dose for further clinical studies.


Assuntos
Epirubicina/farmacocinética , Neoplasias/tratamento farmacológico , Biotransformação , Cromatografia Líquida de Alta Pressão , Esquema de Medicação , Epirubicina/efeitos adversos , Epirubicina/uso terapêutico , Feminino , Humanos , Injeções Intravenosas , Contagem de Leucócitos/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Contagem de Plaquetas/efeitos dos fármacos
12.
Cancer Res ; 51(10): 2524-30, 1991 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-2021932

RESUMO

We report a phase I study in cancer patients being treated with i.v. bolus injections of highly purified lipopolysaccharide (LPS) Salmonella abortus equi. Twenty-four patients with disseminated cancer received escalating doses of LPS at 2-week intervals. Dose escalation was performed in six dose levels treating 3-6 patients at each level. Dose levels 1 and 2 consisted of 0.15 and 0.3 ng/kg, respectively. Further dose escalation up to 5.0 ng/kg was enabled by pretreatment with ibuprofen, which attenuated the constitutional side effects of LPS. The maximum tolerated dose was 4.0 ng/kg with dose-limiting toxicity being World Health Organization grade III hepatic toxicity. Hematological changes included transient decreases in WBCs affecting granulocytes, monocytes, and lymphocytes in a marked different pattern. Endogenous cytokine release occurred in an LPS dose-dependent manner as measured by tumor necrosis factor-alpha, interleukin-6, and macrophage colony-stimulating factor serum levels. Moderate antitumor activity in colorectal cancer was observed in the case of 2 patients. Phase II trials of LPS are currently in progress.


Assuntos
Endotoxinas/toxicidade , Lipopolissacarídeos/toxicidade , Neoplasias/terapia , Esquema de Medicação , Avaliação de Medicamentos , Endotoxinas/administração & dosagem , Endotoxinas/uso terapêutico , Feminino , Humanos , Injeções Intravenosas , Interleucina-6/sangue , Contagem de Leucócitos/efeitos dos fármacos , Lipopolissacarídeos/administração & dosagem , Lipopolissacarídeos/uso terapêutico , Fator Estimulador de Colônias de Macrófagos/sangue , Masculino , Pessoa de Meia-Idade , Contagem de Plaquetas/efeitos dos fármacos , Salmonella , Fator de Necrose Tumoral alfa/análise
13.
Cancer Res ; 52(4): 1013-7, 1992 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-1531320

RESUMO

We previously found that a high-molecular-weight anticancer agent, polystyrene-co-maleic acid conjugated neocarzinostatin (SMANCS), in which two chains of styrene/maleic acid copolymer are conjugated to the anticancer protein neocarzinostatin (NCS), accumulated more selectively in tumor tissue than in normal tissue and was more stable than NCS in blood. These results indicate that SMANCS should have less systemic toxicity and a better therapeutic effect than NCS. In this study, the antitumor activity and adverse effects of SMANCS were compared with those of NCS by using rat mammary tumor induced by 7,12-dimethylbenz[a]anthracene. When tumors of rats, that had received 7,12-dimethylbenz[a]anthracene (20 mg/kg, one dose, p.o. in oily formulation), became palpable usually after 4-20 weeks, SMANCS treatment was initiated. Thirty days after i.v. administration of SMANCS (0.1 mg/kg 3 times and 0.3 mg/kg 3 times), tumors had shrunk in 35 of 37 rats (a mean weight was about 10% of control value; or decreased to about 30% of the value of before treatment in tumor weight); tumor size had not changed in 1 rat, and in the remaining 1 rat the tumor had enlarged. Thirty days after i.v. administration of NCS, tumors had shrunk in 8 of 14 rats, but the tumor size was unchanged in 1 rat and was enlarged in 5. In the control group, all tumors had enlarged. Development of new tumors was completely prevented by the administration of SMANCS. Histological examination of sequential slices of tumor revealed clear finding of degeneration and tumor encapsulation at 30 days after initial administration of SMANCS, with an accompanying fatty degeneration, but these effects were not observed for tumors treated with NCS. Although red blood cell counts and hemoglobin amounts decreased significantly in rats receiving NCS, no such effects were apparent in the SMANCS group.


Assuntos
Anidridos Maleicos/uso terapêutico , Neoplasias Mamárias Experimentais/tratamento farmacológico , Poliestirenos/uso terapêutico , Zinostatina/análogos & derivados , 9,10-Dimetil-1,2-benzantraceno , Animais , Divisão Celular , Contagem de Eritrócitos/efeitos dos fármacos , Feminino , Hemoglobinas/metabolismo , Anidridos Maleicos/toxicidade , Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/patologia , Contagem de Plaquetas/efeitos dos fármacos , Poliestirenos/toxicidade , Ratos , Ratos Endogâmicos , Zinostatina/uso terapêutico , Zinostatina/toxicidade
14.
Cancer Res ; 50(14): 4254-9, 1990 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-2364384

RESUMO

cis-Bis-neodecanoato-trans-R,R-1,2-diaminocyclohexaneplatinum++ +(II) (NDDP) is a liposome dependent cisplatin analogue since the liposome carrier is required for its i.v. administration and for its biological activity. A Phase I study of liposome entrapped NDDP (L-NDDP) was performed using a single i.v. injection every 4 weeks. L-NDDP was prepared and characterized at M. D. Anderson Cancer Center. The maximum tolerated dose of L-NDDP was 312.5 mg/m2. The dose-limiting toxicity was myelosuppression, affecting all three blood cell lineages. The granulocyte nadir occurred on days 14-18, and the platelet nadir consistently earlier (days 11-12). The median day of recovery of blood cell counts was day 21 (range, 18-32). Other toxicities included grade 2 nausea and vomiting, fever consisting of a single temperature spike in most patients, grade 1 diarrhea after 60% of courses, and grade 1-2 malaise lasting for 5-10 days after the infusion in 73% of courses. Transient alanine aminotransferase elevations without clinical relevance were common. No signs of renal dysfunction or ototoxicity were observed. One patient with a preexisting peripheral neuropathy showed some progression of the neuropathy after a cumulative dose of 1605 mg/m2. Except for fever and transient liver dysfunction, no liposome related side effects were observed in spite of the high doses of lipid administered. The blood clearance of L-NDDP fits a two-compartment model at lower doses and a single-compartment model at the maximum tolerated dose, suggesting that saturation of the reticuloendothelial organs occurs at the maximum tolerated dose. Two minimal responses were observed. L-NDDP has a toxicity profile similar to that of carboplatin. Phase II studies to address the issue of how the therapeutic index of platinum compounds is affected by liposome entrapment are being planned.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias/tratamento farmacológico , Compostos Organoplatínicos/administração & dosagem , Portadores de Fármacos/administração & dosagem , Avaliação de Medicamentos , Feminino , Hemoglobinas/metabolismo , Humanos , Contagem de Leucócitos/efeitos dos fármacos , Lipossomos , Masculino , Compostos Organoplatínicos/efeitos adversos , Compostos Organoplatínicos/farmacocinética , Contagem de Plaquetas/efeitos dos fármacos
15.
Cancer Res ; 50(3): 606-14, 1990 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-2404573

RESUMO

A Phase I study of bacterially synthesized recombinant human granulocyte-macrophage colony-stimulating factor (rhGM-CSF) was undertaken in 21 patients with advanced malignancy or neutropenia. rhGM-CSF was administered once daily by i.v. bolus injection (0.3 to 3 micrograms/kg/day) or 2-h i.v. infusion (3 to 20 micrograms/kg day) for 10 days. rhGM-CSF at all i.v. doses caused an immediate transient decrease in circulating neutrophils, eosinophils, and monocytes. By 6 h after rhGM-CSF, circulating leukocyte levels were restored. Daily i.v. bolus dosing (0.3 to 3 micrograms/kg/day) did not elevate leukocyte levels except in one neutropenic patient. Daily 2-h i.v. infusions (10 to 20 micrograms/kg/day) caused a dose-dependent leukocytosis with increased levels of neutrophils (up to 4.3-fold), eosinophils (up to 18-fold), and monocytes (up to 3.5-fold). Marrow aspirates showed increased proportions of promyelocytes and myelocytes during rhGM-CSF administration. Retreatment after 10 days without rhGM-CSF resulted in a more marked leukocytosis at doses greater than or equal to 10 micrograms/kg/day. Platelet levels decreased for the first 3 days and then increased during the first course of rhGM-CSF administration. Two patients with chronic lymphocytic leukemia had a transient reduction in lymphocytosis. Serum cholesterol and albumin levels decreased, and vitamin B12 levels increased during rhGM-CSF treatment. At doses of up to 15 micrograms/kg/day, rhGM-CSF was relatively well tolerated by the patients, but adverse effects included bone pain, lethargy, fever, rash, and weight gain. A first dose reaction characterized by hypoxia and hypotension was identified at dose levels greater than or equal to 1 microgram/kg. Dosing i.v. was less potent at inducing a leukocytosis than previously observed for equivalent s.c. doses and was associated with a higher incidence of generalized rash and first dose reactions. The maximal tolerated dose of i.v. rhGM-CSF was 15 micrograms/kg/day. Phase II studies in which the derived effect is to raise leukocyte levels should be undertaken at rhGM-CSF doses of 3 to 15 micrograms/kg/day.


Assuntos
Fatores Estimuladores de Colônias/administração & dosagem , Substâncias de Crescimento/administração & dosagem , Neoplasias/terapia , Adulto , Idoso , Basófilos , Relação Dose-Resposta a Droga , Avaliação de Medicamentos , Eosinófilos , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Hematopoese/efeitos dos fármacos , Humanos , Infusões Intravenosas , Injeções Subcutâneas , Contagem de Leucócitos/efeitos dos fármacos , Lipídeos/sangue , Linfócitos , Pessoa de Meia-Idade , Monócitos , Neutrófilos , Contagem de Plaquetas/efeitos dos fármacos , Proteínas Recombinantes , Albumina Sérica/metabolismo , Fatores de Tempo
16.
Cancer Res ; 54(16): 4393-7, 1994 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-8044787

RESUMO

Ifosfamide (IFF) is a nitrogen mustard with significant activity against a number of tumors. Since it is a chiral molecule, it has been suggested that enantioselective metabolism could result in different efficacy and toxicity profiles for (R)- and (S)-ifosfamide. Both experimental animal and clinical data suggest that N-dechloroethyl metabolites of (S)-IFF are more significantly associated with neurological toxicity, which may limit therapeutic use of IFF. We have used purified ifosfamide enantiomers to examine the pharmacokinetics; spectrum of toxicity including lethality, weight loss, and myelosuppression; and antitumor effects of the mixture compared to each of the purified enantiomers. In the MatB mammary carcinoma grown in female Fischer rats we demonstrated that the antitumor efficacy appears to be the same for (R)-IFF and (S)-IFF, while the (R)-IFF has greater myelotoxicity. Pharmacokinetic analysis of plasma concentration-time confirms that the (R)-IFF is metabolized to a greater extent than (S)-IFF via the activation pathway. These data suggest that purified (R)-IFF may be an effective way to delivery active cytotoxic drug while limiting the generation of neurotoxic metabolites.


Assuntos
Peso Corporal/efeitos dos fármacos , Medula Óssea/efeitos dos fármacos , Ifosfamida/toxicidade , Neoplasias Mamárias Experimentais/tratamento farmacológico , Animais , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Ifosfamida/análogos & derivados , Ifosfamida/farmacocinética , Ifosfamida/farmacologia , Dose Letal Mediana , Contagem de Leucócitos/efeitos dos fármacos , Contagem de Plaquetas/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344 , Estereoisomerismo
17.
Cancer Res ; 52(3): 521-4, 1992 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-1732038

RESUMO

Piritrexim, an orally administered, lipid-soluble antifolate, was evaluated in a multi-institutional phase I trial in children. The starting dose was 10 mg/m2/dose administered every 8 h daily for 5 days for 3 consecutive weeks, with dose escalations in increments of 5 mg/m2/dose. Eighteen patients (16 with metastatic sarcoma, 1 with acute lymphoblastic leukemia, and 1 with a brainstem glioma), 3.5-20 years of age, with malignancy refractory to therapy, were entered into the study. The dose-limiting toxicities (DLTs), which were myelosuppression and mucositis, occurred in 4 of 4 patients treated at the 25-mg/m2/dose level but in none of the patients treated at the 15- and 20-mg/m2/dose levels. The recommended dose for phase II trials is 20 mg/m2/dose. Pharmacokinetic monitoring was performed in 15 of the 18 children. The area under the concentration-time curve (AUC) was linearly related to the dose administered. Piritrexim was rapidly absorbed, with the median time to peak level occurring 1.5 h after an oral dose. The terminal half-life of piritrexim ranged from 1.5 to 4.5 h. A limited sampling strategy developed earlier, capable of predicting the AUC based on the plasma concentrations at 3 and 6 h after an oral dose, was prospectively tested in this trial and proved to be highly predictive of the AUC (r = 0.98, P = 0.0001). Pharmacodynamic-pharmacokinetic correlations were obtained after combining data from this and the prior phase I pediatric trial. Trough plasma piritrexim concentration strongly correlated with DLT (P = 0.0016). A trough plasma piritrexim concentration greater than 0.5 microM appeared to be predictive of toxicity. Eleven of 15 patients with trough concentrations exceeding this threshold experienced DLTs. Therapeutic drug monitoring may thus play an important role in adjusting the dose and schedule of piritrexim in future trials.


Assuntos
Antineoplásicos/toxicidade , Neoplasias/tratamento farmacológico , Pirimidinas/toxicidade , Adolescente , Adulto , Neoplasias Encefálicas/tratamento farmacológico , Criança , Pré-Escolar , Esquema de Medicação , Avaliação de Medicamentos , Feminino , Glioma/tratamento farmacológico , Humanos , Contagem de Leucócitos/efeitos dos fármacos , Masculino , Contagem de Plaquetas/efeitos dos fármacos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Pirimidinas/administração & dosagem , Pirimidinas/farmacocinética , Pirimidinas/uso terapêutico , Sarcoma/tratamento farmacológico
18.
Cancer Res ; 49(22): 6337-41, 1989 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-2804978

RESUMO

Several chemotherapeutic protocols for the treatment of malignancies include administration of methotrexate (MTX) during or shortly after total anesthesia. Clinical observations in patients treated for breast carcinoma or childhood cancer have shown unexpected myelosuppression and mucosal damage. This phenomenon may be attributed to the synergistic effects of nitrous oxide, which inactivates the cobalamin coenzyme of methionine synthase, and MTX, which inhibits dihydrofolate reductase, on folate metabolism. However, no quantitative data on dose-effect relationships are available regarding the combined toxicity of MTX and N2O. We investigated the effect of exposure to N2O on the toxicity of MTX. Groups of male Wistar rats were exposed to either 50% N2O/50% O2 or air for 12-48 h. Subsequently, a single i.p. injection of 10, 20, 40, or 80 mg MTX/kg body weight was given. Gastrointestinal toxicity resulted in diarrhea and weight loss in all groups for 5 days after MTX administration. Concomitantly, bone marrow depression with leukocytopenia and thrombocytopenia occurred. Exposure to N2O did not alter the plasma clearance of MTX. No substantial liver or kidney toxicity could be detected, but the 50% lethal dose for MTX was reduced from 60 mg/kg to 10 mg/kg if rats had been exposed to N2O for 48 h; the main causes of death were dehydration and bleeding. The administration of 5-formyl-tetrahydrofolate (4 x 10 mg i.p.) but not 5-methyltetrahydrofolate protected completely against the lethal effect of the drug combination. Altogether, cytotoxic effects of MTX on proliferating cells are potentiated by N2O. Therefore, the use of this anesthetic shortly before or during MTX administration should be avoided.


Assuntos
Metotrexato/toxicidade , Óxido Nitroso/farmacologia , Animais , Sistema Digestório/efeitos dos fármacos , Sistema Digestório/patologia , Interações Medicamentosas , Rim/efeitos dos fármacos , Rim/patologia , Dose Letal Mediana , Leucovorina/farmacologia , Contagem de Leucócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Taxa de Depuração Metabólica , Metotrexato/farmacocinética , Contagem de Plaquetas/efeitos dos fármacos , Ratos , Ratos Endogâmicos , Tetra-Hidrofolatos/farmacologia
19.
Cancer Res ; 50(14): 4216-20, 1990 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-2364379

RESUMO

Tumor necrosis factor (TNF) is a pleiotropic cytokine which exerts a wide range of effects when administered in vivo. Using a murine model, we have investigated the effect of pretreatment with 1 microgram (2.6 x 10(4) units) per mouse of recombinant murine TNF-alpha on hematopoietic recovery following administration of cyclophosphamide, 5-fluorouracil, methotrexate, or vinblastine. TNF pretreatment results in enhanced regeneration of circulating neutrophils and hematopoietic progenitors, as measured by in vivo and in vitro assays, in animals given cycle-specific chemotherapeutic agents. The results may suggest that TNF affects cycle kinetics in hematopoietic progenitor cell populations, thus making these cells less prone to the cytocidal effects of the chemotherapeutic agents. As myeloablation is a frequent and often critical side effect following cancer treatment, these findings may have clinical implications.


Assuntos
Medula Óssea/patologia , Ciclofosfamida/toxicidade , Contagem de Eritrócitos/efeitos dos fármacos , Fluoruracila/toxicidade , Hematopoese/efeitos dos fármacos , Contagem de Leucócitos/efeitos dos fármacos , Metotrexato/toxicidade , Contagem de Plaquetas/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Vimblastina/toxicidade , Animais , Medula Óssea/efeitos dos fármacos , Ensaio de Unidades Formadoras de Colônias , Células-Tronco Hematopoéticas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/citologia , Neutrófilos/efeitos dos fármacos , Proteínas Recombinantes/farmacologia
20.
Biochim Biophys Acta ; 1081(1): 33-8, 1991 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-1991153

RESUMO

Evidence suggesting that dietary omega-3 polyunsaturated fatty acids decrease the risk of thrombosis comes mainly from studies involving supplementation with large amounts (15-20 g/day) of fish oil extract. We investigated the inhibition of platelet function by a moderate amount (4 g/d) of ethyl eicosapentaenoate (E-EPA) compared to a concentrated fish oil extract (6 g/d) when given as a supplement to an ordinary diet. We also determined the effects of these supplements on platelet EPA incorporation, thromboxane synthesis, calcium mobilization and fibrinogen binding. After 4 weeks, both omega-3 supplements increased the amount of EPA in platelet phospholipids. The fish oil extract, which contained docosahexaenoic acid (DHA), had increased the amount of DHA also. The total increase in omega-3 fatty acids was similar for both supplements. E-EPA decreased serum cholesterol by 13% and triacylglycerols, 35%; increased the bleeding time by 57% and the threshold dose of collagen needed to induce platelet aggregation by 46%. Thromboxane synthesis in response to collagen was decreased 65% by E-EPA. Thus, the dietary supplement of pure E-EPA was more effective in limiting platelet reactivity than a concentrated fish oil extract providing an equivalent amount of omega-3 fatty acids. As an antithrombotic agent, E-EPA should allow for reasonable daily doses in long-term treatment of cardiovascular disease.


Assuntos
Plaquetas/metabolismo , Gorduras na Dieta/farmacologia , Ácido Eicosapentaenoico/análogos & derivados , Fibrinolíticos/farmacologia , Óleos de Peixe/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Trifosfato de Adenosina/sangue , Adulto , Tempo de Sangramento , Plaquetas/efeitos dos fármacos , Colesterol/sangue , Colágeno/farmacologia , Ácido Eicosapentaenoico/farmacologia , Ácidos Graxos/análise , Humanos , Técnicas In Vitro , Masculino , Fosfolipídeos/sangue , Contagem de Plaquetas/efeitos dos fármacos , Valores de Referência , Tromboxanos/sangue , Triglicerídeos/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA