Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Radiat Res ; 188(6): 615-625, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28985133

RESUMEN

A partial-body irradiation model with approximately 2.5% bone marrow sparing (PBI/BM2.5) was established to determine the radiation dose-response relationships for the prolonged and delayed multi-organ effects of acute radiation exposure. Historically, doses reported to the entire body were assumed to be equal to the prescribed dose at some defined calculation point, and the dose-response relationship for multi-organ injury has been defined relative to the prescribed dose being delivered at this point, e.g., to a point at mid-depth at the level of the xiphoid of the non-human primate (NHP). In this retrospective-dose study, the true distribution of dose within the major organs of the NHP was evaluated, and these doses were related to that at the traditional dose-prescription point. Male rhesus macaques were exposed using the PBI/BM2.5 protocol to a prescribed dose of 10 Gy using 6-MV linear accelerator photons at a rate of 0.80 Gy/min. Point and organ doses were calculated for each NHP from computed tomography (CT) scans using heterogeneous density data. The prescribed dose of 10.0 Gy to a point at midline tissue assuming homogeneous media resulted in 10.28 Gy delivered to the prescription point when calculated using the heterogeneous CT volume of the NHP. Respective mean organ doses to the volumes of nine organs, including the heart, lung, bowel and kidney, were computed. With modern treatment planning systems, utilizing a three-dimensional reconstruction of the NHP's CT images to account for the variations in body shape and size, and using density corrections for each of the tissue types, bone, water, muscle and air, accurate determination of the differences in dose to the NHP can be achieved. Dose and volume statistics can be ascertained for any body structure or organ that has been defined using contouring tools in the planning system. Analysis of the dose delivered to critical organs relative to the total-body target dose will permit a more definitive analysis of organ-specific effects and their respective influence in multiple organ injury.


Asunto(s)
Relación Dosis-Respuesta en la Radiación , Modelos Animales , Órganos en Riesgo/efectos de la radiación , Fotones , Vísceras/efectos de la radiación , Abdomen/efectos de la radiación , Animales , Médula Ósea , Imagenología Tridimensional , Macaca mulatta , Masculino , Tamaño de los Órganos , Tratamientos Conservadores del Órgano , Especificidad de Órganos , Aceleradores de Partículas , Dosificación Radioterapéutica , Planificación de la Radioterapia Asistida por Computador , Radioterapia de Alta Energía , Estudios Retrospectivos , Tórax/efectos de la radiación , Tomografía Computarizada por Rayos X
2.
Drugs Today (Barc) ; 51(9): 537-48, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26488033

RESUMEN

The U.S. Food and Drug Administration (FDA) recently approved Neupogen(®) (filgrastim) for the treatment of patients with radiation-induced myelosuppression following a radiological/nuclear incident. It is the first medical countermeasure currently approved by the FDA for this indication under the criteria of the FDA "animal rule". This article summarizes the consequences of high-dose radiation exposure, a description of the hematopoietic acute radiation syndrome (H-ARS), the use of hematopoietic growth factors in radiation accident victims and current available treatments for H-ARS with an emphasis on the use of Neupogen in this scenario.


Asunto(s)
Síndrome de Radiación Aguda/tratamiento farmacológico , Biosimilares Farmacéuticos/uso terapéutico , Filgrastim/uso terapéutico , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Síndrome de Radiación Aguda/sangre , Síndrome de Radiación Aguda/diagnóstico , Animales , Biosimilares Farmacéuticos/efectos adversos , Filgrastim/efectos adversos , Hematopoyesis/efectos de la radiación , Células Madre Hematopoyéticas/diagnóstico por imagen , Humanos , Traumatismos Experimentales por Radiación , Radiografía , Terrorismo , Resultado del Tratamiento
3.
Radiat Res ; 178(5): 403-13, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22991919

RESUMEN

Conventional daily administration of filgrastim is effective in reducing the duration of severe neutropenia and enhancing survival following lethal radiation, myelosuppressive cytotoxic therapy or myeloablation and stem cell transplantation. A sustained-duration form of filgrastim, pegfilgrastim has significantly simplified scheduling protocols after chemotherapy-induced neutropenia to a single injection while maintaining the therapeutic effectiveness of daily administration of filgrastim. We examined the ability of a single or double (weekly) administration of pegfilgrastim to significantly improve neutrophil recovery in a rhesus macaque model of severe radiation-induced myelosuppression. Animals were exposed to potentially lethal 6 Gy total-body X radiation. After irradiation all animals received supportive care and were administered either pegfilgrastim at 300 µg/kg on day 1 or day 1 and day 7 post exposure, or filgrastim at 10 µg/kg/day initiated on day 1 post exposure and continued daily through neutrophil recovery. Pharmacokinetic parameters and neutrophil-related values for duration of neutropenia, neutrophil nadir, time to recovery to an absolute neutrophil count ≥500/µL or ≥2000/µL, and days of antibiotic support were determined. Effective plasma concentrations of pegfilgrastim were maintained in neutropenic animals until after the onset of hematopoietic recovery, which is consistent with neutrophil-dependent properties of elimination. Administration of pegfilgrastim at day 1 and day 7 was most effective at improving neutrophil recovery compared to daily administration of filgrastim or a single injection of pegfilgrastim on day 1, after severe, radiation-induced myelosuppression in rhesus macaques.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos , Neutrófilos , Protectores contra Radiación , Animales , Esquema de Medicación , Filgrastim , Factor Estimulante de Colonias de Granulocitos/farmacocinética , Factor Estimulante de Colonias de Granulocitos/farmacología , Dosificación Letal Mediana , Macaca mulatta , Masculino , Neutropenia/tratamiento farmacológico , Neutrófilos/efectos de los fármacos , Neutrófilos/efectos de la radiación , Polietilenglicoles , Protectores contra Radiación/farmacocinética , Protectores contra Radiación/farmacología , Proteínas Recombinantes/farmacocinética , Proteínas Recombinantes/farmacología , Rayos X
4.
Bone Marrow Transplant ; 32(4): 399-404, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12900776

RESUMEN

Daily administration of filgrastim decreases the duration of severe neutropenia in the clinical setting. A sustained-duration form of filgrastim, pegfilgrastim, significantly reduces scheduling protocols to a single injection per chemotherapy cycle while maintaining therapeutic efficiency. We examined the ability of a single injection of pegfilgrastim to significantly improve neutrophil recovery following autologous bone marrow transplantation (AuBMT) in rhesus macaques. On day 1, postmyeloablation (920 cGy x-irradiation) and AuBMT, animals received either 0.1% autologous serum for 18 consecutive days (n=13), or single doses of pegfilgrastim via the subcutaneous (s.c.) or intravenous (i.v.) route (300 or 100 micro g/kg), or a single dose of filgrastim at 300 micro g/kg via the s.c. or i.v. route, or filgrastim at 10 micro g/kg via the s.c. route (n=4) on a daily basis (range=days 12-17). Pharmacokinetic parameters and neutrophil recovery were assessed. A single dose of pegfilgrastim via the i.v. or s.c. route was as effective as daily filgrastim administration, resulting in significant improvement of neutrophil recovery after myeloablation and ABuMT. Effective pegfilgrastim plasma concentrations were maintained in neutropenic animals until after the onset of hematopoietic recovery. Enhanced pharmacokinetics in AuBMT cohorts are consistent with self-regulating, neutrophil-mediated clearance.


Asunto(s)
Trasplante de Médula Ósea/métodos , Factor Estimulante de Colonias de Granulocitos/análogos & derivados , Factor Estimulante de Colonias de Granulocitos/farmacología , Factor Estimulante de Colonias de Granulocitos/farmacocinética , Neutrófilos/metabolismo , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Células de la Médula Ósea , Estudios de Cohortes , Citocinas/metabolismo , Filgrastim , Macaca mulatta , Masculino , Neutropenia , Neutrófilos/efectos de los fármacos , Polietilenglicoles , Proteínas Recombinantes/metabolismo , Factores de Tiempo , Acondicionamiento Pretrasplante , Trasplante Autólogo
5.
Stem Cells ; 19(6): 514-21, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11713343

RESUMEN

Leridistim, a member of the myelopoietin family of dual receptor agonists that binds interleukin-3 and G-CSF receptors, has been shown to enhance hematopoietic activity in rhesus monkeys above that observed with either cytokine alone or in combination. This study demonstrated the ability of a pegylated form of leridistim (peg-leridistim), administered s.c., as a single- or two-dose regimen separated by 4 or 7 days, to significantly improve neutrophil recovery following radiation-induced myelosuppression. Rhesus macaques were total body x-irradiated (250 kVp, TBI) to 600 cGy. Following TBI, two groups received peg-leridistim (n = 5) or leridistim (n = 4) at a dose of 600 microg/kg on day 1, while two other groups (both n = 4) received peg-leridistim at 200 microg/kg on day 1 and day 4, or day 1 and day 7. The irradiation controls (n = 7) received 0.1% autologous serum for 18 days. All peg-leridistim treatment schedules significantly improved all neutrophil-related parameters following TBI as compared with nontreated controls and were equivalent in effect when compared among themselves. Administration of a single high dose or two separate lower doses of peg-leridistim significantly improved neutrophil regeneration, in a manner equal to that of conventional daily or abbreviated every-other-day administration of leridistim in this nonhuman primate model of severe myelosuppression.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos , Interleucina-3/farmacología , Neutropenia/prevención & control , Neutrófilos/efectos de los fármacos , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/efectos de la radiación , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de la radiación , Interleucina-3/química , Interleucina-3/farmacocinética , Macaca mulatta , Masculino , Tasa de Depuración Metabólica , Neutropenia/etiología , Neutropenia/patología , Neutrófilos/citología , Neutrófilos/efectos de la radiación , Polietilenglicoles/química , Polietilenglicoles/farmacocinética , Polietilenglicoles/farmacología , Dosis de Radiación , Proteínas Recombinantes de Fusión , Proteínas Recombinantes , Factores de Tiempo , Irradiación Corporal Total/efectos adversos
6.
Stem Cells ; 19(6): 522-33, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11713344

RESUMEN

Leridistim is from the myelopoietin family of proteins, which are dual receptor agonists of the human interleukin-3 and G-CSF receptor complexes. This study investigated the effect of dosage, administration route, and schedule of leridistim to stimulate multilineage hematopoietic recovery in total body irradiated rhesus monkeys. Animals were x-irradiated on day 0 (600 cGy, 250 kVp) and then received, on day 1, leridistim s.c. in an abbreviated, every-other-day schedule at 200 microg/kg, or daily at 50 microg/kg, or i.v. daily or every-other-day schedules at 200 microg/kg dose. Other cohorts received G-CSF (Neupogen((R)) [Filgrastim]) in an every-other-day schedule at 100 microg/kg/day, or autologous serum (0.1%) s.c. daily. Hematopoietic recovery was assessed by bone marrow clonogenic activity, peripheral blood cell nadirs, duration of cytopenias, time to recovery to cellular thresholds, and requirements for clinical support. Leridistim, administered s.c. every other day, or i.v. daily, significantly improved neutrophil, platelet, and lymphocyte nadirs, shortened the respective durations of cytopenia, hastened trilineage hematopoietic recovery, and reduced antibiotic and transfusion requirements. A lower dose of leridistim administered daily s.c. enhanced recovery of neutrophil and platelet parameters but did not affect lymphocyte recovery relative to controls. Leridistim, a novel engineered hematopoietic growth factor administered at the appropriate dose, route and schedule, stimulates multilineage hematopoietic reconstitution in radiation-myelosuppressed nonhuman primates.


Asunto(s)
Células Madre Hematopoyéticas/efectos de los fármacos , Interleucina-3/farmacología , Leucopoyesis/efectos de los fármacos , Receptores de Factor Estimulante de Colonias de Granulocito/agonistas , Receptores de Interleucina-3/agonistas , Animales , Plaquetas/citología , Plaquetas/efectos de los fármacos , Plaquetas/efectos de la radiación , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/efectos de la radiación , Linaje de la Célula , Relación Dosis-Respuesta a Droga , Vías de Administración de Medicamentos , Esquema de Medicación , Eritrocitos/citología , Eritrocitos/efectos de los fármacos , Eritrocitos/efectos de la radiación , Factor Estimulante de Colonias de Granulocitos/farmacología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de la radiación , Inyecciones Intravenosas , Inyecciones Subcutáneas , Interleucina-3/química , Leucopoyesis/efectos de la radiación , Subgrupos Linfocitarios/citología , Subgrupos Linfocitarios/efectos de los fármacos , Subgrupos Linfocitarios/efectos de la radiación , Linfocitos/citología , Linfocitos/efectos de los fármacos , Linfocitos/efectos de la radiación , Macaca mulatta , Masculino , Modelos Animales , Neutropenia/etiología , Neutropenia/prevención & control , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Neutrófilos/efectos de la radiación , Proteínas Recombinantes de Fusión , Proteínas Recombinantes , Trombocitopenia/etiología , Trombocitopenia/prevención & control , Factores de Tiempo , Irradiación Corporal Total/efectos adversos
7.
Clin Cancer Res ; 7(8): 2301-8, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11489805

RESUMEN

Pharmacodynamic measures of neutropenia, such as absolute neutrophil count at nadir and neutrophil survival fraction, may not reflect the overall time course of neutropenia. We developed a pharmacokinetic-pharmacodynamic model to describe and quantify the time course of neutropenia after administration of topotecan to children and to compare this with nonhuman primates (NHPs) as a potential preclinical model of neutropenia. Topotecan was administered as a 30-min infusion daily for 5 days, repeated every 21 days. As part of a Phase I Pediatric Oncology Group study, topotecan was administered at 1.4 and 1.7 mg/m(2)/day without filgrastim (POG), and at 1.7, 2, and 2.4 mg/m(2)/day with filgrastim (POG+G). In NHPs, topotecan was administered at 5, 10, and 20 mg/m(2)/day without filgrastim. A pharmacokinetic-pharmacodynamic model was fit to profiles of topotecan lactone plasma concentrations and neutrophil survival fraction from cycle 1 and used to calculate topotecan lactone area under the plasma concentration-versus-time curve from 0 to 120 h (AUC(LAC)) and the area between the baseline and treatment-related neutrophil survival fraction (ABC) from 0 to 700 h. The mean +/- SD neutrophil survival fraction at nadir for the POG, POG+G, and NHP groups was 0.12 +/- 0.09, 0.11 +/- 0.17, and 0.09 +/- 0.08, respectively (P > 0.05). The mean +/- SD for the ratio of ABC to AUC(LAC) for the POG and NHP groups was 1.02 +/- 0.38 and 0.16 +/- 0.09, respectively (P < 0.05). The model estimate of ABC and the ratio of ABC to AUC(LAC) in children and NHPs may better reflect sensitivity to chemotherapy-induced neutropenia.


Asunto(s)
Neutropenia/patología , Topotecan/farmacocinética , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Área Bajo la Curva , Niño , Preescolar , Ensayos Clínicos Fase I como Asunto , Modelos Animales de Enfermedad , Femenino , Filgrastim , Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Humanos , Masculino , Tasa de Depuración Metabólica , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neutropenia/inducido químicamente , Neutropenia/metabolismo , Neutrófilos/efectos de los fármacos , Proteínas Recombinantes , Factores de Tiempo , Inhibidores de Topoisomerasa I , Topotecan/administración & dosificación , Topotecan/efectos adversos
8.
Stem Cells ; 19(4): 329-38, 2001.
Artículo en Inglés | MEDLINE | ID: mdl-11463953

RESUMEN

Promegapoietin-1a (PMP-1a), a multifunctional agonist for the human interleukin 3 and Mpl receptors, was evaluated for its ability to stimulate hematopoietic reconstitution in nonhuman primates following severe radiation-induced myelosuppression. Animals were total body x-irradiated (250 kVp) to 600 cGy total midline tissue dose. PMP-1a was administered s.c. in several protocols: A) daily (50 microg/kg) for 18 days; B) nine doses (5 microg/kg) every other day for 3 weeks; C) a single high dose (100 microg/kg) at 20 hours, or D) a single high dose (100 microg/kg) at 1 hour following TBI. The irradiation controls received 0.1% autologous serum for 18 consecutive days. Hematopoietic recovery was assessed by bone marrow clonogenic activity, peripheral blood cell nadirs, duration of cytopenias, time to recovery to cellular thresholds, and requirements for clinical support. PMP-1a, irrespective of administration schedule, significantly improved all platelet-related parameters: thrombocytopenia was eliminated, the severity of platelet nadirs was significantly improved, and recovery of platelet counts to > or =20,000/miccrol was significantly reduced in all PMP-1a-treated cohorts. As a consequence, all PMP-1a-treated cohorts were transfusion-independent. Neutrophil regeneration was augmented in all treatment schedules. Additionally, all PMP-1a-treated cohorts showed an improvement in red blood cell nadir and recovery. PMP-1a in conventional or abbreviated schedules induced significant thrombopoietic regeneration relative to the control cohort, whereas significant improvement in neutrophil recovery was schedule-dependent in radiation-myelosuppressed nonhuman primates.


Asunto(s)
Hematopoyesis/efectos de los fármacos , Receptores de Interleucina-3/agonistas , Proteínas Recombinantes de Fusión/farmacología , Trombopoyetina/agonistas , Trombopoyetina/farmacología , Animales , Esquema de Medicación , Combinación de Medicamentos , Eritrocitos/efectos de los fármacos , Eritrocitos/fisiología , Eritrocitos/efectos de la radiación , Hematopoyesis/fisiología , Hematopoyesis/efectos de la radiación , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Interleucina-3 , Macaca mulatta , Masculino , Neutropenia/tratamiento farmacológico , Neutropenia/fisiopatología , Fragmentos de Péptidos , Péptidos/administración & dosificación , Péptidos/farmacología , Ingeniería de Proteínas , Receptores de Interleucina-3/administración & dosificación , Receptores de Interleucina-3/genética , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/farmacocinética , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/fisiopatología , Trombopoyetina/administración & dosificación , Trombopoyetina/genética , Trombopoyetina/farmacocinética
9.
Blood ; 97(6): 1534-42, 2001 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-11238087

RESUMEN

SB-251353 is an N-terminal truncated form of the human CXC chemokine GRObeta. Recombinant SB-251353 was profiled in murine and rhesus monkey peripheral blood stem cell mobilization and transplantation models. SB-251353 rapidly and transiently mobilized hematopoietic stem cells and neutrophils into the peripheral blood after a single subcutaneous injection. Transplantation of equivalent numbers of hematopoietic stem cells mobilized by SB-251353 into lethally irradiated mice resulted in faster neutrophil and platelet recovery than stem cells mobilized by granulocyte colony-stimulating factor (G-CSF). A single injection of SB-251353 in combination with 4 days of G-CSF administration resulted in augmented stem and progenitor cell mobilization 5-fold greater than G-CSF alone. Augmented stem cell mobilization could also be demonstrated in mice when a single injection of SB-251353 was administered with only one-day treatment with G-CSF. In addition, SB-251353, when used as a single agent or in combination with G-CSF, mobilized long-term repopulating stem cells capable of hematopoietic reconstitution of lethally irradiated mice. In rhesus monkeys, a single injection of SB-251353 induced rapid increases in peripheral blood hematopoietic progenitor cells at a 50-fold lower dose than in mice, which indicates a shift in potency. These studies provide evidence that the use of SB-251353 alone or in combination with G-CSF mobilizes hematopoietic stem cells with long-term repopulating ability. In addition, this treatment may (1) reduce the number of apheresis sessions and/or amount of G-CSF required to collect adequate numbers of hematopoietic stem cells for successful peripheral blood cell transplantation and (2) improve hematopoietic recovery after transplantation.


Asunto(s)
Quimiocinas CXC/farmacología , Factores Quimiotácticos/farmacología , Sustancias de Crecimiento/farmacología , Movilización de Célula Madre Hematopoyética/métodos , Células Madre Hematopoyéticas/efectos de los fármacos , Péptidos y Proteínas de Señalización Intercelular , Animales , Plaquetas/citología , Plaquetas/efectos de los fármacos , Quimiocina CXCL1 , Quimiocina CXCL2 , Quimiocinas CXC/administración & dosificación , Quimiocinas CXC/fisiología , Quimiocinas CXC/uso terapéutico , Factores Quimiotácticos/administración & dosificación , Factores Quimiotácticos/fisiología , Factores Quimiotácticos/uso terapéutico , Quimioterapia Combinada , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Sustancias de Crecimiento/fisiología , Sustancias de Crecimiento/uso terapéutico , Hematopoyesis/efectos de los fármacos , Movilización de Célula Madre Hematopoyética/normas , Trasplante de Células Madre Hematopoyéticas/métodos , Trasplante de Células Madre Hematopoyéticas/normas , Humanos , Macaca mulatta , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/farmacología , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Proteínas de Neoplasias/farmacología , Proteínas de Neoplasias/fisiología , Proteínas de Neoplasias/uso terapéutico , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Especificidad de la Especie
10.
Blood ; 95(3): 837-45, 2000 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-10648394

RESUMEN

Myelopoietins (MPOs) constitute a family of engineered, chimeric molecules that bind and activate the IL-3 and G-CSF receptors on hematopoietic cells. This study investigated the in vivo hematopoietic response of rhesus monkeys administered MPO after radiation-induced myelosuppression. Animals were total body irradiated (TBI) in 2 series, with biologically equivalent doses consisting of either a 700 cGy dose of Cobalt-60 ((60)Co) gamma-radiation or 600 cGy, 250 kVp x-irradiation. First series: On day 1 after 700 cGy irradiation, cohorts of animals were subcutaneously (SC) administered MPO at 200 microg/kg/d (n = 4), or 50 microg/kg/d (n = 2), twice daily, or human serum albumin (HSA) (n = 10). Second series: The 600 cGy x-irradiated cohorts of animals were administered either MPO at 200 microg/kg/d, in a daily schedule (n = 4) or 0.1% autologous serum (AS), daily, SC (n = 11) for 23 days. MPO regardless of administration schedule (twice a day or every day) significantly reduced the mean durations of neutropenia (absolute neutrophil count [ANC] < 500/microL) and thrombocytopenia (platelet < 20,000/microL) versus respective control-treated cohorts. Mean neutrophil and platelet nadirs were significantly improved and time to recovery for neutrophils (ANC to < 500/microL) and platelets (PLT < 20,000/microL) were significantly enhanced in the MPO-treated cohorts versus controls. Red cell recovery was further improved relative to control-treated cohorts that received whole blood transfusions. Significant increases in bone marrow-derived clonogenic activity was observed by day 14 after TBI in MPO-treated cohorts versus respective time-matched controls. Thus, MPO, administered daily was as effective as a twice daily schedule for multilineage recovery in nonhuman primates after high-dose, radiation-induced myelosuppression.


Asunto(s)
Enfermedades de la Médula Ósea/etiología , Hematopoyesis/efectos de los fármacos , Factores de Crecimiento de Célula Hematopoyética/uso terapéutico , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Receptores de Factor Estimulante de Colonias de Granulocito/agonistas , Receptores de Interleucina-3/agonistas , Proteínas Recombinantes de Fusión , Irradiación Corporal Total/efectos adversos , Animales , Recuento de Células Sanguíneas/efectos de los fármacos , Transfusión Sanguínea , Linaje de la Célula , Ensayo de Unidades Formadoras de Colonias , Relación Dosis-Respuesta en la Radiación , Diseño de Fármacos , Evaluación Preclínica de Medicamentos , Factor Estimulante de Colonias de Granulocitos , Factores de Crecimiento de Célula Hematopoyética/química , Factores de Crecimiento de Célula Hematopoyética/farmacología , Interleucina-3 , Macaca mulatta , Masculino , Neutropenia/tratamiento farmacológico , Neutropenia/etiología , Ingeniería de Proteínas , Proteínas Recombinantes , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/etiología
11.
Exp Hematol ; 27(10): 1557-68, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10517498

RESUMEN

Myelopoietin (MPO), a multifunctional agonist of interleukin 3 and granulocyte colony-stimulating factor (G-CSF) receptors, was evaluated for its ability to mobilize hematopoietic colony-forming cells (CFC) and CD34+ cells relative to control cytokines in normal nonhuman primates. Additionally, the engraftment potential of MPO-mobilized CD34+ cells was assessed in lethally irradiated rhesus monkeys. Normal rhesus monkeys were administered either MPO (200 microg/kg/day), daniplestim (a high-affinity interleukin 3 receptor agonist) (100 microg/kg/day), G-CSF (100 microg/kg/day), or daniplestim coadministered with G-CSF (100 microg/kg/day each), subcutaneously for 10 consecutive days. The mobilization kinetics were characterized by peripheral blood (PB) complete blood counts, hematopoietic CFC [granulocyte-macrophage CFC (GM-CFC), megakaryocyte CFC (MK-CFC)], and the immunophenotype (CD34+ cells) of PB nucleated cells prior to and on day 3 to days 7, 10, 12, and 14, and at intervals up to day 28 following initiation of cytokine administration. A single large-volume leukapheresis was conducted on day 5 in an additional cohort (n = 10) of MPO-mobilized animals. Eight of these animals were transplanted with two doses of CD34+ cells/kg. A maximum 10-fold increase in PB leukocytes (white blood cells) (from baseline 7.8-12.3 x 10(3)/microL to approximately 90 x 10(3)/microL) was observed over day 7 to day 10 in the MPO, G-CSF, or daniplestim+G-CSF cohorts, whereas daniplestim alone stimulated a less than onefold increase. A sustained, maximal rise in PB-derived GM-CFC/mL was observed over day 4 to day 10 for the MPO-treated cohort, whereas the daniplestim+G-CSF, G-CSF alone, and daniplestim alone treated cohorts were characterized by a mean peak value on days 7, 6, and 18, respectively. Mean peak values for PB-derived GM-CFC/mL were greater for MPO (5,427/mL) than for daniplestim+G-CSF (3,534/mL), G-CSF alone (3,437/mL), or daniplestim alone (155/mL) treated cohorts. Mean peak values for CD34+ cells/mL were noted within day 4 to day 5 of cytokine administration: MPO (255/microL, day 5), daniplestim+G-CSF (47/microL, day 5), G-CSF (182/microL, day 4), and daniplestim (96/microL, day 5). Analysis of the mobilization data as area under the curve indicated that for total CFCs, GM-CFC, MK-CFC, or CD34+ cells, the MPO-treated areas under the curve were greater than those for all other experimental cohorts. A single, large-volume (3.0 x blood volume) leukapheresis at day 5 of MPO administration (PB: CD34+ cell/microL = 438 +/- 140, CFC/mL = 5,170 +/- 140) resulted in collection of sufficient CD34+ cells (4.31 x 10(6)/kg +/- 1.08) and/or total CFCs (33.8 x 10(4)/kg +/- 8.34) for autologous transplantation of the lethally irradiated host. The immunoselected CD34+ cells were transfused into autologous recipients (n = 8) at cell doses of 2 x 10(6)/kg (n = 5), and 4 x 10(6)/kg (n = 3) on the day of apheresis. Successful engraftment occurred with each cell dose. The data demonstrated that MPO is an effective and efficient mobilizer of PB progenitor cells and CD34+ cells, such that a single leukapheresis procedure results in collection of sufficient stem cells for transplantation and long term engraftment of lethally irradiated hosts.


Asunto(s)
Antígenos CD34/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Receptores de Factor Estimulante de Colonias de Granulocito/agonistas , Receptores de Interleucina-3/agonistas , Proteínas Recombinantes de Fusión/farmacología , Animales , Área Bajo la Curva , Recuento de Células Sanguíneas/efectos de los fármacos , División Celular/efectos de los fármacos , Combinación de Medicamentos , Filgrastim , Factor Estimulante de Colonias de Granulocitos/farmacología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Interleucina-3 , Leucocitos/citología , Leucocitos/efectos de los fármacos , Macaca mulatta , Masculino , Fragmentos de Péptidos , Péptidos/farmacología , Proteínas Recombinantes , Factores de Tiempo , Acondicionamiento Pretrasplante
12.
Blood ; 94(7): 2271-86, 1999 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-10498599

RESUMEN

Optimization of mobilization, harvest, and transduction of hematopoietic stem cells is critical to successful stem cell gene therapy. We evaluated the utility of a novel protocol involving Flt3-ligand (Flt3-L) and granulocyte colony-stimulating factor (G-CSF) mobilization of peripheral blood stem cells and retrovirus transduction using hematopoietic growth factors to introduce a reporter gene, murine CD24 (mCD24), into hematopoietic stem cells in nonhuman primates. Rhesus macaques were treated with Flt3-L (200 microgram/kg) and G-CSF (20 microgram/kg) for 7 days and autologous CD34(+) peripheral blood stem cells harvested by leukapheresis. CD34(+) cells were transduced with an MFGS-based retrovirus vector encoding mCD24 using 4 daily transductions with centrifugations in the presence of Flt3-L (100 ng/mL), human stem cell factor (50 ng/mL), and PIXY321 (50 ng/mL) in serum-free medium. An important and novel feature of this study is that enhanced in vivo engraftment of transduced stem cells was achieved by conditioning the animals with a low-morbidity regimen of sublethal irradiation (320 to 400 cGy) on the day of transplantation. Engraftment was monitored sequentially in the bone marrow and blood using both multiparameter flow cytometry and semi-quantitative DNA polymerase chain reaction (PCR). Our data show successful and persistent engraftment of transduced primitive progenitors capable of giving rise to marked cells of multiple hematopoietic lineages, including granulocytes, monocytes, and B and T lymphocytes. At 4 to 6 weeks posttransplantation, 47% +/- 32% (n = 4) of granulocytes expressed mCD24 antigen at the cell surface. Peak in vivo levels of genetically modified peripheral blood lymphocytes approached 35% +/- 22% (n = 4) as assessed both by flow cytometry and PCR 6 to 10 weeks posttransplantation. In addition, naïve (CD45RA(+) and CD62L(+)) CD4(+) and CD8(+) cells were the predominant phenotype of the marked CD3(+) T cells detected at early time points. A high level of marking persisted at between 10% and 15% of peripheral blood leukocytes for 4 months and at lower levels past 6 months in some animals. A cytotoxic T-lymphocyte response against mCD24 was detected in only 1 animal. This degree of persistent long-lived, high-level gene marking of multiple hematopoietic lineages, including naïve T cells, using a nonablative marrow conditioning regimen represents an important step toward the ultimate goal of high-level permanent transduced gene expression in stem cells.


Asunto(s)
Antígenos CD/genética , Marcadores Genéticos , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Glicoproteínas de Membrana , Animales , Antígenos CD/análisis , Linfocitos B/citología , Linfocitos B/fisiología , Células de la Médula Ósea/citología , Antígeno CD24 , Citometría de Flujo/métodos , Factor Estimulante de Colonias de Granulocitos/farmacología , Granulocitos/citología , Granulocitos/fisiología , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Leucaféresis/métodos , Macaca mulatta , Masculino , Proteínas de la Membrana/farmacología , Monocitos/citología , Monocitos/fisiología , Reacción en Cadena de la Polimerasa , Retroviridae , Factor de Células Madre/farmacología , Linfocitos T/citología , Linfocitos T/fisiología , Recolección de Tejidos y Órganos/métodos , Irradiación Corporal Total
13.
Stem Cells ; 16 Suppl 2: 143-54, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-11012186

RESUMEN

This study evaluated the ability of daniplestim, a high affinity interleukin 3 receptor agonist, to enhance the hematopoietic response of Mpl ligand (Mpl-L) administration in nonhuman primates following severe, radiation-induced myelosuppression. Rhesus monkeys were total body x-irradiated (TBI) to 600 cGy, midline tissue dose. Beginning on day 1 post-TBI, animals were s.c. administered daniplestim (100 microg/kg bid; n = 4), Mpl-L (10 microg/kg qd; n = 3), daniplestim (100 microg/kg bid) plus Mpl-L (10 microg/kg qd) (n = 4) or 0.1% autologous serum (AS) (n = 11) for 18 days. CBCs were monitored for 60 d after TBI. The duration of thrombocytopenia (platelet count; PLT <20,000/microl) was significantly decreased by the administration of daniplestim (6.5 d, p = .01), Mpl-L (3.0 d, p = .003) and the coadministered daniplestim/Mpl-L (1.3 d, p = .001) compared to controls (10.4 d). As monotherapy Mpl-L but not daniplestim significantly improved the PLT nadir (21,000/microl, p = .023 and 5,000/microl, p = .266, respectively) compared to the control (3,000/microl). The combined administration of daniplestim and Mpl-L significantly improved the PLT nadir (28,000/microl, p = .007) compared to both the control cohort (3,000/microl) and the daniplestim only cohort (5,000/microl, p = .043). Recovery of PLT to preirradiation values occurred earlier in the daniplestim only (d 21) or the daniplestim/Mpl-L cohorts (d 18) than in the Mpl-L only or control cohorts (d 28, d 29, respectively). The administration of daniplestim or Mpl-L alone neither shortened the duration of neutropenia (ANC<500/microl) compared to the controls (15.8 d, 16.0 d versus 16.2 d, respectively), nor improved the recovery time of neutrophils to baseline values (d 22, d 25, and d 23, respectively). The ANC nadir was significantly improved by daniplestim alone but not Mpl-L administration (76/microl, p = .001 and 50/microl, p = .093, respectively) compared to the controls (8/microl). Coadministration of daniplestim and Mpl-L significantly improved the ANC nadir (196/microl, p = .001) compared to either the AS- or the monotherapy-treated cohorts. Also the duration of neutropenia observed in the AS-controls (16.2 d) was significantly reduced in the daniplestim/Mpl-L cohort (10.8 d, p = .002). The combined administration of daniplestim and Mpl-L significantly improved hematopoietic recovery and further enhanced the stimulatory effect of cytokine monotherapy, as well as reducing clinical support requirements after radiation-induced bone marrow myelosuppression.


Asunto(s)
Médula Ósea/efectos de los fármacos , Citocinas/farmacología , Interacciones Farmacológicas/fisiología , Hematopoyesis/efectos de los fármacos , Péptidos/farmacología , Trombopoyetina/farmacocinética , Animales , Antibacterianos/farmacología , Transfusión Sanguínea , Médula Ósea/metabolismo , Médula Ósea/efectos de la radiación , Cricetinae , Citocinas/metabolismo , Modelos Animales de Enfermedad , Combinación de Medicamentos , Hematopoyesis/fisiología , Hematopoyesis/efectos de la radiación , Terapia de Inmunosupresión/métodos , Interleucina-3 , Macaca mulatta , Masculino , Neutropenia/tratamiento farmacológico , Neutropenia/fisiopatología , Fragmentos de Péptidos , Recuperación de la Función/efectos de los fármacos , Recuperación de la Función/fisiología , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/fisiopatología , Trombopoyetina/metabolismo , Factores de Tiempo
15.
J Clin Invest ; 97(9): 2145-51, 1996 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-8621805

RESUMEN

This study compared the therapeutic potential of recombinant, native versus pegylated megakaryocyte growth and development factor (rMGDF and PEG-rMGDF, respectively), as well as that of the combined administration of PEG-rMGDF and r-methionyl human granulocyte colony-stimulating factor (r-metHuG-CSF) on hematopoietic reconstitution after 700 cGy, 60Co gamma, total body irradiation in nonhuman primates. After total body irradiation, animals received either rMGDF, PEG-rMGDF, r-metHuG-CSF, PEG-rMGDF and r-metHuG-CSF or HSA. Cytokines in all MGDF protocols were administered for 21-23 d. Either rMGDF, PEG-rMGDF, or PEG-rMGDF and r-metHuG-CSF administration significantly diminished the thrombocytopenic duration (platelet count (PLT) < 20,000 per microliter)to o.25, 0, 0.5 d, respectively, and the severity of the PLT nadir (28,000, 43,000, and 30,000 per microliter, respectively) as compared with the controls (12.2 d duration, nadir 4,000 per microliter), and elicited an earlier PLT recovery. Neutrophil regeneration was augmented in all cytokine protocols and combined PEG-rMGDF and r-metHuG-CSF further decreased the duration of neutropenia compared with r-metHuG-CSF alone. These data demonstrated that the administration of PEG-rMGDF significantly induced bone marrow regeneration versus rMGDF, and when combined with r-metHuG-CSF significantly enhanced multilineage hematopoietic recovery with no evidence of lineage competition.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/administración & dosificación , Hematopoyesis/efectos de los fármacos , Megacariocitos/patología , Traumatismos Experimentales por Radiación/sangre , Trombopoyetina/administración & dosificación , Animales , Linaje de la Célula , Ensayo de Unidades Formadoras de Colonias , Hematopoyesis/efectos de la radiación , Humanos , Macaca mulatta , Masculino , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/terapia , Proteínas Recombinantes/administración & dosificación , Irradiación Corporal Total
16.
Blood ; 87(10): 4129-35, 1996 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-8639770

RESUMEN

Combination cytokine therapy continues to be evaluated in an effort to stimulate multilineage hematopoietic reconstitution after bone marrow myelosuppression. This study evaluated the efficacy of combination therapy with the synthetic interleukin-3 receptor agonist, Synthokine-SC55494, and recombinant methionyl human granulocyte colony-stimulating factor (rhG-CSF) on platelet and neutrophil recovery in nonhuman primates exposed to total body 700 cGy 60Co gamma radiation. After irradiation on day (d) 0, cohorts of animals subcutaneously received single-agent protocols of either human serum albumin (HSA; every day [QD], 15 micrograms/kg/d, n = 10), Synthokine (twice daily [BID], 100, micrograms/kg/d, n = 15), rhG-CSF (QD, 10 micrograms/kg/d, n = 5), or a combination of Synthokine and rhG-CSF (BID, 100 and 10 micrograms/kg/d, respectively, n = 5) for 23 days beginning on d1. Complete blood counts were monitored for 60 days postirradiation and the durations of neutropenia (absolute neutrophil count < 500/microL) and thrombocytopenia (platelet count < 20,000/microL) were assessed. Animals were provided clinical support in the form of antibiotics, fresh irradiated whole blood, and fluids. All cytokine protocols significantly (P < .05) reduced the duration thrombocytopenia versus the HSA-treated animals. Only the combination protocol of Synthokine + rhG-CSF and rhG-CSF alone significantly shortened the period neutropenia (P < .05). The combined Synthokine/rhG-CSF protocol significantly improved platelet nadir versus Synthokine alone and HSA controls and neutrophil nadir versus rhG-CSF alone and HSA controls. All cytokine protocols decreased the time to recovery to preirradiation neutrophil and platelet values. The Synthokine/rhG-CSF protocol also reduced the transfusion requirements per treatment group to 0 among 5 animals as compared with 2 among 5 animals for Synthokine alone, 8 among 5 animals for rhG-CSF, and 17 among 10 animals for HSA. These data showed that the combination of Synthokine, SC-55494, and rhG-CSF further decreased the cytopenic periods and nadirs for both platelets and neutrophils relative to Synthokine and rhG-CSF monotherapy and suggest that this combination therapy would be effective against both neutropenia and thrombocytopenia consequent to drug- or radiation- induced myelosuppression.


Asunto(s)
Anemia Aplásica/terapia , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Péptidos/uso terapéutico , Traumatismos Experimentales por Radiación/terapia , Receptores de Interleucina-3/antagonistas & inhibidores , Irradiación Corporal Total/efectos adversos , Anemia Aplásica/etiología , Animales , Transfusión Sanguínea , Terapia Combinada , Evaluación Preclínica de Medicamentos , Quimioterapia Combinada , Filgrastim , Humanos , Interleucina-3 , Leucocitos/efectos de los fármacos , Macaca mulatta , Masculino , Neutropenia/etiología , Neutropenia/terapia , Fragmentos de Péptidos , Proteínas Recombinantes/uso terapéutico , Albúmina Sérica/uso terapéutico , Trombocitopenia/etiología , Trombocitopenia/terapia
17.
Blood ; 87(2): 581-91, 1996 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-8555480

RESUMEN

The synthetic cytokine (Synthokine) SC-55494 is a high-affinity interleukin-3 (IL-3) receptor ligand that stimulates greater in vitro multilineage hematopoietic activity than native IL-3, while inducing no significant increase in inflammatory activity relative to native IL-3. The aim of this study was to investigate the in vivo hematopoietic response of rhesus monkeys receiving Synthokine after radiation-induced marrow aplasia. Administration schedule and dose of Synthokine were evaluated. All animals were total-body irradiated (TBI) with 700 cGy 60Co gamma radiation on day 0. Beginning on day 1, cohorts of animals (n = 5) received Synthokine subcutaneously (SC) twice daily with 25 micrograms/kg/d or 100 micrograms/kg/d for 23 days or 100 micrograms/kg/d for 14 days. Control animals (n = 9) received human serum albumin SC once daily at 15 micrograms/kg/d for 23 days. Complete blood counts were monitored for 60 days postirradiation and the durations of neutropenia (NEUT; absolute neutrophil count [ANC] < 500/microL) and thrombocytopenia (THROM; platelet count < 20,000/microL) were assessed. Synthokine significantly (P < .05) reduced the duration of THROM versus the HSA-treated animals regardless of dose or protocol length. The most striking reduction was obtained in the animals receiving 100 micrograms/kg/d for 23 days (THROM = 3.5 v 12.5 days in HSA control animals). Although the duration of NEUT was not significantly altered, the depth of the nadir was significantly lessened in all animal cohorts treated with Synthokine regardless of dose versus schedule length. Bone marrow progenitor cell cultures indicated a beneficial effect of Synthokine on the recovery of granulocyte-macrophage colony-forming units that was significantly higher at day 24 post-TBI in both cohorts treated at 25 and 100 micrograms/kg/d for 23 days relative to the control animals. Plasma pharmacokinetic parameters were evaluated in both normal and irradiated animals. Pharmacokinetic analysis performed in irradiated animals after 1 week of treatment suggests an effect of repetitive Synthokine schedule and/or TBI on distribution and/or elimination of Synthokine. These data show that the Synthokine, SC55 94, administered therapeutically post-TBI, significantly enhanced platelet recovery and modulated neutrophil nadir and may be clinically useful in the treatment of the myeloablated host.


Asunto(s)
Anemia Aplásica/terapia , Hematopoyesis/efectos de los fármacos , Péptidos/uso terapéutico , Traumatismos Experimentales por Radiación/terapia , Anemia Aplásica/etiología , Animales , Recuento de Células Sanguíneas/efectos de los fármacos , Transfusión Sanguínea , Ensayo de Unidades Formadoras de Colonias , Terapia Combinada , Semivida , Humanos , Interleucina-3/farmacocinética , Interleucina-3/uso terapéutico , Macaca mulatta , Masculino , Neutropenia/etiología , Neutropenia/terapia , Fragmentos de Péptidos , Péptidos/farmacocinética , Péptidos/farmacología , Traumatismos Experimentales por Radiación/etiología , Albúmina Sérica/administración & dosificación , Trombocitopenia/etiología , Trombocitopenia/terapia , Irradiación Corporal Total/efectos adversos
18.
Blood ; 86(1): 54-9, 1995 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-7795256

RESUMEN

Megakaryocyte growth and development factor (MGDF) is a novel cytokine that binds to the c-mpl receptor and stimulates megakaryocyte development in vitro and in vivo. This report describes the ability of recombinant human (r-Hu) MGDF to affect megakaryocytopoiesis in normal nonhuman primates. r-HuMGDF was administered subcutaneously to normal, male rhesus monkeys once per day for 10 consecutive days at dosages of 2.5, 25, or 250 micrograms/kg of body weight. Bone marrow and peripheral blood were assayed for clonogenic activity and peripheral blood counts were monitored. Circulating platelet counts increased significantly (P < .05) for all doses within 6 days of r-HuMGDF administration and reached maximal levels between day 12 and day 14 postcytokine administration. The 2.5, 25.0, and 250.0 micrograms/kg/d doses elicited peak mean platelet counts that were 592%, 670%, and 449% of baseline, respectively. Bone marrow-derived clonogenic data showed significant increases in the concentration of megakaryocyte (MEG)-colony-forming unit (CFU) and granulocyte-erythroid-macrophage-megakaryocyte (GEMM)-CFU, whereas that of granulocyte-macrophage (GM)-CFU and burst-forming unit-erythroid (BFU-e) remained unchanged during the administration of r-HuMGDF. These data show that r-HuMGDF is a potent stimulator of thrombocytopoiesis in the normal nonhuman primate.


Asunto(s)
Hematopoyesis/efectos de los fármacos , Megacariocitos/efectos de los fármacos , Trombopoyetina/farmacología , Animales , Médula Ósea/efectos de los fármacos , Células de la Médula Ósea , Ensayo de Unidades Formadoras de Colonias , Humanos , Macaca mulatta , Masculino , Megacariocitos/citología , Recuento de Plaquetas/efectos de los fármacos , Proteínas Recombinantes/farmacología
19.
Blood ; 84(11): 3675-8, 1994 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-7949122

RESUMEN

The therapeutic efficacy of recombinant human leukemia inhibitory factor (LIF) was examined in a nonhuman primate model of radiation-induced marrow aplasia. Rhesus monkeys received 450 cGy of total-body, 1:1 mixed neutron:gamma radiation. For 23 days thereafter, each monkey received a daily subcutaneous injection of LIF or human serum albumin (HSA) at a dose of 15 micrograms/kg body weight. Complete blood counts and white blood cell differentials were monitored for 60 days postirradiation. Administration of LIF significantly decreased (P < or = .05) the duration of thrombocytopenia (platelet count < 30,000 or 20,000/microL), ie, 9.3 days or 6.3 days, respectively, versus the HSA-treated control monkeys, 12.2 days or 10.2 days, respectively. Treatment with LIF did not alter the duration of neutropenia (absolute neutrophil count < 1,000/microL) as compared with the HSA-treated control monkeys. Cytokine administration did not exacerbate the radiation-induced anemia observed in the HSA-treated control monkeys.


Asunto(s)
Anemia Aplásica/terapia , Inhibidores de Crecimiento/uso terapéutico , Interleucina-6 , Linfocinas/uso terapéutico , Neutropenia/terapia , Traumatismos Experimentales por Radiación/terapia , Proteínas Recombinantes de Fusión/uso terapéutico , Trombocitopenia/terapia , Anemia Aplásica/etiología , Animales , Evaluación Preclínica de Medicamentos , Hematopoyesis/efectos de los fármacos , Factor Inhibidor de Leucemia , Macaca mulatta , Masculino , Neutropenia/etiología , Albúmina Sérica/uso terapéutico , Trombocitopenia/etiología
20.
Blood ; 84(8): 2515-22, 1994 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-7919369

RESUMEN

Using a nonhuman-primate model of radiation-induced bone marrow aplasia, we examined whether the single, concomitant, or sequential administration of recombinant human interleukin-3 (IL-3) and IL-6 would promote bone marrow regeneration measured by an increase in circulating platelets (PLT) and neutrophils (PMN). Rhesus monkeys were irradiated at 450 cGy and were randomly assigned to one of five treatment protocols, receiving IL-6; IL-3; combined IL-6 and IL-3; sequential IL-3 and IL-6; or human serum albumin (HSA) as a control. Cytokines or HSA were administered at total dosages of 15 micrograms/kg/day. Complete blood counts and white blood cell differentials were monitored for 60 days postirradiation. Both IL-3 and IL-6 significantly enhanced the regeneration of PLTs and decreased the duration of thrombocytopenia (P = .005) without affecting PMN recovery. The radiation-induced anemia that was observed in the HSA-treated controls was less severe and resolved more quickly in the IL-6 treated animals. Sequential IL-3/IL-6 significantly increased the production of PLTs when compared with the HSA-treated controls (P = .003) and monkeys receiving concomitant IL-3/IL-6 (P = .041) but did not alter PMN levels significantly (P = .80). Coadministration of IL-6 and IL-3 did not enhance PLT but improved PMN recovery over IL-6 alone. In this primate model of marrow aplasia, IL-6 significantly enhanced the regeneration of PLTs but had no significant effect on PMN production, and did not exacerbate radiation-induced anemia. Furthermore, the use of sequentially administered IL-3 and IL-6 may improve PLT recovery as compared with concurrent IL-3/IL-6 administration, although this protocol is not significantly different in effect than either cytokine alone.


Asunto(s)
Enfermedades de la Médula Ósea/tratamiento farmacológico , Hematopoyesis , Interleucina-3/uso terapéutico , Interleucina-6/uso terapéutico , Traumatismos Experimentales por Radiación , Anemia/tratamiento farmacológico , Anemia/etiología , Animales , Médula Ósea/patología , Enfermedades de la Médula Ósea/etiología , Enfermedades de la Médula Ósea/patología , Quimioterapia Combinada , Interleucina-3/administración & dosificación , Interleucina-6/administración & dosificación , Recuento de Leucocitos , Macaca mulatta , Masculino , Neutropenia/tratamiento farmacológico , Neutropenia/etiología , Recuento de Plaquetas , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/uso terapéutico , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA